WO2006135479A2 - Anti-scarring agents, therapeutic compositions, and use thereof - Google Patents

Anti-scarring agents, therapeutic compositions, and use thereof Download PDF

Info

Publication number
WO2006135479A2
WO2006135479A2 PCT/US2006/013030 US2006013030W WO2006135479A2 WO 2006135479 A2 WO2006135479 A2 WO 2006135479A2 US 2006013030 W US2006013030 W US 2006013030W WO 2006135479 A2 WO2006135479 A2 WO 2006135479A2
Authority
WO
WIPO (PCT)
Prior art keywords
inhibitor
antagonist
compound
agent
receptor
Prior art date
Application number
PCT/US2006/013030
Other languages
French (fr)
Other versions
WO2006135479A3 (en
Inventor
William L. Hunter
Philip M. Toleikis
David M. Gravett
Arpita Maiti
Richard T. Liggins
Aniko Takacs-Cox
Rui Avelar
Pierre E. Signore
Troy A. E. Loss
Anne Hutchinson
Gaye Mcdonald-Jones
Fara Lakhani
Original Assignee
Angiotech International Ag
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Angiotech International Ag filed Critical Angiotech International Ag
Publication of WO2006135479A2 publication Critical patent/WO2006135479A2/en
Publication of WO2006135479A3 publication Critical patent/WO2006135479A3/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61LMETHODS OR APPARATUS FOR STERILISING MATERIALS OR OBJECTS IN GENERAL; DISINFECTION, STERILISATION OR DEODORISATION OF AIR; CHEMICAL ASPECTS OF BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES; MATERIALS FOR BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES
    • A61L27/00Materials for grafts or prostheses or for coating grafts or prostheses
    • A61L27/50Materials characterised by their function or physical properties, e.g. injectable or lubricating compositions, shape-memory materials, surface modified materials
    • A61L27/54Biologically active materials, e.g. therapeutic substances
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61LMETHODS OR APPARATUS FOR STERILISING MATERIALS OR OBJECTS IN GENERAL; DISINFECTION, STERILISATION OR DEODORISATION OF AIR; CHEMICAL ASPECTS OF BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES; MATERIALS FOR BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES
    • A61L29/00Materials for catheters, medical tubing, cannulae, or endoscopes or for coating catheters
    • A61L29/14Materials characterised by their function or physical properties, e.g. lubricating compositions
    • A61L29/16Biologically active materials, e.g. therapeutic substances
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61LMETHODS OR APPARATUS FOR STERILISING MATERIALS OR OBJECTS IN GENERAL; DISINFECTION, STERILISATION OR DEODORISATION OF AIR; CHEMICAL ASPECTS OF BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES; MATERIALS FOR BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES
    • A61L31/00Materials for other surgical articles, e.g. stents, stent-grafts, shunts, surgical drapes, guide wires, materials for adhesion prevention, occluding devices, surgical gloves, tissue fixation devices
    • A61L31/14Materials characterised by their function or physical properties, e.g. injectable or lubricating compositions, shape-memory materials, surface modified materials
    • A61L31/16Biologically active materials, e.g. therapeutic substances
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61LMETHODS OR APPARATUS FOR STERILISING MATERIALS OR OBJECTS IN GENERAL; DISINFECTION, STERILISATION OR DEODORISATION OF AIR; CHEMICAL ASPECTS OF BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES; MATERIALS FOR BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES
    • A61L2300/00Biologically active materials used in bandages, wound dressings, absorbent pads or medical devices
    • A61L2300/40Biologically active materials used in bandages, wound dressings, absorbent pads or medical devices characterised by a specific therapeutic activity or mode of action

Definitions

  • This invention relates generally to devices and compositions that include a therapeutic agent (e.g., a fibrosis-inhibiting agent or an anti- infective agent), and to methods of making and using such compositions.
  • a therapeutic agent e.g., a fibrosis-inhibiting agent or an anti- infective agent
  • tubular _ anatomical structures such as blood vessels, the gastrointestinal tract, the male and female reproductive tract, the urinary tract, sinuses, spinal nerve root canals, lacrimal ducts, Eustachian tubes, the auditory canal, and the respiratory tract
  • stenosis or narrowing
  • Stenosis occurs in response to trauma to the epithelial lining or the entire body tube during the procedure, including virtually any manipulation which attempts to relieve obstruction of the passageway, and is a major factor limiting the effectiveness of invasive treatments for a variety of diseases to be described later.
  • Stenosis (or "restenosis” if the problem recurs after an initially successful attempt to open a blocked passageway) is a form of response to injury leading to wall thickening, narrowing of the lumen, and loss of function in the tissue supplied by the particular passageway.
  • Physical injury during an interventional procedure results in damage to epithelial lining of the tube and the smooth muscle cells (SMCs) that make up the wall.
  • SMCs smooth muscle cells
  • the damaged cells, particularly SMCs release cytokines, which recruit inflammatory cells such as macrophages, lymphocytes and neutrophils (i.e., which are some of the known white blood cells) into the area.
  • the white blood cells in turn release a variety of additional cytokines, growth factors, and tissue degrading enzymes that influence the behavior of the constituent cells of the wall (primarily epithelial cells and SMCs). Stimulation of the SMCs induces them to migrate into the inner aspect of the body passageway (often called the "intima”), proliferate and secrete an extracellar matrix - effectively filling all or parts of the lumen with reactive, fibrous scar tissue. Collectively, this creates a thickening of the intimal layer (known in some tissues as "neointimal hyperplasia") that narrows the lumen of the passageway and can be significant enough to obstruct its lumen.
  • Polymeric compositions particularly those that include synthetic polymers or a combination of synthetic and naturally occurring polymers, have been used in a variety of medical applications, such as the prevention of surgical adhesions, tissue engineering, and as bioadhesive materials.
  • U.S. Patent No. 5,162,430 describes the use of collagen- synthetic polymer conjugates prepared by covalently binding collagen to synthetic hydrophilic polymers such as various derivatives of polyethylene glycol.
  • synthetic hydrophilic polymers such as various derivatives of polyethylene glycol.
  • U.S. Patent No. 5,328,955 various activated forms of polyethylene glycol and various linkages are described, which can be used to produce collagen-synthetic polymer conjugates having a range of physical and chemical properties.
  • EP 0732 109 A1 discloses a crosslinked biomaterial composition that is prepared using a hydrophobic crosslinking agent, or a mixture of hydrophilic and hydrophobic crosslinking agents.
  • U.S. Patent No. 5,614,587 describes bioadhesives that comprise collagen that is crosslinked using a multifunctionally activated synthetic hydrophilic polymer.
  • composition useful in the prevention of surgical adhesions comprising a substrate material and an anti-adhesion binding agent, where the substrate material may comprise collagen and the binding agent may comprise at least one tissue-reactive functional group and at least one substrate-reactive functiona) group.
  • U.S. application Ser. No. 08/476,825, filed Jun. 7, 1995 discloses bioadhesive compositions comprising collagen crosslinked using a multifunctionally activated synthetic hydrophilic polymer, as well as methods of using such compositions to effect adhesion between a first surface and a second surface, wherein at least one of the first and second surfaces may be a native tissue surface.
  • 5,874,500 describes a crosslinked polymer composition that comprises one component having multiple nucleophilic groups and another component having multiple electrophilic groups. Covalently bonding of the nucleophilic and electrophilic groups forms a three dimensional matrix that has a variety of medical uses including tissue adhesion, surface coatings for synthetic implants,, and drug delivery. More recent developments include the addition of a third component having either nucleophilic or electrophilic groups, as is described in U.S. Patent No. 6,458,889 to Trollsas et af. US 5,874,500, US 6,051,648 and US 6,312,725 disclose the in situ crosslinking or crosslinked polymers, in particular poly(ethylene glycol) based polymers, to produce a crosslinked composition.
  • the present invention provides compositions for delivery of selected therapeutic agents via medical implants or implantable medical devices, as well as methods for making and using these implants and devices.
  • drug- coated or drug-impregnated implants and medical devices are provided which reduce fibrosis in the tissue surrounding the device or implant, or inhibit scar development on the device/implant surface, thus enhancing the efficacy the procedure.
  • fibrosis is inhibited by local or systemic release of specific pharmacological agents that become localized to the adjacent tissue.
  • an implant or device is adapted to release an agent that inhibits fibrosis or regeneration through one or more of the mechanisms sited herein.
  • a medical device or implant comprising the step of coating (e.g., spraying, dipping, wrapping, or administering drug through) a medical device or implant.
  • the implant or medical device can be constructed so that the device itself is comprised of materials which inhibit fibrosis in or around the implant.
  • a wide variety of medical devices and implants may be utilized within the context of the present invention, depending on the site and nature of treatment desired.
  • intravascular devices comprising an implant or device, wherein the implant or device is in combination with an agent which inhibits fibrosis in vivo.
  • the implant or device is further coated with a composition or compound, which delays the onset of activity of the fibrosis-inhibiting agent for a period of time after implantation.
  • a composition or compound which delays the onset of activity of the fibrosis-inhibiting agent for a period of time after implantation.
  • agents include heparin, PLGA/MePEG, PLA, and polyethylene glycol.
  • the fibrosis-inhibiting implant or device is activated before, during, or after deployment (e.g., an inactive agent on the device is first activated to one that reduces or inhibits an in vivo fibrotic reaction).
  • a device or implant is coated on one aspect, portion or surface with a composition which inhibits fibrosis, as well as being coated with a composition or compound which promotes scarring on another aspect, portion or surface of the device.
  • agents that promote fibrosis and scarring include silk, wool, silica, bleomycin, neomycin, talcum powder, metallic beryllium, and copper as well as analogues and derivatives thereof.
  • methods for treating patients undergoing surgical, endoscopic or minimally invasive therapies where a medical device or implant is placed as part of the procedure.
  • inhibits fibrosis or stenosis refers to a statistically significant decrease in the amount of scar tissue in or around the device or an improvement in the luminal area of the device/implant, which may or may not result in a permanent prohibition of any complications or failures of the device/implant.
  • the pharmaceutical agents and compositions are utilized to create novel drug-coated implants and medical devices that reduce the foreign body response to implantation and limit the growth of reactive tissue on the surface of, or around in the tissue surrounding the device, such that performance is enhanced.
  • the devices are used to maintain body lumens or passageways such as blood vessels, the gastrointestinal tract, the male and female reproductive tract, the urinary tract, bony foramena (e.g., sinuses, spinal nerve root canals, lacrimal ducts, Eustachian tubes, the auditory canal), and the respiratory tract, where obstruction of the device by scar tissue in the post-procedural period leads to the adverse clinical sequela or failure of the intervention.
  • Medical devices and implants coated with selected pharmaceutical agents designed to prevent scar tissue overgrowth and preserve patency can offer significant clinical advantages over uncoated devices.
  • the present invention is directed to devices that comprise a medical implant and at least one of (i) an anti- scarring agent and (ii) a composition that comprises an anti-scarring agent.
  • the agent is present so as to inhibit scarring that can otherwise occur when the implant is placed within an animal.
  • the present invention is directed to methods wherein both an implant and at least one of (i) an anti-scarring agent and (ii) a composition that comprises an anti- scarring agent, are placed into an animal, and the agent inhibits scarring that can otherwise occur.
  • the present invention provides the following: a device, comprising a medical device and an anti- scarring agent or a composition comprising an anti-scarring agent, wherein the agent inhibits scarring; a device, comprising an intravascular device and an anti-scarring agent or a composition comprising an anti-scarring agent, wherein the agent inhibits scarring; a device, comprising a a gastrointestinal stent and an anti-scarring agent or a composition comprising an anti- scarring agent, wherein the agent inhibits scarring; a device, comprising a tracheal and bronchial stent and an anti-scarring agent or a composition comprising an anti-scarring agent, wherein the agent inhibits scarring; a device, comprising a genital urinary stent and an anti-scarring agent or a composition comprising an anti-scarring agent, wherein the agent inhibits scarring; a device, comprising an ear and nose stent and an anti
  • the anti-fibrotic agent may be present in a composition along with a polymer.
  • the polymer is biodegradable.
  • the polymer is non-biodegradable.
  • the present invention also provides methods.
  • the present invention provides methods whereby a specified device is implanted into an animal, and a specified agent associated with the device inhibits scarring that can otherwise occur.
  • the devices identified herein may be a "specified device”
  • each of the anti-scarring agents identified herein may be an "anti-scarring agent” where the present invention provides, in independent embodiments, for each possible combination of the device and the agent.
  • the agent may be associated with the device prior to the device being placed within the animal.
  • the agent or composition comprising the agent
  • the agent may be coated onto an implant, and the resulting device then placed within the animal.
  • the agent may be independently placed within the animal in the vicinity of where the device is to be, or is being, placed within the animal.
  • the agent may be sprayed or otherwise placed onto the tissue that will be contacting the medical implant or may otherwise undergo scarring.
  • the present invention provides, in independent aspects: a method for inhibiting scarring comprising placing a medical device and an anti-scarring agent or a composition comprising an anti-scarring agent into an animal host, wherein the agent inhibits scarring; a method for inhibiting scarring comprising placing an intravascular device and an anti-scarring agent or a composition comprising an anti-scarring agent into an animal host, wherein the agent inhibits scarring; a method for inhibiting scarring comprising placing a gastrointestinal stent and an anti-scarring agent or a composition comprising an anti-scarring agent into an animal host, wherein the agent inhibits scarring; a method for inhibiting scarring comprising placing a tracheal and bronchial stent and an anti-scarring agent or a composition comprising an anti-scarring agent into an animal host, wherein the agent inhibits scarring; a method for inhibiting scarring comprising placing a genital urinary stent and an anti-scarring agent
  • the agent may be present in a composition along with a polymer.
  • the polymer is biodegradable.
  • the polymer is non-biodegradable.
  • the present invention provides compositions that contain both an anti-fibrotic agent and either a polymer or a pre- polymer, i.e., a compound that forms a polymer.
  • these compositions are formed in-situ when precursors thereof are delivered to a site in the body, or a site on an implant.
  • the compositions of the invention include the crosslinked reaction product that forms when two compounds (a multifunctional polynucleophilic compound and a multifunctional polyelectrophilic compound) are delivered to a site in a host (in other words, a patient) in the presence of an anti-fibrotic agent.
  • the compositions of the invention also include a mixture of anti-fibrotic agent and a polymer, where the composition can be delivered to a site in a patient's body to achieve beneficial affects, e.g., the beneficial affects described herein.
  • the polymers themselves are useful in various methods, including the prevention of surgical adhesions.
  • the present invention provides methods for treating and/or preventing surgical adhesions.
  • the surgical adhesions can be the result of, for example, spinal or neurosurgical procedures, of gynecological procedures, of abdominal procedures, of cardiac procedures, of orthopedic procedures, of reconstructive procedures, and cosmetic procedures.
  • the present invention provides methods for treating or preventing inflammatory arthritis, such as osteoarthritis and rheumatoid arthritis.
  • the method includes delivering to patient in need thereof an anti-fibrotic agent, optionally with a polymer.
  • the present invention provides for the prevention of cartilage loss as can occur, for example after a joint injury.
  • the method includes delivering to the joint of the patient in need therof an anti-fibrotic agent, optionally with a polymer.
  • the present invention provides for treating hypertrophic scars and keloids.
  • the method includes delivering to the scar or keloid of the patient in need thereof an anti-fibrotic agent, optionally with a polymer.
  • the present invention provides a method for the treatment of vascular disease, e.g., stenosis, restenosis or atherosclerosis.
  • the method includes the perivascular delivery of an anti- fibrotic agent.
  • the present invention provides a method for implanting a medical device comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with i) an anti- fibrotic agent, ii) an anti-infective agent, iii) a polymer; iv) a composition comprising an anti-fibrotic agent and a polymer, v) a composition comprising an anti-infective agent and a polymer, or vi) a composition comprising an anti-fibrotic agent, an anti-infective agent and a polymer, and (b) implanting the medical device into the host.
  • the invention provides: a method for implanting a medical device comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with an anti-fibrotic agent, and (b) implanting the medical device into the host; a method for implanting a medical device comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with an anti-infective agent, and (b) implanting the medical device into the host; a method for implanting a medical device comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with a polymer; and (b) implanting the medical device into the host; a method for implanting a medical device comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with a composition comprising an anti-fibrotic agent and
  • the anti-fibrotic agent may be one or more of the following: 1) an anti-fibrotic agent that inhibits cell regeneration, 2) an anti-fibrotic agent that inhibits angiogenesis, 3) an anti-fibrotic agent that inhibits fibroblast migration, 4) an anti-fibrotic agent that inhibits fibroblast proliferation, 5) an anti-fibrotic agent that inhibits deposition of extracellular matrix, 6) an anti- fibrotic agent inhibits tissue remodeling, 7) an adensosine A2A receptor antagonist, 8) an AKT inhibitor, 9) an alpha 2 integrin antagonist, wherein the alpha 2 integrin antagonist is Pharmaprojects No.
  • an alpha 4 integrin antagonist an alpha 4 integrin antagonist
  • 11) an alpha 7 nicotinic receptor agonist 12) an angiogenesis inhibitor selected from the group consisting of AG- 12,958 (Pfizer), ATN-161 (Attenuon LLC), neovastat, an angiogenesis inhibitor from Jerina AG (Germany), NM-3 (Mercian), VGA-1155 (Taisho), FCE-26644 (Pfizer), FCE-26950 (Pfizer), FPMA (Meiji Daries), FR-111142 (Fujisawa), GGTI-298, GM-1306 (Ligand), GPA-1734 (Novartis), NNC-47- 0011 (Novo Nordisk), herbamycin (Nippon Kayaku), lenalidomide (Celegene), IP-10 (NIH), ABT-828 (Abbott), KIN-841 (Tokushima University, Japan), SF-1126 (Semafore Pharmaceutical
  • a KDR inhibitor from LG Life Sciences, CT-6685 and CT-6729 (UCB), KRN-633 and KRN-951 (Kirin Brewery), OSI-930 (OSI Pharmaceuticals), SP-5.2 (Supratek Pharma), SU- 11657 (Pfizer), a Tie-2 antagonist (Hybrigenics), SU 1498 (a VEGF-R inhibitor), a VEGFR-2 kinase inhibitor (Bristol-Myers Squibb), XL-647 (Exelixis), a KDR inhibitor from Abbott Laboratories, sorafenib tosylate, and an analogue or derivative thereof, 39) an endotoxin antagonist, 40) an epothilone and tubulin binder, 41) an estrogen receptor antagonist, 42) an FGF inhibitor, 43) a famexyl transferase inhibitor, 44) a farnesyltransferase inhibitor selected from the group of A-197574 (Abbott),
  • an FLT-3 kinase inhibitor 46a) an FGF receptor kinase inhibitor, 47) a fibrinogen antagonist selected from the group consisting of AUV-201 (Auvation), MG-13926 (Sanofi-Aventis), plasminogen activator (CAS No. 105913-11-9) (from Sanofi-Aventis or UCB), plasminogen activator-2 (tPA-2) (Sanofi-Aventis), pro-urokinase (CAS Np.
  • a heat shock protein 90 antagonist selected from the group consisting of SRN-005 (Sirenade), geldanamycin, NSC-33050 (17- allylaminogeldanamycin; 17-AAG), 17-dimethylaminoethylamino ⁇ 17- demethoxy-geldanamycin (17-DMAG), rifabutin (rifamycin XIV, 1',4- didehydro-1-deoxy-1 ,4-dihydro-5'-(2-methylpropyl)-1-oxo-), radicicol from Humicola fuscoatra (CAS No.
  • an atherosclerosis therapeutic from Lipid Sciences, ATI-16000 (ARYx Therapeutics), KS-01-019 (Kos Pharmaceuticals), Pharmaprojects No. 2197 (Sanofi-Aventi), RP 61969 (Sanofi-Aventis), cerivastatin Na (CAS No.
  • an immunosuppressant selected from the group consisting of teriflunomide (Sanofi Aventis), chlorsulfaquinoxalone (NSC-339004), chlorsulfaquinoxalone sulfate, CS-712 (Sankyo), ismomultin alfa (CAS No.
  • an NF kappa B inhibitor selected from the group consisting of emodin (CAS No. 518-82-1), AVE-0545 or AVE- 0547 (Sanofi-Aventis), bortezomib (CAS No. 179324-69-7) (Millennium Pharmaceuticals), dexanabinol (CAS No. 112924-45-5) (Pharmos), dexlipotam (Viatris), Pharmaprojects No.
  • INDRA OXiGENE
  • IPL- 576092 CAS No. 137571-30-3
  • ⁇ nf ⁇ azyme NFKB decoy
  • NFKB decoy oligo NFKB decoy oligo
  • S5 F005 from Fulcrum Pharmaceuticals
  • P61 Physicaltopharm
  • R-flurbiprofen CAS No.
  • a palmitoyl-protein thioesterase inhibitor 80) a PDGF receptor kinase inhibitor selected from the group consisting of AAL-993, AMN-107, or ABP-309 (Novartis), AMG- 706 (Amgen), BAY-57-9352 (Bayer), CDP-860 (UCB), E-7080 (Eisai), imatinib (CAS No.
  • a peroxisome proliferators- activated receptor agonist selected from the group consisting of (-)- halofenate (Metabolex), AMG-131 (Amgen), antidiabetics from Japan Tobacco, AZD-4619, AZD-8450, AZD-8677 (AstraZeneca), DRF-10945, balaglitazone (Dr Reddy's), CS-00088, CS-00098 (Chipscreen Biosciences), E-3030 (Eisai), etalocib (CAS No.
  • 122320- 73-4 or 155141-29-0) (GlaxoSmithKline), rosiglitazone maleate/glimepir (CAS No. 155141-29-0 and 93479-97-1), AVANDARYL, rosiglitazone maleate/metformin extend (CAS No. 155141-29-0 and 657-24-9), AVANDAMET, rosiglitazone maleate+metformin, AVANDAMET (GlaxoSmithKline), tesaglitazar (AstraZeneca), LBM642, WY-14,643 (CAS No. 50892-23-4), GW7647, fenofibric acid (CAS No.
  • MCC-555 (CAS No. 161600-01-7), GW9662, GW1929, GW501516, L-165,041 (CAS No. 79558-09-1), and an analogue or derivative thereof, 82) a phosphatase inhibitor, 83) a phosphodiesterase (PDE) inhibitor selected from the group consisting of avanafil (Tanabe Seiyaku), dasantafil (CAS No. 569351-91-3) (Schering-Plough), A-906119 (CAS No.
  • PDE phosphodiesterase
  • a phosphodiesterase 111 inhibitor enoximone, a phosphodiesterase IV inhibitor, fosfosal, Atopik (Barrier Therapeutics), triflusal, a phosphodiesterase V inhibitor, and an analogue or derivative thereof
  • a PKC inhibitor 85) a platelet activating factor antagonist, 86) a platelet-derived growth factor receptor kinase inhibitor, 87) a prolyl hydroxylase inhibitor, 88) a polymorphonuclear neutrophil inhibitor, 89) a protein kinase B inhibitor, 90) a protein kinase C stimulant, 91) a purine nucleoside analogue, 92) a purinoreceptor P2X antagonist, 93) a Raf kinase inhibitor, 94) a reversible inhibitor of ErbB1 and ErbB2, 95) a ribonucleoside triphosphate reductase inhibitor,
  • TNF ⁇ antagonist or TACE inhibitor selected from the group consisting of adalimumab (CAS No.
  • TNF antagonists form ProStrakan, and Synergen, TNF inhibitors (Amgen), TNF-alpha antagonists from Dynavax Technologies and Jerina AG (Germany), TNF-alpha inhibitors from IBFB Pharma and Xencor (Xencor), torbafylline (CAS No.
  • brompheniramine (CAS No. 980- 71-2), fexofenadine hydrochloride, promethazine hydrochloride, loratadine, ketotifen fumarate salt, and acrivastine), methylxanthines (e.g., theophylline, theobromine, and caffeine), cimetidine (available under the tradename TAGAMET from SmithKline Beecham Phamaceutical Co., Wilmington, DE), ranitidine (available under the tradename ZANTAC from Warner Lambert Company, Morris Plains, NJ), famotidine (available under the tradename PEPCID from Merck & Co., Whitehouse Station, NJ), nizatidine (available under the tradename AXID from Reliant Pharmaceuticals, Inc., Liberty Corner, NJ), nizatidine, and roxatidine acetate (CAS No.
  • H3 receptor antagonists ⁇ e.g., thioperamide and thioperamide maleate salt
  • antihistamines e.g., tricyclic dibenozoxepins, ethanolamines, ethylenediamines, piperizines, piperidines, and pthalazinones
  • 131 an alpha adrenergic receptor antagonist
  • 132) an anti-psychotic compound
  • 133) a CaM kinase Il inhibitor
  • 134) a G protein agonist 135) an antibiotic selected from the group consisting of apigenin (Cas No. 520-36-5), ampicillin sodium salt (CAS No.
  • a DNA topoisomerase inhibitor selected from the group consisting of ⁇ -lapachone (CAS No. 4707- 32-8), (-)-arctigenin (CAS No. 7770-78-7), aurintricarboxylic acid, and an analogue or derivative thereof, 138) a thromboxane A2 receptor inhibitor selected from the group consisting of BM-531 (CAS No. 284464-46-6), ozagrel hydrochloride (CAS No.
  • a D2 dopamine receptor antagonist 140) a Peptidyl-Prolyl Cis/Trans lsomerase Inhibitor, 141) a dopamine antagonist, an anesthetic compound, 142) a clotting factor, 143) a lysyl hydrolase inhibitor, 144) a muscarinic receptor inhibitor, 145) a superoxide anion generator, 146) a steroid, 147) an antiproliferative agent selected from the group consisting of silibinin (CAS No. 22888-70-6), silymarin (CAS No.
  • temozolomide temozolomide
  • procarbazine HCI and an analogue or derivative thereof
  • 157) a DNA methylation inhibitor 158) a NSAID agent, 159) a peptidylglycine alpha-hydroxylating monooxygenase inhibitor, 160) an MEK1/MEK 2 inhibitor, 161) a NO synthase inhibitor, 162) a retinoic acid receptor antagonist selected from isotretinoin (CAS No.
  • an ACE inhibitor 164) a glycosylation inhibitor, 165) an intracellular calcium influx inhibitor, 166) an anti-emetic agent, 167) an acetylcholinesterase inhibitor, 168) an ALK-5 receptor antagonist, 169) a RAR/RXT antagonist, 170) an elF-2a inhibitor, 171) an S- adenosyl-L-homocysteine hydrolase inhibitor, 172) an estrogen agonist, 173) a serotonin receptor inhibitor, 174) an antithrombotic agent, 175) a tryptase inhibitor, 176) a pesticide, 177) a bone mineralization promoter, 178) a bisphosphonate compound selected from risedronate and an analogue or derivative thereof, 179) an anti-inflammatory compound, 180) a DNA methylation promoter, 181) an anti-spasmodic agent, .182) a protein synthesis inhibitor, 183) an ⁇ -glucosidas
  • Figure 1 schematically depicts the transcriptional regulation of matrix metalloproteinases.
  • Figure 2 is a blot which demonstrates that IL-1 stimulates AP-1 transcriptional activity. _ . . . . . . . . . -
  • Figure 3 is a graph which shows that IL-1 induced binding activity decreased in lysates from chondrocytes which were pretreated with paclitaxel.
  • Figure 4 is a blot which shows that IL-1 induction increases collagenase and stromelysin in RNA levels in chondrocytes, and that this induction can be inhibited by pretreatment with paclitaxel.
  • Figures 5A-H are blots that show the effect of various anti- microtubule agents in inhibiting collagenase expression.
  • Figure 6 is a graph showing the results of a screening assay for assessing the effect of paclitaxel on smooth muscle cell migration.
  • Figure 7 is a graph showing the average rank of joint scores of Hartley guinea pig knees with ACL damage treated with paclitaxel. A reduction in score indicates an improvement in cartilage score. The dose response trend is statistically significant (p ⁇ 0.02).
  • Figures 8A-C are examples of cross sections of Hartley guinea pig knees of control and paclitaxel treated animals.
  • Figure 8A Control speciment showing erosion of cartilage to the bone.
  • Figure 8B Paclitaxel dose 1 (low dose) showing fraying of cartilage.
  • Figure 8C Paclitaxel dose 2 (medium dose) showing minor defects to cartilage.
  • Figures 9A-F are safranin-O stained histological slides of representative synovial tissues from naive (healthy) knees ( Figures 9A and 9D) and knees with arthritis induced by administration of albumin in Freund's complete adjuvant (Figures 9B and 9C) or carrageenan ( Figures 9E and 9F). Arthritic knees received either control ( Figures 9B and 9E) or 20% paclitaxel-loaded microspheres ( Figures 9C and 9F). The data illustrate decreased proteoglycan red staining in arthritic knees treated with control microspheres and the proteoglycan protection properties of the paclitaxel- loaded formulation.
  • Fibrosis or “scarring,” or “fibrotic response” refers to the formation of fibrous (scar) tissue in response to injury or medical intervention.
  • Therapeutic agents which inhibit fibrosis or scarring are referred to herein as “fibrosis-inhibiting agents”, “fibrosis-inhibitors”, “anti- scarring agents”, and the like, where these agents inhibit fibrosis through one or more mechanisms including: inhibiting inflammation or the acute inflammatory response, inhibiting migration or proliferation of connective tissue cells (such as fibroblasts, smooth muscle cells, vascular smooth muscle cells), inhibiting angiogenesis, reducing extracellular matrix (ECM) production or promoting ECM breakdown, and/or inhibiting tissue remodeling.
  • connective tissue cells such as fibroblasts, smooth muscle cells, vascular smooth muscle cells
  • ECM extracellular matrix
  • a body passageway e.g., a blood vessel, the gastrointestinal tract, the respiratory tract, the urinary tract, the female or male reproductive tract, the eustacian tube etc.
  • a body passageway e.g., a blood vessel, the gastrointestinal tract, the respiratory tract, the urinary tract, the female or male reproductive tract, the eustacian tube etc.
  • Implanted refers to having completely or partially placed a device or implant within a host. A device is partially implanted when some of the device reaches, or extends to the outside of, a host.
  • Inhibit fibrosis "reduce fibrosis”, “Inhibits scarring” and the like are used synonymously to refer to the action of agents or compositions which result in a statistically significant decrease in the formation of fibrous tissue that can be expected to occur in the absence of the agent or composition.
  • Anti-infective agent refers to an agent or composition which prevents microrganisms from growing and/or slows the growth rate of microorganisms and/or is directly toxic to microorganisms at or near the site of the agent. These processes would be expected to occur at a statistically significant level at or near the site of the agent or composition relative to the effect in the absence of the agent or composition.
  • Inhibit infection refers to the ability of an agent or composition to prevent microorganisms from accumulating and/or proliferating near or at the site of the agent. These processes would be expected to occur at a statistically significant level at or near the site of the agent or composition relative to the effect in the absence of the agent or composition.
  • “Inhibitor” refers to an agent which prevents a biological process from occurring or slows the rate or degree of occurrence of a biological process.
  • the process may be a general one such as scarring or refer to a specific biological action such as, for example, a molecular process resulting in release of a cytokine.
  • Antagonist refers to an agent which prevents a biological process from occurring or slows the rate or degree of occurrence of a biological process. While the process may be a general one, typically this refers to a drug mechanism where the drug competes with a molecule for an active molecular site or prevents a molecule from interacting with the molecular site. In these situations, the effect is that the molecular process is inhibited.
  • Antist refers to an agent which stimulates a biological process or rate or degree of occurrence of a biological process.
  • the process may be a general one such as scarring or refer to a specific biological action such as, for example, a molecular process resulting in release of a cytokine.
  • Anti-microtubule agents should be understood to include any protein, peptide, chemical, or other molecule which impairs the function of microtubules, for example, through the prevention or stabilization of polymerization.
  • Compounds that stabilize polymerization of microtubules are referred to herein as "microtubule stabilizing agents.”
  • a wide variety of methods may be utilized to determine the anti-microtubule activity of a particular compound, including for example, assays described by Smith et al. ⁇ Cancer Lett 79(2):213-219, 1994) and Mooberry et al., (Cancer Lett. 96(2):261-266, 1995).
  • Medical device “implant”, “”device”, medical device”, “medical implant”, “implant/device” and the like are used synonymously to refer to any object that is designed to be placed partially or wholly within a patient's body for one or more therapeutic or prophylactic purposes such as for restoring physiological function, alleviating symptoms associated with disease, delivering therapeutic agents, and/or repairing, replacing, or augmenting etc. damaged or diseased organs and tissues.
  • some medical devices and implants include materials derived from animals (e.g., "xenografts” such as whole animal organs; animal tissues such as heart valves; naturally occurring or chemically-modified molecules such as collagen, hyaluronic acid, proteins, carbohydrates and others), human donors (e.g., "allografts” such as whole organs; tissues such as bone grafts, skin grafts and others), or from the patients themselves (e.g., "autografts” such as saphenous vein grafts, skin grafts, tendon/ligament/muscle transplants).
  • animals e.g., "xenografts” such as whole animal organs; animal tissues such as heart valves; naturally occurring or chemically-modified molecules such as collagen, hyaluronic acid, proteins, carbohydrates and others
  • human donors e.g., "allografts” such as whole organs; tissues such as bone grafts, skin grafts and others
  • autografts such as sap
  • Release of an agent refers to a statistically significant presence of the agent, or a subcomponent thereof, which has disassociated from the implant/device and/or remains active on the surface of (or within) the device/implant.
  • Biodegradable refers to materials for which the degradation process is at least partially mediated by, and/or performed in, a biological system.
  • Degradation refers to a chain scission process by which a polymer chain is cleaved into oligomers and monomers. Chain scission may occur through various mechanisms, including, for example, by chemical reaction (e.g., hydrolysis) or by a thermal or photolytic process.
  • Polymer degradation may be characterized, for example, using gel permeation chromatography (GPC), which monitors the polymer molecular mass changes during erosion and drug release.
  • GPC gel permeation chromatography
  • Biodegradable also refers to materials may be degraded by an erosion process mediated by, and/or performed in, a biological system.
  • Erosion refers to a process in which material is lost from the bulk.
  • the material may be a monomer, an oligomer, a part of a polymer backbone, or a part of the polymer bulk.
  • Erosion includes (i) surface erosion, in which erosion affects only the surface and not the inner parts of a matrix; and (ii) bulk erosion, in which the entire system is rapidly hydrated and polymer chains are cleaved throughout the matrix.
  • erosion generally occurs by one of three basic mechanisms (see, e.g., Heller, J., CRC Critical Review in Therapeutic Drug Carrier Systems (1984), 1(1), 39-90); Siepmann, J.
  • analogue refers to a chemical compound that is structurally similar to a parent compound, but differs slightly in composition (e.g., one atom or functional group is different, added, or removed).
  • the analogue may or may not have different chemical or physical properties than the original compound and may or may not have improved biological and/or chemical activity.
  • the analogue may be more hydrophilic or it may have altered reactivity as compared to the parent compound.
  • the analogue may mimic the chemical and/or- biologically activity of the parent compound (i.e., it may have similar or identical activity), or, in some cases, may have increased or decreased activity.
  • the analogue may be a naturally or non-naturally occurring (e.g., recombinant) variant of the original compound.
  • An example of an analogue is a mutein (i.e., a protein analogue in which at least one amino acid is deleted, added, or substituted with another amino acid).
  • Other types of analogues include isomers (enantiomers, diasteromers, and the like) and other types of chiral variants of a compound, as well as structural isomers.
  • the analogue may be a branched or cyclic variant of a linear compound.
  • a linear compound may have an analogue that is branched or otherwise substituted to impart certain desirable properties (e.g., improve hydrophilicity or bioavailability).
  • derivative refers to a chemically or biologically modified version of a chemical compound that is structurally similar to a parent compound and (actually or theoretically) derivable from that parent compound.
  • a “derivative” differs from an “analogue” in that a parent compound may be the starting material to generate a "derivative,” whereas the parent compound may not necessarily be used as the starting material to generate an “analogue.”
  • a derivative may or may not have different chemical or physical properties of the parent compound. For example, the derivative may be more hydrophilic or it may have altered reactivity as compared to the parent compound.
  • Derivatization (Ae., modification) may involve substitution of one or more moieties within the molecule (e.g., a change in functional group).
  • a hydrogen may be substituted with a halogen, such as fluorine or chlorine, or a hydroxyl group (-OH) may be replaced with a carboxylic acid moiety (- COOH).
  • derivative also includes conjugates, and prodrugs of a parent compound (i.e., chemically modified derivatives which can be converted into the original compound under physiological conditions).
  • the prodrug may be an inactive form of an active agent. Under physiological conditions, the prodrug may be converted into the active form of the compound.
  • Prodrugs may be formed, for example, by replacing one or two hydrogen atoms on nitrogen atoms by an acyl group (acyl prodrugs) or a carbamate group (carbamate prodrugs).
  • prodrugs More detailed information relating to prodrugs is found, for example, in Fleisher et al., Advanced Drug Delivery Reviews 19 (1996) 115; Design of Prodrugs, H. Bundgaard (ed.), Elsevier, 1985; or H. Bundgaard, Drugs of the Future 16 (1991) 443.
  • derivative is also used to describe all solvates, for example hydrates or adducts (e.g., adducts with alcohols), active metabolites, and salts of the parent compound.
  • the type of salt that may be prepared depends on the nature of the moieties within the compound.
  • acidic groups for example carboxylic acid groups
  • alkali metal salts or alkaline earth metal salts e.g., sodium salts, potassium salts, magnesium salts and calcium salts
  • physiologically tolerable quaternary ammonium ions and acid addition salts with ammonia and physiologically tolerable organic amines such as, for example, triethylamine, ethanolamine or tris-(2-hydroxyethyl)amine.
  • Basic groups can form acid addition salts, for example with inorganic acids such as hydrochloric acid, sulfuric acid or phosphoric acid, or with organic carboxylic acids and sulfonic acids such as acetic acid, citric acid, benzoic acid, maleic acid, fumaric acid, tartaric acid, methanesulfonic acid or p-toluenesulfonic acid.
  • Compounds which simultaneously contain a basic group and an acidic group for example a carboxyl group in addition to basic nitrogen atoms, can be present as zwitterions. Salts can be obtained by customary methods known to those skilled in the art, for example by combining a compound with an inorganic or organic acid or base in a solvent or diluent, or from other salts by cation exchange or anion exchange.
  • Hyaluronic acid or “HA” as used herein refers to all forms of hyaluronic acid that are described or referenced herein, including those that have been processed or chemically or physically modified, as well as hyaluronic acid that has been crosslinked (for example, covalently, ionically, thermally or physically).
  • HA is a glycosaminoglycan composed of a linear chain of about 2500 repeating disaccharide units. Each disaccharide unit is composed of an N-acetylglucosamine residue linked to a glucuronic acid.
  • Hyaluronic acid is a natural substance that is found in the extracellular matrix of many tissues including synovial joint fluid, the vitreous humor of the eye, cartilage, blood vessels, skin and the umbilical cord.
  • Commercial forms of hyaluronic acid having a molecular weight of approximately 1.2 to 1.5 million Daltons (Da) are extracted from rooster combs and other animal sources.
  • Other sources of HA include HA that is isolated from cell culture / fermentation processes. Lower molecular weight HA formulations are also available from a variety of commercial sources.
  • the molecule can be of variable lengths (i.e., different numbers of repeating disaccharide units and different chain branching patterns) and can be modified at several sites (through the addition or subtraction of different functional groups) without deviating from the scope of the present invention.
  • inter-react refers to the formulation of covalent bonds, noncovalent bonds, or both.
  • the term thus includes crosslinking, which involves both intermolecular crosslinks and optionally intramolecular crosslinks as well, arising from the formation of covalent bonds.
  • Covalent bonding between two reactive groups may be direct, in which case an atom in reactive group is directly bound to an atom in the other reactive group, or it may be indirect, through a linking group.
  • Noncovalent bonds include ionic (electrostatic) bonds, hydrogen bonds, or the association of hydrophobic molecular segments, which may be the same or different.
  • a crosslinked matrix may, in addition to covalent bonds, also include such intermolecular and/or intramolecular noncovalent bonds.
  • hydrophilic and hydrophobic are generally defined in terms of an HLB value, i.e., a hydrophilic lipophilic balance.
  • a high HLB value indicates a hydrophilic compound, while a low HLB value characterizes a hydrophobic compound.
  • HLB values are well known in the art, and generally range from 1 to 18.
  • Preferred multifunctional compound cores are hydrophilic, although as long as the multifunctional compound as a whole contains at least one hydrophilic component, crosslinkable hydrophobic components may also be present.
  • synthetic is used to refer to polymers, compounds and other such materials that are “chemically synthesized.”
  • a synthetic material in the present compositions may have a molecular structure that is identical to a naturally occurring material, but the material perse, as incorporated in the compositions of the invention, has been chemically synthesized in the laboratory or industrially.
  • synthetic materials also include semi-synthetic materials, i.e., naturally occurring materials, obtained from a natural source, that have been chemically modified in some way.
  • the synthetic materials herein are purely synthetic, i.e., they are neither semi-synthetic nor have a structure that is identical to that of a naturally occurring material.
  • tissue growth-promoting amount refers to the amount needed in order to stimulate tissue growth to a detectable degree.
  • Tissue in this context, includes connective tissue, bone, cartilage, epidermis and dermis, blood, and other tissues. The actual amount that is determined to be an effective amount will vary depending on factors such as the size, condition, sex and age of the patient and can be more readily determined by the caregiver.
  • compositions of the invention can be injected or otherwise applied to a specific site within a patient's body, e.g., a site in need of augmentation, and allowed to crosslink at the site of injection.
  • Suitable sites will generally be intradermal or subcutaneous regions for augmenting dermal support, at a bone fracture site for bone repair, within sphincter tissue for sphincter augmentation (e.g., for restoration of continence), within a wound or suture, to promote tissue regrowth; and within or adjacent to vessel anastomoses, to promote vessel regrowth.
  • aqueous medium includes solutions, suspensions, dispersions, colloids, and the like containing water.
  • aqueous environment means an environment containing an aqueous medium.
  • dry environment means an environment that does not contain an aqueous medium.
  • alkyl refers to a branched or unbranched saturated hydrocarbon group typically although not necessarily containing 1 to about 24 carbon atoms, such as methyl, ethyl, /7-propyl, isopropyl, ⁇ -butyl, isobutyl, f-butyl, octyl, decyl, and the like, as well as cycloalkyl groups such as cyclopentyl, cyclohexyl and the like. Generally, although again not necessarily, alkyl groups herein contain 1 to about 12 carbon atoms.
  • the term "lower alkyl” intends an alkyl group of one to six carbon atoms, preferably one to four carbon atoms.
  • Substituted alkyl refers to alkyl substituted with one or more substituent groups.
  • Alkylene refers to divalent alkyl, lower alkyl, and substituted alkyl groups, respectively.
  • aryl refers to an aromatic substituent containing a single aromatic ring (monocyclic) or multiple aromatic rings that are fused together, linked covalently, or linked to a common group such as a methylene or ethylene moiety.
  • the common linking group may also be a carbonyl as in benzophenone, an oxygen atom as in diphenylether, or a nitrogen atom as in diphenylamine.
  • Preferred aryl groups contain one aromatic ring or two fused or linked aromatic rings, e.g., phenyl, naphthyl, biphenyl, diphenylether, diphenylamine, benzophenone, and the like.
  • Substituted aryl refers to an aryl moiety substituted with one or more substituent groups
  • heteroatom-containing aryl and “heteroaryl” refer to aryl in which at least one carbon atom is replaced with a heteroatom.
  • arylene and “substituted arylene” refer to divalent aryl and substituted aryl groups as just defined.
  • heteroatom-containing refers to a molecule or molecular fragment in which one or more carbon atoms is replaced with an atom other than carbon, e.g., nitrogen, oxygen, sulfur, phosphorus or silicon.
  • Hydrocarbyl refers to univalent hydrocarbyl radicals containing 1 to about 30 carbon atoms, preferably 1 to about 24 carbon atoms, most preferably 1 to about 12 carbon atoms, including branched or unbranched, saturated or unsaturated species, such as alkyl groups, alkenyl groups, aryl groups, and the like.
  • lower hydrocarbyl intends a hydrocarbyl group of one to six carbon atoms, preferably one to four carbon atoms.
  • hydrocarbylene intends a divalent hydrocarbyl moiety containing 1 to about 30 carbon atoms, preferably 1 to about 24 carbon atoms, most preferably 1 to about 12 carbon atoms, including branched or unbranched, saturated or unsaturated species, or the like.
  • lower hydrocarbylene intends a hydrocarbylene group of one to six carbon atoms, preferably one to four carbon atoms.
  • Substituted hydrocarbyl refers to hydrocarbyl substituted with one or more substituent groups
  • heteroatom-containing hydrocarbyl and heterohydrocarbyl refer to hydrocarbyl in which at least one carbon atom is replaced with a heteroatom.
  • substituted hydrocarbylene refers to hydrocarbylene substituted with one or more substituent groups
  • heteroatom-containing hydrocarbylene and heterohydrocarbylene refer to hydrocarbylene in which at least one carbon atom is replaced with a heteroatom. If not otherwise indicated, “hydrocarbyl” indicates both unsubstituted and substituted hydrocarbyls, “heteroatom-containing hydrocarbyl” indicates both unsubstituted and substituted heteroatom- containing hydrocarbyls and so forth.
  • substituted as in “substituted hydrocarbyl,” “substituted alkyl,” and the like, as alluded to in some of the aforementioned definitions, is meant that in the hydrocarbyl, alkyl, or other moiety, at least one hydrogen atom bound to a carbon atom is replaced with one or more substituents that are functional groups such as alkoxy, hydroxy, halo, nitro, and the like. Unless otherwise indicated, it is to be understood that specified molecular segments can be substituted with one or more substituents that do not compromise a compound's utility.
  • succinimidyl is intended to include unsubstituted succinimidyl as well as sulfosuccinimidyl and other succinimidyl groups substituted on a ring carbon atom, e.g., with alkoxy substituents, polyether substituents, or the like.
  • any concentration ranges, percentage range, or ratio range recited herein are to be understood to include concentrations, percentages or ratios of any integer within that range and fractions thereof, such as one tenth and one hundredth of an integer, unless otherwise indicated.
  • any number range recited herein relating to any physical feature, such as polymer subunits, size or thickness are to be understood to include any integer within the recited range, unless otherwise indicated.
  • the term "about” refers to ⁇ 15% of any indicated structure, value, or range.
  • a and “an” refer to one or more of the indicated items.
  • a polymer refers to both one polymer or a mixture comprising two or more polymers;
  • a multifunctional compound refers not only to a single multifunctional compound but also to a combination of two or more of the same or different multifunctional compounds;
  • a reactive group refers to a combination of reactive groups as well as to a single reactive group, and the like.
  • the present invention provides polymeric compositions which greatly increase the ability to inhibit the formation of reactive scar tissue on, or around, the surface of a device or implant or at a treatment site. Numerous polymeric compositions and therapeutic agents are described herein.
  • compositions e.g., polymers
  • therapeutic agents e.g., drugs, and/or agents that include one or more therapeutic agents, described below.
  • methods for making and methods for utilizing such compositions are also described in more detail below.
  • Therapeutic agents useful in the present invention includes various anti-fibrosis agents, anti-infective agents, and polymers.
  • the present invention discloses pharmaceutical agents which inhibit one or more aspects of the production of excessive fibrous (scar) tissue.
  • Such agents may be readily determined based upon the in vitro and in vivo (animal) models such as those provided in Examples 16-20, 21-28, 29, 38, 39, 42, 43, and 81.
  • Agents which inhibit fibrosis may be identified through in vivo models including inhibition of intimal hyperplasia development in the rat balloon carotid artery model (Examples 21 and 29).
  • the assays set forth in Examples 20 and 28 may be used to determine whether an agent is able to inhibit cell proliferation in fibroblasts and/or smooth muscle cells.
  • the agent has an IC 50 for inhibition of cell proliferation within a range of about 10 "6 to about 10 "10 M. In certain embodiments, the agent may have an IC 50 for inhibition of cell proliferation of less than about 10,000 nM; or less than about 1000 nM; or less than about 100 nM.
  • the assay set forth in Example 24 may be used to determine whether an agent may inhibit migration of fibroblasts and/or smooth muscle cells. In one aspect of the invention, the agent has an IC 50 for inhibition of cell migration within a range of about 10 "6 to about 10 "9 M.
  • the agent may have an IC 50 for inhibition of fibroblast or smooth muscle cell migration of less than about 10,000 nM; or less than about 1000 nM; or less than about 100 nM.
  • Assays set forth herein may be used to determine whether an agent is able to inhibit inflammatory processes, including nitric oxide production in macrophages (Example 16), and/or TNF-alpha production by macrophages (Example 17), and/or IL-1 beta production by macrophages (Example 25), and/or IL-8 production by macrophages (Example 26), and/or inhibition of MCP-1 by macrophages (Example 27).
  • the agent has an IC 50 for inhibition of any one of these inflammatory processes within a range of about 10 "6 to about 10 "10 M. In certain embodiments, the agent may have an IC 50 for any one of these inflammatory processes of less than about 10,000 nM; or less than about 1000 nM; or less than about 100 nM.
  • the assay set forth in Example 22 may be used to determine whether an agent is able to inhibit MMP production. In one aspect of the invention, the agent has an IC 50 for inhibition of MMP production within a range of about 10 "4 to about 10 "8 M.
  • the agent may have an IC 50 for inhibition of MMP production of less than about 10,000 nM; or less than about 1000 nM; or less than about 100 nM.
  • the assay set forth in Example 23 (also known as the CAM assay) may be used to determine whether an agent is able to inhibit angiogenesis.
  • the agent has an IC 50 for inhibition of angiogenesis within a range of about 10 "6 to about 10 "10 M.
  • the agent may have an IC 50 for inhibition of angiogenesis of less than about 10,000 nM; or less than about 1000 nM; or less than about 100 nM.
  • the assay set forth in Example 81 may be used to determine whether an agent is able to inhibit MMP-1.
  • the agent has an IC 50 for inhibition of MMP-1 within a range of about 10 "6 to about 10 "10 M. In certain embodiments, the agent may have an IC50 for inhibition of MMP-1 of less than about 10,000 nM; or less than about 1000 nM; or less than about 100 nM.
  • Agents which reduce the formation of surgical adhesions may be identified through in vivo models including the rabbit surgical adhesions model (Examples 19, 39, 42, and 43) and the rat caecal sidewall model (Example 18). These pharmacologically active agents (described below) can then be delivered at appropriate dosages into to the tissue either alone, or via carriers (described herein), to treat the clinical problems described herein. . . _ _ . .
  • Numerous therapeutic compounds capable of inhibiting fibrosis may be identified as useful in the invention including:
  • the fibrosis-inhibiting compound is an adensosine A2A receptor antagonist (e.g., Sch-63390 (Schering-Plough) or an A2A receptor antagonists from Almirall-Prodesfarma, SCH-58261 (CAS No. 160098-96-4), or an analogue or derivative thereof).
  • an adensosine A2A receptor antagonist e.g., Sch-63390 (Schering-Plough) or an A2A receptor antagonists from Almirall-Prodesfarma, SCH-58261 (CAS No. 160098-96-4), or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is an AKT inhibitor (e.g., PKB inhibitors from DeveloGen, AKT inhibitors from Array BioPharma, Celgene, Merck & Co, Amphora, NeoGenesis Pharmaceuticals, A-443654 (Abbott Laboratories), erucylphosphocholine (AEterna Zentaris), KRX-401 (Keryx), protein kinase B inhibitors from Astex Technology, PX-316 (ProlX), or an analogue or derivative thereof).
  • AKT inhibitor e.g., PKB inhibitors from DeveloGen, AKT inhibitors from Array BioPharma, Celgene, Merck & Co, Amphora, NeoGenesis Pharmaceuticals, A-443654 (Abbott Laboratories), erucylphosphocholine (AEterna Zentaris), KRX-401 (Keryx), protein kinase B inhibitors from Astex Technology, PX-316 (ProlX), or
  • the fibrosis-inhibiting compound is an alpha 2 integrin antagonist (e.g., Pharmaprojects No. 5754 (Merck KGaA), or an analogue or derivative thereof).
  • an alpha 2 integrin antagonist e.g., Pharmaprojects No. 5754 (Merck KGaA), or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is an alpha 4 integrin antagonist (e.g., T-0047 (Tanabe Seiyaku), VLA-4 antagonists from Sanofi-Aventis, Merck & Co., Biogen personal, Uriach, and Molecumetics, alpha 4 integrin antagonists from Genentech), BIO-2421 (Biogen personal), cell adhesion inhibitors from Kaken Pharmaceuticals, CT-737 (Wyeth), CT-767 (Elan), CY-9652 (Epimmune), CY-9701 (Epimmune), fibronectin antagonists from Uriach, integrin alpha4 ⁇ 7 antagonists frin Wilex, Pharmaprojects No.
  • alpha 4 integrin antagonist e.g., T-0047 (Tanabe Seiyaku), VLA-4 antagonists from Sanofi-Aventis, Merck & Co., Biogen personal, and Molecumetics, alpha 4 integrin antagonists from Genentech), BIO
  • the fibrosis-inhibiting compound is an alpha 7 nicotinic receptor agonist (e.g., AZD-0328 (AstraZeneca), galantamine (CAS No. 357-70-0) (Synaptc), MEM-3454 or nicotinic alpha-7 agonist (Memory Pharmaceuticals and Critical Therapeutics), Pharmaprojects No. 4779 (AstraZeneca), PNU-282987 (Pfizer), SSR- 180711 (Sanofi-Aventis), TC-1698 or TC-5280 (Targacept), or an analogue or derivative thereof).
  • AZD-0328 AstraZeneca
  • galantamine CAS No. 357-70-0
  • Synaptc nicotinic alpha-7 agonist
  • Pharmaprojects No. 4779 AstraZeneca
  • PNU-282987 Pfizer
  • SSR- 180711 Sanofi-Aventis
  • the fibrosis-inhibjting compound is an angiogenesis inhibitor (e.g., AG-") 2, 958 (Pfizer), ATN-161 (Attention LLC), neovastat, an angiogenesis inhibitor from Jerina AG (Germany), NM-3 (Mercian), VGA-1155 (Taisho), FCE-26644 (Pfizer), FCE-26950 (Pfizer), FPMA (Meiji Daries), FR-111142 (Fujisawa), GGTI-298, GM-1306 (Ligand), GPA-1734 (Novartis), NNC-47-0011 (Novo Nordisk), herbamycin (Nippon Kayaku), lenalidomide (Celegene), IP-10 (N(H) 1 ABT-828 (Abbott), KIN-841 (Tokushima University, Japan), SF-1126 (Semafore Pharmaceuticals), laminin technology (NIH), CHIR-258 (Chiron), NVP-AEW
  • the fibrosis-inhibiting compound is an apoptosis antagonist (e.g., didemnin B, RGB-286199 (GPC Biotech), 5F- DF-203 (Cancer Research Technology), aplidine, bongkrekic acid, triammonium salt, [6]-gingerol (CAS No. 23513-14-6), or an analogue or derivative thereof).
  • apoptosis antagonist e.g., didemnin B, RGB-286199 (GPC Biotech), 5F- DF-203 (Cancer Research Technology), aplidine, bongkrekic acid, triammonium salt, [6]-gingerol (CAS No. 23513-14-6), or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is an apoptosis activator (e.g., aplidine (CAS No. 137219-37-5) (PharmaMar), canfosfamide hydrochloride (CAS No. 58382-37-74 and 39943-59-6) (Telik), idronoxil (CAS No.
  • apoptosis activator e.g., aplidine (CAS No. 137219-37-5) (PharmaMar), canfosfamide hydrochloride (CAS No. 58382-37-74 and 39943-59-6) (Telik), idronoxil (CAS No.
  • the fibrosis-inhibiting compound is a beta 1 integrin antagonist (e.g., ⁇ -1 integrin antagonists, Berkeley Lab, or an analogue or derivative thereof).
  • a beta 1 integrin antagonist e.g., ⁇ -1 integrin antagonists, Berkeley Lab, or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is a beta tubulin inhibitor (e.g., ZEN-017 (AEterna Zentaris), laulimalide (Kosan Biosciences), or an analogue or derivative thereof).
  • a beta tubulin inhibitor e.g., ZEN-017 (AEterna Zentaris), laulimalide (Kosan Biosciences), or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is an agent that blocks enzyme production in hepatitis C (e.g., merimepodib (Vertex Pharmaceuticals), or an analogue or derivative thereof).
  • hepatitis C e.g., merimepodib (Vertex Pharmaceuticals), or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is a Bruton's tyrosine kinase inhibitor (e.g., a Btk inhibitor from Cellular Genomics, or an analogue or derivative thereof).
  • a Bruton's tyrosine kinase inhibitor e.g., a Btk inhibitor from Cellular Genomics, or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is a calcineurin inhibitor (e.g., tacrolimus (LifeCycle Pharma), or an analogue or derivative thereof).
  • a calcineurin inhibitor e.g., tacrolimus (LifeCycle Pharma)
  • analogue or derivative thereof e.g., tacrolimus (LifeCycle Pharma)
  • the fibrosis-inhibiting compound is a caspase 3 inhibitor (e.g., NM-3 (Mercian), or an analogue or derivative thereof). 15) CC Chemokine Receptor Antagonists
  • the fibrosis-inhibiting compound is a CC chemokine receptor antagonist ⁇ e.g., a chemokine receptor 3 antagonist, a chemokine receptor 6 antagonist, and a chemokine receptor 7 antagonist).
  • CC chemokine receptor antagonists include chemokine antagonists such as the CCR7 antagonists from Neurocrine Biosciences.
  • the fibrosis-inhibiting compound is a CC chemokine receptor antagonist (CCR) 1 , 3, & 5 (e.g., peptide T (Advanced lmmuni T), a CCR3 antagonist from GlaxoSmithKline, a chemokine antagonist (Pharmaprojects No. 6322) from Neurocrine Biosciences or Merck & Co., an HIV therapy agent from ReceptoPharm (Nutra Pharma), Pharmaprojects No. 6129 (Sangamo BioSciences), or an analogue or derivative thereof).
  • CCR CC chemokine receptor antagonist
  • the CCCR antagonist is a CCR2b chemokine receptor antagonist such as RS 102895 (CAS No. 300815-41-2).
  • the fibrosis-inhibiting compound is a cell cycle inhibitor (e.g., SNS-595 (Sunesis), homoharringtonine, or an analogue or derivative thereof).
  • a cell cycle inhibitor e.g., SNS-595 (Sunesis), homoharringtonine, or an analogue or derivative thereof.
  • the cell cycle inhibitor is an anti- microtubule agent (e.g., synthadotin, or an analogue or derivative thereof).
  • cell cycle inhibitor is a microtubule stimulant (e.g., KRX-0403, or an analogue or derivative thereof).
  • the fibrosis-inhibiting compound is a cathepsin B inhibitor (e,g., AM-4299A (Asahi Kasei Pharma), BDI-7800 (Biopharmacopae), a cathepsin B inhibitor from Axys (Celera Genomics), MDL-104903 (CAS No. 180799-56-8) (Sanofi-Aventis), NC-700 (Nippon Chemiphar), Pharmaprojects No. 2332 (Hoffmann-La Roche), Pharmaprojects No. 4884 (Takeda), Pharmaprojects No. 5134 (Nippon Chemiphar), or an analogue or derivative thereof).
  • a cathepsin B inhibitor e,g., AM-4299A (Asahi Kasei Pharma), BDI-7800 (Biopharmacopae), a cathepsin B inhibitor from Axys (Celera Genomics), MDL-104903 (CAS No. 180799-56-8) (San
  • the fibrosis-inhibiting compound is a cathepsin K inhibitor (e.g., 462795 (GlaxoSmithKline), INPL-022-D6 (Amura Therapeutics), or an analogue or derivative thereof).
  • a cathepsin K inhibitor e.g., 462795 (GlaxoSmithKline), INPL-022-D6 (Amura Therapeutics), or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is a cathepsin L Inhibitor (e.g., a cathepsin L inhibitor from Takeda, INPL-022- E10 (Amura Therapeutics), Pharmaprojects No. 5447 (Taiho), or an analogue or derivative thereof).
  • a cathepsin L Inhibitor e.g., a cathepsin L inhibitor from Takeda, INPL-022- E10 (Amura Therapeutics), Pharmaprojects No. 5447 (Taiho), or an analogue or derivative thereof.
  • CD40 Antagonists . .. . .
  • the fibrosis-inhibiting compound is a CD40 antagonists (e.g., 5D12 (Chiron), ABI-793 (Novartis), an anticancer antibody from Chiron, anti-CD40 MAb-2 (Kirin Brewery), anti-CD40 (Eli Lilly), anti-CD40L antibody (UCB), a CD40 inhibitor from Apoxis, CD40 ligand inhibitor from Millennium Pharmaceuticals, a CD40/CAP inhibitor from Snow Brand, CGEN-40 (Compugen), CHIR-12.12 (Chiron), Pharmaprojects No.
  • CD40 antagonists e.g., 5D12 (Chiron), ABI-793 (Novartis), an anticancer antibody from Chiron, anti-CD40 MAb-2 (Kirin Brewery), anti-CD40 (Eli Lilly), anti-CD40L antibody (UCB), a CD40 inhibitor from Apoxis, CD40 ligand inhibitor from Millennium Pharmaceuticals, a CD40/CAP inhibitor from Snow Brand, CGEN-40 (Compugen), CH
  • the fibrosis-inhibiting compound is a chemokine receptor agonist (e.g., a chemokine agonist from NeuroTarget, or an analogue or derivative thereof). 22) Chymase inhibitors
  • the fibrosis-inhibiting compound is a chymase inhibitor ⁇ e.g., BL-3875 (Dainippon), LEX-043 (SuperGen), NK- 3201 (CAS No. 204460-24-2) (Nippon Kayaku), or an analogue or derivative thereof).
  • a chymase inhibitor ⁇ e.g., BL-3875 (Dainippon), LEX-043 (SuperGen), NK- 3201 (CAS No. 204460-24-2) (Nippon Kayaku), or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is a collagenase (interstitial) antagonist (e.g., IBFB-212543 (IBFB Pharma), Pharmaprojects No. 3762 (Sanofi-Aventis), S-0885 (CAS No. 117517-22-3) (Sanofi-Aventis), SC-40827 (CAS No. 101470-42-2) (Pfizer), or an analogue or derivative thereof).
  • IBFB-212543 IBFB Pharma
  • Pharmaprojects No. 3762 Sanofi-Aventis
  • S-0885 CAS No. 117517-22-3
  • SC-40827 CAS No. 101470-42-2
  • the fibrosis-inhibiting compound is a CXCR (2, 4) antagonist (e.g., SB-656933 (GlaxoSmithKline), AMD3100 octahydrochloride (CAS No. 155148-31-5), or an analogue or derivative thereof).
  • CXCR (2, 4) antagonist e.g., SB-656933 (GlaxoSmithKline), AMD3100 octahydrochloride (CAS No. 155148-31-5), or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is a cyclin dependent kinase (CDK) inhibitor.
  • the cyclin dependent kinase inhibitor is a CDK-1 inhibitor.
  • the cyclin dependent kinase inhibitor is a CDK-2 inhibitor.
  • the cyclin dependent kinase inhibitor is a CDK- 4 inhibitor.
  • the cyclin dependent kinase inhibitor is a CDK-6 inhibitor.
  • Representative examples of cyclin dependent kinase inhibitors include CAK1 inhibitors from GPC Biotech and Bristol-Myers Squibb, RGB- 286199 (GPC Biotech), or an analogue or derivative thereof.
  • Additional exemplary cyclin dependent protein kinase inhibitors include an anticancer agent from Astex Technology, a CAK1 inhibitor from GPC Biotech, a CDK inhibitor from Sanofi-Aventis, a CDK1/CDK2 inhibitor from Amgen, a CDK2 inhibitor from SUGEN-2 (Pfizer), a hearing loss therapy agent (Sound Pharmaceuticals), PD-0332991 (Pfizer), RGB-286199 (GPC Biotech), Ro-0505124 (Hoffmann-La Roche), a Ser/Thr kinase inhibitor from Lilly (EIi Lilly), CVT-2584 (CAS No. 199986-75- 9) (CV Therapeutics), CGP 74514A, bohemine, olomoucine (CAS No. 101622-51-9), indole-3-carbinol (CAS No. 700-06-1), and an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is a cyclooxygenase inhibitor (e.g., NS-398 (CAS No. 123653-11-2), ketoprofen, or an analogue or derivative thereof).
  • the cyclooxygenase inhibitor is a COX-1 inhibitor such as triflusal, or an analogue or derivative thereof).
  • the fibrosis-inhibiting compound is a
  • DHFR inhibitor e.g., PDX (Allos Therapeutics), SC12267, sulfamerazine (CAS No. 127-79-7), or an analogue or derivative thereof).
  • the fibrosis-inhibiting compound is a dual integrin inhibitor (e.g., R411 (Roche Pharmaceuticals), or an analogue or derivative thereof).
  • the fibrosis-inhibiting compound is an elastase inhibitor (e.g., orazipone, depelestat (CAS No. 506433-25-6) (Dyax), AE-3763 (Dainippon), or an analogue or derivative thereof).
  • elastase inhibitor e.g., orazipone, depelestat (CAS No. 506433-25-6) (Dyax), AE-3763 (Dainippon), or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is an elongation factor-1 alpha inhibitor (e.g., aplidine, or an analogue or derivative thereof).
  • an elongation factor-1 alpha inhibitor e.g., aplidine, or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is an endothelial growth factor (EGF) antagonist (e.g., neovastat, NM-3 (Mercian), or an analogue or derivative thereof).
  • EGF endothelial growth factor
  • the fibrosis-inhibiting compound is an endothelial growth factor receptor (EGF-R) kinase inhibitor (e.g., sorafenib tosylate (Bayer), AAL-993 (Novartis), ABP-309 (Novartis), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EXEL-2880 (Exelixis), GW-654652 (GlaxoSmithKline), lavendustin A (CAS No.
  • EGF-R endothelial growth factor receptor
  • a KDR inhibitor from LG Life Sciences, CT-6685 or CT-6729 (UCB) 1 KRN-633 or KRN-951 (Kirin Brewery), OSI-930 (OSI Pharmaceuticals), SP-5.2 (Supratek Pharma), SU- 11657 (Pfizer), a Tie-2 antagonist (Hybrigenics), a VEGF-R inhibitor such as SU 1498, a VEGFR-2 kinase inhibitor (Bristol-Myers Squibb), XL-647 (Exelixis), a KDR inhibitor from Abbott Laboratories, or an analogue or derivative thereof).
  • the fibrosis-inhibiting compound is an endothelial growth factor receptor 2 kinase inhibitor (e.g., sorafenib tosylate, or an analogue or derivative thereof).
  • an endothelial growth factor receptor 2 kinase inhibitor e.g., sorafenib tosylate, or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is an endotoxin antagonist (e.g., E5564 (Eisai Pharmaceuticals), or an analogue or derivative thereof).
  • E5564 Esai Pharmaceuticals
  • an analogue or derivative thereof Epothilone and Tubulin Binders
  • the fibrosis-inhibiting compound is an epothilone or tubulin binder (e.g., ixabepilone (BMS), or an analogue or derivative thereof).
  • BMS ixabepilone
  • the fibrosis-inhibiting compound is an estrogen receptor antagonist (e.g., ERB-041 (Wyeth), or an analogue or derivative thereof).
  • the fibrosis-inhibiting compound is a FGF inhibitor (e.g., IDN-5390 (Indena), or an analogue or derivative thereof).
  • FGF inhibitor e.g., IDN-5390 (Indena), or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is an inhibitor of farnexyl transferase (FTI).
  • FTI farnexyl transferase
  • the FTI inhibits the RAS oncogene family.
  • FTI's include SARASAR (from Schering Corporation, Kenilworth, NJ), or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is a famesyltransferase inhibitor (e.g., A-197574 (Abbott), a famesyltransferase inhibitor from Servier, FPTIII (Strathclyde Institute for Drug R), LB-42908 (LG Life Sciences), Pharmaprojects No. 5063 (Genzyme), Pharmaprojects No. 5597 (Ipsen), Yissum Project No. B-1055 (Yissum), or an analogue or derivative thereof).
  • A-197574 Abbott
  • FPTIII Genzyme
  • Pharmaprojects No. 5597 Ipsen
  • Yissum Project No. B-1055 Yissum Project No. B-1055
  • an analogue or derivative thereof FLT-3 Kinase Inhibitors
  • the fibrosis-inhibiting compound is a FLT-3 kinase inhibitor (e.g., Amphora, or an analogue or derivative thereof).
  • the fibrosis-inhibiting compound is a FGF receptor kinase inhibitor (e.g., MED-A300 (Gerolymatos), SSR-128129 (Sanofi-Aventis), TBC-2250 (Encysive Pharmaceuticals), XL-999 (Exelixis), or a FGF receptor kinase inhibitor from Paradigm Therapeutics, or an analogue or derivative thereof).
  • a FGF receptor kinase inhibitor e.g., MED-A300 (Gerolymatos), SSR-128129 (Sanofi-Aventis), TBC-2250 (Encysive Pharmaceuticals), XL-999 (Exelixis), or a FGF receptor kinase inhibitor from Paradigm Therapeutics, or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is a fibrinogen antagonist (e.g., AUV-201 (Auvation), MG-13926 (Sanofi- Aventis), plasminogen activator (CAS No. 105913-11-9) (from Sanofi- Aventis or UCB), plasminogen activator-2 (tPA-2) (Sanofi-Aventis), pro- urokinase (CAS No. 82657-92-9) (Sanofi-Aventis), mevastatin, or an analogue or derivative thereof).
  • a fibrinogen antagonist e.g., AUV-201 (Auvation), MG-13926 (Sanofi- Aventis), plasminogen activator (CAS No. 105913-11-9) (from Sanofi- Aventis or UCB), plasminogen activator-2 (tPA-2) (Sanofi-Aventis), pro- urokinase (CAS No. 82657-92-9) (
  • the fibrosis-inhibiting compound is a heat shock protein 90 antagonist (e.g., SRN-005 (Sirenade), geldanamycin or a derivative thereof, such as NSC-33050 (17-allylaminogeldanamycin; 17-AAG) or 17-dimethylaminoethylamino-17-demethoxy-geldanamycin (17- DMAG), rifabutin (rifamycin XIV, 1 ⁇ 4-didehydro-1-deoxy-1 ,4-dihydro-5'-(2- methylpropyl)-1-oxo-), radicicol, Humicola fuscoatra (CAS No. 12772-57-5), or an analogue or derivative thereof).
  • SRN-005 Serenade
  • geldanamycin or a derivative thereof such as NSC-33050 (17-allylaminogeldanamycin; 17-AAG) or 17-dimethylaminoethylamino-17-demethoxy
  • the fibrosis-inhibiting compound is a histone deacetylase inhibitor (e.g., FK228 (Gloucester), trichostatin A from Streptomyces sp. (CAS No. 58880-19-6), or an analogue or derivative thereof).
  • a histone deacetylase inhibitor e.g., FK228 (Gloucester), trichostatin A from Streptomyces sp. (CAS No. 58880-19-6), or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is an HMGCoA reductase inhibitor (e.g., an atherosclerosis therapeutic from Lipid Sciences, AT ⁇ -16000 (ARYx Therapeutics), KS-01-019 (Kos Pharmaceuticals), Pharmaprojects No. 2197 (Sanofi-Aventi), RP 61969 (Sanofi-Aventis), cerivastatin Na )CAS No. 143201-11-0), or an analogue or derivative thereof).
  • the fibrosis-inhibiting compound is an ICAM inhibitor (e.g., alicaforsen (CAS No. 185229-68-9) (ISIS Pharmaceuticals), an ICAM-5 modulator (such as ICAM-4 from ICOS), or an analogue or derivative thereof). .. . ⁇
  • ICAM inhibitor e.g., alicaforsen (CAS No. 185229-68-9) (ISIS Pharmaceuticals), an ICAM-5 modulator (such as ICAM-4 from ICOS), or an analogue or derivative thereof).
  • the fibrosis-inhibiting compound is an IL-1 , ICE & IRAK antagonist (e.g., CJ-14877 or CP-424174 (Pfizer), NF-61 (Negma-Lerads), or an analogue or derivative thereof).
  • ICE & IRAK antagonist e.g., CJ-14877 or CP-424174 (Pfizer), NF-61 (Negma-Lerads), or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is an IL-2 inhibitor (e.g., AVE 8062 (Sanofi-Aventis), or an analogue or derivative thereof).
  • IL-2 inhibitor e.g., AVE 8062 (Sanofi-Aventis), or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is an immunosuppressant (e.g., teriflunomide (Sanofi Aventis), chlorsulfaquinoxalone (NSC-339004), chlorsulfaquinoxalone sulfate, CS-712 (Sankyo), ismom ⁇ ltin alfa (CAS No. 457913-93-8) (Akzo Nobel), antiallergics from GenPat77, antiinflammatories or AT-005 (Androclus Therapeutics), autoimmune disease therapeutics from EpiVax, BN-007 (Bone), budesonide (CAS No.
  • an immunosuppressant e.g., teriflunomide (Sanofi Aventis), chlorsulfaquinoxalone (NSC-339004), chlorsulfaquinoxalone sulfate, CS-712 (Sankyo), ismom ⁇ ltin alfa (CAS No. 457913
  • the fibrosis-inhibiting compound is IMPDH (inosine monophosphate) (e.g., ribavirin (Hoffmann-La Roche) or an analogue or derivative thereof).
  • IMPDH inosine monophosphate
  • ribavirin Hoffmann-La Roche
  • the fibrosis-inhibiting compound is an integrin antagonist (e.g., 683699 from Glaxo Smith Kline, integrin antagonists from Jerina AG (Germany), or an analogue or derivative thereof).
  • an integrin antagonist e.g., 683699 from Glaxo Smith Kline, integrin antagonists from Jerina AG (Germany), or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is an interleukin antagonist (e.g., dersalazine, or an analogue or derivative thereof). In another embodiment, the fibrosis-inhibiting compound is an interleukin 1 antagonist (e.g., NPI-1302a-3, or an analogue or derivative thereof).
  • the fibrosis-inhibiting compound is an inhibitor of type III receptor tyrosine kinase such as FLT3, PDGRF and c-KIT (e.g., MLN518 (Millenium Pharmaceuticals), or an analogue or derivative thereof).
  • type III receptor tyrosine kinase such as FLT3, PDGRF and c-KIT
  • FLT3, PDGRF and c-KIT e.g., MLN518 (Millenium Pharmaceuticals), or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is an irreversible inhibitor of enzyme methionine aminopeptidase type 2 (e.g., PPI-2458 (Praecis Pharmaceuticals), or analogue or derivative thereof).
  • methionine aminopeptidase type 2 e.g., PPI-2458 (Praecis Pharmaceuticals), or analogue or derivative thereof.
  • the fibrosis-inhibiting compound is an isozyme-selective delta protein kinase C inhibitor (e.g., KAI-9803 (Kai Pharmaceuticals), or an analogue or derivative thereof).
  • KAI-9803 Kai Pharmaceuticals
  • the fibrosis-inhibiting compound is a JAK3 enzyme inhibitor (e.g., CP-690,550 (Pfizer), or an analogue or derivative thereof).
  • a JAK3 enzyme inhibitor e.g., CP-690,550 (Pfizer), or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is a JNK inhibitor (e.g., BF-67192 (BioFocus), XG-101 or XG-102 (Xigen), or an analogue or derivative thereof).
  • JNK inhibitor e.g., BF-67192 (BioFocus), XG-101 or XG-102 (Xigen), or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is a kinase inhibitor (e.g., a kinase inhibitors from EVOTEC, or an analogue or derivative thereof).
  • a kinase inhibitor e.g., a kinase inhibitors from EVOTEC, or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is a kinesin antagonist (e.g., SB-715992 and an antifungal from Optokinetics, or an analogue or derivative thereof).
  • a kinesin antagonist e.g., SB-715992 and an antifungal from Optokinetics, or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is a leukotriene inhibitor or antagonist (e.g., ambicromil (CAS No. 58805-38-2) (Sanofi-Aventis), amelubant (CAS No. 346735-24-8) (Boehringer Ingelheim), DW-1141 (Dong Wha), ebselen (Daiichi Pharmaceutical), ibudilast (Kyorin), leucotriene inhibitors from Sanofi-Aventis, lymphotoxin - beta receptor (LT- ⁇ ) from Biogen pou, Pharmaprojects No. 1535 or 2728 (CAS No.
  • the fibrosis-inhibiting compound is a MAP kinase inhibitor (e.g., SRN-003-556 (Sirenade), AEG-3482 (AEgera), ARRY-142886 (Array BioPharma), CDP-146 (UCB), or analogue or derivative thereof).
  • MAP kinase inhibitor e.g., SRN-003-556 (Sirenade), AEG-3482 (AEgera), ARRY-142886 (Array BioPharma), CDP-146 (UCB), or analogue or derivative thereof.
  • MMPI Matrix Metalloproteinase Inhibitors
  • the fibrosis-inhibiting compound is a matrix metalloproteinase inhibitor.
  • a variety of MMPI's may be used in the practice of the invention.
  • the MMPI is a MMP-1 inhibitor.
  • the MMPI is a MMP-2 inhibitor.
  • the MMPI is a MMP-4, MMP-5, MMP-6, MMP-7, or MMP-8 inhibitor.
  • Representative examples of MMPI's include glucosamine sulfate, neovastat, GM1489 (CAS No. 170905-75-6), XL784 (EXEL-01370784), TNF-a Protease lnhibitor-1 or 2 (TAPM or TAPI-2), galardin, or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is a MCP-CCR2 inhibitor (e.g., MLN 1202 (Millennium Pharmaceuticals) , or an analogue or derivative thereof).
  • MCP-CCR2 inhibitor e.g., MLN 1202 (Millennium Pharmaceuticals) , or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is an mTOR inhibitor ⁇ e.g., temsirolimus (CAS No. 162635-04-3) (Wyeth), or an analogue or derivative thereof).
  • the fibrosis-inhibiting compound is an mTOR kinase inhibitor (e.g., ABT-578 (Abbott), temsirolimus (Wyeth), AP- 23573 (Ariad), or an analogue or derivative thereof).
  • mTOR kinase inhibitor e.g., ABT-578 (Abbott), temsirolimus (Wyeth), AP- 23573 (Ariad), or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is a microtubule inhibitor ⁇ e.g., antibody-maytansinoid conjugates from Biogen pouez, colchicines (MantiCore Pharmaceuticals), anticancer immunoconjugates from Johnson & Johnson, DIME from Octamer, gni-1f (GNI), huC242-DM4 or huMy9-6-DM1 (ImmunoGen), IDN-5404 (Indena), IMO-098 or IMOderm (Imotep), mebendazole (Introgen Therapeutics), microtubule poisons from Cambridge Enterprise, paclitaxel such as LOTAX from Aphios (CAS No.
  • a microtubule inhibitor ⁇ e.g., antibody-maytansinoid conjugates from Biogen pou, colchicines (MantiCore Pharmaceuticals), anticancer immunoconjugates from Johnson & Johnson, DIME from Octamer, gni-1f (GNI), huC
  • the microtubule inhibitor is a microtubule polymerization inhibitor such as vincamine, or an analogue or derivative thereof).
  • the fibrosis-inhibiting compound is a MIF inhibitor (e.g., AVP-13546 (Avanir), an MIF inhibitor from Genzyme, migration stimulation factor D, or an analogue or derivative thereof).
  • MIF inhibitor e.g., AVP-13546 (Avanir)
  • Genzyme a MIF inhibitor from Genzyme
  • migration stimulation factor D a MIF inhibitor from Genzyme
  • an analogue or derivative thereof e.g., a MIF inhibitor from Genzyme, migration stimulation factor D, or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is a MMP (stromolysin) inhibitor (e.g., anticancer tetracycline from Tetragenex, rhostatin (BioAxone), TIMP's from Sanofi-Aventis (CAS No. 86102-31-0), and MMP inhibitors form Cognosci and Tetragenex, or an analogue or derivative thereof).
  • MMP stromolysin
  • NK Neurokinin
  • the fibrosis-inhibiting compound is a neurokinin (NK) antagonist (e.g., anthrotainin (CAS No. 148084-40-6) (Sanofi-Aventis), an IBS thereapeutic such as SLV-332 from ArQuIe, MDL- 105212A (CAS No. 167261-60-1) (Ssanofi-Aventis), Pharmaprojects No. 2744, 3258 (CAS No. 139167-47-8) 4006, 4201 , or 5986 (Sanofi-Aventis), RP 67580 (CAS No. 135911-02-3), SR-144190 (CAS No.
  • NK neurokinin
  • SSR-240600 or SSR-241586 (Sanofi-Aventis), TKA-457 (Novartis), vestipitant mesylate (CAS No. 334476-64-1) (GlaxoSmithKline), Win-64821 (Sanofi-Aventis), PRX-96026 (Predix Pharmaceuticals), or an analogue or derivative thereof).
  • the fibrosis-inhibiting compound is a NF kappa B (NFKB) inhibitor (e.g., emodin (CAS No. 518-82-1), AVE-0545 or AVE-0547 (Sanofi-Aventis), bortezomib (CAS No. 179324-69-7) (Millennium Pharmaceuticals), dexanabinol (CAS No. 112924-45-5) (Pharmos), dexlipotam (Viatris), Pharmaprojects No. 6283 (INDRA) (OXiGENE), IPL-576092 (CAS No.
  • the fibrosis-inhibiting compound is a nitric oxide agonist (e.g., Acclaim, Angx-1039 or Angx-3227 (Angiogenix), CAS-1609 (CAS No. 158590-73-9) (Sanofi-Aventis), GCI-503 (Spear Therapeutics), HCT-3012 (CAS No. 163133-43-5) (NicOx), hydralazine + ISDN (NitroMed), isosorbide dinitrate, Diffutab (CAS No. 87-33-2) (Eurand), isosorbide mononitrate (CAS No.
  • a nitric oxide agonist e.g., Acclaim, Angx-1039 or Angx-3227 (Angiogenix), CAS-1609 (CAS No. 158590-73-9) (Sanofi-Aventis), GCI-503 (Spear Therapeutics), HCT-3012 (CAS No. 163133-43-5) (NicOx), hydral
  • nitroglycerin in the form of a nitroglycerin patch such as DERMATRANS from (Rottapharm), nitroglycerin (CAS No. 55-63-0) (from Cellegy Pharmaceuticals, Forest Laboratories, NovaDel, Schwarz Pharma, and Watson), NO-releasing prodrugs (Inotek), OM-294DP (OM PHARMA), oxdralazine (CAS No.
  • the fibrosis-inhibiting compound is an ornithine decarboxylase inhibitor ⁇ e.g., aplidine, or an analogue or derivative thereof).
  • the fibrosis-inhibiting compound is a p38 MAP kinase inhibitor (e.g., AZD-6703 (AstraZeneca), JX-401 (Jexys Pharmaceuticals), BMS-2 (Bristol-Myers Squibb), a p38 MAP kinase inhibitor from Novartis, a p38-alpha MAP kinase inhibitor from Amphora, Pharmaprojects No. 5704 (Pharmacopeia), SKF86002 (CAS No. 72873-74- 6), RPR-200765A (Sanofi-Aventis), SD-282 (Johnson & Johnson), TAK-715 (Takeda), or an analogue or derivative thereof).
  • a p38 MAP kinase inhibitor e.g., AZD-6703 (AstraZeneca), JX-401 (Jexys Pharmaceuticals), BMS-2 (Bristol-Myers Squibb), a
  • the fibrosis-inhibiting compound is a palmitoyl-protein thioesterase inhibitor (e.g., aplidine, or an analogue or derivative thereof).
  • a palmitoyl-protein thioesterase inhibitor e.g., aplidine, or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is a PDGF receptor kinase inhibitors (e.g., AAL-993, AMN-107, or ABP-309 (Novartis), AMG-706 (Amgen), BAY-57-9352 (Bayer), CDP-860 (UCB), E- 7080 (Eisai), imatinib (CAS No.
  • PDGF receptor kinase inhibitors e.g., AAL-993, AMN-107, or ABP-309 (Novartis), AMG-706 (Amgen), BAY-57-9352 (Bayer), CDP-860 (UCB), E- 7080 (Eisai), imatinib (CAS No.
  • the fibrosis-inhibiting compound is a peroxisome proliferator-activated receptor (PPAR) agonists (e.g., (-)- halofenate (Metabolex), AMG-131 (Amgen), antidiabetics from Japan Tobacco, AZD-4619, AZD-8450, or AZD-8677 (AstraZeneca), DRF-10945 or balaglitazone (Dr Reddy's), CS-00088 or CS-00098 (Chipscreen Biosciences), E-3030 (Eisai), etalocib (CAS No.
  • PPAR peroxisome proliferator-activated receptor
  • rosiglitazone maleate/glimepir (CAS No. 155141-29-0 and 93479-97-1), such as AVANDARYL or rosiglitazone maleate/metformin extend (CAS No. 155141-29-0 and 657-24-9) such as AVANDAMET, or rosiglitazone maleate+metformin, such as AVANDAMET (GlaxoSmithKline), tesaglitazar (AstraZeneca), LBM642, WY-14,643 (CAS No. 50892-23-4), or an analogue or derivative thereof).
  • the PPAR Agonist is a PPAR ⁇ agonist such as GW7647 or fenofibric acid (CAS No. 42017-89-0), a PPAR ⁇ agonist such as MCC-555 (CAS No. 161600-01-7), GW9662 or GW1929, a PPAER ⁇ agonist such as GW501516, a PPAR ⁇ and PPAR ⁇ agonist such L- 165,041 (CAS No. 79558-09-1), or an analogue or derivative thereof.
  • a PPAR ⁇ agonist such as GW7647 or fenofibric acid (CAS No. 42017-89-0)
  • MCC-555 CAS No. 161600-01-7
  • GW9662 or GW1929 a PPAER ⁇ agonist
  • GW501516 a PPAR ⁇ and PPAR ⁇ agonist
  • L- 165,041 CAS No. 79558-09-1
  • the fibrosis-inhibiting compound is a phosphatase inhibitor (e.g., diabetes thereapy such as SQMO3, SQDM38, SQDM60 from Sequenom, Pharmaprojects No. 4191 (Sanofi-Aventis), PRL- 3 inhibitors from Genzyme, WIP1 inhibitors from Amgen, or an analogue or derivative thereof).
  • a phosphatase inhibitor e.g., diabetes thereapy such as SQMO3, SQDM38, SQDM60 from Sequenom, Pharmaprojects No. 4191 (Sanofi-Aventis), PRL- 3 inhibitors from Genzyme, WIP1 inhibitors from Amgen, or an analogue or derivative thereof.
  • PDE Phosphodiesterase
  • the fibrosis-inhibiting compound is a phosphodiesterase (PDE) inhibitor (e.g., avanafil (Tanabe Seiyaku), dasantafil (CAS No. 569351-91-3) (Schering-Plough), A-906119 (CAS No. 134072-58-5) or DL-850 (Sanofi-Aventis), GRC-3015, GRC-3566, or GRC- 3886 (Glenmark), HWA-153 (CAS No.
  • PDE phosphodiesterase
  • the phosphodiesterase inhibitor is a phosphodiesterase III inhibitor (e.g., enoximone, or an analogue or derivative thereof).
  • the phosphodiesterase inhibitor is a phosphodiesterase IV inhibitor (e.g., fosfosal, Atopik (Barrier Therapeutics), triflusal, or an analogue or derivative thereof).
  • the phosphodiesterase inhibitor is a phosphodiesterase V inhibitor.
  • the fibrosis-inhibiting compound is a PKC inhibitor (e.g., HMR-105509 or P-10050 (Sanofi-Aventis), JNJ- 10164830 (Johnson & Johnson), Ro-31-8425 (CAS No. 131848-97-0), NPC- 15437 dihydrochloride (CAS No. 136449-85-9), or an analogue or derivative thereof).
  • PKC inhibitor e.g., HMR-105509 or P-10050 (Sanofi-Aventis), JNJ- 10164830 (Johnson & Johnson), Ro-31-8425 (CAS No. 131848-97-0), NPC- 15437 dihydrochloride (CAS No. 136449-85-9), or an analogue or derivative thereof.
  • the PKC inhibitor is an inhibitor of PKC beta (e.g., ruboxistaurin (EIi Lilly), or an analogue or derivative thereof).
  • the fibrosis-inhibiting compound is a platelet activating factor antagonist (e.g., dersalazine, or an analogue or derivative thereof).
  • a platelet activating factor antagonist e.g., dersalazine, or an analogue or derivative thereof.
  • Platelet-Derived Growth Factor Receptor Kinase Inhibitors In another embodiment, the fibrosis-inhibiting compound is a platelet-derived growth factor receptor kinase inhibitor (e.g., sorafenib tosylate, Raf or Ras inhibitors such as sorafenib tosylate from Bayer and Onyx Pharmaceuticals, or an analogue or derivative thereof).
  • the fibrosis-inhibiting compound is a prolyl hydroxylase inhibitor (e.g., FG-2216 (CAS No. 11096-26-7) or HIF agonists from FibroGen, or an analogue or derivative thereof).
  • FG-2216 CAS No. 11096-26-7
  • HIF agonists from FibroGen or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is a polymorphonuclear neutrophil inhibitor (e.g., orazipone, or an analogue or derivative thereof).
  • a polymorphonuclear neutrophil inhibitor e.g., orazipone, or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is a protein kinase B inhibitor (e.g., Akt-1 inhibitors from Amphora, or an analogue or derivative thereof).
  • Akt-1 inhibitors from Amphora, or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is a protein kinase C stimulant (e.g., bryostatin-1 , or analogue or derivative thereof).
  • a protein kinase C stimulant e.g., bryostatin-1 , or analogue or derivative thereof.
  • the fibrosis-inhibiting compound is a purine nucleoside analogue (e.g., cladrinbine and formulations thereof, such as MYLINAX from Serone SA and IVAX Research Inc. (Miami, FL), or an analogue or derivative thereof).
  • a purine nucleoside analogue e.g., cladrinbine and formulations thereof, such as MYLINAX from Serone SA and IVAX Research Inc. (Miami, FL), or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is a purinoreceptor P2X antagonist (e.g., AZD-9056 (AstraZeneca), R-1554 (Hoffmann-La Roche), AR-C118925XX (AstraZeneca), suramin (CAS No. 129-46-4), P2Y4 receptor from Euroscreen, or an analogue or derivative thereof).
  • AZD-9056 AstraZeneca
  • R-1554 Hoffmann-La Roche
  • AR-C118925XX AstraZeneca
  • suramin CAS No. 129-46-4
  • P2Y4 receptor from Euroscreen or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is a Raf kinase inhibitor (e.g., sorafenib tosylate, or an analogue or derivative thereof).
  • a Raf kinase inhibitor e.g., sorafenib tosylate, or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is a reversible inhibitor (e.g., lapatinib (GSK), or an analogue or derivative thereof).
  • GSK lapatinib
  • the fibrosis-inhibiting compound is a cytoplasmic tyrosine kinase inhibitor such as a SRC inhibitor (e.g., SRN-004 (Sirenade), gallium maltolate (Titan Pharmaceutcals), or an analogue or derivative thereof), or an analogue or derivative thereof).
  • SRC inhibitor e.g., SRN-004 (Sirenade), gallium maltolate (Titan Pharmaceutcals), or an analogue or derivative thereof), or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is a SDF-1 antagonist (e.g., CTCE-9908 (Chemokine Therapeutics), or an analogue or derivative thereof).
  • SDF-1 antagonist e.g., CTCE-9908 (Chemokine Therapeutics), or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is a sheddase inhibitor (e.g., INCB-7839 (Incyte Corporation), or an analogue or derivative thereof).
  • a sheddase inhibitor e.g., INCB-7839 (Incyte Corporation), or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is a SRC inhibitor (e.g., SRN-004 (Sirenade), or an analogue or derivative thereof).
  • SRC inhibitor e.g., SRN-004 (Sirenade), or an analogue or derivative thereof.
  • the SRC inhibitor is a SRC kinase inhibitor (e.g., AZD0530 (AstraZeneca), or an analogue or derivative thereof).
  • the fibrosis-inhibiting compound is a stromelysin inhibitor (e.g., glucosamine sulfate, or an analogue or derivative thereof).
  • a stromelysin inhibitor e.g., glucosamine sulfate, or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is a syk kinase inhibitor (e.g., R406 (Rigel), or an analogue or derivative thereof).
  • a syk kinase inhibitor e.g., R406 (Rigel), or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is a telomerase inhibitor (e.g., AS-1410 (Antisoma), or an analogue or derivative thereof).
  • the fibrosis-inhibiting compound is a TGF beta inhibitor (e.g., pirfenidone (CAS No. 53179-13-8) (MARNAC), tranilast (CAS No. 53902-12-8) (Kissei), IN-1130 (ln2Gen), mannose-6- phosphate (BTG), TGF- ⁇ antagonists from Inflazyme (Pharmaprojects No. 6075), TGF- ⁇ antagonists (e.g., 1090 and 1091 from Sydney; non-industrial source), TGF- ⁇ I receptor kinase inhibitors from EIi Lilly, TGF- ⁇ receptor inhibitors from Johnson & Johnson, or an analogue or derivative thereof).
  • TGF beta inhibitor e.g., pirfenidone (CAS No. 53179-13-8) (MARNAC), tranilast (CAS No. 53902-12-8) (Kissei), IN-1130 (ln2Gen), mannose-6- phosphate (BTG), TGF- ⁇ antagonists from Inflazy
  • the fibrosis-inhibiting compound is a TNF ⁇ antagonist or TACE inhibitors (e.g., adalimumab (CAS No. 331731- 18-1) (Cambridge Antibody Technology), AGIX-4207 (AtheroGenics), AGT-1 (Advanced Biotherapy), an anti-inflammatory from Borean Pharma, Cellzome, or Paradigm Therapeutics, anti-inflammatory vaccine (TNF-alpha kinoid) from Neovacs, humanized anti-TNF antibody or an anti-TNF MAb (CB0006) Celltech (UCB), apratastat (CAS No.
  • TNF ⁇ antagonist or TACE inhibitors e.g., adalimumab (CAS No. 331731- 18-1) (Cambridge Antibody Technology), AGIX-4207 (AtheroGenics), AGT-1 (Advanced Biotherapy), an anti-inflammatory from Borean Pharma, Cellzome, or Paradigm Therapeutics, anti-inflammatory vaccine (TNF-alpha
  • TNF antagonists form ProStrakan, and Synergen, TNF inhibitors (Amgen), TNF-alpha antagonists from Dynavax Technologies and Jerina AG (Germany), TNF-alpha inhibitors from IBFB Pharma and Xencor (Xencor), torbafylline (CAS No. 105102-21-4) (Sanofi-Aventis), UR-1505 (Uriach), VT-346 (Viron Therapeutics), YSIL6 (Y's Therapeutics), YSTH2 (Y's Therapeutics), NPI-1302a-3 (Nereus Pharmaceuticals, a TNF antagonist from Jerina AG (Germany), dersalazine, or an analogue or derivative thereof).
  • the fibrosis-inhibiting compound is a tumor necrosis factor (TNF) antagonist (e.g., anti-inflammatory compounds from Biota Inc., or an analogue or derivative thereof).
  • TNF tumor necrosis factor
  • the fibrosis-inhibiting compound is a Toll receptor antagonist (e.g., E5564 (Eisai Pharmaceuticals), or an analogue or derivative thereof).
  • a Toll receptor antagonist e.g., E5564 (Eisai Pharmaceuticals), or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is a tubulin antagonist (e.g., synthadotin, KRX-0403 (Keryx Biopharmaceuticals), or an analogue or derivative thereof).
  • tubulin antagonist e.g., synthadotin, KRX-0403 (Keryx Biopharmaceuticals), or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is a tyrosine kinase inhibitor (e.g., SU-011248 (e.g., SUTENT from Pfizer Inc. (New York, NY), BMS-354825, PN-355 (Paracelsian Pharmaceuticals), AGN-199659 (Allergan), (e.g., AAL-993 or ABP-309 (Novartis), adaphostin (NIH), AEE-788 (Novartis), AG-013736 (OSI Pharmaceuticals), AG-13736 (Pfizer), ALT-110 (Alteris Therapeutics), AMG-706 (Amgen), anticancer MAbs from Xencor, anti-EGFrvlll MAbs from Abgenix, anti-HER2 MAb from Abiogen, AZD-2171 or AZD-9935 (AstraZeneca), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim),
  • the tyrosine kinase inhibitor is an EGFR tyrosine kinase inhibitor such as EKB-569 (Wyeth), or an analogue or derivative thereof).
  • the fibrosis-inhibiting compound is a VEGF Inhibitor (e.g., AZD2171 (AstraZeneca), or an analogue or derivative thereof).
  • a VEGF Inhibitor e.g., AZD2171 (AstraZeneca), or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is a vitamin D receptor agonist (e.g., BXL-628, BXL-922 (BioXell), or an analogue or derivative thereof).
  • a vitamin D receptor agonist e.g., BXL-628, BXL-922 (BioXell), or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is an histamine receptor antagonist.
  • the histamine receptor antagonists such as H1 , H2, and H3 histamine receptor antagonists, block the production of pro-inflammatory cytokines such as TNFa and IL-1 (e.g., IL-1 ⁇ ).
  • the histamine receptor antagonist inhibit NFkB activation.
  • H1 histamine receptor antagonists include phenothiazines, such as promethazine, and alkylamines, such as chlorpheniramine (CAS No. 7054- 11-7), brompheniramine (CAS No.
  • histamine receptor antagonists include broad spectrum histamine receptor antagonists such as methylxanthines (e.g., theophylline, theobromine, and caffeine).
  • H2 receptor antagonists include those with a histamine-like structure including cimetidine (available under the tradename TAGAMET from SmithKline Beecham Phamaceutical Co., Wilmington, DE), ranitidine (available under the tradename ZANTAC from Warner Lambert Company, Morris Plains, NJ), famotidine (available under the tradename PEPCID from Merck & Co., Whitehouse Station, NJ), nizatidine (available under the tradename AXID from Reliant Pharmaceuticals, Inc., Liberty Corner, NJ), nizatidine, and roxatidine acetate (CAS No. 78628-28-1).
  • cimetidine available under the tradename TAGAMET from SmithKline Beecham Phamaceutical Co., Wilmington, DE
  • ranitidine available under the tradename ZANTAC from Warner Lambert Company, Morris Plains, NJ
  • famotidine available under the tradename PEPCID from Merck & Co., Whitehouse Station, NJ
  • nizatidine available under the tradename AXID from Reliant Pharmaceutical
  • H3 receptor antagonists e.g., thioperamide and thioperamide maleate salt
  • anti-histamines such as tricyclic dibenozoxepins, ethanolamines, ethylenediamines, piperizines, piperidines, and pthalazinones.
  • the fibrosis-inhibiting compound is an alpha adrenergic receptor antagonist.
  • Alpha adrenergic receptor antagonists may inhibit the production of pro-inflammatory cytokines such as TNFa.
  • the alpha adrenergic receptor antagonist may be an alpha-1 and/or an alpha-2 adrenergic receptor antagonist.
  • Representative examples of aipha-1 /alpha-2 antagonists include phenoxybenzamine.
  • the alpha adrenergic receptor antagonist is a haloalkylamine compound or a catecholamine uptake inhibitor.
  • Representative examples of alpha-1 adrenergic receptor antagonists include phenoxybenzamine hydrochloride and prazosin, a piperizinyl quinazoline.
  • alpha-2 adrenergic receptor antagonists include imadazole based compounds such as idazoxan (CAS No. 79944-56-2), idazoxan hydrochloride, and loxapine succinate salt (CAS No. 27833-64-3). Additional examples of alpha adrenergic receptor antagonists include prazosin hydrochloride.
  • the fibrosis-inhibiting compound is an anti-psychotic compound, such as a phenothiazine compound or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is a phenothiazine derivative capable of suppressing the production of pro-inflammatory cytokines such as TNFa and/or IL-1.
  • phenothiazine compounds include chlorpromazine, fluphenazine, trifluorphenazine, mesoridazine, thioridazine, and perphenazine.
  • anti-psychotic compounds include thioxanthines such as chlorprothixene and thiothixene, clozapine, loxapine succinate, and olanzapine.
  • the fibrosis-inhibiting compound is CaM kinase Il inhibitor, such as a lavendustin C, or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is CaM kinase Il inhibitor, such as a lavendustin C, or an analogue or derivative thereof. 109) G Protein Agonist
  • the fibrosis-inhibiting compound is G protein agonist, such as aluminum fluoride, or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is an antibiotic, such as apigenin (Cas No. 520-36-5), ampicillin sodium salt (CAS No. 69-52-3), puromycin, or an analogue or derivative thereof.
  • antibiotic such as apigenin (Cas No. 520-36-5), ampicillin sodium salt (CAS No. 69-52-3), puromycin, or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is an anti-microbial agent, such as brefeldin A (CAS No. 20350-15-6), terbinafine, benzoyl peroxide, pentamidine, ornidazole, imidazole, ketocanazole, sulconazole nitrate salt, or an analogue or derivative thereof.
  • an anti-microbial agent such as brefeldin A (CAS No. 20350-15-6), terbinafine, benzoyl peroxide, pentamidine, ornidazole, imidazole, ketocanazole, sulconazole nitrate salt, or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is DNA topoisomerase I inhibitor, such as ⁇ -lapachone (CAS No. 4707-32-8), or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is DNA topoisomerase Il inhibitor, such as (-)-arctigenin (CAS No. 7770-78-7), aurintricarboxylic acid, or an analogue or derivative thereof.
  • DNA topoisomerase Il inhibitor such as (-)-arctigenin (CAS No. 7770-78-7), aurintricarboxylic acid, or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is thromboxane A2 receptor inhibitor, such as BM-531 (CAS No. 284464-46- 6), ozagrel hydrochloride (CAS No. 78712-43-3), or an analogue or derivative thereof. 113) D2-Dopamine Receptor Antagonist
  • the fibrosis-inhibiting compound is a D2 dopamine receptor antagonist, such as clozapine (CAS No. 5786-21-0), mesoridazine benzenesulfonate, or an analogue or derivative thereof.
  • D2 dopamine receptor antagonist such as clozapine (CAS No. 5786-21-0), mesoridazine benzenesulfonate, or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is a Peptidyl-Prolyl Cis/Trans lsomerase Inhibitor, such as juglone (CAS No. 481-39-0), or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is a dopamine antagonist, such as thiothixene, thioridazine hydrochloride, or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is an anesthetic compound, such as lidocaine (CAS No. 137-58-6), or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is a clotting factor, such as menadione (CAS No. 58-27-5), or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is a lysyl hydrolase inhibitor, such as minoxidil (CAS No. 38304-91-5), or an analogue or derivative thereof.
  • a lysyl hydrolase inhibitor such as minoxidil (CAS No. 38304-91-5)
  • the fibrosis-inhibiting compound is a muscarinic receptor inhibitor, such as perphenazine (CAS No. 58-39-9), or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is a superoxide anion generator, such as plumbagin (CAS No. 481-42-5), or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is a steroid, such as prednisolone, prednisolone 21-acetate (CAS No. 52-21-1), loteprednol etabonate, (CAS No. 82034-46-6), clobetasol propionate, or an analogue or derivative thereof.
  • a steroid such as prednisolone, prednisolone 21-acetate (CAS No. 52-21-1), loteprednol etabonate, (CAS No. 82034-46-6), clobetasol propionate, or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is an antiproliferative agent, such as silibinin (CAS No. 22888-70-6), silymarin (CAS No. 65666-07-1), 1 ,2-hexanediol, dioctyl phthalate (CAS No.
  • zirconium (IV) oxide zirconium (IV) oxide, glycyrrhizic acid, spermidine trihydrochloride or tetrahydrochloride, CGP 74514A, spermine tetrahydrochloride, NG-methyl- L-arginine acetate salt, galardin, halofuginone hydrobromide (HBr), fascaplysin, or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is a diuretic, such as spironolactone (CAS No. 52-01-7), or an analogue or derivative thereof. 124) Anti-Coagulants
  • the fibrosis-inhibiting compound is an anti-coagulant, such as fucoidan from Fucus vesiculosus (CAS No. 9072- 19-9), or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is a cyclic GMP agonist, such as sinitrodil (CAS No. 143248-63-9), or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is an adenylate cyclase agonist, such as histamine (CAS No. 51-45-6), or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is an antioxidant, such as morpholine, phytic acid dipotassium salt, (-)- epigallocatechin or (-)-epigallocatechin gallate from green tea (CAS Nos. 970-74-1 and 1257-08-5, respectively), (-)-epigallocatechin gallate (CAS No. 989-51-5), nobiletin (CAS No. 478-01-3), probucol (CAS No. 23288-49-5), phosphorous acid, hesperetin, L-ascorbyl-2-phosphate, magnesium salt (CAS No. 84309-23-9), catechin, ( ⁇ )-naringenin (CAS No. 67604-48-2), (-)- epicatechin, (-)-epicatechin gallate, 3-hydroxyflavone, (-)-arctigenin, or an analogue or derivative thereof.
  • an antioxidant such as morpholine, phytic acid dipotassium salt, (-)-
  • the fibrosis-inhibiting compound is a nitric oxide synthase inhibitor, such as ammonium pyrrolidinedithiocarbamate (CAS No. 5108-96-3), or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is a reversible nitric oxide synthase inhibitor, such as NB-methyl-L-arginine acetate salt (L-NMMA) (CAS No. 53308-83-1), or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is an antineoplastic agent, such as tirapazamine (CAS No. 27314-97-2), fludarabine (CAS No. 21679-14-1), cladribine, imatinib mesilate, or an analogue or derivative thereof.
  • antineoplastic agent such as tirapazamine (CAS No. 27314-97-2), fludarabine (CAS No. 21679-14-1), cladribine, imatinib mesilate, or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is a DNA synthesis inhibitor, such as S-(2-hydroxy-5-nitrobenyl)-6-thioguanosine or uracilfludarabine phosphate (CAS No. 75607-67-9), 6,11-dihydroxy-5,12- naphthacenedione, or an analogue or derivative thereof.
  • a DNA synthesis inhibitor such as S-(2-hydroxy-5-nitrobenyl)-6-thioguanosine or uracilfludarabine phosphate (CAS No. 75607-67-9), 6,11-dihydroxy-5,12- naphthacenedione, or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is a DNA alkylating agent, such as dacarbazine (CAS No. 4342-03-4), temozolomide, procarbazine HCI, or an analogue or derivative thereof.
  • a DNA alkylating agent such as dacarbazine (CAS No. 4342-03-4), temozolomide, procarbazine HCI, or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is a DNA methylation inhibitor, such as decitabine (CAS No. 2353-33-5), or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is a NSAID agent, such as nabumetone, benzydamine hydrochloride, or an analogue or derivative thereof. 134) Peptidylglycine Alpha-Hydroxylatinq Monooxygenase Inhibitors
  • the fibrosis-inhibiting compound is a peptidylglycine alpha-hydroxylating monooxygenase inhibitor, such as trans- styrylacetic acid, or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is a MEK1/MEK 2 inhibitor, such as U0126 (CAS No. 109511-58-2), or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is an NO synthase inhibitor, such as L-NAME (CAS No. 53308-83-1), NG-Methyl- L-arginine acetate salt, or an analogue or derivative thereof.
  • NO synthase inhibitor such as L-NAME (CAS No. 53308-83-1), NG-Methyl- L-arginine acetate salt, or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is retinoic acid receptor antagonist, such as isotretinoin (CAS No. 4759-48-2), or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is an ACE inhibitor, such as quinapril hydrochloride (CAS No. 85441-61-8), enalapril, or an analogue or derivative thereof.
  • ACE inhibitor such as quinapril hydrochloride (CAS No. 85441-61-8), enalapril, or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is a glycosylation inhibitor, such as aminoguanidine hydrochloride, castanospermine, or an analogue or derivative thereof.
  • a glycosylation inhibitor such as aminoguanidine hydrochloride, castanospermine, or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is an intracellular calcium influx inhibitor, such as TAS-301 (CAS No. 193620-69- 8), or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is an anti-emetic agent, such as amifostine (CAS No. 20537-88-6), or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is an acetylcholinesterase inhibitor, such as (-)-huperzine A (CAS No. 102518-79- 6), or an analogue or derivative thereof.
  • ALK-5 Receptor Antagonists . . _ . _ . . .
  • the fibrosis-inhibiting compound is an ALK-5 receptor antagonist, such as SB 431542 (CAS No. 301836-41-9), or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is a RAR/RXT antagonist, such as 9-cis-retinoic acid, or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is a elF-2a inhibitor, such as salubrinal, or an analogue or derivative thereof. 146) S-Adenosyl-L-Homocysteine Hydrolase Inhibitors
  • the fibrosis-inhibiting compound is a S-adenosyl-L-homocysteine hydrolase inhibitor, such as 3-deazaadenosine, or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is an estrogen agonist, such as coumestrol, bisphenol A, 1-linoleoyl-rac-glycerol (CAS No. 2277-28-3), daidzein (4,7-dihydroxy-iso-flavone), dihexyl phthalate, kaempferol, formononetin, , or an analogue or derivative thereof.
  • estrogen agonist such as coumestrol, bisphenol A, 1-linoleoyl-rac-glycerol (CAS No. 2277-28-3), daidzein (4,7-dihydroxy-iso-flavone), dihexyl phthalate, kaempferol, formononetin, , or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is a serotonin receptor inhibitor, such as amitriptyline hydrochloride, or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is an anti-thrombotic agent, such as geniposidic acid, geniposide, or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is a tryptase inhibitors, such as 2-azetidinone, or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is a pesticide, such as allyl disulfide, or an analogue or derivative thereof. 152) Bone Mineralization Promotor
  • the fibrosis-inhibiting compound is a bone mineralization promotor, such as glycerol 2-phosphate disodium salt hydrate, or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is a bisphosphonate compound, such as risedronate, or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is an anti-inflammatory compound, such as aucubin, cepharanthine, or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is a DNA methylation promotor, such as 5-azacytidine, or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is an anti-spasmodic agent, such as 2-hydroxy-4,6-dimethoxyacetophenone, or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is a protein synthesis inhibitor, such as oxytetracycline hydrochloride, or an analogue or derivative thereof. 158) ⁇ -Glucosidase Inhibitors
  • the fibrosis-inhibiting compound is a ⁇ -glucosidase inhibitor, such as myricetin (CAS No. 529-44-2), or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is a calcium channel blocker, such as verapamil, nitrendipine, or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is a L-type calcium channel blocker, such as nifedipine (CAS No. 21829-25-4), (+)-cis-diltiazem hydrochloride, or an analogue or derivative thereof.
  • L-type calcium channel blocker such as nifedipine (CAS No. 21829-25-4), (+)-cis-diltiazem hydrochloride, or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is a T-type calcium channel blocker, such as penfluridol (CAS No. 26864-56-2), or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is a pyruvate dehydrogenase activator, such as dichloroacetic acid, or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is a prostaglandin inhibitor, such as betulinic acid, or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is a sodium channel inhibitor, such as amiloride hydrochloride hydrate, or an analogue or derivative thereof. 163) Serine Protease Inhibitors
  • the fibrosis-inhibiting compound is a serine protease inhibitor, such as gabexate mesylate, or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is an intracellular calcium flux inhibitor, such as thapsigargin, or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is a JAK2 inhibitor (e.g., AG-490 (CAS No. 134036-52-5), or an analogue or derivative thereof).
  • JAK2 inhibitor e.g., AG-490 (CAS No. 134036-52-5), or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is an androgen inhibitor (e.g., tibolone (CAS No. 5630-53-5), or an analogue or derivative thereof).
  • an androgen inhibitor e.g., tibolone (CAS No. 5630-53-5), or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is an aromatase inhibitor (e.g., letrozole, or an analogue or derivative thereof).
  • an aromatase inhibitor e.g., letrozole, or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is an anti-viral agent, such as imiquimod, or an analogue or derivative thereof. 169) 5-HT Inhibitors
  • the fibrosis-inhibiting compound is a 5-HT inhibitor, such as ketanserin tartrate, amoxapine, or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is a FXR antagonist, such as guggulsterone (CAS No. 95975-55-6), or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is an actin polymerization and stabilization promotor, such as jasplakinolide, or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is an AXOR12 agonist, such as metastin (KiSS-1 (112-121), or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is an angiotensin Il receptor agonist, such as losartan potassium, or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is a platelet aggregation inhibitor, such as clopidogrel, or an analogue or derivative thereof. 175) CB1/CB2 Receptor Agonists
  • the fibrosis-inhibiting compound is a CB1/CB2 receptor agonist, such as HU-210 (CAS No. 112830-95-2), or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is a norepinephrine reuptake inhibitor, such as nortriptyline hydrochloride, or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is a selective serotonin reuptake inhibitor, such as paroxetine maleate, or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is a reducing agent such as VVW-85 (Inotek), or an analogue or derivative thereof.
  • the fibrosis-inhibiting compound is an immunomodulators such as Bay 11-7085, (-)-arctigenin, idazoxan hydrochloride, or an analogue or derivative thereof .
  • immunomodulators such as Bay 11-7085, (-)-arctigenin, idazoxan hydrochloride, or an analogue or derivative thereof .
  • the therapeutic agents useful in the present invention also include anti-infective agents. Such agents may reduce the likelihood of infection upon implantation of the composition or a medical implant and may be used in combination of an anti-fibrosis agent and/or a polymer. Infection is a common complication of the implantation of foreign bodies such as, for example, medical devices and implants. Foreign materials provide an ideal site for micro- organisms to attach and colonize. It is also hypothesized that there is an impairment of host defenses to infection in the microenvironment surrounding a foreign material. These factors make medical implants particularly susceptible to infection and make eradication of such an infection difficult, if not impossible, in most cases. In many cases, an infected implant or device must be surgically removed from the body in order to eradicate the infection.
  • the present invention provides agents (e.g., chemotherapeutic agents) that can be released from a composition, and which have potent antimicrobial activity at extremely low doses.
  • agents e.g., chemotherapeutic agents
  • a wide variety of anti-infective agents can be utilized in combination with the present compositions. Suitable anti-infective agents may be readily determined based upon the assays provided in Example 30).
  • agents that can be, used as anti-infective agents such as: (A) anthracyclines (e.g., doxorubicin and mitoxantrone), (B) fluoropyrimidines (e.g., 5-FU), (C) folic acid antagonists (e.g., methotrexate), (D) podophylotoxins (e.g., etoposide), (E) camptothecins, (F) hydroxyureas, and (G) platinum complexes (e.g., cisplatin).
  • anthracyclines e.g., doxorubicin and mitoxantrone
  • fluoropyrimidines e.g., 5-FU
  • C folic acid antagonists
  • D podophylotoxins
  • E camptothecins
  • F hydroxyureas
  • platinum complexes e.g., cisplatin
  • the therapeutic anti-infective agent is an anthracycline.
  • Anthracyclines have the following general structure, where the R groups may be a variety of organic groups:
  • R groups are as follows: Ri is CH 3 or CH 2 OH; R 2 is daunosamine or H; R 3 and R 4 are independently one of OH, NO 2 , NH 2 , F, Cl, Br, I, CN 1 H or groups derived from these; R 5 is hydrogen, ydroxyl, or methoxy; and R 6-8 are aii hydrogen. Alternatively, R 5 and Re are hydrogen and R 7 and R 8 are alkyl or halogen, or vice versa.
  • Ri may be a conjugated peptide.
  • R 5 may be an ether linked aikyl group.
  • R 5 may be OH or an ether linked alkyl group.
  • R 1 may also be linked to the anthracycline ring by a group other than C(O), such as an alkyl or branched alkyl group having the C(O) linking moiety at its end, such as -CH 2 CH(CH 2 -X)C(O)-R 1 , wherein X is H or an alkyl group (see, e.g., U.S. Patent 4,215,062).
  • R3 may have the following structure:
  • R 9 is OH either in or out of the plane of the ring, or is a second sugar moiety such as R 3 .
  • R 10 may be H or form a secondary amine with a group such as an aromatic group, saturated or partially saturated 5 or 6 membered heterocyclic having at least one ring nitrogen (see U.S. Patent 5,843,903).
  • R 1O may be derived from an amino acid, having the structure -C(O)CH(NHR 11 )(R 12 ), in which R 11 is H, or forms a C 3-4 membered alkylene with R 12 .
  • R 12 may be H, alkyl, aminoalkyl, amino, hydroxyl, mercapto, phenyl, benzyl or methylthio (see U.S. Patent 4,296,105).
  • exemplary anthracyclines are doxorubicin, daunorubicin, idarubicin, epirubicjn, pirarubicin, zorubicin, and carubicin. Suitable compounds have the structures:
  • anthracyclines are anthramycin, mitoxantrone, menogaril, nogalamycin, aclacinomycin A, olivomycin A, chromomycin A 3 , and plicamycin having the structures: R 1 R 2 R,
  • the ant-infective therapeutic agent is a fluoropyrimidine analog, such as 5-fluorouracil, or an analogue or derivative thereof, including carmofur, doxifluridine, emitefur, tegafur, and floxuridine.
  • fluoropyrimidine analog such as 5-fluorouracil
  • an analogue or derivative thereof including carmofur, doxifluridine, emitefur, tegafur, and floxuridine.
  • Exemplary compounds have the structures:
  • fluoropyrimidine analogues include 5-FudR (5- fluoro-deoxyuridine), or an analogue or derivative thereof, including 5- iododeoxyuridine (5-ludR), 5-bromodeoxyuridine (5-BudR), fluorouridine triphosphate (5-FUTP), and fluorodeoxyuridine monophosphate (5-dFUMP).
  • 5-FudR 5- fluoro-deoxyuridine
  • an analogue or derivative thereof including 5- iododeoxyuridine (5-ludR), 5-bromodeoxyuridine (5-BudR), fluorouridine triphosphate (5-FUTP), and fluorodeoxyuridine monophosphate (5-dFUMP).
  • Exemplary compounds have the structures:
  • fluoropyrimidine analogues include N3-alkylated analogues of 5-fluorouracil (Kozai et a/., J. Chem. Soc, Perkin Trans.
  • the anti-infective therapeutic agent is a folic acid antagonist, such as methotrexate or derivatives or analogues thereof, including edatrexate, trimetrexate, raltitrexed, piritrexim, denopterin, tomudex, and pteropterin.
  • Methotrexate analogues have the following general structure:
  • R group may be selected from organic groups, particularly those groups set forth in U.S. Patent Nos. 5,166,149 and 5,382,582.
  • R 1 may be N
  • R 2 may be N or C(CH 3 )
  • R 3 and R 3 ' may H or alkyl, e.g., CH 3
  • R 4 may be a single bond or NR, where R is H or alkyl group.
  • R ⁇ ( 6, 8 nriay be H, OCH 3 , or alternately they can be halogens or hydro groups.
  • R 7 is a side chain of the general structure:
  • the carboxyl groups in the side chain may be esterified or form a salt such as a Zn 2+ salt.
  • Rg and Rio can be NH 2 or may be alkyl substituted.
  • Exemplary folic acid antagonist compounds have the structures:
  • N-( ⁇ -aminoacyl) methotrexate derivatives Cheung et al., Pteridines 3(1 -2): 101 -2, 1992
  • biotin methotrexate derivatives Fean et al., Pteridines 3(1-2):131-2, 1992
  • D-glutamic acid or D- erythrou, threo-4-fluoroglutamic acid methotrexate analogues McGuire et al., Biochem. Pharmacol.
  • N-(L- ⁇ - aminoacyl) methotrexate derivatives (Cheung et al., Heterocycles 2 ⁇ (2):751- 8, 1989), meta and ortho isomers of aminopterin (Rosowsky ef al., J. Med. Chem. 32(12):2582, 1989), hydroxymethy Methotrexate (DE 267495), ⁇ - fluoromethotrexate (McGuire et al., Cancer Res. 49(16):4517-25, 1989), polyglutamyl methotrexate derivatives (Kumar ef a/., Cancer Res.
  • the anti-infective therapeutic agent is a podophyllotoxin, or a derivative or an analogue thereof.
  • exemplary compounds of this type are etoposide or teniposide, which have the following structures:
  • podophyllotoxins include Cu(II)-VP-16 (etoposide) complex (Tawa et al., Bioorg. Med. Chem. 6(7): 1003-1008, 1998), pyrrolecarboxamidino-bearing etoposide analogues (Ji et al., Bioorg. Med. Chem. Lett. 7(5):607-612, 1997), 4 ⁇ -amino etoposide analogues (Hu, University of North Carolina Dissertation, 1992), ⁇ -lactone ring-modified arylamino etoposide analogues (Zhou et al., J. Med. Chem.
  • the anti-infective therapeutic agent is camptothecin, or an analogue or derivative thereof.
  • Camptothecins have the following general structure.
  • X is typically O, but can be other groups, e.g., NH in the case of 21-lactam derivatives.
  • Ri is typically H or OH, but may be other groups, e.g., a terminally hydroxylated C 1 - 3 alkane.
  • R 2 is typically H or an amino containing group such as (CHs) 2 NHCH 2 , but may be other groups e.g., NO 2 , NH 2 , halogen (as disclosed in, e.g., U.S. Patent 5,552,156) or a short alkane containing these groups.
  • R 3 is typically H or a short alkyl such as C 2 H 5 .
  • R 4 is typically H but may be other groups, e.g., a methylenedioxy group with R 1 .
  • camptothecin compounds include topotecan, irinotecan (CPT-11), 9-aminocamptothecin, 21-lactam-20(S)-camptothecin, 10,11-methylenedioxycamptothecin, SN-38, 9-nitrocamptothecin, 10- hydroxycamptothecin.
  • Exemplary compounds have the structures:
  • Camptothecins have the five rings shown here.
  • the ring labeled E must be intact (the lactone rather than carboxylate form) for maximum activity and minimum toxicity.
  • Camptothecins are believed to function as topoisomerase I inhibitors and/or DNA cleavage agents.
  • the anti-infective therapeutic agent of the present invention may be a hydroxyurea.
  • Hydroxyureas have the following general structure:
  • Suitable hydroxyureas are disclosed in, for example, U.S. Patent No. 6,080,874, wherein Ri is: and R 2 is an alkyl group having 1-4 carbons and R 3 is one of H, acyl, methyl, ethyl, and mixtures thereof, such as a methylether.
  • R 1 is a cycloalkenyl group, for example N-(3-(5-(4- fluorophenylthio)-furyl)-2-cyclopenten-1-yl)N-hydroxyurea
  • R 2 is H or an alkyl group having 1 to 4 carbons and R 3 is H
  • X is H or a cation.
  • Suitable hydroxyureas are disclosed in, e.g., U.S. Patent No. 4,299,778, wherein Ri is a phenyl group substituted with one or more fluorine atoms; R 2 is a cyclopropyl group; and R 3 and X is H.
  • n is 0-2 and Y is an alkyl group.
  • the hydroxyurea has the structure:
  • the anti-infective therapeutic agent is a platinum compound.
  • suitable platinum complexes may be of Pt(II) or Pt(IV) and have this basic structure:
  • X and Y are anionic leaving groups such as sulfate, phosphate, carboxylate, and halogen; R 1 and R 2 are alkyl, amine, amino alkyl any may be further substituted, and are basically inert or bridging groups.
  • Z 1 and Z 2 are non-existent.
  • Z 1 and Z 2 may be anionic groups such as halogen, hydroxy, carboxylate, ester, sulfate or phosphate. See, e.g., U.S. Patent Nos. 4,588,831 and 4,250,189.
  • Suitable platinum complexes may contain multiple Pt atoms. See, e.g., U.S. Patent Nos. 5,409,915 and 5,380,897.
  • platinum compounds are cisplatin, carboplatin, oxaliplatin, and miboplatin having the structuresf
  • platinum compounds include (CPA) 2 Pt(DOLYM) and (DACH)Pt(DOLYM) cisplatin (Choi et al., Arch. Pharmacal Res. 22(2):151-156, 1999), Cis-(PtCI 2 (4,7-H-5-methyl-7- oxo)1, 2,4(triazolo(1,5-a)pyrimidine) 2 ) (Navarro et al., J. Med. Chem. 47(3):332-338, 1998), (Pt(cis-1 ,4-DACH)(trans-CI 2 )(CBDCA)) . Y 2 MeOH cisplatin (Shamsuddin et al., Inorg. Chem.
  • the anti-infective therapeutic agent is a quinolone antibacterial agent.
  • quinolone antibacterial agents include garenoxacin (Schering Plough) or an analogue or derivative thereof.
  • Drug dose administered from the present compositions for prevention or inhibition of infection in accordance with the present invention will depend on a variety of factors, including the type of formulation, the location of the treatment site, and the type of condition being treated. However, certain principles can be applied in the application of this art. Drug dose can be calculated as a function of dose per unit area (of the treatment site), total drug dose administered can be measured and appropriate surface concentrations of active drug can be determined. Drugs are to be used .at concentrations that range from several times more than to 50%, 20%, 10%, 5%, or even less than 1% of the concentration typically used in a single anti-infective systemic dose application.
  • the anti- infective agent is released from the composition in effective concentrations in a time period that may be measured from the time of infiltration into tissue adjacent to the device, which ranges from about less than 1 day to about 180 days.
  • the release time may also be from about less than 1 day to about 180 days; from about 7 days to about 14 days; from about 14 days to about 28 days; from about 28 days to about 56 days; from about 56 days to about 90 days; from about 90 days to about 180 days.
  • the exemplary anti-infective agents used alone or in combination, should be administered under the following dosing guidelines.
  • the total amount (dose) of anti-infective agent in the composition can be in the range of about 0.01 ⁇ g-1 ⁇ g, or about 1 ⁇ g-10 ⁇ g, or about 10 ⁇ g-1 mg, or about 1 mg to 10 mg, or about 10 mg-100 mg, or about 100 mg to 250 mg, or about 250 mg-1000 mg.
  • the dose (amount) of anti-infective agent per unit area of device or tissue surface to which the agent is applied may be in the range of about 0.01 ⁇ g/mm 2 - 1 ⁇ g/mm 2 , or about 1 ⁇ g/mm 2 - 10 ⁇ g/mm 2 , or about 10 ⁇ g/mm 2 - 100 ⁇ g/mm 2 , or about 100 ⁇ g/mm 2 to 250 ⁇ g/mm 2 , or about 250 ⁇ g/mm 2 - 1000 ⁇ g/mm 2 .
  • the above dosing parameters should be utilized in combination with the release rate of the drug from the composition such that a minimum concentration of about 10 "8 M to 10 "7 M, or about 10 "7 M to lO '6 M about 10 "6 M to 10 "5 M or about 10 "5 M to 10 '4 M of the agent is maintained on the tissue surface.
  • the total dose of doxorubicin applied to the device or implant should not exceed 25 mg (range of 0.1 ⁇ g to 25 mg). In a particularly preferred embodiment, the total amount of drug applied should be in the range of 1 ⁇ g to 5 mg.
  • the dose per unit area i.e., the amount of drug as a function of the surface area of the portion of the implant to which drug is applied and/or incorporated should fall within the range of 0.01 ⁇ g - 100 ⁇ g per mm 2 of surface area.
  • doxorubicin should be applied to the implant surface at a dose of 0.1 ⁇ g/mm 2 - 10 ⁇ g/mm 2 .
  • the above dosing parameters should be utilized in combination with the release rate of the drug from the implant surface such that a minimum concentration of 10 "7 - 10 "4 M of doxorubicin is maintained on the surface. It is necessary to insure that surface drug concentrations exceed concentrations of doxorubicin known to be lethal to multiple species of bacteria and fungi (i.e., are in excess of 10 "4 M; although for some embodiments lower concentrations are sufficient).
  • doxorubicin is released from the surface of the implant such that anti-infective activity is maintained for a period ranging from several hours to several months.
  • the drug is released in effective concentrations for a period ranging from 1 week - 6 months.
  • analogues and derivatives of doxorubicin (as described previously) with similar functional activity can be utilized for the purposes of this invention; the above dosing parameters are then adjusted according to the relative potency of the analogue or derivative as compared to the parent compound (e.g., a compound twice as potent as doxorubicin is administered at half the above parameters, a compound half as potent as doxorubicin is administered at twice the above parameters, etc.).
  • the total dose of mitoxantrone applied should not exceed 5 mg (range of 0.01 ⁇ g to 5 mg). In a particularly preferred embodiment, the total amount of drug applied should be in the range of- 0.-1 ⁇ g to 1 mg.
  • the dose per unit area i.e., the amount of drug as a function of the surface area of the portion of the implant to which drug is applied and/or incorporated should fall within the range of 0.01 ⁇ g - 20 ⁇ g per mm 2 of surface area.
  • mitoxantrone should be applied to the implant surface at a dose of 0.05 ⁇ g/mm 2 - 3 ⁇ g/mm 2 .
  • the above dosing parameters should be utilized in combination with the release rate of the drug from the implant surface such that a minimum concentration of 10 "5 - 10 "6 M of mitoxantrone is maintained. It is necessary to insure that drug concentrations on the implant surface exceed concentrations of mitoxantrone known to be lethal to multiple species of bacteria and fungi (i.e., are in excess of 10 "5 M; although for some embodiments lower drug levels will be sufficient).
  • mitoxantrone is released from the surface of the implant such that anti-infective activity is maintained for a period ranging from several hours to several months.
  • the drug is released in effective concentrations for a period ranging from 1 week - 6 months.
  • analogues and derivatives of mitoxantrone (as described previously) with similar functional activity can be utilized for the purposes of this invention; the above dosing parameters are then adjusted according to the relative potency of the analogue or derivative as compared to the parent compound (e.g., a compound twice as potent as mitoxantrone is administered at half the above parameters, a compound half as potent as mitoxantrone is administered at twice the above parameters, etc.).
  • the total dose of 5-fluorouracil applied should not exceed 250 mg (range of 1.0 ⁇ g to 250 mg). In a particularly preferred-embodiment, the total amount of drug applied should be in the range of 10 ⁇ g to 25 mg.
  • the dose per unit area i.e., the amount of drug as a function of the surface area of the portion of the implant to which drug is applied and/or incorporated
  • 5- fluorouracil should be applied to the implant surface at a dose of 1.0 ⁇ g/mm 2 - 50 ⁇ g/mm 2 .
  • the above dosing parameters should be utilized in combination with the release rate of the drug from the implant surface such that a minimum concentration of 10 "4 - 10 "7 M of 5-fluorouracil is maintained. It is necessary to insure that surface drug concentrations exceed concentrations of 5-fluorouracil known to be lethal to numerous species of bacteria and fungi (Ae., are in excess of 10 "4 M; although for some embodiments lower drug levels will be sufficient).
  • 5-fluorouracil is released from the implant surface such that anti-infective activity is maintained for a period ranging from several hours to several months.
  • the drug is released in effective concentrations for a period ranging from 1 week - 6 months.
  • analogues and derivatives of 5-fluorouracil (as described previously) with similar functional activity can be utilized for the purposes of this invention; the above dosing parameters are then adjusted according to the relative potency of the analogue or derivative as compared to the parent compound (e.g., a compound twice as potent as 5-fluorouracil is administered at half the above parameters, a compound half as potent as 5-fluorouracil is administered at twice the above parameters, etc.).
  • the total dose of etoposide applied should not exceed 25 mg (range of 0.1 ⁇ g to 25 mg). In a particularly preferred embodiment, the total amount of drug applied should be in the range of 1 ⁇ g to 5 mg.
  • the dose per unit area i.e., the amount of drug as a function of the surface area of the portion of the implant to which drug is applied and/or incorporated should fall within the range of 0.01 ⁇ g - 100 ⁇ g per mm 2 of surface area.
  • etoposide should be applied to the implant surface at a dose of 0.1 ⁇ g/mm 2 - 10 ⁇ g/mm 2 .
  • the above dosing parameters should be utilized in combination with the release rate of the drug from the implant surface such that a concentration of 10 "5 - 10 ⁇ 6 M of etoposide is maintained. It is necessary to insure that surface drug concentrations exceed concentrations of etoposide known to be lethal to a variety of bacteria and fungi (i.e., are in excess of 10 "5 M; although for some embodiments lower drug levels will be sufficient).
  • etoposide is released from the surface of the implant such that anti-infective activity is maintained for a period ranging from several hours to several months.
  • the drug is released in effective concentrations for a period ranging from 1 week - 6 months.
  • analogues and derivatives of etoposide (as described previously) with similar functional activity can be utilized for the purposes of this invention; the above dosing parameters are then adjusted according to the relative potency of the analogue or derivative as compared to the parent compound (e.g., a compound twice as potent as etoposide is administered at half the above parameters, a compound half as potent as etoposide is administered at twice the above parameters, etc.).
  • anthracyclines e.g., doxorubicin or mitoxantrone
  • fluoropyrimidines e.g., 5-fluorouracil
  • folic acid antagonists e.g., methotrexate
  • podophylotoxins e.g., etoposide
  • quinolones can be utilized to enhance the antibacterial activity of the composition.
  • the therapeutic agents useful in the present invention may also include various polymers. Such polymers may be used alone to be effective in certain applications (e.g., treating or preventing surgical adhesions) or in combination of an anti-fibrosis agent and/or an anti- infective agent to facilitate the delivery of, or to provide a sustained release formulation of, the anti-fibrosis agent and/or the anti-infective agent. Detailed descriptions of exemplary polymers are provided below in the section regarding pharmaceutical compositions, especially in the section regarding sustained release formulations.
  • the present invention in another aspect, provides pharmaceutical compositions that comprise a fibrosis-inhibiting agent and/or anti-infective agent.
  • the pharmaceutical compositions further comprise a polymer, an additional therapeutic agent, a pharmaceutical excipient, and/or an agent that facilitates the delivery of the therapeutic agents or compositions.
  • compositions of the present invention may comprise a polymer that itself is a therapeutic agent.
  • the compositions of the present invention may comprise a polymer that facilitates the delivery of a therapeutic agent or forms a sustained release formuation for a therapeutic agent.
  • compositions that comprise polymers may further comprise additional agents (e.g., pharmaceutical exicipents, echogenic agents, etc.).
  • the composition may be or include a hydrophilic polymer gel that has anti-thrombogenic properties.
  • a composition can be in the form of a coating that can comprise a hydrophilic, biodegradable polymer that is physically removed from the surface of the device over time, thus reducing adhesion of platelets to the device surface.
  • the gel composition can include a polymer or a blend of polymers.
  • the anti-thrombotic composition can include a crosslinked gel formed from a combination of molecules (e.g., PEG) having two or more terminal electrophilic groups and two or more nucleophilic groups.
  • desired therapeutic agents may be admixed with, blended with, conjugated to, or, otherwise modified to contain a polymer composition (which may be either biodegradable or non- biodegradable) or a non-polymeric composition in order to release the therapeutic agent over a prolonged period of time.
  • a polymer composition which may be either biodegradable or non- biodegradable
  • a non-polymeric composition in order to release the therapeutic agent over a prolonged period of time.
  • localized delivery as well as localized sustained delivery of the fibrosis-inhibiting and/or anti-infective agent may be required.
  • a desired therapeutic agent may be admixed with, blended with, conjugated to, or, otherwise modified to contain a polymeric composition (which may be either biodegradable or nonbiodegradable) or non-polymeric composition in order to release the therapeutic agent over a period of time.
  • a polymeric composition which may be either biodegradable or nonbiodegradable
  • non-polymeric composition in order to release the therapeutic agent over a period of time.
  • biodegradable polymers suitable for the delivery of the aforementioned therapeutic agents include albumin, collagen, gelatin, hyaluronic acid, starch, cellulose and cellulose derivatives (e.g., regenerated cellulose, methylcellulose, hydroxypropylcellulose, hydroxypropylmethylcellulose, carboxymethylcellulose, cellulose acetate phthalate, cellulose acetate succinate, hydroxypropylmethylcellulose phthalate), casein, dextrans, polysaccharides, fibrinogen, poly(ether ester) multiblock copolymers, based on poly(ethylene glycol) and poly ⁇ utylene terephthalate), tyrosine-derived polycarbonates (e.g., U.S. Patent No.
  • X is
  • non-degradable polymers suitable for the delivery of fibrosis-inhibiting agents include poly(ethylene-co-vinyl acetate) ("EVA") copolymers, non-degradable polyesters, such as poly(ethylene terephthalate), silicone rubber, acrylic polymers (polyacrylate, polyacrylic acid, polymethylacrylic acid, polymethylmethacrylate, poly(butyl methacrylate)), poly(alkylcynoacrylate) (e.g., poly(ethylcyanoacrylate), poly(butylcyanoacrylate) poly(hexylcyanoacrylate) poly(octylcyanoacrylate)), -acrylic resin, polyethylene, polypropylene, polyamides (nylon 6,6), polyurethanes (e.g., CHRONOFLEX AR, CHRONOFLEX AL, BIONATE, and PELLETHANE), poly(ester urethanes), poly(ether urethanes), poly(ester-(est
  • Polymers may also be developed which are either anionic (e.g., alginate, carrageenan, carboxymethyl cellulose, poly(acrylamido-2-methyl propane sulfonic acid) and copolymers thereof, poly(methacrylic acid and copolymers thereof and poly(acrylic acid) and copolymers thereof, as well as blends thereof, or cationic (e.g., chitosan, poly-L-lysine, polyethylenimine, and poly(allyl amine)) and blends, copolymers and branched polymers thereof (see generally, Dunn et al., J. Applied Polymer ScL 50:353-365, 1993; Cascone et al., J.
  • anionic e.g., alginate, carrageenan, carboxymethyl cellulose, poly(acrylamido-2-methyl propane sulfonic acid) and copolymers thereof, poly(methacrylic acid and copolymers thereof and poly(acrylic acid
  • preferred polymeric carriers include poly(ethylene-co-vinyl acetate), polyurethanes (e.g., CHRONOFLEX AR, CHRONOFLEX AL, BIONATE, and PELLETHANE), poly (D,L-lactic acid) oligomers and polymers, poly (L-lactic acid) oligomers and polymers, poly (glycolic acid), copolymers of lactic acid and glycolic acid, poly (caprolactone), poly (valerolactone), polyanhydrides, copolymers of poly (caprolactone) or poly (lactic acid) with a polyethylene glycol (e.g., MePEG), poly(alkylene oxide)-poly(ester) block copolymers (e.g., X-Y, X-Y-X or Y-X- Y, R-(Y-X) n , R-(X-Y) n where X is a polyalkylene oxide and Y is a polyester (e.g., CH
  • polysaccharides such as hyaluronic acid, chitosan and fucans, and copolymers of polysaccharides with degradable polymers, as well as blends thereof.
  • Other representative polymers capable of sustained localized delivery of anti-infective and/or fibrosis-inhibiting therapeutic agents include carboxylic polymers, polyacetates, polycarbonates, polyethers, polyethylenes, polyvinylbutyrals, polysilanes, polyureas, polyoxides, polystyrenes, polysulfides, polysulfones, polysulfonides, polyvinylhalides, pyrrolidones, rubbers, thermal-setting polymers, cross-linkable acrylic and methacrylic polymers, ethylene acrylic acid copolymers, styrene acrylic copolymers, vinyl acetate polymers and copolymers, vinyl acetal polymers and copolymers, epoxies, melamines, other amino resins, phenolic polymers, and copolymers thereof, water-insoluble cellulose ester polymers (including cellulose acetate propionate, cellulose acetate, cellulose acetate butyrate,
  • polymers as described herein can also be blended or copolymerized in various compositions as required to deliver therapeutic doses of fibrosis-inhibiting agents.
  • polymers as described herein can also be blended or copolymerized in various compositions as required to deliver therapeutic doses of biologically active agents (such as anit-infective agents).
  • biologically active agents such as anit-infective agents
  • Polymeric carriers for anti-infective and/or fibrosis-inhibiting therapeutic agents can be fashioned in a variety of forms, with desired release characteristics and/or with specific properties depending upon the composition being utilized.
  • polymeric carriers may be fashioned to release a therapeutic agent upon exposure to a specific triggering event such as pH (see, e.g., Heller et al., "Chemically Self- Regulated Drug Delivery Systems," in Polymers in Medicine III, Elsevier Science Publishers B.V., Amsterdam, 1988, pp. 175-188; Kang et al., J. Applied Polymer ScL 48:343-354, 1993; Dong et al., J.
  • pH-sensitive polymers include poly (acrylic acid) and its derivatives (including for example, homopolymers such as poly(aminocarboxylic acid); poly(acrylic acid); poly(methyl acrylic acid), copolymers of such homopolymers, and copolymers of poly(acrylic acid) and/or acrylate or acrylamide lmonomers such as those discussed above.
  • pH sensitive polymers include polysaccharides such as cellulose acetate phthalate; hydroxypropylmethylcellulose phthalate; hydroxypropylmethylcellulose acetate succinate; cellulose acetate trimellilate; and chitosan.
  • pH sensitive polymers include any mixture of a pH sensitive polymer and a water-soluble polymer.
  • ant-infective and/or fibrosis-inhibiting therapeutic agents can be delivered via polymeric carriers which are temperature sensitive (see, e.g., Chen et al., "Novel Hydrogels of a Temperature- Sensitive PLURONIC Grafted to a Bioadhesive Polyacrylic Acid Backbone for Vaginal Drug Delivery," in Proceed. Intern. Symp. Control. ReI. Bioact. Mater. 22:167-168, Controlled Release Society, Inc., 1995; Okano, "Molecular Design of Stimuli-Responsive Hydrogels for Temporal Controlled Drug Delivery," in Proceed. Intern. Symp. Control. ReI. Bioact. Mater.
  • thermogelling polymers and the gelatin temperature (LCST ( 0 C)
  • thermogelling polymers may be made by preparing copolymers between (among) monomers of the above, or by combining such homopolymers with other water-soluble polymers such as acrylmonomers (e.g., acrylic acid and derivatives thereof, such as methylacrylic acid, acrylate monomers and derivatives thereof, such as butyl methacrylate, butyl acrylate, lauryl acrylate, and acrylamide monomers and derivatives thereof, such as N-butyl acrylamide and acrylamide).
  • acrylmonomers e.g., acrylic acid and derivatives thereof, such as methylacrylic acid, acrylate monomers and derivatives thereof, such as butyl methacrylate, butyl acrylate, lauryl acrylate, and acrylamide monomers and derivatives thereof, such as N-butyl acrylamide and acrylamide.
  • thermogelling polymers include cellulose ether derivatives such as hydroxypropyl cellulose, 41 0 C; methyl cellulose, 55°C; hydroxypropylmethyl cellulose, 66°C; and ethylhydroxyethyl cellulose, polyalkylene oxide-polyester block copolymers of the structure X-Y, Y-X-Y and X-Y-X where X in a polyalkylene oxide and Y is a biodegradable polyester (e.g., PLG-PEG-PLG) and PLURONICs such as F-127, 10 - 15°C; L-122, 19 0 C; L-92, 26 0 C; L-81 , 20 0 C; and L-61 , 24°C.
  • PLG-PEG-PLG biodegradable polyester
  • PLURONICs such as F-127, 10 - 15°C; L-122, 19 0 C; L-92, 26 0 C; L-81 , 20 0
  • patents relating to thermally gelling polymers and the preparation include U.S. Patent Nos. 6,451,346; 6,201 ,072; 6,117,949; 6,004,573; 5,702,717; and 5,484,610; and PCT Publication Nos. WO 99/07343; WO 99/18142; WO 03/17972; WQ . . 01/82970; WO 00/18821 ; WO 97/15287; WO 01/41735; WO 00/00222 and WO 00/38651.
  • Anti-infective and/or fibrosis-inhibiting therapeutic agents may be linked by occlusion in the polymer matrix, dissolution in the polymer, bound by covalent linkages, bound by ionic interactions, or encapsulated in microcapsules.
  • therapeutic compositions are provided in non-capsular formulations such as microspheres (ranging from nanometers to micrometers in size), pastes, threads of various size, films, or sprays.
  • the anti-scarring agent may be incorporated into biodegradable magnetic nanospheres.
  • the nanospheres may be used, for example, to replenish an anti-scarring agent into an implanted intravascular device, such as a stent containing a weak magnetic alloy (see, e.g., Z. Forbes, B. B. Yellen, G. Friedman, K. Barbee. "An approach to targeted drug delivery based on uniform magnetic fields," IEEE Trans. Magn. 39(5): 3372-3377 (2003)).
  • an implanted intravascular device such as a stent containing a weak magnetic alloy
  • therapeutic compositions of anti-infective and/or fibrosis-inhibiting agents may be fashioned in the form of microspheres, microparticles and/or nanoparticles having any size ranging from 50 nm to 500 ⁇ m, depending upon the particular use. These compositions can be.
  • compositions can be formed by spray-drying methods, milling methods, coacervation methods, W/O emulsion methods, W/O/W emulsion methods, and solvent evaporation methods.
  • these compositions can include microemulsions, emulsions, liposomes and micelles.
  • such compositions may also be readily applied as a "spray", which solidifies into a film or coating for use as a device/implant surface coating or to line the tissues of the implantation site.
  • Such sprays may be prepared from microspheres of a wide array of sizes, including for example, from 0.1 ⁇ m to 3 ⁇ m, from 10 ⁇ m to 30 ⁇ m, and from 30 ⁇ m to 100 ⁇ m.
  • compositions that include anti-infective and/or anti-fibrosis agents may also be prepared in a variety of "paste" or gel forms.
  • therapeutic compositions are provided which are liquid at one temperature ⁇ e.g., temperature greater than 37°C, such as 40 0 C, 45°C, 5O 0 C, 55 0 C or 60 0 C), and solid or semi-solid at another temperature (e.g., ambient body temperature, or any temperature lower than 37°C).
  • temperature e.g., temperature greater than 37°C, such as 40 0 C, 45°C, 5O 0 C, 55 0 C or 60 0 C
  • solid or semi-solid e.g., ambient body temperature, or any temperature lower than 37°C.
  • Such "thermopastes” may be readily made utilizing a variety of techniques (see, e.g., PCT Publication WO 98/24427).
  • pastes may be applied as a liquid, which solidify in vivo due to dissolution of a water-soluble component of the paste and precipitation of encapsulated drug into the aqueous body environment.
  • These "pastes” and “gels” containing therapeutic agents are particularly useful for application to the surface of tissues that will be in contact with the implant or device.
  • the therapeutic compositions of the present invention may be formed as a film or tube.
  • These films or tubes can be porous or non-porous.
  • films or tubes are generally less than 5, 4, 3, 2, or 1 mm thick, more preferably less than 0.75 mm, 0.5 mm, 0.25 mm, or, 0.10 mm thick.
  • Films or tubes can also be generated of thicknesses less than 50 ⁇ m, 25 ⁇ m or 10 ⁇ m.
  • Such films are preferably flexible with a good tensile strength (e.g., greater than 50, preferably greater than 100, and more preferably greater than 150 or 200 N/cm 2 ), good adhesive properties (Ae., adheres to moist or wet surfaces), and have controlled permeability.
  • Fibrosis-inhibiting agents contained in polymeric films are particularly useful for application to the surface of a device or implant as well as to the surface of tissue, cavity or an organ.
  • polymeric carriers are provided which are adapted to contain and release a hydrophobic ant-infective and/or fibrosis-inhibiting compound, and/or the carrier containing the hydrophobic compound in combination with a carbohydrate, protein or polypeptide.
  • the polymeric carrier contains or comprises regions, pockets, or granules of one or more hydrophobic compounds.
  • hydrophobic compounds may be incorporated within a matrix which contains the hydrophobic therapeutic compound, followed by incorporation of the matrix within the polymeric carrier.
  • matrices can be utilized in this regard, including for example, carbohydrates and polysaccharides such as starch, cellulose, dextran, methylcellulose, sodium alginate, heparin, chitosan and hyaluronic acid, proteins or polypeptides such as albumin, collagen and gelatin.
  • hydrophobic compounds may be contained within a hydrophobic core, and this core contained within a hydrophilic shell.
  • the anti-infective and/or fibrosis-inhibiting therapeutic agent may be delivered as a solution.
  • the therapeutic agent can be incorporated directly into the solution to provide a homogeneous solution or dispersion.
  • the solution is an aqueous solution.
  • the aqueous solution may further include buffer salts, as well as viscosity modifying agents (e.g., hyaluronic acid, alginates, carboxymethylcellulose (CMC), and the like).
  • the solution can include a biocompatible solvent or liquid oligomers and/or polymers, such as ethanol, DMSO, glycerol, PEG-200, PEG-300 or NMP.
  • compositions may further comprise a polymer such a degradable polyester, where the polyester may comprise the residues of one or more of the monomers selected from lactide, lactic acid, glycolide, glycolic acid, e-caprolactone, gamma-caprolactone, hydroxy valeric acid, hydroxybutyric acid, beta- butyrolactone, gamma-butyrolactone, gamma-valerolactone, y- decanolactone, ⁇ -decanolactone, trimethylene carbonate, 1 ,4-dioxane-2- one or 1 ,5-dioxepan-2one, or block copolymers of the form X-Y, Y-X-Y, R- (Y-X) n , R-(X-Y) n and X-Y-X (where X in a polyalkylene oxide (e.g., poly(ethylene glycol, poly(propylene glycol) and block copolymers of poly(
  • the therapeutic anti- infective and/or fibrosis-inhibiting agent can further comprise a secondary carrier.
  • the secondary carrier can be in the form of microspheres (e.g., PLGA, PLLA, PDLLA, PCL, gelatin, polydioxanone, poly(alkylcyanoacrylate)), nanospheres (PLGA, PLLA, PDLLA, PCL, gelatin, polydioxanone, poly(alkylcyanoacrylate)), liposomes, emulsions, microemulsions, micelles (SDS, block copolymers of the form X-Y, Y-X-Y, R-(Y-X) n , R-(X-Y) n and X-Y-X (where X in a polyalkylene oxide (e.g., poly(ethylene glycol, poly ⁇ ropylene glycol) and block copolymers of poly(ethylene oxide) and poly(propylene oxide) (e.g., X
  • hydroxypropyl cyclodextrin (Cserhati and Hollo, Int. J. Pharm. 108:69-75, 1994), liposomes (see, e.g., Sharma et al., Cancer Res. 53:5877-5881, 1993; Sharma and Straubinger, Pharm. Res. 11 (60):889- 896, 1994; WO 93/18751; U.S. Patent No. 5,242,073), liposome/gel (WO 94/26254), nanocapsules (Bartoli et a!., J.
  • polymeric carriers can be materials that are formed in situ.
  • the precursors can be monomers or macromers that contain unsaturated groups that can be polymerized and/or cross-linked.
  • the monomers or macromers can then, for example, be injected into the treatment area or onto the surface of the treatment area and polymerized in situ using a radiation source (e.g., visible or UV light) or a free radical system (e.g., potassium persulfate and ascorbic acid or iron and hydrogen peroxide).
  • a radiation source e.g., visible or UV light
  • a free radical system e.g., potassium persulfate and ascorbic acid or iron and hydrogen peroxide
  • compositions that undergo free radical polymerization reactions are described in WO 01/44307, WO 01/68720, WO 02/072166, WO 03/043552, WO 93/17669, WO 00/64977; U.S. Patent Nos. 5,900,245, 6,051 ,248, 6,083,524, 6,177,095, 6,201 ,065, 6,217,894, 6,639,014, 6,352,710, 6,410,645, 6,531 ,147, 5,567,435, 5,986,043, 6,602,975; U.S. Patent Application Publication Nos. 2002/012796A1, 2002/0127266A1 , 2002/0151650A1, 2003/0104032A1 , 2002/0091229A1 , and 2003/0059906A1.
  • compositions that can be administered as liquids, but subsequently form hydrogels at the site of administration.
  • Such in situ hydrogel forming compositions can be administered as liquids from a variety of different devices, and are more adaptable for administration to any site, since they are not preformed.
  • Examples of in situ forming hydrogels include photoactivatable mixtures of water-soluble co-polyester prepolymers and polyethylene glycol to create hydrogel barriers.
  • Block copolymers of polyalkylene oxide polymers e.g., PLURONIC compounds from BASF Corporation, Mount Olive, NJ
  • poloxamers have been designed that are soluble in cold water, but form insoluble hydrogels that adhere to tissues at body temperature (Leach, et al., Am. J. Obstet. Gynecol. 162:1317-1319 (1990)).
  • the present invention provides for polymeric crosslinked matrices, and polymeric carriers, that may be used to assist in the prevention of the formation or growth of fibrous connective tissue.
  • the composition may contain and deliver fibrosis-inhibiting agents in the vicinity of the implanted device.
  • the following compositions are particularly useful when it is desired to infiltrate around the device, with or without a fibrosis-inhibiting agent.
  • Such polymeric materials may be prepared from, e.g., (a) synthetic materials, (b) naturally-occurring materials, or (c) mixtures of synthetic and naturally occurring materials.
  • the matrix may be prepared from, e.g., (a) a one-component, i.e., self-reactive, compound, or (b) two or more compounds that are reactive with one another.
  • these materials are fluid prior to delivery, and thus can be sprayed or otherwise extruded from a delivery device (e.g., a syringe) in order to deliver the composition.
  • a delivery device e.g., a syringe
  • the component materials react with each other, and/or with the body, to provide the desired affect.
  • materials that are reactive with one another must be kept separated prior to delivery to the patient, and are mixed together just prior to being delivered to the patient, in order that they maintain a fluid form prior to delivery.
  • the components of the matrix are delivered in a liquid state to the desired site in the body, whereupon in situ polymerization occurs.
  • crosslinked polymer compositions are prepared by reacting a first synthetic polymer containing two or more nucleophilic groups with a second synthetic polymer containing two or more electrophilic groups, where the electrophilic groups are capable of covalently binding with the nucleophilic groups.
  • the first and second polymers are each non-immunogenic.
  • the matrices are not susceptible to enzymatic cleavage by, e.g., a matrix metalloproteinase (e.g., collagenase) and are therefore expected to have greater long-term persistence in vivo than collagen-based compositions.
  • polymer refers inter alia to polyalkyls, polyamino acids, polyalkyleneoxides and polysaccharides. Additionally, for external or oral use, the polymer may be polyacrylic acid or carbopol.
  • synthetic polymer refers to polymers that are not naturally occurring and that are produced via chemical synthesis. As such, naturally occurring proteins such as collagen and naturally occurring polysaccharides such as hyaluronic acid are specifically excluded. Synthetic collagen, and synthetic hyaluronic acid, and their derivatives, are included.
  • Multifunctionally activated synthetic polymers Synthetic polymers containing either nucleophilic or electrophilic groups are also referred to herein as "multifunctionally activated synthetic polymers.”
  • multifunctionally activated refers to synthetic polymers which have, or have-been chemically modified to have, two or more nucleophilic or electrophilic groups which are capable of reacting with one another (i.e., the nucleophilic groups react with the electrophilic groups) to form covalent bonds.
  • Types of multifunctionally activated synthetic polymers include difunctionally activated, tetrafunctionally activated, and star-branched polymers.
  • Multifunctionally activated synthetic polymers for use in the present invention must contain at least two, more preferably, at least three, functional groups in order to form a three-dimensional crosslinked network with synthetic polymers containing multiple nucleophilic groups (i.e., "multi- nucleophilic polymers"). In other words, they must be at least difunctionally activated, and are more preferably trifunctionally or tetrafunctionally activated. If the first synthetic polymer is a difunctionally activated synthetic polymer, the second synthetic polymer must contain three or more functional groups in order to obtain a three-dimensional crosslinked network. Most preferably, both the first and the second synthetic polymer contain at least three functional groups.
  • Multi-nucleophilic polymers Synthetic polymers containing multiple nucleophilic groups are also referred to generically herein as "multi-nucleophilic polymers.”
  • multi-nucleophilic polymers must contain at least two, more preferably, at least three, nucleophilic groups. If a synthetic polymer containing only two nucleophilic groups is used, a synthetic polymer containing three or more electrophilic groups must be used in order to obtain a three-dimensional crosslinked network.
  • Preferred multi-nucleophilic polymers for use in the compositions and methods of the present invention include synthetic polymers that contain, or have been modified to contain, multiple nucleophilic groups such as primary amino groups and thiol groups.
  • Preferred multi-nucleophilic polymers include: (i) synthetic polypeptides that have been synthesized to contain two or more primary amino groups or thiol groups; and (ii)_polyethylene glycols that have been-modified to contain two or more primary amino groups or thiol groups.
  • reaction of a thiol group with an electrophilic group tends to proceed more slowly than reaction of a primary amino group with an electrophilic group.
  • the multi-nucleophilic polypeptide is a synthetic polypeptide that has been synthesized to incorporate amino acid residues containing primary amino groups (such as lysine) and/or amino acids containing thiol groups (such as cysteine).
  • Poly(lysine) a synthetically produced polymer of the amino acid lysine (145 MW), is particularly preferred.
  • Poly(lysine)s have been prepared having anywhere from 6 to about 4,000 primary amino groups, corresponding to molecular weights of about 870 to about 580,000.
  • Poly(lysine)s for use in the present invention preferably have a molecular weight within the range of about 1 ,000 to about 300,000; more preferably, within the range of about 5,000 to about 100,000; most preferably, within the range of about 8,000 to about 15,000.
  • Poly(lysine)s of varying molecular weights are commercially available from Peninsula Laboratories, Inc. (Belmont, Calif.) and Aldrich Chemical (Milwaukee, Wl).
  • Polyethylene glycol can be chemically modified to contain multiple primary amino or thiol groups according to methods set forth, for example, in Chapter 22 of Poly(ethylene Glycol) Chemistry: Biotechnical and Biomedical Applications, J. Milton Harris, ed., Plenum Press, N.Y. (1992). Polyethylene glycols which have been modified to contain two or more primary amino groups are referred to herein as "multi-amino PEGs.” Polyethylene glycols which have been modified to contain two or more thiol groups are referred to herein as "multi-thiol PEGs.” As used herein, the term "polyethylene glycol(s)" includes modified and or derivatized polyethylene glycol(s).
  • Multi-amino PEGs useful in the present invention include Huntsman's Jeffamine diamines ("D” series) and triamines ("T” series), which contain two and three primary amino groups per molecule, respectively.
  • Polyamines such as ethylenediamine (H 2 N-CH 2 -CH 2 -NH2), tetramethylenediamine (H 2 N-(C H 2 ⁇ -NH 2 ), pentamethylenediamine (cadaverine) (H 2 N-(CH 2 ) S -NH 2 ), hexamethylenediamine (H 2 N-(CH 2 J 6 -NH 2 ), di(2-aminoethyl)amine (HN-(CH 2 -CH 2 -NH 2 ⁇ ), and tris(2-aminoethyl)amine (N-(CH 2 -CH 2 -NH 2 ) 3 ) may also be used as the synthetic polymer containing multiple nucleophillc groups.
  • ethylenediamine H 2 N-CH 2 -CH 2 -NH2
  • tetramethylenediamine H 2 N-(C H 2 ⁇ -NH 2
  • pentamethylenediamine cadaverine
  • Multi-electrophilic polymers Synthetic polymers containing multiple electrophilic groups are also referred to herein as "multi-electrophilic polymers.”
  • the multifunctionally activated synthetic polymers must contain at least two, more preferably, at least three, electrophilic groups in order to form a three-dimensional crosslinked network with multi- nucleophilic polymers.
  • Preferred multi-electrophilic polymers for use in the compositions of the invention are polymers which contain two or more succinimidyl groups capable of forming covalent bonds with nucleophilic groups on other molecules.
  • Succinimidyl groups are highly reactive with materials containing primary amino (NH 2 ) groups, such as multi-amino PEG, poly(lysine), or collagen.
  • Succinimidyl groups are slightly less reactive with materials containing thiol (SH) groups, such as multi-thiol PEG or synthetic polypeptides containing multiple cysteine residues.
  • succinimidyl groups As used herein, the term "containing two or more succinimidyl groups” is meant to encompass polymers which are preferably commercially available containing two or more succinimidyl groups, as well as those that must be chemically derivatized to contain two or more succinimidyl groups.
  • succinimidyl group is intended to encompass sulfosuccinimidyl groups and other such variations of the "generic" succinimidyl group. The presence of the sodium sulfite moiety on the sulfosuccinimidyl group serves to increase the solubility of the polymer.
  • Hydrophilic polymers and, in particular, various derivatized polyethylene glycols are preferred for use in the compositions of the present invention.
  • PEG refers to polymers having the repeating structure (OCH 2 -CHb) n - Structures for some specific, tetrafunctionally activated forms of PEG are shown in FIGS. 4 to 13 of U.S. Patent 5,874,500, incorporated herein by reference.
  • suitable PEGS include PEG succinimidyl propionate (SE-PEG), PEG succinimidyl succinamide (SSA-PEG), and PEG succinimidyl carbonate (SC-PEG).
  • the crosslinked matrix is formed in situ by reacting pentaerythritol poly(ethylene glycol)ether tetra-sulfhydryl] (4-armed thiol PEG) and pentaerythritol poly(ethylene glycol)ether tetra-succinimidyl glutarate] (4-armed NHS PEG) as reactive reagents. Structures for these reactants are shown in U.S. Patent 5,874,500.
  • Each of these materials has a core with a structure that may be seen by adding ethylene oxide-derived residues to each of the hydroxyl groups in pentaerythritol, and then derivatizing the terminal hydroxyl groups (derived from the ethylene oxide) to contain either thiol groups (so as to form 4-armed thiol PEG) or N- hydroxysuccinimydyl groups (so as to form 4-armed NHS PEG), optionally with a linker group present between the ethylene oxide derived backbone and the reactive functional group, where this product is commercially available as COSEAL from Angiotech Pharmaceuticals Inc.
  • a group "D" may be present in one or both of these molecules, as discussed in more detail below.
  • preferred activated polyethylene glycol derivatives for use in the invention contain succinimidyl groups as the reactive group.
  • different activating groups can be attached at sites along the length of the PEG molecule.
  • PEG can be derivatized to form functionally activated PEG propionaldehyde (A-PEG), or functionally activated PEG glycidyl ether (E-PEG), or functionally activated PEG-isocyanate (I-PEG), or functionally activated PEG-vinylsulfone (V- PEG).
  • Hydrophobic polymers can also be used to prepare the compositions of the present invention.
  • Hydrophobic polymers for use in the present invention preferably contain, or can be derivatized to contain, two or more electrophilic groups, such as succinimidyl groups, most preferably, two, three, or four electrophilic groups.
  • electrophilic groups such as succinimidyl groups, most preferably, two, three, or four electrophilic groups.
  • hydrophobic polymer refers to polymers which contain a relatively small proportion of oxygen or nitrogen atoms.
  • Hydrophobic polymers which already contain two or more succinimidyl groups include, without limitation, disuccinimidyl suberate (DSS), bis(sulfosuccinimidyl) suberate (BS3), dithiobis(succinimidylpropionate) (DSP), bis(2-succinimidooxycarbonyloxy) ethyl sulfone (BSOCOES), and 3,3'-dithiobis(sulfosuccinimidylpropionate (DTSPP), and their analogs and derivatives.
  • DSS disuccinimidyl suberate
  • BS3 bis(sulfosuccinimidyl) suberate
  • DSP dithiobis(succinimidylpropionate)
  • BSOCOES bis(2-succinimidooxycarbonyloxy) ethyl sulfone
  • DTSPP 3,3'-dithiobis(sulfosucc
  • Preferred hydrophobic polymers for use in the invention generally have a carbon chain that is no longer than about 14 carbons.
  • Polymers having carbon chains substantially longer than 14 carbons generally have very poor solubility in aqueous solutions and, as such, have very long reaction times when mixed with aqueous solutions of synthetic polymers containing multiple nucleophilic groups.
  • polyacids can be derivatized to contain two or more functional groups, such as succinimidyl groups.
  • Polyacids for use in the present invention include, without limitation, trimethylolpropane-based tricarboxylic acid, di(trimethylol propane)-based tetracarboxylic acid, heptanedioic acid, octanedioic acid (suberic acid), and hexadecanedioic acid (thapsic acid). Many of these polyacids are commercially available from DuPont Chemical Company (Wilmington, DE).
  • polyacids can be chemically derivatized to contain two or more succinimidyl groups by reaction with an appropriate molar amount of N-hydroxysuccinimide (NHS) in the presence of N 1 N 1 - dicyclohexylcarbodiimide (DCC).
  • NHS N-hydroxysuccinimide
  • DCC N 1 N 1 - dicyclohexylcarbodiimide
  • Polyalcohols such as trimethylolpropane and di(trimethylol propane) can be converted to carboxylic acid form using various methods, then further derivatized by reaction with NHS in the presence of DCC to produce trifunctionally and tetrafunctionally activated polymers, respectively, as described in U.S. application Ser. No. 08/403,358.
  • Polyacids such as heptanedioic acid (HOOC-(CH 2 ) 5 -COOH), octanedioic acid (HOOC-(CH 2 ) 6 - COOH), and hexadecanedioic acid (HOOC-(CH 2 )i 4 -COOH) are derivatized by the addition of succinimidyl groups to produce difunctionally activated polymers.
  • heptanedioic acid HOOC-(CH 2 ) 5 -COOH
  • octanedioic acid HOOC-(CH 2 ) 6 - COOH
  • hexadecanedioic acid HOOC-(CH 2 )i 4 -COOH
  • Polyamines such as ethylenediamine, tetramethylenediamine, pentamethylenediamine (cadaverine), hexamethylenediamine, bis (2- aminoethyl)amine, and tris(2-aminoethyl)amine can be chemically derivatized to polyacids, which can then be derivatized to contain two or more succinimidyl groups by reacting with the appropriate molar amounts of N-hydroxysuccinimide in the presence of DCC, as described in U.S. application Ser. No. 08/403,358. Many of these polyamines are commercially available from DuPont Chemical Company.
  • the first synthetic polymer will contain multiple nucleophilic groups (represented below as “X”) and it will react with the second synthetic polymer containing multiple electrophilic groups (represented below as “Y”), resulting in a covalently bound polymer network, as follows:
  • X and Y may be the same or different, i.e., a synthetic polymer may have two different electrophilic groups, or two different nucleophilic groups, such as with glutathione.
  • the backbone of at least one of the synthetic polymers comprises alkylene oxide residues, e.g., residues from ethylene oxide, propylene oxide, and mixtures thereof.
  • alkylene oxide residues e.g., residues from ethylene oxide, propylene oxide, and mixtures thereof.
  • 'backbone' refers to a significant portion of the polymer.
  • the synthetic polymer containing alkylene oxide residues may be described by the formula X-polymer-X or Y-polymer-Y, wherein X and Y are as defined above, and the term "polymer” represents - (CH 2 CH 2 O) n - or -(CH(CH 3 )CH 2 O) n - or -(CH 2 -CH 2 -O) n -(CH(CH 3 )CH 2 -O) n -. In these cases the synthetic polymer would be difunctional.
  • the required functional group X or Y is commonly coupled to the polymer backbone by a linking group (represented below as "Q"), many of which are known or possible.
  • Q a linking group
  • Exemplary Q groups include -O-(CH 2 ) n -; -S-(CH 2 ) n -; -NH- (CHz) n -; -O 2 C-NH-(CH 2 ) n -; -O 2 C-(CH 2 ) n -; -O 2 C-(CR 1 H) n -; and -0-R 2 -CO-NH-, which provide synthetic polymers of the partial structures: polymer-O- (CH 2 ) n -(X or Y); polymer-S-(CH 2 ) n -(X or Y); polymer-NH-(CH 2 ) n -(X or Y); polymer-O 2 C-NH-(CH 2 ) n -(X or Y); polymer-O 2 C-(CH 2 ) n -(X or Y); polymer-O 2 C-(CH 2 ) n -(X or Y); polymer-O 2 C-
  • n 1-10
  • R 1 H or alkyl (Ae., CH 3 , C 2 H 5 , etc.);
  • R 2 CH 2 , or CO-NH-CH 2 CH 2 ; and Q 1 and Q 2 may be the same or different.
  • D An additional group, represented below as "D" can be inserted between the polymer and the linking group, if present.
  • D group One purpose of such a D group is to affect the degradation rate of the crosslinked polymer composition in vivo, for example, to increase the degradation rate, or to decrease the degradation rate. This may be useful in many instances, for example, when drug has been incorporated into the matrix, and it is desired to increase or decrease polymer degradation rate so as to influence a drug delivery profile in the desired direction.
  • An illustration of a crosslinking reaction involving first and second synthetic polymers each having D and Q groups is shown below.
  • Some useful biodegradable groups "D” include polymers formed from one or more ⁇ -hydroxy acids, e.g., lactic acid, glycolic acid, and the cyclization products thereof (e.g., lactide, glycolide), ⁇ -caprolactone, and amino acids.
  • the polymers may be referred to as polylactide, polyglycolide, poly(co-lactide-glycolide); poly- ⁇ -caprolactone, polypeptide (also known as poly amino acid, for example, various di- or tri-peptides) and poly(anhydride)s.
  • a first synthetic polymer containing multiple nucleophilic groups is mixed with a second synthetic polymer containing multiple electrophilic groups. Formation of a three-dimensional crosslinked network occurs as a result of the reaction between the nucleophilic groups on the first synthetic polymer and the electrophilic groups on the second synthetic polymer.
  • the concentrations of the first synthetic polymer and the second synthetic polymer used to prepare the compositions of the present invention will vary depending upon a number of factors, including the types and molecular weights of the particular synthetic polymers used and the desired end use application.
  • it is preferably used at a concentration in the range of about 0.5 to about 20 percent by weight of the final composition, while the second synthetic polymer is used at a concentration in the range of about 0.5 to about 20 percent by weight of the final composition.
  • a final composition having a total weight of 1 gram (1000 milligrams) would contain between about 5 to about 200 milligrams of multi- amino PEG, and between about 5 to about 200 milligrams of the second synthetic polymer.
  • compositions intended for use in tissue augmentation will generally employ concentrations of first and second synthetic polymer that fall toward the higher end of the preferred concentration range.
  • Compositions intended for use as bioadhesives or in adhesion prevention do not need to be as firm and may therefore contain lower polymer concentrations.
  • the second synthetic polymer is generally stored and used in sterile, dry form to prevent the loss of crosslinking ability due to hydrolysis which typically occurs upon exposure of such electrophilic groups to aqueous media.
  • Processes for preparing synthetic hydrophilic polymers containing multiple electrophylic groups in sterile, dry form are set forth in U.S. Patent 5,643,464.
  • the dry synthetic polymer may be compression molded into a thin sheet or membrane, which can then be sterilized using gamma or, preferably, e-beam irradiation. The resulting dry membrane or sheet can be cut to the desired size or chopped into smaller size particulates.
  • polymers containing multiple nucleophilic groups are generally not water-reactive and can therefore be stored in aqueous solution.
  • one or both of the electrophilic- or nucleophilic-terminated polymers described above can be combined with a synthetic or naturally occurring polymer.
  • the presence of the synthetic or naturally occurring polymer may enhance the mechanical and/or adhesive properties of the in situ forming compositions.
  • Naturally occurring polymers, and polymers derived from naturally occurring polymer that may be included in in situ forming materials include naturally occurring proteins, such as collagen, collagen derivatives (such as methylated collagen), fibrinogen, thrombin, albumin, fibrin, and derivatives of and naturally occurring polysaccharides, such as glycosaminoglycans, including deacetylated and desulfated glycosaminoglycan derivatives.
  • a composition comprising naturally-occurring protein and both of the first and second synthetic polymer as described above is used to form the crosslinked matrix according to the present invention.
  • a composition comprising collagen and both of the first and second synthetic polymer as described above is used to form the crosslinked matrix according to the present invention.
  • a composition comprising methylated collagen and both of the first and second synthetic polymer as described above is used to form the crosslinked matrix according to the present invention.
  • a composition comprising fibrinogen and both of the first and second synthetic polymer as described above is used to form the crosslinked matrix according to the present invention.
  • a composition comprising thrombin and both of the first and second synthetic polymer as described above is used to form the crosslinked matrix according to the present invention.
  • a composition comprising albumin and both of the first and second synthetic polymer as described above is used to form the crosslinked matrix according to the present invention.
  • a composition comprising fibrin and both of the first and second synthetic polymer as described above is used to form the crosslinked matrix according to the present invention.
  • a composition comprising naturally occurring polysaccharide and both of the first and second synthetic polymer as described above is used to form the crosslinked matrix according to the present invention.
  • a composition comprising glycosaminoglycan and both of the first and second synthetic polymer as described above is used to form the crosslinked matrix according to the present invention.
  • a composition comprising deacetylated glycosaminoglycan and both of the first and second synthetic polymer as described above is used to form the crosslinked matrix according to the present invention.
  • a composition comprising desulfated glycosaminoglycan and both of the first and second synthetic polymer as described above is used to form the crosslinked matrix according to the present invention.
  • a composition comprising naturally-occurring protein and the first synthetic polymer as described above is used to form the crosslinked matrix according to the present invention.
  • a composition comprising collagen and the first synthetic polymer as described above is used to form the crosslinked matrix according to the present invention.
  • a composition comprising methylated collagen and the first synthetic polymer as described above is used to form the crosslinked matrix according to the present invention.
  • a composition comprising fibrinogen and the first synthetic polymer as described above is used to form the crosslinked matrix according to the present invention.
  • a composition comprising thrombin and the first synthetic polymer as described above is used to form the crosslinked matrix according to the present invention.
  • a composition comprising albumin and the first synthetic polymer as described above is used to form the crosslinked matrix according to the present invention.
  • a composition comprising fibrin and the first synthetic polymer as described above is used to form the crosslinked matrix according to the present invention.
  • a composition comprising naturally occurring polysaccharide and the first synthetic polymer as described above is used to form the crosslinked matrix according to the present invention.
  • a composition comprising glycosaminoglycan and the first synthetic polymer as described above is used to form the crosslinked matrix according to the present invention.
  • a composition comprising deacetylated glycosaminoglycan and the first synthetic polymer as described above is used to form the crosslinked matrix according to the present invention.
  • a composition comprising desulfated glycosaminoglycan and the first synthetic polymer as described above is used to form the crosslinked matrix according to the present invention.
  • a composition comprising naturally-occurring protein and the second synthetic polymer as described above is used to form the crosslinked matrix according to the present invention.
  • a composition comprising collagen and the second synthetic polymer as described above is used to form the crosslinked matrix according to the present invention.
  • a composition comprising methylated collagen and the second synthetic polymer as described above is used to form the crosslinked matrix according to the present invention.
  • a composition comprising fibrinogen and the second synthetic polymer as described above is used to form the crosslinked matrix according to the present invention.
  • a composition comprising thrombin and the second synthetic polymer as described above is used to form the crosslinked matrix according to the present invention.
  • a composition comprising albumin and the second synthetic polymer as described above is used to form the crosslinked matrix according to the present invention.
  • a composition comprising fibrin and the second synthetic polymer as described above is used to form the crosslinked matrix according to the present invention.
  • a composition comprising naturally occurring polysaccharide and the second synthetic polymer as described above is used to form the crosslinked matrix according to the present invention.
  • a composition comprising glycosaminoglycan and the second synthetic polymer as described above is used to form the crosslinked matrix according to the present invention.
  • a composition comprising deacetylated glycosaminoglycan and the second synthetic polymer as described above is used to form the crosslinked matrix according to the present invention.
  • a composition comprising desulfated glycosaminoglycan and the second synthetic polymer as described above is used to form the crosslinked matrix according to the present invention.
  • the presence of protein or polysaccharide components which contain functional groups that can react with the functional groups on multiple activated synthetic polymers can result in formation of a crosslinked synthetic polymer-naturally occurring polymer matrix upon mixing and/or crosslinking of the synthetic polymer(s).
  • the electrophilic groups on the second synthetic polymer will react with the primary amino groups on these components, as well as the nucleophilic groups on the first synthetic polymer, to cause these other components to become part of the polymer matrix.
  • lysine-rich proteins such as collagen may be especially reactive with electrophilic groups on synthetic polymers.
  • the naturally occurring protein is polymer may be collagen.
  • collagen or “collagen material” refers to all forms of collagen, including those which have been processed or otherwise modified and is intended to encompass collagen of any type, from any source, including, but not limited to, collagen extracted from tissue or produced recombinantly, collagen analogues, collagen derivatives, modified collagens, and denatured collagens, such as gelatin.
  • collagen from any source may be included in the compositions of the invention; for example, collagen may be extracted and purified from human or other mammalian source, such as bovine or porcine corium and human placenta, or may be recombinantly or otherwise produced.
  • human or other mammalian source such as bovine or porcine corium and human placenta
  • the preparation of purified, substantially non-antigenic collagen in solution from bovine skin is well known in the art.
  • U.S. Patent No. 5,428,022 discloses methods of extracting and purifying collagen from the human placenta.
  • U.S. Patent No. 5,667,839 discloses methods of producing recombinant human collagen in the milk of transgenic animals, including transgenic cows.
  • Collagen of any type including, but not limited to, types I, II, III, IV, or any combination thereof, may be used in the compositions of the invention, although type I is generally preferred.
  • Either atelopeptide or telopeptide-containing collagen may be used; however, when collagen from a xenogeneic source, such as bovine collagen, is used, atelopeptide collagen is generally preferred, because of its reduced immunogenicity compared to telopeptide-containing collagen.
  • Collagen that has not been previously crosslinked by methods such as heat, irradiation, or chemical crosslinking agents is preferred for use in the compositions of the invention, although previously crosslinked collagen may be used.
  • Non-crosslinked atelopeptide fibrillar collagen is commercially available from lnamed Aesthetics (Santa Barbara, CA) at collagen concentrations of 35 mg/ml and 65 mg/ml under the trademarks ZYDERM I Collagen and ZYDERM Il Collagen, respectively.
  • Glutaraldehyde crosslinked atelopeptide fibrillar collagen is commercially available from lnamed Corporation (Santa Barbara, CA) at a collagen concentration of 35 mg/ml under the trademark ZYPLAST Collagen.
  • Collagens for use in the present invention are generally in aqueous suspension at a concentration between about 20 mg/ml to about 120 mg/ml; preferably, between about 30 mg/ml to about 90 mg/ml.
  • nonfibrillar collagen may be preferred for use in compositions that are intended for use as bioadhesives.
  • nonfibrillar collagen refers to any modified or unmodified collagen material that is in substantially nonfibrillar form at pH 7, as indicated by optical clarity of an aqueous suspension of the collagen.
  • Collagen that is already in nonfibrillar form may be used in the compositions of the invention.
  • nonfibrillar collagen is intended to encompass collagen types that are nonfibrillar in native form, as well as collagens that have been chemically modified such that they are in nonfibrillar form at or around neutral pH.
  • Collagen types that are nonfibrillar (or microfibrillar) in native form include types IV, Vl, and VII.
  • Chemically modified collagens that are in nonfibrillar form at neutral pH include succinylated collagen and methylated collagen, both of which can be prepared according to the methods described in U.S. Pat. No. 4,164,559, issued Aug. 14, 1979, to Miyata et al., which is hereby incorporated by reference in its entirety. Due to its inherent tackiness, methylated collagen is particularly preferred for use in bioadhesive compositions, as disclosed in U.S. application Ser. No. 08/476,825.
  • Collagens for use in the crosslinked polymer compositions of the present invention may start out in fibrillar form, then be rendered nonfibrillar by the addition of one or more fiber disassembly agent.
  • the fiber disassembly agent must be present in an amount sufficient to render the collagen substantially nonfibrillar at pH 7, as described above.
  • Fiber disassembly agents for use in the present invention include, without limitation, various biocompatible alcohols, amino acids (e.g., arginine), inorganic salts (e.g., sodium chloride and potassium chloride), and carbohydrates (e.g., various sugars including sucrose).
  • the polymer may be collagen or a collagen derivative, for example methylated collagen.
  • An example of an in situ forming composition uses pentaerythritol poly(ethylene glycol)ether tetra- sulfhydryl] (4-armed thiol PEG), pentaerythritol poly(ethylene glycol)ether tetra-succinimidyl glutarate] (4-armed NHS PEG) and methylated collagen as the reactive reagents.
  • This composition when mixed with the appropriate buffers can produce a crosslinked hydrogel.
  • the naturally occurring polymer may be a glycosaminoglycan.
  • Glycosaminoglycans e.g., hyaluronic acid
  • glycosaminoglycan may be derivatized.
  • glycosaminoglycans can be chemically derivatized by, e.g., deacetylation, desulfation, or both in order to contain primary amino groups available for reaction with electrophilic groups on synthetic polymer molecules.
  • Glycosaminoglycans that can be derivatized according to either or both of the aforementioned methods include the following: hyaluronic acid, chondroitin sulfate A, chondroitin sulfate B (dermatan sulfate), chondroitin sulfate C, chitin (can be derivatized to chitosan), keratan sulfate, keratosulfate, and heparin.
  • Derivatization of glycosaminoglycans by deacetylation and/or desulfation and covalent binding of the resulting glycosaminoglycan derivatives with synthetic hydrophilic polymers is described in further detail in commonly assigned, allowed U.S. patent application Ser. No. 08/146,843, filed Nov. 3, 1993.
  • the collagen is added to the first synthetic polymer, then the collagen and first synthetic polymer are mixed thoroughly to achieve a homogeneous composition.
  • the second synthetic polymer is then added and mixed into the collagen/first synthetic polymer mixture, where it will covalently bind to primary amino groups or thiol groups on the first synthetic polymer and primary amino groups on the collagen, resulting in the formation of a homogeneous crosslinked network.
  • Various deacetylated and/or desulfated glycosaminoglycan derivatives can be incorporated into the composition in a similar manner as that described above for collagen.
  • hydrocolloids such as carboxymethylcellulose may promote tissue adhesion and/or swellability.
  • compositions of the present invention having two synthetic polymers may be administered before, during or after crosslinking of the first and second synthetic polymer.
  • Certain uses, which are discussed in greater detail below, such as tissue augmentation, may require the compositions to be crosslinked before administration, whereas other applications, such as tissue adhesion, require the compositions to be administered before crosslinking has reached "equilibrium.”
  • the point at which crosslinking has reached equilibrium is defined herein as the point at which the composition no longer feels tacky or sticky to the touch.
  • the first synthetic polymer and second synthetic polymer may be contained within separate barrels of a dual-compartment syringe.
  • the two synthetic polymers do not actually mix until the point at which the two polymers are extruded from the tip of the syringe needle into the patient's tissue.
  • This allows the vast majority of the crosslinking reaction to occur in situ, avoiding the problem of needle blockage which commonly occurs if the two synthetic polymers are mixed too early and crosslinking between the two components is already too advanced prior to delivery from the syringe needle.
  • the use of a dual-compartment syringe, as described above, allows for the use of smaller diameter needles, which is advantageous when performing soft tissue augmentation in delicate facial tissue, such as that surrounding the eyes.
  • first synthetic polymer and second synthetic polymer may be mixed according to the methods described above prior to delivery to the tissue site, then injected to the desired tissue site immediately (preferably, within about 60 seconds) following mixing.
  • the first synthetic polymer and second synthetic polymer are mixed, then extruded and allowed to crosslink into a sheet or other solid form.
  • the crosslinked solid is then dehydrated to remove substantially all unbound water.
  • the resulting dried solid may be ground or comminuted into particulates, then suspended in a nonaqueous fluid carrier, including, without limitation, hyaluronic acid, dextran sulfate, dextran, succinylated noncrosslinked collagen, methylated noncrosslinked collagen, glycogen, glycerol, dextrose, maltose, triglycerides of fatty acids (such as corn oil, soybean oil, and sesame oil), and egg yolk phospholipid.
  • a nonaqueous fluid carrier including, without limitation, hyaluronic acid, dextran sulfate, dextran, succinylated noncrosslinked collagen, methylated noncrosslinked collagen, glycogen, glycerol, dextrose, maltose, trigly
  • the suspension of particulates can be injected through a small-gauge needle to a tissue site. Once inside the tissue, the crosslinked polymer particulates will rehydrate and swell in size at least five-fold.
  • the first and/or second synthetic polymers may be combined with a hydrophilic polymer, e.g., collagen or methylated collagen, to form a composition useful in the present invention.
  • a hydrophilic polymer e.g., collagen or methylated collagen
  • the compositions useful in the present invention include a hydrophilic polymer in combination with two or more crosslinkable components. This embodiment is described in further detail in this section.
  • the Hydrophilic Polymer Component is a Hydrophilic Polymer Component
  • the hydrophilic polymer component may be a synthetic or naturally occurring hydrophilic polymer.
  • Naturally occurring hydrophilic polymers include, but are not limited to: proteins such as collagen and derivatives thereof, fibronectin, albumins, globulins, fibrinogen, and fibrin, with collagen particularly preferred; carboxylated polysaccharides such as -polymannuronic acid and polygalacturonic acid; aminated polysaccharides, particularly the glycosaminoglycans, e.g., hyaluronic acid, chitin, chondroitin sulfate A, B, or C, keratin sulfate, keratosulfate and heparin; and activated polysaccharides such as dextran and starch derivatives.
  • Collagen e.g., methylated collagen
  • glycosaminoglycans are preferred naturally occurring hydrophilic polymers for use herein.
  • collagen from any source may be used in the composition of the method; for example, collagen may be extracted and purified from human or other mammalian source, such as bovine or porcine corium and human placenta, or may be recombinantly or otherwise produced.
  • human or other mammalian source such as bovine or porcine corium and human placenta
  • the preparation of purified, substantially non-antigenic collagen in solution from bovine skin is well known in the art. See, e.g., U.S. Pat. No. 5,428,022, to Palefsky et al., which discloses methods of extracting and purifying collagen from the human placenta. See also U.S. Patent No. 5,667,839, to Berg, which discloses methods of producing recombinant human collagen in the milk of transgenic animals, including transgenic cows.
  • the term "collagen” or "collagen material” as used herein refers to all forms of collagen, including those that have been processed or otherwise modified.
  • Collagen of any type including, but not limited to, types I, II, III, IV, or any combination thereof, may be used in the compositions of the invention, although type I is generally preferred.
  • Either atelopeptide or telopeptide-containing collagen may be used; however, when collagen from a source, such as bovine collagen, is used, atelopeptide collagen is generally preferred, because of its reduced immunogenicity compared to telopeptide-containing collagen.
  • Collagen that has not been previously crosslinked by methods such as heat, irradiation, or chemical crosslinking agents is preferred for use in the compositions of the invention, although previously crosslinked collagen may be used.
  • Non-crosslinked atelopeptide fibrillar collagen is commercially available from McGhan Medical Corporation (Santa Barbara, Calif.) at collagen concentrations of 35 mg/ml and 65 mg/ml under the trademarks ZYDERM ® I Collagen and ZYDERM ® Il Collagen, respectively.
  • Glutaraldehyde-crosslinked atelopeptide fibrillar collagen is commercially available from McGhan Medical Corporation at a collagen concentration of 35 mg/ml under the trademark ZYPLAST ® .
  • Collagens for use in the present invention are generally, although not necessarily, in aqueous suspension at a concentration between about 20 mg/ml to about 120 mg/ml, preferably between about 30 mg/ml to about 90 mg/ml.
  • denatured collagen commonly known as gelatin
  • Gelatin may have the added benefit of being degradable faster than collagen.
  • nonfibrillar collagen refers to any modified or unmodified collagen material that is in substantially nonfibrillar form at pH 7, as indicated by optical clarity of an aqueous suspension of the collagen.
  • Collagen that is already in nonfibrillar form may be used in the compositions of the invention.
  • nonfibrillar collagen is intended to encompass collagen types that are nonfibrillar in native form, as well as collagens that have been chemically modified such that they are in nonfibrillar form at or around neutral pH.
  • Collagen types that are nonfibrillar (or microfibrillar) in native form include types IV, Vl, and VII.
  • Chemically modified collagens that are in nonfibrillar form at neutral pH include succinylated collagen, propylated collagen, ethylated collagen, methylated collagen, and the like, both of which can be prepared according to the methods described in U.S. Pat. No. 4,164,559, to Miyata et al., which is hereby incorporated by reference in its entirety. Due to its inherent tackiness, methylated collagen is particularly preferred, as disclosed in U.S. Patent No. 5,614,587 to Rhee et al. Gollagens-for use in the crosslinkable compositions of the present invention may start out in fibrillar form, then be rendered nonfibrillar by the addition of one or more fiber disassembly agents.
  • Fiber disassembly agent must be present in an amount sufficient to render the collagen substantially nonfibrillar at pH 7, as described above.
  • Fiber disassembly agents for use in the present invention include, without limitation, various biocompatible alcohols, amino acids, inorganic salts, and carbohydrates, with biocompatible alcohols being particularly preferred.
  • Preferred biocompatible alcohols include glycerol and propylene glycol.
  • Non-biocompatible alcohols such as ethanol, methanol, and isopropanol, are not preferred for use in the present invention, due to their potentially deleterious effects on the body of the patient receiving them.
  • Preferred amino acids include arginine.
  • Preferred inorganic salts include sodium chloride and potassium chloride.
  • carbohydrates such as various sugars including sucrose, may be used in the practice of the present invention, they are not as preferred as other types of fiber disassembly agents because they can have cytotoxic effects in vivo.
  • fibrillar collagen has less surface area and a lower concentration of reactive groups than nonfibrillar, fibrillar collagen is less preferred.
  • fibrillar collagen or mixtures of nonfibrillar and fibrillar collagen, may be preferred for use in compositions intended for long-term persistence in vivo, if optical clarity is not a requirement.
  • Synthetic hydrophilic polymers may also be used in the present invention.
  • Useful synthetic hydrophilic polymers include, but are not limited to: polyalkylene oxides, particularly polyethylene glycol and poly(ethylene oxide)-poly(propylene oxide) copolymers, including block and random copolymers; polyols such as glycerol, polyglycerol (particularly highly branched polyglycerol), propylene glycol and trimethylene glycol substituted with one or more polyalkylene oxides, e.g., mono-, di- and tri- pplyoxyethylated glycerol,, mono- and di-polyoxyethylated propylene glycol, and mono- and di-polyoxyethylated trimethylene glycol; polyoxyethylated sorbitol, polyoxyethylated glucose; acrylic acid polymers and analogs and copolymers thereof, such as polyacrylic acid perse, polymethacrylic acid, poly(hydroxyethy
  • compositions of the invention also comprise a plurality of crosslinkable components.
  • Each of the crosslinkable components participates in a reaction that results in a crosslinked matrix.
  • the crosslinkable components Prior to completion of the crosslinking reaction, the crosslinkable components provide the necessary adhesive qualities that enable the methods of the invention.
  • the crosslinkable components are selected so that crosslinking gives rise to a biocompatible, nonimmunogenic matrix useful in a variety of contexts including adhesion prevention, biologically active agent delivery, tissue augmentation, and other applications.
  • the crosslinkable components of the invention comprise: a component A, which has m nucleophilic groups, wherein m > 2 and a component B, which has n electrophilic groups capable of reaction with the m nucleophilic groups, wherein n > 2 and m + n > 4.
  • An optional third component, optional component C which has at least one functional group that is either electrophilic and capable of reaction with the nucleophilic groups of component A, or nucleophilic and capable of reaction with the electrophilic groups of component B may also be present.
  • the total number of functional groups present on components A, B and C, when present, in combination is > 5; that is, the total functional groups given by m + n + p must be > 5, where p is the number of functional groups on component C and, as indicated, is > 1.
  • Each of the components is biocompatible and nonimmunogenic, and at least one component is comprised of a hydrophilic polymer.
  • the composition may contain additional crosslinkable components D, E, F, etc., having one or more reactive nucleophilic or electrophilic groups and thereby participate in formation of the crosslinked biomaterial via covalent bonding to other components.
  • the m nucleophilic groups on component A may all be the same, or, alternatively, A may contain two or more different nucleophilic groups.
  • the n electrophilic groups on component B may all be the same, or two or more different electrophilic groups may be present.
  • the functional group(s) on optional component C if nucleophilic, may or may not be the same as the nucleophilic groups on component A, and, conversely, if electrophilic, the functional group(s) on optional component C may or may not be the same as the electrophilic groups on component B.
  • R 1 , R 2 and R 3 are independently selected from the group consisting of C 2 to C 14 hydrocarbyl, heteroatom-containing C 2 to Ci 4 hydrocarbyl, hydrophilic polymers, and hydrophobic polymers, providing that at least one of R 1 , R 2 and R 3 is a hydrophilic polymer, preferably a synthetic hydrophilic polymer;
  • X represents one of the m nucleophilic groups of component A, and the various X moieties on A may be the same or different;
  • Y represents one of the n electrophilic groups of component B, and the various Y moieties on A may be the same or different;
  • Fn represents a functional group on optional component C
  • Q 1 , Q 2 and Q 3 are linking groups; m ⁇ 2, n ⁇ 2, m + n is > 4, q, and r are independently zero or 1 , and when optional component C is present, p ⁇ 1 , and s is independently zero or 1.
  • X may be virtually any nucleophilic group, so long as reaction can occur with the electrophilic group Y.
  • Y may be virtually any electrophilic group, so long as reaction can take place with X.
  • the only limitation is a practical one, in that reaction between X and Y should be fairly rapid and take place automatically upon admixture with an aqueous medium, without need for heat or potentially toxic or non-biodegradable reaction catalysts or other chemical reagents. It is also preferred although not essential that reaction occur without need for ultraviolet or other radiation.
  • the reactions between X and Y should be complete in under 60 minutes, preferably under 30 minutes. Most preferably, the reaction occurs in about 5 to 15 minutes or less.
  • nucleophilic groups suitable as X include, but are not limited to, -NH 2 , -NHR 4 , -N(R 4 ) 2 , -SH, -OH, -COOH, -C 6 H 4 -OH, -PH 2 , - PHR 5 , -P(R 5 ) 2 , -NH-NH 2 , -CO-NH-NH 2 , -C 5 H 4 N, etc.
  • R 4 and R 5 are hydrocarbyl, typically alkyl or monocyclic aryl, preferably alkyl, and most preferably lower alkyl.
  • Organometallic moieties are also useful nucleophilic groups for the purposes of the invention, particularly those that act as carbanion donors.
  • Organometallic nucleophiles are not, however, preferred.
  • organometallic moieties include: Grignard functionalities - R 6 MgHaI wherein R 6 is a carbon atom (substituted or unsubstituted), and Hal is halo, typically bromo, iodo or chloro, preferably bromo; and lithium- containing functionalities, typically alkyllithium groups; sodium-containing functionalities.
  • nucleophilic groups must be activated with a base so as to be capable of reaction with an electrophile.
  • the composition when there are nucleophilic sulfhydryl and hydroxyl groups in the crosslinkable composition, the composition must be admixed with an aqueous base in order to remove a proton and provide an -S " or -O " species to enable reaction with an electrophile.
  • a nonnucleophilic base is preferred.
  • the base may be present as a component of a buffer solution. Suitable bases and corresponding crosslinking reactions are described infra in Section E.
  • electrophilic groups provided within the crosslinkable composition i.e., on component B, must be made so that reaction is possible with the specific nucleophilic groups.
  • the Y groups are selected so as to react with amino groups.
  • the corresponding electrophilic groups are sulfhydryl-reactive groups, and the like.
  • a carboxylic acid group perse is not susceptible to reaction with a nucleophilic amine
  • components containing carboxylic acid groups must be activated so as to be amine-reactive. Activation may be accomplished in a variety of ways, but often involves reaction with a suitable hydroxyl-containing compound in the presence of a dehydrating agent such as dicyclohexylcarbodiimide (DCC) or dicyclohexylurea (DHU).
  • a dehydrating agent such as dicyclohexylcarbodiimide (DCC) or dicyclohexylurea (DHU).
  • a carboxylic acid can be reacted with an alkoxy-substituted N-hydroxy- succinimide or N-hydroxysulfosuccinimide in the presence of DCC to form reactive electrophilic groups, the N-hydroxysuccinimide ester and the N- hydroxysulfosuccinimide ester, respectively.
  • Carboxylic acids may also be activated by reaction with an acyl halide such as an acyl chloride (e.g., acetyl chloride), to provide a reactive anhydride group.
  • a carboxylic acid may be converted to an acid chloride group using, e.g., thionyl chloride or an acyl chloride capable of an exchange reaction. Specific reagents and procedures used to carry out such activation reactions will be known to those of ordinary skill in the art and are described in the pertinent texts and literature.
  • the electrophilic groups present on Y are groups that react with a sulfhydryl moiety.
  • Such reactive groups include those that form thioester linkages upon reaction with a sulfhydryl group, such as those described in PCT Publication No. WO 00/62827 to Wallace et al.
  • such "sulfhydryl reactive" groups include, but are not limited to: mixed anhydrides; ester derivatives of phosphorus; ester derivatives of p-nitrophenol, p- nitrothiophenol and pentafluorophenol; esters of substituted hydroxylamines, including N-hydroxyphthalimide esters, N-hydroxysuccinimide esters, N- hydroxysulfosuccinimide esters, and N-hydroxyglutarimide esters; esters of 1 -hydroxybenzotriazole; 3-hydroxy-3,4-dihydro-benzotriazin-4-one; 3- hydroxy-3,4-dihydro-quinazoline-4-one; carbonylimidazole derivatives; acid chlorides; ketenes; and isocyanates.
  • auxiliary reagents can also be used to facilitate bond formation, e.g., 1-ethyl- 3-[3-dimethylaminopropyl]carbodiimide can be used to facilitate coupling of sulfhydryl groups to carboxyl-containing groups.
  • sulfhydryl reactive groups that form thioester linkages
  • various other sulfhydryl reactive functionalities can be utilized that form other types of linkages.
  • compounds that contain methyl imidate derivatives form imido-thioester linkages with sulfhydryl groups.
  • sulfhydryl reactive groups can be employed that form disulfide bonds with sulfhydryl groups; such groups generally have the structure -S- S-Ar where Ar is a substituted or unsubstituted nitrogen-containing heteroaromatic moiety or a non-heterocyclic aromatic group substituted with an electron-withdrawing moiety, such that Ar may be, for example, 4- pyridinyl, o-nitrophenyl, m-nitrophenyl, p-nitrophenyl, 2,4-dinitrophenyl, 2- nitro-4-benzoic acid, 2-nitro-4-pyridinyl, etc.
  • auxiliary reagents i.e., mild oxidizing agents such as hydrogen peroxide, can be used to facilitate disulfide bond formation.
  • sulfhydryl reactive groups forms thioether bonds with sulfhydryl groups.
  • groups include, inter alia, maleimido, substituted maleimido, haloalkyl, epoxy, imino, and aziridino, as well as olefins (including conjugated olefins) such as ethenesulfonyl, etheneimino, acrylate, methacrylate, and ⁇ , ⁇ -unsaturated aldehydes and ketones.
  • This class of sulfhydryl reactive groups are particularly preferred as the thioether bonds may provide faster crosslinking and longer in vivo stability.
  • the electrophilic functional groups on the remaining component(s) must react with hydroxy! groups.
  • the hydroxyl group may be activated as described above with respect to carboxylic acid groups, or it may react directly in the presence of base with a sufficiently reactive electrophile such as an epoxide group, an aziridine group, an acyl halide, or an anhydride.
  • suitable electrophilic functional groups for reaction therewith are those containing carbonyl groups, including, by way of example, ketones and aldehydes.
  • a carboxylic acid group can act as a nucleophile in the presence of a fairly strong base, but generally acts as an electrophile allowing nucleophilic attack at the carbonyl carbon and concomitant replacement of the hydroxyl group with the incoming nucleophile.
  • covalent linkages in the crosslinked structure that result upon covalent binding of specific nucleophilic components to specific electrophilic components in the crosslinkable composition include, solely by way of example, the following (the optional linking groups Q 1 and Q 2 are omitted for clarity):
  • the functional groups X and Y and FN on optional component C may be directly attached to the compound core (R 1 , R 2 or R 3 on optional component C, respectively), or they may be indirectly attached through a linking group, with longer linking groups also termed "chain extenders.”
  • chain extenders In structural formulae (I), (II) and (III), the optional linking groups are represented by Q 1 , Q 2 and Q 3 , wherein the linking groups are present when q, r and s are equal to 1 (with R, X, Y, Fn, m n and p as defined previously).
  • linking groups are well known in the art. See, for example, International Patent Publication No. WO 97/22371. Linking groups are useful to avoid steric hindrance problems that are sometimes associated with the formation of direct linkages between molecules. Linking groups may additionally be used to link several multifunctionally activated compounds together to make larger molecules. In a preferred embodiment, a linking group can be used to alter the degradative properties of the compositions after administration and resultant gel formation. For example, linking groups can be incorporated into components A, B, or optional component C to promote hydrolysis, to discourage hydrolysis, or to provide a site for enzymatic degradation.
  • linking groups that provide hydrolyzable sites, include, inter alia: ester linkages; anhydride linkages, such as obtained by incorporation of glutarate and succinate; ortho ester linkages; ortho carbonate linkages such as trimethylene carbonate; amide linkages; phosphoester linkages; ⁇ -hydroxy acid linkages, such as may be obtained by incorporation of lactic acid and glycolic acid; lactone-based linkages, such as may be obtained by incorporation of caprolactone, valerolactone, y- butyrolactone and p-dioxanone; and amide linkages such as in a dimeric, oligomeric, or poly(amino acid) segment.
  • non-degradable linking groups include succinimide, propionic acid and carboxymethylate linkages. See, for example, PCT WO 99/07417.
  • enzymatically degradable linkages include Leu-Gly-Pro-Ala, which is degraded by collagenase; and Gly-Pro-Lys, which is degraded by plasmin.
  • Linking groups can also enhance or suppress the reactivity of the various nucleophilic and electrophilic groups.
  • electron- withdrawing groups within one or two carbons of a sulfhydryl group would be expected to diminish its effectiveness in coupling, due to a lowering of nucleophilicity. Carbon-carbon double bonds and carbonyl groups will also have such an effect.
  • electron-withdrawing groups adjacent to a carbonyl group e.g., the reactive carbonyl of glutaryl-N- hydroxysuccinimidyl
  • sterically bulky groups in the vicinity of a functional group can be used to diminish reactivity and thus coupling rate as a result of steric hindrance.
  • n is generally in the range of 1 to about 10
  • R 7 is generally hydrocarbyl, typically alkyl or aryl, preferably alkyl, and most preferably lower alkyl
  • R 8 is hydrocarbylene, heteroatom-containing hydrocarbylene, substituted hydrocarbylene, or substituted heteroatom- containing hydrocarbylene) typically alkylene or arylene (again, optionally substituted and/or containing a heteroatom), preferably lower alkylene (e.g., methylene, ethylene, n-propylene, n-butylene, etc.), phenylene, or amidoalkylene (e.g., -(CO)-NH-CH 2 ).
  • lower alkylene e.g., methylene, ethylene, n-propylene, n-butylene, etc.
  • phenylene or amidoalkylene (e.g., -(CO)-NH-CH 2 ).
  • linking groups are as follows: If higher molecular weight components are to be used, they preferably have biodegradable linkages as described above, so that fragments larger than 20,000 mol. wt. are not generated during resorption in the body. In addition, to promote water miscibility and/or solubility, it may be desired to add sufficient electric charge or hydrophilicity. Hydrophilic groups can be easily introduced using known chemical synthesis, so long as they do not give rise to unwanted swelling or an undesirable decrease in compressive strength. In particular, polyalkoxy segments may weaken gel strength.
  • the Component Core is preferably have biodegradable linkages as described above, so that fragments larger than 20,000 mol. wt. are not generated during resorption in the body. In addition, to promote water miscibility and/or solubility, it may be desired to add sufficient electric charge or hydrophilicity. Hydrophilic groups can be easily introduced using known chemical synthesis, so long as they do not give rise to unwanted swelling or an undesirable decrease in compressive strength. In particular, polyalk
  • each crosslinkable component is comprised of the molecular structure to which the nucleophilic or electrophilic groups are bound.
  • each molecular core of the reactive components of the crosslinkable composition is generally selected from synthetic and naturally occurring hydrophilic polymers, hydrophobic polymers, and C 2 -C 14 hydrocarbyl groups zero to 2 heteroatoms selected from N, O and S, with the proviso that at least one of the crosslinkable components A, B, and optionally C, comprises a molecular core of a synthetic hydrophilic polymer.
  • at least one of A and B comprises a molecular core of a synthetic hydrophilic polymer.
  • hydrophilic Crosslinkable Components In one aspect, the crosslinkable component(s) is (are) hydrophilic polymers.
  • hydrophilic polymer refers to a synthetic polymer having an average molecular weight and composition effective to render the polymer "hydrophilic" as defined above.
  • synthetic crosslinkable hydrophilic polymers useful herein include, but are not limited to: polyalkylene oxides, particularly polyethylene glycol and poly(ethylene oxide)-poly(propylene oxide) copolymers, including block and random copolymers; polyols such as glycerol, polyglycerol (particularly highly branched polyglycerol), propylene glycol and trimethylene glycol substituted with one or more polyalkylene oxides, e.g., mono-, di- and tri-polyoxyethylated glycerol, mono- and di-polyoxyethylated propylene glycol, and mono- and di-polyoxyethylated trimethylene glycol; polyoxyethylated sorbitol, polyoxyethylated glucose; acrylic acid polymers and analogs and copolymers thereof, such as polyacrylic acid perse, polymethacrylic acid, poly(hydroxyethyl-methacrylate), poly(hydroxyethy
  • the synthetic crosslinkable hydrophilic polymer may be a homopolymer, a block copolymer, a random copolymer, or a graft copolymer.
  • the polymer may be linear or branched, and if branched, may be minimally to highly branched, dendrimeric, hyperbranched, or a star polymer.
  • the polymer may include biodegradable segments and blocks, either distributed throughout the polymer's molecular structure or present as a single block, as in a block copolymer. Biodegradable segments are those that degrade so as to break covalent bonds. Typically, biodegradable segments are segments that are hydrolyzed in the presence of water and/or enzymatically cleaved in situ.
  • Biodegradable segments may be composed of small molecular segments such as ester linkages, anhydride linkages, ortho ester linkages, ortho carbonate linkages, amide linkages, phosphonate linkages, etc.
  • Larger biodegradable "blocks" will generally be composed of oligomeric or polymeric segments incorporated within the hydrophilic polymer.
  • Illustrative oligomeric and polymeric segments that are biodegradable include, by way of example, poly(amino acid) segments, poly(orthoester) segments, poly(orthocarbonate) segments, and the like.
  • Suitable synthetic crosslinkable hydrophilic polymers include chemically synthesized polypeptides, particularly polynucleophilic polypeptides that have been synthesized to incorporate amino acids containing primary amino groups (such as lysine) and/or amino acids containing thiol groups (such as cysteine).
  • Poly(lysine) a synthetically produced polymer of the amino acid lysine (145 MW), is particularly preferred.
  • Poly(lysine)s have been prepared having anywhere from 6 to about 4,000 primary amino groups, corresponding to molecular weights of about 870 to about 580,000.
  • Poly(lysine)s for use in the present invention preferably have a molecular weight within the range of about 1 ,000 to about 300,000, more preferably within the range of about 5,000 to about 100,000, and most preferably, within the range of about 8,000 to about 15,000.
  • Poly(lysine)s of varying molecular weights are commercially available from Peninsula Laboratories, Inc. (Belmont, Calif.).
  • the synthetic crosslinkable hydrophilic polymer may be a homopolymer, a block copolymer, a random copolymer, or a graft copolymer.
  • the polymer may be linear or branched, and if branched, may be minimally to highly branched, dendrimeric, hyperbranched, or a star polymer.
  • the polymer may include biodegradable segments and blocks, either distributed throughout the polymer's molecular structure or present as a single block, as in a block copolymer. Biodegradable segments are those that degrade so as to break covalent bonds. Typically, biodegradable segments are segments that are hydrolyzed in the presence of water and/or enzymatically cleaved in situ.
  • Biodegradable segments may be composed of small molecular segments such as ester linkages, anhydride linkages, ortho ester linkages, ortho carbonate linkages, amide linkages, phosphonate linkages, etc.
  • Larger biodegradable "blocks" will generally be composed of oligomeric or polymeric segments incorporated within the hydrophilic polymer.
  • Illustrative oligomeric and polymeric segments that are biodegradable include, by way of example, poly(amino acid) segments, poly(orthoester) segments, poly(orthocarbonate) segments, and the like.
  • preferred synthetic crosslinkable hydrophilic polymers are polyethylene glycol (PEG) and polyglycerol (PG), particularly highly branched polyglycerol.
  • PEG polyethylene glycol
  • PG polyglycerol
  • Various forms of PEG are extensively used in the modification of biologically active molecules because PEG lacks toxicity, antigenicity, and immunogenicity (i.e., is biocompatible), can be formulated so as to have a wide range of solubilities, and do not typically interfere with the enzymatic activities and/or conformations of peptides.
  • a particularly preferred synthetic crosslinkable hydrophilic polymer for certain applications is a polyethylene glycol (PEG) having a molecular weight within the range of about 100 to about 100,000 mol. wt., although for highly branched PEG, far higher molecular weight polymers can be employed — up to 1 ,000,000 or more - providing that biodegradable sites are incorporated ensuring that all degradation products will have a molecular weight of less than about 30,000.
  • the preferred molecular weight is about 1,000 to about 20,000 mol. wt., more preferably within the range of about 7,500 to about 20,000 mol. wt.
  • the polyethylene glycol has a molecular weight of approximately 10,000 mol. wt.
  • Naturally occurring crosslinkable hydrophilic polymers include, but are not limited to: proteins such as collagen, fibronectin, albumins, globulins, fibrinogen, and fibrin, with collagen particularly preferred; carboxylated polysaccharides such as polymannuronic acid and polygalacturonic acid; aminated polysaccharides, particularly the glycosaminoglycans, e.g., hyaluronic acid, chitin, chondroitin sulfate A, B, or C, keratin sulfate, keratosulfate and heparin; and activated polysaccharides such as dextran and starch derivatives.
  • proteins such as collagen, fibronectin, albumins, globulins, fibrinogen, and fibrin, with collagen particularly preferred
  • carboxylated polysaccharides such as polymannuronic acid and polygalacturonic acid
  • aminated polysaccharides particularly the glycosaminoglycans
  • Collagen and glycosaminoglycans are examples of naturally occurring hydrophilic polymers for use herein, with methylated collagen being a preferred hydrophilic polymer.
  • Any of the hydrophilic polymers herein must contain, or be activated to contain, functional groups, i.e., nucleophilic or electrophilic groups, which enable crosslinking. Activation of PEG is discussed below; it is to be understood, however, that the following discussion is for purposes of illustration and analogous techniques may be employed with other polymers.
  • Activated forms of PEG are commercially available, and are also easily prepared using known methods. For example, see Chapter 22 of Poly(ethylene Glycol) Chemistry: Biotechnical and Biomedical Applications, J. Milton Harris, ed., Plenum Press, NY (1992); and Shearwater Polymers, Inc. Catalog, Polyethylene Glycol Derivatives, Huntsville, Alabama (1997-1998).
  • FIGS. 1 to 10 of U.S. Patent 5,874,500 Structures for some specific, tetrafunctionally activated forms of PEG are shown in FIGS. 1 to 10 of U.S. Patent 5,874,500, as are generalized reaction products obtained by reacting the activated PEGs with multi-amino PEGs, i.e., a PEG with two or more primary amino groups.
  • the activated PEGs illustrated have a pentaerythritol (2,2-bis(hydroxymethyl)- 1 ,3-propanediol) core.
  • Such activated PEGs are readily prepared by conversion of the exposed hydroxyl groups in the PEGylated polyol (i.e., the terminal hydroxyl groups on the PEG chains) to carboxylic acid groups (typically by reaction with an anhydride in the presence of a nitrogenous base), followed by esterification with N-hydroxysuccinimide, N-hydroxysulfosuccinimide, or the like, to give the polyfunctionally activated PEG.
  • the crosslinkable compositions of the invention can also include hydrophobic polymers, although for most uses hydrophilic polymers are preferred.
  • Hydrophilic polymers are examples of two hydrophobic polymers that can be used.
  • With other hydrophobic polymers only short-chain oligomers should be used, containing at most about 14 carbon atoms, to avoid solubility-related problems during reaction.
  • the molecular core of one or more of the crosslinkable components can also be a low molecular weight compound, Ae., a C 2 -C 14 hydrocarbyl group containing zero to 2 heteroatoms selected from N, O, S and combinations thereof.
  • a molecular core can be substituted with nucleophilic groups or with electrophilic groups.
  • the component may be, for example, ethylenediamine (H 2 N-CH 2 CH 2 -NH 2 ), tetramethylenediamine (H 2 N-(CH 4 )- NH 2 ), pentamethylenediamine (cadaverine) (H 2 N-(CH 5 )-NH 2 ), hexamethylenediamine (H 2 N-(CH 6 )-NH 2 ), bis(2-aminoethyl)amine (HN- [CH 2 CH 2 -NH 2 I 2 ), or tris(2-aminoethyl)amine (N-[CH 2 CH 2 -NH 2 J 3 ).
  • ethylenediamine H 2 N-CH 2 CH 2 -NH 2
  • tetramethylenediamine H 2 N-(CH 4 )- NH 2
  • pentamethylenediamine cadaverine
  • H 2 N-(CH 5 )-NH 2 hexamethylenediamine
  • H 2 N-(CH 6 )-NH 2 bis(
  • Low molecular weight diols and polyols include trimethylolpropane, di(trimethylol propane), pentaerythritol, and diglycerol, all of which require activation with a base in order to facilitate their reaction as nucleophiles.
  • Such diols and polyols may also be functionalized to provide di- and poly-carboxylic acids, functional groups that are, as noted earlier herein, also useful as nucleophiles under certain conditions.
  • Polyacids for use in the present compositions include, without limitation, trimethylolpropane-based tricarboxylic acid, di(trimethylol propane)-based tetracarboxylic acid, heptanedioic acid, octanedioic acid (suberic acid), and hexadecanedioic acid (thapsic acid), all of which are commercially available and/or readily synthesized using known techniques.
  • Low molecular weight di- and poly-electrophiles include, for example, disuccinimidyl suberate (DSS), bis(sulfosuccinimidyl) suberate (BS 3 ), dithiobis(succinimidylpropionate) (DSP), bis(2- succinimidooxycarbonyloxy) ethyl sulfone (BSOCOES), and 3,3'- dithiobis(sulfosuccinimidylpropionate (DTSPP), and their analogs and derivatives.
  • DSS disuccinimidyl suberate
  • BS 3 bis(sulfosuccinimidyl) suberate
  • DSP dithiobis(succinimidylpropionate)
  • BSOCOES bis(2- succinimidooxycarbonyloxy) ethyl sulfone
  • DTSPP 3,3'- dithiobis(sulfosuccinimidylpropionat
  • di- and poly-electrophiles can also be synthesized from di- and polyacids, for example by reaction with an appropriate molar amount of N-hydroxysuccinimide in the presence of DCC.
  • Polyols such as trimethylolpropane and di(trimethylol propane) can be converted to carboxylic acid form using various known techniques, then further derivatized by reaction with NHS in the presence of DCC to produce trifunctiona ⁇ ly and tetrafunctionally activated polymers.
  • Suitable delivery systems for the homogeneous dry powder composition (containing at least two crosslinkable polymers) and the two buffer solutions may involve a multi-compartment spray device, where one or more compartments contains the powder and one or more compartments contain the buffer solutions needed to provide for the aqueous environment, so that the composition is exposed to the aqueous environment as it leaves the compartment.
  • a multi-compartment spray device where one or more compartments contains the powder and one or more compartments contain the buffer solutions needed to provide for the aqueous environment, so that the composition is exposed to the aqueous environment as it leaves the compartment.
  • Many devices that are adapted for delivery of multi- component tissue sealants/hemostatic agents are well known in the art and can also be used in the practice of the present invention.
  • the composition can be delivered using any type of controllable extrusion system, or it can be delivered manually in the form of a dry powder, and exposed to the aqueous environment at the site of administration.
  • the homogeneous dry powder composition and the two buffer solutions may be conveniently formed under aseptic conditions by placing each of the three ingredients (dry powder, acidic buffer solution and basic buffer solution) into separate syringe barrels.
  • the composition, first buffer solution and second buffer solution can be housed separately in a multiple-compartment syringe system having a multiple barrels, a mixing head, and an exit orifice.
  • the first buffer solution can be added to the barrel housing the composition to dissolve the composition and form a homogeneous solution, which is then extruded into the mixing head.
  • the second buffer solution can be simultaneously extruded into the mixing head.
  • the resulting composition can then be extruded through the orifice onto a surface.
  • the syringe barrels holding the dry powder and the basic buffer may be part of a dual-syringe system, e.g., a double barrel syringe as described in U.S. Patent 4,359,049 to Redl et al.
  • the acid buffer can be added to the syringe barrel that also holds the dry powder, so as to produce the homogeneous solution.
  • the acid buffer may be added (e.g., injected) into the syringe barrel holding the dry powder to thereby produce a homogeneous solution of the first and second components. This homogeneous solution can then be extruded into a mixing head, while the basic buffer is simultaneously extruded into the mixing head.
  • the homogeneous solution and the basic buffer are mixed together to thereby form a reactive mixture.
  • the reactive mixture is extruded through an orifice and onto a surface (e.g., tissue), where a film is formed, which can function as a sealant or a barrier, or the like.
  • the reactive mixture begins forming a three- dimensional matrix immediately upon being formed by the mixing of the homogeneous solution and the basic buffer in the mixing head.
  • the reactive mixture is preferably extruded from the mixing head onto the tissue very quickly after it is formed so that the three-dimensional matrix forms on, and is able to adhere to, the tissue.
  • Other systems for combining two reactive liquids are well known in the art, and include the systems described in U.S.
  • the electrophilic component or components are generally stored and used in sterile, dry form to prevent hydrolysis.
  • Processes for preparing synthetic hydrophilic polymers containing multiple electrophilic groups in sterile, dry form are set forth in commonly assigned U.S. Patent No. 5,643,464 to Rhee et al.
  • the dry synthetic polymer may be compression molded into a thin sheet or membrane, which can then be sterilized using gamma or, preferably, e-beam irradiation. The resulting dry membrane or sheet can be cut to the desired size or chopped into smaller size particulates.
  • Components containing multiple nucleophilic groups are generally not water-reactive and can therefore be stored either dry or in aqueous solution. If stored as a dry, particulate, solid, the various components of the crosslinkable composition may be blended and stored in a single container. Admixture of all components with water, saline, or other aqueous media should not occur until immediately prior to use.
  • the crosslinking components can be mixed together in a single aqueous medium in which they are both unreactive, i.e., such as in a low pH buffer. Thereafter, they can be sprayed onto the targeted tissue site along with a high pH buffer, after which they will rapidly react and form a gel.
  • Suitable liquid media for storage of crosslinkable compositions include aqueous buffer solutions such as monobasic sodium phosphate/dibasic sodium phosphate, sodium carbonate/sodium bicarbonate, glutamate or acetate, at a concentration of 0.5 to 300 mM.
  • a sulfhydryl-reactive component such as PEG substituted with maleimido groups or succinimidyl esters is prepared in water or a dilute buffer, with a pH of between around 5 to 6.
  • Buffers with pKs between about 8 and 10.5 for preparing a polysulfhydryl component such as sulfhydryl-PEG are useful to achieve fast gelation time of compositions containing mixtures of sulfhydryl-PEG and SG-PEG.
  • These include carbonate, borate and AMPSO (3-[(1 ,1-dimethyl-2-hydroxyethyl)amino]2-hydroxy-propane-sulfonic acid).
  • a pH of around 5 to 9 is preferred for the liquid medium used to prepare the sulfhydryl PEG.
  • the polymer composition may include collagen in combination with fibrinogen and/or thrombin.
  • an aqueous composition may include a fibrinogen and FXIII, particularly plasma, collagen in an amount sufficient to thicken the composition, thrombin in an amount sufficient to catalyze polymerization of fibrinogen present in the composition, and Ca 2+ and, optionally, an antifibrinolytic agent in amount sufficient to retard degradation of the resulting adhesive clot.
  • the composition may be formulated as a two-part composition that may be mixed together just prior to use, in which fibrinogen/FXIII and collagen constitute the first component, and thrombin together with an antifibrinolytic agent, and Ca 2+ constitute the second component.
  • Plasma which provides a source of fibrinogen
  • the plasma may be obtained from the patient for which the composition is to be delivered.
  • the plasma can be used "as is" after standard preparation which includes centrifuging out cellular components of blood.
  • the plasma can be further processed to concentrate the fibrinogen to prepare a plasma cryoprecipitate.
  • the plasma cryoprecipitate can be prepared by freezing the plasma for at least about an hour at about -20 0 C, and then storing the frozen plasma overnight at about 4 0 C. to slowly thaw.
  • the thawed plasma is centrifuged and the plasma cryoprecipitate is harvested by removing approximately four-fifths of the plasma to provide a cryoprecipitate comprising the remaining one-fifth of the plasma.
  • fibrinogen/FXIII preparations may be used, such as cryoprecipitate, patient autologous fibrin sealant, fibrinogen analogs or other single donor or commercial fibrin sealant materials.
  • Approximately 0.5 ml to about 1.0 ml of either the plasma or the plasma-cryoprecipitate provides about 1 to 2 ml of adhesive composition which is sufficient for use in middle ear surgery.
  • Other plasma proteins e.g., albumin, plasminogen, von Willebrands factor, Factor VIII, etc.
  • Collagen preferably hypoallergenic collagen
  • the collagen may be atelbpeptide collagen or telopeptide collagen, e.g., native collagen.
  • the collagen augments the fibrin by acting as a macromolecular lattice work or scaffold to which the fibrin network adsorbs. This gives more strength and durability to the resulting glue clot with a relatively low concentration of fibrinogen in comparison to the various concentrated autogenous fibrinogen glue formulations (i.e., AFGs).
  • the form of collagen which is employed may be described as at least "near native" in its structural characteristics. It may be further characterized as resulting in insoluble fibers at a pH above 5; unless crosslinked or as part of a complex composition, e.g., bone, it will generally consist of a minor amount by weight of fibers with diameters greater than 50 nm, usually from about 1 to 25 volume % and there will be substantially little, if any, change in the helical structure of the fibrils.
  • the collagen composition must be able to enhance gelation in the surgical adhesion composition. A number of commercially available collagen preparations may be used.
  • ZYDERM Collagen Implant has a fibrillar diameter distribution consisting of 5 to 10 nm diameter fibers at 90% volume content and the remaining 10% with greater than about 50 nm diameter fibers.
  • ZCI is available as a fibrillar slurry and solution in phosphate buffered isotonic saline, pH 7.2, and is injectable with fine gauge needles.
  • ZYPLAST As distinct from ZCI 1 cross-linked collagen available as ZYPLAST may be employed.
  • ZYPL-AST is essentially an exogenously crosslinked (glutaraldehyde) version of ZCI. The material has a somewhat higher content of greater than about 50 nm diameter fibrils and remains insoluble over a wide pH range. Crosslinking has the effect of mimicking in vivo endogenous crosslinking found in many tissues.
  • Thrombin acts as a catalyst for fibrinogen to provide fibrin, an insoluble polymer and is present in the composition in an amount sufficient to catalyze polymerization of fibrinogen present in the patient plasma. Thrombin also activates FXIII, a plasma protein that catalyzes covalent crosslinks in fibrin, rendering the resultant clot insoluble.
  • FXIII a plasma protein that catalyzes covalent crosslinks in fibrin, rendering the resultant clot insoluble.
  • the thrombin is present in the adhesive composition in concentration of from about 0.01 to about 1000 or greater NIH units (NlHu) of activity, usually about i to about 500 NIHu, most usually about 200 to about 500 NIHu.
  • the thrombin can be from a variety of host animal sources, conveniently bovine.
  • Thrombin is commercially available from a variety of sources including Parke-Davis, usually lyophilized with buffer salts and stabilizers in vials which provide thrombin activity ranging from about 1000 NIHu to 10,000 NIHu.
  • the thrombin is usually prepared by reconstituting the powder by the addition of either sterile distilled water or isotonic saline. Alternately, thrombin analogs or reptile-sourced coagulants may be used.
  • the composition may additionally comprise an effective amount of an antifibrinolytic agent to enhance the integrity of the glue clot as the healing processes occur.
  • antifibrinolytic agents include aprotinin, C1 -esterase inhibitor and ⁇ -amino-n-caproic acid (EACA).
  • ⁇ -amino-n-caproic acid the only antifibrinolytic agent approved by the FDA, is effective at a concentration of from about 5 mg/ml to about 40 mg/ml of the final adhesive composition, more usually from about 20 to about 30 mg/ml.
  • EACA is commercially available as a solution having a concentration of about 250 mg/ml. Conveniently, the commercial solution is diluted with distilled water to provide a solution of the desired concentration. That solution is desirably used to reconstitute lyophilized thrombin to the desired thrombin concentration.
  • in situ forming materials based on the crosslinking of proteins are described, e.g., in U.S. Patent Nos. RE38158; 4,839,345; 5,514,379, 5,583,114; 6,458,147; 6,371 ,975; 5,290,552; 6,096,309; U.S. Patent Application Publication Nos. 2002/0161399; 2001/0018598 and PCT Publication Nos. WO 03/090683; WO 01/45761 ; WO 99/66964 and WO 96/03159).
  • the therapeutic agent is released from a crosslinked matrix formed, at least in part, from a self-reactive compound.
  • a self-reactive compound comprises a core substituted with a minimum of three reactive groups.
  • the reactive groups may be directed attached to the core of the compound, or the reactive groups may be indirectly attached to the compound's core, e.g., the reactive groups are joined to the core through one or more linking groups.
  • Each of the three reactive groups that are necessarily present in a self-reactive compound can undergo a bond-forming reaction with at least one of the remaining two reactive groups.
  • the term "self-reactive” is not intended to mean that each self-reactive compound necessarily reacts with itself, but rather that when a plurality of identical self-reactive compounds are in combination and undergo a crosslinking reaction, then these compounds will react with one another to form the matrix.
  • the compounds are "self-reactive" in the sense that they can react with other compounds having the identical chemical structure as themselves.
  • the self-reactive compound comprises at least four components: a core and three reactive groups.
  • the self-reactive compound can be characterized by the formula (I), where R is the core, the reactive groups are represented by X 1 , X 2 and X 3 , and a linker (L) is optionally present between the core and a functional group.
  • the core R is a polyvalent moiety having attachment to at least three groups (i.e., it is at least trivalent) and may be, or may contain, for example, a hydrophilic polymer, a hydrophobic polymer, an amphiphilic polymer, a C 2-I4 hydrocarbyl, or a C 2 - 14 hydrocarbyl which is heteroatom- containing.
  • the linking groups L 1 , L 2 , and L 3 may be the same or different.
  • the designators p, q and r are either 0 (when no linker is present) or 1 (when a linker is present).
  • the reactive groups X 1 , X 2 and X 3 may be the same or different.
  • Each of these reactive groups reacts with at least one other reactive group to form a three-dimensional matrix. Therefore X 1 can react with X 2 and/or X 3 , X 2 can react with X 1 and/or X 3 , X 3 can react with X 1 and/or X 2 and so forth.
  • a trivalent core will be directly or indirectly bonded to three functional groups, a tetravalent core will be directly or indirectly bonded to four functional groups, etc.
  • each side chain typically has one reactive group.
  • the invention also encompasses self-reactive compounds where the side chains contain more than one reactive group.
  • the self-reactive compound has the formula (II): [ X' - (L 4 ) a - Y' - (L 5 ) b ] c R 1 where: a and b are integers from 0-1; c is an integer from 3-12; R 1 is selected from hydrophilic polymers, hydrophobic polymers, amphiphilic polymers, C 2- i 4 hydrocarbyls, and heteroatom-containing C 2-M hydrocarbyls; X' and Y' are reactive groups and can be the same or different; and L 4 and L 5 are linking groups.
  • Each reactive group inter-reacts with the other reactive group to form a three-dimensional matrix.
  • the compound is essentially non-reactive in an initial environment but is rendered reactive upon exposure to a modification in the initial environment that provides a modified environment such that a plurality of the self-reactive compounds inter-react in the modified environment to form a three-dimensional matrix.
  • R is a hydrophilic polymer.
  • X' is a nucleophilic group and Y' is an electrophilic group.
  • J ne L following self-reactive compound is one example of a compound of formula (II):
  • R 4 has the formula:
  • a and b are 1; c is 4; the core R' is the hydrophilic polymer, tetrafunctionally activated polyethylene glycol, (C(CH 2 - O-) 4 ; X 1 is the electrophilic reactive group, succinimidyl; Y' is the nucleophilic reactive group -CH-NH 2 ; L 4 is -C(O)-O-; and L 5 is -(CH 2 - C H 2 -O-C H 2 ) x -C H 2 -
  • the reactive groups are selected so that the compound is essentially non-reactive in an initial environment. Upon exposure to a specific modification in the initial environment, providing a modified environment, the compound is rendered reactive and a plurality of self- reactive compounds are then able to inter-react in the modified environment to form a three-dimensional matrix. Examples of modification in the initial environment are detailed below, but include the addition of an aqueous medium, a change in pH, exposure to ultraviolet radiation, a change in temperature, or contact with a redox initiator.
  • the core and reactive groups can also be selected so as to provide a compound that has one of more of the following features: are biocompatible, are non-immunogenic, and do not leave any toxic, inflammatory or immunogenic reaction products at the site of administration. Similarly, the core and reactive groups can also be selected so as to provide a resulting matrix that has one or more of these features.
  • the self-reactive compounds inter-react form a three-dimensional matrix.
  • the term "substantially immediately” is intended to mean within less than five minutes, preferably within less than two minutes, and the term “immediately” is intended to mean within less than one minute, preferably within less than 30 seconds.
  • the self-reactive compound and resulting matrix are not subject to enzymatic cleavage by matrix metalloproteinases such as collagenase, and are therefore not readily degradable in vivo.
  • the self-reactive compound may be readily tailored, in terms of the selection and quantity of each component, to enhance certain properties, e.g., compression strength, swellability, tack, hydrophilicity, optical clarity, and the like.
  • R is a hydrophilic polymer.
  • X is a nucleophilic group
  • Y is an electrophilic group
  • Z is either an electrophilic or a nucleophilic group. Additional embodiments are detailed below.
  • a higher degree of inter-reaction e.g., crosslinking
  • n be an integer from 2-12.
  • the compounds may be the same or different.
  • the self-reactive compound Prior to use, the self-reactive compound is stored in an initial environment that insures that the compound remain essentially non-reactive until use. Upon modification of this environment, the compound is rendered reactive and a plurality of compounds will then inter-react to form the desired matrix.
  • the initial environment, as well as the modified environment, is thus determined by the nature of the reactive groups involved.
  • the number of reactive groups can be the same or different. However, in one embodiment of the invention, the number of reactive groups are approximately equal. As used in this context, the term “approximately” refers to a 2:1 to 1 :2 ratio of moles of one reactive group to moles of a different reactive groups. A 1 :1 :1 molar ratio of reactive groups is generally preferred.
  • the concentration of the self-reactive compounds in the modified environment when liquid in nature, will be in the range of about 1 to 50 wt%, generally about 2 to 40 wt%.
  • the preferred concentration of the compound in the liquid will depend on a number of factors, including the type of compound (Ae., type of molecular core and reactive groups), its molecular weight, and the end use of the resulting three-dimensional matrix. For example, use of higher concentrations of the compounds, or using highly functionalized compounds, will result in the formation of a more tightly crosslinked network, producing a stiffer, more robust gel. As such, compositions intended for use in tissue augmentation will generally employ concentrations of self-reactive compounds that fall toward the higher end of the preferred concentration range.
  • compositions intended for use as bioadhesives or in adhesion prevention do not need to be as firm and may therefore contain lower concentrations of the self-reactive compounds.
  • the reactive groups are electrophilic and nucleophilic groups, which undergo a nucleophilic substitution reaction, a nucleophilic addition reaction, or both.
  • electrophilic refers to a reactive group that is susceptible to nucleophilic attack, i.e., susceptible to reaction with an incoming nucieophilic group.
  • Electrophilic groups herein are positively charged or electron-deficient, typically electron-deficient.
  • nucleophilic refers to a reactive group that is electron rich, has an unshared pair of electrons acting as a reactive site, and reacts with a positively charged or electron-deficient site.
  • the modification in the initial environment comprises the addition of an aqueous medium and/or a change in pH.
  • X1 (also referred to herein as X) can be a nucleophilic group and X2 (also referred to herein as Y) can be an electrophilic group or vice versa, and X3 (also referred to herein as Z) can be either an electrophilic or a nucleophilic group.
  • X may be virtually any nucleophilic group, so long as reaction can occur with the electrophilic group Y and also with Z, when Z is electrophilic (Z E L).
  • Y may be virtually any electrophilic group, so long as reaction can take place with X and also with Z when Z is nucleophilic (ZNU)-
  • ZNU nucleophilic
  • the reactions between X and Y, and between either X and ZEL or Y and Z NU are complete in under 60 minutes, preferably under 30 minutes. Most preferably, the reaction occurs in about 5 to 15 minutes or less.
  • nucleophilic groups suitable as X or F ⁇ INU include, but are not limited to: -NH 2 , -NHR 1 , -N(R 1 ) 2 , -SH, -OH, -COOH, -C 6 H 4 -OH, -H, -PH 2 ,
  • R 1 is a hydrocarbyl group and each R1 may be the same or different.
  • R 1 is typically alkyl or monocyclic aryl, preferably alkyl, and most preferably lower alkyl.
  • Organometallic moieties are also useful nucleophilic groups for the purposes of the invention, particularly those that act as carbanion donors.
  • organometallic moieties include: Grignard functionalities - R 2 MgHaI wherein R 2 is a carbon atom (substituted or unsubstituted), and Hal is halo, typically bromo, iodo or chloro, preferably bromo; and lithium- containing functionalities, typically alkyllithium groups; sodium-containing functionalities.
  • nucleophilic groups must be activated with a base so as to be capable of reaction with an electrophilic group.
  • the compound when there are nucleophilic sulfhydryl and hydroxyl groups in the self-reactive compound, the compound must be admixed with an aqueous base in order to remove a proton and provide an -S " or -O " species to enable reaction with the electrophilic group.
  • a non-nucleophilic base is preferred.
  • the base may be present as a component of a buffer solution. Suitable bases and corresponding crosslinking reactions are described herein.
  • electrophilic groups provided on the self- reactive compound must be made so that reaction is possible with the specific nucleophilic groups.
  • the Y and any Z E L groups are selected so as to react with amino groups.
  • the corresponding electrophilic groups are sulfhydryl-reactive groups, and the like.
  • the amine-reactive groups contain an electrophilically reactive carbonyl group susceptible to nucleophilic attack by a primary or secondary amine, for example the carboxylic acid esters and aldehydes noted above, as well as carboxyl groups (-COOH).
  • a carboxylic acid group per se is not susceptible to reaction with a nucleophilic amine
  • components containing carboxylic acid groups must be activated so as to be amine-reactive. Activation may be accomplished in a variety of ways, but often involves reaction with a suitable hydroxyl-containing compound in the presence of a dehydrating agent such as dicyclohexylcarbodiimide (DCC) or dicyclohexylurea (DHU).
  • a dehydrating agent such as dicyclohexylcarbodiimide (DCC) or dicyclohexylurea (DHU).
  • a carboxylic acid can be reacted with an alkoxy-substituted N- hydroxy-succinimide or N-hydroxysulfosuccinimide in the presence of DCC to form reactive electrophilic groups, the N-hydroxysuccinimide ester and the N-hydroxysulfosuccinimide ester, respectively.
  • Carboxylic acids may also be activated by reaction with an acyl halide such as an acyl chloride (e.g., acetyl chloride), to provide a reactive anhydride group.
  • a carboxylic acid may be converted to an acid chloride group using, e.g., thionyl chloride or an acyl chloride capable of an exchange reaction.
  • thionyl chloride or an acyl chloride capable of an exchange reaction Specific reagents and procedures used to carry out such activation reactions will be known to those of ordinary skill in the art and are described in the pertinent texts and literature.
  • the amine-reactive groups are selected from succinimidyl ester (-O(CO)-N(COCH 2 ) 2 ), sulfosuccinimidyl ester (-O(CO)-N(COCH 2 ) 2 -S(O) 2 OH), maleimido (-N(COCH) 2 ), epoxy, isocyanato, thioisocyanato, and ethenesulfonyl.
  • the electrophilic groups present on Y and ZE L are groups that react with a sulfhydryl moiety.
  • Such reactive groups include those that form thioester linkages upon reaction with a sulfhydryl group, such as those described in WO 00/62827 to Wallace et al.
  • sulfhydryl reactive groups include, but are not limited to: mixed anhydrides; ester derivatives of phosphorus; ester derivatives of p-nitrophenol, p-nitrothiophenol and pentafluorophenol; esters of substituted hydroxylamines, including N-hydroxyphthalimide esters, N- hydroxysuccinimide esters, N-hydroxysulfosuccinimide esters, and N- hydroxyglutarimide esters; esters of 1-hydroxybenzotriazole; 3-hydroxy-3,4- dihydro-benzotriazin-4-one; 3-hydroxy-3,4-dihydro-quinazoline-4-one; carbonylimidazole derivatives; acid chlorides; ketenes; and isocyanates.
  • auxiliary reagents can also be used to facilitate bond formation, e.g., 1-ethyl-3-[3- dimethylaminopropyljcarbodiimide can be used to facilitate coupling of sulfhydryl groups to carboxyl-containing groups.
  • sulfhydryl reactive groups that form thioester linkages
  • various other sulfhydryl reactive functionalities can be utilized that form other types of linkages.
  • compounds that contain methyl imidate derivatives form imido-thioester linkages with sulfhydryl groups.
  • sulfhydryl reactive groups can be employed that form disulfide bonds with sulfhydryl groups; such groups generally have the structure -S- S-Ar where Ar is a substituted or unsubstituted nitrogen-containing heteroaromatic moiety or a non-heterocyclic aromatic group substituted with an electron-withdrawing moiety, such that Ar may be, for example, 4- pyridinyl, o-nitrophenyl, m-nitrophenyl, p-nitrophenyl, 2,4-dinitrophenyl, 2- nitro-4-benzoic acid, 2-nitro-4-pyridinyl, etc.
  • auxiliary reagents Ae., mild oxidizing agents such as hydrogen peroxide, can be used to facilitate disulfide bond formation.
  • sulfhydryl reactive groups forms thioether bonds with sulfhydryl groups.
  • groups include, inter alia, maleimido, substituted maleimido, haloalkyl, epoxy, imino, and aziridino, as well as olefins (including conjugated olefins) such as ethenesulfonyl, etheneimino, acrylate, methacrylate, and ⁇ , ⁇ -unsaturated aldehydes and ketones.
  • the electrophilic functional groups on the remaining component(s) must react with hydroxyl groups.
  • the hydroxyl group may be activated as described above with respect to carboxylic acid groups, or it may react directly in the presence of base with a sufficiently reactive electrophilic group such as an epoxide group, an aziridine group, an acyl halide, an anhydride, and so forth.
  • Suitable electrophilic functional groups for reaction therewith are those containing carbonyl groups, including, by way of example, ketones and aldehydes.
  • a carboxylic acid group can act as a nucleophilic group in the presence of a fairly strong base, but generally acts as an electrophilic group allowing nucleophilic attack at the carbonyl carbon and concomitant replacement of the hydroxyl group with the incoming nucleophilic group.
  • the initial environment typically can be dry and sterile. Since electrophilic groups react with water, storage in sterile, dry form will prevent hydrolysis.
  • the dry synthetic polymer may be compression molded into a thin sheet or membrane, which can then be sterilized using gamma or e-beam irradiation. The resulting dry membrane or sheet can be cut to the desired size or chopped into smaller size particulates.
  • the modification of a dry initial environment will typically comprise the addition of an aqueous medium.
  • the initial environment can be an aqueous medium such as in a low pH buffer, i.e., having a pH less than about 6.0, in which both electrophilic and nucleophilic groups are non-reactive.
  • aqueous buffer solutions such as monobasic sodium phosphate/dibasic sodium phosphate, sodium carbonate/sodium bicarbonate, glutamate or acetate, at a concentration of 0.5 to 300 mM.
  • Modification of an initial low pH aqueous environment will typically comprise increasing the pH to at least pH 7.0, more preferably increasing the pH to at least pH 9.5.
  • the modification of a dry initial environment comprises dissolving the self-reactive compound in a first buffer solution having a pH within the range of about 1.0 to 5.5 to form a homogeneous solution, and (ii) adding a second buffer solution having a pH within the range of about 6.0 to 11.0 to the homogeneous solution.
  • the buffer solutions are aqueous and can be any pharmaceutically acceptable basic or acid composition.
  • the term "buffer" is used in a general sense to refer to an acidic or basic aqueous solution, where the solution may or may not be functioning to provide a buffering effect (i.e., resistance to change in pH upon addition of acid or base) in the compositions of the present invention.
  • the self-reactive compound can be in the form of a homogeneous dry powder.
  • This powder is then combined with a buffer solution having a pH within the range of about 1.0 to 5.5 to form a homogeneous acidic aqueous solution, and this solution is then combined with a buffer solution having a pH within the range of about 6.0 to 11.0 to form a reactive solution.
  • 0.375 grams of the dry powder can be combined with 0.75 grams of the acid buffer to provide, after mixing, a homogeneous solution, where this solution is combined with 1.1 grams of the basic buffer to provide a reactive mixture that substantially immediately forms a three-dimensional matrix.
  • Acidic buffer solutions having a pH within the range of about 1.0 to 5.5 include by way of illustration and not limitation, solutions of: citric acid, hydrochloric acid, phosphoric acid, sulfuric acid, AMPSO (3-[(1,1- dimethyl-2-hydroxyethyl)amino]2-hydroxy-propane-sulfonic acid), acetic acid, lactic acid, and combinations thereof.
  • the acidic buffer solution is a solution of citric acid, hydrochloric acid, phosphoric acid, sulfuric acid, and combinations thereof.
  • the acidic buffer preferably has a pH such that it retards the reactivity of the nucleophilic groups on the core.
  • a pH of 2.1 is generally sufficient to retard the nucleophilicity of thiol groups.
  • a lower pH is typically preferred when the core contains amine groups as the nucleophilic groups.
  • the acidic buffer is an acidic solution that, when contacted with nucleophilic groups, renders those nucleophilic groups relatively non-nucleophilic.
  • An exemplary acidic buffer is a solution of hydrochloric acid, having a concentration of about 6.3 mM and a pH in the range of 2.1 to 2.3.
  • This buffer may be prepared by combining concentrated hydrochloric acid with water, i.e., by diluting concentrated hydrochloric acid with water.
  • this buffer A may also be conveniently prepared by diluting 1.23 grams of concentrated hydrochloric acid to a volume of 2 liters, or diluting 1.84 grams of concentrated hydrochloric acid to a volume to 3 liters, or diluting 2.45 grams of concentrated hydrochloric acid to a volume of 4 liters, or diluting 3.07 grams concentrated hydrochloric acid to a volume of 5 liters, or diluting 3.68 grams of concentrated hydrochloric acid to a volume to 6 liters.
  • the concentrated acid is preferably added to water.
  • Basic buffer solutions having a pH within the range of about 6.0 to 11.0 include by way of illustration and not limitation, solutions of: glutamate, acetate, carbonate and carbonate salts (e.g., sodium carbonate, sodium carbonate monohydrate and sodium bicarbonate), borate, phosphate and phosphate salts (e.g., monobasic sodium phosphate monohydrate and dibasic sodium phosphate), and combinations thereof.
  • the basic buffer solution is a solution of carbonate salts, phosphate salts, and combinations thereof.
  • the basic buffer is an aqueous solution that neutralizes the effect of the acidic buffer, when it is added to the homogeneous solution of the compound and first buffer, so that the nucleophilic groups on the core regain their nucleophilic character (that has been masked by the action of the acidic buffer), thus allowing the nucleophilic groups to inter-react with the electrophilic groups on the core.
  • An exemplary basic buffer is an aqueous solution of carbonate and phosphate salts.
  • This buffer may be prepared by combining a base solution with a salt solution.
  • the salt solution may be prepared by combining 34.7 g of monobasic sodium phosphate monohydrate, 49.3 g of sodium carbonate monohydrate, and sufficient water to provide a solution volume of 2 liter.
  • a 6 liter solution may be prepared by combining 104.0 g of monobasic sodium phosphate monohydrate, 147.94 g of sodium carbonate monohydrate, and sufficient water to provide 6 liter of the salt solution.
  • the basic buffer may be prepared by combining 7.2 g of sodium hydroxide with 180.0 g of water.
  • the basic buffer is typically prepared by adding the base solution as needed to the salt solution, ultimately to provide a mixture having the desired pH, e.g., a pH of 9.65 to 9.75.
  • the basic species present in the basic buffer should be sufficiently basic to neutralize the acidity provided by the acidic buffer, but should not be so nucleophilic itself that it will react substantially with the electrophilic groups on the core. For this reason, relatively "soft" bases such as carbonate and phosphate are preferred in this embodiment of the invention.
  • a three-dimensional matrix of the present invention may combine an admixture of the self-reactive compound with a first, acidic, buffer (e.g., an acid solution, e.g., a dilute hydrochloric acid solution) to form a homogeneous solution.
  • a first, acidic, buffer e.g., an acid solution, e.g., a dilute hydrochloric acid solution
  • This homogeneous solution is mixed with a second, basic, buffer (e.g., a basic solution, e.g., an aqueous solution containing phosphate and carbonate salts) whereupon the reactive groups on the core of the self-reactive compound substantially immediately inter-react with one another to form a three-dimensional matrix.
  • a basic, buffer e.g., a basic solution, e.g., an aqueous solution containing phosphate and carbonate salts
  • the reactive groups are vinyl groups such as styrene derivatives, which undergo a radical polymerization upon initiation with a redox initiator.
  • redox refers to a reactive group that is susceptible to oxidation-reduction activation.
  • vinyl refers to a reactive group that is activated by a redox initiator, and forms a radical upon reaction.
  • X, Y and Z can be the same or different vinyl groups, for example, methacrylic groups.
  • the initial environment typically will be an aqueous environment.
  • the modification of the initial environment involves the addition of a redox initiator.
  • the reactive groups undergo an oxidative coupling reaction.
  • X, Y and Z can be a halo group such as chloro, with an adjacent electron-withdrawing group on the halogen-bearing carbon (e.g., on the "L" linking group).
  • exemplary electron-withdrawing groups include nitro, aryl, and so forth.
  • the modification in the initial environment comprises a change in pH.
  • a base such as KOH
  • the self-reactive compounds then undergo a de-hydro, chloro coupling reaction, forming a double bond between the carbon atoms, as illustrated below:
  • the initial environment typically can be can be dry and sterile, or a non-basic medium.
  • the modification of the initial environment will typically comprise the addition of a base.
  • the reactive groups are photoinitiated groups.
  • the modification in the initial environment comprises exposure to ultraviolet radiation.
  • X can be an azide (-N 3 ) group and Y can be an alkyl group such as -CH(CH 3 ) 2 or vice versa. Exposure to ultraviolet radiation will then form a bond between the groups to provide for the following linkage: -NH-C(CH 3 ) 2 -CH 2 -.
  • X can be a benzophenone (-(C 6 H 4 )-C(O)- (CeH 5 )) group and Y can be an alkyl group such as -CH(CH 3 ) 2 or vice versa. Exposure to ultraviolet radiation will then form a bond between the groups to provide for the following linkage:

Abstract

The present invention provides devices or implants that comprise anti-scarring agents, methods or making such devices or implants, and methods of inhibiting fibrosis between the devices or implants and tissue surrounding the devices or implants. The present invention also provides compositions that comprise anti-fibrotic agents, and their uses in various medical applications including the prevention of surgical adhesions, treatment of inflammatory arthritis, treatment of scars and keloids, the treatment of vascular disease, and the prevention of cartilage loss.

Description

ANTI-SCARRING AGENTS, THERAPEUTIC COMPOSITIONS, AND USE
THEREOF
BACKGROUND OF THE INVENTION
Field of the Invention
This invention relates generally to devices and compositions that include a therapeutic agent (e.g., a fibrosis-inhibiting agent or an anti- infective agent), and to methods of making and using such compositions.
Description of the Related Art
The clinical function of numerous medical implants and devices is dependent upon the device being able to effectively maintain an anatomical, or surgically created, space or passageway. Unfortunately, many devices implanted in the body are subject to a "foreign body" response from the surrounding host tissues. In particular, injury to tubular _ anatomical structures (such as blood vessels, the gastrointestinal tract, the male and female reproductive tract, the urinary tract, sinuses, spinal nerve root canals, lacrimal ducts, Eustachian tubes, the auditory canal, and the respiratory tract) from surgery and/or injury created by the implantation of medical devices can lead to a well known clinical problem called "stenosis" (or narrowing). Stenosis occurs in response to trauma to the epithelial lining or the entire body tube during the procedure, including virtually any manipulation which attempts to relieve obstruction of the passageway, and is a major factor limiting the effectiveness of invasive treatments for a variety of diseases to be described later.
Stenosis (or "restenosis" if the problem recurs after an initially successful attempt to open a blocked passageway) is a form of response to injury leading to wall thickening, narrowing of the lumen, and loss of function in the tissue supplied by the particular passageway. Physical injury during an interventional procedure results in damage to epithelial lining of the tube and the smooth muscle cells (SMCs) that make up the wall. The damaged cells, particularly SMCs, release cytokines, which recruit inflammatory cells such as macrophages, lymphocytes and neutrophils (i.e., which are some of the known white blood cells) into the area. The white blood cells in turn release a variety of additional cytokines, growth factors, and tissue degrading enzymes that influence the behavior of the constituent cells of the wall (primarily epithelial cells and SMCs). Stimulation of the SMCs induces them to migrate into the inner aspect of the body passageway (often called the "intima"), proliferate and secrete an extracellar matrix - effectively filling all or parts of the lumen with reactive, fibrous scar tissue. Collectively, this creates a thickening of the intimal layer (known in some tissues as "neointimal hyperplasia") that narrows the lumen of the passageway and can be significant enough to obstruct its lumen.
Polymeric compositions, particularly those that include synthetic polymers or a combination of synthetic and naturally occurring polymers, have been used in a variety of medical applications, such as the prevention of surgical adhesions, tissue engineering, and as bioadhesive materials. U.S. Patent No. 5,162,430 describes the use of collagen- synthetic polymer conjugates prepared by covalently binding collagen to synthetic hydrophilic polymers such as various derivatives of polyethylene glycol. In a related patent, U.S. Patent No. 5,328,955, various activated forms of polyethylene glycol and various linkages are described, which can be used to produce collagen-synthetic polymer conjugates having a range of physical and chemical properties. U.S. Patent No. 5,324,775 also describes synthetic hydrophilic polyethylene glycol conjugates, but the conjugates involve naturally occurring polymers such as polysaccharides. EP 0732 109 A1 discloses a crosslinked biomaterial composition that is prepared using a hydrophobic crosslinking agent, or a mixture of hydrophilic and hydrophobic crosslinking agents. U.S. Patent No. 5,614,587 describes bioadhesives that comprise collagen that is crosslinked using a multifunctionally activated synthetic hydrophilic polymer. U.S. application Ser. No. 08/403,360, filed Mar. 14, 1995, discloses a composition useful in the prevention of surgical adhesions comprising a substrate material and an anti-adhesion binding agent, where the substrate material may comprise collagen and the binding agent may comprise at least one tissue-reactive functional group and at least one substrate-reactive functiona) group. U.S. application Ser. No. 08/476,825, filed Jun. 7, 1995, discloses bioadhesive compositions comprising collagen crosslinked using a multifunctionally activated synthetic hydrophilic polymer, as well as methods of using such compositions to effect adhesion between a first surface and a second surface, wherein at least one of the first and second surfaces may be a native tissue surface. U.S. Patent No. 5,874,500 describes a crosslinked polymer composition that comprises one component having multiple nucleophilic groups and another component having multiple electrophilic groups. Covalently bonding of the nucleophilic and electrophilic groups forms a three dimensional matrix that has a variety of medical uses including tissue adhesion, surface coatings for synthetic implants,, and drug delivery. More recent developments include the addition of a third component having either nucleophilic or electrophilic groups, as is described in U.S. Patent No. 6,458,889 to Trollsas et af. US 5,874,500, US 6,051,648 and US 6,312,725 disclose the in situ crosslinking or crosslinked polymers, in particular poly(ethylene glycol) based polymers, to produce a crosslinked composition. West and Hubbell, Biomaterials (1995) 16:1153-1156, disclose the prevention of post-operative adhesions using a photopolymerized polyethylene glycol-co-lactic acid diacrylate hydrogel and a physically crosslinked polyethylene glycol-co-polypropylene glycol hydrogel, POLOXAMER 407 (BASF Corporation, Mount Olive, NJ). Polymerizable cyanoacrylates have also been described for use as tissue adhesives (Ellis, et al., J. Otolaryngol. 19:68-72 (1990)). Two-part synthetic polymer compositions have been described that, when mixed together, form covalent bonds with one another, as well as with exposed tissue surfaces (PCTWO 97/22371 , which corresponds to U.S. application Ser. No. 08/769,806 U.S. Pat. No. 5,874,500).
BRIEF SUMMARY OF THE INVENTION
Briefly stated, in one aspect, the present invention provides compositions for delivery of selected therapeutic agents via medical implants or implantable medical devices, as well as methods for making and using these implants and devices. Within one aspect of the invention, drug- coated or drug-impregnated implants and medical devices are provided which reduce fibrosis in the tissue surrounding the device or implant, or inhibit scar development on the device/implant surface, thus enhancing the efficacy the procedure. Within various embodiments, fibrosis is inhibited by local or systemic release of specific pharmacological agents that become localized to the adjacent tissue.
The repair of tissues following a mechanical or surgical intervention involves two distinct processes: (1) regeneration (the replacement of injured cells by cells of the same type and (2) fibrosis (the replacement of injured cells by connective tissue). There are four general components to the process of fibrosis (or scarring) including: formation of new blood vessels (angiogenesis), migration and proliferation of connective tissue cells (such as fibroblasts or smooth muscle cells),- deposition of extracellular matrix (ECM), and remodeling (maturation and organization of the fibrous tissue). Within one embodiment of the invention, an implant or device is adapted to release an agent that inhibits fibrosis or regeneration through one or more of the mechanisms sited herein.
Within yet other aspects of the present invention, methods are provided for manufacturing a medical device or implant, comprising the step of coating (e.g., spraying, dipping, wrapping, or administering drug through) a medical device or implant. Additionally, the implant or medical device can be constructed so that the device itself is comprised of materials which inhibit fibrosis in or around the implant. A wide variety of medical devices and implants may be utilized within the context of the present invention, depending on the site and nature of treatment desired.
Within related aspects of the present invention, intravascular devices, gastrointestinal stents, tracheal and bronchial stents, genital urinary stents, ear and nose stents, ear ventilation tubes, intraocular implants, devices for treating hypertropic scar or keloid, vascular grafts, hemodialysis access devices, devices comprinsing a film or a mesh, glaucoma drainage devices, prosthetic heart valves or components thereof, penile implants, endotracheal or tracheostomy tubes, peritoneal dialysis catheters, central nervous system shunts or pressure monitor devices, inferior vena cava filters, gastrointestinal devices, central venous catheters, ventricular assist devices, spinal implants, implants that provide surgical adhesion barriers, and the like are provided comprising an implant or device, wherein the implant or device is in combination with an agent which inhibits fibrosis in vivo.
Within various embodiments of the invention, the implant or device is further coated with a composition or compound, which delays the onset of activity of the fibrosis-inhibiting agent for a period of time after implantation. Representative examples of such agents include heparin, PLGA/MePEG, PLA, and polyethylene glycol. Within further embodiments the fibrosis-inhibiting implant or device is activated before, during, or after deployment (e.g., an inactive agent on the device is first activated to one that reduces or inhibits an in vivo fibrotic reaction).
Within various embodiments of the invention, a device or implant is coated on one aspect, portion or surface with a composition which inhibits fibrosis, as well as being coated with a composition or compound which promotes scarring on another aspect, portion or surface of the device. Representative examples of agents that promote fibrosis and scarring include silk, wool, silica, bleomycin, neomycin, talcum powder, metallic beryllium, and copper as well as analogues and derivatives thereof. Also provided by the present invention are methods for treating patients undergoing surgical, endoscopic or minimally invasive therapies where a medical device or implant is placed as part of the procedure. As utilized herein, it should be understood that "inhibits fibrosis or stenosis" refers to a statistically significant decrease in the amount of scar tissue in or around the device or an improvement in the luminal area of the device/implant, which may or may not result in a permanent prohibition of any complications or failures of the device/implant.
The pharmaceutical agents and compositions are utilized to create novel drug-coated implants and medical devices that reduce the foreign body response to implantation and limit the growth of reactive tissue on the surface of, or around in the tissue surrounding the device, such that performance is enhanced. In many instances, the devices are used to maintain body lumens or passageways such as blood vessels, the gastrointestinal tract, the male and female reproductive tract, the urinary tract, bony foramena (e.g., sinuses, spinal nerve root canals, lacrimal ducts, Eustachian tubes, the auditory canal), and the respiratory tract, where obstruction of the device by scar tissue in the post-procedural period leads to the adverse clinical sequela or failure of the intervention. Medical devices and implants coated with selected pharmaceutical agents designed to prevent scar tissue overgrowth and preserve patency can offer significant clinical advantages over uncoated devices.
For example, in one aspect the present invention is directed to devices that comprise a medical implant and at least one of (i) an anti- scarring agent and (ii) a composition that comprises an anti-scarring agent. The agent is present so as to inhibit scarring that can otherwise occur when the implant is placed within an animal. In another aspect the present invention is directed to methods wherein both an implant and at least one of (i) an anti-scarring agent and (ii) a composition that comprises an anti- scarring agent, are placed into an animal, and the agent inhibits scarring that can otherwise occur. These and other aspects of the invention are summarized below.
Thus, in various independent aspects, the present invention provides the following: a device, comprising a medical device and an anti- scarring agent or a composition comprising an anti-scarring agent, wherein the agent inhibits scarring; a device, comprising an intravascular device and an anti-scarring agent or a composition comprising an anti-scarring agent, wherein the agent inhibits scarring; a device, comprising a a gastrointestinal stent and an anti-scarring agent or a composition comprising an anti- scarring agent, wherein the agent inhibits scarring; a device, comprising a tracheal and bronchial stent and an anti-scarring agent or a composition comprising an anti-scarring agent, wherein the agent inhibits scarring; a device, comprising a genital urinary stent and an anti-scarring agent or a composition comprising an anti-scarring agent, wherein the agent inhibits scarring; a device, comprising an ear and nose stent and an anti-scarring agent or a composition comprising an anti-scarring agent, wherein the agent inhibits scarring; a device, comprising an ear ventilation tube and an anti- scarring agent or a composition comprising an anti-scarring agent, wherein the agent inhibits scarring; a device, comprising an intraocular implant and an anti-scarring agent or a composition comprising an anti-scarring agent, wherein the agent inhibits scarring; a device, comprising a medical device for treating hypertropic scar or keloid and an anti-scarring agent or a composition comprising an anti-scarring agent, wherein the agent inhibits scarring; device, comprising a vascular graft and an anti-scarring agent or a composition comprising an anti-scarring agent, wherein the agent inhibits scarring; a device, comprising a hemodialysis access device and an anti- scarring agent or a composition comprising an anti-scarring agent, wherein the agent inhibits scarring; a device, comprising a device comprising a film or a mesh and an anti-scarring agent or a composition comprising an anti- scarring agent, wherein the agent inhibits scarring; a device, comprising glaucoma drainage device and an anti-scarring agent or a composition comprising an anti-scarring agent, wherein the agent inhibits scarring; a device, comprising prosthetic heart valve or component thereof and an anti- scarring agent or a composition comprising an anti-scarring agent, wherein the agent inhibits scarring; a device, comprising a penile implant and an anti-scarring agent or a composition comprising an anti-scarring agent, wherein the agent inhibits scarring; a device, comprising an endotracheal or tracheostomy tube and an anti-scarring agent or a composition comprising an anti-scarring agent, wherein the agent inhibits scarring; a device, comprising a peritoneal dialysis catheter and an anti-scarring agent or a composition comprising an anti-scarring agent, wherein the agent inhibits scarring; a device, comprising a central nervous system shunt or pressure monitor device and an anti-scarring agent or a composition comprising an anti-scarring agent, wherein the agent inhibits scarring; a device, comprising inferior vena cava filter and an anti-scarring agent or a composition comprising an anti-scarring agent, wherein the agent inhibits scarring; a device, comprising a gastrointestinal device and an anti-scarring agentor a composition comprising an anti-scarring agent, wherein the agent inhibits scarring; a device, comprising a central venous catheter and an anti- scarring agent or a composition comprising an anti-scarring agent, wherein the agent inhibits scarring; a device, comprising a ventricular assist device and an anti-scarring agent or a composition comprising an anti-scarring agent, wherein the agent inhibits scarring; a device, comprising a spinal implant and an anti-scarring agent or a composition comprising an anti- scarring agent, wherein the agent inhibits scarring; a device, comprising an implant that provides a surgical adhesion barrier and an anti-scarring agent or a composition comprising an anti-scarring agent, wherein the agent inhibits scarring. These and other devices are described in more detail herein.
In additional aspects, for each of the aforementioned devices combined with each of the anti-fibrotic agents disclosed herein, it is, for each combination, independently disclosed that the anti-fibrotic agent may be present in a composition along with a polymer. In one embodiment of this aspect, the polymer is biodegradable. In another embodiment of this aspect, the polymer is non-biodegradable. Other features and characteristics of the polymer, which may serve to describe the present invention for every combination of device and agent described above, are set forth in greater detail herein.
In addition to devices, the present invention also provides methods. For example, in additional aspects of the present invention, for each of the aforementioned devices, and for each of the aforementioned combinations of the devices with the anti-scarring agents, the present invention provides methods whereby a specified device is implanted into an animal, and a specified agent associated with the device inhibits scarring that can otherwise occur. Each of the devices identified herein may be a "specified device", and each of the anti-scarring agents identified herein may be an "anti-scarring agent", where the present invention provides, in independent embodiments, for each possible combination of the device and the agent.
The agent may be associated with the device prior to the device being placed within the animal. For example, the agent (or composition comprising the agent) may be coated onto an implant, and the resulting device then placed within the animal. In addition, or alternatively, the agent may be independently placed within the animal in the vicinity of where the device is to be, or is being, placed within the animal. For example, the agent may be sprayed or otherwise placed onto the tissue that will be contacting the medical implant or may otherwise undergo scarring. To this end, the present invention provides, in independent aspects: a method for inhibiting scarring comprising placing a medical device and an anti-scarring agent or a composition comprising an anti-scarring agent into an animal host, wherein the agent inhibits scarring; a method for inhibiting scarring comprising placing an intravascular device and an anti-scarring agent or a composition comprising an anti-scarring agent into an animal host, wherein the agent inhibits scarring; a method for inhibiting scarring comprising placing a gastrointestinal stent and an anti-scarring agent or a composition comprising an anti-scarring agent into an animal host, wherein the agent inhibits scarring; a method for inhibiting scarring comprising placing a tracheal and bronchial stent and an anti-scarring agent or a composition comprising an anti-scarring agent into an animal host, wherein the agent inhibits scarring; a method for inhibiting scarring comprising placing a genital urinary stent and an anti-scarring agent or a composition comprising an anti-scarring agent into an animal host, wherein the agent inhibits scarring; a method for inhibiting scarring comprising placing an ear and nose stent and an anti-scarring agent or a composition comprising an anti-scarring agent into an animal host, wherein the agent inhibits scarring; a method for inhibiting scarring comprising placing an ear ventilation tube.and an anti-scarring agent or a composition comprising an anti-scarring agent into an animal host, wherein the agent inhibits scarring; a method for inhibiting scarring comprising placing an intraocular implant and an anti- - scarring agent or a composition comprising an anti-scarring agent into an animal host, wherein the agent inhibits scarring; a method for inhibiting scarring comprising placing a medical device for treating hypertropic scar or keloid and an anti-scarring agent or a composition comprising an anti- scarring agent into an animal host, wherein the agent inhibits scarring; a method for inhibiting scarring comprising placing a vascular graft and an anti-scarring agent or a composition comprising an anti-scarring agent into an animal host, wherein the agent inhibits scarring; a method for inhibiting scarring comprising placing a hemodialysis access device and an anti- scarring agent or a composition comprising an anti-scarring agent into an animal host, wherein the agent inhibits scarring; a method for inhibiting scarring comprising placing a medical device comprising a film or a mesh and an anti-scarring agent or a composition comprising an anti-scarring agent into an animal host, wherein the agent inhibits scarring; a method for inhibiting scarring comprising placing a glaucoma drainage device and an anti-scarring agent or a composition comprising an anti-scarring agent into an animal host, wherein the agent inhibits scarring; a method for inhibiting scarring comprising placing prosthetic heart valve or component thereof and an anti-scarring agent or a composition comprising an anti-scarring agent into an animal host, wherein the agent inhibits scarring; a method for inhibiting scarring comprising placing a penile implant and an anti-scarring agent or a composition comprising an anti-scarring agent into an animal host, wherein the agent inhibits scarring; a method for inhibiting scarring comprising placing an endotracheal or tracheostomy tube and an anti- scarring agent or a composition comprising an anti-scarring agent into an animal host, wherein the agent inhibits scarring; a method for inhibiting scarring comprising placing a peritoneal dialysis catheter and an anti- scarring agent or a composition comprising an anti-scarring agent into an animal host, wherein the agent inhibits scarring; a method for inhibiting scarring comprising placing a central nervous system shunt or pressure monitor device and an anti-scarring agent or a composition comprising an anti-scarring agent into an animal host, wherein the agent inhibits scarring; a method for inhibiting scarring comprising placing inferioOr vena cava filter and an anti-scarring agent or a composition comprising an anti-scarring agent into an animal host, wherein the agent inhibits scarring; a method for inhibiting scarring comprising placing a gastrointestinal device and an anti- scarring agent or a composition comprising an anti-scarring agent into an animal host, wherein the agent inhibits scarring; a method for inhibiting scarring comprising placing a central venous catheter and an anti-scarring agent or a composition comprising an anti-scarring agent into an animal host, wherein the agent inhibits scarring; a method for inhibiting scarring comprising placing a ventricular assist device and an anti-scarring agent or a composition comprising an anti-scarring agent into an animal host, wherein the agent inhibits scarring; a method for inhibiting scarring comprising placing a spinal implant and an anti-scarring agent or a composition comprising an anti-scarring agent into an animal host, wherein the agent inhibits scarring; a method for inhibiting scarring comprising placing an implant that provides surgical barrier and an anti-scarring agent or a composition comprising an anti-scarring agent into an animal host, wherein the agent inhibits scarring.
In additional aspects, for each of the aforementioned methods used in combination with each of the aforementioned agents, it is, for each combination, independently disclosed that the agent may be present in a composition along with a polymer. In one embodiment of this aspect, the polymer is biodegradable. In another embodiment of this aspect, the polymer is non-biodegradable. Other features and characteristics of the polymer, which may serve to describe the present invention for every combination of device and agent described above, are set forth in greater detail herein.
In other aspects, the present invention provides compositions that contain both an anti-fibrotic agent and either a polymer or a pre- polymer, i.e., a compound that forms a polymer. In one embodiment, these compositions are formed in-situ when precursors thereof are delivered to a site in the body, or a site on an implant. For example, the compositions of the invention include the crosslinked reaction product that forms when two compounds (a multifunctional polynucleophilic compound and a multifunctional polyelectrophilic compound) are delivered to a site in a host (in other words, a patient) in the presence of an anti-fibrotic agent. However, the compositions of the invention also include a mixture of anti-fibrotic agent and a polymer, where the composition can be delivered to a site in a patient's body to achieve beneficial affects, e.g., the beneficial affects described herein.
In some instances, the polymers themselves are useful in various methods, including the prevention of surgical adhesions.
In another aspect, the present invention provides methods for treating and/or preventing surgical adhesions. The surgical adhesions can be the result of, for example, spinal or neurosurgical procedures, of gynecological procedures, of abdominal procedures, of cardiac procedures, of orthopedic procedures, of reconstructive procedures, and cosmetic procedures.
In another aspect, the present invention provides methods for treating or preventing inflammatory arthritis, such as osteoarthritis and rheumatoid arthritis. The method includes delivering to patient in need thereof an anti-fibrotic agent, optionally with a polymer.
In another aspect, the present invention provides for the prevention of cartilage loss as can occur, for example after a joint injury. The method includes delivering to the joint of the patient in need therof an anti-fibrotic agent, optionally with a polymer.
In another aspect, the present invention provides for treating hypertrophic scars and keloids. The method includes delivering to the scar or keloid of the patient in need thereof an anti-fibrotic agent, optionally with a polymer.
In another aspect, the present invention provides a method for the treatment of vascular disease, e.g., stenosis, restenosis or atherosclerosis. The method includes the perivascular delivery of an anti- fibrotic agent.
In one aspect, the present invention provides a method for implanting a medical device comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with i) an anti- fibrotic agent, ii) an anti-infective agent, iii) a polymer; iv) a composition comprising an anti-fibrotic agent and a polymer, v) a composition comprising an anti-infective agent and a polymer, or vi) a composition comprising an anti-fibrotic agent, an anti-infective agent and a polymer, and (b) implanting the medical device into the host.
Optionally, in separate aspects, the invention provides: a method for implanting a medical device comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with an anti-fibrotic agent, and (b) implanting the medical device into the host; a method for implanting a medical device comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with an anti-infective agent, and (b) implanting the medical device into the host; a method for implanting a medical device comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with a polymer; and (b) implanting the medical device into the host; a method for implanting a medical device comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with a composition comprising an anti-fibrotic agent and a polymer, and (b) implanting the medical device into the host; a method for implanting a medical device comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with a composition comprising an anti-infective agent and a polymer, and (b) implanting the medical device into the host; and a method for implanting a medical device comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with a composition comprising an.anti-fibrotic .. agent, an anti-infective agent and a polymer, and (b) implanting the medical device into the host.
In each of the aforementioned devices, compositions, methods of making the aforementioned devices or compositions, and methods of using the aforementioned devices or compostions, the present invention provides that the anti-fibrotic agent may be one or more of the following: 1) an anti-fibrotic agent that inhibits cell regeneration, 2) an anti-fibrotic agent that inhibits angiogenesis, 3) an anti-fibrotic agent that inhibits fibroblast migration, 4) an anti-fibrotic agent that inhibits fibroblast proliferation, 5) an anti-fibrotic agent that inhibits deposition of extracellular matrix, 6) an anti- fibrotic agent inhibits tissue remodeling, 7) an adensosine A2A receptor antagonist, 8) an AKT inhibitor, 9) an alpha 2 integrin antagonist, wherein the alpha 2 integrin antagonist is Pharmaprojects No. 5754 (Merck KgaA), 10) an alpha 4 integrin antagonist, 11) an alpha 7 nicotinic receptor agonist, 12) an angiogenesis inhibitor selected from the group consisting of AG- 12,958 (Pfizer), ATN-161 (Attenuon LLC), neovastat, an angiogenesis inhibitor from Jerina AG (Germany), NM-3 (Mercian), VGA-1155 (Taisho), FCE-26644 (Pfizer), FCE-26950 (Pfizer), FPMA (Meiji Daries), FR-111142 (Fujisawa), GGTI-298, GM-1306 (Ligand), GPA-1734 (Novartis), NNC-47- 0011 (Novo Nordisk), herbamycin (Nippon Kayaku), lenalidomide (Celegene), IP-10 (NIH), ABT-828 (Abbott), KIN-841 (Tokushima University, Japan), SF-1126 (Semafore Pharmaceuticals), laminin technology (NIH), CHIR-258 (Chiron), NVP-AEW541 (Novartis), NVP-AEW541 (Novartis), Vt16907 (Alchemia), OXI-8007 (Oxigene), EG-3306 (Ark Therapeutics), Maspin (Arriva), ABT-567 (Abbott), PPI-2458 (Praecis Pharmaceuticals), CC-5079, CC-4089 (Celgene), HIF-1 alpha inhibitors (Xenova), S-247 (Pfizer), AP-23573 (Ariad), AZD-9935 (Astra Zeneca), mebendazole (Introgen Therapeutics), MetAP-2 inhibitors (GlaxoSmithKline), AG-615 (Angiogene Pharmaceuticals), Tie-2 antagonists (Hybrigenics), NC-381 , CYC-381, NC-169, NC-219, NC-383, NC-384, NC-407 (Lorus Therapeutics), ATN-224 (Attenuon), ON-01370 (Onconova), Vitronectin antagonists (Amgen), SDX-103 (Salmedix), Vitronectin antagonists (Shire), CHP (Riemser), TEK (Amgen), Anecortave acetate (Alcon), T46.2 (Matrix Therapeutics), HG-2 (Heptagen), TEM antagonists (Genzyme), Oxi-4500 (Oxigene), ATN-161 (Attenuon), WX-293 (Wilex), M-2025 (Metris Therapeutics), Alphastatin (BioActa), YH-16 (Yantai Rongchang), BIBF- 1120 (Boehringer Ingelheim), BAY-57-9352 (Bayer), AS-1404 (Cancer Research Technology), SC-77964 (Pfizer), glycomimetics (BioTie Therapies), TIE-2 Inhibitors (Ontogen), DIMI, Octamer (Octamer), ABR- 215050 (Active Biotech), ABT-518 (Abbott), KDR inhibitors (Abbott), BSF- 466895 (Abbott), SCH-221153 (Schering-Plough), DAC:antiangiogenic (ConjuChem), TFPI (EntreMed), AZD-2171 (Astra-Zenaca), CDC-394 (Celgene), LY290293 (EIi Lilly), IDN-5390 (Indena), Kdr Kinase Inhibitors (Merck), CT-113020, CT-116433, CT-116563, CT-31890, CT-32228) (Cell Therapeutics), A-299620 (Abbott), TWEAK Inhibitor (Amgen), VEGF modulators (Johnson and Johnson), Tum-N53, tumstatin (Genzyme), Thios- 1 , Thios-2 (Thios Pharmaceuticals), MV-6401 (Miravant Medical Technologies), Spisulosine (PharmaMar), CEP-7055 (Cephalon), AUV-201 (Auvation), LM-609 (EIi Lilly), SKF-106615 (AnorMED), Oglufanide disodium (Cytran), BW-114 (Pharminox), Calreticulin (NIH), WX-678 (Wilex), SD-7784 (Pfizer), WX-UK1 (Wilex), SH-268 (Schering AG), 2-Me-PGA (Celgene), S- 137 (Pfizer), ZD-6126 (Angiogene Pharmaceuticals), SG-292 (SignalGen), Benefin (Lane Labs), A6, A36 (Angstrom), SB-2723005 (GlaxoSmithKline), SC-7 (Cell Therapeutics), ZEN-014 (AEterna Zentaris), 2-methoxyestradiol (EntreMed), NK-130119 (Nippon Kayaku), CC-10004 (Celgene), AVE- 8062A (Ajinomoto), Tacedinaline (Pfizer), Actinonin (Tokyo Metropolitan Institute of Medical Science), Lenalidomide (Celgene), VGA-1155, BTO-956 (SRI International), ER-68203-00 (Eisai), CT-6685 (UCB), JKC-362 (Phoenix Pharmaceuticals), DMI-3798 (DMI Biosciences, Angiomate (Ipsen), ZD-6474 (AstraZeneca), CEP-5214 (Cephalon), Canstatin (Genzyme), NM-3 (Mercian), Oridigm (MediQuest Therapeutics), Exherin (Adherex), BLS-0597 (Boston Life Sciences), PTC-299 (PTC Therapeutics), NPI-2358 (Nereus Pharmaceuticals), CGP-79787 (Novartis), AEE-788 (Novartis), CKD-732 (Chong Kun Dang), CP-564959 (OSI Pharmaceuticals), CM-101 (CarboMed), CT-2584, CT3501 (Cell Therapeutics), combretastatin and analogues and derivatives thereof (Oxigene), Rebimastat (Bristol-Meyers Squibb), Dextrin 2-sulfate (ML Laboratories), Cilengitide (Merk KGaA), NSC-706704 (Pharminox), KRN- 951 (Kirin Brewery), Ukrain, NSC-631570 (Nowicky Pharma), Tecogalan sodium (Daiichi Pharmaceutical), Tz-93 (Tsumura), TBC-1635 (Encysive Pharmaceuticals), TAN-1120 (Takeda), Semaxanib (Pfizer), BDI-7800 (Biopharmacopae), SD-186, SD-983 (Bristol-Meyers Squibb), SB-223245 (GlaxoSmithKline), SC-236 (Pfizer), RWJ-590973 (Johnson and Johnson), ILX-1850 (Genzyme), SC-68488, S-836 (Pfizer), CG-55069-11 (CuraGen), Ki-23057 (Kirin Brewery), CCX-700 (Chemoentryx), Pegaptanib octasodium (Giled Sciences), ANGIOCOL (available from Biostratum Inc.), or an analogue or derivative thereof, 13) an apoptosis antagonist, 14) an apoptosis activator, 15) a beta 1 integrin antagonist, 16) a beta tubulin inhibitor, 17) a blocker of enzyme production in Hepatitis C, 18) a Bruton's tyrosine kinase inhibitor, 19) a calcineurin inhibitor, 20) a caspase 3 inhibitor, 21) a CC chemokine receptor antagonist, 22) a cell cycle inhibitor selected from the group consisting of SNS-595 (Sunesis), synthadotin, KRX- 0403, homoharringtonine, and an analogue or derivative thereof, 23) a cathepsin B inhibitor, 24) a cathepsin K inhibitor, wherein the cathepsin K inhibitor is 462795 (GlaxoSmithKline), INPL-022-D6 (Amura Therapeutics), or an analogue or derivative thereof, 25) a cathepsin L inhibitor, 26) a CD40 antagonist, 27) a chemokine receptor agonist, 28) a chymase inhibitor, 29) a collagenase antagonist, 30) a CXCR antagonist, 31) a cyclin dependent kinase inhibitor selected from the group consisting of a CDK-1 inhibitor, a CDK-2 inhibitor, a CDK- 4 inhibitor, a CDK-6 inhibitor, a CAK1 inhibitor from GPC Biotech and Bristol-Myers Squibb, RGB-286199 (GPC Biotech), an anticancer agent from Astex Technology, a CAK1 inhibitor from GPC Biotech, a CDK inhibitor from Sanofi-Aventis, a CDK1/CDK2 inhibitor from Amgen, a CDK2 inhibitor from SUGEN-2 (Pfizer), a hearing loss therapy agent (Sound Pharmaceuticals), PD-0332991 (Pfizer), RGB-286199 (GPC Biotech), Ro-0505124 (Hoffmann-La Roche), a Ser/Thr kinase inhibitor from Lilly (EIi Lilly), CVT-2584 (CAS No. 199986-75-9) (CV Therapeutics), CGP 74514A, bohemine, olomoucine (CAS No. 101622-51-9), indole-3-carbinol (CAS No. 700-06-1), and an analogue or derivative thereof, 32) a cyclooxygenase 1 inhibitor, 33) a DHFR inhibitor, 34) a dual integrin inhibitor, 35) an elastase inhibitor, 36) an elongation factor-1 alpha inhibitor, 37) an endothelial growth factor antagonist, 38) an endothelial growth factor receptor kinase inhibitor selected from the group consisting of sorafenib tosylate (Bayer), AAL-993 (Novartis), ABP-309 (Novartis), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EXEL-2880 (Exelixis), GW-654652 (GlaxoSmithKline), lavendustin A (CAS No. 125697-92-9), a KDR inhibitor from LG Life Sciences, CT-6685 and CT-6729 (UCB), KRN-633 and KRN-951 (Kirin Brewery), OSI-930 (OSI Pharmaceuticals), SP-5.2 (Supratek Pharma), SU- 11657 (Pfizer), a Tie-2 antagonist (Hybrigenics), SU 1498 (a VEGF-R inhibitor), a VEGFR-2 kinase inhibitor (Bristol-Myers Squibb), XL-647 (Exelixis), a KDR inhibitor from Abbott Laboratories, sorafenib tosylate, and an analogue or derivative thereof, 39) an endotoxin antagonist, 40) an epothilone and tubulin binder, 41) an estrogen receptor antagonist, 42) an FGF inhibitor, 43) a famexyl transferase inhibitor, 44) a farnesyltransferase inhibitor selected from the group of A-197574 (Abbott), a farnesyltransferase inhibitor from Servier, FPTIII (Strathclyde Institute for Drug R), LB-42908 (LG Life Sciences), Pharmaprojects No. 5063 (Genzyme), Pharmaprojects No. 5597 (Ipsen), Yissum Project No. B-1055 (Yissum), and an analogue or derivative thereof, 45) an FLT-3 kinase inhibitor, 46a) an FGF receptor kinase inhibitor, 47) a fibrinogen antagonist selected from the group consisting of AUV-201 (Auvation), MG-13926 (Sanofi-Aventis), plasminogen activator (CAS No. 105913-11-9) (from Sanofi-Aventis or UCB), plasminogen activator-2 (tPA-2) (Sanofi-Aventis), pro-urokinase (CAS Np. 82657-92-9) (Sanofi-Aventis), mevastatin, and an analogue or derivative thereof, 48) a heat shock protein 90 antagonist selected from the group consisting of SRN-005 (Sirenade), geldanamycin, NSC-33050 (17- allylaminogeldanamycin; 17-AAG), 17-dimethylaminoethylamino~17- demethoxy-geldanamycin (17-DMAG), rifabutin (rifamycin XIV, 1',4- didehydro-1-deoxy-1 ,4-dihydro-5'-(2-methylpropyl)-1-oxo-), radicicol from Humicola fuscoatra (CAS No. 12772-57-5), and an analogue or derivative thereof, 49) a histone deacetylase inhibitor, 50) an HMGCoA reductase inhibitor selected from the group consisting of an atherosclerosis therapeutic from Lipid Sciences, ATI-16000 (ARYx Therapeutics), KS-01-019 (Kos Pharmaceuticals), Pharmaprojects No. 2197 (Sanofi-Aventi), RP 61969 (Sanofi-Aventis), cerivastatin Na (CAS No. 143201-11-0), and an analogue or derivative thereof, 51) an ICAM inhibitor, 52) an IL, ICE and IRAK antagonist, wherein the antagonist is a CJ-14877, CP-424174 (Pfizer), NF- 61 (Negma-Lerads), and an analogue or derivative thereof, 53) an IL-2 inhibitor, 54) an immunosuppressant selected from the group consisting of teriflunomide (Sanofi Aventis), chlorsulfaquinoxalone (NSC-339004), chlorsulfaquinoxalone sulfate, CS-712 (Sankyo), ismomultin alfa (CAS No. 457913-93-8) (Akzo Nobel), antiallergics from GenPat77, antiinflammatories or AT-005 (Androclus Therapeutics), autoimmune disease therapeutics from EpiVax, BN-007 (Bone), budesonide (CAS No. 51333-22- 3) (MAP Pharmaceuticals), CO-14 (Genzyme), edratide (CAS No. 433922- 67-9) (Teva), EP-314 (Enanta), eprovafen (CAS No. 101335-99-3) (Sanofi- Aventis), HWA-131 (CAS No. 118788-41-3) (Sanofi-Aventis), immunomodulators from MerLion Pharmaceuticals, immunosuppressives from Alchemia, IPL-12 (Inflazyme), MDL-9563 (CAS No. 27086-86-8) (Sanofi-Aventis), Pharmaprojects No. 2330 (Sanofi-Aventis), Pharmaprojects No. 6426 (Abgenix), PXS-25 (Pharmaxis), rosmarinic acid (CAS No. 20283-92-5) (Sanofi-Aventis), RP 42927 or RP 54745 (CAS No. 135330-08-4) (Sanofi-Aventis), SGN-35 (Seattle Genetics), ST-1959 (Sigma-Tau), type I diabetes therapy from SYNX Pharma, U NIL-88 (Debiopharm), VP-025 (Vasogen), VR-694 (Vectura), PRTX-001 (Protalex), and an analogue or derivative thereof, 55) an IMPDH (inosine monophosphate), 56) an integrin antagonist, 57) an interleukin antagonist, 58) an inhibitor of type III receptor tyrosine kinase, 59) an irreversible inhibitor of enzyme methionine aminopeptidase type 2, 60) an isozyme selective delta protein kinase C inhibitor, 61) a JAK3 enzyme inhibitor, 62) a JNK inhibitor, 63) a kinase inhibitor, 64) a kinesin antagonist, 65) a leukotriene inhibitor and antagonist selected from the group consisting of ambicromil (CAS No. 58805-38-2) (Sanofi-Aventis), amelubant (CAS No. 346735-24-8) (Boehringer Ingelheim), DW-1141 (Dong Wha), ebselen (Daiichi Pharmaceutical), ibudilast (Kyorin), leucotriene inhibitors from Sanofi-Aventis, lymphotoxin-beta receptor (LT-β) from Biogen Idee, Pharmaprojects No. 1535 and 2728 (CAS No. 119340-33-9) (Sanofi- Aventis), R-112 (Rigel), Rev-5367 (CAS No. 92532-05-3) (Sanofi-Aventis), RG-14893 (CAS No. 141835-49-6) (Sanofi-Aventis), RG-5901-A (CAS No. 101910-24-1), 92532-23-5, RP 66153 (CAS No. 142422-79-5), RP 66364 (CAS No.186912-92-5), RP 69698 (CAS No. 141748-00-7) (Sanofi-Aventis), SC-411930 (Pfizer), SC-41930 (CAS No. 120072-59-5) (Pfizer), SC-50605 (CAS No. 138828-39-4) (Pfizer), SC-51146 (CAS No. 141059-52-1), SC- 53228 (CAS No. 153633-01-3) (Pfizer), spaglumic acid (ZY-15106) (CAS No.3106-85-2), 80619-64-3 (Novartis), tipredane (CAS No. 85197-77-9) (Bristol-Myers Squibb), U-75302 (CAS No. 119477-85-9) (Pfizer), and analogue or derivative thereof, 66) a MAP kinase inhibitor, 67) a matrix metalloproteinase inhibitor, 68) an MCP-CCR2 inhibitor, 69) an mTOR inhibitor, 70) an mTOR kinase inhibitor,71) a microtubule inhibitor selected from the group consisting of antibody-maytansinoid conjugates from Biogen Idee, colchicines (MantiCore Pharmaceuticals), anticancer immunoconjugates from Johnson & Johnson, DIME from Octamer, gni-1f (GNI), huC242-DM4, huMy9-6-DM1 (ImmunoGen), IDN-5404 (Indena), IMO-098, IMOderm (Imotep), mebendazole (Introgen Therapeutics), microtubule poisons from Cambridge Enterprise, paclitaxel such as LOTAX from Aphios (CAS No. 33069-62-4), Genexol-PM from Samyang, Pharmaprojects No. 6383 (Tapestry Pharmaceuticals), RPR-112378 (Sanofi-Aventis), SGN-75 (Seattle Genetics), SPL-7435 (Starpharma), SSR- 250411 (Sanofi-Aventis), trastuzumab-DM1 (Genentech), vinorelbine, dolastatin 15 (CAS No. 123884-00-4), vincamine, and an analogue or derivative thereof, 72) an MIF inhibitor, 73) an MMP inhibitor, 74) a neurokinin (NK) antagonist selected from the group consisting of anthrotainin (CAS No. 148084-40-6) (Sanofi-Aventis), an IBS thereapeutic from ArQuIe, MDL-105212A (CAS No. 167261-60-1) (Ssanofi-Aventis), Pharmaprojects No. 2744, 3258 (CAS No. 139167-47-8) 4006, 4201, or 5986 (Sanofi-Aventis), RP 67580 (CAS No. 135911-02-3), SR-144190 (CAS No. 201152-86-5), SSR-240600, SSR-241586 (Sanofi-Aventis), TKA-457 (Novartis), vestipitant mesylate (CAS No. 334476-64-1) (GlaxoSmithKline), Win-64821 (Sanofi-Aventis), PRX-96026 (Predix Pharmaceuticals), and an analogue or derivative thereof, 75) an NF kappa B inhibitor selected from the group consisting of emodin (CAS No. 518-82-1), AVE-0545 or AVE- 0547 (Sanofi-Aventis), bortezomib (CAS No. 179324-69-7) (Millennium Pharmaceuticals), dexanabinol (CAS No. 112924-45-5) (Pharmos), dexlipotam (Viatris), Pharmaprojects No. 6283 (INDRA) (OXiGENE), IPL- 576092 (CAS No. 137571-30-3) (\nf\azyme), NFKB decoy (Corgentech), NFKB decoy oligo (AnGes MG), NFKB's from Ariad, osteoporosis treatments or S5 (F005) from Fulcrum Pharmaceuticals, P61 (Phytopharm), R-flurbiprofen (CAS No. 5104-49-4) (Encore Pharmaceuticals), Bay 11- 7085, and an analogue or derivative thereof, 76) a nitric oxide agonist, 77) an ornithine decarboxylase inhibitor, 78) a p38 MAP kinase inhibitor selected from the group consisting of AZD-6703 (AstraZeneca), JX-401 (Jexys Pharmaceuticals), BMS-2 (Bristol-Myers Squibb), a p38 MAP kinase inhibitor from Novartis, a p38-alpha MAP kinase inhibitor from Amphora, Pharmaprojects No. 5704 (Pharmacopeia), SKF86002 (CAS No. 72873-74- 6), RPR-200765A (Sanofi-Aventis), SD-282 (Johnson & Johnson), TAK-715 (Takeda), and an analogue or derivative thereof, 79) a palmitoyl-protein thioesterase inhibitor, 80) a PDGF receptor kinase inhibitor selected from the group consisting of AAL-993, AMN-107, or ABP-309 (Novartis), AMG- 706 (Amgen), BAY-57-9352 (Bayer), CDP-860 (UCB), E-7080 (Eisai), imatinib (CAS No. 152459-95-5) (Novartis), OSI-930 (OSI Pharmaceuticals), RPR-127963E (Sanofi-Aventis), RWJ-540973 (Johnson & Johnson), sorafenib tosylate (Bayer), SlM 1657 (Pfizer), tandutinib (CAS No. 387867- 13-2) (Millennium Pharmaceuticals), vatalanib (Novartis), ZK-CDK (Schering AG), and an analogue or derivative thereof, 81) a peroxisome proliferators- activated receptor agonist selected from the group consisting of (-)- halofenate (Metabolex), AMG-131 (Amgen), antidiabetics from Japan Tobacco, AZD-4619, AZD-8450, AZD-8677 (AstraZeneca), DRF-10945, balaglitazone (Dr Reddy's), CS-00088, CS-00098 (Chipscreen Biosciences), E-3030 (Eisai), etalocib (CAS No. 161172-51-6) (EIi Lilly), GSK-641597 (Ligand), GSK-677954 (GlaxoSmithKline), GW-409544 (Ligand), GW- 590735 (GlaxoSmithKline), K-111 (Hoffmann-La Roche), LY-518674 (EIi Lilly), LY-674 (Ligand), LY-929 (Ligand), MC-3001 , MC-3002 (MaxoCore Pharmaceuticals), metformin HCI + pioglitazone (CAS No. 1115-70-4 and 112529-15-4), ACTOPLUS MET from Andrx), muraglitazar (CAS No. 331741-94-7) (Bristol-Myers Squibb), naveglitazar (Ligand), oleoylethanolamide (Kadmus Pharmaceuticals), ONO-5129, pioglitazone hydrochloride (CAS No. 111025-46-8 and 112529-15-4) (Takeda), PLX-204 (Plexxikon), PPAR agonists from Genfit, PPAR delta agonists from EIi Lilly, PPAR-alpha agonists from CrystalGenomics, PPAR-gamma modulators and PPAR-β modulators from CareX, rosiglitazone maleate (CAS No. 122320- 73-4 or 155141-29-0) (GlaxoSmithKline), rosiglitazone maleate/glimepir (CAS No. 155141-29-0 and 93479-97-1), AVANDARYL, rosiglitazone maleate/metformin extend (CAS No. 155141-29-0 and 657-24-9), AVANDAMET, rosiglitazone maleate+metformin, AVANDAMET (GlaxoSmithKline), tesaglitazar (AstraZeneca), LBM642, WY-14,643 (CAS No. 50892-23-4), GW7647, fenofibric acid (CAS No. 42017-89-0), MCC-555 (CAS No. 161600-01-7), GW9662, GW1929, GW501516, L-165,041 (CAS No. 79558-09-1), and an analogue or derivative thereof, 82) a phosphatase inhibitor, 83) a phosphodiesterase (PDE) inhibitor selected from the group consisting of avanafil (Tanabe Seiyaku), dasantafil (CAS No. 569351-91-3) (Schering-Plough), A-906119 (CAS No. 134072-58-5), DL-850 (Sanofi- Aventis), GRC-3015, GRC-3566, GRC-3886 (Glenmark), HWA-153 (CAS No. 56395-66-5) (Sanofi-Aventis), hydroxypumafentrine (Altana), IBFB- 130011 , IBFB-14-016, IBFB-140301 , IBFB-150007, IBFB-211913 (IBFB Pharma), L-826141 (Merck & Co), medorinone (CAS No. 88296-61-1) (Sanofi-Aventis), MEM-1917 (Memory Pharmaceuticals), ND-1251 (Neuro3d), PDE inhibitors from ICOS, PDE IV inhibitors from Memory Pharmaceuticals and CrystalGenomics, Pharmaprojects No. 2742 and 6141 (Sanofi-Aventis), QAD-171 (Novartis), RHC-2963 (CAS No. 76993-12-9 and 76993-14-1), RPR-117658, RPR-122818 derivatives, SR-24870 , and RPR- 132294 (Sanofi-Aventis), SK-350 (ln2Gen), stroke therapy agents from deCODE Genetics, TAS-203 (Taiho), tofimilast (CAS No. 185954-27-2) (Pfizer), UK-371800 (Pfizer), WIN-65579 (CAS No. 158020-82-7) (Sanofi- Aventis), IBFB-130020 (IBFB Pharma), OPC-6535 (CAS No. 145739-56-6) (Otsuka), theobromine (CAS No. 83-67-0), papverine hydrochloride (CAS No. 61-25-6), quercetin dehydrate (CAS No. 6151-25-3), YM 976 (CAS No. 191219-80-4), irsogladine (CAS No. 57381-26-7), a phosphodiesterase 111 inhibitor, enoximone, a phosphodiesterase IV inhibitor, fosfosal, Atopik (Barrier Therapeutics), triflusal, a phosphodiesterase V inhibitor, and an analogue or derivative thereof, 84) a PKC inhibitor, 85) a platelet activating factor antagonist, 86) a platelet-derived growth factor receptor kinase inhibitor, 87) a prolyl hydroxylase inhibitor, 88) a polymorphonuclear neutrophil inhibitor, 89) a protein kinase B inhibitor, 90) a protein kinase C stimulant, 91) a purine nucleoside analogue, 92) a purinoreceptor P2X antagonist, 93) a Raf kinase inhibitor, 94) a reversible inhibitor of ErbB1 and ErbB2, 95) a ribonucleoside triphosphate reductase inhibitor, 96) an SDF-1 antagonist, 97) a sheddase inhibitor, 98) an SRC inhibitor, 99) a stromelysin inhibitor, 100) an Syk kinase inhibitor, 101) a telomerase inhibitor, 102) a TGF beta inhibitor selected from the group consisting of pirfenidone (CAS No. 53179-13-8) (MARNAC), tranilast (CAS No. 53902-12-8) (Kissei), IN- 1130 (ln2Gen), mannose-6-phosphate (BTG), TGF-β antagonists from Inflazyme (Pharmaprojects No. 6075), TGF-β antagonists from Sydney, non-industrial source), TGF-β I receptor kinase inhibitors from EIi Lilly, TGF- β receptor inhibitors from Johnson & Johnson, and an analogue or derivative thereof, 103) a TNFα antagonist or TACE inhibitor selected from the group consisting of adalimumab (CAS No. 331731-18-1) (Cambridge Antibody Technology), AGIX-4207 (AtheroGenics), AGT-1 (Advanced Biotherapy), an anti-inflammatory from Borean Pharma, Cellzome, or Paradigm Therapeutics, anti-inflammatory vaccine (TNF-alpha kinoid) from Neovacs, humanized anti-TNF antibody or an anti-TNF MAb (CB0006) Celltech (UCB)1 apratastat (CAS No. 287405-51-0) (Wyeth), BMS-561392 (Bristol-Myers Squibb), BN-006 (Bone), certolizumab pegol (CAS No. 428863-50-7 or CH-138 (UCB), cilomilast (CAS No. 153259-65-5) (GlaxoSmithKline), CR-1 (Nuada Pharmaceuticals), CRx-119 (CombinatoRx), D-5410 (UCB), dacopafant (CAS No. 125372-33-0) (Sanofi- Aventis), dersalazine (CAS No. 188913-57-7/188913-58-8) (Uriach), etanercept (CAS No. 185243-69-0) (Amgen), ethyl pyruvate (Critical (Critical Therapeutics), golimumab (CAS No. 476181-74-5) (Johnson & Johnson), hormono-immunotherapy from Ipsen, CDP571 {e.g., Humicade from UCB), IC-485 (ICOS), infliximab (CAS No. 170277-31-3) (Johnson & Johnson), IP- 751 (Manhattan Pharmaceuticals), ISIS-104838 (CAS No. 250755-32-9) (ISIS Pharmaceuticals), lenalidomide (CAS No. 191732-72-6) (Celgene), lentinan (CAS No. 37339-90-5) (Ajinomoto), MDL-201112 (CAS No. 142130-73-2) (Sanofi-Aventis), medroxyprogesterone (CAS No. 520-85-4) (InKine Pharmaceutical), N-acetylcysteine (CAS No. 616-91-1) (Zambon), NBE-P2 (DIREVO Biotech), nerelimomab (CAS No. 162774-06-3) (Chiron), OM-294DP (OM PHARMA), onercept (CAS No. 199685-57-9) (Yeda), PASSTNF-alpha (Verigen), pentoxifylline or oxypentifylline (Sanofi-Aventis), Pharmaprojects No. 4091 , 4241 , 4295, or 4488 (Sanofi-Aventis), Pharmaprojects No. 5480 (Amgen), Pharmaprojects No. 6749 (Cengent), pirfenidone (CAS No. 53179-13-8) (MARNAC), RPR-132294 (Sanofi- Aventis), S5 (F002) (Fulcrum Pharmaceuticals), simvastatin (CAS No. 79902-63-9) (Merck & Co), STA-6292 (Synta Pharmaceuticals), tacrolimus (CAS No. 104987-11-3) (Fujisawa LifeCycle Pharma), talactoferrin alfa (CAS No. 308240-58-6) (Agennix), thalidomide (CAS No. 50-35-1) (Celgene), TNF antagonists form ProStrakan, and Synergen, TNF inhibitors (Amgen), TNF-alpha antagonists from Dynavax Technologies and Jerina AG (Germany), TNF-alpha inhibitors from IBFB Pharma and Xencor (Xencor), torbafylline (CAS No. 105102-21-4) (Sanofi-Aventis), UR-1505 (Uriach), VT-346 (Viron Therapeutics), YSIL6 (Y's Therapeutics), YSTH2 (Y's Therapeutics), NPI-1302a-3 (Nereus Pharmaceuticals, a TNF antagonist from Jerina AG (Germany), dersalazine, and an analogue or derivative thereof, 104) a tumor necrosis factor antagonist, 105) a Toll receptor inhibitor, 106) a tubulin antagonist, 107) a tyrosine kinase inhibitor selected from the group consisting of SU-011248, SUTENT from Pfizer Inc. (New York, NY), BMS-354825, PN-355 (Paracelsian Pharmaceuticals), AGN-199659 (Allergan), AAL-993 or ABP-309 (Novartis), adaphostin (NIH), AEE-788 (Novartis), AG-013736 (OSI Pharmaceuticals), AG-13736 (Pfizer), ALT-110 (Alteris Therapeutics), AMG-706 (Amgen), anticancer MAbs from Xencor, anti-EGFrvlll MAbs from Abgenix, anti-HER2 MAb from Abiogen, AZD-2171 or AZD-9935 (AstraZeneca), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), CEP-5214 (Cephalon), CEP-7055 (Cephalon), cetuximab (ImClone Systems), CHIR-200131 and CHIR-258 (Chiron), CP- 547632 (OSI Pharmaceuticals), CP-724714 (Pfizer), CT-301 (Creabilis Therapeutics), D-69491 (Baxter International), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EGFR/ErbB2 inhibitors from Array BioPharma, erlotinib (CAS No. 183319-69-9) (OSI Pharmaceuticals), EXEL-2880 (Exelixis), FK-778 (Sanofi-Aventis) , gefitinib (CAS No. 184475-35-2) (AstraZeneca), GW-2286 or GW-654652 (GlaxoSmithKline), her2/neu antigen from AlphaVax, HER- 2/neu inhibitor from Generex, Herzyme (Medipad) (Sirna Therapeutics), HKI-272 (Wyeth), HuMax-EGFr (Genmab), idronoxil (CAS No. 81267-65-4) (Novogen), IGF-1 inhibitors from Ontogen, IMC-11F8 (ImClone Systems), kahalalide F (CAS No. 149204-42-2) (PharmaMar), KDR inhibitor from LG Life Sciences, KDR inhibitors from Abbott Laboratories, KDR kinase inhibitors (UCB), Kdr kinase inhibitors from Merck & Co, KRN-633 and KRN- 951 (Kirin Brewery), KSB-102 (Xenova), lapatinib ditosylate (CAS No. 388082-78-8) (GlaxoSmithKline), matuzumab (Merck KGaA), MDX-214 (Medarex), ME-103 (Pharmexa), MED-A300 (Gerolymatos), MNAC-13 (Lay Line Genomics), nimotuzumab (Center of Molecular Immunology), NSC- 330507 or NSC-707545 (NIH), NV-50 (Novogen), OSI-930 (OSI Pharmaceuticals), panitumumab (Abgenix), pelitinib (CAS No. 287933-82-7) (Wyeth), pertuzumab (CAS No. 380610-27-5) (Genentech), Pharmaprojects No. 3985 (Sanofi-Aventis), prostate cancer therapeutics from Sequenom (SQPC35, SQPC36, SQPC90), removab and remoxab (Trion Pharma), RG- 13022 (CAS No. 136831-48-6), RG-13291 (CAS No. 138989-50-1), or RG- 14620 (CAS No. 136831-49-7) (Sanofi-Aventis), RM-6427 (Romark), RNAi breast cancer therapy from Benitec, RP 53801 (CAS No. 125882-88-4) (Sanofi-Aventis), sorafenib tosylate (Bayer), SU-11657 (Pfizer), Tie-2 antagonists from Semaia (Hybrigenics), Tie-2 inhibitors from Ontogen, trastuzumab (CAS No. 180288-69-1) (Genentech), tyrosine kinase inhibitors from Sanofi-Aventis, U3-1287, U3-1565, U3-1784, U3-1800 (U3 Pharma), vatalanib (Novartis), VEGFR-2 kinase inhibitor from Bristol-Myers Squibb, XL-647 (Exelixis), ZD-6474 (AstraZeneca), ZK-CDK (Schering AG), an EGFR tyrosine kinase inhibitor, EKB-569 (Wyeth), herbimycin A, and an analogue or derivative thereof, 108) a VEGF inhibitor, 109) a vitamin D receptor agonist, 110) ZD-6474 (an angiogenesis inhibitor), 111) AP-23573 (an mTOR inhibitor), 112) synthadotin (a tubulin antagonist), 113) S-0885 (a collagenase inhibitor), 114) aplidine (an elongation factor-1 alpha inhibitor), 115) ixabepilone (an epithilone), 116) IDN-5390 (an angiogenesis inhibitor and an FGF inhibitor), 117) SB-2723005 (an angiogenesis inhibitor), 118) ABT-518 (an angiogenesis inhibitor), 119) combretastatin (an angiogenesis inhibitor), 120) anecortave acetate (an angiogenesis inhibitor), 121) SB- 715992 (a kinesin antagonist), 122) temsirolimus (an mTOR inhibitor), and 123) adalimumab (a TNFα antagonist), 124) erucylphosphocholine (an ATK inhibitor), 125) alphastatin (an angiogenesis inhibitor), 126) bortezomib (an NF Kappa B inhibitor), 127) etanercept (a TNFα antagonist and TACE inhibitor), 128) humicade (a TNFα inhibitor), and 129) gefitinib (a tyrosine kinase inhibitor), 130) a histamine receptor antagonist selected from the group consisting of phenothiazines (e.g., promethazine), alkylamines (e.g., chlorpheniramine (CAS No. 7054-11-7), brompheniramine (CAS No. 980- 71-2), fexofenadine hydrochloride, promethazine hydrochloride, loratadine, ketotifen fumarate salt, and acrivastine), methylxanthines (e.g., theophylline, theobromine, and caffeine), cimetidine (available under the tradename TAGAMET from SmithKline Beecham Phamaceutical Co., Wilmington, DE), ranitidine (available under the tradename ZANTAC from Warner Lambert Company, Morris Plains, NJ), famotidine (available under the tradename PEPCID from Merck & Co., Whitehouse Station, NJ), nizatidine (available under the tradename AXID from Reliant Pharmaceuticals, Inc., Liberty Corner, NJ), nizatidine, and roxatidine acetate (CAS No. 78628-28-1), H3 receptor antagonists {e.g., thioperamide and thioperamide maleate salt), and antihistamines (e.g., tricyclic dibenozoxepins, ethanolamines, ethylenediamines, piperizines, piperidines, and pthalazinones), 131) an alpha adrenergic receptor antagonist, 132) an anti-psychotic compound, 133) a CaM kinase Il inhibitor, 134) a G protein agonist, 135) an antibiotic selected from the group consisting of apigenin (Cas No. 520-36-5), ampicillin sodium salt (CAS No. 69-52-3), puromycin, and an analogue or derivative thereof, 136) an anti-microbial agent, 137) a DNA topoisomerase inhibitor selected from the group consisting of β-lapachone (CAS No. 4707- 32-8), (-)-arctigenin (CAS No. 7770-78-7), aurintricarboxylic acid, and an analogue or derivative thereof, 138) a thromboxane A2 receptor inhibitor selected from the group consisting of BM-531 (CAS No. 284464-46-6), ozagrel hydrochloride (CAS No. 78712-43-3), and an analogue or derivative thereof, 139) a D2 dopamine receptor antagonist, 140) a Peptidyl-Prolyl Cis/Trans lsomerase Inhibitor, 141) a dopamine antagonist, an anesthetic compound, 142) a clotting factor, 143) a lysyl hydrolase inhibitor, 144) a muscarinic receptor inhibitor, 145) a superoxide anion generator, 146) a steroid, 147) an antiproliferative agent selected from the group consisting of silibinin (CAS No. 22888-70-6), silymarin (CAS No. 65666-07-1), 1,2- hexanediol, dioctyl phthalate (CAS No. 117-81-7), zirconium (IV) oxide, glycyrrhizic acid, spermidine trihydrochloride, tetrahydrochloride, CGP 74514, spermine tetrahydrochloride, NG-methyl-L-arginine acetate salt, galardin, and an analogue or derivative thereof, 148) a diuretic, 149) an anticoagulant, 150) a cyclic GMP agonist, 151) an adenylate cyclase agonist, 152) an antioxidant, 153) a nitric oxide synthase inhibitor, 154) an antineoplastic agent selected from tirapazamine (CAS No. 27314-97-2), fludarabine (CAS No. 21679-14-1), cladribine, imatinib mesilate, and an analogue or derivative thereof, 155) a DNA synthesis inhibitor, 156) a DNA alkylating agent selected from dacarbazine (CAS No. 4342-03-4), temozolomide, procarbazine HCI, and an analogue or derivative thereof, 157) a DNA methylation inhibitor, 158) a NSAID agent, 159) a peptidylglycine alpha-hydroxylating monooxygenase inhibitor, 160) an MEK1/MEK 2 inhibitor, 161) a NO synthase inhibitor, 162) a retinoic acid receptor antagonist selected from isotretinoin (CAS No. 4759-48-2) and an analogue or derivative thereof, 163) an ACE inhibitor, 164) a glycosylation inhibitor, 165) an intracellular calcium influx inhibitor, 166) an anti-emetic agent, 167) an acetylcholinesterase inhibitor, 168) an ALK-5 receptor antagonist, 169) a RAR/RXT antagonist, 170) an elF-2a inhibitor, 171) an S- adenosyl-L-homocysteine hydrolase inhibitor, 172) an estrogen agonist, 173) a serotonin receptor inhibitor, 174) an antithrombotic agent, 175) a tryptase inhibitor, 176) a pesticide, 177) a bone mineralization promoter, 178) a bisphosphonate compound selected from risedronate and an analogue or derivative thereof, 179) an anti-inflammatory compound, 180) a DNA methylation promoter, 181) an anti-spasmodic agent, .182) a protein synthesis inhibitor, 183) an α-glucosidase inhibitor, 184) a calcium channel blocker, 185) a pyruvate dehydrogenase activator, 186) a prostaglandin inhibitor, 187) a sodium channel inhibitor, 188) a serine protease inhibitor, 189) an intracellular calcium flux inhibitor, 190) a JAK2 inhibitor; 191) an androgen inhibitor, 192) an aromatase inhibitor, 193) an anti-viral agent, 194) a 5-HT inhibitor, 195) an FXR antagonist, 196) an actin polymerization and stabilization promoter, 197) an AXOR12 agonist, 198) an angiotensin Il receptor agonist, 199) a platelet aggregation inhibitor, 200) a CB1/CB2 receptor agonist, 201) a norepinephrine reuptake inhibitor, 202) a selective serotonin reuptake inhibitor, 203) a reducing agent, 204) Isotretinoin, 205) radicicol, 206) clobetasol propionate, 207) homoharringtonine, 208) trichostatin A, 209) brefeldin A, 210) thapsigargin, 211) dolastatin 15, 212) cerivastatin, 213) jasplakinolide, 214) herbimycin A, 215) pirfenidone, 216) vinorelbine, 217) 17-DMAG, 218) tacrolimus, 219) loteprednol etabonate, 220) juglone, 221) prednisolone, 222) puromycin, 223) 3-BAABE, 224) cladribine, 225) mannose-6-phosphate, 226) 5-azacytidine, 227) Ly333531 (ruboxistaurin), 228) simvastatin, and 229) an immuno-modulator selected from Bay 11-7085, (-)-arctigenin, idazoxan hydrochloride, and an analogue or derivative thereof. These and other agents are described in more detail herein.
These and other aspects of the present invention will become evident upon reference to the following detailed description and attached drawings. In addition, various references are set forth herein which describe in more detail certain procedures and/or compositions, and are therefore incorporated by reference in the entirety.
BRIEF DESCRIPTION OF THE DRAWINGS
Figure 1 schematically depicts the transcriptional regulation of matrix metalloproteinases.
Figure 2 is a blot which demonstrates that IL-1 stimulates AP-1 transcriptional activity. _ . . . . . . . . . .. . -
Figure 3 is a graph which shows that IL-1 induced binding activity decreased in lysates from chondrocytes which were pretreated with paclitaxel.
Figure 4 is a blot which shows that IL-1 induction increases collagenase and stromelysin in RNA levels in chondrocytes, and that this induction can be inhibited by pretreatment with paclitaxel.
Figures 5A-H are blots that show the effect of various anti- microtubule agents in inhibiting collagenase expression.
Figure 6 is a graph showing the results of a screening assay for assessing the effect of paclitaxel on smooth muscle cell migration.
Figure 7 is a graph showing the average rank of joint scores of Hartley guinea pig knees with ACL damage treated with paclitaxel. A reduction in score indicates an improvement in cartilage score. The dose response trend is statistically significant (p < 0.02). Figures 8A-C are examples of cross sections of Hartley guinea pig knees of control and paclitaxel treated animals. Figure 8A. Control speciment showing erosion of cartilage to the bone. Figure 8B. Paclitaxel dose 1 (low dose) showing fraying of cartilage. Figure 8C. Paclitaxel dose 2 (medium dose) showing minor defects to cartilage.
Figures 9A-F are safranin-O stained histological slides of representative synovial tissues from naive (healthy) knees (Figures 9A and 9D) and knees with arthritis induced by administration of albumin in Freund's complete adjuvant (Figures 9B and 9C) or carrageenan (Figures 9E and 9F). Arthritic knees received either control (Figures 9B and 9E) or 20% paclitaxel-loaded microspheres (Figures 9C and 9F). The data illustrate decreased proteoglycan red staining in arthritic knees treated with control microspheres and the proteoglycan protection properties of the paclitaxel- loaded formulation.
DETAILED DESCRIPTION OF THE INVENTION - -
Definitions
Prior to setting forth the invention, it may be helpful to an understanding thereof to first set forth definitions of certain terms that are used herein.
"Fibrosis," or "scarring," or "fibrotic response" refers to the formation of fibrous (scar) tissue in response to injury or medical intervention. Therapeutic agents which inhibit fibrosis or scarring are referred to herein as "fibrosis-inhibiting agents", "fibrosis-inhibitors", "anti- scarring agents", and the like, where these agents inhibit fibrosis through one or more mechanisms including: inhibiting inflammation or the acute inflammatory response, inhibiting migration or proliferation of connective tissue cells (such as fibroblasts, smooth muscle cells, vascular smooth muscle cells), inhibiting angiogenesis, reducing extracellular matrix (ECM) production or promoting ECM breakdown, and/or inhibiting tissue remodeling. When scarring occurs in a confined space (e.g., within a lumen) following surgery or instrumentation (including implantation of a medical device or implant), such that a body passageway (e.g., a blood vessel, the gastrointestinal tract, the respiratory tract, the urinary tract, the female or male reproductive tract, the eustacian tube etc.) is partially or completely obstructed by scar tissue, this is referred to as "stenosis" (narrowing). When scarring subsequently occurs to re-occlude a body passageway after it was initially successfully opened by a surgical intervention (such as placement of a medical device or implant), this is referred to as "restenosis."
"Host", "person", "subject", "patient" and the like are used synonymously to refer to the living being into which a device or implant of the present invention is implanted.
"Implanted" refers to having completely or partially placed a device or implant within a host. A device is partially implanted when some of the device reaches, or extends to the outside of, a host.
"Inhibit fibrosis", "reduce fibrosis", "Inhibits scarring" and the like are used synonymously to refer to the action of agents or compositions which result in a statistically significant decrease in the formation of fibrous tissue that can be expected to occur in the absence of the agent or composition.
"Anti-infective agent" refers to an agent or composition which prevents microrganisms from growing and/or slows the growth rate of microorganisms and/or is directly toxic to microorganisms at or near the site of the agent. These processes would be expected to occur at a statistically significant level at or near the site of the agent or composition relative to the effect in the absence of the agent or composition.
"Inhibit infection" refers to the ability of an agent or composition to prevent microorganisms from accumulating and/or proliferating near or at the site of the agent. These processes would be expected to occur at a statistically significant level at or near the site of the agent or composition relative to the effect in the absence of the agent or composition.
"Inhibitor" refers to an agent which prevents a biological process from occurring or slows the rate or degree of occurrence of a biological process. The process may be a general one such as scarring or refer to a specific biological action such as, for example, a molecular process resulting in release of a cytokine.
"Antagonist" refers to an agent which prevents a biological process from occurring or slows the rate or degree of occurrence of a biological process. While the process may be a general one, typically this refers to a drug mechanism where the drug competes with a molecule for an active molecular site or prevents a molecule from interacting with the molecular site. In these situations, the effect is that the molecular process is inhibited.
"Agonist" refers to an agent which stimulates a biological process or rate or degree of occurrence of a biological process. The process may be a general one such as scarring or refer to a specific biological action such as, for example, a molecular process resulting in release of a cytokine.
"Anti-microtubule agents" should be understood to include any protein, peptide, chemical, or other molecule which impairs the function of microtubules, for example, through the prevention or stabilization of polymerization. Compounds that stabilize polymerization of microtubules are referred to herein as "microtubule stabilizing agents." A wide variety of methods may be utilized to determine the anti-microtubule activity of a particular compound, including for example, assays described by Smith et al. {Cancer Lett 79(2):213-219, 1994) and Mooberry et al., (Cancer Lett. 96(2):261-266, 1995).
"Medical device", "implant", ""device", medical device", "medical implant", "implant/device" and the like are used synonymously to refer to any object that is designed to be placed partially or wholly within a patient's body for one or more therapeutic or prophylactic purposes such as for restoring physiological function, alleviating symptoms associated with disease, delivering therapeutic agents, and/or repairing, replacing, or augmenting etc. damaged or diseased organs and tissues. While normally composed of biologically compatible synthetic materials (e.g., medical-grade stainless steel, titanium and other metals; polymers such as polyurethane, silicon, PLA, PLGA and other materials) that are exogenous, some medical devices and implants include materials derived from animals (e.g., "xenografts" such as whole animal organs; animal tissues such as heart valves; naturally occurring or chemically-modified molecules such as collagen, hyaluronic acid, proteins, carbohydrates and others), human donors (e.g., "allografts" such as whole organs; tissues such as bone grafts, skin grafts and others), or from the patients themselves (e.g., "autografts" such as saphenous vein grafts, skin grafts, tendon/ligament/muscle transplants). Representative examples of medical devices that are of particular utility in the present invention include,,but are not restricted to, vascular stents, gastrointestinal stents, tracheal/bronchial stents, genital- urinary stents, ENT stents, intra-articular implants, intraocular lenses, implants for hypertrophic scars and keloids, vascular grafts, anastomotic connector devices, implantable sensors, implantable pumps, soft tissue implants (e.g., cosmetic implants and implants for reconstructive surgery), implantable electrical devices, such as implantable neurostimulators and implantable electrical leads, surgical adhesion barriers, glaucoma drainage devices, surgical films and meshes, prosthetic heart valves, tympanostomy tubes, penile implants, endotracheal and tracheostomy tubes, peritoneal dialysis catheters, intracranial pressure monitors, vena cava filters, central venous catheters (CVCs), ventricular assist devices (e.g., LVAD), spinal prostheses, urinary (Foley) catheters, prosthetic bladder sphincters, orthopedic implants, and gastrointestinal drainage tubes. "Chondroprotection" refers to the prevention of cartilage loss. Cartilage is formed from chondrocytes, and chondroprotection is the protection of the chondrocytes so that they do not die.
"Release of an agent" refers to a statistically significant presence of the agent, or a subcomponent thereof, which has disassociated from the implant/device and/or remains active on the surface of (or within) the device/implant.
"Biodegradable" refers to materials for which the degradation process is at least partially mediated by, and/or performed in, a biological system. "Degradation" refers to a chain scission process by which a polymer chain is cleaved into oligomers and monomers. Chain scission may occur through various mechanisms, including, for example, by chemical reaction (e.g., hydrolysis) or by a thermal or photolytic process. Polymer degradation may be characterized, for example, using gel permeation chromatography (GPC), which monitors the polymer molecular mass changes during erosion and drug release. Biodegradable also refers to materials may be degraded by an erosion process mediated by, and/or performed in, a biological system. "Erosion" refers to a process in which material is lost from the bulk. In the case of a polymeric system, the material may be a monomer, an oligomer, a part of a polymer backbone, or a part of the polymer bulk. Erosion includes (i) surface erosion, in which erosion affects only the surface and not the inner parts of a matrix; and (ii) bulk erosion, in which the entire system is rapidly hydrated and polymer chains are cleaved throughout the matrix. Depending on the type of polymer, erosion generally occurs by one of three basic mechanisms (see, e.g., Heller, J., CRC Critical Review in Therapeutic Drug Carrier Systems (1984), 1(1), 39-90); Siepmann, J. et al., Adv. Drug Del. Rev. (2001), 48, 229-247): (1) water-soluble polymers that have been insolubilized by covalent cross-links and that solubilize as the cross-links or the backbone undergo a hydrolytic cleavage; (2) polymers that are initially water insoluble are solubilized by hydrolysis, ionization, or pronation of a pendant group; and (3) hydrophobic polymers are converted to small water-soluble molecules by backbone cleavage. Techniques for characterizing erosion include thermal analysis (e.g., DSC), X-ray diffraction, scanning electron microscopy (SEM), electron paramagnetic resonance spectroscopy (EPR), NMR imaging, and recording mass loss during an erosion experiment. For microspheres, photon correlation spectroscopy (PCS) and other particles size measurement techniques may be applied to monitor the size evolution of erodible devices versus time.
As used herein, "analogue" refers to a chemical compound that is structurally similar to a parent compound, but differs slightly in composition (e.g., one atom or functional group is different, added, or removed). The analogue may or may not have different chemical or physical properties than the original compound and may or may not have improved biological and/or chemical activity. For example, the analogue may be more hydrophilic or it may have altered reactivity as compared to the parent compound. The analogue may mimic the chemical and/or- biologically activity of the parent compound (i.e., it may have similar or identical activity), or, in some cases, may have increased or decreased activity. The analogue may be a naturally or non-naturally occurring (e.g., recombinant) variant of the original compound. An example of an analogue is a mutein (i.e., a protein analogue in which at least one amino acid is deleted, added, or substituted with another amino acid). Other types of analogues include isomers (enantiomers, diasteromers, and the like) and other types of chiral variants of a compound, as well as structural isomers. The analogue may be a branched or cyclic variant of a linear compound. For example, a linear compound may have an analogue that is branched or otherwise substituted to impart certain desirable properties (e.g., improve hydrophilicity or bioavailability).
As used herein, "derivative" refers to a chemically or biologically modified version of a chemical compound that is structurally similar to a parent compound and (actually or theoretically) derivable from that parent compound. A "derivative" differs from an "analogue" in that a parent compound may be the starting material to generate a "derivative," whereas the parent compound may not necessarily be used as the starting material to generate an "analogue." A derivative may or may not have different chemical or physical properties of the parent compound. For example, the derivative may be more hydrophilic or it may have altered reactivity as compared to the parent compound. Derivatization (Ae., modification) may involve substitution of one or more moieties within the molecule (e.g., a change in functional group). For example, a hydrogen may be substituted with a halogen, such as fluorine or chlorine, or a hydroxyl group (-OH) may be replaced with a carboxylic acid moiety (- COOH). The term "derivative" also includes conjugates, and prodrugs of a parent compound (i.e., chemically modified derivatives which can be converted into the original compound under physiological conditions). For example, the prodrug may be an inactive form of an active agent. Under physiological conditions, the prodrug may be converted into the active form of the compound. Prodrugs may be formed, for example, by replacing one or two hydrogen atoms on nitrogen atoms by an acyl group (acyl prodrugs) or a carbamate group (carbamate prodrugs). More detailed information relating to prodrugs is found, for example, in Fleisher et al., Advanced Drug Delivery Reviews 19 (1996) 115; Design of Prodrugs, H. Bundgaard (ed.), Elsevier, 1985; or H. Bundgaard, Drugs of the Future 16 (1991) 443. The term "derivative" is also used to describe all solvates, for example hydrates or adducts (e.g., adducts with alcohols), active metabolites, and salts of the parent compound. The type of salt that may be prepared depends on the nature of the moieties within the compound. For example, acidic groups, for example carboxylic acid groups, can form, for example, alkali metal salts or alkaline earth metal salts (e.g., sodium salts, potassium salts, magnesium salts and calcium salts, and also salts with physiologically tolerable quaternary ammonium ions and acid addition salts with ammonia and physiologically tolerable organic amines such as, for example, triethylamine, ethanolamine or tris-(2-hydroxyethyl)amine). Basic groups can form acid addition salts, for example with inorganic acids such as hydrochloric acid, sulfuric acid or phosphoric acid, or with organic carboxylic acids and sulfonic acids such as acetic acid, citric acid, benzoic acid, maleic acid, fumaric acid, tartaric acid, methanesulfonic acid or p-toluenesulfonic acid. Compounds which simultaneously contain a basic group and an acidic group, for example a carboxyl group in addition to basic nitrogen atoms, can be present as zwitterions. Salts can be obtained by customary methods known to those skilled in the art, for example by combining a compound with an inorganic or organic acid or base in a solvent or diluent, or from other salts by cation exchange or anion exchange.
"Hyaluronic acid" or "HA" as used herein refers to all forms of hyaluronic acid that are described or referenced herein, including those that have been processed or chemically or physically modified, as well as hyaluronic acid that has been crosslinked (for example, covalently, ionically, thermally or physically). HA is a glycosaminoglycan composed of a linear chain of about 2500 repeating disaccharide units. Each disaccharide unit is composed of an N-acetylglucosamine residue linked to a glucuronic acid. Hyaluronic acid is a natural substance that is found in the extracellular matrix of many tissues including synovial joint fluid, the vitreous humor of the eye, cartilage, blood vessels, skin and the umbilical cord. Commercial forms of hyaluronic acid having a molecular weight of approximately 1.2 to 1.5 million Daltons (Da) are extracted from rooster combs and other animal sources. Other sources of HA include HA that is isolated from cell culture / fermentation processes. Lower molecular weight HA formulations are also available from a variety of commercial sources. The molecule can be of variable lengths (i.e., different numbers of repeating disaccharide units and different chain branching patterns) and can be modified at several sites (through the addition or subtraction of different functional groups) without deviating from the scope of the present invention. The term "inter-react" refers to the formulation of covalent bonds, noncovalent bonds, or both. The term thus includes crosslinking, which involves both intermolecular crosslinks and optionally intramolecular crosslinks as well, arising from the formation of covalent bonds. Covalent bonding between two reactive groups may be direct, in which case an atom in reactive group is directly bound to an atom in the other reactive group, or it may be indirect, through a linking group. Noncovalent bonds include ionic (electrostatic) bonds, hydrogen bonds, or the association of hydrophobic molecular segments, which may be the same or different. A crosslinked matrix may, in addition to covalent bonds, also include such intermolecular and/or intramolecular noncovalent bonds.
When referring to polymers, the terms "hydrophilic" and "hydrophobic" are generally defined in terms of an HLB value, i.e., a hydrophilic lipophilic balance. A high HLB value indicates a hydrophilic compound, while a low HLB value characterizes a hydrophobic compound. HLB values are well known in the art, and generally range from 1 to 18. Preferred multifunctional compound cores are hydrophilic, although as long as the multifunctional compound as a whole contains at least one hydrophilic component, crosslinkable hydrophobic components may also be present.
The term "synthetic" is used to refer to polymers, compounds and other such materials that are "chemically synthesized." For example, a synthetic material in the present compositions may have a molecular structure that is identical to a naturally occurring material, but the material perse, as incorporated in the compositions of the invention, has been chemically synthesized in the laboratory or industrially. "Synthetic" materials also include semi-synthetic materials, i.e., naturally occurring materials, obtained from a natural source, that have been chemically modified in some way. Generally, however, the synthetic materials herein are purely synthetic, i.e., they are neither semi-synthetic nor have a structure that is identical to that of a naturally occurring material. The term "effective amount" refers to the amount of composition required in order to obtain the effect desired. For example, a "tissue growth-promoting amount" of a composition refers to the amount needed in order to stimulate tissue growth to a detectable degree. Tissue, in this context, includes connective tissue, bone, cartilage, epidermis and dermis, blood, and other tissues. The actual amount that is determined to be an effective amount will vary depending on factors such as the size, condition, sex and age of the patient and can be more readily determined by the caregiver.
The term "in situ" as used herein means at the site of administration. Thus, compositions of the invention can be injected or otherwise applied to a specific site within a patient's body, e.g., a site in need of augmentation, and allowed to crosslink at the site of injection. Suitable sites will generally be intradermal or subcutaneous regions for augmenting dermal support, at a bone fracture site for bone repair, within sphincter tissue for sphincter augmentation (e.g., for restoration of continence), within a wound or suture, to promote tissue regrowth; and within or adjacent to vessel anastomoses, to promote vessel regrowth.
The term "aqueous medium" includes solutions, suspensions, dispersions, colloids, and the like containing water. The term "aqueous environment" means an environment containing an aqueous medium. Similarly, the term "dry environment" means an environment that does not contain an aqueous medium.
With regard to nomenclature pertinent to molecular structures, the following definitions apply:
The term "alkyl" as used herein refers to a branched or unbranched saturated hydrocarbon group typically although not necessarily containing 1 to about 24 carbon atoms, such as methyl, ethyl, /7-propyl, isopropyl, π-butyl, isobutyl, f-butyl, octyl, decyl, and the like, as well as cycloalkyl groups such as cyclopentyl, cyclohexyl and the like. Generally, although again not necessarily, alkyl groups herein contain 1 to about 12 carbon atoms. The term "lower alkyl" intends an alkyl group of one to six carbon atoms, preferably one to four carbon atoms. "Substituted alkyl" refers to alkyl substituted with one or more substituent groups. "Alkylene," "lower alkylene" and "substituted alkylene" refer to divalent alkyl, lower alkyl, and substituted alkyl groups, respectively.
The term "aryl" as used herein, and unless otherwise specified, refers to an aromatic substituent containing a single aromatic ring (monocyclic) or multiple aromatic rings that are fused together, linked covalently, or linked to a common group such as a methylene or ethylene moiety. The common linking group may also be a carbonyl as in benzophenone, an oxygen atom as in diphenylether, or a nitrogen atom as in diphenylamine. Preferred aryl groups contain one aromatic ring or two fused or linked aromatic rings, e.g., phenyl, naphthyl, biphenyl, diphenylether, diphenylamine, benzophenone, and the like. "Substituted aryl" refers to an aryl moiety substituted with one or more substituent groups, and the terms "heteroatom-containing aryl" and "heteroaryl" refer to aryl in which at least one carbon atom is replaced with a heteroatom. The terms "arylene" and "substituted arylene" refer to divalent aryl and substituted aryl groups as just defined.
The term "heteroatom-containing" as in a "heteroatom- containing hydrocarbyl group" refers to a molecule or molecular fragment in which one or more carbon atoms is replaced with an atom other than carbon, e.g., nitrogen, oxygen, sulfur, phosphorus or silicon.
"Hydrocarbyl" refers to univalent hydrocarbyl radicals containing 1 to about 30 carbon atoms, preferably 1 to about 24 carbon atoms, most preferably 1 to about 12 carbon atoms, including branched or unbranched, saturated or unsaturated species, such as alkyl groups, alkenyl groups, aryl groups, and the like. The term "lower hydrocarbyl" intends a hydrocarbyl group of one to six carbon atoms, preferably one to four carbon atoms. The term "hydrocarbylene" intends a divalent hydrocarbyl moiety containing 1 to about 30 carbon atoms, preferably 1 to about 24 carbon atoms, most preferably 1 to about 12 carbon atoms, including branched or unbranched, saturated or unsaturated species, or the like. The term "lower hydrocarbylene" intends a hydrocarbylene group of one to six carbon atoms, preferably one to four carbon atoms. "Substituted hydrocarbyl" refers to hydrocarbyl substituted with one or more substituent groups, and the terms "heteroatom-containing hydrocarbyl" and "heterohydrocarbyl" refer to hydrocarbyl in which at least one carbon atom is replaced with a heteroatom. Similarly, "substituted hydrocarbylene" refers to hydrocarbylene substituted with one or more substituent groups, and the terms "heteroatom-containing hydrocarbylene" and "heterohydrocarbylene" refer to hydrocarbylene in which at least one carbon atom is replaced with a heteroatom. If not otherwise indicated, "hydrocarbyl" indicates both unsubstituted and substituted hydrocarbyls, "heteroatom-containing hydrocarbyl" indicates both unsubstituted and substituted heteroatom- containing hydrocarbyls and so forth.
By "substituted" as in "substituted hydrocarbyl," "substituted alkyl," and the like, as alluded to in some of the aforementioned definitions, is meant that in the hydrocarbyl, alkyl, or other moiety, at least one hydrogen atom bound to a carbon atom is replaced with one or more substituents that are functional groups such as alkoxy, hydroxy, halo, nitro, and the like. Unless otherwise indicated, it is to be understood that specified molecular segments can be substituted with one or more substituents that do not compromise a compound's utility. For example, "succinimidyl" is intended to include unsubstituted succinimidyl as well as sulfosuccinimidyl and other succinimidyl groups substituted on a ring carbon atom, e.g., with alkoxy substituents, polyether substituents, or the like.
Any concentration ranges, percentage range, or ratio range recited herein are to be understood to include concentrations, percentages or ratios of any integer within that range and fractions thereof, such as one tenth and one hundredth of an integer, unless otherwise indicated. Also, any number range recited herein relating to any physical feature, such as polymer subunits, size or thickness, are to be understood to include any integer within the recited range, unless otherwise indicated. As used herein, the term "about" refers to ± 15% of any indicated structure, value, or range.
"A" and "an" refer to one or more of the indicated items. For example, "a" polymer refers to both one polymer or a mixture comprising two or more polymers; "a multifunctional compound " refers not only to a single multifunctional compound but also to a combination of two or more of the same or different multifunctional compounds; "a reactive group" refers to a combination of reactive groups as well as to a single reactive group, and the like.
As discussed above, the present invention provides polymeric compositions which greatly increase the ability to inhibit the formation of reactive scar tissue on, or around, the surface of a device or implant or at a treatment site. Numerous polymeric compositions and therapeutic agents are described herein.
The present invention provides for the combination of compositions (e.g., polymers) which include one or more therapeutic agents, described below. Also described in more detail below are methods for making and methods for utilizing such compositions.
Therapeutic Agents
Therapeutic agents useful in the present invention includes various anti-fibrosis agents, anti-infective agents, and polymers.
Anti-fibrosis Agents
In one aspect, the present invention discloses pharmaceutical agents which inhibit one or more aspects of the production of excessive fibrous (scar) tissue. Such agents may be readily determined based upon the in vitro and in vivo (animal) models such as those provided in Examples 16-20, 21-28, 29, 38, 39, 42, 43, and 81. Agents which inhibit fibrosis may be identified through in vivo models including inhibition of intimal hyperplasia development in the rat balloon carotid artery model (Examples 21 and 29). The assays set forth in Examples 20 and 28 may be used to determine whether an agent is able to inhibit cell proliferation in fibroblasts and/or smooth muscle cells. In one aspect of the invention, the agent has an IC50 for inhibition of cell proliferation within a range of about 10"6 to about 10"10 M. In certain embodiments, the agent may have an IC50 for inhibition of cell proliferation of less than about 10,000 nM; or less than about 1000 nM; or less than about 100 nM. The assay set forth in Example 24 may be used to determine whether an agent may inhibit migration of fibroblasts and/or smooth muscle cells. In one aspect of the invention, the agent has an IC50 for inhibition of cell migration within a range of about 10"6 to about 10"9M. In certain embodiments, the agent may have an IC50 for inhibition of fibroblast or smooth muscle cell migration of less than about 10,000 nM; or less than about 1000 nM; or less than about 100 nM. Assays set forth herein may be used to determine whether an agent is able to inhibit inflammatory processes, including nitric oxide production in macrophages (Example 16), and/or TNF-alpha production by macrophages (Example 17), and/or IL-1 beta production by macrophages (Example 25), and/or IL-8 production by macrophages (Example 26), and/or inhibition of MCP-1 by macrophages (Example 27). In one aspect of the invention, the agent has an IC50 for inhibition of any one of these inflammatory processes within a range of about 10"6 to about 10"10M. In certain embodiments, the agent may have an IC50 for any one of these inflammatory processes of less than about 10,000 nM; or less than about 1000 nM; or less than about 100 nM. The assay set forth in Example 22 may be used to determine whether an agent is able to inhibit MMP production. In one aspect of the invention, the agent has an IC50 for inhibition of MMP production within a range of about 10"4 to about 10"8M. In certain embodiments, the agent may have an IC50 for inhibition of MMP production of less than about 10,000 nM; or less than about 1000 nM; or less than about 100 nM. The assay set forth in Example 23 (also known as the CAM assay) may be used to determine whether an agent is able to inhibit angiogenesis. In one aspect of the invention, the agent has an IC50 for inhibition of angiogenesis within a range of about 10"6 to about 10"10M. In certain embodiments, the agent may have an IC50 for inhibition of angiogenesis of less than about 10,000 nM; or less than about 1000 nM; or less than about 100 nM. The assay set forth in Example 81 may be used to determine whether an agent is able to inhibit MMP-1. In one aspect of the invention, the agent has an IC50 for inhibition of MMP-1 within a range of about 10"6 to about 10"10M. In certain embodiments, the agent may have an IC50 for inhibition of MMP-1 of less than about 10,000 nM; or less than about 1000 nM; or less than about 100 nM. Agents which reduce the formation of surgical adhesions may be identified through in vivo models including the rabbit surgical adhesions model (Examples 19, 39, 42, and 43) and the rat caecal sidewall model (Example 18). These pharmacologically active agents (described below) can then be delivered at appropriate dosages into to the tissue either alone, or via carriers (described herein), to treat the clinical problems described herein. . . _ _ . .
Numerous therapeutic compounds capable of inhibiting fibrosis may be identified as useful in the invention including:
1) Adensosine A2A receptor antagonist
In another embodiment, the fibrosis-inhibiting compound is an adensosine A2A receptor antagonist (e.g., Sch-63390 (Schering-Plough) or an A2A receptor antagonists from Almirall-Prodesfarma, SCH-58261 (CAS No. 160098-96-4), or an analogue or derivative thereof).
2) AKT inhibitor
In another embodiment, the fibrosis-inhibiting compound is an AKT inhibitor (e.g., PKB inhibitors from DeveloGen, AKT inhibitors from Array BioPharma, Celgene, Merck & Co, Amphora, NeoGenesis Pharmaceuticals, A-443654 (Abbott Laboratories), erucylphosphocholine (AEterna Zentaris), KRX-401 (Keryx), protein kinase B inhibitors from Astex Technology, PX-316 (ProlX), or an analogue or derivative thereof).
3) Alpha 2 Inteqrin Antagonist
In another embodiment, the fibrosis-inhibiting compound is an alpha 2 integrin antagonist (e.g., Pharmaprojects No. 5754 (Merck KGaA), or an analogue or derivative thereof).
4) Alpha 4 Integrin Antagonist
In another embodiment, the fibrosis-inhibiting compound is an alpha 4 integrin antagonist (e.g., T-0047 (Tanabe Seiyaku), VLA-4 antagonists from Sanofi-Aventis, Merck & Co., Biogen Idee, Uriach, and Molecumetics, alpha 4 integrin antagonists from Genentech), BIO-2421 (Biogen Idee), cell adhesion inhibitors from Kaken Pharmaceuticals, CT-737 (Wyeth), CT-767 (Elan), CY-9652 (Epimmune), CY-9701 (Epimmune), fibronectin antagonists from Uriach, integrin alpha4β7 antagonists frin Wilex, Pharmaprojects No. 5972 (UCB), Pharmaprojects No. 6603 (Wyeth), TBC- 3342, TBC-772, and TBC-3486 (Encysive Pharmaceuticals), TBC-4746 (Schering-Plough), or a VLA4/VCAM inhibitor (Elan Pharmaceuticals), ZD- 7349 (AstraZeneca), or an analogue or derivative thereof).
5) Alpha 7 Nicotinic Receptor Agonist
In another embodiment, the fibrosis-inhibiting compound is an alpha 7 nicotinic receptor agonist (e.g., AZD-0328 (AstraZeneca), galantamine (CAS No. 357-70-0) (Synaptec), MEM-3454 or nicotinic alpha-7 agonist (Memory Pharmaceuticals and Critical Therapeutics), Pharmaprojects No. 4779 (AstraZeneca), PNU-282987 (Pfizer), SSR- 180711 (Sanofi-Aventis), TC-1698 or TC-5280 (Targacept), or an analogue or derivative thereof). 6) Angiogenesis Inhibitors
In one embodiment, the fibrosis-inhibjting compound is an angiogenesis inhibitor (e.g., AG-") 2, 958 (Pfizer), ATN-161 (Attention LLC), neovastat, an angiogenesis inhibitor from Jerina AG (Germany), NM-3 (Mercian), VGA-1155 (Taisho), FCE-26644 (Pfizer), FCE-26950 (Pfizer), FPMA (Meiji Daries), FR-111142 (Fujisawa), GGTI-298, GM-1306 (Ligand), GPA-1734 (Novartis), NNC-47-0011 (Novo Nordisk), herbamycin (Nippon Kayaku), lenalidomide (Celegene), IP-10 (N(H)1 ABT-828 (Abbott), KIN-841 (Tokushima University, Japan), SF-1126 (Semafore Pharmaceuticals), laminin technology (NIH), CHIR-258 (Chiron), NVP-AEW541 (Novartis), NVP-AEW541 (Novartis), Vt16907 (Alchemia), OXI-8007 (Oxigene), EG- 3306 (Ark Therapeutics), Maspin (Arriva), ABT-567 (Abbott), PPI-2458 (Praecis Pharmaceuticals), CC-5079, CC-4089 (Celgene), HIF-1 alpha inhibitors (Xenova), S-247 (Pfizer), AP-23573 (Ariad), AZD-9935 (Astra Zeneca), mebendazole (Introgen Therapeutics), MetAP-2 inhibitors (GlaxoSmithKline), AG-615 (Angiogene Pharmaceuticals), Tie-2 antagonists (Hybrigenics), NC-381, CYC-381, NC-169, NC-219, NC-383, NC-384, NC- 407 (Lorus Therapeutics), ATN-224 (Attenuon), ON-01370 (Onconova), Vitronectin antagonists (Amgen), SDX-103 (Salmedix), Vitronectin antagonists (Shire), CHP (Riemser), TEK (Amgen), Anecortave acetate (Alcon), T46.2 (Matrix Therapeutics), HG-2 (Heptagen), TEM antagonists (Genzyme), Oxi-4500 (Oxigene), ATN-161 (Attenuon), WX-293 (Wilex), M- 2025 (Metris Therapeutics), Alphastatin (BioActa), YH-16 (Yantai Rongchang), BIBF-1120 (Boehringer Ingelheim), BAY-57-9352 (Bayer), AS- 1404 (Cancer Research Technology), SC-77964 (Pfizer), glycomimetics (BioTie Therapies), TIE-2 Inhibitors (Ontogen), DIMI, Octamer (Octamer), ABR-215050 (Active Biotech), ABT-518 (Abbott), KDR inhibitors (Abbott), BSF-466895 (Abbott), SCH-221153 (Schering-Plough), DAC:antiangiogenic (ConjuChem), TFPI (EntreMed), AZD-2171 (Astra-Zenaca), CDC-394 (Celgene), LY290293 (EIi Lilly), IDN-5390 (Indena), Kdr Kinase Inhibitors (Merck), CT-113020, CT-116433, CT-116563, CT-31890, CT-32228) (Cell Therapeutics), A-299620 (Abbott), TWEAK Inhibitor (Amgen), VEGF modulators (Johnson and Johnson), Tum-N53, tumstatin (Genzyme), Thios- 1, Thios-2 (Thios Pharmaceuticals), MV-6401 (Miravant Medical Technologies), Spisulosine (PharmaMar), CEP-7055 (Cephalon), AUV-201 (Auvation), LM-609 (EIi Lilly), SKF-106615 (AnorMED), Oglufanide disodium (Cytran), BW-114 (Pharminox), Calreticulin (NIH), WX-678 (Wilex), SD-7784 (Pfizer), WX-UK1 (Wilex), SH-268 (Schering AG), 2-Me-PGA (Celgene), S- 137 (Pfizer), ZD-6126 (Angiogene Pharmaceuticals), SG-292 (SignalGen), Benefin (Lane Labs), A6, A36 (Angstrom), SB-2723005 (GlaxoSmithKline), SC-7 (Cell Therapeutics), ZEN-014 (AEterna Zentaris), 2-methoxyestradiol (EntreMed), NK-130119 (Nippon Kayaku), CC-10004 (Celgene), AVE- 8062A (Ajinomoto), Tacedinaline (Pfizer), Actinonin (Tokyo Metropolitan Institute of Medical Science), Lenalidomide (Celgene), VGA-1155, BTO-956 (SRI International), ER-68203-00 (Eisai), CT-6685 (UCB), JKC-362 (Phoenix Pharmaceuticals), DMI-3798 (DMI Biosciences, Angfomate (Ipsen), ZD-6474 (AstraZeneca), CEP-5214 (Cephalon), Canstatin (Genzyme), NM-3 (Mercian), Oridigm (MediQuest Therapeutics), Exherin (Adherex), BLS-0597 (Boston Life Sciences), PTC-299 (PTC Therapeutics), NPI-2358 (Nereus Pharmaceuticals), CGP-79787 (Novartis), AEE-788 (Novartis), CKD-732 (Chong Kun Dang), CP-564959 (OSI Pharmaceuticals), CM-101 (CarboMed), CT-2584, CT3501 (Cell Therapeutics), combretastatin and analogues and derivatives thereof (such as combretastatin A-1, A-2, A-3, A-4, A-5, A-6, B-1, B-2, B-3, B-4, D-1 , D-2, and combretastatin A-4 phosphate (Oxigene)), Rebimastat (Bristol-Meyers Squibb), Dextrin 2-sulfate (ML Laboratories), Cilengitide (Merk KGaA), NSC- 706704 (Pharminox), KRN-951 (Kirin Brewery), Ukrain, NSC-631570 (Nowicky Pharma), Tecogalan sodium (Daiichi Pharmaceutical), Tz-93 (Tsumura), TBC-1635 (Encysive Pharmaceuticals), TAN-1120 (Takeda), Semaxanib (Pfizer), BDI-7800 (Biopharmacopae), SD-186, SD-983 (Bristol- Meyers Squibb), SB-223245 (GlaxoSmithKline), SC-236 (Pfizer), RWJ- 590973 (Johnson and Johnson), ILX-1850 (Genzyme), SC-68488, S-836 (Pfizer), CG-55069-11 (CuraGen), Ki-23057 (Kirin Brewery), CCX-700 (Chemoentryx), Pegaptanib octasodium (Giled Sciences), or an analogue or derivative thereof). In other embodiments, the angiogenesis inhibitor may be a recombinant anti-angiogenic compound such as ANGIOCOL (available from Biostratum Inc., Durham, NC).
7) Apoptosis Antagonists
In another embodiment, the fibrosis-inhibiting compound is an apoptosis antagonist (e.g., didemnin B, RGB-286199 (GPC Biotech), 5F- DF-203 (Cancer Research Technology), aplidine, bongkrekic acid, triammonium salt, [6]-gingerol (CAS No. 23513-14-6), or an analogue or derivative thereof).
8) Apoptosis Activators
In another embodiment, the fibrosis-inhibiting compound is an apoptosis activator (e.g., aplidine (CAS No. 137219-37-5) (PharmaMar), canfosfamide hydrochloride (CAS No. 58382-37-74 and 39943-59-6) (Telik), idronoxil (CAS No. 81267-65-4) (Novogen), OSI-461 (OSI Pharmaceuticals), DE-098 (Santen), ARQ-550RP (ArQuIe), ABJ-879 (Novartis), adaphostin (NIH), anticancer agents from Apogenix Biotechnology and Momenta Pharmaceuticals, anti-PARP-1 or anti-PARP-2 (Octamer), BA-1037 (BioAxone), CP-248 (CAS No. 200803-37-8) (OSI Pharmaceuticals), EM- 1421 (Erimos), lPI-504 (Infinity Pharmaceuticals), KP-372-1 (QLT), MPC- 6827 (Maxim), MT-103 (Medisyn Technologies), MX-116407 or MX-126374 (Maxim), NPI-0052 (Nereus Pharmaceuticals), NVP-AEW541 (Novartis), PARP inhibitor from Agouron (Pfizer), R-306465 (Johnson & Johnson), TG- 100-33 (TargeGen), a XIAP inhibitor from AEgera, ZEN-011 (AEterna Zentaris), canertinib dihydrochloride (CAS No. 289499-45-2) (Pfizer), BH31- 1, 3-BAABE, or an analogue or derivative thereof). 9) Beta 1 Inteqrin Antagonist
In another embodiment, the fibrosis-inhibiting compound is a beta 1 integrin antagonist (e.g., β-1 integrin antagonists, Berkeley Lab, or an analogue or derivative thereof).
10) Beta Tubulin Inhibitor
In another embodiment, the fibrosis-inhibiting compound is a beta tubulin inhibitor (e.g., ZEN-017 (AEterna Zentaris), laulimalide (Kosan Biosciences), or an analogue or derivative thereof).
11) Blockers of Enzyme Production in Hepatitis C
In another embodiment, the fibrosis-inhibiting compound is an agent that blocks enzyme production in hepatitis C (e.g., merimepodib (Vertex Pharmaceuticals), or an analogue or derivative thereof).
12) Bruton's Tyrosine Kinase Inhibitor
In another embodiment, the fibrosis-inhibiting compound is a Bruton's tyrosine kinase inhibitor (e.g., a Btk inhibitor from Cellular Genomics, or an analogue or derivative thereof).
13) Calcineurin Inhibitors
In another embodiment, the fibrosis-inhibiting compound is a calcineurin inhibitor (e.g., tacrolimus (LifeCycle Pharma), or an analogue or derivative thereof).
14) Caspase 3 Inhibitors
In another embodiment, the fibrosis-inhibiting compound is a caspase 3 inhibitor (e.g., NM-3 (Mercian), or an analogue or derivative thereof). 15) CC Chemokine Receptor Antagonists
In another embodiment, the fibrosis-inhibiting compound is a CC chemokine receptor antagonist {e.g., a chemokine receptor 3 antagonist, a chemokine receptor 6 antagonist, and a chemokine receptor 7 antagonist). Representative examples of CC chemokine receptor antagonists include chemokine antagonists such as the CCR7 antagonists from Neurocrine Biosciences.
In a related embodiment, the fibrosis-inhibiting compound is a CC chemokine receptor antagonist (CCR) 1 , 3, & 5 (e.g., peptide T (Advanced lmmuni T), a CCR3 antagonist from GlaxoSmithKline, a chemokine antagonist (Pharmaprojects No. 6322) from Neurocrine Biosciences or Merck & Co., an HIV therapy agent from ReceptoPharm (Nutra Pharma), Pharmaprojects No. 6129 (Sangamo BioSciences), or an analogue or derivative thereof).
In certain embodiments, the CCCR antagonist is a CCR2b chemokine receptor antagonist such as RS 102895 (CAS No. 300815-41-2).
16) Cell Cycle Inhibitors
In another embodiment, the fibrosis-inhibiting compound is a cell cycle inhibitor (e.g., SNS-595 (Sunesis), homoharringtonine, or an analogue or derivative thereof).
In certain embodiments, the cell cycle inhibitor is an anti- microtubule agent (e.g., synthadotin, or an analogue or derivative thereof).
In certain embodiments, cell cycle inhibitor is a microtubule stimulant (e.g., KRX-0403, or an analogue or derivative thereof).
17) Cathepsin B Inhibitor
In another embodiment, the fibrosis-inhibiting compound is a cathepsin B inhibitor (e,g., AM-4299A (Asahi Kasei Pharma), BDI-7800 (Biopharmacopae), a cathepsin B inhibitor from Axys (Celera Genomics), MDL-104903 (CAS No. 180799-56-8) (Sanofi-Aventis), NC-700 (Nippon Chemiphar), Pharmaprojects No. 2332 (Hoffmann-La Roche), Pharmaprojects No. 4884 (Takeda), Pharmaprojects No. 5134 (Nippon Chemiphar), or an analogue or derivative thereof).
18) Cathepsin K Inhibitor
In another embodiment, the fibrosis-inhibiting compound is a cathepsin K inhibitor (e.g., 462795 (GlaxoSmithKline), INPL-022-D6 (Amura Therapeutics), or an analogue or derivative thereof).
19) Cathepsin L Inhibitor
In another embodiment, the fibrosis-inhibiting compound is a cathepsin L Inhibitor (e.g., a cathepsin L inhibitor from Takeda, INPL-022- E10 (Amura Therapeutics), Pharmaprojects No. 5447 (Taiho), or an analogue or derivative thereof).
20) CD40 Antagonists . .. . .
In another embodiment, the fibrosis-inhibiting compound is a CD40 antagonists (e.g., 5D12 (Chiron), ABI-793 (Novartis), an anticancer antibody from Chiron, anti-CD40 MAb-2 (Kirin Brewery), anti-CD40 (Eli Lilly), anti-CD40L antibody (UCB), a CD40 inhibitor from Apoxis, CD40 ligand inhibitor from Millennium Pharmaceuticals, a CD40/CAP inhibitor from Snow Brand, CGEN-40 (Compugen), CHIR-12.12 (Chiron), Pharmaprojects No. 5163 (Nippon Kayaku), ruplizumab (Biogen Idee), SGN-40 (Seattle Genetics), TNX-100 (Akzo Nobel), toralizumab (CAS No. 252662-47-8) (Biogen Idee), or an analogue or derivative thereof).
21) Chemokine Receptor Agonists
In another embodiment, the fibrosis-inhibiting compound is a chemokine receptor agonist (e.g., a chemokine agonist from NeuroTarget, or an analogue or derivative thereof). 22) Chymase inhibitors
In another embodiment, the fibrosis-inhibiting compound is a chymase inhibitor {e.g., BL-3875 (Dainippon), LEX-043 (SuperGen), NK- 3201 (CAS No. 204460-24-2) (Nippon Kayaku), or an analogue or derivative thereof).
23) Collaαenase (Interstitial) Antagonists
In another embodiment, the fibrosis-inhibiting compound is a collagenase (interstitial) antagonist (e.g., IBFB-212543 (IBFB Pharma), Pharmaprojects No. 3762 (Sanofi-Aventis), S-0885 (CAS No. 117517-22-3) (Sanofi-Aventis), SC-40827 (CAS No. 101470-42-2) (Pfizer), or an analogue or derivative thereof).
24) CXCR (2. 4) Antagonists
In another embodiment, the fibrosis-inhibiting compound is a CXCR (2, 4) antagonist (e.g., SB-656933 (GlaxoSmithKline), AMD3100 octahydrochloride (CAS No. 155148-31-5), or an analogue or derivative thereof).
25) Cvclin Dependent Kinase Inhibitors
In another embodiment, the fibrosis-inhibiting compound is a cyclin dependent kinase (CDK) inhibitor. In certain embodiments, the cyclin dependent kinase inhibitor is a CDK-1 inhibitor. In certain embodiments, the cyclin dependent kinase inhibitor is a CDK-2 inhibitor. In certain embodiments, the cyclin dependent kinase inhibitor is a CDK- 4 inhibitor. In certain embodiments, the cyclin dependent kinase inhibitor is a CDK-6 inhibitor. Representative examples of cyclin dependent kinase inhibitors include CAK1 inhibitors from GPC Biotech and Bristol-Myers Squibb, RGB- 286199 (GPC Biotech), or an analogue or derivative thereof.
Additional exemplary cyclin dependent protein kinase inhibitors include an anticancer agent from Astex Technology, a CAK1 inhibitor from GPC Biotech, a CDK inhibitor from Sanofi-Aventis, a CDK1/CDK2 inhibitor from Amgen, a CDK2 inhibitor from SUGEN-2 (Pfizer), a hearing loss therapy agent (Sound Pharmaceuticals), PD-0332991 (Pfizer), RGB-286199 (GPC Biotech), Ro-0505124 (Hoffmann-La Roche), a Ser/Thr kinase inhibitor from Lilly (EIi Lilly), CVT-2584 (CAS No. 199986-75- 9) (CV Therapeutics), CGP 74514A, bohemine, olomoucine (CAS No. 101622-51-9), indole-3-carbinol (CAS No. 700-06-1), and an analogue or derivative thereof.
26) Cvclooxyqenase Inhibitors
In another embodiment, the fibrosis-inhibiting compound is a cyclooxygenase inhibitor (e.g., NS-398 (CAS No. 123653-11-2), ketoprofen, or an analogue or derivative thereof). In some embodiments, the cyclooxygenase inhibitor is a COX-1 inhibitor such as triflusal, or an analogue or derivative thereof).
27) Dihydroorotate Dehydrogenase Inhibitor (PHFR) Inhibitors In another embodiment, the fibrosis-inhibiting compound is a
DHFR inhibitor (e.g., PDX (Allos Therapeutics), SC12267, sulfamerazine (CAS No. 127-79-7), or an analogue or derivative thereof).
28) Dual lntegrin Inhibitor
In another embodiment, the fibrosis-inhibiting compound is a dual integrin inhibitor (e.g., R411 (Roche Pharmaceuticals), or an analogue or derivative thereof).
29) Elastase Inhibitors
In another embodiment, the fibrosis-inhibiting compound is an elastase inhibitor (e.g., orazipone, depelestat (CAS No. 506433-25-6) (Dyax), AE-3763 (Dainippon), or an analogue or derivative thereof). 30) Elongation Factor-1 Alpha Inhibitors
In another embodiment, the fibrosis-inhibiting compound is an elongation factor-1 alpha inhibitor (e.g., aplidine, or an analogue or derivative thereof).
31) Endothelial Growth Factor Antagonists
In another embodiment, the fibrosis-inhibiting compound is an endothelial growth factor (EGF) antagonist (e.g., neovastat, NM-3 (Mercian), or an analogue or derivative thereof).
32) Endothelial Growth Factor Receptor Kinase Inhibitors
In another embodiment, the fibrosis-inhibiting compound is an endothelial growth factor receptor (EGF-R) kinase inhibitor (e.g., sorafenib tosylate (Bayer), AAL-993 (Novartis), ABP-309 (Novartis), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EXEL-2880 (Exelixis), GW-654652 (GlaxoSmithKline), lavendustin A (CAS No. 125697-92-9), a KDR inhibitor from LG Life Sciences, CT-6685 or CT-6729 (UCB)1 KRN-633 or KRN-951 (Kirin Brewery), OSI-930 (OSI Pharmaceuticals), SP-5.2 (Supratek Pharma), SU- 11657 (Pfizer), a Tie-2 antagonist (Hybrigenics), a VEGF-R inhibitor such as SU 1498, a VEGFR-2 kinase inhibitor (Bristol-Myers Squibb), XL-647 (Exelixis), a KDR inhibitor from Abbott Laboratories, or an analogue or derivative thereof).
In another embodiment, the fibrosis-inhibiting compound is an endothelial growth factor receptor 2 kinase inhibitor (e.g., sorafenib tosylate, or an analogue or derivative thereof).
33) Endotoxin Antagonists
In another embodiment, the fibrosis-inhibiting compound is an endotoxin antagonist (e.g., E5564 (Eisai Pharmaceuticals), or an analogue or derivative thereof). 34) Epothilone and Tubulin Binders
In another embodiment, the fibrosis-inhibiting compound is an epothilone or tubulin binder (e.g., ixabepilone (BMS), or an analogue or derivative thereof).
35) Estrogen Receptor Antagonists
In another embodiment, the fibrosis-inhibiting compound is an estrogen receptor antagonist (e.g., ERB-041 (Wyeth), or an analogue or derivative thereof).
36) FGF Inhibitors
In another embodiment, the fibrosis-inhibiting compound is a FGF inhibitor (e.g., IDN-5390 (Indena), or an analogue or derivative thereof).
37) Farnexy) Transferase Inhibitors
In another embodiment, the fibrosis-inhibiting compound is an inhibitor of farnexyl transferase (FTI). In certain embodiments, the FTI inhibits the RAS oncogene family. Examples of FTI's include SARASAR (from Schering Corporation, Kenilworth, NJ), or an analogue or derivative thereof.
38) Famesyltransferase Inhibitors
In another embodiment, the fibrosis-inhibiting compound is a famesyltransferase inhibitor (e.g., A-197574 (Abbott), a famesyltransferase inhibitor from Servier, FPTIII (Strathclyde Institute for Drug R), LB-42908 (LG Life Sciences), Pharmaprojects No. 5063 (Genzyme), Pharmaprojects No. 5597 (Ipsen), Yissum Project No. B-1055 (Yissum), or an analogue or derivative thereof). 39) FLT-3 Kinase Inhibitors
In another embodiment, the fibrosis-inhibiting compound is a FLT-3 kinase inhibitor (e.g., Amphora, or an analogue or derivative thereof).
40) FGF Receptor Kinase Inhibitors
In another embodiment, the fibrosis-inhibiting compound is a FGF receptor kinase inhibitor (e.g., MED-A300 (Gerolymatos), SSR-128129 (Sanofi-Aventis), TBC-2250 (Encysive Pharmaceuticals), XL-999 (Exelixis), or a FGF receptor kinase inhibitor from Paradigm Therapeutics, or an analogue or derivative thereof).
41) Fibrinogen Antagonists
In another embodiment, the fibrosis-inhibiting compound is a fibrinogen antagonist (e.g., AUV-201 (Auvation), MG-13926 (Sanofi- Aventis), plasminogen activator (CAS No. 105913-11-9) (from Sanofi- Aventis or UCB), plasminogen activator-2 (tPA-2) (Sanofi-Aventis), pro- urokinase (CAS No. 82657-92-9) (Sanofi-Aventis), mevastatin, or an analogue or derivative thereof).
42) Heat Shock Protein 90 Antagonists
In another embodiment, the fibrosis-inhibiting compound is a heat shock protein 90 antagonist (e.g., SRN-005 (Sirenade), geldanamycin or a derivative thereof, such as NSC-33050 (17-allylaminogeldanamycin; 17-AAG) or 17-dimethylaminoethylamino-17-demethoxy-geldanamycin (17- DMAG), rifabutin (rifamycin XIV, 1\4-didehydro-1-deoxy-1 ,4-dihydro-5'-(2- methylpropyl)-1-oxo-), radicicol, Humicola fuscoatra (CAS No. 12772-57-5), or an analogue or derivative thereof).
43) Histone Deacetylase Inhibitors
In another embodiment, the fibrosis-inhibiting compound is a histone deacetylase inhibitor (e.g., FK228 (Gloucester), trichostatin A from Streptomyces sp. (CAS No. 58880-19-6), or an analogue or derivative thereof).
44) HMGCoA Reductase Inhibitors
In another embodiment, the fibrosis-inhibiting compound is an HMGCoA reductase inhibitor (e.g., an atherosclerosis therapeutic from Lipid Sciences, ATΪ-16000 (ARYx Therapeutics), KS-01-019 (Kos Pharmaceuticals), Pharmaprojects No. 2197 (Sanofi-Aventi), RP 61969 (Sanofi-Aventis), cerivastatin Na )CAS No. 143201-11-0), or an analogue or derivative thereof).
45) ICAM Inhibitors
In another embodiment, the fibrosis-inhibiting compound is an ICAM inhibitor (e.g., alicaforsen (CAS No. 185229-68-9) (ISIS Pharmaceuticals), an ICAM-5 modulator (such as ICAM-4 from ICOS), or an analogue or derivative thereof). .. . ~
46) IL-1 , ICE & IRAK Antagonists
In another embodiment, the fibrosis-inhibiting compound is an IL-1 , ICE & IRAK antagonist (e.g., CJ-14877 or CP-424174 (Pfizer), NF-61 (Negma-Lerads), or an analogue or derivative thereof).
47) IL-2 Inhibitors
In another embodiment, the fibrosis-inhibiting compound is an IL-2 inhibitor (e.g., AVE 8062 (Sanofi-Aventis), or an analogue or derivative thereof).
48) Immunosuppressants
Jn another embodiment, the fibrosis-inhibiting compound is an immunosuppressant (e.g., teriflunomide (Sanofi Aventis), chlorsulfaquinoxalone (NSC-339004), chlorsulfaquinoxalone sulfate, CS-712 (Sankyo), ismomυltin alfa (CAS No. 457913-93-8) (Akzo Nobel), antiallergics from GenPat77, antiinflammatories or AT-005 (Androclus Therapeutics), autoimmune disease therapeutics from EpiVax, BN-007 (Bone), budesonide (CAS No. 51333-22-3) (MAP Pharmaceuticals), CO-14 (Genzyme), edratide (CAS No. 433922-67-9) (Teva), EP-314 (Enanta), eprovafen (CAS No. 101335-99-3) (Sanofi-Aventis), HWA-131 (CAS No. 118788-41-3) (Sanofi-Aventis), immunomodulators from MerLion Pharmaceuticals, immunosuppressives from Alchemia, IPL-12 (Inflazyme), MDL-9563 (CAS No. 27086-86-8) (Sanofi-Aventis), Pharmaprojects No. 2330 (Sanofi-Aventis), Pharmaprojects No. 6426 (Abgenix), PXS-25 (Pharmaxis), rosmarinic acid (CAS No. 20283-92-5) (Sanofi-Aventis), RP 42927 or RP 54745 (CAS No. 135330-08-4) (Sanofi-Aventis), SGN-35 (Seattle Genetics), ST-1959 (Sigma-Tau), type I diabetes therapy from SYNX Pharma, UNIL-88 (Debiopharm), VP-025 (Vasogen), VR-694 (Vectura), PRTX-001 (Protalex), or an analogue or derivative thereof).
49) IMPDH (inosine monophosphate)
In another embodiment, the fibrosis-inhibiting compound is IMPDH (inosine monophosphate) (e.g., ribavirin (Hoffmann-La Roche) or an analogue or derivative thereof).
50) Inteqrin Antagonists
In another embodiment, the fibrosis-inhibiting compound is an integrin antagonist (e.g., 683699 from Glaxo Smith Kline, integrin antagonists from Jerina AG (Germany), or an analogue or derivative thereof).
51) lnterleukin Antagonists
In another embodiment, the fibrosis-inhibiting compound is an interleukin antagonist (e.g., dersalazine, or an analogue or derivative thereof). In another embodiment, the fibrosis-inhibiting compound is an interleukin 1 antagonist (e.g., NPI-1302a-3, or an analogue or derivative thereof).
52) Inhibitors of Type III Receptor Tyrosine Kinases
In another embodiment, the fibrosis-inhibiting compound is an inhibitor of type III receptor tyrosine kinase such as FLT3, PDGRF and c-KIT (e.g., MLN518 (Millenium Pharmaceuticals), or an analogue or derivative thereof).
53) Irreversible Inhibitors of Enzyme Methionine Aminopeptidase Type 2
In another embodiment, the fibrosis-inhibiting compound is an irreversible inhibitor of enzyme methionine aminopeptidase type 2 (e.g., PPI-2458 (Praecis Pharmaceuticals), or analogue or derivative thereof).
54) Isozvme-Selective Delta Protein Kinase C Inhibitors
In another embodiment, the fibrosis-inhibiting compound is an isozyme-selective delta protein kinase C inhibitor (e.g., KAI-9803 (Kai Pharmaceuticals), or an analogue or derivative thereof).
55) JAK3 Enzyme Inhibitors
In another embodiment, the fibrosis-inhibiting compound is a JAK3 enzyme inhibitor (e.g., CP-690,550 (Pfizer), or an analogue or derivative thereof).
56) JNK Inhibitors
In another embodiment, the fibrosis-inhibiting compound is a JNK inhibitor (e.g., BF-67192 (BioFocus), XG-101 or XG-102 (Xigen), or an analogue or derivative thereof). 57) Kinase Inhibitors
In another embodiment, the fibrosis-inhibiting compound is a kinase inhibitor (e.g., a kinase inhibitors from EVOTEC, or an analogue or derivative thereof).
58) Kinesin Antagonist
In another embodiment, the fibrosis-inhibiting compound is a kinesin antagonist (e.g., SB-715992 and an antifungal from Optokinetics, or an analogue or derivative thereof).
59) Leukotriene Inhibitors and Antagonists
In another embodiment, the fibrosis-inhibiting compound is a leukotriene inhibitor or antagonist (e.g., ambicromil (CAS No. 58805-38-2) (Sanofi-Aventis), amelubant (CAS No. 346735-24-8) (Boehringer Ingelheim), DW-1141 (Dong Wha), ebselen (Daiichi Pharmaceutical), ibudilast (Kyorin), leucotriene inhibitors from Sanofi-Aventis, lymphotoxin - beta receptor (LT-β) from Biogen Idee, Pharmaprojects No. 1535 or 2728 (CAS No. 119340-33-9) (Sanofi-Aventis), R-112 (Rigel), Rev-5367 (CAS No. 92532-05-3) (Sanofi-Aventis), RG-14893 (CAS No. 141835-49-6) (Sanofi- Aventis), RG-5901-A (CAS No. 101910-24-1), 92532-23-5, RP 66153 (CAS No. 142422-79-5), RP 66364 (CAS No.186912-92-5), or RP 69698 (CAS No. 141748-00-7) (Sanofi-Aventis), SC-411930 (Pfizer), SC-41930 (CAS No. 120072-59-5) (Pfizer), SC-50605 (CAS No. 138828-39-4) (Pfizer), SC- 51146 (CAS No. 141059-52-1) or SC-53228 (CAS No. 153633-01-3) (Pfizer), spaglumic acid (ZY-15106) (CAS No.3106-85-2) or 80619-64-3 (Novartis), tipredane (CAS No. 85197-77-9) (Bristol-Myers Squibb), U- 75302 (CAS No. 119477-85-9) (Pfizer), or analogue or derivative thereof).
60) MAP Kinase Inhibitors
In another embodiment, the fibrosis-inhibiting compound is a MAP kinase inhibitor (e.g., SRN-003-556 (Sirenade), AEG-3482 (AEgera), ARRY-142886 (Array BioPharma), CDP-146 (UCB), or analogue or derivative thereof).
61) Matrix Metalloproteinase Inhibitors (MMPI)
In another embodiment, the fibrosis-inhibiting compound is a matrix metalloproteinase inhibitor. A variety of MMPI's may be used in the practice of the invention. In one embodiment, the MMPI is a MMP-1 inhibitor. In another embodiment, the MMPI is a MMP-2 inhibitor. In other embodiments, the MMPI is a MMP-4, MMP-5, MMP-6, MMP-7, or MMP-8 inhibitor. Representative examples of MMPI's include glucosamine sulfate, neovastat, GM1489 (CAS No. 170905-75-6), XL784 (EXEL-01370784), TNF-a Protease lnhibitor-1 or 2 (TAPM or TAPI-2), galardin, or an analogue or derivative thereof.
62) MCP- CCR2 Inhibitors
In another embodiment, the fibrosis-inhibiting compound is a MCP-CCR2 inhibitor (e.g., MLN 1202 (Millennium Pharmaceuticals), or an analogue or derivative thereof).
63) mTOR Inhibitor
In another embodiment, the fibrosis-inhibiting compound is an mTOR inhibitor {e.g., temsirolimus (CAS No. 162635-04-3) (Wyeth), or an analogue or derivative thereof).
64) mTOR Kinase Inhibitor
In another embodiment, the fibrosis-inhibiting compound is an mTOR kinase inhibitor (e.g., ABT-578 (Abbott), temsirolimus (Wyeth), AP- 23573 (Ariad), or an analogue or derivative thereof). 65) Microtubule Inhibitors
In another embodiment, the fibrosis-inhibiting compound is a microtubule inhibitor {e.g., antibody-maytansinoid conjugates from Biogen Idee, colchicines (MantiCore Pharmaceuticals), anticancer immunoconjugates from Johnson & Johnson, DIME from Octamer, gni-1f (GNI), huC242-DM4 or huMy9-6-DM1 (ImmunoGen), IDN-5404 (Indena), IMO-098 or IMOderm (Imotep), mebendazole (Introgen Therapeutics), microtubule poisons from Cambridge Enterprise, paclitaxel such as LOTAX from Aphios (CAS No. 33069-62-4), Genexol-PM from Samyang, Pharmaprojects No. 6383 (Tapestry Pharmaceuticals), RPR-112378 (Sanofi-Aventis), SGN-75 (Seattle Genetics), SPL-7435 (Starpharma), SSR- 250411 (Sanofi-Aventis), trastuzumab-DM1 (Genentech), vinorelbine, dolastatin 15 (CAS No. 123884-00-4) or an analogue or derivative thereof).
In certain embodiments, the microtubule inhibitor is a microtubule polymerization inhibitor such as vincamine, or an analogue or derivative thereof).
66) MIF Inhibitors
In another embodiment, the fibrosis-inhibiting compound is a MIF inhibitor (e.g., AVP-13546 (Avanir), an MIF inhibitor from Genzyme, migration stimulation factor D, or an analogue or derivative thereof).
67) MMP (Stromolvsin) Inhibitors
In another embodiment, the fibrosis-inhibiting compound is a MMP (stromolysin) inhibitor (e.g., anticancer tetracycline from Tetragenex, rhostatin (BioAxone), TIMP's from Sanofi-Aventis (CAS No. 86102-31-0), and MMP inhibitors form Cognosci and Tetragenex, or an analogue or derivative thereof). 68) Neurokinin (NK) Antagonist
In another embodiment, the fibrosis-inhibiting compound is a neurokinin (NK) antagonist (e.g., anthrotainin (CAS No. 148084-40-6) (Sanofi-Aventis), an IBS thereapeutic such as SLV-332 from ArQuIe, MDL- 105212A (CAS No. 167261-60-1) (Ssanofi-Aventis), Pharmaprojects No. 2744, 3258 (CAS No. 139167-47-8) 4006, 4201 , or 5986 (Sanofi-Aventis), RP 67580 (CAS No. 135911-02-3), SR-144190 (CAS No. 201152-86-5), SSR-240600 or SSR-241586 (Sanofi-Aventis), TKA-457 (Novartis), vestipitant mesylate (CAS No. 334476-64-1) (GlaxoSmithKline), Win-64821 (Sanofi-Aventis), PRX-96026 (Predix Pharmaceuticals), or an analogue or derivative thereof).
69) NF kappa B Inhibitors
In another embodiment, the fibrosis-inhibiting compound is a NF kappa B (NFKB) inhibitor (e.g., emodin (CAS No. 518-82-1), AVE-0545 or AVE-0547 (Sanofi-Aventis), bortezomib (CAS No. 179324-69-7) (Millennium Pharmaceuticals), dexanabinol (CAS No. 112924-45-5) (Pharmos), dexlipotam (Viatris), Pharmaprojects No. 6283 (INDRA) (OXiGENE), IPL-576092 (CAS No. 137571-30-3) (Inflazyme), NFKB decoy (Corgentech), NFKB decoy oligo (AnGes MG), NFKB's from Ariad, osteoporosis treatments or S5 (F005) from Fulcrum Pharmaceuticals, P61 (Phytopharm), R-flurbiprofen (CAS No. 5104-49-4) (Encore Pharmaceuticals), Bay 11-7085, or an analogue or derivative thereof).
70) Nitric Oxide Agonists
In another embodiment, the fibrosis-inhibiting compound is a nitric oxide agonist (e.g., Acclaim, Angx-1039 or Angx-3227 (Angiogenix), CAS-1609 (CAS No. 158590-73-9) (Sanofi-Aventis), GCI-503 (Spear Therapeutics), HCT-3012 (CAS No. 163133-43-5) (NicOx), hydralazine + ISDN (NitroMed), isosorbide dinitrate, Diffutab (CAS No. 87-33-2) (Eurand), isosorbide mononitrate (CAS No. 16051-77-7) from AstraZeneca, Schering AGor Schwarz Pharma, LA-419 (Lacer), molsidomine (CAS No. 25717-80- 0) (from Takeda and Therabel), NCX-1000, NCX-2057, or NCX-4040 (NicOx), nitric oxide (ProStrakan), nitroglycerin in the form of a nitroglycerin patch, such as DERMATRANS from (Rottapharm), nitroglycerin (CAS No. 55-63-0) (from Cellegy Pharmaceuticals, Forest Laboratories, NovaDel, Schwarz Pharma, and Watson), NO-releasing prodrugs (Inotek), OM-294DP (OM PHARMA), oxdralazine (CAS No. 27464-23-9) (Sanofi-Aventis), pirsidomine (CAS No. 132722-74-8) (Sanofi-Aventis), prostaglandin and NO donor (Cellegy Pharmaceuticals), upidosin derivatives (Recordati), or an analogue or derivative thereof).
71) Ornithine Decarboxylase Inhibitors
In another embodiment, the fibrosis-inhibiting compound is an ornithine decarboxylase inhibitor {e.g., aplidine, or an analogue or derivative thereof).
72) p38 MAP Kinase Inhibitors
In another embodiment, the fibrosis-inhibiting compound is a p38 MAP kinase inhibitor (e.g., AZD-6703 (AstraZeneca), JX-401 (Jexys Pharmaceuticals), BMS-2 (Bristol-Myers Squibb), a p38 MAP kinase inhibitor from Novartis, a p38-alpha MAP kinase inhibitor from Amphora, Pharmaprojects No. 5704 (Pharmacopeia), SKF86002 (CAS No. 72873-74- 6), RPR-200765A (Sanofi-Aventis), SD-282 (Johnson & Johnson), TAK-715 (Takeda), or an analogue or derivative thereof).
73) Palmitoyl-Protein Thioesterase Inhibitors
In another embodiment, the fibrosis-inhibiting compound is a palmitoyl-protein thioesterase inhibitor (e.g., aplidine, or an analogue or derivative thereof). 74) PDGF Receptor Kinase Inhibitors
In another embodiment, the fibrosis-inhibiting compound is a PDGF receptor kinase inhibitors (e.g., AAL-993, AMN-107, or ABP-309 (Novartis), AMG-706 (Amgen), BAY-57-9352 (Bayer), CDP-860 (UCB), E- 7080 (Eisai), imatinib (CAS No. 152459-95-5) (Novartis), OSI-930 (OSI Pharmaceuticals), RPR-127963E (Sanofi-Aventis), RWJ-540973 (Johnson & Johnson), sorafenib tosylate (Bayer), SU-11657 (Pfizer), tandutinib (CAS No. 387867-13-2) (Millennium Pharmaceuticals), vatalanib (Novartis), ZK- CDK (Schering AG), or an analogue or derivative thereof).
75) Peroxisome Proliferator-Activated Receptor Agonists
In another embodiment, the fibrosis-inhibiting compound is a peroxisome proliferator-activated receptor (PPAR) agonists (e.g., (-)- halofenate (Metabolex), AMG-131 (Amgen), antidiabetics from Japan Tobacco, AZD-4619, AZD-8450, or AZD-8677 (AstraZeneca), DRF-10945 or balaglitazone (Dr Reddy's), CS-00088 or CS-00098 (Chipscreen Biosciences), E-3030 (Eisai), etalocib (CAS No. 161172-51-6) (EIi Lilly), GSK-641597 (Ligand), GSK-677954 (GlaxoSmithKline), GW-409544 (Ligand), GW-590735 (GlaxoSmithKline), K-111 (Hoffmann-La Roche), LY- 518674 (EIi Lilly), LY-674 (Ligand), LY-929 (Ligand), MC-3001 or MC-3002 (MaxoCore Pharmaceuticals), metformin HCI + pioglitazone (CAS No. 1115- 70-4 and 112529-15-4) (such as ACTOPLUS MET from Andrx), muraglitazar (CAS No. 331741-94-7) (Bristol-Myers Squibb), naveglitazar (Ligand), oleoylethanolamide (Kadmus Pharmaceuticals), ONO-5129, pioglitazone hydrochloride (CAS No. 111025-46-8 and 112529-15-4) (Takeda), PLX-204 (Plexxikon), PPAR agonists from Genfit, PPAR delta agonists from EIi Lilly, PPAR-alpha agonists from CrystalGenomics, PPAR- gamma modulators and PPAR-β modulators from CareX, rosiglitazone maleate (CAS No. 122320-73-4 or 155141-29-0) (GlaxoSmithKline), rosiglitazone maleate/glimepir (CAS No. 155141-29-0 and 93479-97-1), such as AVANDARYL or rosiglitazone maleate/metformin extend (CAS No. 155141-29-0 and 657-24-9) such as AVANDAMET, or rosiglitazone maleate+metformin, such as AVANDAMET (GlaxoSmithKline), tesaglitazar (AstraZeneca), LBM642, WY-14,643 (CAS No. 50892-23-4), or an analogue or derivative thereof).
In certain embodiments, the PPAR Agonist is a PPARα agonist such as GW7647 or fenofibric acid (CAS No. 42017-89-0), a PPAR γ agonist such as MCC-555 (CAS No. 161600-01-7), GW9662 or GW1929, a PPAERδ agonist such as GW501516, a PPARβ and PPARδ agonist such L- 165,041 (CAS No. 79558-09-1), or an analogue or derivative thereof.
76) Phosphatase Inhibitor
In another embodiment, the fibrosis-inhibiting compound is a phosphatase inhibitor (e.g., diabetes thereapy such as SQMO3, SQDM38, SQDM60 from Sequenom, Pharmaprojects No. 4191 (Sanofi-Aventis), PRL- 3 inhibitors from Genzyme, WIP1 inhibitors from Amgen, or an analogue or derivative thereof).
77) Phosphodiesterase (PDE) Inhibitors
In another embodiment, the fibrosis-inhibiting compound is a phosphodiesterase (PDE) inhibitor (e.g., avanafil (Tanabe Seiyaku), dasantafil (CAS No. 569351-91-3) (Schering-Plough), A-906119 (CAS No. 134072-58-5) or DL-850 (Sanofi-Aventis), GRC-3015, GRC-3566, or GRC- 3886 (Glenmark), HWA-153 (CAS No. 56395-66-5) (Sanofi-Aventis), hydroxypumafentrine (Altana), IBFB-130011, IBFB-14-016, IBFB-140301 , IBFB-150007, or IBFB-211913 (IBFB Pharma), L-826141 (Merck & Co), medorinone (CAS No. 88296-61-1) (Sanofi-Aventis), MEM-1917 (Memory Pharmaceuticals), ND-1251 (Neuro3d), PDE inhibitors from ICOS, PDE IV inhibitors from Memory Pharmaceuticals and CrystalGenomics, Pharmaprojects No. 2742 and 6141 (Sanofi-Aventis), QAD-171 (Novartis), RHC-2963 (CAS No. 76993-12-9 and 76993-14-1), RPR-117658, RPR- 122818 derivatives, SR-24870 , and RPR-132294 (Sanofi-Aventis), SK-350 (ln2Gen), stroke therapy agents from deCODE Genetics, TAS-203 (Taiho), tofimilast (CAS No. 185954-27-2) (Pfizer), UK-371800 (Pfizer), WIN-65579 (CAS No. 158020-82-7) (Sanofi-Aventis), IBFB-130020 (IBFB Pharma), OPC-6535 (CAS No. 145739-56-6) (Otsuka), theobromine (CAS No. 83-67- 0), papverine hydrochloride (CAS No. 61-25-6), quercetin dehydrate (CAS No. 6151-25-3), YM 976 (CAS No. 191219-80-4), irsogladine (CAS No. 57381-26-7), or an analogue or derivative thereof).
In one embodiment, the phosphodiesterase inhibitor is a phosphodiesterase III inhibitor (e.g., enoximone, or an analogue or derivative thereof). In other embodiments, the phosphodiesterase inhibitor is a phosphodiesterase IV inhibitor (e.g., fosfosal, Atopik (Barrier Therapeutics), triflusal, or an analogue or derivative thereof). In other embodiments, the phosphodiesterase inhibitor is a phosphodiesterase V inhibitor.
78) PKC Inhibitor
In another embodiment, the fibrosis-inhibiting compound is a PKC inhibitor (e.g., HMR-105509 or P-10050 (Sanofi-Aventis), JNJ- 10164830 (Johnson & Johnson), Ro-31-8425 (CAS No. 131848-97-0), NPC- 15437 dihydrochloride (CAS No. 136449-85-9), or an analogue or derivative thereof).
In one embodiment, the PKC inhibitor is an inhibitor of PKC beta (e.g., ruboxistaurin (EIi Lilly), or an analogue or derivative thereof).
79) Platelet Activating Factor Antagonists
In another embodiment, the fibrosis-inhibiting compound is a platelet activating factor antagonist (e.g., dersalazine, or an analogue or derivative thereof). 80) Platelet-Derived Growth Factor Receptor Kinase Inhibitors In another embodiment, the fibrosis-inhibiting compound is a platelet-derived growth factor receptor kinase inhibitor (e.g., sorafenib tosylate, Raf or Ras inhibitors such as sorafenib tosylate from Bayer and Onyx Pharmaceuticals, or an analogue or derivative thereof).
81) Prolyl Hydroxylase Inhibitors
In another embodiment, the fibrosis-inhibiting compound is a prolyl hydroxylase inhibitor (e.g., FG-2216 (CAS No. 11096-26-7) or HIF agonists from FibroGen, or an analogue or derivative thereof).
82) Polymorphonuclear Neutrophil Inhibitors
In another embodiment, the fibrosis-inhibiting compound is a polymorphonuclear neutrophil inhibitor (e.g., orazipone, or an analogue or derivative thereof).
83) Protein Kinase B Inhibitors
In another embodiment, the fibrosis-inhibiting compound is a protein kinase B inhibitor (e.g., Akt-1 inhibitors from Amphora, or an analogue or derivative thereof).
84) Protein Kinase C Stimulants
In another embodiment, the fibrosis-inhibiting compound is a protein kinase C stimulant (e.g., bryostatin-1 , or analogue or derivative thereof).
85) Purine Nucleoside Analogues
In another embodiment, the fibrosis-inhibiting compound is a purine nucleoside analogue (e.g., cladrinbine and formulations thereof, such as MYLINAX from Serone SA and IVAX Research Inc. (Miami, FL), or an analogue or derivative thereof). 86) Purinoreceptor P2X Antagonist
In another embodiment, the fibrosis-inhibiting compound is a purinoreceptor P2X antagonist (e.g., AZD-9056 (AstraZeneca), R-1554 (Hoffmann-La Roche), AR-C118925XX (AstraZeneca), suramin (CAS No. 129-46-4), P2Y4 receptor from Euroscreen, or an analogue or derivative thereof).
87) Raf Kinase Inhibitors
In another embodiment, the fibrosis-inhibiting compound is a Raf kinase inhibitor (e.g., sorafenib tosylate, or an analogue or derivative thereof).
88) Reversible Inhibitors of ErbB1 and ERbB2
In another embodiment, the fibrosis-inhibiting compound is a reversible inhibitor (e.g., lapatinib (GSK), or an analogue or derivative thereof).
89) Ribonucleoside Triphosphate Reductase Inhibitors
In another embodiment, the fibrosis-inhibiting compound is a cytoplasmic tyrosine kinase inhibitor such as a SRC inhibitor (e.g., SRN-004 (Sirenade), gallium maltolate (Titan Pharmaceutcals), or an analogue or derivative thereof), or an analogue or derivative thereof).
90) SDF-1 Antagonists
In another embodiment, the fibrosis-inhibiting compound is a SDF-1 antagonist (e.g., CTCE-9908 (Chemokine Therapeutics), or an analogue or derivative thereof).
91) Sheddase Inhibitor In another embodiment, the fibrosis-inhibiting compound is a sheddase inhibitor (e.g., INCB-7839 (Incyte Corporation), or an analogue or derivative thereof).
92) SRC Inhibitors
In another embodiment, the fibrosis-inhibiting compound is a SRC inhibitor (e.g., SRN-004 (Sirenade), or an analogue or derivative thereof).
In certain embodiments, the SRC inhibitor is a SRC kinase inhibitor (e.g., AZD0530 (AstraZeneca), or an analogue or derivative thereof).
93) Stromelvsin Inhibitors
In another embodiment, the fibrosis-inhibiting compound is a stromelysin inhibitor (e.g., glucosamine sulfate, or an analogue or derivative thereof).
94) Syk Kinase Inhibitors
In another embodiment, the fibrosis-inhibiting compound is a syk kinase inhibitor (e.g., R406 (Rigel), or an analogue or derivative thereof).
95) Telomerase Inhibitors
In another embodiment, the fibrosis-inhibiting compound is a telomerase inhibitor (e.g., AS-1410 (Antisoma), or an analogue or derivative thereof).
96) TGF Beta Inhibitors
In another embodiment, the fibrosis-inhibiting compound is a TGF beta inhibitor (e.g., pirfenidone (CAS No. 53179-13-8) (MARNAC), tranilast (CAS No. 53902-12-8) (Kissei), IN-1130 (ln2Gen), mannose-6- phosphate (BTG), TGF-β antagonists from Inflazyme (Pharmaprojects No. 6075), TGF-β antagonists (e.g., 1090 and 1091 from Sydney; non-industrial source), TGF-β I receptor kinase inhibitors from EIi Lilly, TGF-β receptor inhibitors from Johnson & Johnson, or an analogue or derivative thereof).
97) TNFα Antagonists and TACE Inhibitors
In another embodiment, the fibrosis-inhibiting compound is a TNFα antagonist or TACE inhibitors (e.g., adalimumab (CAS No. 331731- 18-1) (Cambridge Antibody Technology), AGIX-4207 (AtheroGenics), AGT-1 (Advanced Biotherapy), an anti-inflammatory from Borean Pharma, Cellzome, or Paradigm Therapeutics, anti-inflammatory vaccine (TNF-alpha kinoid) from Neovacs, humanized anti-TNF antibody or an anti-TNF MAb (CB0006) Celltech (UCB), apratastat (CAS No. 287405-51-0) (Wyeth), BMS-561392 (Bristol-Myers Squibb), BN-006 (Bone), certolizumab pegol (CAS No. 428863-50-7 or CH-138 (UCB), cilomilast (CAS No. 153259-65-5) (GlaxoSmithKline), CFM (Nuada Pharmaceuticals), CRx- 11.9 (CombinatoRx), D-5410 (UCB), dacopafant (CAS No. 125372-33-0) (Sanofi- Aventis), dersalazine (CAS No. 188913-57-7/188913-58-8) (Uriach), etanercept (CAS No. 185243-69-0) (Amgen), ethyl pyruvate (Critical (Critical Therapeutics), golimumab (CAS No. 476181-74-5) (Johnson & Johnson), hormono-immunotherapy from Ipsen, CDP571 (e.g., humicade from UCB), IC-485 (ICOS), infliximab (CAS No. 170277-31-3) (Johnson & Johnson), IP- 751 (Manhattan Pharmaceuticals), ISIS-104838 (CAS No. 250755-32-9) (ISIS Pharmaceuticals), lenalidomide (CAS No. 191732-72-6) (Celgene), lentinan (CAS No. 37339-90-5) (Ajinomoto), MDL-201112 (CAS No. 142130-73-2) (Sanofi-Aventis), medroxyprogesterone (CAS No. 520-85-4) (InKine Pharmaceutical), N-acetylcysteine (CAS No. 616-91-1) (Zambon), NBE-P2 (DIREVO Biotech), nerelimomab (CAS No. 162774-06-3) (Chiron), OM-294DP (OM PHARMA), onercept (CAS No. 199685-57-9) (Yeda), PASSTNF-alpha (Verigen), pentoxifylline or oxypentifylline (Sanofi-Aventis), Pharmaprojects No. 4091, 4241, 4295, or 4488 (Sanofi-Aventis), Pharmaprojects No. 5480 (Amgen), Pharmaprojects No. 6749 (Cengent), pirfenidone (CAS No. 53179-13-8) (MARNAC), RPR-132294 (Sanofi- Aventis), S5 (F002) (Fulcrum Pharmaceuticals), simvastatin (CAS No. 79902-63-9) (Merck & Co), STA-6292 (Synta Pharmaceuticals), tacrolimus (CAS No. 104987-11-3) (from Fujisawa LifeCycle Pharma), talactoferrin alfa (CAS No. 308240-58-6) (Agennix), thalidomide (CAS No. 50-35-1) (Celgene), TNF antagonists form ProStrakan, and Synergen, TNF inhibitors (Amgen), TNF-alpha antagonists from Dynavax Technologies and Jerina AG (Germany), TNF-alpha inhibitors from IBFB Pharma and Xencor (Xencor), torbafylline (CAS No. 105102-21-4) (Sanofi-Aventis), UR-1505 (Uriach), VT-346 (Viron Therapeutics), YSIL6 (Y's Therapeutics), YSTH2 (Y's Therapeutics), NPI-1302a-3 (Nereus Pharmaceuticals, a TNF antagonist from Jerina AG (Germany), dersalazine, or an analogue or derivative thereof).
98) Tumor Necrosis Factor Antagonists
In another embodiment, the fibrosis-inhibiting compound is a tumor necrosis factor (TNF) antagonist (e.g., anti-inflammatory compounds from Biota Inc., or an analogue or derivative thereof).
99) Toll Receptor Antagonists
In another embodiment, the fibrosis-inhibiting compound is a Toll receptor antagonist (e.g., E5564 (Eisai Pharmaceuticals), or an analogue or derivative thereof).
100) Tubulin Antagonist
In another embodiment, the fibrosis-inhibiting compound is a tubulin antagonist (e.g., synthadotin, KRX-0403 (Keryx Biopharmaceuticals), or an analogue or derivative thereof). 101) Tyrosine Kinase Inhibitors
In another embodiment, the fibrosis-inhibiting compound is a tyrosine kinase inhibitor (e.g., SU-011248 (e.g., SUTENT from Pfizer Inc. (New York, NY), BMS-354825, PN-355 (Paracelsian Pharmaceuticals), AGN-199659 (Allergan), (e.g., AAL-993 or ABP-309 (Novartis), adaphostin (NIH), AEE-788 (Novartis), AG-013736 (OSI Pharmaceuticals), AG-13736 (Pfizer), ALT-110 (Alteris Therapeutics), AMG-706 (Amgen), anticancer MAbs from Xencor, anti-EGFrvlll MAbs from Abgenix, anti-HER2 MAb from Abiogen, AZD-2171 or AZD-9935 (AstraZeneca), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), CEP-5214 (Cephalon), CEP-7055 (Cephalon), cetuximab (ImCIone Systems), CHIR-200131 and CHIR-258 (Chiron), CP-547632 (OSI Pharmaceuticals), CP-724714 (Pfizer), CT-301 (Creabilis Therapeutics), D-69491 (Baxter International), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EGFR/ErbB2 inhibitors from Array BioPharma, erlotinib (CAS No. 183319-69-9) (OSI Pharmaceuticals), EXEL-2880 (Exelixis), FK-778 (Sanofi-Aventis) , gefitinib (CAS No. 184475-35-2) (AstraZeneca), GW-2286 or GW-654652 (GlaxoSmithKline), her2/neu antigen from AlphaVax, HER-2/neu inhibitor from Generex, Herzyme (Medipad) (Sima Therapeutics), HKI-272 (Wyeth), HuMax-EGFr (Genmab), idronoxil (CAS No. 81267-65-4) (Novogen), IGF-1 inhibitors from Ontogen, IMC-11F8 (ImCIone Systems), kahalalide F (CAS No. 149204-42-2) (PharmaMar), KDR inhibitor from LG Life Sciences, KDR inhibitors from Abbott Laboratories, KDR kinase inhibitors (UCB), Kdr kinase inhibitors from Merck & Co, KRN-633 and KRN-951 (Kirin Brewery), KSB-102 (Xenova), lapatinib ditosylate (CAS No. 388082-78-8) (GlaxoSmithKline), matuzumab (Merck KGaA), MDX-214 (Medarex), ME-103 (Pharmexa), MED-A300 (Gerolymatos), MNAC-13 (Lay Line Genomics), nimotuzumab (Center of Molecular Immunology), NSC-330507 or NSC-707545 (NIH), NV-50 (Novogen), OSI-930 (OSI Pharmaceuticals), panitumumab (Abgenix), pelitinib (CAS No. 287933-82-7) (Wyeth), pertuzumab (CAS No. 380610-27- 5) (Genentech), Pharmaprojects No. 3985 (Sanofi-Aventis), prostate cancer therapeutics from Sequenom (SQPC35, SQPC36, SQPC90), removab and remoxab (Trion Pharma), RG-13022 (CAS No. 136831-48-6), RG-13291 (CAS No. 138989-50-1), or RG-14620 (CAS No. 136831-49-7) (Sanofi- Aventis), RM-6427 (Romark), RNAi breast cancer therapy from Benitec, RP 53801 (CAS No. 125882-88-4) (Sanofi-Aventis), sorafenib tosylate (Bayer), SlM 1657 (Pfizer), Tie-2 antagonists from Semaia (Hybrigenics), Tie-2 inhibitors from Ontogen, trastuzumab (CAS No. 180288-69-1) (Genentech), tyrosine kinase inhibitors from Sanofi-Aventis, U3-1287, U3-1565, U3-1784, or U3-1800 (U3 Pharma), vatalanib (Novartis), VEGFR-2 kinase inhibitor from Bristol-Myers Squibb, XL-647 (Exeiixis), ZD-6474 (AstraZeneca), ZK- CDK (Schering AG), herbimycin A, or an analogue or derivative thereof).
In certain embodiments, the tyrosine kinase inhibitor is an EGFR tyrosine kinase inhibitor such as EKB-569 (Wyeth), or an analogue or derivative thereof).
102) VEGF Inhibitors
In another embodiment, the fibrosis-inhibiting compound is a VEGF Inhibitor (e.g., AZD2171 (AstraZeneca), or an analogue or derivative thereof).
103) Vitamin D Receptor Agonists
In another embodiment, the fibrosis-inhibiting compound is a vitamin D receptor agonist (e.g., BXL-628, BXL-922 (BioXell), or an analogue or derivative thereof).
104) Histamine Receptor Antagonists
In another embodiment, the fibrosis-inhibiting compound is an histamine receptor antagonist. Certain embodiments, the histamine receptor antagonists, such as H1 , H2, and H3 histamine receptor antagonists, block the production of pro-inflammatory cytokines such as TNFa and IL-1 (e.g., IL-1β). In certain embodiments, the histamine receptor antagonist inhibit NFkB activation. Representative examples of H1 histamine receptor antagonists include phenothiazines, such as promethazine, and alkylamines, such as chlorpheniramine (CAS No. 7054- 11-7), brompheniramine (CAS No. 980-71-2), fexofenadine hydrochloride, promethazine hydrochloride, loratadine, ketotifen fumarate salt, and acrivastine. Other examples of histamine receptor antagonists include broad spectrum histamine receptor antagonists such as methylxanthines (e.g., theophylline, theobromine, and caffeine). Representative examples of H2 receptor antagonists include those with a histamine-like structure including cimetidine (available under the tradename TAGAMET from SmithKline Beecham Phamaceutical Co., Wilmington, DE), ranitidine (available under the tradename ZANTAC from Warner Lambert Company, Morris Plains, NJ), famotidine (available under the tradename PEPCID from Merck & Co., Whitehouse Station, NJ), nizatidine (available under the tradename AXID from Reliant Pharmaceuticals, Inc., Liberty Corner, NJ), nizatidine, and roxatidine acetate (CAS No. 78628-28-1). Additional examples include H3 receptor antagonists (e.g., thioperamide and thioperamide maleate salt) and anti-histamines such as tricyclic dibenozoxepins, ethanolamines, ethylenediamines, piperizines, piperidines, and pthalazinones.
105) Alpha Adrenergic Receptor Antagonists
In another embodiment, the fibrosis-inhibiting compound is an alpha adrenergic receptor antagonist. Alpha adrenergic receptor antagonists may inhibit the production of pro-inflammatory cytokines such as TNFa. The alpha adrenergic receptor antagonist may be an alpha-1 and/or an alpha-2 adrenergic receptor antagonist. Representative examples of aipha-1 /alpha-2 antagonists include phenoxybenzamine. In certain embodiments, the alpha adrenergic receptor antagonist is a haloalkylamine compound or a catecholamine uptake inhibitor. Representative examples of alpha-1 adrenergic receptor antagonists include phenoxybenzamine hydrochloride and prazosin, a piperizinyl quinazoline. Representative examples of alpha-2 adrenergic receptor antagonists include imadazole based compounds such as idazoxan (CAS No. 79944-56-2), idazoxan hydrochloride, and loxapine succinate salt (CAS No. 27833-64-3). Additional examples of alpha adrenergic receptor antagonists include prazosin hydrochloride.
106) Anti-Psychotic Compounds
In another embodiment, the fibrosis-inhibiting compound is an anti-psychotic compound, such as a phenothiazine compound or an analogue or derivative thereof. In some embodiments, the fibrosis-inhibiting compound is a phenothiazine derivative capable of suppressing the production of pro-inflammatory cytokines such as TNFa and/or IL-1. Representative examples of phenothiazine compounds include chlorpromazine, fluphenazine, trifluorphenazine, mesoridazine, thioridazine, and perphenazine. Other examples of anti-psychotic compounds include thioxanthines such as chlorprothixene and thiothixene, clozapine, loxapine succinate, and olanzapine.
107) CaM Kinase Il Inhibitor
In another embodiment, the fibrosis-inhibiting compound is CaM kinase Il inhibitor, such as a lavendustin C, or an analogue or derivative thereof.
108) CaM Kinase Il Inhibitor
In another embodiment, the fibrosis-inhibiting compound is CaM kinase Il inhibitor, such as a lavendustin C, or an analogue or derivative thereof. 109) G Protein Agonist
In another embodiment, the fibrosis-inhibiting compound is G protein agonist, such as aluminum fluoride, or an analogue or derivative thereof.
110) Antibiotics and Anti-Microbials
In another embodiment, the fibrosis-inhibiting compound is an antibiotic, such as apigenin (Cas No. 520-36-5), ampicillin sodium salt (CAS No. 69-52-3), puromycin, or an analogue or derivative thereof.
In another embodiment, the fibrosis-inhibiting compound is an anti-microbial agent, such as brefeldin A (CAS No. 20350-15-6), terbinafine, benzoyl peroxide, pentamidine, ornidazole, imidazole, ketocanazole, sulconazole nitrate salt, or an analogue or derivative thereof.
111) PNA Topoisomerase Inhibitors
In another embodiment, the fibrosis-inhibiting compound is DNA topoisomerase I inhibitor, such as β-lapachone (CAS No. 4707-32-8), or an analogue or derivative thereof.
In another embodiment, the fibrosis-inhibiting compound is DNA topoisomerase Il inhibitor, such as (-)-arctigenin (CAS No. 7770-78-7), aurintricarboxylic acid, or an analogue or derivative thereof.
112) Thromboxane A2 Receptor Inhibitor
In another embodiment, the fibrosis-inhibiting compound is thromboxane A2 receptor inhibitor, such as BM-531 (CAS No. 284464-46- 6), ozagrel hydrochloride (CAS No. 78712-43-3), or an analogue or derivative thereof. 113) D2-Dopamine Receptor Antagonist
In another embodiment, the fibrosis-inhibiting compound is a D2 dopamine receptor antagonist, such as clozapine (CAS No. 5786-21-0), mesoridazine benzenesulfonate, or an analogue or derivative thereof.
114) Peptidyl-Prolyl Cis/Trans lsomerase Inhibitor
In another embodiment, the fibrosis-inhibiting compound is a Peptidyl-Prolyl Cis/Trans lsomerase Inhibitor, such as juglone (CAS No. 481-39-0), or an analogue or derivative thereof.
115) Dopamine Antagonists
In another embodiment, the fibrosis-inhibiting compound is a dopamine antagonist, such as thiothixene, thioridazine hydrochloride, or an analogue or derivative thereof.
116) Anesthetics
In another embodiment, the fibrosis-inhibiting compound is an anesthetic compound, such as lidocaine (CAS No. 137-58-6), or an analogue or derivative thereof.
117) Clotting Factors
In another embodiment, the fibrosis-inhibiting compound is a clotting factor, such as menadione (CAS No. 58-27-5), or an analogue or derivative thereof.
118) Lysyl Hydrolase Inhibitor
In another embodiment, the fibrosis-inhibiting compound is a lysyl hydrolase inhibitor, such as minoxidil (CAS No. 38304-91-5), or an analogue or derivative thereof. 119) Muscarinic Receptor Inhibitor
In another embodiment, the fibrosis-inhibiting compound is a muscarinic receptor inhibitor, such as perphenazine (CAS No. 58-39-9), or an analogue or derivative thereof.
120) Superoxide Anion Generator
In another embodiment, the fibrosis-inhibiting compound is a superoxide anion generator, such as plumbagin (CAS No. 481-42-5), or an analogue or derivative thereof.
121) Steroids
In another embodiment, the fibrosis-inhibiting compound is a steroid, such as prednisolone, prednisolone 21-acetate (CAS No. 52-21-1), loteprednol etabonate, (CAS No. 82034-46-6), clobetasol propionate, or an analogue or derivative thereof.
122) Anti-Proliferative Agents
In another embodiment, the fibrosis-inhibiting compound is an antiproliferative agent, such as silibinin (CAS No. 22888-70-6), silymarin (CAS No. 65666-07-1), 1 ,2-hexanediol, dioctyl phthalate (CAS No. 117-81- 7), zirconium (IV) oxide, glycyrrhizic acid, spermidine trihydrochloride or tetrahydrochloride, CGP 74514A, spermine tetrahydrochloride, NG-methyl- L-arginine acetate salt, galardin, halofuginone hydrobromide (HBr), fascaplysin, or an analogue or derivative thereof.
123) Diuretics
In another embodiment, the fibrosis-inhibiting compound is a diuretic, such as spironolactone (CAS No. 52-01-7), or an analogue or derivative thereof. 124) Anti-Coagulants
In another embodiment, the fibrosis-inhibiting compound is an anti-coagulant, such as fucoidan from Fucus vesiculosus (CAS No. 9072- 19-9), or an analogue or derivative thereof.
125) Cyclic GMP Agonists
In another embodiment, the fibrosis-inhibiting compound is a cyclic GMP agonist, such as sinitrodil (CAS No. 143248-63-9), or an analogue or derivative thereof.
126) Adenylate Cyclase Agonist
In another embodiment, the fibrosis-inhibiting compound is an adenylate cyclase agonist, such as histamine (CAS No. 51-45-6), or an analogue or derivative thereof.
127) Antioxidants
In another embodiment, the fibrosis-inhibiting compound is an antioxidant, such as morpholine, phytic acid dipotassium salt, (-)- epigallocatechin or (-)-epigallocatechin gallate from green tea (CAS Nos. 970-74-1 and 1257-08-5, respectively), (-)-epigallocatechin gallate (CAS No. 989-51-5), nobiletin (CAS No. 478-01-3), probucol (CAS No. 23288-49-5), phosphorous acid, hesperetin, L-ascorbyl-2-phosphate, magnesium salt (CAS No. 84309-23-9), catechin, (±)-naringenin (CAS No. 67604-48-2), (-)- epicatechin, (-)-epicatechin gallate, 3-hydroxyflavone, (-)-arctigenin, or an analogue or derivative thereof.
128) Nitric Oxide Synthase Inhibitors
In another embodiment, the fibrosis-inhibiting compound is a nitric oxide synthase inhibitor, such as ammonium pyrrolidinedithiocarbamate (CAS No. 5108-96-3), or an analogue or derivative thereof. In another embodiment, the fibrosis-inhibiting compound is a reversible nitric oxide synthase inhibitor, such as NB-methyl-L-arginine acetate salt (L-NMMA) (CAS No. 53308-83-1), or an analogue or derivative thereof.
129) Anti-Neoplastic Agents
In another embodiment, the fibrosis-inhibiting compound is an antineoplastic agent, such as tirapazamine (CAS No. 27314-97-2), fludarabine (CAS No. 21679-14-1), cladribine, imatinib mesilate, or an analogue or derivative thereof.
130) DNA Synthesis Inhibitors
In another embodiment, the fibrosis-inhibiting compound is a DNA synthesis inhibitor, such as S-(2-hydroxy-5-nitrobenyl)-6-thioguanosine or uracilfludarabine phosphate (CAS No. 75607-67-9), 6,11-dihydroxy-5,12- naphthacenedione, or an analogue or derivative thereof.
131) DNA Alkylating Agents
In another embodiment, the fibrosis-inhibiting compound is a DNA alkylating agent, such as dacarbazine (CAS No. 4342-03-4), temozolomide, procarbazine HCI, or an analogue or derivative thereof.
132) DNA Methylation Inhibitors
In another embodiment, the fibrosis-inhibiting compound is a DNA methylation inhibitor, such as decitabine (CAS No. 2353-33-5), or an analogue or derivative thereof.
133) NSAID Agents
In another embodiment, the fibrosis-inhibiting compound is a NSAID agent, such as nabumetone, benzydamine hydrochloride, or an analogue or derivative thereof. 134) Peptidylglycine Alpha-Hydroxylatinq Monooxygenase Inhibitors
In another embodiment, the fibrosis-inhibiting compound is a peptidylglycine alpha-hydroxylating monooxygenase inhibitor, such as trans- styrylacetic acid, or an analogue or derivative thereof.
135) MEK1/MEK2 Inhibitors
In another embodiment, the fibrosis-inhibiting compound is a MEK1/MEK 2 inhibitor, such as U0126 (CAS No. 109511-58-2), or an analogue or derivative thereof.
136) NO Synthase Inhibitors
In another embodiment, the fibrosis-inhibiting compound is an NO synthase inhibitor, such as L-NAME (CAS No. 53308-83-1), NG-Methyl- L-arginine acetate salt, or an analogue or derivative thereof.
137) Retinoic Acid Receptor Antagonists
In another embodiment, the fibrosis-inhibiting compound is retinoic acid receptor antagonist, such as isotretinoin (CAS No. 4759-48-2), or an analogue or derivative thereof.
138) ACE Inhibitors
In another embodiment, the fibrosis-inhibiting compound is an ACE inhibitor, such as quinapril hydrochloride (CAS No. 85441-61-8), enalapril, or an analogue or derivative thereof.
139) Glvcosylation Inhibitors
In another embodiment, the fibrosis-inhibiting compound is a glycosylation inhibitor, such as aminoguanidine hydrochloride, castanospermine, or an analogue or derivative thereof. 140) Intracellular Calcium Influx Inhibitors
In another embodiment, the fibrosis-inhibiting compound is an intracellular calcium influx inhibitor, such as TAS-301 (CAS No. 193620-69- 8), or an analogue or derivative thereof.
141) Anti-Emetic Agents
In another embodiment, the fibrosis-inhibiting compound is an anti-emetic agent, such as amifostine (CAS No. 20537-88-6), or an analogue or derivative thereof.
142) Acetylcholinesterase Inhibitors
In another embodiment, the fibrosis-inhibiting compound is an acetylcholinesterase inhibitor, such as (-)-huperzine A (CAS No. 102518-79- 6), or an analogue or derivative thereof.
143) ALK-5 Receptor Antagonists . . _ . _ . . .
In another embodiment, the fibrosis-inhibiting compound is an ALK-5 receptor antagonist, such as SB 431542 (CAS No. 301836-41-9), or an analogue or derivative thereof.
144) RAR/RXR Antagonists
In another embodiment, the fibrosis-inhibiting compound is a RAR/RXT antagonist, such as 9-cis-retinoic acid, or an analogue or derivative thereof.
145) EIF-2a Inhibitors
In another embodiment, the fibrosis-inhibiting compound is a elF-2a inhibitor, such as salubrinal, or an analogue or derivative thereof. 146) S-Adenosyl-L-Homocysteine Hydrolase Inhibitors
In another embodiment, the fibrosis-inhibiting compound is a S-adenosyl-L-homocysteine hydrolase inhibitor, such as 3-deazaadenosine, or an analogue or derivative thereof.
147) Estrogen Agonists
In another embodiment, the fibrosis-inhibiting compound is an estrogen agonist, such as coumestrol, bisphenol A, 1-linoleoyl-rac-glycerol (CAS No. 2277-28-3), daidzein (4,7-dihydroxy-iso-flavone), dihexyl phthalate, kaempferol, formononetin, , or an analogue or derivative thereof.
148) Serotonin Receptor Inhibitors
In another embodiment, the fibrosis-inhibiting compound is a serotonin receptor inhibitor, such as amitriptyline hydrochloride, or an analogue or derivative thereof.
149) Anti-Thrombotic Agents
In another embodiment, the fibrosis-inhibiting compound is an anti-thrombotic agent, such as geniposidic acid, geniposide, or an analogue or derivative thereof.
150) Tryptase Inhibitors
In another embodiment, the fibrosis-inhibiting compound is a tryptase inhibitors, such as 2-azetidinone, or an analogue or derivative thereof.
151) Pesticides
In another embodiment, the fibrosis-inhibiting compound is a pesticide, such as allyl disulfide, or an analogue or derivative thereof. 152) Bone Mineralization Promotor
In another embodiment, the fibrosis-inhibiting compound is a bone mineralization promotor, such as glycerol 2-phosphate disodium salt hydrate, or an analogue or derivative thereof.
153) Bisphosphonate Compounds
In another embodiment, the fibrosis-inhibiting compound is a bisphosphonate compound, such as risedronate, or an analogue or derivative thereof.
154) Anti-Inflammatory Compounds
In another embodiment, the fibrosis-inhibiting compound is an anti-inflammatory compound, such as aucubin, cepharanthine, or an analogue or derivative thereof.
155) DNA Methylation Promotors
In another embodiment, the fibrosis-inhibiting compound is a DNA methylation promotor, such as 5-azacytidine, or an analogue or derivative thereof.
156) Anti-Spasmodic Agents
In another embodiment, the fibrosis-inhibiting compound is an anti-spasmodic agent, such as 2-hydroxy-4,6-dimethoxyacetophenone, or an analogue or derivative thereof.
157) Protein Synthesis Inhibitors
In another embodiment, the fibrosis-inhibiting compound is a protein synthesis inhibitor, such as oxytetracycline hydrochloride, or an analogue or derivative thereof. 158) α-Glucosidase Inhibitors
In another embodiment, the fibrosis-inhibiting compound is a α-glucosidase inhibitor, such as myricetin (CAS No. 529-44-2), or an analogue or derivative thereof.
159) Calcium Channel Blockers
In another embodiment, the fibrosis-inhibiting compound is a calcium channel blocker, such as verapamil, nitrendipine, or an analogue or derivative thereof.
In another embodiment, the fibrosis-inhibiting compound is a L-type calcium channel blocker, such as nifedipine (CAS No. 21829-25-4), (+)-cis-diltiazem hydrochloride, or an analogue or derivative thereof.
In another embodiment, the fibrosis-inhibiting compound is a T-type calcium channel blocker, such as penfluridol (CAS No. 26864-56-2), or an analogue or derivative thereof.
160) Pyruvate Dehydrogenase Activators
In another embodiment, the fibrosis-inhibiting compound is a pyruvate dehydrogenase activator, such as dichloroacetic acid, or an analogue or derivative thereof.
161) Prostaglandin Inhibitors
In another embodiment, the fibrosis-inhibiting compound is a prostaglandin inhibitor, such as betulinic acid, or an analogue or derivative thereof.
162) Sodium Channel Inhibitors
In another embodiment, the fibrosis-inhibiting compound is a sodium channel inhibitor, such as amiloride hydrochloride hydrate, or an analogue or derivative thereof. 163) Serine Protease Inhibitors
In another embodiment, the fibrosis-inhibiting compound is a serine protease inhibitor, such as gabexate mesylate, or an analogue or derivative thereof.
164) Intracellular Calcium Flux Inhibitors
In another embodiment, the fibrosis-inhibiting compound is an intracellular calcium flux inhibitor, such as thapsigargin, or an analogue or derivative thereof.
165) JAK2 Inhibitors
In another embodiment, the fibrosis-inhibiting compound is a JAK2 inhibitor (e.g., AG-490 (CAS No. 134036-52-5), or an analogue or derivative thereof).
166) Androgen Inhibitors
In another embodiment, the fibrosis-inhibiting compound is an androgen inhibitor (e.g., tibolone (CAS No. 5630-53-5), or an analogue or derivative thereof).
167) Aromatase Inhibitors
In another embodiment, the fibrosis-inhibiting compound is an aromatase inhibitor (e.g., letrozole, or an analogue or derivative thereof).
168) Anti-Viral Agents
In another embodiment, the fibrosis-inhibiting compound is an anti-viral agent, such as imiquimod, or an analogue or derivative thereof. 169) 5-HT Inhibitors
In another embodiment, the fibrosis-inhibiting compound is a 5-HT inhibitor, such as ketanserin tartrate, amoxapine, or an analogue or derivative thereof.
170) FXR Antagonists
In another embodiment, the fibrosis-inhibiting compound is a FXR antagonist, such as guggulsterone (CAS No. 95975-55-6), or an analogue or derivative thereof.
171) Actin Polymerization and Stabilization Promotors
In another embodiment, the fibrosis-inhibiting compound is an actin polymerization and stabilization promotor, such as jasplakinolide, or an analogue or derivative thereof.
172) AXOR 12 Agonists
In another embodiment, the fibrosis-inhibiting compound is an AXOR12 agonist, such as metastin (KiSS-1 (112-121), or an analogue or derivative thereof.
173) Angiotensin Il Receptor Antagonists
In another embodiment, the fibrosis-inhibiting compound is an angiotensin Il receptor agonist, such as losartan potassium, or an analogue or derivative thereof.
174) Platelet Aggregation Inhibitors
In another embodiment, the fibrosis-inhibiting compound is a platelet aggregation inhibitor, such as clopidogrel, or an analogue or derivative thereof. 175) CB1/CB2 Receptor Agonists
In another embodiment, the fibrosis-inhibiting compound is a CB1/CB2 receptor agonist, such as HU-210 (CAS No. 112830-95-2), or an analogue or derivative thereof.
176) Norepinephrine Reuptake Inhibitors
In another embodiment, the fibrosis-inhibiting compound is a norepinephrine reuptake inhibitor, such as nortriptyline hydrochloride, or an analogue or derivative thereof.
177) Selective Serotonin Reuptake Inhibitors
In another embodiment, the fibrosis-inhibiting compound is a selective serotonin reuptake inhibitor, such as paroxetine maleate, or an analogue or derivative thereof.
178) Reducing Agents
In another embodiment, the fibrosis-inhibiting compound is a reducing agent such as VVW-85 (Inotek), or an analogue or derivative thereof.
179) Immuno-modulators
In another embodiment, the fibrosis-inhibiting compound is an immunomodulators such as Bay 11-7085, (-)-arctigenin, idazoxan hydrochloride, or an analogue or derivative thereof .Anti-Infective Agents
The therapeutic agents useful in the present invention also include anti-infective agents. Such agents may reduce the likelihood of infection upon implantation of the composition or a medical implant and may be used in combination of an anti-fibrosis agent and/or a polymer. Infection is a common complication of the implantation of foreign bodies such as, for example, medical devices and implants. Foreign materials provide an ideal site for micro- organisms to attach and colonize. It is also hypothesized that there is an impairment of host defenses to infection in the microenvironment surrounding a foreign material. These factors make medical implants particularly susceptible to infection and make eradication of such an infection difficult, if not impossible, in most cases. In many cases, an infected implant or device must be surgically removed from the body in order to eradicate the infection.
The present invention provides agents (e.g., chemotherapeutic agents) that can be released from a composition, and which have potent antimicrobial activity at extremely low doses. A wide variety of anti-infective agents can be utilized in combination with the present compositions. Suitable anti-infective agents may be readily determined based upon the assays provided in Example 30). Discussed in more detail below are several representative examples of agents that can be, used as anti-infective agents, such as: (A) anthracyclines (e.g., doxorubicin and mitoxantrone), (B) fluoropyrimidines (e.g., 5-FU), (C) folic acid antagonists (e.g., methotrexate), (D) podophylotoxins (e.g., etoposide), (E) camptothecins, (F) hydroxyureas, and (G) platinum complexes (e.g., cisplatin).
1) Anthracvclines
In one aspect, the therapeutic anti-infective agent is an anthracycline. Anthracyclines have the following general structure, where the R groups may be a variety of organic groups:
Figure imgf000092_0001
According to U.S. Patent 5,594,158, suitable R groups are as follows: Ri is CH3 or CH2OH; R2 is daunosamine or H; R3 and R4 are independently one of OH, NO2, NH2, F, Cl, Br, I, CN1 H or groups derived from these; R5 is hydrogen, ydroxyl, or methoxy; and R6-8 are aii hydrogen. Alternatively, R5 and Re are hydrogen and R7 and R8 are alkyl or halogen, or vice versa.
According to U.S. Patent 5,843,903, Ri may be a conjugated peptide. According to U.S. Patent 4,296,105, R5 may be an ether linked aikyl group. According to U.S. Patent 4,215,062, R5 may be OH or an ether linked alkyl group. R1 may also be linked to the anthracycline ring by a group other than C(O), such as an alkyl or branched alkyl group having the C(O) linking moiety at its end, such as -CH2CH(CH2-X)C(O)-R1, wherein X is H or an alkyl group (see, e.g., U.S. Patent 4,215,062). R2 may alternately be a group linked by the functional group =N-NHC(O)-Y, where Y is a group such as a phenyl or substituted phenyl ring. Alternately R3 may have the following structure:
Figure imgf000093_0001
in which R9 is OH either in or out of the plane of the ring, or is a second sugar moiety such as R3. R10 may be H or form a secondary amine with a group such as an aromatic group, saturated or partially saturated 5 or 6 membered heterocyclic having at least one ring nitrogen (see U.S. Patent 5,843,903). Alternately, R1O may be derived from an amino acid, having the structure -C(O)CH(NHR11)(R12), in which R11 is H, or forms a C3-4 membered alkylene with R12. R12 may be H, alkyl, aminoalkyl, amino, hydroxyl, mercapto, phenyl, benzyl or methylthio (see U.S. Patent 4,296,105). Exemplary anthracyclines are doxorubicin, daunorubicin, idarubicin, epirubicjn, pirarubicin, zorubicin, and carubicin. Suitable compounds have the structures:
Figure imgf000094_0001
Ri K2 Rs doxorubicin: OCH3 C(O)CH2OH OH out of ring plane epirubicin:
(4' epimer of OCH3 C(O)CH2OH OH in ring plane doxorubicin) daunorubicin: OCH3 C(O)CH3 OH out of ring plane idarubicin: H C(O)CH3 OH out of ring plane pirarubicin: OCH3 C(O)CH2OH
Figure imgf000094_0002
zorubicin: OCH3 C(CH3X=N)NHC(O)C6H5 OH carubicin: OH C(O)CH3 OH out of ring plane
Other suitable anthracyclines are anthramycin, mitoxantrone, menogaril, nogalamycin, aclacinomycin A, olivomycin A, chromomycin A3, and plicamycin having the structures: R1 R2 R,
Menogaril H OCH3 H
Nogalarrycin σsugar H COOCH3
Figure imgf000095_0001
Mitoxanlrone
Figure imgf000095_0003
Olivomycin A COCH(CH3J2 CH3 COCH3 H
ChroiTDmycin Aa COCH3 CH3 COCH3 CH3
Plicarrycin H H H CH3
Figure imgf000095_0002
Other representative antbracyclinesjnclude, FCE 23762 doxorubicin derivative (Quaglia ef a/., J. Uq. Chromatogr. 17(18):3911-3923, 1994), annamycin (Zou et al., J. Pharm. Sci. 82(11): 1151 -1154, 1993), rυboxyl (Rapoport et al., J. Controlled Release 5S(2):153-162, 1999), anthracycline disaccharide doxorubicin analogue (Pratesi et al., CHn. Cancer Res. 4(11):2833-2839, 1998), N-(trifluoroacetyl)doxorubicin and 4'-O~acetyl- N-(trifluoroacetyl)doxorubicin (Berube & Lepage, Synth. Commun. 28(6): 1109-1116, 1998), 2-pyrrolinodoxorubicin (Nagy et a/., Proc. Nat'l Acad. Sci. U.S.A. 95(4): 1794-1799, 1998), disaccharide doxorubicin analogues (Arcamone et al., J. Nat'l Cancer Inst 89(16):1217-1223, 1997), 4-demethoxy-7-O-(2,6-dideoxy-4-O-(2,3,6-trideoxy-3-amino-α-L-lyxo- hexopyranosy[)-α-L-lyxo-hexopyranosyi)adriamicinone doxorubicin disaccharide analogue (Monteagudo et al., Carbohydr. Res. 300(1):11-16, 1997), 2-pyrrolinodoxorubicin (Nagy et al., Proc. Nat'l Acad. Sci. U. S. A. 94(2):652-656, 1997), morpholinyl doxorubicin analogues (Duran etal., Cancer Chemother. Pharmacol. 38(3):210-216, 1996), enamϊnomalonyl-β- alanine doxorubicin derivatives (Seitz et al., Tetrahedron Lett. 36(9):1413- 16, 1995), cephalosporin doxorubicin derivatives (Vrudhula et al., J. Med. Cherri. 38(8):1380-5, 1995), hydroxyrubicin (Solary et al., Int. J. Cancer 58(1):85-94, 1994), methoxymorpholino doxorubicin derivative (Kuhl et al., Cancer Chemother. Pharmacol. 33(1):10-16, 1993), (6- maleimidocaproyl)hydrazone doxorubicin derivative (Willner et al., Bioconjugate Chem. 4(6):521-7, 1993), N-(5,5-diacetoxypent-1-yl) doxorubicin (Cherif & Farquhar, J. Med. Chem. 35(17):3208-14, 1992), FCE 23762 methoxymorpholinyl doxorubicin derivative (Ripamonti etal., Br. J. Cancer 65(5):703-7 , 1992), N-hydroxysuccinimide ester doxorubicin derivatives (Demant et al., Biochim. Biophys. Acta 1118(1): 83-90, 1991), polydeoxynucleotide doxorubicin derivatives (Ruggiero et al., Biochim. Biophys. Acta 1129(3):294-302, 1991), morpholinyl doxorubicin derivatives (EPA 434960), mitoxantrone doxorubicin analogue (Krapcho et al., J. Med. Chem. 34(8):2373~80. 1991), AD198 doxorubicin analogue (Traganos et al., Cancer Res. 57(14):3682-9, 1991), 4-demethoxy-3'-N- trifluoroacetyldoxorubicin (Horton et al., Drug Des. Delivery 6(2): 123-9, 1990), 4'-epidoxorubicin (Drzewoski et al., Pol. J. Pharmacol. Pharm. 40(2):159-65, 1988; Weenen et al., Eur. J. Cancer Clin. Oncol. 20(7):919- 26, 1984), alkylating cyanomorpholino doxorubicin derivative (Scudder et al., J. Nat'l Cancer Inst. S0(16):1294-8, 1988), deoxydihydroiodooxorubicin (EPA 275966), adriblastin (Kalishevskaya et al., Vestn. Mosk. Univ., 16(Biol. 1):21-7, 1988), 4'-deoxydoxorubicin (Schoelzel et al., Leuk. Res. 70(12):1455-9, 1986), 4-demethyoxy-4'-o-methyldoxorubicin (Giuliani et al., Proc. Int. Congr. Chemother. 16:285-70-285-77, 1983), 3'-deamino-3'- hydroxydoxorubicin (Horton et al., J. Antibiot. 37(8):853-8, 1984), 4- demethyoxy doxorubicin analogues (Barbieri et al., Drugs Exp. Clin. Res. 10(2):85-90, 1984), N-L-leucyl doxorubicin derivatives (Trouet et al., Anthracyclines (Proc. Int. Symp. Tumor Pharmacother.), 179-81, 1983), 3'- deamino-3'-(4-methoxy-1-piperidinyl) doxorubicin derivatives (U.S. 4,314,054), 3'-deamino-3'-(4-mortholinyl) doxorubicin derivatives (U.S. 4,301 ,277), 4'-deoxydoxorubicin and 4'-o-methyldoxorubicin (Giuliani et ai, Int. J. Cancer 27(1):5-13, 1981), aglycone doxorubicin derivatives (Chan & Watson, J. Pharm. Sci. 67(12):1748-52, 1978), SM 5887 {Pharma Japan 1468:20, 1995), MX-2 (Pharma Japan 1420:19, 1994), 4'-deoxy-13(S)- dihydro-4'-iododoxorubicin (EP 275966), morpholinyl doxorubicin derivatives (EPA 434960), 3'-deamino-3'-(4-methoxy-1~piperidinyl) doxorubicin derivatives (U.S. 4,314,054), doxorubicin-14-valerate, morpholinodoxorubicin (U.S. 5,004,606), 3'-deamino-3'-(3"-cyano-4"- morpholinyl doxorubicin; 3'~deamino-3'-(3"-cyano-4"-morpholinyl)-13- dihydoxorubicin; (3'-deamino-3'-(3"-cyano-4"-morpholiny() daunorubicin; 3'- deamino-3'-(3"-cyano-4"-morpholinyl)-3-dihydrodaunorubicin; and 3'- deamino-3'-(4"-morpholinyl-5-iminodoxorubicin and derivatives (U.S. 4,585,859), 3'-deamino-3'-(4-methoxy-1~piperidinyl) doxorubicin derivatives (U.S. 4,314,054) and 3-deamino-3-(4-morpholinyl) doxorubicin derivatives (U.S. 4,301 ,277).
2) Fluoropyrimidine analogues
In another aspect, the ant-infective therapeutic agent is a fluoropyrimidine analog, such as 5-fluorouracil, or an analogue or derivative thereof, including carmofur, doxifluridine, emitefur, tegafur, and floxuridine. Exemplary compounds have the structures:
Figure imgf000097_0001
Figure imgf000098_0001
5-fluoroυracil H H carmofur C(O)NH(CH2)5CH3 H doxifluridine Ai H floxuridine A2 H emitefur CH2OCH2CH3 B tegafur C H
Figure imgf000098_0002
Other suitable fluoropyrimidine analogues include 5-FudR (5- fluoro-deoxyuridine), or an analogue or derivative thereof, including 5- iododeoxyuridine (5-ludR), 5-bromodeoxyuridine (5-BudR), fluorouridine triphosphate (5-FUTP), and fluorodeoxyuridine monophosphate (5-dFUMP). Exemplary compounds have the structures:
Figure imgf000098_0003
5-fluoro-2'-deoxyuridine: R = F 5-bromo-2'-deoxyuridine: R = Br 5-iodo~2'-deoxyuridine: R = I Other representative examples of fluoropyrimidine analogues include N3-alkylated analogues of 5-fluorouracil (Kozai et a/., J. Chem. Soc, Perkin Trans. 7(19):3145-3146, 1998), 5-fluorouracil derivatives with 1 ,4- oxaheteroepane moieties (Gomez et a/., Tetrahedron 54(43):13295-13312, 1998), 5-fluorouraciJ and nucleoside analogues (Li, Anticancer Res. 77(1A):21-27, 1997), cis- and trans-5-fluoro-5,6-dihydro-6-alkoxyuracil (Van der Wilt et a/., Br. J. Cancer 68(4) :702-7, 1993), cyclopentane 5-fluorouracil analogues (Hronowski & Szarek, Can. J. Chem. 70(4): 1162-9, 1992), A-OT- fluorouracil (Zhang et al., Zongguo Yiyao Gongye Zazhi 20(11):513-15, 1989), N4-trimethoxybenzoyl-5'-deoxy-5-fluorocytidine and 5'-deoxy-5- fluorouridine (Miwa et al., Chem. Pharm. Bull. 38(4):998-1003, 1990), 1- hexylcarbamoyl-5-fluorouracil (Hoshi et al., J. Pharmacobio-Dun. 3(9):478- 81 , 1980; Maehara et al., Chemotherapy (Basel) 34(6):484-9, 1988), B-3839 (Prajda et al., In Vivo 2(2):151-4, 1988), uracil-1-(2-tetrahydrofuryl)-5- fluorouracil (Anai et al., Oncology 45(3): 144-7, 1988), 1 -(2'-deoxy-2'-fluoro- β-D-arabinofuranosyi)-5-fiuorouracii (Suzuko et a/., MoI. Pharmacol. 37(3):301-6, 1987), doxifluridine (Matuura et al., Oyo Yakuri 29(5):803-31, 1985), 5'-deoxy-5-fluorouridine (Bollag & Hartmann, Eur. J. Cancer 76(4):427-32, 1980), i-acetyl-S-O-toluyl-δ-fluorouracil (Okada, Hiroshima J. Med. Sci. 2S(1):49-66, 1979), 5-fluorouracil-m-formylbenzene-sulfonate (JP 55059173), N'-(2-furanidyl)~5-fluorouracil (JP 53149985) and 1-(2- tetrahydrofuryl)-5-fluorouracil (JP 52089680).
These compounds are believed to function as therapeutic agents by serving as antimetabolites of pyrimidine.
3) Folic acid antagonists
In another aspect, the anti-infective therapeutic agent is a folic acid antagonist, such as methotrexate or derivatives or analogues thereof, including edatrexate, trimetrexate, raltitrexed, piritrexim, denopterin, tomudex, and pteropterin. Methotrexate analogues have the following general structure:
Figure imgf000100_0001
The identity of the R group may be selected from organic groups, particularly those groups set forth in U.S. Patent Nos. 5,166,149 and 5,382,582. For example, R1 may be N, R2 may be N or C(CH3), R3 and R3' may H or alkyl, e.g., CH3, R4 may be a single bond or NR, where R is H or alkyl group. Rδ(6,8 nriay be H, OCH3, or alternately they can be halogens or hydro groups. R7 is a side chain of the general structure:
Figure imgf000100_0002
wherein n = 1 for methotrexate, n = 3 for pteropterin. The carboxyl groups in the side chain may be esterified or form a salt such as a Zn2+ salt. Rg and Rio can be NH2 or may be alkyl substituted.
Exemplary folic acid antagonist compounds have the structures:
Figure imgf000100_0003
Figure imgf000101_0001
methotrexate NH2 N N H N(CH3) H H A (n=1) H edatrexate NH2 N N H CH(CH2CH3) H H A (n=1) H trimetrexate NH2 CH C(CH3) H NH H OCH3 OCH3 OCH3 pteropterin OH N N H NH H H A (n=3) H denopterin OH N N CH3 N(CH3) H H A (n-1) H peritrexim NH2 N C(CH3) H single bond OCH3 H H OCH3
Figure imgf000101_0002
Figure imgf000101_0003
Other representative examples include 6-S aminoacyloxymethyl mercaptopurine derivatives (Harada et al., Chem. Pharm. Bull. 43(10):793-6, 1995), 6-mercaptopurine (6-MP) (Kashida et al., Biol. Pharm. Bull. 78(11): 1492-7, 1995), 7,8-po)ymethyleneimidazo-1 ,3,2- diazaphosphorines (Nilov et a/., Mendeleev Commun. 2:67, 1995), azathioprine (Chifotides et al., J. Inorg. Biochem. 56(4):249-64, 1994), methyl-D-glucopyranoside mercaptopurine derivatives (Da Silva et al., Eur. J. Med. Chem. 29(2): 149-52, 1994) and s-alkynyl mercaptopurine derivatives (Ratsino et al., Khim.-Farm. Zh. 75(8):65-7, 1981); indoline ring and a modified ornithine or glutamic acid-bearing methotrexate derivatives (Matsuoka et al., Chem. Pharm. Bull. 45(7): 1146-1150, 1997), alkyl- substituted benzene ring C bearing methotrexate derivatives (Matsuoka et al, Chem. Pharm. Bull. 44(12):2287-2293, 1996), benzoxazine or benzothiazine moiety-bearing methotrexate derivatives (Matsuoka et al., J. Med. Chem. 40(1): 105-111, 1997), 10-deazaaminopterin analogues (DeGraw e/ a/., J. Med. Chem. 40(3): 370-376, 1997), 5-deazaaminopterin and 5,10-dideazaaminopterin methotrexate analogues (Piper ef al., J. Med. Chem. 40(3):377-384, 1997), indoline moiety-bearing methotrexate derivatives (Matsuoka et al., Chem. Pharm. Bull. 44(J)-A 332-1337, 1996), lipophilic amide methotrexate derivatives (Pignatello et al., World Meet. Pharm., Biopharm. Pharm. Techno!., 563-4, 1995), L-threo-(2S,4S)-4- fluoroglutamic acid and DL-3,3-difluoroglutamic acid-containing methotrexate analogues (Hart ef al., J. Med. Chem. 39(1):56-65, 1996), methotrexate tetrahydroquinazoline analogue (Gangjee, et al., J. Heterocycl. Chem. 32(1):243-8, 1995), N-(α-aminoacyl) methotrexate derivatives (Cheung et al., Pteridines 3(1 -2): 101 -2, 1992), biotin methotrexate derivatives (Fan et al., Pteridines 3(1-2):131-2, 1992), D-glutamic acid or D- erythrou, threo-4-fluoroglutamic acid methotrexate analogues (McGuire et al., Biochem. Pharmacol. 42(12):2400-3, 1991), β,γ-methano methotrexate analogues (Rosowsky et al., Pteridines 2(3):133-9, 1991), 10- deazaaminopterin (10-EDAM) analogue (Braakhuis et al., Chem. Biol. Pteridines, Proe. Int. Sy mp. Pteridines Folic Acid Deriv., 1027-30, .1989), γ- tetrazole methotrexate analogue (Kalman ef al., Chem. Biol. Pteridines, Proc. Int. Symp. Pteridines Folic Acid Deriv. , 1154-7, 1989), N-(L-α- aminoacyl) methotrexate derivatives (Cheung et al., Heterocycles 2δ(2):751- 8, 1989), meta and ortho isomers of aminopterin (Rosowsky ef al., J. Med. Chem. 32(12):2582, 1989), hydroxymethy Methotrexate (DE 267495), γ- fluoromethotrexate (McGuire et al., Cancer Res. 49(16):4517-25, 1989), polyglutamyl methotrexate derivatives (Kumar ef a/., Cancer Res. 46(10):5020-3, 1986), gem-diphosphonate methotrexate analogues (WO 88/06158), α- and γ-substituted methotrexate analogues (Tsushima et al., Tetrahedron 44(17) :5375-87, 1988), 5-methyl~5-deaza methotrexate analogues (4,725,687), Nδ-acyl-Nα-(4-amino-4-deoxypteroyl)-L-ornithine derivatives (Rosowsky et al., J. Med. Chem. 37(7): 1332-7, 1988), 8-deaza methotrexate analogues (Kuehl ef al., Cancer Res. 4S(6):1481-8, 1988), acivicin methotrexate analogue (Rosowsky et al., J. Med. Chem. 30(8): 1463-9, 1987), polymeric platinol methotrexate derivative (Carraher ef al., Polym. ScI. Technol. (Plenum), 35(Adv. Biomed. Po/y/7?.J:311-24, 1987), methotrexate-γ-dimyristoylphophatidylethanolamine (Kinsky et al., Biochim. Biophys. Acta 977(2):211-18, 1987), methotrexate polyglutamate analogues (Rosowsky et al., Chem. Biol. Pteridines, Pteridines Folic Acid Deriv., Proc. Int. Symp. Pteridines Folic Acid Deriv.: Chem., Biol. Clin. Aspects: 985-8, 1986), poly-γ-glutamyl methotrexate derivatives (Kisliuk et al., Chem. Biol. Pteridines, Pteridines Folic Acid Deriv., Proc. Int. Symp. Pteridines Folic Acid Deriv.: Chem., Biol. Clin. Aspects: 989-92, 1986), deoxyuridylate methotrexate derivatives (Webber et al., Chem. Biol. Pteridines, Pteridines Folic Acid Deriv., Proc. Int. Symp. Pteridines Folic Acid Deriv.: Chem., Biol. Clin. Aspects: 659-62, 1986), iodoacetyl lysine methotrexate analogue (Delcamp et al., Chem. Biol. Pteridines, Pteridines Folic Acid Deriv., Proc. Int. Symp. Pteridines Folic Acid Deriv.: Chem., Biol. Clin. Aspects: 807-9, 1986), 2,.omega.-diaminoalkanoid acid-containing methotrexate analogues (McGuire et al., Biochem. Pharmacol. 35(15):2607-13, 1986), polyglutamate methotrexate derivatives (Kamen & Winick, Methods Enzymol. 722(Vitam. Coenzymes, Pt. G):339-46, 1986), 5-methyl-5-deaza analogues (Piper et al., J. Med. Chem. 29(6): 1080-7, 1986), quinazoline methotrexate analogue (Mastropaolo et al., J. Med. Chem. 29(1):155-8, 1986), pyrazine methotrexate analogue (Lever & Vestal, J. Heterocycl. Chem. 22(1):5-6, 1985), cysteic acid and homocysteic acid methotrexate analogues (4,490,529), γ-tert-butyl methotrexate esters (Rosowsky et al., J. Med. Chem. 28(5):660-7, 1985), fluorinated methotrexate analogues (Tsushima et al., Heterocycles 23(1):45-9, 1985), folate methotrexate analogue (Trombe, J. Bacteήol. 760(3):849-53, 1984), phosphonoglutamic acid analogues (Sturtz & Guillamot, Eur. J. Med. Chem.-Chim. Ther. 19(3):267-73, 1984), poly (L-lysine) methotrexate conjugates (Rosowsky et al., J. Med. Chem. 27(7):888-93, 1984), dilysine and trilysine methotrexate derivates (Forsch & Rosowsky, J. Org. Chem. 49(7): 1305-9, 1984), 7-hydroxymethotrexate (Fabre et al., Cancer Res. 43(10):4648-52, 1983), poly-γ-glutamyl methotrexate analogues (Piper & Montgomery, Adv. Exp. Med. Biol., 163(FoIyI Antifolyl Polyglutamates):95-W0, 1983), β'.δ1- dichloromethotrexate (Rosowsky & Yu, J. Med. Chem. 26(10): 1448-52, 1983), diazoketone and chloromethylketone methotrexate analogues (Gangjee et al., J. Pharm. ScL 77(6):717-19, 1982), 10- propargylaminopterin and alkyl methotrexate homologs (Piper et al., J. Med. Chem. 25(7):877-80, 1982), lectin derivatives of methotrexate (Lin et al., JNCI 66(3):523-8, 1981), polyglutamate methotrexate derivatives (Galivan, MoI. Pharmacol. 17(1): 105-10, 1980), halogentated methotrexate derivatives (Fox, JNCI 58(4):J955-8, 1977), 8-alkyl-7,8-dihydro analogues (Chaykovsky et al., J. Med. Chem. 20(10):J 1323-7, 1977), 7-methyl methotrexate derivatives and dichloromethotrexate (Rosowsky & Chen, J. Med. Chem. 77(12):J1308-11, 1974), lipophilic methotrexate derivatives and 3',5'-dichloromethotrexate (Rosowsky, J. Med. Chem. 76(10):J1190-3, 1973), deaza amethopterin analogues (Montgomery et al., Ann. N.Y. Acad. Sci. 786:J227-34, 1971), MX068 (Pharma Japan, 1658:18, 1999) and cysteic acid and homocysteic acid methotrexate analogues (EPA 0142220).
These compounds are believed to act as antimetabolites of folic acid.
4) Podophyllotoxins
In another aspect, the anti-infective therapeutic agent is a podophyllotoxin, or a derivative or an analogue thereof. Exemplary compounds of this type are etoposide or teniposide, which have the following structures:
Figure imgf000104_0001
Other representative examples of podophyllotoxins include Cu(II)-VP-16 (etoposide) complex (Tawa et al., Bioorg. Med. Chem. 6(7): 1003-1008, 1998), pyrrolecarboxamidino-bearing etoposide analogues (Ji et al., Bioorg. Med. Chem. Lett. 7(5):607-612, 1997), 4β-amino etoposide analogues (Hu, University of North Carolina Dissertation, 1992), γ-lactone ring-modified arylamino etoposide analogues (Zhou et al., J. Med. Chem. 37(2):287-92, 1994), N-glucosyl etoposide analogue (Allevi et al., Tetrahedron Lett. 34(45):7313-16, 1993), etoposide A-ring analogues (Kadow ef al., Bioorg. Med. Chem. Lett. 2(1):17-22, 1992), 4'-deshydroxy-4'- methyl etoposide (Saulnier et al., Bioorg. Med. Chem. Lett. 2(10):1213-18, 1992), pendulum ring etoposide analogues (Sinha et al., Eur. J. Cancer 26(5):590-3, 1990) and E-ring desoxy etoposide analogues (Saulnier et al., J. Med. Chem. 32(7): 1418-20, 1989).
These compounds are believed to act as topoisomerase Il inhibitors and/or DNA cleaving agents.
5) Camptothecins
In another aspect, the anti-infective therapeutic agent is camptothecin, or an analogue or derivative thereof. Camptothecins have the following general structure.
Figure imgf000105_0001
In this structure, X is typically O, but can be other groups, e.g., NH in the case of 21-lactam derivatives. Ri is typically H or OH, but may be other groups, e.g., a terminally hydroxylated C1-3 alkane. R2 is typically H or an amino containing group such as (CHs)2NHCH2, but may be other groups e.g., NO2, NH2, halogen (as disclosed in, e.g., U.S. Patent 5,552,156) or a short alkane containing these groups. R3 is typically H or a short alkyl such as C2H5. R4 is typically H but may be other groups, e.g., a methylenedioxy group with R1.
Exemplary camptothecin compounds include topotecan, irinotecan (CPT-11), 9-aminocamptothecin, 21-lactam-20(S)-camptothecin, 10,11-methylenedioxycamptothecin, SN-38, 9-nitrocamptothecin, 10- hydroxycamptothecin. Exemplary compounds have the structures:
Figure imgf000106_0001
SN-38: OH H C2H5
X: O for most analogs, NH for 21 -lactam analogs
Camptothecins have the five rings shown here. The ring labeled E must be intact (the lactone rather than carboxylate form) for maximum activity and minimum toxicity.
Camptothecins are believed to function as topoisomerase I inhibitors and/or DNA cleavage agents.
6) Hydroxyureas
The anti-infective therapeutic agent of the present invention may be a hydroxyurea. Hydroxyureas have the following general structure:
Figure imgf000106_0002
Suitable hydroxyureas are disclosed in, for example, U.S. Patent No. 6,080,874, wherein Ri is:
Figure imgf000107_0001
and R2 is an alkyl group having 1-4 carbons and R3 is one of H, acyl, methyl, ethyl, and mixtures thereof, such as a methylether.
Other suitable hydroxyureas are disclosed in, e.g., U.S. Patent No. 5,665,768, wherein R1 is a cycloalkenyl group, for example N-(3-(5-(4- fluorophenylthio)-furyl)-2-cyclopenten-1-yl)N-hydroxyurea; R2 is H or an alkyl group having 1 to 4 carbons and R3 is H; X is H or a cation.
Other suitable hydroxyureas are disclosed in, e.g., U.S. Patent No. 4,299,778, wherein Ri is a phenyl group substituted with one or more fluorine atoms; R2 is a cyclopropyl group; and R3 and X is H.
Other suitable hydroxyureas are disclosed in, e.g., U.S. Patent No. 5,066,658, wherein R2 and R3 together with the adjacent nitrogen form:
Figure imgf000107_0002
where in m is 1 or 2, n is 0-2 and Y is an alkyl group.
In one aspect, the hydroxyurea has the structure:
Figure imgf000107_0003
Hydroxyurea
These compounds are thought to function by inhibiting DNA synthesis. 7) Platinum complexes
In another aspect, the anti-infective therapeutic agent is a platinum compound. In general, suitable platinum complexes may be of Pt(II) or Pt(IV) and have this basic structure:
Figure imgf000108_0001
wherein X and Y are anionic leaving groups such as sulfate, phosphate, carboxylate, and halogen; R1 and R2 are alkyl, amine, amino alkyl any may be further substituted, and are basically inert or bridging groups. For Pt(II) complexes Z1 and Z2 are non-existent. For Pt(IV) Z1 and Z2 may be anionic groups such as halogen, hydroxy, carboxylate, ester, sulfate or phosphate. See, e.g., U.S. Patent Nos. 4,588,831 and 4,250,189.
Suitable platinum complexes may contain multiple Pt atoms. See, e.g., U.S. Patent Nos. 5,409,915 and 5,380,897. For example bisplatinum and triplatinum complexes of the type:
Figure imgf000109_0001
Exemplary platinum compounds are cisplatin, carboplatin, oxaliplatin, and miboplatin having the structuresf
Figure imgf000109_0002
Cisplatin Carboplatin
Figure imgf000109_0003
Oxaliplatin
Figure imgf000109_0004
Other representative platinum compounds include (CPA)2Pt(DOLYM) and (DACH)Pt(DOLYM) cisplatin (Choi et al., Arch. Pharmacal Res. 22(2):151-156, 1999), Cis-(PtCI2(4,7-H-5-methyl-7- oxo)1, 2,4(triazolo(1,5-a)pyrimidine)2) (Navarro et al., J. Med. Chem. 47(3):332-338, 1998), (Pt(cis-1 ,4-DACH)(trans-CI2)(CBDCA)) . Y2MeOH cisplatin (Shamsuddin et al., Inorg. Chem. 36(25):5969-5971 , 1997), 4- pyridoxate diammine hydroxy platinum (Tokunaga et al., Pharm. Sci. 3(7):353-356, 1997), Pt(II) ... Pt(II) (Pt2(NHCHN(C(CH2)(CH3)))4) (Navarro et al., Inorg. Chem. 35(26):7829-7835, 1996), 254-S cisplatin analogue (Koga et al., Neurol. Res. 78(3):244-247, 1996), o-phenylenediamine ligand bearing cisplatin analogues (Koeckerbauer & Bednarski, J. Inorg. Biochem. 62(4):281-298, 1996), trans, cis-(Pt(OAc)2l2(en)) (Kratochwil et al., J. Med. Chem. 39(13):2499-2507, 1996), estrogenic 1,2-diarylethylenediamine ligand (with sulfur-containing amino acids and glutathione) bearing cisplatin analogues (Bednarski, J. Inorg. Biochem. 62(1):75, 1996), cis-1 ,4- diaminocyclohexane cisplatin analogues (Shamsuddin et al., J. Inorg. Biochem. 67(4):291-301 , 1996), 5' orientational isomer of cis-(Pt(NH3)(4- aminoTEMP-O){d(GpG)}) (Dunham & Lippard, J. Am. Chem. Soc. 777(43): 10702-12, 1995), chelating diamine-bearing cisplatin analogues (Koeckerbauer & Bednarski, J. Pharm. Sci. 84(7):819-23, 1995), 1 ,2- diarylethyleneamine ligand-bearing cisplatin analogues (Otto et al., J. Cancer Res. Clin. Oncol. 727(1 ):31 -8, 1995), (ethylenediamine)platinum(ll) complexes (Pasini et al., J. Chem. Soc, Dalton Trans. 4:579-85, 1995), Cl- 973 cisplatin analogue (Yang et al., Int. J. Oncol. 5(3):597-602, 1994), cis- diaminedichloroplatinum(ll) and its analogues cis-1 ,1- cyclobutanedicarbosylato(2R)-2-methyl-1 ,4-butanediamineplatinum(ll) and cis-diammine(glycolato)platinum (Claycamp & Zimbrick, J. Inorg. Biochem. 26(4):257-67, 1986; Fan et al., Cancer Res. 48(11):3135-9, 1988; Heiger- Bernays et al., Biochemistry 29(36):8461-6, 1990; Kikkawa et al., J. Exp. Clin. Cancer Res. 72(4):233-40, 1993; Murray et a/., Biochemistry 37(47):11812-17, 1992; Takahashi et al., Cancer Chemother. Pharmacol. 33(1):31-5, 1993), cϊs-amine-cyclohexylamine-dichloroplatinum(ll) (Yoshida et al., Biochem. Pharmacol. 48(4):793-9, 1994), gem-diphosphonate cisplatin analogues (FR 2683529), (meso-1,2-bis(2,6-dichloro-4- hydroxyplenyl)ethylenediamine) dichloroplatinum(ll) (Bednarski et al., J. Med. Chem. 35(23):4479-85, 1992), cisplatin analogues containing a tethered dansyl group (Hartwig et al., J. Am. Chem. Soc. 774(21):8292-3, 1992), platinum(ll) polyamines (Siegmann et al., Inorg. Met.-Containing Polym. Mater., (Proc. Am. Chem. Soc. Int. Symp.), 335-61 , 1990), cis- (3H)dichloro(ethylenediamine)platinum(ll) (Eastman, Anal. Biochem. 797(2):311-15, 1991), trans-diamminedichloroplatinum(ll) and cis- (Pt(NH3)2(N3-cytosine)CI) (Bellon & Lippard, Biophys. Chem. 35(2-3): 179- 88, 1990), 3H-cis-1,2-diaminocyclohexanedichloroplatinum(ll) and 3H-cis- 1 ,2-diaminocyclohexanemalonatoplatinum (II) (Oswald etal., Res. Commun. Chem. Pathol. Pharmacol. 64(1):41-58, 1989), diaminocarboxylatoplatinum (EPA 296321), trans-(D,1)-1 ,2-diaminocyclohexane carrier ligand-bearing platinum analogues (Wyrick & Chaney, J. Labelled Compd. Radiopharm. 25(4):349-57, 1988), aminoalkylaminoanthraquinone-derived cisplatin analogues (Kitov et al., Eur. J. Med. Chem. 23(4):381-3, 1988), spiroplatin, carboplatin, iproplatin and JM40 platinum analogues (Schroyen et al., Eur. J. Cancer Clin. Oncol. 24(8): 1309-12, 1988), bidentate tertiary diamine- containing cisplatinum derivatives (Orbell et al., Inorg. Chim. Acta 152(2): 125-34, 1988), platinum(ll), platinum(IV) (Liu & Wang, Shandong Yike Daxue Xuehao 24(1):35-41 , 1986), cis-diammine(1,1- cyclobutanedicarboxylato-)platinum(ll) (carboplatin, JM8) and ethylenediammine-malonatoplatinum(ll) (JM40) (Begg et al., Radiother. Oncol. 9(2):157-65, 1987), JM8 and JM9 cisplatin analogues (Harstrick et al., Int. J. Androl. 70(1); 139-45, 1987), (NPr4)2((PtCL4).cis-(PtCI2- (NH2Me)2)) (Brammer et al., J. Chem. Soc, Chem. Commun. 6:443-5, 1987), aliphatic tricarboxylic acid platinum complexes (EPA 185225), and cis-dichloro(amino acid)(tert-butylamine)platinum(ll) complexes (Pasini & Bersanetti, Inorg. Chim. Acta 107(4)259-67, 1985). These compounds are thought to function by binding to DNA, i.e., acting as alkylating agents of DNA. 8) Other Anti-Infective Agents
In another aspect, the anti-infective therapeutic agent is a quinolone antibacterial agent. Representative examples of quinolone antibacterial agents include garenoxacin (Schering Plough) or an analogue or derivative thereof.
Dosages of Anti-Infective Agents
The drug dose administered from the present compositions for prevention or inhibition of infection in accordance with the present invention will depend on a variety of factors, including the type of formulation, the location of the treatment site, and the type of condition being treated. However, certain principles can be applied in the application of this art. Drug dose can be calculated as a function of dose per unit area (of the treatment site), total drug dose administered can be measured and appropriate surface concentrations of active drug can be determined. Drugs are to be used .at concentrations that range from several times more than to 50%, 20%, 10%, 5%, or even less than 1% of the concentration typically used in a single anti-infective systemic dose application. In certain aspects, the anti- infective agent is released from the composition in effective concentrations in a time period that may be measured from the time of infiltration into tissue adjacent to the device, which ranges from about less than 1 day to about 180 days. Generally, the release time may also be from about less than 1 day to about 180 days; from about 7 days to about 14 days; from about 14 days to about 28 days; from about 28 days to about 56 days; from about 56 days to about 90 days; from about 90 days to about 180 days.
The exemplary anti-infective agents, used alone or in combination, should be administered under the following dosing guidelines. The total amount (dose) of anti-infective agent in the composition can be in the range of about 0.01 μg-1 μg, or about 1 μg-10 μg, or about 10 μg-1 mg, or about 1 mg to 10 mg, or about 10 mg-100 mg, or about 100 mg to 250 mg, or about 250 mg-1000 mg. The dose (amount) of anti-infective agent per unit area of device or tissue surface to which the agent is applied may be in the range of about 0.01 μg/mm2 - 1 μg/mm2, or about 1 μg/mm2 - 10 μg/mm2, or about 10 μg/mm2 - 100 μg/mm2, or about 100 μg/mm2 to 250 μg/mm2, or about 250 μg/mm2 - 1000 μg/mm2. As different compositions will release the anti-infective agent at differing rates, the above dosing parameters should be utilized in combination with the release rate of the drug from the composition such that a minimum concentration of about 10"8 M to 10"7 M, or about 10"7 M to lO'6 M about 10"6 M to 10"5 M or about 10"5 M to 10'4 M of the agent is maintained on the tissue surface.
(a) Anthracvclines. Utilizing the anthracycline doxorubicin as an example, whether applied as a polymer coating, incorporated into the polymers which make up the implant components, or applied without a carrier polymer, the total dose of doxorubicin applied to the device or implant should not exceed 25 mg (range of 0.1 μg to 25 mg). In a particularly preferred embodiment, the total amount of drug applied should be in the range of 1 μg to 5 mg. The dose per unit area (i.e., the amount of drug as a function of the surface area of the portion of the implant to which drug is applied and/or incorporated) should fall within the range of 0.01 μg - 100 μg per mm2 of surface area. In a particularly preferred embodiment, doxorubicin should be applied to the implant surface at a dose of 0.1 μg/mm2 - 10 μg/mm2. As different polymer and non-polymer coatings will release doxorubicin at differing rates, the above dosing parameters should be utilized in combination with the release rate of the drug from the implant surface such that a minimum concentration of 10"7- 10"4 M of doxorubicin is maintained on the surface. It is necessary to insure that surface drug concentrations exceed concentrations of doxorubicin known to be lethal to multiple species of bacteria and fungi (i.e., are in excess of 10"4 M; although for some embodiments lower concentrations are sufficient). In a preferred embodiment, doxorubicin is released from the surface of the implant such that anti-infective activity is maintained for a period ranging from several hours to several months. In a particularly preferred embodiment the drug is released in effective concentrations for a period ranging from 1 week - 6 months. It should be readily evident based upon the discussions provided herein that analogues and derivatives of doxorubicin (as described previously) with similar functional activity can be utilized for the purposes of this invention; the above dosing parameters are then adjusted according to the relative potency of the analogue or derivative as compared to the parent compound (e.g., a compound twice as potent as doxorubicin is administered at half the above parameters, a compound half as potent as doxorubicin is administered at twice the above parameters, etc.).
Utilizing mitoxantrone as another example of an anthracycline, whether applied as a polymer coating, incorporated into the polymers which make up the device or implant, or applied without a carrier polymer, the total dose of mitoxantrone applied should not exceed 5 mg (range of 0.01 μg to 5 mg). In a particularly preferred embodiment, the total amount of drug applied should be in the range of- 0.-1 μg to 1 mg. The dose per unit area (i.e., the amount of drug as a function of the surface area of the portion of the implant to which drug is applied and/or incorporated) should fall within the range of 0.01 μg - 20 μg per mm2 of surface area. In a particularly preferred embodiment, mitoxantrone should be applied to the implant surface at a dose of 0.05 μg/mm2 - 3 μg/mm2. As different polymer and non-polymer coatings will release mitoxantrone at differing rates, the above dosing parameters should be utilized in combination with the release rate of the drug from the implant surface such that a minimum concentration of 10"5 - 10"6 M of mitoxantrone is maintained. It is necessary to insure that drug concentrations on the implant surface exceed concentrations of mitoxantrone known to be lethal to multiple species of bacteria and fungi (i.e., are in excess of 10"5 M; although for some embodiments lower drug levels will be sufficient). In a preferred embodiment, mitoxantrone is released from the surface of the implant such that anti-infective activity is maintained for a period ranging from several hours to several months. In a particularly preferred embodiment the drug is released in effective concentrations for a period ranging from 1 week - 6 months. It should be readily evident based upon the discussions provided herein that analogues and derivatives of mitoxantrone (as described previously) with similar functional activity can be utilized for the purposes of this invention; the above dosing parameters are then adjusted according to the relative potency of the analogue or derivative as compared to the parent compound (e.g., a compound twice as potent as mitoxantrone is administered at half the above parameters, a compound half as potent as mitoxantrone is administered at twice the above parameters, etc.).
(b) Fluoropyrimidines Utilizing the fluoropyrimidine 5- fluorouracil as an example, whether applied as a polymer coating, incorporated into the polymers which make up the device or implant, or applied without a carrier polymer, the total dose of 5-fluorouracil applied should not exceed 250 mg (range of 1.0 μg to 250 mg). In a particularly preferred-embodiment, the total amount of drug applied should be in the range of 10 μg to 25 mg. The dose per unit area (i.e., the amount of drug as a function of the surface area of the portion of the implant to which drug is applied and/or incorporated) should fall within the range of 0.1 μg - 1 mg per mm2 of surface area. In a particularly preferred embodiment, 5- fluorouracil should be applied to the implant surface at a dose of 1.0 μg/mm2 - 50 μg/mm2. As different polymer and non-polymer coatings will release 5- fluorouracil at differing rates, the above dosing parameters should be utilized in combination with the release rate of the drug from the implant surface such that a minimum concentration of 10"4- 10"7 M of 5-fluorouracil is maintained. It is necessary to insure that surface drug concentrations exceed concentrations of 5-fluorouracil known to be lethal to numerous species of bacteria and fungi (Ae., are in excess of 10"4 M; although for some embodiments lower drug levels will be sufficient). In a preferred embodiment, 5-fluorouracil is released from the implant surface such that anti-infective activity is maintained for a period ranging from several hours to several months. In a particularly preferred embodiment the drug is released in effective concentrations for a period ranging from 1 week - 6 months. It should be readily evident based upon the discussions provided herein that analogues and derivatives of 5-fluorouracil (as described previously) with similar functional activity can be utilized for the purposes of this invention; the above dosing parameters are then adjusted according to the relative potency of the analogue or derivative as compared to the parent compound (e.g., a compound twice as potent as 5-fluorouracil is administered at half the above parameters, a compound half as potent as 5-fluorouracil is administered at twice the above parameters, etc.).
(c) Podophylotoxins Utilizing the podophylotoxin etoposide as an example, whether applied as a polymer coating, incorporated into the polymers which make up the device or implant, or applied without a carrier polymer, the total dose of etoposide applied should not exceed 25 mg (range of 0.1 μg to 25 mg). In a particularly preferred embodiment, the total amount of drug applied should be in the range of 1 μg to 5 mg. The dose per unit area (i.e., the amount of drug as a function of the surface area of the portion of the implant to which drug is applied and/or incorporated) should fall within the range of 0.01 μg - 100 μg per mm2 of surface area. In a particularly preferred embodiment, etoposide should be applied to the implant surface at a dose of 0.1 μg/mm2 - 10 μg/mm2. As different polymer and non-polymer coatings will release etoposide at differing rates, the above dosing parameters should be utilized in combination with the release rate of the drug from the implant surface such that a concentration of 10"5 - 10~6 M of etoposide is maintained. It is necessary to insure that surface drug concentrations exceed concentrations of etoposide known to be lethal to a variety of bacteria and fungi (i.e., are in excess of 10"5 M; although for some embodiments lower drug levels will be sufficient). In a preferred embodiment, etoposide is released from the surface of the implant such that anti-infective activity is maintained for a period ranging from several hours to several months. In a particularly preferred embodiment the drug is released in effective concentrations for a period ranging from 1 week - 6 months. It should be readily evident based upon the discussions provided herein that analogues and derivatives of etoposide (as described previously) with similar functional activity can be utilized for the purposes of this invention; the above dosing parameters are then adjusted according to the relative potency of the analogue or derivative as compared to the parent compound (e.g., a compound twice as potent as etoposide is administered at half the above parameters, a compound half as potent as etoposide is administered at twice the above parameters, etc.).
It should be readily evident based upon the discussions provided herein that combinations of anthracyclines (e.g., doxorubicin or mitoxantrone), fluoropyrimidines (e.g., 5-fluorouracil), folic acid antagonists (e.g., methotrexate), podophylotoxins (e.g., etoposide), and/or quinolones can be utilized to enhance the antibacterial activity of the composition.
Polymers _ . . . _ .
The therapeutic agents useful in the present invention may also include various polymers. Such polymers may be used alone to be effective in certain applications (e.g., treating or preventing surgical adhesions) or in combination of an anti-fibrosis agent and/or an anti- infective agent to facilitate the delivery of, or to provide a sustained release formulation of, the anti-fibrosis agent and/or the anti-infective agent. Detailed descriptions of exemplary polymers are provided below in the section regarding pharmaceutical compositions, especially in the section regarding sustained release formulations.
Pharmaceutical Compositions
The present invention, in another aspect, provides pharmaceutical compositions that comprise a fibrosis-inhibiting agent and/or anti-infective agent. In certain embodiments, the pharmaceutical compositions further comprise a polymer, an additional therapeutic agent, a pharmaceutical excipient, and/or an agent that facilitates the delivery of the therapeutic agents or compositions.
Compositions That Comprise Polymers
In certain embodiments, the compositions of the present invention may comprise a polymer that itself is a therapeutic agent. In certain other embodiments, the compositions of the present invention may comprise a polymer that facilitates the delivery of a therapeutic agent or forms a sustained release formuation for a therapeutic agent. In certain embodiments, compositions that comprise polymers may further comprise additional agents (e.g., pharmaceutical exicipents, echogenic agents, etc.).
For instance, the composition may be or include a hydrophilic polymer gel that has anti-thrombogenic properties. Such a composition can be in the form of a coating that can comprise a hydrophilic, biodegradable polymer that is physically removed from the surface of the device over time, thus reducing adhesion of platelets to the device surface. The gel composition can include a polymer or a blend of polymers. Representative examples include alginates, chitosan and chitosan sulfate, hyaluronic acid, dextran sulfate, PLURONIC polymers (e.g., F-127 or F87), chain extended PLURONIC polymers, various polyester-polyether block copolymers of various configurations (e.g., AB, ABA, or BAB, where A is a polyester such as PLA, PGA, PLGA, PCL or the like), examples of which include MePEG- PLA, PLA-PEG-PLA, and the like). In one embodiment, the anti-thrombotic composition can include a crosslinked gel formed from a combination of molecules (e.g., PEG) having two or more terminal electrophilic groups and two or more nucleophilic groups.
Sustained-Release Preparations of Therapeutic Agents In certain embodiments, desired therapeutic agents may be admixed with, blended with, conjugated to, or, otherwise modified to contain a polymer composition (which may be either biodegradable or non- biodegradable) or a non-polymeric composition in order to release the therapeutic agent over a prolonged period of time. For many of the aforementioned embodiments, localized delivery as well as localized sustained delivery of the fibrosis-inhibiting and/or anti-infective agent may be required. For example, a desired therapeutic agent may be admixed with, blended with, conjugated to, or, otherwise modified to contain a polymeric composition (which may be either biodegradable or nonbiodegradable) or non-polymeric composition in order to release the therapeutic agent over a period of time.
Representative examples of biodegradable polymers suitable for the delivery of the aforementioned therapeutic agents include albumin, collagen, gelatin, hyaluronic acid, starch, cellulose and cellulose derivatives (e.g., regenerated cellulose, methylcellulose, hydroxypropylcellulose, hydroxypropylmethylcellulose, carboxymethylcellulose, cellulose acetate phthalate, cellulose acetate succinate, hydroxypropylmethylcellulose phthalate), casein, dextrans, polysaccharides, fibrinogen, poly(ether ester) multiblock copolymers, based on poly(ethylene glycol) and polyφutylene terephthalate), tyrosine-derived polycarbonates (e.g., U.S. Patent No. 6,120,491), poly(hydroxyl acids), poly(D,L-lactide), poly(D,L-lactide-co- glycolide), poly(glycolide), poly(hydroxybutyrate), polydioxanone, poly(alkylcarbonate) and poly(orthoesters), polyesters, poly(hydroxyvaleric acid), polydioxanone, polyesters, poly(malic acid), poly(tartronic acid), poly(acrylamides), polyanhydrides, polyphosphazenes, poly(amino acids), poly(alkylene oxide)-poly(ester) block copolymers (e.g., X-Y, X-Y-X, Y-X-Y, R-(Y-X)n, or R-(X-Y)n, where X is a polyalkylene oxide (e.g., poly(ethylene glycol, polyφropylene glycol) and block copolymers of poly(ethylene oxide) and polypropylene oxide) (e.g., PLURONIC and PLURONIC R series of polymers from BASF Corporation, Mount Olive, NJ) and Y is a polyester, where the polyester may comprise the residues of one or more of the monomers selected from lactide, lactic acid, glycolide, glycolic acid, e- caprolactone, gamma-caprolactone, hydroxyvaleric acid, hydroxybutyric acid, beta-butyrolactone, gamma-butyrolactone, gamma-valerolactone, y- decanolactone, δ-decanolactone, trimethylene carbonate, 1 ,4-dioxane-2- one or 1 ,5-dioxepan-2one (e.g., PLGA, PLA, PCL, polydioxanone and copolymers thereof) and R is a multifunctional initiator), and the copolymers as well as blends thereof (see generally, Ilium, L., Davids, S. S. (eds.) "Polymers in Controlled Drug Delivery" Wright, Bristol, 1987; Arshady, J. Controlled Release 17:1-22, 1991 ; Pitt, Int. J. Phar. 59:173-196, 1990; Holland et al., J. Controlled Release 4:155-0180, 1986).
Representative examples of non-degradable polymers suitable for the delivery of fibrosis-inhibiting agents include poly(ethylene-co-vinyl acetate) ("EVA") copolymers, non-degradable polyesters, such as poly(ethylene terephthalate), silicone rubber, acrylic polymers (polyacrylate, polyacrylic acid, polymethylacrylic acid, polymethylmethacrylate, poly(butyl methacrylate)), poly(alkylcynoacrylate) (e.g., poly(ethylcyanoacrylate), poly(butylcyanoacrylate) poly(hexylcyanoacrylate) poly(octylcyanoacrylate)), -acrylic resin, polyethylene, polypropylene, polyamides (nylon 6,6), polyurethanes (e.g., CHRONOFLEX AR, CHRONOFLEX AL, BIONATE, and PELLETHANE), poly(ester urethanes), poly(ether urethanes), poly(ester-urea), cellulose esters (e.g., nitrocellulose), polyethers (poly(ethylene oxide), polyφropylene oxide), polyoxyalkylene ether block copolymers based on ethylene oxide and propylene oxide such as the PLURONIC polymers (e.g., F-127 or F87) from BASF Corporation (Mount Olive, NJ), and poly(tetramethylene glycol), styrene-based polymers (polystyrene, poly(styrene sulfonic acid), poly(styrene)-block- poly(isobutylene)-block-poly(styrene), poly(styrene)-poly(isoprene) block copolymers), and vinyl polymers (polyvinylpyrrolidone, polyvinyl alcohol), polyvinyl acetate phthalate) as well as copolymers and blends thereof. Polymers may also be developed which are either anionic (e.g., alginate, carrageenan, carboxymethyl cellulose, poly(acrylamido-2-methyl propane sulfonic acid) and copolymers thereof, poly(methacrylic acid and copolymers thereof and poly(acrylic acid) and copolymers thereof, as well as blends thereof, or cationic (e.g., chitosan, poly-L-lysine, polyethylenimine, and poly(allyl amine)) and blends, copolymers and branched polymers thereof (see generally, Dunn et al., J. Applied Polymer ScL 50:353-365, 1993; Cascone et al., J. Materials Sci.: Materials in Medicine 5:770-77 '4, 1994; Shiraishi et al., Biol. Pharm. Bull. 76(11):1164-1168, 1993; Thacharodi and Rao, Int'I J. Pharm. -/20:115-118, 1995; Miyazaki et al., InVU. Pharm. 118:257-263, 1995).
Some examples of preferred polymeric carriers include poly(ethylene-co-vinyl acetate), polyurethanes (e.g., CHRONOFLEX AR, CHRONOFLEX AL, BIONATE, and PELLETHANE), poly (D,L-lactic acid) oligomers and polymers, poly (L-lactic acid) oligomers and polymers, poly (glycolic acid), copolymers of lactic acid and glycolic acid, poly (caprolactone), poly (valerolactone), polyanhydrides, copolymers of poly (caprolactone) or poly (lactic acid) with a polyethylene glycol (e.g., MePEG), poly(alkylene oxide)-poly(ester) block copolymers (e.g., X-Y, X-Y-X or Y-X- Y, R-(Y-X)n, R-(X-Y)n where X is a polyalkylene oxide and Y is a polyester (e.g., polyester can comprise the residues of one or more of the monomers selected from lactide, lactic acid, glycolide, glycolic acid, e-caprolactone, gamma-caprolactone, hydroxyvaleric acid, hydroxybutyric acid, beta- butyrolactone, gamma-butyrolactone, gamma-valerolactone, v- decanolactone, δ-decanolactone, trimethylene carbonate, 1 ,4-dioxane-2- one or 1 ,5-dioxepan-2one.), R is a multifunctional initiator and copolymers as well as blends thereof), nitrocellulose, silicone rubbers, poly(styrene)block-poly(isobutylene)-block-poly(styrene), poly(acrylate) polymers and blends, admixtures, or co-polymers of any of the above. Other preferred polymers include collagen, poly(alkylene oxide)-based polymers, polysaccharides such as hyaluronic acid, chitosan and fucans, and copolymers of polysaccharides with degradable polymers, as well as blends thereof.
Other representative polymers capable of sustained localized delivery of anti-infective and/or fibrosis-inhibiting therapeutic agents include carboxylic polymers, polyacetates, polycarbonates, polyethers, polyethylenes, polyvinylbutyrals, polysilanes, polyureas, polyoxides, polystyrenes, polysulfides, polysulfones, polysulfonides, polyvinylhalides, pyrrolidones, rubbers, thermal-setting polymers, cross-linkable acrylic and methacrylic polymers, ethylene acrylic acid copolymers, styrene acrylic copolymers, vinyl acetate polymers and copolymers, vinyl acetal polymers and copolymers, epoxies, melamines, other amino resins, phenolic polymers, and copolymers thereof, water-insoluble cellulose ester polymers (including cellulose acetate propionate, cellulose acetate, cellulose acetate butyrate, cellulose nitrate, cellulose acetate phthalate, and mixtures thereof), polyvinylpyrrolidone, polyethylene glycols, polyethylene oxide, polyvinyl alcohol, polyethers, polysaccharides, hydrophilic polyurethane, polyhydroxyacrylate, dextran, xanthan, hydroxypropyl cellulose, and homopolymers and copolymers of N-vinylpyrrolidone, N-vinyllactam, N-vinyl butyrolactam, N-vinyl caprolactam, other vinyl compounds having polar pendant groups, acrylate and methacrylate having hydrophilic esterifying groups, hydroxyacrylate, and acrylic acid, and combinations thereof; cellulose esters and ethers, ethyl cellulose, hydroxyethyl cellulose, cellulose nitrate, cellulose acetate, cellulose acetate butyrate, cellulose acetate propionate, natural and synthetic elastomers, rubber, acetal, styrene polybutadiene, acrylic resin, polyvinylidene chloride, polycarbonate, homopolymers and copolymers of vinyl compounds, polyvinylchloride, and polyvinylchloride acetate.
Representative examples of patents relating to drug-delivery polymers and their preparation include PCT Publication Nos. WO 98/19713, WO 01/17575, WO 01/41821, WO 01/41822, and WO 01/15526 (as well as the corresponding U.S. applications), U.S. Patent Nos. 4,500,676, 4,582,865, 4,629,623, 4,636,524, 4,713,448, 4,795,741 , 4,913,743, 5,069,899, 5,099,013, 5,128,326, 5,143,724, 5,153,174, 5,246,698, 5,266,563, 5,399,351, 5,525,348, 5,800,412, 5,837,226, 5,942,555, 5,997,517, 6,007,833, 6,071 ,447, 6,090,995, 6,106,473, 6,110,483, 6,121 ,027, 6,156,345, 6,214,901 , 6,368,611 6,630,155, 6,528,080, RE37.950, 6,46,1631 , 6,143,314, 5,990,194, 5,792,469, 5,780,044, 5,759,563, 5,744,153, 5,739,176, 5,733,950, 5,681 ,873, 5,599,552, 5,340,849, 5,278,202, 5,278,201 , 6,589,549, 6,287,588, 6,201 ,072, 6,117,949, 6,004,573, 5,702,717, 6,413,539, 5,714,159, 5,612,052, and U.S. Patent Application Publication Nos. 2003/0068377, 2002/0192286, 2002/0076441, and 2002/0090398.
It should be obvious to one of skill in the art that the polymers as described herein can also be blended or copolymerized in various compositions as required to deliver therapeutic doses of fibrosis-inhibiting agents.
It should be also obvious to one of skill in the art that the polymers as described herein can also be blended or copolymerized in various compositions as required to deliver therapeutic doses of biologically active agents (such as anit-infective agents).
Polymeric carriers for anti-infective and/or fibrosis-inhibiting therapeutic agents can be fashioned in a variety of forms, with desired release characteristics and/or with specific properties depending upon the composition being utilized. For example, polymeric carriers may be fashioned to release a therapeutic agent upon exposure to a specific triggering event such as pH (see, e.g., Heller et al., "Chemically Self- Regulated Drug Delivery Systems," in Polymers in Medicine III, Elsevier Science Publishers B.V., Amsterdam, 1988, pp. 175-188; Kang et al., J. Applied Polymer ScL 48:343-354, 1993; Dong et al., J. Controlled Release 19:171-178, 1992; Dong and Hoffman, J. Controlled Release 75:141-152, 1991; Kim et al., J. Controlled Release 28:143-152, 1994; Comejo-Bravo et al., J. Controlled Release 33:223-229, 1995; Wu and Lee, Pharm. Res. 70(10):1544-1547, 1993; Serres et al., Pharm. Res. 73(2): 196-201 , 1996; Peppas, "Fundamentals of pH- and Temperature-Sensitive Delivery Systems," in Gurny et al. (eds.), Pulsatile Drug Delivery, Wissenschaftliche Verlagsgesellschaft mbH, Stuttgart, 1993, pp. 41-55; Doelker, "Cellulose Derivatives," 1993, in Peppas and Langer (eds.), Biopolymers I, Springer- Verlag, Berlin). Representative examples of pH-sensitive polymers include poly (acrylic acid) and its derivatives (including for example, homopolymers such as poly(aminocarboxylic acid); poly(acrylic acid); poly(methyl acrylic acid), copolymers of such homopolymers, and copolymers of poly(acrylic acid) and/or acrylate or acrylamide lmonomers such as those discussed above. Other pH sensitive polymers include polysaccharides such as cellulose acetate phthalate; hydroxypropylmethylcellulose phthalate; hydroxypropylmethylcellulose acetate succinate; cellulose acetate trimellilate; and chitosan. Yet other pH sensitive polymers include any mixture of a pH sensitive polymer and a water-soluble polymer.
Likewise, ant-infective and/or fibrosis-inhibiting therapeutic agents can be delivered via polymeric carriers which are temperature sensitive (see, e.g., Chen et al., "Novel Hydrogels of a Temperature- Sensitive PLURONIC Grafted to a Bioadhesive Polyacrylic Acid Backbone for Vaginal Drug Delivery," in Proceed. Intern. Symp. Control. ReI. Bioact. Mater. 22:167-168, Controlled Release Society, Inc., 1995; Okano, "Molecular Design of Stimuli-Responsive Hydrogels for Temporal Controlled Drug Delivery," in Proceed. Intern. Symp. Control. ReI. Bioact. Mater. 22:111-112, Controlled Release Society, Inc., 1995; Johnston et al., Pharm. Res. 9(3):425-433, 1992; Tung, Int'l J. Pharm. 707:85-90, 1994; Harsh and Gehrke, J. Controlled Release 77:175-186, 1991; Bae et al., Pharm. Res. 8(4):531-537, 1991 ; Dinarvand and D'Emanuele, J. Controlled Release 36:221-227, 1995; Yu and Grainger, "Novel Thermo-sensitive Amphiphilic Gels: Poly N-isopropylacrylamide-co-sodium acrylate-co-n-N- alkylacrylamide Network Synthesis and Physicochemical Characterization," Dept. of Chemical & Biological ScL, Oregon Graduate Institute of Science & Technology, Beaverton, OR, pp. 820-821; Zhou and Smid, "Physical Hydrogels of Associative Star Polymers," Polymer Research Institute, Dept. of Chemistry, College of Environmental Science and Forestry, State Univ. of New York, Syracuse, NY, pp. 822-823; Hoffman et al., "Characterizing Pore Sizes and Water 'Structure' in Stimuli-Responsive Hydrogels," Center for Bioengineering, Univ. of Washington, Seattle, WA, p. 828; Yu and Grainger, "Thermo-sensitive Swelling Behavior in Crosslinked N-isopropylacrylamide Networks: Cationic, Anionic and Ampholytic Hydrogels," Dept. of Chemical & Biological Sci., Oregon Graduate Institute of Science & Technology, Beaverton, OR, pp. 829-830; Kim et al., Pharm. Res. 9(3):283-290, 1992; Bae et al., Pharm. Res. 8(5):624-628, 1991; Kono et al., J. Controlled Release 30:69-75, 1994; Yoshida et al., J. Controlled Release 32:97-102, 1994; Okano et al., J. Controlled Release 36:125-133, 1995; Chun and Kim, J. Controlled Release 38:39-47, 1996; D'Emanuele and Dinarvand, Int'l J. Pharm. 118:237-242, 1995; Katono et al., J. Controlled Release 76:215-228, 1991; Hoffman, "Thermally Reversible Hydrogels Containing Biologically Active Species," in Migliaresi et al. (eds.), Polymers in Medicine III, Elsevier Science Publishers B.V., Amsterdam, 1988, pp. 161-167; Hoffman, "Applications of Thermally Reversible Polymers and Hydrogels in Therapeutics and Diagnostics," in Third International Symposium on Recent Advances in Drug Delivery Systems, Salt Lake City, UT, Feb. 24-27, 1987, pp. 297-305; Gutowska et al., J. Controlled Release 22:95-104, 1992; Palasis and Gehrke, J. Controlled Release 78:1-12, 1992; Paavola et al., Pharm. Res. 72(12): 1997-2002, 1995).
Representative examples of thermogelling polymers, and the gelatin temperature (LCST (0C)) include homopolymers such as poly(N-methyl-N-n-propylacrylamide), 19.8; poly(N-n-propylacrylamide), 21.5; poly(N-methyl-N-isopropylacrylamide), 22.3; poly(N-n-propylmethacrylamide), 28.0; poIy(N-isopropylacrylamide), 30.9; poly(N, n-diethylacrylamide), 32.0; poly(N-isopropylmethacrylamide), 44.0; poly(N-cyclopropylacrylamide), 45.5; poly(N-ethylmethyacrylamide), 50.0; poly(N-methyl-N-ethylacrylamide), 56.0; poly(N-cyclopropylmethacrylamide), 59.0; poly(N-ethylacrylamide), 72.0. Moreover thermogelling polymers may be made by preparing copolymers between (among) monomers of the above, or by combining such homopolymers with other water-soluble polymers such as acrylmonomers (e.g., acrylic acid and derivatives thereof, such as methylacrylic acid, acrylate monomers and derivatives thereof, such as butyl methacrylate, butyl acrylate, lauryl acrylate, and acrylamide monomers and derivatives thereof, such as N-butyl acrylamide and acrylamide).
Other representative examples of thermogelling polymers include cellulose ether derivatives such as hydroxypropyl cellulose, 410C; methyl cellulose, 55°C; hydroxypropylmethyl cellulose, 66°C; and ethylhydroxyethyl cellulose, polyalkylene oxide-polyester block copolymers of the structure X-Y, Y-X-Y and X-Y-X where X in a polyalkylene oxide and Y is a biodegradable polyester (e.g., PLG-PEG-PLG) and PLURONICs such as F-127, 10 - 15°C; L-122, 190C; L-92, 260C; L-81 , 200C; and L-61 , 24°C.
Representative examples of patents relating to thermally gelling polymers and the preparation include U.S. Patent Nos. 6,451,346; 6,201 ,072; 6,117,949; 6,004,573; 5,702,717; and 5,484,610; and PCT Publication Nos. WO 99/07343; WO 99/18142; WO 03/17972; WQ . . 01/82970; WO 00/18821 ; WO 97/15287; WO 01/41735; WO 00/00222 and WO 00/38651.
Anti-infective and/or fibrosis-inhibiting therapeutic agents may be linked by occlusion in the polymer matrix, dissolution in the polymer, bound by covalent linkages, bound by ionic interactions, or encapsulated in microcapsules. Within certain embodiments of the invention, therapeutic compositions are provided in non-capsular formulations such as microspheres (ranging from nanometers to micrometers in size), pastes, threads of various size, films, or sprays. In one aspect, the anti-scarring agent may be incorporated into biodegradable magnetic nanospheres. The nanospheres may be used, for example, to replenish an anti-scarring agent into an implanted intravascular device, such as a stent containing a weak magnetic alloy (see, e.g., Z. Forbes, B. B. Yellen, G. Friedman, K. Barbee. "An approach to targeted drug delivery based on uniform magnetic fields," IEEE Trans. Magn. 39(5): 3372-3377 (2003)). Within certain aspects of the present invention, therapeutic compositions of anti-infective and/or fibrosis-inhibiting agents may be fashioned in the form of microspheres, microparticles and/or nanoparticles having any size ranging from 50 nm to 500 μm, depending upon the particular use. These compositions can be. These compositions can be formed by spray-drying methods, milling methods, coacervation methods, W/O emulsion methods, W/O/W emulsion methods, and solvent evaporation methods. In other aspects, these compositions can include microemulsions, emulsions, liposomes and micelles. Alternatively, such compositions may also be readily applied as a "spray", which solidifies into a film or coating for use as a device/implant surface coating or to line the tissues of the implantation site. Such sprays may be prepared from microspheres of a wide array of sizes, including for example, from 0.1 μm to 3 μm, from 10 μm to 30 μm, and from 30 μm to 100 μm.
Therapeutic compositions that include anti-infective and/or anti-fibrosis agents may also be prepared in a variety of "paste" or gel forms. For example, within one embodiment of the invention, therapeutic compositions are provided which are liquid at one temperature {e.g., temperature greater than 37°C, such as 400C, 45°C, 5O0C, 550C or 600C), and solid or semi-solid at another temperature (e.g., ambient body temperature, or any temperature lower than 37°C). Such "thermopastes" may be readily made utilizing a variety of techniques (see, e.g., PCT Publication WO 98/24427). Other pastes may be applied as a liquid, which solidify in vivo due to dissolution of a water-soluble component of the paste and precipitation of encapsulated drug into the aqueous body environment. These "pastes" and "gels" containing therapeutic agents are particularly useful for application to the surface of tissues that will be in contact with the implant or device.
Within yet other aspects of the invention, the therapeutic compositions of the present invention may be formed as a film or tube. These films or tubes can be porous or non-porous. Preferably, such films or tubes are generally less than 5, 4, 3, 2, or 1 mm thick, more preferably less than 0.75 mm, 0.5 mm, 0.25 mm, or, 0.10 mm thick. Films or tubes can also be generated of thicknesses less than 50 μm, 25 μm or 10 μm. Such films are preferably flexible with a good tensile strength (e.g., greater than 50, preferably greater than 100, and more preferably greater than 150 or 200 N/cm2), good adhesive properties (Ae., adheres to moist or wet surfaces), and have controlled permeability. Fibrosis-inhibiting agents contained in polymeric films are particularly useful for application to the surface of a device or implant as well as to the surface of tissue, cavity or an organ.
Within further aspects of the present invention, polymeric carriers are provided which are adapted to contain and release a hydrophobic ant-infective and/or fibrosis-inhibiting compound, and/or the carrier containing the hydrophobic compound in combination with a carbohydrate, protein or polypeptide. Within certain embodiments, the polymeric carrier contains or comprises regions, pockets, or granules of one or more hydrophobic compounds. For example, within one embodiment of the invention, hydrophobic compounds may be incorporated within a matrix which contains the hydrophobic therapeutic compound, followed by incorporation of the matrix within the polymeric carrier. A variety of matrices can be utilized in this regard, including for example, carbohydrates and polysaccharides such as starch, cellulose, dextran, methylcellulose, sodium alginate, heparin, chitosan and hyaluronic acid, proteins or polypeptides such as albumin, collagen and gelatin. Within alternative embodiments, hydrophobic compounds may be contained within a hydrophobic core, and this core contained within a hydrophilic shell.
The anti-infective and/or fibrosis-inhibiting therapeutic agent may be delivered as a solution. The therapeutic agent can be incorporated directly into the solution to provide a homogeneous solution or dispersion. In certain embodiments, the solution is an aqueous solution. The aqueous solution may further include buffer salts, as well as viscosity modifying agents (e.g., hyaluronic acid, alginates, carboxymethylcellulose (CMC), and the like). In another aspect of the invention, the solution can include a biocompatible solvent or liquid oligomers and/or polymers, such as ethanol, DMSO, glycerol, PEG-200, PEG-300 or NMP. These compositions may further comprise a polymer such a degradable polyester, where the polyester may comprise the residues of one or more of the monomers selected from lactide, lactic acid, glycolide, glycolic acid, e-caprolactone, gamma-caprolactone, hydroxy valeric acid, hydroxybutyric acid, beta- butyrolactone, gamma-butyrolactone, gamma-valerolactone, y- decanolactone, δ-decanolactone, trimethylene carbonate, 1 ,4-dioxane-2- one or 1 ,5-dioxepan-2one, or block copolymers of the form X-Y, Y-X-Y, R- (Y-X)n, R-(X-Y)n and X-Y-X (where X in a polyalkylene oxide (e.g., poly(ethylene glycol, poly(propylene glycol) and block copolymers of poly(ethylene oxide) and polypropylene oxide) (e.g., PLURONIC and PLURONIC R series of polymers from BASF Corporation, Mount Olive, NJ) and Y is a biodegradable polyester, where the polyester may comprise the residues of one or more of the monomers selected from lactide, lactic acid, glycolide, glycolic acid, e-caprolactone, gamma-caprolactone, hydroxy valeric acid, hydroxybutyric acid, beta-butyrolactone, gamma- butyrolactone, gamma-valerolactone, γ-decanolactone, δ-decanolactone, trimethylene carbonate, 1 ,4-dioxane-2-one or 1 ,5-dioxepan-2one (e.g., PLG- PEG-PLG) and R is a multifunctional initiator).
Within another aspect of the invention, the therapeutic anti- infective and/or fibrosis-inhibiting agent can further comprise a secondary carrier. The secondary carrier can be in the form of microspheres (e.g., PLGA, PLLA, PDLLA, PCL, gelatin, polydioxanone, poly(alkylcyanoacrylate)), nanospheres (PLGA, PLLA, PDLLA, PCL, gelatin, polydioxanone, poly(alkylcyanoacrylate)), liposomes, emulsions, microemulsions, micelles (SDS, block copolymers of the form X-Y, Y-X-Y, R-(Y-X)n, R-(X-Y)n and X-Y-X (where X in a polyalkylene oxide (e.g., poly(ethylene glycol, polyφropylene glycol) and block copolymers of poly(ethylene oxide) and poly(propylene oxide) (e.g., PLURONIC and PLURONIC R series of polymers from BASF Corporation, Mount Olive, NJ) and Y is a biodegradable polyester, where the polyester may comprise the residues of one or more of the monomers selected from lactide, lactic acid, glycolide, glycolic acid, e-caprolactone, gamma-caprolactone, hydroxyvaleric acid, hydroxybutyric acid, beta-butyrolactone, gamma- butyrolactone, gamma-valerolactone, γ-decanolactone, δ-decanolactone, trimethylene carbonate, 1 ,4-dioxane-2-one or 1 ,5-dioxepan-2one (e.g., PLG- PEG-PLG) and R is a multifunctional initiator), zeolites or cyclodextrins. Other carriers that may likewise be utilized to contain and deliver anti-infective and/or fibrosis-inhibiting therapeutic agents described herein include: hydroxypropyl cyclodextrin (Cserhati and Hollo, Int. J. Pharm. 108:69-75, 1994), liposomes (see, e.g., Sharma et al., Cancer Res. 53:5877-5881, 1993; Sharma and Straubinger, Pharm. Res. 11 (60):889- 896, 1994; WO 93/18751; U.S. Patent No. 5,242,073), liposome/gel (WO 94/26254), nanocapsules (Bartoli et a!., J. Microencapsulation 7(2):191-197, 1990), micelles (Alkan-Onyuksel et al., Pharm. Res. ; 11(2):206-212, 1994), implants (Jampel et al., Invest. Ophthalm. Vis. Science 34(11):3076-3083, 1993; Walter et al., Cancer Res. 54:22017-2212, 1994), nanoparticles (Violante and Lanzafame PAACR), nanoparticles - modified (U.S. Patent No. 5,145,684), nanoparticles (surface modified) (U.S. Patent No. 5,399,363), micelle (surfactant) (U.S. Patent No. 5,403,858), synthetic phospholipid compounds (U.S. Patent No. 4,534,899), gas borne dispersion (U.S. Patent No. 5,301,664), liquid emulsions, foam, spray, gel, lotion, cream, ointment, dispersed vesicles, particles or droplets solid- or liquid- aerosols, microemulsions (U.S. Patent No. 5,330,756), polymeric shell (nano- and micro- capsule) (U.S. Patent No. 5,439,686), emulsion (Tarr et al., Pharm Res. 4: 62-165, 1987), nanospheres (Hagan et al., Proc. Intern. Symp. Control ReI. Bioact. Mater. 22, 1995; Kwon et al., Pharm Res. 12(2): 192-195; Kwon et al., Pharm Res. 70(7):970-974; Yokoyama et al., J. Contr. ReI. 32:269-277, 1994; Gref et al., Science 263:1600-1603, 1994; Bazile et al., J. Pharm. Sci. 84:493-498, 1994) and implants (U.S. Patent No. 4,882,168).
Within another aspect of the present invention, polymeric carriers can be materials that are formed in situ. In one embodiment, the precursors can be monomers or macromers that contain unsaturated groups that can be polymerized and/or cross-linked. The monomers or macromers can then, for example, be injected into the treatment area or onto the surface of the treatment area and polymerized in situ using a radiation source (e.g., visible or UV light) or a free radical system (e.g., potassium persulfate and ascorbic acid or iron and hydrogen peroxide). The polymerization step can be performed immediately prior to, simultaneously to or post injection of the reagents into the treatment site. Representative examples of compositions that undergo free radical polymerization reactions are described in WO 01/44307, WO 01/68720, WO 02/072166, WO 03/043552, WO 93/17669, WO 00/64977; U.S. Patent Nos. 5,900,245, 6,051 ,248, 6,083,524, 6,177,095, 6,201 ,065, 6,217,894, 6,639,014, 6,352,710, 6,410,645, 6,531 ,147, 5,567,435, 5,986,043, 6,602,975; U.S. Patent Application Publication Nos. 2002/012796A1, 2002/0127266A1 , 2002/0151650A1, 2003/0104032A1 , 2002/0091229A1 , and 2003/0059906A1.
In certain aspects, it is desirable to use compositions that can be administered as liquids, but subsequently form hydrogels at the site of administration. Such in situ hydrogel forming compositions can be administered as liquids from a variety of different devices, and are more adaptable for administration to any site, since they are not preformed. Examples of in situ forming hydrogels include photoactivatable mixtures of water-soluble co-polyester prepolymers and polyethylene glycol to create hydrogel barriers. Block copolymers of polyalkylene oxide polymers (e.g., PLURONIC compounds from BASF Corporation, Mount Olive, NJ) and poloxamers have been designed that are soluble in cold water, but form insoluble hydrogels that adhere to tissues at body temperature (Leach, et al., Am. J. Obstet. Gynecol. 162:1317-1319 (1990)).
In certain embodiments, the present invention provides for polymeric crosslinked matrices, and polymeric carriers, that may be used to assist in the prevention of the formation or growth of fibrous connective tissue. The composition may contain and deliver fibrosis-inhibiting agents in the vicinity of the implanted device. The following compositions are particularly useful when it is desired to infiltrate around the device, with or without a fibrosis-inhibiting agent. Such polymeric materials may be prepared from, e.g., (a) synthetic materials, (b) naturally-occurring materials, or (c) mixtures of synthetic and naturally occurring materials. The matrix may be prepared from, e.g., (a) a one-component, i.e., self-reactive, compound, or (b) two or more compounds that are reactive with one another. Typically, these materials are fluid prior to delivery, and thus can be sprayed or otherwise extruded from a delivery device (e.g., a syringe) in order to deliver the composition. After delivery, the component materials react with each other, and/or with the body, to provide the desired affect. In some instances, materials that are reactive with one another must be kept separated prior to delivery to the patient, and are mixed together just prior to being delivered to the patient, in order that they maintain a fluid form prior to delivery. In a preferred aspect of the invention, the components of the matrix are delivered in a liquid state to the desired site in the body, whereupon in situ polymerization occurs.
First and Second Synthetic Polymers In one embodiment, crosslinked polymer compositions (in other words, crosslinked matrices) are prepared by reacting a first synthetic polymer containing two or more nucleophilic groups with a second synthetic polymer containing two or more electrophilic groups, where the electrophilic groups are capable of covalently binding with the nucleophilic groups. In one embodiment, the first and second polymers are each non-immunogenic. In another embodiment, the matrices are not susceptible to enzymatic cleavage by, e.g., a matrix metalloproteinase (e.g., collagenase) and are therefore expected to have greater long-term persistence in vivo than collagen-based compositions.
As used herein, the term "polymer" refers inter alia to polyalkyls, polyamino acids, polyalkyleneoxides and polysaccharides. Additionally, for external or oral use, the polymer may be polyacrylic acid or carbopol. As used herein, the term "synthetic polymer" refers to polymers that are not naturally occurring and that are produced via chemical synthesis. As such, naturally occurring proteins such as collagen and naturally occurring polysaccharides such as hyaluronic acid are specifically excluded. Synthetic collagen, and synthetic hyaluronic acid, and their derivatives, are included. Synthetic polymers containing either nucleophilic or electrophilic groups are also referred to herein as "multifunctionally activated synthetic polymers." The term "multifunctionally activated" (or, simply, "activated") refers to synthetic polymers which have, or have-been chemically modified to have, two or more nucleophilic or electrophilic groups which are capable of reacting with one another (i.e., the nucleophilic groups react with the electrophilic groups) to form covalent bonds. Types of multifunctionally activated synthetic polymers include difunctionally activated, tetrafunctionally activated, and star-branched polymers.
Multifunctionally activated synthetic polymers for use in the present invention must contain at least two, more preferably, at least three, functional groups in order to form a three-dimensional crosslinked network with synthetic polymers containing multiple nucleophilic groups (i.e., "multi- nucleophilic polymers"). In other words, they must be at least difunctionally activated, and are more preferably trifunctionally or tetrafunctionally activated. If the first synthetic polymer is a difunctionally activated synthetic polymer, the second synthetic polymer must contain three or more functional groups in order to obtain a three-dimensional crosslinked network. Most preferably, both the first and the second synthetic polymer contain at least three functional groups.
Synthetic polymers containing multiple nucleophilic groups are also referred to generically herein as "multi-nucleophilic polymers." For use in the present invention, multi-nucleophilic polymers must contain at least two, more preferably, at least three, nucleophilic groups. If a synthetic polymer containing only two nucleophilic groups is used, a synthetic polymer containing three or more electrophilic groups must be used in order to obtain a three-dimensional crosslinked network.
Preferred multi-nucleophilic polymers for use in the compositions and methods of the present invention include synthetic polymers that contain, or have been modified to contain, multiple nucleophilic groups such as primary amino groups and thiol groups. Preferred multi-nucleophilic polymers include: (i) synthetic polypeptides that have been synthesized to contain two or more primary amino groups or thiol groups; and (ii)_polyethylene glycols that have been-modified to contain two or more primary amino groups or thiol groups. In general, reaction of a thiol group with an electrophilic group tends to proceed more slowly than reaction of a primary amino group with an electrophilic group.
In one embodiment, the multi-nucleophilic polypeptide is a synthetic polypeptide that has been synthesized to incorporate amino acid residues containing primary amino groups (such as lysine) and/or amino acids containing thiol groups (such as cysteine). Poly(lysine), a synthetically produced polymer of the amino acid lysine (145 MW), is particularly preferred. Poly(lysine)s have been prepared having anywhere from 6 to about 4,000 primary amino groups, corresponding to molecular weights of about 870 to about 580,000.
Poly(lysine)s for use in the present invention preferably have a molecular weight within the range of about 1 ,000 to about 300,000; more preferably, within the range of about 5,000 to about 100,000; most preferably, within the range of about 8,000 to about 15,000. Poly(lysine)s of varying molecular weights are commercially available from Peninsula Laboratories, Inc. (Belmont, Calif.) and Aldrich Chemical (Milwaukee, Wl).
Polyethylene glycol can be chemically modified to contain multiple primary amino or thiol groups according to methods set forth, for example, in Chapter 22 of Poly(ethylene Glycol) Chemistry: Biotechnical and Biomedical Applications, J. Milton Harris, ed., Plenum Press, N.Y. (1992). Polyethylene glycols which have been modified to contain two or more primary amino groups are referred to herein as "multi-amino PEGs." Polyethylene glycols which have been modified to contain two or more thiol groups are referred to herein as "multi-thiol PEGs." As used herein, the term "polyethylene glycol(s)" includes modified and or derivatized polyethylene glycol(s).
Various forms of multi-amino PEG are commercially available from Shearwater Polymers (Huntsville, Ala.) and from Huntsman Chemical Company (Utah) under the name "Jeffamine." Multi-amino PEGs useful in the present invention-include Huntsman's Jeffamine diamines ("D" series) and triamines ("T" series), which contain two and three primary amino groups per molecule, respectively.
Polyamines such as ethylenediamine (H2N-CH2-CH2-NH2), tetramethylenediamine (H2N-(C H2^-NH2), pentamethylenediamine (cadaverine) (H2N-(CH2)S-NH2), hexamethylenediamine (H2N-(CH2J6-NH2), di(2-aminoethyl)amine (HN-(CH2-CH2-NH2^), and tris(2-aminoethyl)amine (N-(CH2-CH2-NH2)3) may also be used as the synthetic polymer containing multiple nucleophillc groups.
Synthetic polymers containing multiple electrophilic groups are also referred to herein as "multi-electrophilic polymers." For use in the present invention, the multifunctionally activated synthetic polymers must contain at least two, more preferably, at least three, electrophilic groups in order to form a three-dimensional crosslinked network with multi- nucleophilic polymers. Preferred multi-electrophilic polymers for use in the compositions of the invention are polymers which contain two or more succinimidyl groups capable of forming covalent bonds with nucleophilic groups on other molecules. Succinimidyl groups are highly reactive with materials containing primary amino (NH2) groups, such as multi-amino PEG, poly(lysine), or collagen. Succinimidyl groups are slightly less reactive with materials containing thiol (SH) groups, such as multi-thiol PEG or synthetic polypeptides containing multiple cysteine residues.
As used herein, the term "containing two or more succinimidyl groups" is meant to encompass polymers which are preferably commercially available containing two or more succinimidyl groups, as well as those that must be chemically derivatized to contain two or more succinimidyl groups. As used herein, the term "succinimidyl group" is intended to encompass sulfosuccinimidyl groups and other such variations of the "generic" succinimidyl group. The presence of the sodium sulfite moiety on the sulfosuccinimidyl group serves to increase the solubility of the polymer.
Hydrophilic polymers and, in particular, various derivatized polyethylene glycols, are preferred for use in the compositions of the present invention. As used herein, the term "PEG" refers to polymers having the repeating structure (OCH2-CHb)n- Structures for some specific, tetrafunctionally activated forms of PEG are shown in FIGS. 4 to 13 of U.S. Patent 5,874,500, incorporated herein by reference. Examples of suitable PEGS include PEG succinimidyl propionate (SE-PEG), PEG succinimidyl succinamide (SSA-PEG), and PEG succinimidyl carbonate (SC-PEG). In one aspect of the invention, the crosslinked matrix is formed in situ by reacting pentaerythritol poly(ethylene glycol)ether tetra-sulfhydryl] (4-armed thiol PEG) and pentaerythritol poly(ethylene glycol)ether tetra-succinimidyl glutarate] (4-armed NHS PEG) as reactive reagents. Structures for these reactants are shown in U.S. Patent 5,874,500. Each of these materials has a core with a structure that may be seen by adding ethylene oxide-derived residues to each of the hydroxyl groups in pentaerythritol, and then derivatizing the terminal hydroxyl groups (derived from the ethylene oxide) to contain either thiol groups (so as to form 4-armed thiol PEG) or N- hydroxysuccinimydyl groups (so as to form 4-armed NHS PEG), optionally with a linker group present between the ethylene oxide derived backbone and the reactive functional group, where this product is commercially available as COSEAL from Angiotech Pharmaceuticals Inc. Optionally, a group "D" may be present in one or both of these molecules, as discussed in more detail below.
As discussed above, preferred activated polyethylene glycol derivatives for use in the invention contain succinimidyl groups as the reactive group. However, different activating groups can be attached at sites along the length of the PEG molecule. For example, PEG can be derivatized to form functionally activated PEG propionaldehyde (A-PEG), or functionally activated PEG glycidyl ether (E-PEG), or functionally activated PEG-isocyanate (I-PEG), or functionally activated PEG-vinylsulfone (V- PEG).
Hydrophobic polymers can also be used to prepare the compositions of the present invention. Hydrophobic polymers for use in the present invention preferably contain, or can be derivatized to contain, two or more electrophilic groups, such as succinimidyl groups, most preferably, two, three, or four electrophilic groups. As used herein, the term "hydrophobic polymer" refers to polymers which contain a relatively small proportion of oxygen or nitrogen atoms.
Hydrophobic polymers which already contain two or more succinimidyl groups include, without limitation, disuccinimidyl suberate (DSS), bis(sulfosuccinimidyl) suberate (BS3), dithiobis(succinimidylpropionate) (DSP), bis(2-succinimidooxycarbonyloxy) ethyl sulfone (BSOCOES), and 3,3'-dithiobis(sulfosuccinimidylpropionate (DTSPP), and their analogs and derivatives. The above-referenced polymers are commercially available from Pierce (Rockford, III.), under catalog Nos. 21555, 21579, 22585, 21554, and 21577, respectively.
Preferred hydrophobic polymers for use in the invention generally have a carbon chain that is no longer than about 14 carbons. Polymers having carbon chains substantially longer than 14 carbons generally have very poor solubility in aqueous solutions and, as such, have very long reaction times when mixed with aqueous solutions of synthetic polymers containing multiple nucleophilic groups.
Certain polymers, such as polyacids, can be derivatized to contain two or more functional groups, such as succinimidyl groups. Polyacids for use in the present invention include, without limitation, trimethylolpropane-based tricarboxylic acid, di(trimethylol propane)-based tetracarboxylic acid, heptanedioic acid, octanedioic acid (suberic acid), and hexadecanedioic acid (thapsic acid). Many of these polyacids are commercially available from DuPont Chemical Company (Wilmington, DE). According to a general method, polyacids can be chemically derivatized to contain two or more succinimidyl groups by reaction with an appropriate molar amount of N-hydroxysuccinimide (NHS) in the presence of N1N1- dicyclohexylcarbodiimide (DCC).
Polyalcohols such as trimethylolpropane and di(trimethylol propane) can be converted to carboxylic acid form using various methods, then further derivatized by reaction with NHS in the presence of DCC to produce trifunctionally and tetrafunctionally activated polymers, respectively, as described in U.S. application Ser. No. 08/403,358. Polyacids such as heptanedioic acid (HOOC-(CH2)5-COOH), octanedioic acid (HOOC-(CH2)6- COOH), and hexadecanedioic acid (HOOC-(CH2)i4-COOH) are derivatized by the addition of succinimidyl groups to produce difunctionally activated polymers.
Polyamines such as ethylenediamine, tetramethylenediamine, pentamethylenediamine (cadaverine), hexamethylenediamine, bis (2- aminoethyl)amine, and tris(2-aminoethyl)amine can be chemically derivatized to polyacids, which can then be derivatized to contain two or more succinimidyl groups by reacting with the appropriate molar amounts of N-hydroxysuccinimide in the presence of DCC, as described in U.S. application Ser. No. 08/403,358. Many of these polyamines are commercially available from DuPont Chemical Company.
In a preferred embodiment, the first synthetic polymer will contain multiple nucleophilic groups (represented below as "X") and it will react with the second synthetic polymer containing multiple electrophilic groups (represented below as "Y"), resulting in a covalently bound polymer network, as follows:
Polymer-Xm + Polymer-Yn → Polymer-Z-Polymer wherein m < 2, n < 2, and m + n ≤ 5; where exemplary X groups include -NH2, -SH, -OH, -PH2, CO- NH-NH2, etc., where the X groups may be the same or different in polymer-
Xm! where exemplary Y groups include -CO2-N(COCH2^, -CO2H, - CHO, -CHOCH2 (epoxide), -N=C=O, -SO2-CH=CH2, -N(COCH)2 (i.e., a five- membered heterocyclic ring with a double bond present between the two CH groups), -S-S-(CsH4N), etc., where the Y groups may be the same or different in polymer-Yn; and where Z is the functional group resulting from the union of a nucleophilic group (X) and an electrophilic group (Y).
As noted above, it is also contemplated by the present invention that X and Y may be the same or different, i.e., a synthetic polymer may have two different electrophilic groups, or two different nucleophilic groups, such as with glutathione.
In one embodiment, the backbone of at least one of the synthetic polymers comprises alkylene oxide residues, e.g., residues from ethylene oxide, propylene oxide, and mixtures thereof. The term 'backbone' refers to a significant portion of the polymer.
For example, the synthetic polymer containing alkylene oxide residues may be described by the formula X-polymer-X or Y-polymer-Y, wherein X and Y are as defined above, and the term "polymer" represents - (CH2CH2 O)n- or -(CH(CH3)CH2 O)n- or -(CH2-CH2-O)n-(CH(CH3)CH2-O)n-. In these cases the synthetic polymer would be difunctional.
The required functional group X or Y is commonly coupled to the polymer backbone by a linking group (represented below as "Q"), many of which are known or possible. There are many ways to prepare the various functionalized polymers, some of which are listed below:
Polymer-Qi-X + Polymer-Q2-Y → Polymer-Qi-Z-Q2-Polymer
Exemplary Q groups include -O-(CH2)n-; -S-(CH2)n-; -NH- (CHz)n-; -O2C-NH-(CH2)n-; -O2C-(CH2)n-; -O2C-(CR1H)n-; and -0-R2-CO-NH-, which provide synthetic polymers of the partial structures: polymer-O- (CH2)n-(X or Y); polymer-S-(CH2)n-(X or Y); polymer-NH-(CH2)n-(X or Y); polymer-O2C-NH-(CH2)n-(X or Y); polymer-O2C-(CH2)n-(X or Y); polymer-O2C-(CR1 H)n-(X or Y); and polymer-O-R2-CO-NH-(X or Y), respectively. In these structures, n = 1-10, R1 = H or alkyl (Ae., CH3, C2H5, etc.); R2 = CH2, or CO-NH-CH2CH2; and Q1 and Q2 may be the same or different.
For example, when Q2 = OCH2CH2 (there is no Qi in this case); Y = -CO2-N(COCH2)2; and X = -NH2, -SH, or -OH, the resulting reactions and Z groups would be as follows:
Polymer-NH2 + Polymer-O-CH2-CH2-CO2-N(COCH2)2 → PoIymer-NH-CO-CH2-CH2-O-Polymer;
Polymer-SH + Polymer-O-CH2-CH2-CO2-N(COCH2)2 → Polymer-S-COCH2CH2-O-Polymer; and
Polymer-OH + Polymer-O-CH2-CH2-CO2-N(COCH2)2 → Polymer-O-COCH2CH2-O-Polymer.
An additional group, represented below as "D", can be inserted between the polymer and the linking group, if present. One purpose of such a D group is to affect the degradation rate of the crosslinked polymer composition in vivo, for example, to increase the degradation rate, or to decrease the degradation rate. This may be useful in many instances, for example, when drug has been incorporated into the matrix, and it is desired to increase or decrease polymer degradation rate so as to influence a drug delivery profile in the desired direction. An illustration of a crosslinking reaction involving first and second synthetic polymers each having D and Q groups is shown below.
Polymer-D-Q-X + Polymer-D-Q-Y → Polymer-D-Q-Z-Q-D- Polymer
Some useful biodegradable groups "D" include polymers formed from one or more α-hydroxy acids, e.g., lactic acid, glycolic acid, and the cyclization products thereof (e.g., lactide, glycolide), ε-caprolactone, and amino acids. The polymers may be referred to as polylactide, polyglycolide, poly(co-lactide-glycolide); poly-ε-caprolactone, polypeptide (also known as poly amino acid, for example, various di- or tri-peptides) and poly(anhydride)s.
In a general method for preparing the crosslinked polymer compositions used in the context of the present invention, a first synthetic polymer containing multiple nucleophilic groups is mixed with a second synthetic polymer containing multiple electrophilic groups. Formation of a three-dimensional crosslinked network occurs as a result of the reaction between the nucleophilic groups on the first synthetic polymer and the electrophilic groups on the second synthetic polymer.
The concentrations of the first synthetic polymer and the second synthetic polymer used to prepare the compositions of the present invention will vary depending upon a number of factors, including the types and molecular weights of the particular synthetic polymers used and the desired end use application. In general, when using multi-amino PEG as the first synthetic polymer, it is preferably used at a concentration in the range of about 0.5 to about 20 percent by weight of the final composition, while the second synthetic polymer is used at a concentration in the range of about 0.5 to about 20 percent by weight of the final composition. For example, a final composition having a total weight of 1 gram (1000 milligrams) would contain between about 5 to about 200 milligrams of multi- amino PEG, and between about 5 to about 200 milligrams of the second synthetic polymer.
Use of higher concentrations of both first and second synthetic polymers will result in the formation of a more tightly crosslinked network, producing a stiffer, more robust gel. Compositions intended for use in tissue augmentation will generally employ concentrations of first and second synthetic polymer that fall toward the higher end of the preferred concentration range. Compositions intended for use as bioadhesives or in adhesion prevention do not need to be as firm and may therefore contain lower polymer concentrations.
Because polymers containing multiple electrophilic groups will also react with water, the second synthetic polymer is generally stored and used in sterile, dry form to prevent the loss of crosslinking ability due to hydrolysis which typically occurs upon exposure of such electrophilic groups to aqueous media. Processes for preparing synthetic hydrophilic polymers containing multiple electrophylic groups in sterile, dry form are set forth in U.S. Patent 5,643,464. For example, the dry synthetic polymer may be compression molded into a thin sheet or membrane, which can then be sterilized using gamma or, preferably, e-beam irradiation. The resulting dry membrane or sheet can be cut to the desired size or chopped into smaller size particulates. In contrast, polymers containing multiple nucleophilic groups are generally not water-reactive and can therefore be stored in aqueous solution.
In certain embodiments, one or both of the electrophilic- or nucleophilic-terminated polymers described above can be combined with a synthetic or naturally occurring polymer. The presence of the synthetic or naturally occurring polymer may enhance the mechanical and/or adhesive properties of the in situ forming compositions. Naturally occurring polymers, and polymers derived from naturally occurring polymer that may be included in in situ forming materials include naturally occurring proteins, such as collagen, collagen derivatives (such as methylated collagen), fibrinogen, thrombin, albumin, fibrin, and derivatives of and naturally occurring polysaccharides, such as glycosaminoglycans, including deacetylated and desulfated glycosaminoglycan derivatives.
In one aspect, a composition comprising naturally-occurring protein and both of the first and second synthetic polymer as described above is used to form the crosslinked matrix according to the present invention. In one aspect, a composition comprising collagen and both of the first and second synthetic polymer as described above is used to form the crosslinked matrix according to the present invention. In one aspect, a composition comprising methylated collagen and both of the first and second synthetic polymer as described above is used to form the crosslinked matrix according to the present invention. In one aspect, a composition comprising fibrinogen and both of the first and second synthetic polymer as described above is used to form the crosslinked matrix according to the present invention. In one aspect, a composition comprising thrombin and both of the first and second synthetic polymer as described above is used to form the crosslinked matrix according to the present invention. In one aspect, a composition comprising albumin and both of the first and second synthetic polymer as described above is used to form the crosslinked matrix according to the present invention. In one aspect, a composition comprising fibrin and both of the first and second synthetic polymer as described above is used to form the crosslinked matrix according to the present invention. In one aspect, a composition comprising naturally occurring polysaccharide and both of the first and second synthetic polymer as described above is used to form the crosslinked matrix according to the present invention. In one aspect, a composition comprising glycosaminoglycan and both of the first and second synthetic polymer as described above is used to form the crosslinked matrix according to the present invention. In one aspect, a composition comprising deacetylated glycosaminoglycan and both of the first and second synthetic polymer as described above is used to form the crosslinked matrix according to the present invention. In one aspect, a composition comprising desulfated glycosaminoglycan and both of the first and second synthetic polymer as described above is used to form the crosslinked matrix according to the present invention.
In one aspect, a composition comprising naturally-occurring protein and the first synthetic polymer as described above is used to form the crosslinked matrix according to the present invention. In one aspect, a composition comprising collagen and the first synthetic polymer as described above is used to form the crosslinked matrix according to the present invention. In one aspect, a composition comprising methylated collagen and the first synthetic polymer as described above is used to form the crosslinked matrix according to the present invention. In one aspect, a composition comprising fibrinogen and the first synthetic polymer as described above is used to form the crosslinked matrix according to the present invention. In one aspect, a composition comprising thrombin and the first synthetic polymer as described above is used to form the crosslinked matrix according to the present invention. In one aspect, a composition comprising albumin and the first synthetic polymer as described above is used to form the crosslinked matrix according to the present invention. In one aspect, a composition comprising fibrin and the first synthetic polymer as described above is used to form the crosslinked matrix according to the present invention. In one aspect, a composition comprising naturally occurring polysaccharide and the first synthetic polymer as described above is used to form the crosslinked matrix according to the present invention. In one aspect, a composition comprising glycosaminoglycan and the first synthetic polymer as described above is used to form the crosslinked matrix according to the present invention. In one aspect, a composition comprising deacetylated glycosaminoglycan and the first synthetic polymer as described above is used to form the crosslinked matrix according to the present invention. In one aspect, a composition comprising desulfated glycosaminoglycan and the first synthetic polymer as described above is used to form the crosslinked matrix according to the present invention.
In one aspect, a composition comprising naturally-occurring protein and the second synthetic polymer as described above is used to form the crosslinked matrix according to the present invention. In one aspect, a composition comprising collagen and the second synthetic polymer as described above is used to form the crosslinked matrix according to the present invention. In one aspect, a composition comprising methylated collagen and the second synthetic polymer as described above is used to form the crosslinked matrix according to the present invention. In one aspect, a composition comprising fibrinogen and the second synthetic polymer as described above is used to form the crosslinked matrix according to the present invention. In one aspect, a composition comprising thrombin and the second synthetic polymer as described above is used to form the crosslinked matrix according to the present invention. In one aspect, a composition comprising albumin and the second synthetic polymer as described above is used to form the crosslinked matrix according to the present invention. In one aspect, a composition comprising fibrin and the second synthetic polymer as described above is used to form the crosslinked matrix according to the present invention. In one aspect, a composition comprising naturally occurring polysaccharide and the second synthetic polymer as described above is used to form the crosslinked matrix according to the present invention. In one aspect, a composition comprising glycosaminoglycan and the second synthetic polymer as described above is used to form the crosslinked matrix according to the present invention. In one aspect, a composition comprising deacetylated glycosaminoglycan and the second synthetic polymer as described above is used to form the crosslinked matrix according to the present invention. In one aspect, a composition comprising desulfated glycosaminoglycan and the second synthetic polymer as described above is used to form the crosslinked matrix according to the present invention. The presence of protein or polysaccharide components which contain functional groups that can react with the functional groups on multiple activated synthetic polymers can result in formation of a crosslinked synthetic polymer-naturally occurring polymer matrix upon mixing and/or crosslinking of the synthetic polymer(s). In particular, when the naturally occurring polymer (protein or polysaccharide) also contains nucleophilic groups such as primary amino groups, the electrophilic groups on the second synthetic polymer will react with the primary amino groups on these components, as well as the nucleophilic groups on the first synthetic polymer, to cause these other components to become part of the polymer matrix. For example, lysine-rich proteins such as collagen may be especially reactive with electrophilic groups on synthetic polymers.
In one aspect, the naturally occurring protein is polymer may be collagen. As used herein, the term "collagen" or "collagen material" refers to all forms of collagen, including those which have been processed or otherwise modified and is intended to encompass collagen of any type, from any source, including, but not limited to, collagen extracted from tissue or produced recombinantly, collagen analogues, collagen derivatives, modified collagens, and denatured collagens, such as gelatin.
In general, collagen from any source may be included in the compositions of the invention; for example, collagen may be extracted and purified from human or other mammalian source, such as bovine or porcine corium and human placenta, or may be recombinantly or otherwise produced. The preparation of purified, substantially non-antigenic collagen in solution from bovine skin is well known in the art. U.S. Patent No. 5,428,022 discloses methods of extracting and purifying collagen from the human placenta. U.S. Patent No. 5,667,839, discloses methods of producing recombinant human collagen in the milk of transgenic animals, including transgenic cows. Collagen of any type, including, but not limited to, types I, II, III, IV, or any combination thereof, may be used in the compositions of the invention, although type I is generally preferred. Either atelopeptide or telopeptide-containing collagen may be used; however, when collagen from a xenogeneic source, such as bovine collagen, is used, atelopeptide collagen is generally preferred, because of its reduced immunogenicity compared to telopeptide-containing collagen.
Collagen that has not been previously crosslinked by methods such as heat, irradiation, or chemical crosslinking agents is preferred for use in the compositions of the invention, although previously crosslinked collagen may be used. Non-crosslinked atelopeptide fibrillar collagen is commercially available from lnamed Aesthetics (Santa Barbara, CA) at collagen concentrations of 35 mg/ml and 65 mg/ml under the trademarks ZYDERM I Collagen and ZYDERM Il Collagen, respectively. Glutaraldehyde crosslinked atelopeptide fibrillar collagen is commercially available from lnamed Corporation (Santa Barbara, CA) at a collagen concentration of 35 mg/ml under the trademark ZYPLAST Collagen.
Collagens for use in the present invention are generally in aqueous suspension at a concentration between about 20 mg/ml to about 120 mg/ml; preferably, between about 30 mg/ml to about 90 mg/ml.
Because of its tacky consistency, nonfibrillar collagen may be preferred for use in compositions that are intended for use as bioadhesives. The term "nonfibrillar collagen" refers to any modified or unmodified collagen material that is in substantially nonfibrillar form at pH 7, as indicated by optical clarity of an aqueous suspension of the collagen.
Collagen that is already in nonfibrillar form may be used in the compositions of the invention. As used herein, the term "nonfibrillar collagen" is intended to encompass collagen types that are nonfibrillar in native form, as well as collagens that have been chemically modified such that they are in nonfibrillar form at or around neutral pH. Collagen types that are nonfibrillar (or microfibrillar) in native form include types IV, Vl, and VII.
Chemically modified collagens that are in nonfibrillar form at neutral pH include succinylated collagen and methylated collagen, both of which can be prepared according to the methods described in U.S. Pat. No. 4,164,559, issued Aug. 14, 1979, to Miyata et al., which is hereby incorporated by reference in its entirety. Due to its inherent tackiness, methylated collagen is particularly preferred for use in bioadhesive compositions, as disclosed in U.S. application Ser. No. 08/476,825.
Collagens for use in the crosslinked polymer compositions of the present invention may start out in fibrillar form, then be rendered nonfibrillar by the addition of one or more fiber disassembly agent. The fiber disassembly agent must be present in an amount sufficient to render the collagen substantially nonfibrillar at pH 7, as described above. Fiber disassembly agents for use in the present invention include, without limitation, various biocompatible alcohols, amino acids (e.g., arginine), inorganic salts (e.g., sodium chloride and potassium chloride), and carbohydrates (e.g., various sugars including sucrose).
In one aspect, the polymer may be collagen or a collagen derivative, for example methylated collagen. An example of an in situ forming composition uses pentaerythritol poly(ethylene glycol)ether tetra- sulfhydryl] (4-armed thiol PEG), pentaerythritol poly(ethylene glycol)ether tetra-succinimidyl glutarate] (4-armed NHS PEG) and methylated collagen as the reactive reagents. This composition, when mixed with the appropriate buffers can produce a crosslinked hydrogel. (See, e.g., U.S. Patent Nos. 5,874,500; 6,051,648; 6,166,130; 5,565,519 and 6,312,725J.
In another aspect, the naturally occurring polymer may be a glycosaminoglycan. Glycosaminoglycans, e.g., hyaluronic acid, contain both anionic and cationic functional groups along each polymeric chain, which can form intramolecular and/or intermolecular ionic crosslinks, and are responsible for the thixotropic (or shear thinning) nature of hyaluronic acid.
In certain aspects, the glycosaminoglycan may be derivatized. For example, glycosaminoglycans can be chemically derivatized by, e.g., deacetylation, desulfation, or both in order to contain primary amino groups available for reaction with electrophilic groups on synthetic polymer molecules. Glycosaminoglycans that can be derivatized according to either or both of the aforementioned methods include the following: hyaluronic acid, chondroitin sulfate A, chondroitin sulfate B (dermatan sulfate), chondroitin sulfate C, chitin (can be derivatized to chitosan), keratan sulfate, keratosulfate, and heparin. Derivatization of glycosaminoglycans by deacetylation and/or desulfation and covalent binding of the resulting glycosaminoglycan derivatives with synthetic hydrophilic polymers is described in further detail in commonly assigned, allowed U.S. patent application Ser. No. 08/146,843, filed Nov. 3, 1993.
In general, the collagen is added to the first synthetic polymer, then the collagen and first synthetic polymer are mixed thoroughly to achieve a homogeneous composition. The second synthetic polymer is then added and mixed into the collagen/first synthetic polymer mixture, where it will covalently bind to primary amino groups or thiol groups on the first synthetic polymer and primary amino groups on the collagen, resulting in the formation of a homogeneous crosslinked network." Various deacetylated and/or desulfated glycosaminoglycan derivatives can be incorporated into the composition in a similar manner as that described above for collagen. In addition, the introduction of hydrocolloids such as carboxymethylcellulose may promote tissue adhesion and/or swellability.
Administration of the Crosslinked Synthetic Polymer
Compositions
The compositions of the present invention having two synthetic polymers may be administered before, during or after crosslinking of the first and second synthetic polymer. Certain uses, which are discussed in greater detail below, such as tissue augmentation, may require the compositions to be crosslinked before administration, whereas other applications, such as tissue adhesion, require the compositions to be administered before crosslinking has reached "equilibrium." The point at which crosslinking has reached equilibrium is defined herein as the point at which the composition no longer feels tacky or sticky to the touch.
In order to administer the composition prior to crosslinking, the first synthetic polymer and second synthetic polymer may be contained within separate barrels of a dual-compartment syringe. In this case, the two synthetic polymers do not actually mix until the point at which the two polymers are extruded from the tip of the syringe needle into the patient's tissue. This allows the vast majority of the crosslinking reaction to occur in situ, avoiding the problem of needle blockage which commonly occurs if the two synthetic polymers are mixed too early and crosslinking between the two components is already too advanced prior to delivery from the syringe needle. The use of a dual-compartment syringe, as described above, allows for the use of smaller diameter needles, which is advantageous when performing soft tissue augmentation in delicate facial tissue, such as that surrounding the eyes.
Alternatively, the first synthetic polymer and second synthetic polymer may be mixed according to the methods described above prior to delivery to the tissue site, then injected to the desired tissue site immediately (preferably, within about 60 seconds) following mixing.
In another embodiment of the invention, the first synthetic polymer and second synthetic polymer are mixed, then extruded and allowed to crosslink into a sheet or other solid form. The crosslinked solid is then dehydrated to remove substantially all unbound water. The resulting dried solid may be ground or comminuted into particulates, then suspended in a nonaqueous fluid carrier, including, without limitation, hyaluronic acid, dextran sulfate, dextran, succinylated noncrosslinked collagen, methylated noncrosslinked collagen, glycogen, glycerol, dextrose, maltose, triglycerides of fatty acids (such as corn oil, soybean oil, and sesame oil), and egg yolk phospholipid. The suspension of particulates can be injected through a small-gauge needle to a tissue site. Once inside the tissue, the crosslinked polymer particulates will rehydrate and swell in size at least five-fold. Hydrophilic Polymer + Plurality of Crosslinkable
Components
As mentioned above, the first and/or second synthetic polymers may be combined with a hydrophilic polymer, e.g., collagen or methylated collagen, to form a composition useful in the present invention. In one general embodiment, the compositions useful in the present invention include a hydrophilic polymer in combination with two or more crosslinkable components. This embodiment is described in further detail in this section.
The Hydrophilic Polymer Component:
The hydrophilic polymer component may be a synthetic or naturally occurring hydrophilic polymer. Naturally occurring hydrophilic polymers include, but are not limited to: proteins such as collagen and derivatives thereof, fibronectin, albumins, globulins, fibrinogen, and fibrin, with collagen particularly preferred; carboxylated polysaccharides such as -polymannuronic acid and polygalacturonic acid; aminated polysaccharides, particularly the glycosaminoglycans, e.g., hyaluronic acid, chitin, chondroitin sulfate A, B, or C, keratin sulfate, keratosulfate and heparin; and activated polysaccharides such as dextran and starch derivatives. Collagen (e.g., methylated collagen) and glycosaminoglycans are preferred naturally occurring hydrophilic polymers for use herein.
In general, collagen from any source may be used in the composition of the method; for example, collagen may be extracted and purified from human or other mammalian source, such as bovine or porcine corium and human placenta, or may be recombinantly or otherwise produced. The preparation of purified, substantially non-antigenic collagen in solution from bovine skin is well known in the art. See, e.g., U.S. Pat. No. 5,428,022, to Palefsky et al., which discloses methods of extracting and purifying collagen from the human placenta. See also U.S. Patent No. 5,667,839, to Berg, which discloses methods of producing recombinant human collagen in the milk of transgenic animals, including transgenic cows. Unless otherwise specified, the term "collagen" or "collagen material" as used herein refers to all forms of collagen, including those that have been processed or otherwise modified.
Collagen of any type, including, but not limited to, types I, II, III, IV, or any combination thereof, may be used in the compositions of the invention, although type I is generally preferred. Either atelopeptide or telopeptide-containing collagen may be used; however, when collagen from a source, such as bovine collagen, is used, atelopeptide collagen is generally preferred, because of its reduced immunogenicity compared to telopeptide-containing collagen.
Collagen that has not been previously crosslinked by methods such as heat, irradiation, or chemical crosslinking agents is preferred for use in the compositions of the invention, although previously crosslinked collagen may be used. Non-crosslinked atelopeptide fibrillar collagen is commercially available from McGhan Medical Corporation (Santa Barbara, Calif.) at collagen concentrations of 35 mg/ml and 65 mg/ml under the trademarks ZYDERM® I Collagen and ZYDERM® Il Collagen, respectively. Glutaraldehyde-crosslinked atelopeptide fibrillar collagen is commercially available from McGhan Medical Corporation at a collagen concentration of 35 mg/ml under the trademark ZYPLAST®.
Collagens for use in the present invention are generally, although not necessarily, in aqueous suspension at a concentration between about 20 mg/ml to about 120 mg/ml, preferably between about 30 mg/ml to about 90 mg/ml.
Although intact collagen is preferred, denatured collagen, commonly known as gelatin, can also be used in the compositions of the invention. Gelatin may have the added benefit of being degradable faster than collagen.
Because of its greater surface area and greater concentration of reactive groups, nonfibrillar collagen is generally preferred. The term "nonfibrillar collagen" refers to any modified or unmodified collagen material that is in substantially nonfibrillar form at pH 7, as indicated by optical clarity of an aqueous suspension of the collagen.
Collagen that is already in nonfibrillar form may be used in the compositions of the invention. As used herein, the term "nonfibrillar collagen" is intended to encompass collagen types that are nonfibrillar in native form, as well as collagens that have been chemically modified such that they are in nonfibrillar form at or around neutral pH. Collagen types that are nonfibrillar (or microfibrillar) in native form include types IV, Vl, and VII.
Chemically modified collagens that are in nonfibrillar form at neutral pH include succinylated collagen, propylated collagen, ethylated collagen, methylated collagen, and the like, both of which can be prepared according to the methods described in U.S. Pat. No. 4,164,559, to Miyata et al., which is hereby incorporated by reference in its entirety. Due to its inherent tackiness, methylated collagen is particularly preferred, as disclosed in U.S. Patent No. 5,614,587 to Rhee et al. Gollagens-for use in the crosslinkable compositions of the present invention may start out in fibrillar form, then be rendered nonfibrillar by the addition of one or more fiber disassembly agents. The fiber disassembly agent must be present in an amount sufficient to render the collagen substantially nonfibrillar at pH 7, as described above. Fiber disassembly agents for use in the present invention include, without limitation, various biocompatible alcohols, amino acids, inorganic salts, and carbohydrates, with biocompatible alcohols being particularly preferred. Preferred biocompatible alcohols include glycerol and propylene glycol. Non-biocompatible alcohols, such as ethanol, methanol, and isopropanol, are not preferred for use in the present invention, due to their potentially deleterious effects on the body of the patient receiving them. Preferred amino acids include arginine. Preferred inorganic salts include sodium chloride and potassium chloride. Although carbohydrates, such as various sugars including sucrose, may be used in the practice of the present invention, they are not as preferred as other types of fiber disassembly agents because they can have cytotoxic effects in vivo.
As fibrillar collagen has less surface area and a lower concentration of reactive groups than nonfibrillar, fibrillar collagen is less preferred. However, as disclosed in U.S. Patent 5,614,587, fibrillar collagen, or mixtures of nonfibrillar and fibrillar collagen, may be preferred for use in compositions intended for long-term persistence in vivo, if optical clarity is not a requirement.
Synthetic hydrophilic polymers may also be used in the present invention. Useful synthetic hydrophilic polymers include, but are not limited to: polyalkylene oxides, particularly polyethylene glycol and poly(ethylene oxide)-poly(propylene oxide) copolymers, including block and random copolymers; polyols such as glycerol, polyglycerol (particularly highly branched polyglycerol), propylene glycol and trimethylene glycol substituted with one or more polyalkylene oxides, e.g., mono-, di- and tri- pplyoxyethylated glycerol,, mono- and di-polyoxyethylated propylene glycol, and mono- and di-polyoxyethylated trimethylene glycol; polyoxyethylated sorbitol, polyoxyethylated glucose; acrylic acid polymers and analogs and copolymers thereof, such as polyacrylic acid perse, polymethacrylic acid, poly(hydroxyethyl-methacrylate), poly(hydroxyethylacrylate), poly(methylalkylsulfoxide methacrylate), poly(methylalkylsulfoxide acrylate) and copolymers of any of the foregoing, and/or with additional acrylate species such as aminoethyl acrylate and mono-2-(acryloxy)-ethyl succinate; polymaleic acid; poly(acrylamides) such as polyacrylamide perse, poly(methacrylamide), poly(dimethylacrylamide), and poly(N-isopropyl- acrylamide); poly(olefinic alcohol)s such as polyvinyl alcohol); poly(N-vinyl lactams) such as polyvinyl pyrrolidone), poly(N-vinyl caprolactam), and copolymers thereof; polyoxazolines, including poly(methyloxazoline) and poly(ethyloxazoline); and polyvinylamines. It must be emphasized that the aforementioned list of polymers is not exhaustive, and a variety of other synthetic hydrophilic polymers may be used, as will be appreciated by those skilled in the art.
The Crosslinkable Components:
The compositions of the invention also comprise a plurality of crosslinkable components. Each of the crosslinkable components participates in a reaction that results in a crosslinked matrix. Prior to completion of the crosslinking reaction, the crosslinkable components provide the necessary adhesive qualities that enable the methods of the invention.
The crosslinkable components are selected so that crosslinking gives rise to a biocompatible, nonimmunogenic matrix useful in a variety of contexts including adhesion prevention, biologically active agent delivery, tissue augmentation, and other applications. The crosslinkable components of the invention comprise: a component A, which has m nucleophilic groups, wherein m > 2 and a component B, which has n electrophilic groups capable of reaction with the m nucleophilic groups, wherein n > 2 and m + n > 4. An optional third component, optional component C, which has at least one functional group that is either electrophilic and capable of reaction with the nucleophilic groups of component A, or nucleophilic and capable of reaction with the electrophilic groups of component B may also be present. Thus, the total number of functional groups present on components A, B and C, when present, in combination is > 5; that is, the total functional groups given by m + n + p must be > 5, where p is the number of functional groups on component C and, as indicated, is > 1. Each of the components is biocompatible and nonimmunogenic, and at least one component is comprised of a hydrophilic polymer. Also, as will be appreciated, the composition may contain additional crosslinkable components D, E, F, etc., having one or more reactive nucleophilic or electrophilic groups and thereby participate in formation of the crosslinked biomaterial via covalent bonding to other components.
The m nucleophilic groups on component A may all be the same, or, alternatively, A may contain two or more different nucleophilic groups. Similarly, the n electrophilic groups on component B may all be the same, or two or more different electrophilic groups may be present. The functional group(s) on optional component C, if nucleophilic, may or may not be the same as the nucleophilic groups on component A, and, conversely, if electrophilic, the functional group(s) on optional component C may or may not be the same as the electrophilic groups on component B.
Accordingly, the components may be represented by the structural formulae
(I) R 1HQ1 ]q-X)m (component A),
(H) R 2HQ2 IrY)n (component B), and
(III) R 3HQ3 VFn)p (optional component C), wherein:
R1, R2 and R3 are independently selected from the group consisting of C2 to C14 hydrocarbyl, heteroatom-containing C2 to Ci4 hydrocarbyl, hydrophilic polymers, and hydrophobic polymers, providing that at least one of R1, R2 and R3 is a hydrophilic polymer, preferably a synthetic hydrophilic polymer;
X represents one of the m nucleophilic groups of component A, and the various X moieties on A may be the same or different;
Y represents one of the n electrophilic groups of component B, and the various Y moieties on A may be the same or different;
Fn represents a functional group on optional component C;
Q1, Q2 and Q3 are linking groups; m ≥ 2, n ≥ 2, m + n is > 4, q, and r are independently zero or 1 , and when optional component C is present, p ≥ 1 , and s is independently zero or 1. Reactive Groups:
X may be virtually any nucleophilic group, so long as reaction can occur with the electrophilic group Y. Analogously, Y may be virtually any electrophilic group, so long as reaction can take place with X. The only limitation is a practical one, in that reaction between X and Y should be fairly rapid and take place automatically upon admixture with an aqueous medium, without need for heat or potentially toxic or non-biodegradable reaction catalysts or other chemical reagents. It is also preferred although not essential that reaction occur without need for ultraviolet or other radiation. Ideally, the reactions between X and Y should be complete in under 60 minutes, preferably under 30 minutes. Most preferably, the reaction occurs in about 5 to 15 minutes or less.
Examples of nucleophilic groups suitable as X include, but are not limited to, -NH2, -NHR4, -N(R4)2, -SH, -OH, -COOH, -C6H4-OH, -PH2, - PHR5, -P(R5)2, -NH-NH2, -CO-NH-NH2, -C5H4N, etc. wherein R4 and R5 are hydrocarbyl, typically alkyl or monocyclic aryl, preferably alkyl, and most preferably lower alkyl. Organometallic moieties are also useful nucleophilic groups for the purposes of the invention, particularly those that act as carbanion donors. Organometallic nucleophiles are not, however, preferred. Examples of organometallic moieties include: Grignard functionalities - R6MgHaI wherein R6 is a carbon atom (substituted or unsubstituted), and Hal is halo, typically bromo, iodo or chloro, preferably bromo; and lithium- containing functionalities, typically alkyllithium groups; sodium-containing functionalities.
It will be appreciated by those of ordinary skill in the art that certain nucleophilic groups must be activated with a base so as to be capable of reaction with an electrophile. For example, when there are nucleophilic sulfhydryl and hydroxyl groups in the crosslinkable composition, the composition must be admixed with an aqueous base in order to remove a proton and provide an -S" or -O" species to enable reaction with an electrophile. Unless it is desirable for the base to participate in the crosslinking reaction, a nonnucleophilic base is preferred. In some embodiments, the base may be present as a component of a buffer solution. Suitable bases and corresponding crosslinking reactions are described infra in Section E.
The selection of electrophilic groups provided within the crosslinkable composition, i.e., on component B, must be made so that reaction is possible with the specific nucleophilic groups. Thus, when the X moieties are amino groups, the Y groups are selected so as to react with amino groups. Analogously, when the X moieties are sulfhydryl moieties, the corresponding electrophilic groups are sulfhydryl-reactive groups, and the like.
By way of example, when X is amino (generally although not necessarily primary amino), the electrophilic groups present on Y are amino reactive groups such as, but not limited to: (1) carboxylic acid esters, including cyclic esters and "activated" esters; (2) acid chloride groups (-CO- Cl); (3) anhydrides (-(CO)-O-(CO)-R); (4) ketones and aldehydes, including α,β-unsaturated aldehydes and ketones such as -CH=CH-CH=O and - CH=CH-C(CH3)=O; (5) halides; (6) isocyanate (-N=C=O); (7) isothiocyanate (-N=C=S); (8) epoxides; (9) activated hydroxyl groups (e.g., activated with conventional activating agents such as carbonyldiimidazole or sulfonyl chloride); and (10) olefins, including conjugated olefins, such as ethenesulfonyl (-SO2CH=CH2) and analogous functional groups, including acrylate (-CO2-C=CH2), methacrylate (-CO2-C(CH3)=CH2)), ethyl acrylate (- CO2-C(CH2CHs)=CH2), and ethyleneimino (-CH=CH-C=NH). Since a carboxylic acid group perse is not susceptible to reaction with a nucleophilic amine, components containing carboxylic acid groups must be activated so as to be amine-reactive. Activation may be accomplished in a variety of ways, but often involves reaction with a suitable hydroxyl-containing compound in the presence of a dehydrating agent such as dicyclohexylcarbodiimide (DCC) or dicyclohexylurea (DHU). For example, a carboxylic acid can be reacted with an alkoxy-substituted N-hydroxy- succinimide or N-hydroxysulfosuccinimide in the presence of DCC to form reactive electrophilic groups, the N-hydroxysuccinimide ester and the N- hydroxysulfosuccinimide ester, respectively. Carboxylic acids may also be activated by reaction with an acyl halide such as an acyl chloride (e.g., acetyl chloride), to provide a reactive anhydride group. In a further example, a carboxylic acid may be converted to an acid chloride group using, e.g., thionyl chloride or an acyl chloride capable of an exchange reaction. Specific reagents and procedures used to carry out such activation reactions will be known to those of ordinary skill in the art and are described in the pertinent texts and literature.
Analogously, when X is sulfhydryl, the electrophilic groups present on Y are groups that react with a sulfhydryl moiety. Such reactive groups include those that form thioester linkages upon reaction with a sulfhydryl group, such as those described in PCT Publication No. WO 00/62827 to Wallace et al. As explained in detail therein, such "sulfhydryl reactive" groups include, but are not limited to: mixed anhydrides; ester derivatives of phosphorus; ester derivatives of p-nitrophenol, p- nitrothiophenol and pentafluorophenol; esters of substituted hydroxylamines, including N-hydroxyphthalimide esters, N-hydroxysuccinimide esters, N- hydroxysulfosuccinimide esters, and N-hydroxyglutarimide esters; esters of 1 -hydroxybenzotriazole; 3-hydroxy-3,4-dihydro-benzotriazin-4-one; 3- hydroxy-3,4-dihydro-quinazoline-4-one; carbonylimidazole derivatives; acid chlorides; ketenes; and isocyanates. With these sulfhydryl reactive groups, auxiliary reagents can also be used to facilitate bond formation, e.g., 1-ethyl- 3-[3-dimethylaminopropyl]carbodiimide can be used to facilitate coupling of sulfhydryl groups to carboxyl-containing groups.
In addition to the sulfhydryl reactive groups that form thioester linkages, various other sulfhydryl reactive functionalities can be utilized that form other types of linkages. For example, compounds that contain methyl imidate derivatives form imido-thioester linkages with sulfhydryl groups. Alternatively, sulfhydryl reactive groups can be employed that form disulfide bonds with sulfhydryl groups; such groups generally have the structure -S- S-Ar where Ar is a substituted or unsubstituted nitrogen-containing heteroaromatic moiety or a non-heterocyclic aromatic group substituted with an electron-withdrawing moiety, such that Ar may be, for example, 4- pyridinyl, o-nitrophenyl, m-nitrophenyl, p-nitrophenyl, 2,4-dinitrophenyl, 2- nitro-4-benzoic acid, 2-nitro-4-pyridinyl, etc. In such instances, auxiliary reagents, i.e., mild oxidizing agents such as hydrogen peroxide, can be used to facilitate disulfide bond formation.
Yet another class of sulfhydryl reactive groups forms thioether bonds with sulfhydryl groups. Such groups include, inter alia, maleimido, substituted maleimido, haloalkyl, epoxy, imino, and aziridino, as well as olefins (including conjugated olefins) such as ethenesulfonyl, etheneimino, acrylate, methacrylate, and α,β-unsaturated aldehydes and ketones. This class of sulfhydryl reactive groups are particularly preferred as the thioether bonds may provide faster crosslinking and longer in vivo stability.
When X is -OH, the electrophilic functional groups on the remaining component(s) must react with hydroxy! groups. The hydroxyl group may be activated as described above with respect to carboxylic acid groups, or it may react directly in the presence of base with a sufficiently reactive electrophile such as an epoxide group, an aziridine group, an acyl halide, or an anhydride.
When X is an organometallic nucleophile such as a Grignard functionality or an alkyllithium group, suitable electrophilic functional groups for reaction therewith are those containing carbonyl groups, including, by way of example, ketones and aldehydes.
It will also be appreciated that certain functional groups can react as nucleophiles or as electrophiles, depending on the selected reaction partner and/or the reaction conditions. For example, a carboxylic acid group can act as a nucleophile in the presence of a fairly strong base, but generally acts as an electrophile allowing nucleophilic attack at the carbonyl carbon and concomitant replacement of the hydroxyl group with the incoming nucleophile.
The covalent linkages in the crosslinked structure that result upon covalent binding of specific nucleophilic components to specific electrophilic components in the crosslinkable composition include, solely by way of example, the following (the optional linking groups Q1 and Q2 are omitted for clarity):
TABLE
Figure imgf000161_0001
Figure imgf000162_0001
Linking Groups:
The functional groups X and Y and FN on optional component C may be directly attached to the compound core (R1, R2 or R3 on optional component C, respectively), or they may be indirectly attached through a linking group, with longer linking groups also termed "chain extenders." In structural formulae (I), (II) and (III), the optional linking groups are represented by Q1, Q2 and Q3, wherein the linking groups are present when q, r and s are equal to 1 (with R, X, Y, Fn, m n and p as defined previously).
Suitable linking groups are well known in the art. See, for example, International Patent Publication No. WO 97/22371. Linking groups are useful to avoid steric hindrance problems that are sometimes associated with the formation of direct linkages between molecules. Linking groups may additionally be used to link several multifunctionally activated compounds together to make larger molecules. In a preferred embodiment, a linking group can be used to alter the degradative properties of the compositions after administration and resultant gel formation. For example, linking groups can be incorporated into components A, B, or optional component C to promote hydrolysis, to discourage hydrolysis, or to provide a site for enzymatic degradation.
Examples of linking groups that provide hydrolyzable sites, include, inter alia: ester linkages; anhydride linkages, such as obtained by incorporation of glutarate and succinate; ortho ester linkages; ortho carbonate linkages such as trimethylene carbonate; amide linkages; phosphoester linkages; α-hydroxy acid linkages, such as may be obtained by incorporation of lactic acid and glycolic acid; lactone-based linkages, such as may be obtained by incorporation of caprolactone, valerolactone, y- butyrolactone and p-dioxanone; and amide linkages such as in a dimeric, oligomeric, or poly(amino acid) segment. Examples of non-degradable linking groups include succinimide, propionic acid and carboxymethylate linkages. See, for example, PCT WO 99/07417. Examples of enzymatically degradable linkages include Leu-Gly-Pro-Ala, which is degraded by collagenase; and Gly-Pro-Lys, which is degraded by plasmin.
Linking groups can also enhance or suppress the reactivity of the various nucleophilic and electrophilic groups. For example, electron- withdrawing groups within one or two carbons of a sulfhydryl group would be expected to diminish its effectiveness in coupling, due to a lowering of nucleophilicity. Carbon-carbon double bonds and carbonyl groups will also have such an effect. Conversely, electron-withdrawing groups adjacent to a carbonyl group (e.g., the reactive carbonyl of glutaryl-N- hydroxysuccinimidyl) would increase the reactivity of the carbonyl carbon with respect to an incoming nucleophile. By contrast, sterically bulky groups in the vicinity of a functional group can be used to diminish reactivity and thus coupling rate as a result of steric hindrance.
By way of example, particular linking groups and corresponding component structure are indicated in the following Table:
TABLE
Figure imgf000164_0001
Figure imgf000165_0001
In the above Table, n is generally in the range of 1 to about 10, R7 is generally hydrocarbyl, typically alkyl or aryl, preferably alkyl, and most preferably lower alkyl, and R8 is hydrocarbylene, heteroatom-containing hydrocarbylene, substituted hydrocarbylene, or substituted heteroatom- containing hydrocarbylene) typically alkylene or arylene (again, optionally substituted and/or containing a heteroatom), preferably lower alkylene (e.g., methylene, ethylene, n-propylene, n-butylene, etc.), phenylene, or amidoalkylene (e.g., -(CO)-NH-CH2).
Other general principles that should be considered with respect to linking groups are as follows: If higher molecular weight components are to be used, they preferably have biodegradable linkages as described above, so that fragments larger than 20,000 mol. wt. are not generated during resorption in the body. In addition, to promote water miscibility and/or solubility, it may be desired to add sufficient electric charge or hydrophilicity. Hydrophilic groups can be easily introduced using known chemical synthesis, so long as they do not give rise to unwanted swelling or an undesirable decrease in compressive strength. In particular, polyalkoxy segments may weaken gel strength. The Component Core:
The "core" of each crosslinkable component is comprised of the molecular structure to which the nucleophilic or electrophilic groups are bound. Using the formulae (I) R1-[Q1]q-X)m, for component A, (II) R2(-[Q2]r Y)n for component B, and (III)
R3(-[Q3]S-Fn)p for optional component C, the "core" groups are R1, R2 and R3. Each molecular core of the reactive components of the crosslinkable composition is generally selected from synthetic and naturally occurring hydrophilic polymers, hydrophobic polymers, and C2-C14 hydrocarbyl groups zero to 2 heteroatoms selected from N, O and S, with the proviso that at least one of the crosslinkable components A, B, and optionally C, comprises a molecular core of a synthetic hydrophilic polymer. In a preferred embodiment, at least one of A and B comprises a molecular core of a synthetic hydrophilic polymer.
Hydrophilic Crosslinkable Components - In one aspect, the crosslinkable component(s) is (are) hydrophilic polymers. The term "hydrophilic polymer" as used herein refers to a synthetic polymer having an average molecular weight and composition effective to render the polymer "hydrophilic" as defined above. As discussed above, synthetic crosslinkable hydrophilic polymers useful herein include, but are not limited to: polyalkylene oxides, particularly polyethylene glycol and poly(ethylene oxide)-poly(propylene oxide) copolymers, including block and random copolymers; polyols such as glycerol, polyglycerol (particularly highly branched polyglycerol), propylene glycol and trimethylene glycol substituted with one or more polyalkylene oxides, e.g., mono-, di- and tri-polyoxyethylated glycerol, mono- and di-polyoxyethylated propylene glycol, and mono- and di-polyoxyethylated trimethylene glycol; polyoxyethylated sorbitol, polyoxyethylated glucose; acrylic acid polymers and analogs and copolymers thereof, such as polyacrylic acid perse, polymethacrylic acid, poly(hydroxyethyl-methacrylate), poly(hydroxyethylacrylate), poly(methylalkylsulfoxide methacrylate), poly(methylalkylsulfoxide acrylate) and copolymers of any of the foregoing, and/or with additional acrylate species such as aminoethyl acrylate and mono-2-(acryloxy)-ethyl succinate; polymaleic acid; poly(acrylamides) such as polyacrylamide perse, poly(methacrylamide), poly(dimethylacrylamide), and poly(N-isopropyl-acrylamide); poly(olefinic alcohol)s such as polyvinyl alcohol); poly(N-vinyl lactams) such as polyvinyl pyrrolidone), poly(N-vinyl caprolactam), and copolymers thereof; polyoxazolines, including poly(methyloxazoline) and poly(ethyloxazoline); and polyvinylamines. It must be emphasized that the aforementioned list of polymers is not exhaustive, and a variety of other synthetic hydrophilic polymers may be used, as will be appreciated by those skilled in the art.
The synthetic crosslinkable hydrophilic polymer may be a homopolymer, a block copolymer, a random copolymer, or a graft copolymer. In addition, the polymer may be linear or branched, and if branched, may be minimally to highly branched, dendrimeric, hyperbranched, or a star polymer. The polymer may include biodegradable segments and blocks, either distributed throughout the polymer's molecular structure or present as a single block, as in a block copolymer. Biodegradable segments are those that degrade so as to break covalent bonds. Typically, biodegradable segments are segments that are hydrolyzed in the presence of water and/or enzymatically cleaved in situ. Biodegradable segments may be composed of small molecular segments such as ester linkages, anhydride linkages, ortho ester linkages, ortho carbonate linkages, amide linkages, phosphonate linkages, etc. Larger biodegradable "blocks" will generally be composed of oligomeric or polymeric segments incorporated within the hydrophilic polymer. Illustrative oligomeric and polymeric segments that are biodegradable include, by way of example, poly(amino acid) segments, poly(orthoester) segments, poly(orthocarbonate) segments, and the like. Other suitable synthetic crosslinkable hydrophilic polymers include chemically synthesized polypeptides, particularly polynucleophilic polypeptides that have been synthesized to incorporate amino acids containing primary amino groups (such as lysine) and/or amino acids containing thiol groups (such as cysteine). Poly(lysine), a synthetically produced polymer of the amino acid lysine (145 MW), is particularly preferred. Poly(lysine)s have been prepared having anywhere from 6 to about 4,000 primary amino groups, corresponding to molecular weights of about 870 to about 580,000. Poly(lysine)s for use in the present invention preferably have a molecular weight within the range of about 1 ,000 to about 300,000, more preferably within the range of about 5,000 to about 100,000, and most preferably, within the range of about 8,000 to about 15,000. Poly(lysine)s of varying molecular weights are commercially available from Peninsula Laboratories, Inc. (Belmont, Calif.).
The synthetic crosslinkable hydrophilic polymer may be a homopolymer, a block copolymer, a random copolymer, or a graft copolymer. In addition, the polymer may be linear or branched, and if branched, may be minimally to highly branched, dendrimeric, hyperbranched, or a star polymer. The polymer may include biodegradable segments and blocks, either distributed throughout the polymer's molecular structure or present as a single block, as in a block copolymer. Biodegradable segments are those that degrade so as to break covalent bonds. Typically, biodegradable segments are segments that are hydrolyzed in the presence of water and/or enzymatically cleaved in situ. Biodegradable segments may be composed of small molecular segments such as ester linkages, anhydride linkages, ortho ester linkages, ortho carbonate linkages, amide linkages, phosphonate linkages, etc. Larger biodegradable "blocks" will generally be composed of oligomeric or polymeric segments incorporated within the hydrophilic polymer. Illustrative oligomeric and polymeric segments that are biodegradable include, by way of example, poly(amino acid) segments, poly(orthoester) segments, poly(orthocarbonate) segments, and the like.
Although a variety of different synthetic crosslinkable hydrophilic polymers can be used in the present compositions, as indicated above, preferred synthetic crosslinkable hydrophilic polymers are polyethylene glycol (PEG) and polyglycerol (PG), particularly highly branched polyglycerol. Various forms of PEG are extensively used in the modification of biologically active molecules because PEG lacks toxicity, antigenicity, and immunogenicity (i.e., is biocompatible), can be formulated so as to have a wide range of solubilities, and do not typically interfere with the enzymatic activities and/or conformations of peptides. A particularly preferred synthetic crosslinkable hydrophilic polymer for certain applications is a polyethylene glycol (PEG) having a molecular weight within the range of about 100 to about 100,000 mol. wt., although for highly branched PEG, far higher molecular weight polymers can be employed — up to 1 ,000,000 or more - providing that biodegradable sites are incorporated ensuring that all degradation products will have a molecular weight of less than about 30,000. For most PEGs, however, the preferred molecular weight is about 1,000 to about 20,000 mol. wt., more preferably within the range of about 7,500 to about 20,000 mol. wt. Most preferably, the polyethylene glycol has a molecular weight of approximately 10,000 mol. wt.
Naturally occurring crosslinkable hydrophilic polymers include, but are not limited to: proteins such as collagen, fibronectin, albumins, globulins, fibrinogen, and fibrin, with collagen particularly preferred; carboxylated polysaccharides such as polymannuronic acid and polygalacturonic acid; aminated polysaccharides, particularly the glycosaminoglycans, e.g., hyaluronic acid, chitin, chondroitin sulfate A, B, or C, keratin sulfate, keratosulfate and heparin; and activated polysaccharides such as dextran and starch derivatives. Collagen and glycosaminoglycans are examples of naturally occurring hydrophilic polymers for use herein, with methylated collagen being a preferred hydrophilic polymer. Any of the hydrophilic polymers herein must contain, or be activated to contain, functional groups, i.e., nucleophilic or electrophilic groups, which enable crosslinking. Activation of PEG is discussed below; it is to be understood, however, that the following discussion is for purposes of illustration and analogous techniques may be employed with other polymers.
With respect to PEG, first of all, various functionalized polyethylene glycols have been used effectively in fields such as protein modification (see Abuchowski et al., Enzymes as Drugs, John Wiley & Sons: New York, N.Y. (1981) pp. 367-383; and Dreborg et al., Crit. Rev. Therap. Drug Carrier Syst. (1990) 6:315), peptide chemistry (see Mutter et al., The Peptides, Academic: New York, N.Y. 2:285-332; and Zalipsky et al., Int. J. Peptide Protein Res. (1987) 30:740), and the synthesis of polymeric drugs (see Zalipsky et al., Eur. Polym. J. (1983) 19:1177; and Ouchi et al., J. Macromol. Sci. Chem. (1987) A24:1011).
Activated forms of PEG, including multifunctionally activated PEG, are commercially available, and are also easily prepared using known methods. For example, see Chapter 22 of Poly(ethylene Glycol) Chemistry: Biotechnical and Biomedical Applications, J. Milton Harris, ed., Plenum Press, NY (1992); and Shearwater Polymers, Inc. Catalog, Polyethylene Glycol Derivatives, Huntsville, Alabama (1997-1998).
Structures for some specific, tetrafunctionally activated forms of PEG are shown in FIGS. 1 to 10 of U.S. Patent 5,874,500, as are generalized reaction products obtained by reacting the activated PEGs with multi-amino PEGs, i.e., a PEG with two or more primary amino groups. The activated PEGs illustrated have a pentaerythritol (2,2-bis(hydroxymethyl)- 1 ,3-propanediol) core. Such activated PEGs, as will be appreciated by those in the art, are readily prepared by conversion of the exposed hydroxyl groups in the PEGylated polyol (i.e., the terminal hydroxyl groups on the PEG chains) to carboxylic acid groups (typically by reaction with an anhydride in the presence of a nitrogenous base), followed by esterification with N-hydroxysuccinimide, N-hydroxysulfosuccinimide, or the like, to give the polyfunctionally activated PEG.
Hydrophobic Polymers:
The crosslinkable compositions of the invention can also include hydrophobic polymers, although for most uses hydrophilic polymers are preferred. Polylactic acid and polyglycolic acid are examples of two hydrophobic polymers that can be used. With other hydrophobic polymers, only short-chain oligomers should be used, containing at most about 14 carbon atoms, to avoid solubility-related problems during reaction.
Low Molecular Weight Components:
As indicated above, the molecular core of one or more of the crosslinkable components can also be a low molecular weight compound, Ae., a C2-C14 hydrocarbyl group containing zero to 2 heteroatoms selected from N, O, S and combinations thereof. Such a molecular core can be substituted with nucleophilic groups or with electrophilic groups.
When the low molecular weight molecular core is substituted with primary amino groups, the component may be, for example, ethylenediamine (H2N-CH2CH2-NH2), tetramethylenediamine (H2N-(CH4)- NH2), pentamethylenediamine (cadaverine) (H2N-(CH5)-NH2), hexamethylenediamine (H2N-(CH6)-NH2), bis(2-aminoethyl)amine (HN- [CH2CH2-NH2I2), or tris(2-aminoethyl)amine (N-[CH2CH2-NH2J3).
Low molecular weight diols and polyols include trimethylolpropane, di(trimethylol propane), pentaerythritol, and diglycerol, all of which require activation with a base in order to facilitate their reaction as nucleophiles. Such diols and polyols may also be functionalized to provide di- and poly-carboxylic acids, functional groups that are, as noted earlier herein, also useful as nucleophiles under certain conditions. Polyacids for use in the present compositions include, without limitation, trimethylolpropane-based tricarboxylic acid, di(trimethylol propane)-based tetracarboxylic acid, heptanedioic acid, octanedioic acid (suberic acid), and hexadecanedioic acid (thapsic acid), all of which are commercially available and/or readily synthesized using known techniques.
Low molecular weight di- and poly-electrophiles include, for example, disuccinimidyl suberate (DSS), bis(sulfosuccinimidyl) suberate (BS3), dithiobis(succinimidylpropionate) (DSP), bis(2- succinimidooxycarbonyloxy) ethyl sulfone (BSOCOES), and 3,3'- dithiobis(sulfosuccinimidylpropionate (DTSPP), and their analogs and derivatives. The aforementioned compounds are commercially available from Pierce (Rockford, III.). Such di- and poly-electrophiles can also be synthesized from di- and polyacids, for example by reaction with an appropriate molar amount of N-hydroxysuccinimide in the presence of DCC. Polyols such as trimethylolpropane and di(trimethylol propane) can be converted to carboxylic acid form using various known techniques, then further derivatized by reaction with NHS in the presence of DCC to produce trifunctionaϊly and tetrafunctionally activated polymers.
Delivery Systems:
Suitable delivery systems for the homogeneous dry powder composition (containing at least two crosslinkable polymers) and the two buffer solutions may involve a multi-compartment spray device, where one or more compartments contains the powder and one or more compartments contain the buffer solutions needed to provide for the aqueous environment, so that the composition is exposed to the aqueous environment as it leaves the compartment. Many devices that are adapted for delivery of multi- component tissue sealants/hemostatic agents are well known in the art and can also be used in the practice of the present invention. Alternatively, the composition can be delivered using any type of controllable extrusion system, or it can be delivered manually in the form of a dry powder, and exposed to the aqueous environment at the site of administration. The homogeneous dry powder composition and the two buffer solutions may be conveniently formed under aseptic conditions by placing each of the three ingredients (dry powder, acidic buffer solution and basic buffer solution) into separate syringe barrels. For example, the composition, first buffer solution and second buffer solution can be housed separately in a multiple-compartment syringe system having a multiple barrels, a mixing head, and an exit orifice. The first buffer solution can be added to the barrel housing the composition to dissolve the composition and form a homogeneous solution, which is then extruded into the mixing head. The second buffer solution can be simultaneously extruded into the mixing head. Finally, the resulting composition can then be extruded through the orifice onto a surface.
For example, the syringe barrels holding the dry powder and the basic buffer may be part of a dual-syringe system, e.g., a double barrel syringe as described in U.S. Patent 4,359,049 to Redl et al. In this embodiment, the acid buffer can be added to the syringe barrel that also holds the dry powder, so as to produce the homogeneous solution. In other words, the acid buffer may be added (e.g., injected) into the syringe barrel holding the dry powder to thereby produce a homogeneous solution of the first and second components. This homogeneous solution can then be extruded into a mixing head, while the basic buffer is simultaneously extruded into the mixing head. Within the mixing head, the homogeneous solution and the basic buffer are mixed together to thereby form a reactive mixture. Thereafter, the reactive mixture is extruded through an orifice and onto a surface (e.g., tissue), where a film is formed, which can function as a sealant or a barrier, or the like. The reactive mixture begins forming a three- dimensional matrix immediately upon being formed by the mixing of the homogeneous solution and the basic buffer in the mixing head. Accordingly, the reactive mixture is preferably extruded from the mixing head onto the tissue very quickly after it is formed so that the three-dimensional matrix forms on, and is able to adhere to, the tissue. Other systems for combining two reactive liquids are well known in the art, and include the systems described in U.S. Patent Nos. 6,454,786 to Holm et al.; 6,461 ,325 to Delmotte et al.; 5,585,007 to Antanavich et al.; 5,116,315 to Capozzi et al.; and 4,631,055 to Redl et al.
Storage and Handling:
Because crosslinkable components containing electrophilic groups react with water, the electrophilic component or components are generally stored and used in sterile, dry form to prevent hydrolysis. Processes for preparing synthetic hydrophilic polymers containing multiple electrophilic groups in sterile, dry form are set forth in commonly assigned U.S. Patent No. 5,643,464 to Rhee et al. For example, the dry synthetic polymer may be compression molded into a thin sheet or membrane, which can then be sterilized using gamma or, preferably, e-beam irradiation. The resulting dry membrane or sheet can be cut to the desired size or chopped into smaller size particulates.
Components containing multiple nucleophilic groups are generally not water-reactive and can therefore be stored either dry or in aqueous solution. If stored as a dry, particulate, solid, the various components of the crosslinkable composition may be blended and stored in a single container. Admixture of all components with water, saline, or other aqueous media should not occur until immediately prior to use.
In an alternative embodiment, the crosslinking components can be mixed together in a single aqueous medium in which they are both unreactive, i.e., such as in a low pH buffer. Thereafter, they can be sprayed onto the targeted tissue site along with a high pH buffer, after which they will rapidly react and form a gel.
Suitable liquid media for storage of crosslinkable compositions include aqueous buffer solutions such as monobasic sodium phosphate/dibasic sodium phosphate, sodium carbonate/sodium bicarbonate, glutamate or acetate, at a concentration of 0.5 to 300 mM. In general, a sulfhydryl-reactive component such as PEG substituted with maleimido groups or succinimidyl esters is prepared in water or a dilute buffer, with a pH of between around 5 to 6. Buffers with pKs between about 8 and 10.5 for preparing a polysulfhydryl component such as sulfhydryl-PEG are useful to achieve fast gelation time of compositions containing mixtures of sulfhydryl-PEG and SG-PEG. These include carbonate, borate and AMPSO (3-[(1 ,1-dimethyl-2-hydroxyethyl)amino]2-hydroxy-propane-sulfonic acid). In contrast, using a combination of maleimidyl PEG and sulfhydryl- PEG, a pH of around 5 to 9 is preferred for the liquid medium used to prepare the sulfhydryl PEG.
Collagen + Fibrinogen and/or Thrombin (e.g., Costasis) In yet another aspect, the polymer composition may include collagen in combination with fibrinogen and/or thrombin. (See, e.g., U.S. Patent Nos. 5,290,552; 6,096,309; and 5,997,811). For example, an aqueous composition may include a fibrinogen and FXIII, particularly plasma, collagen in an amount sufficient to thicken the composition, thrombin in an amount sufficient to catalyze polymerization of fibrinogen present in the composition, and Ca2+ and, optionally, an antifibrinolytic agent in amount sufficient to retard degradation of the resulting adhesive clot. The composition may be formulated as a two-part composition that may be mixed together just prior to use, in which fibrinogen/FXIII and collagen constitute the first component, and thrombin together with an antifibrinolytic agent, and Ca2+ constitute the second component.
Plasma, which provides a source of fibrinogen, may be obtained from the patient for which the composition is to be delivered. The plasma can be used "as is" after standard preparation which includes centrifuging out cellular components of blood. Alternatively, the plasma can be further processed to concentrate the fibrinogen to prepare a plasma cryoprecipitate. The plasma cryoprecipitate can be prepared by freezing the plasma for at least about an hour at about -20 0C, and then storing the frozen plasma overnight at about 4 0C. to slowly thaw. The thawed plasma is centrifuged and the plasma cryoprecipitate is harvested by removing approximately four-fifths of the plasma to provide a cryoprecipitate comprising the remaining one-fifth of the plasma. Other fibrinogen/FXIII preparations may be used, such as cryoprecipitate, patient autologous fibrin sealant, fibrinogen analogs or other single donor or commercial fibrin sealant materials. Approximately 0.5 ml to about 1.0 ml of either the plasma or the plasma-cryoprecipitate provides about 1 to 2 ml of adhesive composition which is sufficient for use in middle ear surgery. Other plasma proteins (e.g., albumin, plasminogen, von Willebrands factor, Factor VIII, etc.) may or may not be present in the fibrinogen/FXII separation due to wide variations in the formulations and methods to derive them.
Collagen, preferably hypoallergenic collagen, is present in the composition in an amount sufficient to thicken the composition and augment the cohesive properties of the preparation. The collagen may be atelbpeptide collagen or telopeptide collagen, e.g., native collagen. In addition to thickening the composition, the collagen augments the fibrin by acting as a macromolecular lattice work or scaffold to which the fibrin network adsorbs. This gives more strength and durability to the resulting glue clot with a relatively low concentration of fibrinogen in comparison to the various concentrated autogenous fibrinogen glue formulations (i.e., AFGs).
The form of collagen which is employed may be described as at least "near native" in its structural characteristics. It may be further characterized as resulting in insoluble fibers at a pH above 5; unless crosslinked or as part of a complex composition, e.g., bone, it will generally consist of a minor amount by weight of fibers with diameters greater than 50 nm, usually from about 1 to 25 volume % and there will be substantially little, if any, change in the helical structure of the fibrils. In addition, the collagen composition must be able to enhance gelation in the surgical adhesion composition. A number of commercially available collagen preparations may be used. ZYDERM Collagen Implant (ZCI) has a fibrillar diameter distribution consisting of 5 to 10 nm diameter fibers at 90% volume content and the remaining 10% with greater than about 50 nm diameter fibers. ZCI is available as a fibrillar slurry and solution in phosphate buffered isotonic saline, pH 7.2, and is injectable with fine gauge needles. As distinct from ZCI1 cross-linked collagen available as ZYPLAST may be employed. ZYPL-AST is essentially an exogenously crosslinked (glutaraldehyde) version of ZCI. The material has a somewhat higher content of greater than about 50 nm diameter fibrils and remains insoluble over a wide pH range. Crosslinking has the effect of mimicking in vivo endogenous crosslinking found in many tissues.
Thrombin acts as a catalyst for fibrinogen to provide fibrin, an insoluble polymer and is present in the composition in an amount sufficient to catalyze polymerization of fibrinogen present in the patient plasma. Thrombin also activates FXIII, a plasma protein that catalyzes covalent crosslinks in fibrin, rendering the resultant clot insoluble. Usually the thrombin is present in the adhesive composition in concentration of from about 0.01 to about 1000 or greater NIH units (NlHu) of activity, usually about i to about 500 NIHu, most usually about 200 to about 500 NIHu. The thrombin can be from a variety of host animal sources, conveniently bovine. Thrombin is commercially available from a variety of sources including Parke-Davis, usually lyophilized with buffer salts and stabilizers in vials which provide thrombin activity ranging from about 1000 NIHu to 10,000 NIHu. The thrombin is usually prepared by reconstituting the powder by the addition of either sterile distilled water or isotonic saline. Alternately, thrombin analogs or reptile-sourced coagulants may be used.
The composition may additionally comprise an effective amount of an antifibrinolytic agent to enhance the integrity of the glue clot as the healing processes occur. A number of antifibrinolytic agents are well known and include aprotinin, C1 -esterase inhibitor and ε-amino-n-caproic acid (EACA). ε-amino-n-caproic acid, the only antifibrinolytic agent approved by the FDA, is effective at a concentration of from about 5 mg/ml to about 40 mg/ml of the final adhesive composition, more usually from about 20 to about 30 mg/ml. EACA is commercially available as a solution having a concentration of about 250 mg/ml. Conveniently, the commercial solution is diluted with distilled water to provide a solution of the desired concentration. That solution is desirably used to reconstitute lyophilized thrombin to the desired thrombin concentration.
Other examples of in situ forming materials based on the crosslinking of proteins are described, e.g., in U.S. Patent Nos. RE38158; 4,839,345; 5,514,379, 5,583,114; 6,458,147; 6,371 ,975; 5,290,552; 6,096,309; U.S. Patent Application Publication Nos. 2002/0161399; 2001/0018598 and PCT Publication Nos. WO 03/090683; WO 01/45761 ; WO 99/66964 and WO 96/03159).
Self-Reactive Compounds
In one aspect, the therapeutic agent is released from a crosslinked matrix formed, at least in part, from a self-reactive compound. As used herein, a self-reactive compound comprises a core substituted with a minimum of three reactive groups. The reactive groups may be directed attached to the core of the compound, or the reactive groups may be indirectly attached to the compound's core, e.g., the reactive groups are joined to the core through one or more linking groups.
Each of the three reactive groups that are necessarily present in a self-reactive compound can undergo a bond-forming reaction with at least one of the remaining two reactive groups. For clarity it is mentioned that when these compounds react to form a crosslinked matrix, it will most often happen that reactive groups on one compound will reactive with reactive groups on another compound. That is, the term "self-reactive" is not intended to mean that each self-reactive compound necessarily reacts with itself, but rather that when a plurality of identical self-reactive compounds are in combination and undergo a crosslinking reaction, then these compounds will react with one another to form the matrix. The compounds are "self-reactive" in the sense that they can react with other compounds having the identical chemical structure as themselves.
The self-reactive compound comprises at least four components: a core and three reactive groups. In one embodiment, the self-reactive compound can be characterized by the formula (I), where R is the core, the reactive groups are represented by X1, X2 and X3, and a linker (L) is optionally present between the core and a functional group.
Figure imgf000179_0001
The core R is a polyvalent moiety having attachment to at least three groups (i.e., it is at least trivalent) and may be, or may contain, for example, a hydrophilic polymer, a hydrophobic polymer, an amphiphilic polymer, a C2-I4 hydrocarbyl, or a C2-14 hydrocarbyl which is heteroatom- containing. The linking groups L1, L2, and L3 may be the same or different. The designators p, q and r are either 0 (when no linker is present) or 1 (when a linker is present). The reactive groups X1, X2 and X3 may be the same or different. Each of these reactive groups reacts with at least one other reactive group to form a three-dimensional matrix. Therefore X1 can react with X2 and/or X3, X2 can react with X1 and/or X3, X3 can react with X1 and/or X2 and so forth. A trivalent core will be directly or indirectly bonded to three functional groups, a tetravalent core will be directly or indirectly bonded to four functional groups, etc.
Each side chain typically has one reactive group. However, the invention also encompasses self-reactive compounds where the side chains contain more than one reactive group. Thus, in another embodiment of the invention, the self-reactive compound has the formula (II): [ X' - (L4)a - Y' - (L5)b ] c R1 where: a and b are integers from 0-1; c is an integer from 3-12; R1 is selected from hydrophilic polymers, hydrophobic polymers, amphiphilic polymers, C2-i4 hydrocarbyls, and heteroatom-containing C2-M hydrocarbyls; X' and Y' are reactive groups and can be the same or different; and L4 and L5 are linking groups. Each reactive group inter-reacts with the other reactive group to form a three-dimensional matrix. The compound is essentially non-reactive in an initial environment but is rendered reactive upon exposure to a modification in the initial environment that provides a modified environment such that a plurality of the self-reactive compounds inter-react in the modified environment to form a three-dimensional matrix. In one preferred embodiment, R is a hydrophilic polymer. In another preferred embodiment, X' is a nucleophilic group and Y' is an electrophilic group.
Jne L following self-reactive compound is one example of a compound of formula (II):
Figure imgf000180_0001
where R4 has the formula:
Figure imgf000180_0002
Thus, in formula (II), a and b are 1; c is 4; the core R' is the hydrophilic polymer, tetrafunctionally activated polyethylene glycol, (C(CH2- O-)4; X1 is the electrophilic reactive group, succinimidyl; Y' is the nucleophilic reactive group -CH-NH2; L4 is -C(O)-O-; and L5 is -(CH2- C H2-O-C H2)x-C H2-
0-C(O)-(CHz)2-.
The self-reactive compounds of the invention are readily synthesized by techniques that are well known in the art. An exemplary synthesis is set forth below:
Figure imgf000181_0001
Mitsunobo or DCC
Figure imgf000181_0002
H2, Pd/C
Figure imgf000182_0001
The reactive groups are selected so that the compound is essentially non-reactive in an initial environment. Upon exposure to a specific modification in the initial environment, providing a modified environment, the compound is rendered reactive and a plurality of self- reactive compounds are then able to inter-react in the modified environment to form a three-dimensional matrix. Examples of modification in the initial environment are detailed below, but include the addition of an aqueous medium, a change in pH, exposure to ultraviolet radiation, a change in temperature, or contact with a redox initiator. The core and reactive groups can also be selected so as to provide a compound that has one of more of the following features: are biocompatible, are non-immunogenic, and do not leave any toxic, inflammatory or immunogenic reaction products at the site of administration. Similarly, the core and reactive groups can also be selected so as to provide a resulting matrix that has one or more of these features.
In one embodiment of the invention, substantially immediately or immediately upon exposure to the modified environment, the self-reactive compounds inter-react form a three-dimensional matrix. The term "substantially immediately" is intended to mean within less than five minutes, preferably within less than two minutes, and the term "immediately" is intended to mean within less than one minute, preferably within less than 30 seconds.
In one embodiment, the self-reactive compound and resulting matrix are not subject to enzymatic cleavage by matrix metalloproteinases such as collagenase, and are therefore not readily degradable in vivo. Further, the self-reactive compound may be readily tailored, in terms of the selection and quantity of each component, to enhance certain properties, e.g., compression strength, swellability, tack, hydrophilicity, optical clarity, and the like.
In one preferred embodiment, R is a hydrophilic polymer. In another preferred embodiment, X is a nucleophilic group, Y is an electrophilic group and Z is either an electrophilic or a nucleophilic group. Additional embodiments are detailed below.
A higher degree of inter-reaction, e.g., crosslinking, may be useful when a less swellable matrix is desired or increased compressive strength is desired. In those embodiments, it may be desirable to have n be an integer from 2-12. In addition, when a plurality of self-reactive compounds are utilized, the compounds may be the same or different. a. Reactive Groups
Prior to use, the self-reactive compound is stored in an initial environment that insures that the compound remain essentially non-reactive until use. Upon modification of this environment, the compound is rendered reactive and a plurality of compounds will then inter-react to form the desired matrix. The initial environment, as well as the modified environment, is thus determined by the nature of the reactive groups involved.
The number of reactive groups can be the same or different. However, in one embodiment of the invention, the number of reactive groups are approximately equal. As used in this context, the term "approximately" refers to a 2:1 to 1 :2 ratio of moles of one reactive group to moles of a different reactive groups. A 1 :1 :1 molar ratio of reactive groups is generally preferred.
In general, the concentration of the self-reactive compounds in the modified environment, when liquid in nature, will be in the range of about 1 to 50 wt%, generally about 2 to 40 wt%. The preferred concentration of the compound in the liquid will depend on a number of factors, including the type of compound (Ae., type of molecular core and reactive groups), its molecular weight, and the end use of the resulting three-dimensional matrix. For example, use of higher concentrations of the compounds, or using highly functionalized compounds, will result in the formation of a more tightly crosslinked network, producing a stiffer, more robust gel. As such, compositions intended for use in tissue augmentation will generally employ concentrations of self-reactive compounds that fall toward the higher end of the preferred concentration range. Compositions intended for use as bioadhesives or in adhesion prevention do not need to be as firm and may therefore contain lower concentrations of the self-reactive compounds. i) Electrophilic and Nucleophilic Reactive Groups In one embodiment of the invention, the reactive groups are electrophilic and nucleophilic groups, which undergo a nucleophilic substitution reaction, a nucleophilic addition reaction, or both. The term "electrophilic" refers to a reactive group that is susceptible to nucleophilic attack, i.e., susceptible to reaction with an incoming nucieophilic group. Electrophilic groups herein are positively charged or electron-deficient, typically electron-deficient. The term "nucleophilic" refers to a reactive group that is electron rich, has an unshared pair of electrons acting as a reactive site, and reacts with a positively charged or electron-deficient site. For such reactive groups, the modification in the initial environment comprises the addition of an aqueous medium and/or a change in pH.
In one embodiment of the invention, X1 (also referred to herein as X) can be a nucleophilic group and X2 (also referred to herein as Y) can be an electrophilic group or vice versa, and X3 (also referred to herein as Z) can be either an electrophilic or a nucleophilic group.
X may be virtually any nucleophilic group, so long as reaction can occur with the electrophilic group Y and also with Z, when Z is electrophilic (ZEL). Analogously, Y may be virtually any electrophilic group, so long as reaction can take place with X and also with Z when Z is nucleophilic (ZNU)- The only limitation is a practical one, in that reaction between X and Y, and X and ZEL, or Y and ZNu should be fairly rapid and take place automatically upon admixture with an aqueous medium, without need for heat or potentially toxic or non-biodegradable reaction catalysts or other chemical reagents. It is also preferred although not essential that reaction occur without need for ultraviolet or other radiation. In one embodiment, the reactions between X and Y, and between either X and ZEL or Y and ZNU, are complete in under 60 minutes, preferably under 30 minutes. Most preferably, the reaction occurs in about 5 to 15 minutes or less. Examples of nucleophilic groups suitable as X or FΓINU include, but are not limited to: -NH2, -NHR1, -N(R1)2, -SH, -OH, -COOH, -C6H4-OH, -H, -PH2,
-PHR1, -P(R1)2, -NH-NH2, -CO-NH-NH2, -C5H4N, etc. wherein R1 is a hydrocarbyl group and each R1 may be the same or different. R1 is typically alkyl or monocyclic aryl, preferably alkyl, and most preferably lower alkyl. Organometallic moieties are also useful nucleophilic groups for the purposes of the invention, particularly those that act as carbanion donors. Examples of organometallic moieties include: Grignard functionalities - R2MgHaI wherein R2 is a carbon atom (substituted or unsubstituted), and Hal is halo, typically bromo, iodo or chloro, preferably bromo; and lithium- containing functionalities, typically alkyllithium groups; sodium-containing functionalities.
It will be appreciated by those of ordinary skill in the art that certain nucleophilic groups must be activated with a base so as to be capable of reaction with an electrophilic group. For example, when there are nucleophilic sulfhydryl and hydroxyl groups in the self-reactive compound, the compound must be admixed with an aqueous base in order to remove a proton and provide an -S" or -O" species to enable reaction with the electrophilic group. Unless it is desirable for the base to participate in the reaction, a non-nucleophilic base is preferred. In some embodiments, the base may be present as a component of a buffer solution. Suitable bases and corresponding crosslinking reactions are described herein.
The selection of electrophilic groups provided on the self- reactive compound, must be made so that reaction is possible with the specific nucleophilic groups. Thus, when the X reactive groups are amino groups, the Y and any ZEL groups are selected so as to react with amino groups. Analogously, when the X reactive groups are sulfhydryl moieties, the corresponding electrophilic groups are sulfhydryl-reactive groups, and the like. In general, examples of electrophilic groups suitable as Y or ZEL include, but are not limited to, -CO-CI, -(CO)-O-(CO)-R (where R is an alkyl group), -CH=CH-CH=O and -CH=CH-C(CH3)=O, halo, -N=C=O, -N=C=S, -SO2CH=CH2, -0(CO)-C=CH2, -O(CO)-C(CH3)=CH2, -S-S-(C5H4N), -0(CO)-C(CH2CHs)=CH2, -CH=CH-C=NH, -COOH, -(CO)O-N(COCH2)2, -CHO, -(CO)O-N(COCH2)2-S(O)2OH, and -N(COCH)2.
When X is amino (generally although not necessarily primary amino), the electrophilic groups present on Y and ZEL are amine-reactive groups. Exemplary amine-reactive groups include, by way of example and not limitation, the following groups, or radicals thereof: (1) carboxylic acid esters, including cyclic esters and "activated" esters; (2) acid chloride groups (-CO-CI); (3) anhydrides (-(CO)-O-(CO)-R, where R is an alkyl group); (4) ketones and aldehydes, including α,β-unsaturated aldehydes and ketones such as -CH=CH-CH=O and -CH=CH-C(CH3)=O; (5) halo groups; (6) isocyanate group (-N=C=O); (7) thioisocyanato group (-N=C=S); (8) epoxides; (9) activated hydroxyl groups (e.g., activated with conventional activating agents such as carbonyldiimidazole or sulfonyl chloride); and (10) olefins, including conjugated olefins, such as ethenesulfonyl (-SO2CH=CH2) and analogous functional groups, including acrylate (-0(CO)-C=CH2), methacrylate (-O(CO)-C(CH3)=CH2), ethyl acrylate (-O(CO)-C(CH2CH3)=CH2), and ethyleneimino (-CH=CH-C=NH).
In one embodiment the amine-reactive groups contain an electrophilically reactive carbonyl group susceptible to nucleophilic attack by a primary or secondary amine, for example the carboxylic acid esters and aldehydes noted above, as well as carboxyl groups (-COOH).
Since a carboxylic acid group per se is not susceptible to reaction with a nucleophilic amine, components containing carboxylic acid groups must be activated so as to be amine-reactive. Activation may be accomplished in a variety of ways, but often involves reaction with a suitable hydroxyl-containing compound in the presence of a dehydrating agent such as dicyclohexylcarbodiimide (DCC) or dicyclohexylurea (DHU). For example, a carboxylic acid can be reacted with an alkoxy-substituted N- hydroxy-succinimide or N-hydroxysulfosuccinimide in the presence of DCC to form reactive electrophilic groups, the N-hydroxysuccinimide ester and the N-hydroxysulfosuccinimide ester, respectively. Carboxylic acids may also be activated by reaction with an acyl halide such as an acyl chloride (e.g., acetyl chloride), to provide a reactive anhydride group. In a further example, a carboxylic acid may be converted to an acid chloride group using, e.g., thionyl chloride or an acyl chloride capable of an exchange reaction. Specific reagents and procedures used to carry out such activation reactions will be known to those of ordinary skill in the art and are described in the pertinent texts and literature.
Accordingly, in one embodiment, the amine-reactive groups are selected from succinimidyl ester (-O(CO)-N(COCH2)2), sulfosuccinimidyl ester (-O(CO)-N(COCH2)2-S(O)2OH), maleimido (-N(COCH)2), epoxy, isocyanato, thioisocyanato, and ethenesulfonyl.
Analogously, when X is sulfhydryl, the electrophilic groups present on Y and ZEL are groups that react with a sulfhydryl moiety. Such reactive groups include those that form thioester linkages upon reaction with a sulfhydryl group, such as those described in WO 00/62827 to Wallace et al. As explained in detail therein, sulfhydryl reactive groups include, but are not limited to: mixed anhydrides; ester derivatives of phosphorus; ester derivatives of p-nitrophenol, p-nitrothiophenol and pentafluorophenol; esters of substituted hydroxylamines, including N-hydroxyphthalimide esters, N- hydroxysuccinimide esters, N-hydroxysulfosuccinimide esters, and N- hydroxyglutarimide esters; esters of 1-hydroxybenzotriazole; 3-hydroxy-3,4- dihydro-benzotriazin-4-one; 3-hydroxy-3,4-dihydro-quinazoline-4-one; carbonylimidazole derivatives; acid chlorides; ketenes; and isocyanates. With these sulfhydryl reactive groups, auxiliary reagents can also be used to facilitate bond formation, e.g., 1-ethyl-3-[3- dimethylaminopropyljcarbodiimide can be used to facilitate coupling of sulfhydryl groups to carboxyl-containing groups.
In addition to the sulfhydryl reactive groups that form thioester linkages, various other sulfhydryl reactive functionalities can be utilized that form other types of linkages. For example, compounds that contain methyl imidate derivatives form imido-thioester linkages with sulfhydryl groups. Alternatively, sulfhydryl reactive groups can be employed that form disulfide bonds with sulfhydryl groups; such groups generally have the structure -S- S-Ar where Ar is a substituted or unsubstituted nitrogen-containing heteroaromatic moiety or a non-heterocyclic aromatic group substituted with an electron-withdrawing moiety, such that Ar may be, for example, 4- pyridinyl, o-nitrophenyl, m-nitrophenyl, p-nitrophenyl, 2,4-dinitrophenyl, 2- nitro-4-benzoic acid, 2-nitro-4-pyridinyl, etc. In such instances, auxiliary reagents, Ae., mild oxidizing agents such as hydrogen peroxide, can be used to facilitate disulfide bond formation.
Yet another class of sulfhydryl reactive groups forms thioether bonds with sulfhydryl groups. Such groups include, inter alia, maleimido, substituted maleimido, haloalkyl, epoxy, imino, and aziridino, as well as olefins (including conjugated olefins) such as ethenesulfonyl, etheneimino, acrylate, methacrylate, and α,β-unsaturated aldehydes and ketones.
When X is -OH, the electrophilic functional groups on the remaining component(s) must react with hydroxyl groups. The hydroxyl group may be activated as described above with respect to carboxylic acid groups, or it may react directly in the presence of base with a sufficiently reactive electrophilic group such as an epoxide group, an aziridine group, an acyl halide, an anhydride, and so forth.
When X is an organometallic nucleophilic group such as a Grignard functionality or an alkyllithium group, suitable electrophilic functional groups for reaction therewith are those containing carbonyl groups, including, by way of example, ketones and aldehydes.
It will also be appreciated that certain functional groups can react as nucleophilic or as electrophilic groups, depending on the selected reaction partner and/or the reaction conditions. For example, a carboxylic acid group can act as a nucleophilic group in the presence of a fairly strong base, but generally acts as an electrophilic group allowing nucleophilic attack at the carbonyl carbon and concomitant replacement of the hydroxyl group with the incoming nucleophilic group.
These, as well as other embodiments are illustrated below, where the covalent linkages in the matrix that result upon covalent binding of specific nucleophilic reactive groups to specific electrophilic reactive groups on the self-reactive compound include, solely by way of example, the following Table:
Table
Figure imgf000190_0001
Figure imgf000191_0001
For self-reactive compounds containing electrophilic and nucleophilic reactive groups, the initial environment typically can be dry and sterile. Since electrophilic groups react with water, storage in sterile, dry form will prevent hydrolysis. The dry synthetic polymer may be compression molded into a thin sheet or membrane, which can then be sterilized using gamma or e-beam irradiation. The resulting dry membrane or sheet can be cut to the desired size or chopped into smaller size particulates. The modification of a dry initial environment will typically comprise the addition of an aqueous medium.
In one embodiment, the initial environment can be an aqueous medium such as in a low pH buffer, i.e., having a pH less than about 6.0, in which both electrophilic and nucleophilic groups are non-reactive. Suitable liquid media for storage of such compounds include aqueous buffer solutions such as monobasic sodium phosphate/dibasic sodium phosphate, sodium carbonate/sodium bicarbonate, glutamate or acetate, at a concentration of 0.5 to 300 mM. Modification of an initial low pH aqueous environment will typically comprise increasing the pH to at least pH 7.0, more preferably increasing the pH to at least pH 9.5. In another embodiment the modification of a dry initial environment comprises dissolving the self-reactive compound in a first buffer solution having a pH within the range of about 1.0 to 5.5 to form a homogeneous solution, and (ii) adding a second buffer solution having a pH within the range of about 6.0 to 11.0 to the homogeneous solution. The buffer solutions are aqueous and can be any pharmaceutically acceptable basic or acid composition. The term "buffer" is used in a general sense to refer to an acidic or basic aqueous solution, where the solution may or may not be functioning to provide a buffering effect (i.e., resistance to change in pH upon addition of acid or base) in the compositions of the present invention. For example, the self-reactive compound can be in the form of a homogeneous dry powder. This powder is then combined with a buffer solution having a pH within the range of about 1.0 to 5.5 to form a homogeneous acidic aqueous solution, and this solution is then combined with a buffer solution having a pH within the range of about 6.0 to 11.0 to form a reactive solution. For example, 0.375 grams of the dry powder can be combined with 0.75 grams of the acid buffer to provide, after mixing, a homogeneous solution, where this solution is combined with 1.1 grams of the basic buffer to provide a reactive mixture that substantially immediately forms a three-dimensional matrix.
Acidic buffer solutions having a pH within the range of about 1.0 to 5.5, include by way of illustration and not limitation, solutions of: citric acid, hydrochloric acid, phosphoric acid, sulfuric acid, AMPSO (3-[(1,1- dimethyl-2-hydroxyethyl)amino]2-hydroxy-propane-sulfonic acid), acetic acid, lactic acid, and combinations thereof. In a preferred embodiment, the acidic buffer solution, is a solution of citric acid, hydrochloric acid, phosphoric acid, sulfuric acid, and combinations thereof. Regardless of the precise acidifying agent, the acidic buffer preferably has a pH such that it retards the reactivity of the nucleophilic groups on the core. For example, a pH of 2.1 is generally sufficient to retard the nucleophilicity of thiol groups. A lower pH is typically preferred when the core contains amine groups as the nucleophilic groups. In general, the acidic buffer is an acidic solution that, when contacted with nucleophilic groups, renders those nucleophilic groups relatively non-nucleophilic.
An exemplary acidic buffer is a solution of hydrochloric acid, having a concentration of about 6.3 mM and a pH in the range of 2.1 to 2.3. This buffer may be prepared by combining concentrated hydrochloric acid with water, i.e., by diluting concentrated hydrochloric acid with water. Similarly, this buffer A may also be conveniently prepared by diluting 1.23 grams of concentrated hydrochloric acid to a volume of 2 liters, or diluting 1.84 grams of concentrated hydrochloric acid to a volume to 3 liters, or diluting 2.45 grams of concentrated hydrochloric acid to a volume of 4 liters, or diluting 3.07 grams concentrated hydrochloric acid to a volume of 5 liters, or diluting 3.68 grams of concentrated hydrochloric acid to a volume to 6 liters. For safety reasons, the concentrated acid is preferably added to water.
Basic buffer solutions having a pH within the range of about 6.0 to 11.0, include by way of illustration and not limitation, solutions of: glutamate, acetate, carbonate and carbonate salts (e.g., sodium carbonate, sodium carbonate monohydrate and sodium bicarbonate), borate, phosphate and phosphate salts (e.g., monobasic sodium phosphate monohydrate and dibasic sodium phosphate), and combinations thereof. In a preferred embodiment, the basic buffer solution is a solution of carbonate salts, phosphate salts, and combinations thereof.
In general, the basic buffer is an aqueous solution that neutralizes the effect of the acidic buffer, when it is added to the homogeneous solution of the compound and first buffer, so that the nucleophilic groups on the core regain their nucleophilic character (that has been masked by the action of the acidic buffer), thus allowing the nucleophilic groups to inter-react with the electrophilic groups on the core.
An exemplary basic buffer is an aqueous solution of carbonate and phosphate salts. This buffer may be prepared by combining a base solution with a salt solution. The salt solution may be prepared by combining 34.7 g of monobasic sodium phosphate monohydrate, 49.3 g of sodium carbonate monohydrate, and sufficient water to provide a solution volume of 2 liter. Similarly, a 6 liter solution may be prepared by combining 104.0 g of monobasic sodium phosphate monohydrate, 147.94 g of sodium carbonate monohydrate, and sufficient water to provide 6 liter of the salt solution. The basic buffer may be prepared by combining 7.2 g of sodium hydroxide with 180.0 g of water. The basic buffer is typically prepared by adding the base solution as needed to the salt solution, ultimately to provide a mixture having the desired pH, e.g., a pH of 9.65 to 9.75.
In general, the basic species present in the basic buffer should be sufficiently basic to neutralize the acidity provided by the acidic buffer, but should not be so nucleophilic itself that it will react substantially with the electrophilic groups on the core. For this reason, relatively "soft" bases such as carbonate and phosphate are preferred in this embodiment of the invention.
To illustrate the preparation of a three-dimensional matrix of the present invention, one may combine an admixture of the self-reactive compound with a first, acidic, buffer (e.g., an acid solution, e.g., a dilute hydrochloric acid solution) to form a homogeneous solution. This homogeneous solution is mixed with a second, basic, buffer (e.g., a basic solution, e.g., an aqueous solution containing phosphate and carbonate salts) whereupon the reactive groups on the core of the self-reactive compound substantially immediately inter-react with one another to form a three-dimensional matrix.
//) Redox Reactive Groups
In one embodiment of the invention, the reactive groups are vinyl groups such as styrene derivatives, which undergo a radical polymerization upon initiation with a redox initiator. The term "redox" refers to a reactive group that is susceptible to oxidation-reduction activation. The term "vinyl" refers to a reactive group that is activated by a redox initiator, and forms a radical upon reaction. X, Y and Z can be the same or different vinyl groups, for example, methacrylic groups.
For self-reactive compounds containing vinyl reactive groups, the initial environment typically will be an aqueous environment. The modification of the initial environment involves the addition of a redox initiator.
Hi) Oxidative Coupling Reactive Groups In one embodiment of the invention, the reactive groups undergo an oxidative coupling reaction. For example, X, Y and Z can be a halo group such as chloro, with an adjacent electron-withdrawing group on the halogen-bearing carbon (e.g., on the "L" linking group). Exemplary electron-withdrawing groups include nitro, aryl, and so forth.
For such reactive groups, the modification in the initial environment comprises a change in pH. For example, in the presence of a base such as KOH, the self-reactive compounds then undergo a de-hydro, chloro coupling reaction, forming a double bond between the carbon atoms, as illustrated below:
Cl
I H I
Ar-C R C-Ar I
A1 I ^OH Ar-C R C-Ar
Cl C| KOH c, T,
+ ci c"Ar
C-Ar
I Ar-C R CH-Ar
Ar-C R C-Ar Cl Cl
Cl Cl
For self-reactive compounds containing oxidative coupling reactive groups, the initial environment typically can be can be dry and sterile, or a non-basic medium. The modification of the initial environment will typically comprise the addition of a base. /V) Photoinitiated Reactive Groups In one embodiment of the invention, the reactive groups are photoinitiated groups. For such reactive groups, the modification in the initial environment comprises exposure to ultraviolet radiation.
In one embodiment of the invention, X can be an azide (-N3) group and Y can be an alkyl group such as -CH(CH3)2 or vice versa. Exposure to ultraviolet radiation will then form a bond between the groups to provide for the following linkage: -NH-C(CH3)2-CH2-. In another embodiment of the invention, X can be a benzophenone (-(C6H4)-C(O)- (CeH5)) group and Y can be an alkyl group such as -CH(CH3)2 or vice versa. Exposure to ultraviolet radiation will then form a bond between the groups to provide for the following linkage:
Figure imgf000196_0001
For self-reactive compounds containing photoinitiated reactive groups, the initial environment typically will be in an ultraviolet radiation- shielded environment. This can be for example, storage within a container that is impermeable to ultraviolet radiation.
The modification of the initial environment will typically comprise exposure to ultraviolet radiation.
v) Temperature-sensitive Reactive Groups In one embodiment of the invention, the reactive groups are temperature-sensitive groups, which undergo a thermochemical reaction. For such reactive groups, the modification in the initial environment thus comprises a change in temperature. The term "temperature-sensitive" refers to a reactive group that is chemically inert at one temperature or temperature range and reactive at a different temperature or temperature range. In one embodiment of the invention, X, Y, and Z are the same or different vinyl groups.
For self-reactive compounds containing reactive groups that are temperature-sensitive, the initial environment typically will be within the range of about 10 to 3O0C.
The modification of the initial environment will typically comprise changing the temperature to within the range of about 20 to 4O0C.
b. Linking Groups
The reactive groups may be directly attached to the core, or they may be indirectly attached through a linking group, with longer linking groups also termed "chain extenders." In the formula (I) shown above, the optional linker groups are represented by L1, L2, and L3, wherein the linking groups are present when p, q and r are equal to 1.
Suitable linking groups are well known in the art. See, for example, WO 97/22371 to'Rhee et"al. Linking groups are useful to avoid steric hindrance problems that can sometimes associated with the formation of direct linkages between molecules. Linking groups may additionally be used to link several self-reactive compounds together to make larger molecules. In one embodiment, a linking group can be used to alter the degradative properties of the compositions after administration and resultant gel formation. For example, linking groups can be used to promote hydrolysis, to discourage hydrolysis, or to provide a site for enzymatic degradation.
Examples of linking groups that provide hydrolyzable sites, include, inter alia: ester linkages; anhydride linkages, such as those obtained by incorporation of glutarate and succinate; ortho ester linkages; ortho carbonate linkages such as trimethylene carbonate; amide linkages; phosphoester linkages; α-hydroxy acid linkages, such as those obtained by incorporation of lactic acid and glycolic acid; lactone-based linkages, such as those obtained by incorporation of caprolactone, valerolactone, y- butyrolactone and p-dioxanone; and amide linkages such as in a dimeric, oligomeric, or poly(amino acid) segment. Examples of non-degradable linking groups include succinimide, propionic acid and carboxymethylate linkages. See, for example, WO 99/07417 to Coury et al. Examples of enzymatically degradable linkages include Leu-Gly-Pro-Ala, which is degraded by collagenase; and Gly-Pro-Lys, which is degraded by plasmin.
Linking groups can also be included to enhance or suppress the reactivity of the various reactive groups. For example, electron- withdrawing groups within one or two carbons of a sulfhydryl group would be expected to diminish its effectiveness in coupling, due to a lowering of nucleophilicity. Carbon-carbon double bonds and carbonyl groups will also have such an effect. Conversely, electron-withdrawing groups adjacent to a carbonyl group (e.g., the reactive carbonyl of glutaryl-N- hydroxysuccinimidyl) would increase the reactivity of the carbonyl carbon with respect to an incoming nucleophilic group. By contrast, sterically bulky groups in the vicinity of a reactive group can be used to diminish reactivity and thus reduce the coupling rate as a result of steric hindrance.
By way of example, particular linking groups and corresponding formulas are indicated in the following Table:
Table
Figure imgf000198_0001
Figure imgf000199_0001
In the above Table, x is generally in the range of 1 to about 10; R2 is generally hydrocarbyl, typically alkyl or aryl, preferably alkyl, and most preferably lower alkyl; and R3 is hydrocarbylene, heteroatom-containing hydrocarbylene, substituted hydrocarbylene, or substituted heteroatom- containing hydrocarbylene) typically alkylene or arylene (again, optionally substituted and/or containing a heteroatom), preferably lower alkylene (e.g., methylene, ethylene, n-propylene, n-butylene, etc.), phenylene, or amidoalkylene (e.g., -(CO)-NH-CH2). Other general principles that should be considered with respect to linking groups are as follows. If a higher molecular weight self- reactive compound is to be used, it will preferably have biodegradable linkages as described above, so that fragments larger than 20,000 mol. wt. are not generated during resorption in the body. In addition, to promote water miscibility and/or solubility, it may be desired to add sufficient electric charge or hydrophilicity. Hydrophilic groups can be easily introduced using known chemical synthesis, so long as they do not give rise to unwanted swelling or an undesirable decrease in compressive strength. In particular, polyalkoxy segments may weaken gel strength.
c. The Core
The "core" of each self-reactive compound is comprised of the molecular structure to which the reactive groups are bound. The molecular core can a polymer, which includes synthetic polymers and naturally occurring polymers: In one embodfment, the core is a polymer containing repeating monomer units. The polymers can be hydrophilic, hydrophobic, or amphiphilic. The molecular core can also be a low molecular weight components such as a C2-U hydrocarbyl or a heteroatom-containing C2-14 hydrocarbyl. The heteroatom-containing C2-I4 hydrocarbyl can have 1 or 2 heteroatoms selected from N, O and S. In a preferred embodiment, the self- reactive compound comprises a molecular core of a synthetic hydrophilic polymer.
/) Hydrophilic Polymers
As mentioned above, the term "hydrophilic polymer" as used herein refers to a polymer having an average molecular weight and composition that naturally renders, or is selected to render the polymer as a whole "hydrophilic." Preferred polymers are highly pure or are purified to a highly pure state such that the polymer is or is treated to become pharmaceutically pure. Most hydrophilic polymers can be rendered water soluble by incorporating a sufficient number of oxygen (or less frequently nitrogen) atoms available for forming hydrogen bonds in aqueous solutions.
Synthetic hydrophilic polymers may be homopolymers, block copolymers including di-block and tri-block copolymers, random copolymers, or graft copolymers. In addition, the polymer may be linear or branched, and if branched, may be minimally to highly branched, dendrimeric, hyperbranched, or a star polymer. The polymer may include biodegradable segments and blocks, either distributed throughout the polymer's molecular structure or present as a single block, as in a block copolymer. Biodegradable segments preferably degrade so as to break covalent bonds. Typically, biodegradable segments are segments that are hydrolyzed in the presence of water and/or enzymatically cleaved in situ. Biodegradable segments may be composed of small molecular segments such as ester linkages, anhydride linkages, ortho ester linkages, ortho carbonate linkages, amide linkages, phosphonate linkages, etc. Larger biodegradable "blocks" will generally be composed of όligomeric or polymeric segments incorporated within the hydrophilic polymer. Illustrative oligomeric and polymeric segments that are biodegradable include, by way of example, poly(amino acid) segments, poly(orthoester) segments, poly(orthocarbonate) segments, and the like. Other biodegradable segments that may form part of the hydrophilic polymer core include polyesters such as polylactide, polyethers such as polyalkylene oxide, polyamides such as a protein, and polyurethanes. For example, the core of the self-reactive compound can be a diblock copolymer of tetrafunctionally activated polyethylene glycol and polylactide.
Synthetic hydrophilic polymers that are useful herein include, but are not limited to: polyalkylene oxides, particularly polyethylene glycol (PEG) and poly(ethylene oxide)-poly(propylene oxide) copolymers, including block and random copolymers; polyols such as glycerol, polyglycerol (PG) and particularly highly branched polyglycerol, propylene glycol; poly(oxyalkylene)-substituted diols, and poly(oxyalkylene)-substituted polyols such as mono-, di- and tri-polyoxyethylated glycerol, mono- and di- polyoxyethylated propylene glycol, and mono- and di-polyoxyethylated trimethylene glycol; polyoxyethylated sorbitol, polyoxyethylated glucose; poly(acrylic acids) and analogs and copolymers thereof, such as polyacrylic acid perse, polymethacrylic acid, poly(hydroxyethylmethacrylate), poly(hydroxyethylacrylate), poly(methylalkylsulfoxide methacrylates), poly(methylalkylsulfoxide acrylates) and copolymers of any of the foregoing, and/or with additional acrylate species such as aminoethyl acrylate and mono-2-(acryloxy)-ethyl succinate; polymaleic acid; poly(acrylamides) such as polyacrylamide per se, poly(methacrylamide), poly(dimethylacrylamide), poly(N-isopropyl-acrylamide), and copolymers thereof; poly(olefinic alcohols) such as polyvinyl alcohols) and copolymers thereof; poly(N-vinyl lactams) such as polyvinyl pyrrolidones), poly(N-vinyl caprolactams), and copolymers thereof; polyoxazolines, including poly(methyloxazoline) and poly(ethyloxazoline); and polyvinylamines; as well as copolymers of any of the foregoing. It must be emphasized that the aforementioned list of polymers is not exhaustive, and a variety of other synthetic hydrophilic polymers may be used, as will be appreciated by those skilled in the art.
Those of ordinary skill in the art will appreciate that synthetic polymers such as polyethylene glycol cannot be prepared practically to have exact molecular weights, and that the term "molecular weight" as used herein refers to the weight average molecular weight of a number of molecules in any given sample, as commonly used in the art. Thus, a sample of PEG 2,000 might contain a statistical mixture of polymer molecules ranging in weight from, for example, 1 ,500 to 2,500 daltons with one molecule differing slightly from the next over a range. Specification of a range of molecular weights indicates that the average molecular weight may be any value between the limits specified, and may include molecules outside those limits. Thus, a molecular weight range of about 800 to about 20,000 indicates an average molecular weight of at least about 800, ranging up to about 20 kDa. Other suitable synthetic hydrophilic polymers include chemically synthesized polypeptides, particularly polynucleophilic polypeptides that have been synthesized to incorporate amino acids containing primary amino groups (such as lysine) and/or amino acids containing thiol groups (such as cysteine). Poly(lysine), a synthetically produced polymer of the amino acid lysine (145 MW), is particularly preferred. Poly(lysine)s have been prepared having anywhere from 6 to about 4,000 primary amino groups, corresponding to molecular weights of about 870 to about 580,000. Poly(Iysine)s for use in the present invention preferably have a molecular weight within the range of about 1 ,000 to about 300,000, more preferably within the range of about 5,000 to about 100,000, and most preferably, within the range of about 8,000 to about 15,000. Poly(lysine)s of varying molecular weights are commercially available from Peninsula Laboratories, Inc. (Belmont, Calif.).
Although a variety of different synthetic hydrophilic polymers can be used in the present compounds, preferred synthetic hydrophilic polymers are PEG and PG, particularly highly branched PG. Various forms of PEG are extensively used in the modification of biologically active molecules because PEG lacks toxicity, antigenicity, and immunogenicity (Ae., is biocompatible), can be formulated so as to have a wide range of solubilities, and does not typically interfere with the enzymatic activities and/or conformations of peptides. A particularly preferred synthetic hydrophilic polymer for certain applications is a PEG having a molecular weight within the range of about 100 to about 100,000, although for highly branched PEG, far higher molecular weight polymers can be employed, up to 1 ,000,000 or more, providing that biodegradable sites are incorporated ensuring that all degradation products will have a molecular weight of less than about 30,000. For most PEGs, however, the preferred molecular weight is about 1 ,000 to about 20,000, more preferably within the range of about 7,500 to about 20,000. Most preferably, the polyethylene glycol has a molecular weight of approximately 10,000. Naturally occurring hydrophilic polymers include, but are not limited to: proteins such as collagen, fibronectin, albumins, globulins, fibrinogen, fibrin and thrombin, with collagen particularly preferred; carboxylated polysaccharides such as polymannuronic acid and polygalacturonic acid; aminated polysaccharides, particularly the glycosaminoglycans, e.g., hyaluronic acid, chitin, chondroitin sulfate A, B, or C, keratin sulfate, keratosulfate and heparin; and activated polysaccharides such as dextran and starch derivatives. Collagen and glycosaminoglycans are preferred naturally occurring hydrophilic polymers for use herein.
Unless otherwise specified, the term "collagen" as used herein refers to all forms of collagen, including those, which have been processed or otherwise modified. Thus, collagen from any source may be used in the compounds of the invention; for example, collagen may be extracted and purified from human or other mammalian source, such as bovine or porcine corium and human placenta, or may be recombinantly or otherwise produced. The preparation of purified, substantially non-antigenic collagen in solution from bovine skin is well known in the art. For example, U.S. Patent No. 5,428,022 to Palefsky et al. discloses methods of extracting and purifying collagen from the human placenta, and U.S. Patent No. 5,667,839 to Berg discloses methods of producing recombinant human collagen in the milk of transgenic animals, including transgenic cows. Non-transgenic, recombinant collagen expression in yeast and other cell lines) is described in U.S. Patent No. 6,413,742 to Olsen et al., 6,428,978 to Olsen et al., and 6,653,450 to Berg et al.
Collagen of any type, including, but not limited to, types I1 II1 III, IV, or any combination thereof, may be used in the compounds of the invention, although type I is generally preferred. Either atelopeptide or telopeptide-containing collagen may be used; however, when collagen from a natural source, such as bovine collagen, is used, atelopeptide collagen is generally preferred, because of its reduced immunogenicity compared to telopeptide-containing collagen. Collagen that has not been previously crosslinked by methods such as heat, irradiation, or chemical crosslinking agents is preferred for use in the invention, although previously crosslinked collagen may be used.
Collagens for use in the present invention are generally, although not necessarily, in aqueous suspension at a concentration between about 20 mg/ml to about 120 mg/ml, preferably between about 30 mg/ml to about 90 mg/ml. Although intact collagen is preferred, denatured collagen, commonly known as gelatin, can also be used. Gelatin may have the added benefit of being degradable faster than collagen.
Nonfibrillar collagen is generally preferred for use in compounds of the invention, although fibrillar collagens may also be used. The term "nonfibrillar collagen" refers to any modified or unmodified collagen material that is in substantially nonfibrillar form, i.e., molecular collagen that is not tightly associated with other collagen molecules so as to form fibers. Typically, a solution of nonfibrillar collagen is more transparent than is a solution of fibrillar collagen. Collagen types that are nonfibrillar (or microfibrillar) in native form include types IV, Vl, and VII.
Chemically modified collagens that are in nonfibrillar form at neutral pH include succinylated collagen and methylated collagen, both of which can be prepared according to the methods described in U.S. Patent No. 4,164,559 to Miyata et al. Methylated collagen, which contains reactive amine groups, is a preferred nucleophile-containing component in the compositions of the present invention. In another aspect, methylated collagen is a component that is present in addition to first and second components in the matrix-forming reaction of the present invention. Methylated collagen is described in, for example, in U.S. Patent No. 5,614,587 to Rhee et al.
Collagens for use in the compositions of the present invention may start out in fibrillar form, then can be rendered nonfibrillar by the addition of one or more fiber disassembly agent. The fiber disassembly agent must be present in an amount sufficient to render the collagen substantially nonfibrillar at pH 7, as described above. Fiber disassembly agents for use in the present invention include, without limitation, various biocompatible alcohols, amino acids, inorganic salts, and carbohydrates, with biocompatible alcohols being particularly preferred. Preferred biocompatible alcohols include glycerol and propylene glycol. Non- biocompatible alcohols, such as ethanol, methanol, and isopropanol, are not preferred for use in the present invention, due to their potentially deleterious effects on the body of the patient receiving them. Preferred amino acids include arginine. Preferred inorganic salts include sodium chloride and potassium chloride. Although carbohydrates, such as various sugars including sucrose, may be used in the practice of the present invention, they are not as preferred as other types of fiber disassembly agents because they can have cytotoxic effects in vivo.
Fibrillar collagen is less preferred for use in the compounds of the invention. However, as disclosed in U.S. Patent No. 5,614,587 to Rhee et'al., fibrillar collagen, or mixtures of nonfibrillar and fibrillar collagen, may be preferred for use in compounds intended for long-term persistence in vivo.
H) Hydrophobic Polymers
The core of the self-reactive compound may also comprise a hydrophobic polymer, including low molecular weight polyfunctional species, although for most uses hydrophilic polymers are preferred. Generally, "hydrophobic polymers" herein contain a relatively small proportion of oxygen and/or nitrogen atoms. Preferred hydrophobic polymers for use in the invention generally have a carbon chain that is no longer than about 14 carbons. Polymers having carbon chains substantially longer than 14 carbons generally have very poor solubility in aqueous solutions and, as such, have very long reaction times when mixed with aqueous solutions of synthetic polymers containing, for example, multiple nucleophilic groups. Thus, use of short-chain oligomers can avoid solubility-related problems during reaction. Polylactic acid and polyglycolic acid are examples of two particularly suitable hydrophobic polymers.
Hi) Amphiphilic Polymers
Generally, amphiphilic polymers have a hydrophilic portion and a hydrophobic (or lipophilic) portion. The hydrophilic portion can be at one end of the core and the hydrophobic portion at the opposite end, or the hydrophilic and hydrophobic portions may be distributed randomly (random copolymer) or in the form of sequences or grafts (block copolymer) to form the amphiphilic polymer core of the self-reactive compound. The hydrophilic and hydrophobic portions may include any of the aforementioned hydrophilic and hydrophobic polymers.
Alternately, the amphiphilic polymer core can be a hydrophilic polymer that has been modified with hydrophobic moieties (e.g., alkylated PEG or a hydrophilic polymer modified with one or more fatty chains ), or a hydrophobic polymer that has been modified with hydrophilic moieties (e.g., "PEGylated" phospholipids such as polyethylene glycolated phospholipids).
iv) Low Molecular Weight Components As indicated above, the molecular core of the self-reactive compound can also be a low molecular weight compound, defined herein as being a C2-M hydrocarbyl or a heteroatom-containing C2-U hydrocarbyl, which contains 1 to 2 heteroatoms selected from N, O, S and combinations thereof. Such a molecular core can be substituted with any of the reactive groups described herein.
Alkanes are suitable C2-U hydrocarbyl molecular cores. Exemplary alkanes, for substituted with a nucleophilic primary amino group and a Y electrophilic group, include, ethyleneamine (H2N-CH2CH2-Y), tetramethyleneamine (H2N-(C H4)-Y), pentamethyleneamine (H2N-(CH5)-Y), and hexamethyleneamine (H2N-(CHe)-Y). Low molecular weight diols and polyols are also suitable C2-I4 hydrocarbyls and include trimethylolpropane, di(trimethylol propane), pentaerythritol, and diglycerol. Polyacids are also suitable C2-I4 hydrocarbyls, and include trimethylolpropane-based tricarboxylic acid, di(trimethylol propane)-based tetracarboxylic acid, heptanedioic acid, octanedioic acid (suberic acid), and hexadecanedioic acid (thapsic acid).
Low molecular weight di- and poly-electrophiles are suitable heteroatom-containing C2-14 hydrocarbyl molecular cores. These include, for example, disuccinimidyl suberate (DSS), bis(sulfosuccinimidyl) suberate (BS3), dithiobis(succinimidylpropionate) (DSP), bis(2- succinimidooxycarbonyloxy) ethyl sulfone (BSOCOES), and 3,3'- dithiobis(sulfosuccinimidylpropionate (DTSPP), and their analogs and derivatives.
In one embodiment of the invention, the self-reactive compound of the invention comprises a low-molecular weight material core, with a plurality of acrylate moieties and a plurality of thiol groups.
d. Preparation
The self-reactive compounds are readily synthesized to contain a hydrophilic, hydrophobic or amphiphilic polymer core or a low molecular weight core, functionalized with the desired functional groups, i.e., nucleophilic and electrophilic groups, which enable crosslinking. For example, preparation of a self-reactive compound having a polyethylene glycol (PEG) core is discussed below. However, it is to be understood that the following discussion is for purposes of illustration and analogous techniques may be employed with other polymers.
With respect to PEG, first of all, various functionalized PEGs have been used effectively in fields such as protein modification (see Abuchowski et a!., Enzymes as Drugs, John Wiley & Sons: New York, N.Y. (1981) pp. 367-383; and Dreborg et al. (1990) Crit Rev. Therap. Drug Carrier Syst. 6:315), peptide chemistry (see Mutter et al., The Peptides, Academic: New York, N.Y. 2:285-332; and Zalipsky et al. (1987) Int. J. Peptide Protein Res. 30:740), and the synthesis of polymeric drugs (see Zalipsky et al. (1983) Eur. Polym. J. 19:1177; and Ouchi et al. (1987) J. Macromol. Sci. Chem. A24:1011).
Functionalized forms of PEG, including multi-functionalized PEG, are commercially available, and are also easily prepared using known methods. For example, see Chapter 22 of Poly(ethylene Glycol) Chemistry: Biotechnical and Biomedical Applications, J. Milton Harris, ed., Plenum Press, NY (1992).
Multi-functionalized forms of PEG are of particular interest and include, PEG succinimidyl glutarate, PEG succinimidyl propionate, succinimidyl butylate, PEG succinimidyl acetate, PEG succinimidyl succinamide, PEG succinimidyl carbonate, PEG propionaldehyde, PEG glycidyl ether, PEG-isocyanate, and PEG-vinylsulfone. Many such forms of PEG are described in U.S. Patent No. 5,328,955 and 6,534,591 , both to Rhee et al. Similarly, various forms of mϋlti-amino PEG are commercially available from sources such as PEG Shop, a division of SunBio of South Korea (www.sunbio.com), Nippon Oil and Fats (Yebisu Garden Place Tower, 20-3 Ebisu 4-chome, Shibuya-ku, Tokyo), Nektar Therapeutics (San Carlos, California, formerly Shearwater Polymers, Huntsville, Alabama) and from Huntsman's Performance Chemicals Group (Houston, Texas) under the name Jeffamine® polyoxyalkyleneamines. Multi-amino PEGs useful in the present invention include the Jeffamine diamines ("D" series) and triamines ("T" series), which contain two and three primary amino groups per molecule. Analogous poly(sulfhydryl) PEGs are also available from Nektar Therapeutics, e.g., in the form of pentaerythritol poly(ethylene glycol) ether tetra-sulfhydryl (molecular weight 10,000). These multi-functionalized forms of PEG can then be modified to include the other desired reactive groups.
Reaction with succinimidyl groups to convert terminal hydroxyl groups to reactive esters is one technique for preparing a core with electrophilic groups. This core can then be modified include nucleophilic groups such as primary amines, thiols, and hydroxyl groups. Other agents to convert hydroxyl groups include carbonyldiimidazole and sulfonyl chloride. However, as discussed herein, a wide variety of electrophilic groups may be advantageously employed for reaction with corresponding nucleophilic groups. Examples of such electrophilic groups include acid chloride groups; anhydrides, ketones, aldehydes, isocyanate, isothiocyanate, epoxides, and olefins, including conjugated olefins such as ethenesulfony! (-SO2CH=CH2) and analogous functional groups.
Other in situ Crosslinkinq Materials
Numerous other types of in situ forming materials have been described which may be used in combination with an anti-scarring agent in accordance with the invention. The in situ forming material may be a biocompatible crosslinked polymer that is formed from water soluble precursors having electrophilic and nucleophilic groups capable of reacting" and crosslinking in situ (see, e.g., U.S. Patent No. 6,566,406). The in situ forming material may be hydrogel that may be formed through a combination of physical and chemical crosslinking processes, where physical crosslinking is mediated by one or more natural or synthetic components that stabilize the hydrogel-forming precursor solution at a deposition site for a period of time sufficient for more resilient chemical crosslinks to form (see, e.g., U.S. Patent No. 6,818,018). The in situ forming material may be formed upon exposure to an aqueous fluid from a physiological environment from dry hydrogel precursors (see, e.g., U.S. Patent No. 6,703,047). The in situ forming material may be a hydrogel matrix that provides controlled release of relatively low molecular weight therapeutic species by first dispersing or dissolving the therapeutic species within relatively hydrophobic rate modifying agents to form a mixture; the mixture is formed into microparticles that are dispersed within bioabsorbable hydrogels, so as to release the water soluble therapeutic agents in a controlled fashion (see, e.g., 6,632,457). The in situ forming material may be a multi-component hydrogel system (see, e.g., U.S. Patent No. 6,379, 373). The in situ forming material may be a multi-arm block copolymer that includes a central core molecule, such as a residue of a polyol, and at least three copolymer arms covalently attached to the central core molecule, each copolymer arm comprising an inner hydrophobic polymer segment covalently attached to the central core molecule and an outer hydrophilic polymer segment covalently attached to the hydrophobic polymer segment, wherein the central core molecule and the hydrophobic polymer segment define a hydrophobic core region (see, e.g., 6,730,334). The in situ forming material may include a gel-forming macromer that includes at least four polymeric blocks, at least two of which are hydrophobic and at least one of which is hydrophilic, and including a crosslinkable group (see, e.g., 6,639,014). The in situ forming material may be a water-soluble macromer that includes at least one hydrolysable linkage formed from carbonate or dioxanone groups, at least one water-soluble polymeric block, and at least one polymerizable group (see, e.g., U.S. Patent No. 6,177,095). The in situ forming material may comprise polyoxyalkylene block copolymers that form weak physical crosslinks to provide gels having a paste-like consistency at physiological temperatures, (see, e.g., U.S. Patent No. 4,911,926). The in situ forming material may be a thermo-irreversible gel made from polyoxyalkylene polymers and ionic polysaccharides (see, e.g., U.S. Patent No. 5,126,141). The in situ forming material may be a gel forming composition that includes chitin derivatives (see, e.g., U.S. Patent No. 5,093,319), chitosan-coagulum (see, e.g., U.S. Patent No. 4,532,134), or hyaluronic acid (see, e.g., U.S. Patent No. 4,141 ,973). The in situ forming material may be an in situ modification of alginate (see, e.g., U.S. Patent No. 5,266,326 ). The in situ forming material may be formed from ethylenically unsaturated water soluble macromers that can be crosslinked in contact with tissues, cells, and bioactive molecules to form gels (see, e.g., U.S. Patent No. 5,573,934). The in situ forming material may include urethane prepolymers used in combination with an unsaturated cyano compound containing a cyano group attached to a carbon atom, such as cyano(meth)acrylic acids and esters thereof (see, e.g., U.S. Patent No. 4,740,534). The in situ forming material may be a biodegradable hydrogel that polymerizes by a photoinitiated free radical polymerization from water soluble macromers (see, e.g., U.S. Patent No. 5,410,016). The \n situ forming material may be formed from a two component mixture including a first part comprising a serum albumin protein in an aqueous buffer having a pH in a range of about 8.0-11.0, and a second part comprising a water- compatible or water-soluble bifunctional crosslinking agent, (see, e.g., U.S. Patent No. 5,583,114).
In another aspect, in situ forming materials that can be used include those based on the crosslinking of proteins. For example, the in situ forming material may be a biodegradable hydrogel composed of a recombinant or natural human serum albumin and poly(ethylene) glycol polymer solution whereby upon mixing the solution cross-links to form a mechanical non-liquid covering structure which acts as a sealant. See e.g., U.S. Patent No. 6,458,147 and 6,371 ,975. The in situ forming material may be composed of two separate mixtures based on fibrinogen and thrombin which are dispensed together to form a biological adhesive when intermixed either prior to or on the application site to form a fibrin sealant. See e.g., U.S. Patent No. 6,764,467. The in situ forming material may be composed of ultrasonically treated collagen and albumin which form a viscous material that develops adhesive properties when crosslinked chemically with glutaraldehyde and amino acids or peptides. See e.g., U.S. Patent No. 6,310,036. The in situ forming material may be a hydrated adhesive gel composed of an aqueous solution consisting essentially of a protein having amino groups at the side chains (e.g., gelatin, albumin) which is crosslinked with an N-hydroxyimidoester compound. See e.g., U.S. Patent No. 4,839,345. The in situ forming material may be a hydrogel prepared from a protein or polysaccharide backbone (e.g., albumin or polymannuronic acid) bonded to a cross-linking agent (e.g., polyvalent derivatives of polyethylene or polyalkylene glycol). See e.g., U.S. Patent No. 5,514,379. The in situ forming material may be composed of a polymerizable collagen composition that is applied to the tissue and then exposed to an initiator to polymerize the collagen to form a seal over a wound opening in the tissue. See e.g., U.S. Patent No. 5,874,537. The in situ forming material may be a two component mixture composed of a protein (e.g., serum albumin) in an aqueous buffer having a pH in the range of about 8.0-11.0 and a water- soluble bifunctional polyethylene oxide type crosslinking agent, which transforms from a liquid to a strong, flexible bonding composition to seal tissue in situ. See e.g., U.S. Patents 5,583,114 and RE38158 and PCT Publication No. WO 96/03159. The in situ forming material may be composed of a protein, a surfactant, and a lipid in a liquid carrier, which is crosslinked by adding a crosslinker and used as a sealant or bonding agent in situ. See e.g., U.S. Patent Application No. 2004/0063613A1 and PCT Publication Nos. WO 01/45761 and WO 03/090683. The in situ forming material may be composed of two enzyme-free liquid components that are mixed by dispensing the components into a catheter tube deployed at the vascular puncture site, wherein, upon mixing, the two liquid components chemically cross-link to form a mechanical non-liquid matrix that seals a vascular puncture site. See e.g., U.S. Patent Application Nos. 2002/0161399A1 and 2001 /0018598A1. The in situ forming material may be a cross-linked albumin composition composed of an albumin preparation and a carbodiimide preparation which are mixed under conditions that permit crosslinking of the albumin for use as a bioadhesive or sealant. See e.g., PCT Publication No. WO 99/66964. The in situ forming material may be composed of collagen and a peroxidase and hydrogen peroxide, such that the collagen is crosslinked to from a semi-solid gel that seals a wound. See e.g., PCT Publication No. WO 01/35882.
In another aspect, in situ forming materials that can be used include those based on isocyanate or isothiocyanate capped polymers. For example, the in situ forming material may be composed of isocyanate- capped polymers that are liquid compositions which form into a solid adhesive coating by in situ polymerization and crosslinking upon contact with body fluid or tissue. See e.g., PCT Publication No. WO 04/021983. The in situ forming material may be a moisture-curing sealant composition composed of an active isocyanato-terminated isocyanate prepolymer containing a polyol component with a molecular weight of 2,000 to 20,000 and an isocyanurating catalyst agent. See e.g., U.S. Patent No. 5,206,331.
In another embodiment, the reagents can undergo an electrophilic-nucleophilic reaction to produce a crosslinked matrix. Polymers containing and/or terminated with nucleophilic groups such as amine, sulfhydryl, hydroxyl, -PH2 or CO-NH-NH2 can be used as the nucleophilic reagents and polymers containing and/or terminated with electrophilic groups such as succinimidyl, carboxylic acid, aldehyde, epoxide, isocyanate, vinyl, vinyl sulfone, maleimide, -S-S-(C5H4N) or activated esters, such as are used in peptide synthesis can be used as the electrophilic reagents. For example, a 4-armed thiol derivatized poly(ethylene glycol) (e.g., pentaerythritol poly(ethylene glycol)ether tetra-sulfhydryl) can be reacted with a 4 armed NHS-derivatized polyethylene glycol (e.g., pentaerythritol poly(ethylene glycol)ether tetra-succinimidyl glutarate) under basic conditions (pH > about 8). Representative examples of compositions that undergo such electrophilic-nucleophilic crosslinking reactions are described, for example, in U.S. Patent. Nos. 5,752,974; 5,807,581; 5,874,500; 5,936,035; 6,051 ,648; 6,165,489; 6,312,725; 6,458,889; 6,495,127; 6,534,591; 6,624,245; 6,566,406; 6,610,033; 6,632,457; and PCT Application Publication Nos. WO 04/060405 and WO 04/060346.
In another embodiment, the electrophilic- or nucleophilic- terminated polymers can further comprise a polymer that can enhance the mechanical and/or adhesive properties of the in situ forming compositions. This polymer can be a degradable or non-degradable polymer. For example, the polymer may be collagen or a collagen derivative, for example methylated collagen. An example of an in situ forming composition uses pentaerythritol poly(ethylene glycol)ether tetra-sulfhydryl) (4-armed thiol PEG), pentaerythritol poly(ethylene glycol)ether tetra-succinimidyl glutarate) (4-armed NHS PEG) and methylated collagen as the reactive reagents. This composition, when mixed with the appropriate buffers can produce a crosslinked hydrogel. (See, e.g., U.S. Patent Nos. 5,874,500; 6,051 ,648; 6,166,130; 5,565,519 and 6,312,725J.
In another embodiment, the reagents that can form a covalent bond with the tissue to which it is applied may be used. Polymers containing and/or terminated with electrophilic groups such as succinimidyl, aldehyde, epoxide, isocyanate, vinyl, vinyl sulfone, maleimide, -S-S-(CsH4N) or activated esters, such as are used in peptide synthesis may be used as the reagents. For example, a 4 armed NHS-derivatized polyethylene glycol (e.g., pentaerythritol poly(ethylene glycol)ether tetra-succinimidyl glutarate) may be applied to the tissue in the solid form or in a solution form. In the preferred embodiment, the 4 armed NHS-derivatized polyethylene glycol is applied to the tissue under basic conditions (pH > about 8). Other representative examples of compositions of this nature that may be used are disclosed in PCT Application Publication No. WO 04/060405 and WO 04/060346, and U.S. Patent Application No. 10/749,123.
In another embodiment, the in situ forming material polymer can be a polyester. Polyesters that can be used in in situ forming compositions include poly(hydroxyesters). In another embodiment, the polyester can comprise the residues of one or more of the monomers selected from lactide, lactic acid, glycolide, glycolic acid, e-caprolactone, gamma-caprolactone, hydroxy valeric acid, hydroxybutyric acid, beta- butyrolactone, gamma-butyrolactone, gamma-valerolactone, γ~ decanolactone, δ-decanolactone, trimethylene carbonate, 1,4-dioxane-2- one or 1 ,5-dioxepan-2one. Representative examples of these types of compositions are described in U.S. Patent. Nos. 5,874,500; 5,936,035; 6,312,725; 6,495,127 and PCT Publication Nos. WO 2004/028547. In another embodiment, the electrophilic-terminated polymer can be partially or completely replaced by a small molecule or oligomer that comprises an electrophilic group (e.g., disuccinimidyl glutarate).
In another embodiment, the nucleophilic-terminated polymer can be partially or completely replaced by a small molecule or oligomer that comprises a nucleophilic group (e.g., dicysteine, dilysine, trilysine, etc.).
Other examples of in situ forming materials that can be used include those based on the crosslinking of proteins (described in, for example, U.S. Patent Nos. RE38158; 4,839,345; 5,514,379, 5,583,114; 6,310,036; 6,458,147; 6,371 ,975; US Patent Application Publication Nos. 2004/0063613A1 , 2002/0161399A1 , and 2001/0018598A1 , and PCT Publication Nos. WO 03/090683, WO 01/45761 , WO 99/66964, and WO 96/03159) and those based on isocyanate or isothiocyanate capped polymers (see, e.g., PCT Publication No. WO 04/021983).
Other examples of in situ forming materials can include reagents that comprise one or more cyanoacrylate groups. These reagents can be used to prepare a poly(alkylcyanoacrylate) or poly(carboxyalkylcyanoacrylate) (e.g., poly(ethylcyanoacrylate), poly(butylcyanoacrylate), poly(isobutylcyanoacrylate), poly(hexylcyanoacrylate), poly(methoxypropylcyanoacrylate), and poly(octylcyanoacrylate)).
Examples of commercially available cyanoacrylates that can be used in the present invention include DERMABOND, INDERMIL, GLUSTITCH, VETBOND, HISTOACRYL, TISSUMEND, HISTOACRYL BLUE and ORABASE SOOTHE-N-SEAL LIQUID PROTECTANT.
In another embodiment, the cyanoacrylate compositions may further comprise additives to stabilize the reagents and/or alter the rate of reaction of the cyanoacrylate, and/or plasticize the poly(cyanoacrylate), and/or alter the rate of degradation of the poly(cyanoacrylate). For example, a trimethylene carbonate based polymer or an oxalate polymer of poly(ethylene glycol) or a ε-caprolactone based copolymer may be mixed with a 2-alkoxyalkylcyanoacrylate (e.g., 2-methoxypropyIcyanoacrylate). Representative examples of these compositions are described in U.S. Patent Nos. 5,350,798 and 6,299,631.
In another embodiment, the cyanoacrylate composition can be prepared by capping heterochain polymers with a cyanoacrylate group. The cyanoacrylate-capped heterochain polymer preferably has at least two cyanoacrylate ester groups per chain. The heterochain polymer can comprise an absorbable poly(ester), poly(ester-carbonate), poly(ether- carbonate) and poly(ether-ester). The poly(ether-ester)s described in U.S. Patent Nos. 5,653,992 and 5,714,159 can also be used as the heterochain polymers. A triaxial poly(ε-caprolactone-co-trimethylene carbonate) is an example of a poly(ester-carbonate) that can be used. The heterochain polymer may be a polyether. Examples of polyethers that can be used include poly(ethylene glycol), polyφropylene glycol) and block copolymers of poly(ethylene glycol) and poly(propylene glycol) (e.g., PLURONICS group of polymers including but not limited to PLURONIC F127 or F68). Representative examples of these compositions are described in U.S. Patent No. 6,699,940.
Within another aspect of the invention, the biologically active ant-infective and/or fibrosis-inhibiting agent can be delivered with a non- polymeric compound (e.g., a carrier). These non-polymeric carriers can include sucrose derivatives (e.g., sucrose acetate isobutyrate, sucrose oleate), sterols such as cholesterol, stigmasterol, β-sitosterol, and estradiol; cholesteryl esters such as cholesteryl stearate; Ci2 -C24 fatty acids such as lauric acid, myristic acid, palmitic acid, stearic acid, arachidic acid, behenic acid, and lignoceric acid; Ci8 -C36 mono-, di- and triacylglycerides such as glyceryl monooleate, glyceryl monolinoleate, glyceryl monolaurate, glyceryl monodocosanoate, glyceryl monomyristate, glyceryl monodicenoate, glyceryl dipalmitate, glyceryl didocosanoate, glyceryl dimyristate, glyceryl didecenoate, glyceryl tridocosanoate, glyceryl trimyristate, glyceryl tridecenoate, glycerol tristearate and mixtures thereof; sucrose fatty acid esters such as sucrose distearate and sucrose palmitate; sorbitan fatty acid esters such as sorbitan monostearate, sorbitan monopalmitate and sorbitan tristearate; Ci6 -C18 fatty alcohols such as cetyl alcohol, myristyl alcohol, stearyl alcohol, and cetostearyl alcohol; esters of fatty alcohols and fatty acids such as cetyl palmitate and cetearyl palmitate; anhydrides of fatty acids such as stearic anhydride; phospholipids including phosphatidylcholine (lecithin), phosphatidylserine, phosphatidylethanolamine, phosphatidylinositol, and lysoderivatives thereof; sphingosine and derivatives thereof; spingomyelins such as stearyl, palmitoyl, and tricosanyl spingomyelins; ceramides such as stearyl and palmitoyl ceramides; glycosphingolipids; lanolin and lanolin alcohols, calcium phosphate, sintered and unscintered hydoxyapatite, zeolites; and combinations and mixtures thereof.
Representative examples of patents relating to non-polymeric delivery systems and the preparation include U.S. Patent Nos. 5,736,152; 5,888,533; 6,120,789; 5,968,542; and 5,747,058.
Within certain embodiments of the invention, the therapeutic compositions are provided that include (i) a fibrosis-inhibiting agent and/or (ii) an anti-infective agent. The therapeutic compositions may include one or more additional therapeutic agents (such as described above), for example, anti-inflammatory agents, anti-thrombotic agents, and/ or anti-platelet agents. Other agents that may be combined with the therapeutic compositions include, e.g., additional ingredients such as surfactants (e.g., PLURONICS, such as F-127, L-122, L-101, L-92, L-81 , and L-61), preservatives, anti-oxidants.
In one aspect, the present invention provides compositions comprising i) an anti-fibrotic agent and ii) a polymer or a compound that forms a polymer in situ. The following are some, but by no means all, of the preferred anti-fibrotic agents and classes of anti-fibrotic agents that may be included in the inventive compositions:
1a) an anti-fibrotic agent that inhibits cell regeneration, 2a) an anti-fibrotic agent that inhibits angiogenesis, 3a) an anti-fibrotic agent that inhibits fibroblast migration, 4a) an anti-fibrotic agent that inhibits fibroblast proliferation, 5a) an anti-fibrotic agent that inhibits deposition of extracellular matrix,
6a) an anti-fibrotic agent inhibits tissue remodeling, 7a) an adensosine A2A receptor antagonist, 8a) an AKT inhibitor,
9a) an alpha 2 integrin antagonist, wherein the alpha 2 integrin antagonist is Pharmaprojects No. 5754 (Merck KgaA), 10a) an alpha 4 integrin antagonist, 11a) an alpha 7 nicotinic receptor agonist, 12a) an angiogenesis inhibitor selected from the group consisting of AG-12,958 (Pfizer), ATN-161 (Attenuon LLC), neovastat, an angiogenesis inhibitor from Jerina AG (Germany), NM-3 (Mercian), VGA- 1155 (Taisho), FCE-26644 (Pfizer), FCE-26950 (Pfizer), FPMA (Meiji Daries), FR-111142 (Fujisawa), GGTI-298, GM-1306 (Ligand), GPA-1734 (Novartis), NNC-47-0011 (Novo Nordisk), herbamycin (Nippon Kayaku), lenalidomide (Celegene), IP-10 (NIH), ABT-828 (Abbott), KIN-841 (Tokushima University, Japan), SF-1126 (Semafore Pharmaceuticals), laminin technology (NIH), CHIR-258 (Chiron), NVP-AEW541 (Novartis), NVP-AEW541 (Novartis), Vt16907 (Alchemia), OXI-8007 (Oxigene), EG- 3306 (Ark Therapeutics), Maspin (Arriva), ABT-567 (Abbott), PPI-2458 (Praecis Pharmaceuticals), CC-5079, CC-4089 (Celgene), HIF-1 alpha inhibitors (Xenova), S-247 (Pfizer), AP-23573 (Ariad), AZD-9935 (Astra Zeneca), mebendazole (Introgen Therapeutics), MetAP-2 inhibitors (GlaxoSmithKline), AG-615 (Angiogene Pharmaceuticals), Tie-2 antagonists (Hybrigenics), NC-381 , CYC-381 , NC-169, NC-219, NC-383, NC-384, NC- 407 (Lorus Therapeutics), ATN-224 (Attenuon), ON-01370 (Onconova), Vitronectin antagonists (Amgen), SDX-103 (Salmedix), Vitronectin antagonists (Shire), CHP (Riemser), TEK (Amgen), Anecortave acetate (Alcon), T46.2 (Matrix Therapeutics), HG-2 (Heptagen), TEM antagonists (Genzyme), Oxi-4500 (Oxigene), ATN-161 (Attenuon), WX-293 (Wilex), M- 2025 (Metris Therapeutics), Alphastatin (BioActa), YH-16 (Yantai Rongchang), BIBF-1120 (Boehringer Ingelheim), BAY-57-9352 (Bayer), AS- 1404 (Cancer Research Technology), SC-77964 (Pfizer), glycomimetics (BioTie Therapies), TIE-2 Inhibitors (Ontogen), DIMI, Octamer (Octamer), ABR-215050 (Active Biotech), ABT-518 (Abbott), KDR inhibitors (Abbott), BSF-466895 (Abbott), SCH-221153 (Schering-Plough), DAC:antiangiogenic (ConjuChem), TFPI (EntreMed), AZD-2171 (Astra-Zenaca), CDC-394 (Celgene), LY290293 (EIi Lilly), IDN-5390 (Indena), Kdr Kinase Inhibitors (Merck), CT-113020, CT-116433, CT-116563, CT-31890, CT-32228) (Cell Therapeutics), A-299620 (Abbott), TWEAK Inhibitor (Amgen), VEGF modulators (Johnson and Johnson), Tum-N53, tumstatin (Genzyme), Thios- 1 , Thios-2 (Thios Pharmaceuticals), MV-6401 (Miravant Medical Technologies), Spisulosine (PharmaMar), CEP-7055 (Cephalon), AUV-201 (Auvation),~LM-609 (EIi Lilly), SKF-106615 (AnorMED), Oglufanide disodium (Cytran), BW-114 (Pharminox), Calreticulin (NIH), WX-678 (Wilex), SD-7784 (Pfizer), WX-UK1 (Wilex), SH-268 (Schering AG), 2-Me-PGA (Celgene), S- 137 (Pfizer), ZD-6126 (Angiogene Pharmaceuticals), SG-292 (SignalGen), Benefin (Lane Labs), A6, A36 (Angstrom), SB-2723005 (GlaxoSmithKline), SC-7 (Cell Therapeutics), ZEN-014 (AEterna Zentaris), 2-methoxyestradiol (EntreMed), NK-130119 (Nippon Kayaku), CC-10004 (Celgene), AVE- 8062A (Ajinomoto), Tacedinaline (Pfizer), Actinonin (Tokyo Metropolitan Institute of Medical Science), Lenalidomide (Celgene), VGA-1155, BTO-956 (SRI International), ER-68203-00 (Eisai), CT-6685 (UCB), JKC-362 (Phoenix Pharmaceuticals), DMI-3798 (DMI Biosciences, Angiomate (Ipsen), ZD-6474 (AstraZeneca), CEP-5214 (Cephalon), Canstatin (Genzyme), NM-3 (Mercian), Oridigm (MediQuest Therapeutics), Exherin (Adherex), BLS-0597 (Boston Life Sciences), PTC-299 (PTC Therapeutics), NPI-2358 (Nereus Pharmaceuticals), CGP-79787 (Novartis), AEE-788 (Novartis), CKD-732 (Chong Kun Dang), CP-564959 (OSI Pharmaceuticals), CM-101 (CarboMed), CT-2584, CT3501 (Cell Therapeutics), combretastatin and analogues and derivatives thereof (Oxigene), Rebimastat (Bristol-Meyers Squibb), Dextrin 2-sulfate (ML Laboratories), Cilengitide (Merk KGaA), NSC-706704 (Pharminox), KRN- 951 (Kirin Brewery), Ukrain, NSC-631570 (Nowicky Pharma), Tecogalan sodium (Daiichi Pharmaceutical), Tz-93 (Tsumura), TBC-1635 (Encysive Pharmaceuticals), TAN-1120 (Takeda), Semaxanib (Pfizer), BDI-7800 (Biopharmacopae), SD-186, SD-983 (Bristol-Meyers Squibb), SB-223245 (GlaxoSmithKline), SC-236 (Pfizer), RWJ-590973 (Johnson and Johnson), ILX-1850 (Genzyme), SC-68488, S-836 (Pfizer), CG-55069-11 (CuraGen), Ki-23057 (Kirin Brewery), CCX-700 (Chemoentryx), Pegaptanib octasodium (Giled Sciences), ANGIOCOL (available from Biostratum Inc.), or an analogue or derivative thereof,
13a) an apoptosis antagonist,
14a) an apoptosis activator,
15a) a beta 1 integrin antagonist,
16a) a beta tubulin inhibitor,
17a) a blocker of enzyme production in Hepatitis C,
18a) a Bruton's tyrosine kinase inhibitor,
19a) a calcineurin inhibitor,
20a) a caspase 3 inhibitor,
21a) a CC chemokine receptor antagonist,
22a) a cell cycle inhibitor selected from the group consisting of SNS-595 (Sunesis), synthadotin, KRX-0403, homoharringtonine, and an analogue or derivative thereof,
23a) a cathepsin B inhibitor,
24a) a cathepsin K inhibitor, wherein the cathepsin K inhibitor is 462795 (GlaxoSmithKline), INPL-022-D6 (Amura Therapeutics), or an analogue or derivative thereof,
25a) a cathepsin L inhibitor,
26a) a CD40 antagonist, 27a) a chemokine receptor agonist,
28a) a chymase inhibitor,
29a) a collagenase antagonist,
30a) a CXCR antagonist,
31a) a cyclin dependent kinase inhibitor selected from the group consisting of a CDK-1 inhibitor, a CDK-2 inhibitor, a CDK- 4 inhibitor, a CDK-6 inhibitor, a CAK1 inhibitor from GPC Biotech and Bristol-Myers Squibb, RGB-286199 (GPC Biotech), an anticancer agent from Astex Technology, a CAK1 inhibitor from GPC Biotech, a CDK inhibitor from Sanofi-Aventis, a CDK1/CDK2 inhibitor from Amgen, a CDK2 inhibitor from SUGEN-2 (Pfizer), a hearing loss therapy agent (Sound Pharmaceuticals), PD-0332991 (Pfizer), RGB-286199 (GPC Biotech), Ro-0505124 (Hoffmann- La Roche), a Ser/Thr kinase inhibitor from Lilly (EIi Lilly), CVT-2584 (CAS No. 199986-75-9) (CV Therapeutics), CGP 74514A, bohemine, olomoucine (CAS No. 101622-51-9), indole-3-carbinol (CAS No. 700-06-1), and an analogue or derivative thereof,
32a) a cyclooxygenase 1 inhibitor,
33a) a DHFR inhibitor,
34a) a dual integrin inhibitor,
35a) an elastase inhibitor,
36a) an elongation factor-1 alpha inhibitor,
37a) an endothelial growth factor antagonist,
38a) an endothelial growth factor receptor kinase inhibitor selected from the group consisting of sorafenib tosylate (Bayer), AAL-993 (Novartis), ABP-309 (Novartis), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EXEL- 2880 (Exelixis), GW-654652 (GlaxoSmithKline), lavendustin A (CAS No. 125697-92-9), a KDR inhibitor from LG Life Sciences, CT-6685 and CT- 6729 (UCB), KRN-633 and KRN-951 (Kirin Brewery), OSI-930 (OSI Pharmaceuticals), SP-5.2 (Supratek Pharma), SU-11657 (Pfizer), a Tie-2 antagonist (Hybrigenics), SU 1498 (a VEGF-R inhibitor), a VEGFR-2 kinase inhibitor (Bristol-Myers Squibb), XL-647 (Exelixis), a KDR inhibitor from Abbott Laboratories, sorafenib tosylate, and an analogue or derivative thereof,
39a) an endotoxin antagonist,
40a) an epothilone and tubulin binder,
41a) an estrogen receptor antagonist,
42a) an FGF inhibitor,
43a) a famexyl transferase inhibitor,
44a) a famesyltransferase inhibitor selected from the group of A-197574 (Abbott), a famesyltransferase inhibitor from Servier, FPTIII (Strathclyde Institute for Drug R), LB-42908 (LG Life Sciences), Pharmaprojects No. 5063 (Genzyme), Pharmaprojects No. 5597 (Ipsen), Yissum Project No. B-1055 (Yissum), and an analogue or derivative thereof,
45a) an FLT-3 kinase inhibitor,
46a) an FGF receptor kinase inhibitor,
47a) a fibrinogen antagonist selected from the group consisting of AUV-201 (Auvation), MG-13926 (Sanofi-Aventis), plasminogen activator (CAS No. 105913-11-9) (from Sanofi-Aventis or UCB), plasminogen activator-2 (tPA-2) (Sanofi-Aventis), pro-urokinase (CAS No. 82657-92-9) (Sanofi-Aventis), mevastatin, and an analogue or derivative thereof,
48a) a heat shock protein 90 antagonist selected from the group consisting of SRN-005 (Sirenade), geldanamycin, NSC-33050 (17- allylaminogeldanamycin; 17-AAG), 17-dimethylaminoethylamino-17- demethoxy-geldanamycin (17-DMAG), rifabutin (rifamycin XIV, 1',4- didehydro-1-deoxy-1 ,4-dihydro-5'-(2-methylpropyl)-1-oxo-), radicicol from Humicola fuscoatra (CAS No. 12772-57-5), and an analogue or derivative thereof,
49a) a histone deacetylase inhibitor,
50a) an HMGCoA reductase inhibitor selected from the group consisting of an atherosclerosis therapeutic from Lipid Sciences, ATI-16000 (ARYx Therapeutics), KS-01-019 (Kos Pharmaceuticals), Pharmaprojects No. 2197 (Sanofi-Aventi), RP 61969 (Sanofi-Aventis), cerivastatin Na (CAS No. 143201-11-0), and an analogue or derivative thereof,
51a) an ICAM inhibitor,
52a) an IL, ICE and IRAK antagonist, wherein the antagonist is a CJ-14877, CP-424174 (Pfizer), NF-61 (Negma-Lerads), and an analogue or derivative thereof,
53a) an IL-2 inhibitor,
54a) an immunosuppressant selected from the group consisting of teriflunomide (Sanofi Aventis), chlorsulfaquinoxalone (NSC- 339004), chlorsulfaquinoxalone sulfate, CS-712 (Sankyo), ismomultin alfa (CAS No. 457913-93-8) (Akzo Nobel), antiallergics from GenPat77, antiinflammatories or AT-005 (Androclus Therapeutics), autoimmune disease therapeutics from EpiVax, BN-007 (Bone), budesonide (CAS No. 51333-22- 3) (MAP Pharmaceuticals), CO-14 (Genzyme), edratide (CAS No. 433922- 67-9) (Teva), EP-314 (Enanta), eprovafen (CAS No. 101335-99-3) (Sanofi- Aventis), HWA-131 (CAS No. 118788-41-3) (Sanofi-Aventis), immunomodulators from MerLion Pharmaceuticals, immunosuppressives from Alchemia, IPL-12 (Inflazyme), MDL-9563 (CAS No. 27086-86-8) (Sanofi-Aventis), Pharmaprojects No. 2330 (Sanofi-Aventis), Pharmaprojects No. 6426 (Abgenix), PXS-25 (Pharmaxis), rosmarinic acid (CAS No. 20283-92-5) (Sanofi-Aventis), RP 42927 or RP 54745 (CAS No. 135330-08-4) (Sanofi-Aventis), SGN-35 (Seattle Genetics), ST-1959 (Sigma-Tau), type I diabetes therapy from SYNX Pharma, UNIL-88 (Debiopharm), VP-025 (Vasogen), VR-694 (Vectura), PRTX-001 (Protalex), and an analogue or derivative thereof,
55a) an IMPDH (inosine monophosphate),
56a) an integrin antagonist,
57a) an interleukin antagonist,
58a) an inhibitor of type III receptor tyrosine kinase, 59a) an irreversible inhibitor of enzyme methionine aminopeptidase type 2,
60a) an isozyme selective delta protein kinase C inhibitor,
61a) a JAK3 enzyme inhibitor,
62a) a JNK inhibitor,
63a) a kinase inhibitor,
64a) a kinesin antagonist,
65a) a leukotriene inhibitor and antagonist selected from the group consisting of ambicromil (CAS No. 58805-38-2) (Sanofi-Aventis), amelubant (CAS No. 346735-24-8) (Boehringer Ingelheim), DW-1141 (Dong Wha), ebselen (Daiichi Pharmaceutical), ibudilast (Kyorin), leucotriene inhibitors from Sanofi-Aventis, lymphotoxin-beta receptor (LT-β) from Biogen Idee, Pharmaprojects No. 1535 and 2728 (CAS No. 119340-33-9) (Sanofi- Aventis), R-112 (Rigel), Rev-5367 (CAS No. 92532-05-3) (Sanofi-Aventis), RG-14893 (CAS No. 141835-49-6) (Sanofi-Aventis), RG-5901-A (CAS No. 101910-24-1), 92532-23-5, RP 66153 (CAS No. 142422-79-5), RP 66364 (CAS No.186912-92-5), RP 69698 (CAS No. 141748-00-7) (Sanofi-Aventis), SC-411930 (Pfizer), SC-41930 (CAS No. 120072-59-5) (Pfizer), SC-50605 (CAS No. 138828-39-4) (Pfizer), SC-51146 (CAS No. 141059-52-1), SC- 53228 (CAS No. 153633-01-3) (Pfizer), spaglumic acid (ZY-15106) (CAS No.3106-85-2), 80619-64-3 (Novartis), tipredane (CAS No. 85197-77-9) (Bristol-Myers Squibb), U-75302 (CAS No. 119477-85-9) (Pfizer), and analogue or derivative thereof,
66a) a MAP kinase inhibitor,
67a) a matrix metalloproteinase inhibitor,
68a) an MCP-CCR2 inhibitor,
69a) an mTOR inhibitor,
70a) an mTOR kinase inhibitor,
71a) a microtubule inhibitor selected from the group consisting of antibody-maytansinoid conjugates from Biogen Idee, colchicines (MantiCore Pharmaceuticals), anticancer immunoconjugates from Johnson & Johnson, DIME from Octamer, gni-1f (GNI), huC242-DM4, huMy9-6-DM1 (ImmunoGen), IDN-5404 (Indena), IMO-098, IMOderm (Imotep), mebendazole (Introgen Therapeutics), microtubule poisons from Cambridge Enterprise, paclitaxel such as LOTAX from Aphios (CAS No. 33069-62-4), Genexol-PM from Samyang, Pharmaprojects No. 6383 (Tapestry Pharmaceuticals), RPR-112378 (Sanofi-Aventis), SGN-75 (Seattle Genetics), SPL-7435 (Starpharma), SSR-250411 (Sanofi-Aventis), trastuzumab-DM1 (Genentech), vinorelbine, dolastatin 15 (CAS No. 123884-00-4), vincamine, and an analogue or derivative thereof,
72a) an MIF inhibitor,
73a) an MMP inhibitor,
74a) a neurokinin (NK) antagonist selected from the group consisting of anthrotainin (CAS No. 148084-40-6) (Sanofi-Aventis), an IBS thereapeutic from ArQuIe, MDL-105212A (CAS No. 167261-60-1) (Ssanofi- Aventis), Pharmaprojects No. 2744, 3258 (CAS No. 139167-47-8) 4006, 4201 , or 5986 (Sanofi-Aventis), RP 67580 (CAS No. 135911-02-3), SR- 144190 (CAS No. 201152-86-5), SSR-240600, SSR-241586 (Sanofi- Aventis), TKA-457 (Novartis), vestipitant mesylate (CAS No. 334476-64-1) (GlaxoSmithKline), Win-64821 (Sanofi-Aventis), PRX-96026 (Predix Pharmaceuticals), and an analogue or derivative thereof,
75a) an NF kappa B inhibitor selected from the group consisting of emodin (CAS No. 518-82-1), AVE-0545 or AVE-0547 (Sanofi- Aventis), bortezomib (CAS No. 179324-69-7) (Millennium Pharmaceuticals), dexanabinol (CAS No. 112924-45-5) (Pharmos), dexlipotam (Viatris), Pharmaprojects No. 6283 (INDRA) (OXiGENE), IPL-576092 (CAS No. 137571-30-3) (Inflazyme), NFKB decoy (Corgentech), NFKB decoy oligo (AnGes MG), NFKB's from Ariad, osteoporosis treatments or S5 (F005) from Fulcrum Pharmaceuticals, P61 (Phytopharm), R-flurbiprofen (CAS No. 5104-49-4) (Encore Pharmaceuticals), Bay 11-7085, and an analogue or derivative thereof,
76a) a nitric oxide agonist, 77a) an ornithine decarboxylase inhibitor, 78a) a p38 MAP kinase inhibitor selected from the group consisting of AZD-6703 (AstraZeneca), JX-401 (Jexys Pharmaceuticals), BMS-2 (Bristol-Myers Squibb), a p38 MAP kinase inhibitor from Novartis, a p38-alpha MAP kinase inhibitor from Amphora, Pharmaprojects No. 5704 (Pharmacopeia), SKF86002 (CAS No. 72873-74-6), RPR-200765A (Sanofi- Aventis), SD-282 (Johnson & Johnson), TAK-715 (Takeda), and an analogue or derivative thereof,
79a) a palmitoyl-protein thioesterase inhibitor, 80a) a PDGF receptor kinase inhibitor selected from the group consisting of AAL-993, AMN-107, or ABP-309 (Novartis), AMG-706 (Amgen), BAY-57-9352 (Bayer), CDP-860 (UCB), E-7080 (Eisai), imatinib (CAS No. 152459-95-5) (Novartis), OSI-930 (OSI Pharmaceuticals), RPR- 127963E (Sanofi-Aventis), RWJ-540973 (Johnson & Johnson), sorafenib tosylate (Bayer), SU-11657 (Pfizer), tandutinib (CAS No. 387867-13-2) (Millennium Pharmaceuticals), vatalanib (Novartis), ZK-CDK (Schering AG), and an analogue or derivative thereof,
81a) a peroxisome proliferators-activated receptor agonist selected from the group consisting of (-)-halofenate (Metabolex), AMG-131 (Amgen), antidiabetics from Japan Tobacco, AZD-4619, AZD-8450, AZD- 8677 (AstraZeneca), DRF-10945, balaglitazone (Dr Reddy's), CS-00088, CS-00098 (Chipscreen Biosciences), E-3030 (Eisai), etalocib (CAS No. 161172-51-6) (EIi Lilly), GSK-641597 (Ligand), GSK-677954 (GlaxoSmithKline), GW-409544 (Ligand), GW-590735 (GlaxoSmithKline), K- 111 (Hoffmann-La Roche), LY-518674 (EIi Lilly), LY-674 (Ligand), LY-929 (Ligand), MC-3001 , MC-3002 (MaxoCore Pharmaceuticals), metformin HCI + pioglitazone (CAS No. 1115-70-4 and 112529-15-4), ACTOPLUS MET from Andrx), muragiitazar (CAS No. 331741-94-7) (Bristol-Myers Squibb), naveglitazar (Ligand), oleoylethanolamide (Kadmus Pharmaceuticals), ONO-5129, pioglitazone hydrochloride (CAS No. 111025-46-8 and 112529- 15-4) (Takeda), PLX-204 (Plexxikon), PPAR agonists from Genfit, PPAR delta agonists from EIi Lilly, PPAR-alpha agonists from CrystalGenomics, PPAR-gamma modulators and PPAR-β modulators from CareX, rosiglitazone maleate (CAS No. 122320-73-4 or 155141-29-0) (GlaxoSmithKline), rosiglitazone maleate/glimepir (CAS No. 155141-29-0 and 93479-97-1), AVANDARYL, rosiglitazone maleate/metformin extend (CAS No. 155141-29-0 and 657-24-9), AVANDAMET, rosiglitazone maieate+metformin, AVANDAMET (GlaxoSmithKline), tesaglitazar (AstraZeneca), LBM642, WY-14,643 (CAS No. 50892-23-4), GW7647, fenofibric acid (CAS No. 42017-89-0), MCC-555 (CAS No. 161600-01-7), GW9662, GW1929, GW501516, L-165,041 (CAS No. 79558-09-1), and an analogue or derivative thereof,
82a) a phosphatase inhibitor,
83a) a phosphodiesterase (PDE) inhibitor selected from the group consisting of avanafil (Tanabe Seiyaku), dasantafil (CAS No. 569351- 91-3) (Schering-Plough), A-906119 (CAS No. 134072-58-5), DL-850 (Sanofi-Aventis)rGRC-3015, GRC-3566, GRC-3886 (Glenmark), HWA-153 (CAS No. 56395-66-5) (Sanofi-Aventis), hydroxypumafentrine (Altana), IBFB-130011 , IBFB-14-016, IBFB-140301 , IBFB-150007, IBFB-211913 (IBFB Pharma), L-826141 (Merck & Co), medorinone (CAS No. 88296-61-1) (Sanofi-Aventis), MEM-1917 (Memory Pharmaceuticals), ND-1251 (Neuro3d), PDE inhibitors from ICOS, PDE IV inhibitors from Memory Pharmaceuticals and CrystalGenomics, Pharmaprojects No. 2742 and 6141 (Sanofi-Aventis), QAD-171 (Novartis), RHC-2963 (CAS No. 76993-12-9 and 76993-14-1), RPR-117658, RPR-122818 derivatives, SR-24870 , and RPR- 132294 (Sanofi-Aventis), SK-350 (ln2Gen), stroke therapy agents from deCODE Genetics, TAS-203 (Taiho), tofimilast (CAS No. 185954-27-2) (Pfizer), UK-371800 (Pfizer), WIN-65579 (CAS No. 158020-82-7) (Sanofi- Aventis), IBFB-130020 (IBFB Pharma), OPC-6535 (CAS No. 145739-56-6) (Otsuka), theobromine (CAS No. 83-67-0), papverine hydrochloride (CAS No. 61-25-6), quercetin dehydrate (CAS No. 6151-25-3), YM 976 (CAS No. 191219-80-4), irsogladine (CAS No. 57381-26-7), a phosphodiesterase III inhibitor, enoximone, a phosphodiesterase IV inhibitor, fosfosal, Atopik (Barrier Therapeutics), triflusal, a phosphodiesterase V inhibitor, and an analogue or derivative thereof,
84a) a PKC inhibitor,
85a) a platelet activating factor antagonist,
86a) a platelet-derived growth factor receptor kinase inhibitor,
87a) a prolyl hydroxylase inhibitor,
88a) a polymorphonuclear neutrophil inhibitor,
89a) a protein kinase B inhibitor,
90a) a protein kinase C stimulant,
91a) a purine nucleoside analogue,
92a) a purinoreceptor P2X antagonist,
93a) a Raf kinase inhibitor,
94a) a reversible inhibitor of ErbB1 and ErbB2,
95a) a ribonucleoside triphosphate reductase inhibitor,
96a) an SDF-T antagonist,
97a) a sheddase inhibitor,
98a) an SRC inhibitor,
99a) a stromelysin inhibitor,
100a) an Syk kinase inhibitor,
101a) a telomerase inhibitor,
102a) a TGF beta inhibitor selected from the group consisting of pirfenidone (CAS No. 53179-13-8) (MARNAC), tranilast (CAS No. 53902- 12-8) (Kissei), IN-1130 (ln2Gen), mannose-6-phosphate (BTG), TGF-β antagonists from Inflazyme (Pharmaprojects No. 6075), TGF-β antagonists from Sydney, non-industrial source), TGF-β I receptor kinase inhibitors from EIi Lilly, TGF-β receptor inhibitors from Johnson & Johnson, and an analogue or derivative thereof,
103a) a TNFα antagonist or TACE inhibitor selected from the group consisting of adalimumab (CAS No. 331731-18-1) (Cambridge Antibody Technology), AGIX-4207 (AtheroGenics), AGT-1 (Advanced Biotherapy), an anti-inflammatory from Borean Pharma, Cellzome, or Paradigm Therapeutics, anti-inflammatory vaccine (TNF-alpha kinoid) from Neovacs, humanized anti-TNF antibody or an anti-TNF MAb (CB0006) Celltech (UCB), apratastat (CAS No. 287405-51-0) (Wyeth), BMS-561392 (Bristol-Myers Squibb), BN-006 (Bone), certolizumab pegol (CAS No. 428863-50-7 or CH-138 (UCB), cilomilast (CAS No. 153259-65-5) (GlaxoSmithKline), CR-1 (Nuada Pharmaceuticals), CRx-119 (CombinatoRx), D-5410 (UCB), dacopafant (CAS No. 125372-33-0) (Sanofi- Aventis), dersalazine (CAS No. 188913-57-7/188913-58-8) (Uriach), etanercept (CAS No. 185243-69-0) (Amgen), ethyl pyruvate (Critical (Critical Therapeutics), golimumab (CAS No. 476181-74-5) (Johnson & Johnson), hormono-immunotherapy from Ipsen, CDP571 (e.g., Humicade from UCB), IC-485 (ICOS), infliximab (CAS No. 170277-31-3) (Johnson & Johnson), IP- 751 (Manhattan Pharmaceuticals), ISIS-104838 (CAS No. 250755-32-9) (ISIS Pharmaceuticals), lenalidomide (CAS No. 191732-72-6) (Celgene), lentinan (CAS No. 37339-90-5) (Ajinomoto), MDL-201112 (CAS No. 142130-73-2) (Sanofi-Aventis), medroxyprogesterone (CAS No. 520-85-4) (InKine Pharmaceutical), N-acetylcysteine (CAS No. 616-91-1) (Zambon), NBE-P2 (DIREVO Biotech), nerelimomab (CAS No. 162774-06-3) (Chiron), OM-294DP (OM PHARMA), onercept (CAS No. 199685-57-9) (Yeda), PASSTN F-alpha (Verigen), pentoxifylline or oxypentifylline (Sanofi-Aventis), Pharmaprojects No. 4091 , 4241 , 4295, or 4488 (Sanofi-Aventis), Pharmaprojects No. 5480 (Amgen), Pharmaprojects No. 6749 (Cengent), pirfenidone (CAS No. 53179-13-8) (MARNAC), RPR-132294 (Sanofi- Aventis), S5 (F002) (Fulcrum Pharmaceuticals), simvastatin (CAS No. 79902-63-9) (Merck & Co), STA-6292 (Synta Pharmaceuticals), tacrolimus (CAS No. 104987-11-3) (Fujisawa LifeCycle Pharma), talactoferrin alfa (CAS No. 308240-58-6) (Agennix), thalidomide (CAS No. 50-35-1) (Celgene), TNF antagonists form ProStrakan, and Synergen, TNF inhibitors (Amgen), TNF-alpha antagonists from Dynavax Technologies and Jerina AG (Germany), TNF-alpha inhibitors from IBFB Pharma and Xencor (Xencor), torbafylline (CAS No. 105102-21-4) (Sanofi-Aventis), UR-1505 (Uriach), VT-346 (Viron Therapeutics), YSIL6 (Y's Therapeutics), YSTH2 (Y's Therapeutics), NPI-1302a-3 (Nereus Pharmaceuticals, a TNF antagonist from Jerina AG (Germany), dersalazine, and an analogue or derivative thereof,
104a) a tumor necrosis factor antagonist,
105a) a Toll receptor inhibitor,
106a) a tubulin antagonist,
107a) a tyrosine kinase inhibitor selected from the group consisting of SU-011248, SUTENT from Pfizer Inc. (New York, NY), BMS- 354825, PN-355 (Paracelsian Pharmaceuticals), AGN-199659 (Allergan), AAL-993 or ABP-309 (Novartis), adaphostin (NIH), AEE-788 (Novartis), AG- 013736 (OSI Pharmaceuticals), AG-13736 (Pfizer), ALT-110 (Alteris Therapeutics), AMG-706 (Amgen), anticancer MAbs from Xencor, anti- EGFrvlll MAbs from Abgenix, anti-HER2 MAb from Abiogen, AZD-2171 or AZD-9935 (AstraZeneca), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), CEP-5214 (Cephalon), CEP-7055 (Cephalon), cetuximab (ImClone Systems), CHIR-200131 and CHIR-258 (Chiron), CP-547632 (OSI Pharmaceuticals), CP-724714 (Pfizer), CT-301 (Creabilis Therapeutics), D- 69491 (Baxter International), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EGFR/ErbB2 inhibitors from Array BioPharma, erlotinib (CAS No. 183319- 69-9) (OSI Pharmaceuticals), EXEL-2880 (Exelixis), FK-778 (Sanofi- Aventis) , gefitinib (CAS No. 184475-35-2) (AstraZeneca), GW-2286 or GW- 654652 (GlaxoSmithKline), her2/neu antigen from AlphaVax, HER-2/neu inhibitor from Generex, Herzyme (Medipad) (Sirna Therapeutics), HKI-272 (Wyeth), HuMax-EGFr (Genmab), idronoxil (CAS No. 81267-65-4) (Novogen), IGF-1 inhibitors from Ontogen, IMC-11F8 (ImClone Systems), kahalalide F (CAS No. 149204-42-2) (PharmaMar), KDR inhibitor from LG Life Sciences, KDR inhibitors from Abbott Laboratories, KDR kinase inhibitors (UCB), Kdr kinase inhibitors from Merck & Co, KRN-633 and KRN- 951 (Kirin Brewery), KSB-102 (Xenova), lapatinib ditosylate (CAS No. 388082-78-8) (GlaxoSmithKline), matuzumab (Merck KGaA), MDX-214 (Medarex), ME-103 (Pharmexa), MED-A300 (Gerolymatos), MNAC-13 (Lay Line Genomics), nimotuzumab (Center of Molecular Immunology), NSC- 330507 or NSC-707545 (NIH), NV-50 (Novogen), OSI-930 (OSI Pharmaceuticals), panitumumab (Abgenix), pelitinib (CAS No. 287933-82-7) (Wyeth), pertuzumab (CAS No. 380610-27-5) (Genentech), Pharmaprojects No. 3985 (Sanofi-Aventis), prostate cancer therapeutics from Sequenom (SQPC35, SQPC36, SQPC90), removab and remoxab (Trion Pharma), RG- 13022 (CAS No. 136831-48-6), RG-13291 (CAS No. 138989-50-1), or RG- 14620 (CAS No. 136831-49-7) (Sanofi-Aventis), RM-6427 (Romark), RNAi breast cancer therapy from Benitec, RP 53801 (CAS No. 125882-88-4) (Sanofi-Aventis), sorafenib tosylate (Bayer), SU-11657 (Pfizer), Tie-2 antagonists from Semaia (Hybrigenics), Tie-2 inhibitors from Ontogen, trastuzumab (CAS No. 180288-69-1) (Genentech), tyrosine kinase inhibitors from Sanofi-Aventis, U3-1287, U3-1565, U3-1784, U3-1800 (U3 Pharma), vatalanib (Novartis), VEGFR-2 kinase inhibitor from Bristol-Myers Squibb, XL-647 (Exelixis), ZD-6474 (AstraZeneca), ZK-CDK (Schering AG), an EGFR tyrosine kinase inhibitor, EKB-569 (Wyeth), herbimycin A, and an analogue or derivative thereof,
108a) a VEGF inhibitor,
109a) a vitamin D receptor agonist,
110a) ZD-6474 (an angiogenesis inhibitor),
111a) AP-23573 (an mTOR inhibitor),
112a) synthadotin (a tubulin antagonist),
113a) S-0885 (a collagenase inhibitor),
114a) aplidine (an elongation factor-1 alpha inhibitor),
115a) ixabepilone (an epithilone),
116a) IDN-5390 (an angiogenesis inhibitor and an FGF inhibitor),
117a) SB-2723005 (an angiogenesis inhibitor),
118a) ABT-518 (an angiogenesis inhibitor), 119a) combretastatin (an angiogenesis inhibitor),
120a) anecortave acetate (an angiogenesis inhibitor),
121a) SB-715992 (a kinesin antagonist),
122a) temsirolimus (an mTOR inhibitor),
123a) adalimumab (a TNFα antagonist),
124a) erucylphosphocholine (an ATK inhibitor),
125a) alphastatin (an angiogenesis inhibitor),
126a) bortezomib (an NF Kappa B inhibitor),
127a) etanercept (a TNFα antagonist and TACE inhibitor),
128a) humicade (a TNFα inhibitor),
129a) gefitinib (a tyrosine kinase inhibitor),
130a) a histamine receptor antagonist selected from the group consisting of phenothiazines (e.g., promethazine), alkylamines (e.g., chlorpheniramine (CAS No. 7054-11-7), brompheniramine (CAS No. 980- 71-2), fexofenadine hydrochloride, promethazine hydrochloride, loratadine, ketotifen fumarate salt, and acrivastine), methylxanthines (e.g., theophylline, theobromine, and caffeine), cimetidine (available under the tradename TAGAMET from SmithKline Beecham Phamaceutical Co., Wilmington, DE), ranitidine (available under the tradename ZANTAC from Warner Lambert Company, Morris Plains, NJ), famotidine (available under the tradename PEPCID from Merck & Co., Whitehouse Station, NJ), nizatidine (available under the tradename AXID from Reliant Pharmaceuticals, Inc., Liberty Corner, NJ), nizatidine, and roxatidine acetate (CAS No. 78628-28-1), H3 receptor antagonists (e.g., thioperamide and thioperamide maleate salt), and anti-histamines (e.g., tricyclic dibenozoxepins, ethanolamines, ethylenediamines, piperizines, piperidines, and pthalazinones),
131a) an alpha adrenergic receptor antagonist,
132a) an anti-psychotic compound,
133a) a CaM kinase Il inhibitor,
134a) a G protein agonist, 135a) an antibiotic selected from the group consisting of apigenin (Cas No. 520-36-5), ampicillin sodium salt (CAS No. 69-52-3), puromycin, and an analogue or derivative thereof,
136a) an anti-microbial agent,
137a) a DNA topoisomerase inhibitor selected from the group consisting of β-lapachone (CAS No. 4707-32-8), (-)-arctigenin (CAS No. 7770-78-7), aurintricarboxylic acid, and an analogue or derivative thereof,
138a) a thromboxane A2 receptor inhibitor selected from the group consisting of BM-531 (CAS No. 284464-46-6), ozagrel hydrochloride (CAS No. 78712-43-3), and an analogue or derivative thereof,
139a) a D2 dopamine receptor antagonist,
140a) a Peptidyl-Prolyl Cis/Trans lsomerase Inhibitor,
141a) a dopamine antagonist, an anesthetic compound,
142a) a clotting factor,
143a) a lysyl hydrolase inhibitor,
144a) a muscarinic receptor inhibitor,
145a) a superoxide anion generator,
146a) a steroid,
147a) an anti-proliferative agent selected from the group consisting of silibinin (CAS No. 22888-70-6), silymarin (CAS No. 65666-07- 1), 1 ,2-hexanediol, dioctyl phthalate (CAS No. 117-81-7), zirconium (IV) oxide, glycyrrhizic acid, spermidine trihydrochloride, tetrahydrochloride, CGP 74514, spermine tetrahydrochloride, NG-methyl-L-arginine acetate salt, galardin, and an analogue or derivative thereof,
148a) a diuretic,
149a) an anti-coagulant,
150a) a cyclic GMP agonist,
151a) an adenylate cyclase agonist,
152a) an antioxidant,
153a) a nitric oxide synthase inhibitor, 154a) an anti-neoplastic agent selected from tirapazamine (CAS No. 27314-97-2), fludarabine (CAS No. 21679-14-1), cladribine, imatinib mesilate, and an analogue or derivative thereof,
155a) a DNA synthesis inhibitor,
156a) a DNA alkylating agent selected from dacarbazine (CAS No. 4342-03-4), temozolomide, procarbazine HCI, and an analogue or derivative thereof,
157a) a DNA methylation inhibitor,
158a) a NSAID agent,
159a) a peptidylglycine alpha-hydroxylating monooxygenase inhibitor,
160a) an MEK1/MEK 2 inhibitor,
161a) a NO synthase inhibitor,
162a) a retinoic acid receptor antagonist selected from isotretinoin (CAS No. 4759-48-2) and an analogue or derivative thereof, ~ 163a) an ACE inhibitor;
164a) a glycosylation inhibitor,
165a) an intracellular calcium influx inhibitor,
166a) an anti-emetic agent,
167a) an acetylcholinesterase inhibitor,
168a) an ALK-5 receptor antagonist,
169a) a RAR/RXT antagonist,
170a) an elF-2a inhibitor,
171a) an S-adenosyl-L-homocysteine hydrolase inhibitor,
172a) an estrogen agonist,
173a) a serotonin receptor inhibitor,
174a) an anti-thrombotic agent,
175a) a tryptase inhibitor,
176a) a pesticide,
177a) a bone mineralization promoter, 178a) a bisphosphonate compound selected from risedronate and an analogue or derivative thereof,
179a) an anti-inflammatory compound, 180a) a DNA methylation promoter, 181a) an anti-spasmodic agent, 182a) a protein synthesis inhibitor, 183a) an α-glucosidase inhibitor, 184a) a calcium channel blocker, 185a) a pyruvate dehydrogenase activator, 186a) a prostaglandin inhibitor, 187a) a sodium channel inhibitor, 188a) a serine protease inhibitor, 189a) an intracellular calcium flux inhibitor, 190a) a JAK2 inhibitor; 191a) an androgen inhibitor, ~ 192a) an aromatase inhibitor, 193a) an anti-viral agent, 194a) a 5-HT inhibitor, 195a) an FXR antagonist,
196a) an actin polymerization and stabilization promoter, 197a) an AXOR12 agonist, 198a) an angiotensin Il receptor agonist, 199a) a platelet aggregation inhibitor, 200a) a CB1/CB2 receptor agonist, 201a) a norepinephrine reuptake inhibitor, 202a) a selective serotonin reuptake inhibitor, 203a) a reducing agent, 204a) isotretinoin, 205a) radicicol, 206a) clobetasol propionate, 207a) homoharringtonine, 208a) trichostatin A, 209a) brefeldin A, 210a) thapsigargin, 211a) dolastatin 15, 212a) cerivastatin, 213a) jasplakinolide, 214a) herbimycin A, 215a) pirfenidone, 216a) vinorelbine, 217a) 17-DMAG, 218a) tacrolimus, 219a) loteprednol etabonate, 220a) juglone, 221a) prednisolone, 222a) pυromycin, 223a) 3-BAABE, 224a) cladribine, 225a) mannose-6-phosphate, 226a) 5-azacytidine, 227a) Ly333531 (ruboxistaurin), 228a) simvastatin, and
229a) an immuno-modulator selected from Bay 11-7085, (-)- arctigenin, idazoxan hydrochloride, and an analogue or derivative thereof.
As mentioned above, the present invention provides compositions comprising each of the foregoing 229 (i.e., 1a through 229a) listed anti-fibrotic agents or classes of anti-fibrotic agents, with each of the following 97 (i.e., 1b through 97b) polymers and compounds:
1b. A crosslinked polymer.
2b. A polymer that reacts with mammalian tissue.
3b. A polymer that is a naturally occurring polymer.
4b. A polymer that is a protein. 5b. A polymer that is a carbohydrate.
6b. A polymer that is biodegradable.
7b. A polymer that is crosslinked and biodegradable.
8b. A polymer that nonbiodegradable.
9b. Collagen.
10b. Methylated collagen.
11b. Fibrinogen.
12b. Thrombin.
13b. Albumin.
14b. Plasminogen.
15b. von Willebrands factor.
16b. Factor VIII.
17b. Hypoallergenic collagen.
18b. Atelopeptidic collagen.
19b. Telopeptide collagen.
20b. Crosslinked collagen.
21b. Aprotinin.
22b. Gelatin.
23b. A protein conjugate.
24b. A gelatin conjugate.
25b. Hyaluronic acid.
26b. A hyaluronic acid derivative.
27b. A synthetic polymer.
28b. A polymer formed from reactants comprising a synthetic isocyanate-containing compound.
29b. A synthetic isocyanate-containing compound.
30b. A polymer formed from reactants comprising a synthetic thiol-containing compound.
31 b. A synthetic thiol-containing compound.
32b. A polymer formed from reactants comprising a synthetic compound containing at least two thiol groups. 33b. A synthetic compound containing at least two thiol groups.
34b. A polymer formed from reactants comprising a synthetic compound containing at least three thiol groups.
35b. A synthetic compound containing at least three thiol groups.
36b. A polymer formed from reactants comprising a synthetic compound containing at least four thiol groups.
37b. A synthetic compound containing at least four thiol groups.
38b. A polymer formed from reactants comprising a synthetic amino-containing compound.
39b. A synthetic amino-containing compound.
40b. A polymer formed from reactants comprising a synthetic compound containing at least two amino groups.
41b. A synthetic compound containing at least two amino groups.
42b. A polymer formed from reactants comprising a synthetic compound containing at least three amino groups.
43b. A synthetic compound containing at least three amino groups.
44b. A polymer formed from reactants comprising a synthetic compound containing at least four amino groups.
45b. A synthetic compound containing at least four amino groups.
46b. A polymer formed from reactants comprising a synthetic compound comprising a carbonyl-oxygen-succinimidyl group.
47b. A synthetic compound comprising a carbonyl-oxygen- succinimidyl group.
48b. A polymer formed from reactants comprising a synthetic compound comprising at least two carbonyl-oxygen-succinimidyl groups. 49b. A synthetic compound comprising at least two carbonyl- oxygen-succinimidyl groups.
50b. A polymer formed from reactants comprising a synthetic compound comprising at least three carbonyl-oxygen-succinimidyl groups.
51b. A synthetic compound comprising at least three carbonyl-oxygen-succinimidyl groups.
52b. A polymer formed from reactants comprising a synthetic compound comprising at least four carbonyl-oxygen-succinimidyl groups.
53b. A synthetic compound comprising at least four carbonyl-oxygen-succinimidyl groups.
54b. A polymer formed from from reactants comprising a synthetic polyalkylene oxide-containing compound.
55b. A synthetic polyalkylene oxide-containing compound.
56b. A polymer formed from reactants comprising a synthetic compound comprising both polyalkylene oxide and biodegradable polyester blocks. - - - . _
57b. A synthetic compound comprising both polyalkylene oxide and biodegradable polyester blocks.
58b. A polymer formed from reactants comprising a synthetic polyalkylene oxide-containing compound having reactive amino groups.
59b. A synthetic polyalkylene oxide-containing compound having reactive amino groups.
60b. A polymer formed from reactants comprising a synthetic polyalkylene oxide-containing compound having reactive thiol groups.
61b. A synthetic polyalkylene oxide-containing compound having reactive thiol groups.
62b. A polymer formed from reactants comprising a synthetic polyalkylene oxide-containing compound having reactive carbonyl-oxygen- succinimidyl groups.
63b. A synthetic polyalkylene oxide-containing compound having reactive carbonyl-oxygen-succinimidyl groups. 64b. A polymer formed from reactants comprising a synthetic compound comprising a biodegradable polyester block.
65b. A synthetic compound comprising a biodegradable polyester block.
66b. A polymer formed from reactants comprising a synthetic polymer formed in whole or part from lactic acid or lactide.
67b. A synthetic polymer formed in whole or part from lactic acid or lactide.
68b. A polymer formed from reactants comprising a synthetic polymer formed in whole or part from glycolic acid or glycolide.
69b. A synthetic polymer formed in whole or part from glycolic acid or glycolide.
70b. A polymer formed from reactants comprising polylysine.
71b. Polylysine.
72b. A polymer formed from reactants comprising (a) protein and (b) a compound comprising a polyalkylene oxide portion.
73b. A polymer formed from reactants comprising (a) protein and (b) polylysine.
74b. A polymer formed from reactants comprising (a) protein and (b) a compound having at least four thiol groups.
75b. A polymer formed from reactants comprising (a) protein and (b) a compound having at least four amino groups.
76b. A polymer formed from reactants comprising (a) protein and (b) a compound having at least four carbonyl-oxygen-succinimide groups.
77b. A polymer formed from reactants comprising (a) protein and (b) a compound having a biodegradable region formed from reactants selected from lactic acid, lactide, glycolic acid, glycolide, and epsilon- caprolactone.
78b. A polymer formed from reactants comprising (a) collagen and (b) a compound comprising a polyalkylene oxide portion. 79b. A polymer formed from reactants comprising (a) collagen and (b) polylysine.
80b. A polymer formed from reactants comprising (a) collagen and (b) a compound having at least four thiol groups.
81b. A polymer formed from reactants comprising (a) collagen and (b) a compound having at least four amino groups.
82b. A polymer formed from reactants comprising (a) collagen and (b) a compound having at least four carbonyl-oxygen- succinimide groups.
83b. A polymer formed from reactants comprising (a) collagen and (b) a compound having a biodegradable region formed from reactants selected from lactic acid, lactide, glycolic acid, glycolide, and epsilon-caprolactone.
84b. A polymer formed from reactants comprising (a) methylated collagen and (b) a compound comprising a polyalkylene oxide portion.- -. . . . .
85b. A polymer formed from reactants comprising (a) methylated collagen and (b) polylysine.
86b. A polymer formed from reactants comprising (a) methylated collagen and (b) a compound having at least four thiol groups.
87b. A polymer formed from reactants comprising (a) methylated collagen and (b) a compound having at least four amino groups.
88b. A polymer formed from reactants comprising (a) methylated collagen and (b) a compound having at least four carbonyl- oxygen-succinimide groups.
89b. A polymer formed from reactants comprising (a) methylated collagen and (b) a compound having a biodegradable region formed from reactants selected from lactic acid, lactide, glycolic acid, glycolide, and epsilon-caprolactone.
90b. A polymer formed from reactants comprising hyaluronic acid. 91 b. A polymer formed from reactants comprising a hyaluronic acid derivative.
92b. A polymer formed from reactants comprising pentaerythritol poly(ethylene glycol)ether tetra-sulfhydryl of number average molecular weight between 3,000 and 30,000.
93b. Pentaerythritol poly(ethylene glycol)ether tetra- sulfhydryl of number average molecular weight between 3,000 and 30,000.
94b. A polymer formed from reactants comprising pentaerythritol poly(ethylene glycol)ether tetra-amino of number average molecular weight between 3,000 and 30,000.
95b. Pentaerythritol poly(ethylene glycol)ether tetra-amino of number average molecular weight between 3,000 and 30,000.
96b. A polymer formed from reactants comprising (a) a synthetic compound having a number average molecular weight between 3,000 and 30,000 and comprising a polyalkylene oxide region and multiple nucleophilic groups, and (b) a synthetic compound having a number average molecular weight between 3,000 and 30,000 and comprising a polyalkylene oxide region and multiple electrophilic groups.
97b. A mixture of (a) a synthetic compound having a number average molecular weight between 3,000 and 30,000 and comprising a polyalkylene oxide region and multiple nucleophilic groups, and (b) a synthetic compound having a number average molecular weight between 3,000 and 30,000 and comprising a polyalkylene oxide region and multiple electrophilic groups.
As mentioned above, the present invention provides compositions comprising each of the foregoing 229 (1a through 229a) listed anti-fibrotic agents or classes of anti-fibrotic agents, with each of the foregoing 97 (1b through 97b) polymers and compounds: Thus, in separate aspects, the invention provides 229 times 97 = 22213 described compositions. In other words, each of the following is a distinct aspect of the present invention: 1a+1b, 2a+1b, 3a+1b, 4a+1b, 5a+1b, 6a+1b, 7a+1b, 8a+1b, 9a+1b, 10a+1b, 11a+1b, 12a+1b, 13a+1b, 14a+1b, 15a+1b, 16a+1b, 17a+1b, 18a+1b, 19a+1b, 20a+1b, 21a+1b, 22a+1b, 23a+1b, 24a+1b, 25a+1b, 26a+1b, 27a+1b, 28a+1b, 29a+1b, 30a+1b, 31a+1b, 32a+1b, 33a+1b, 34a+1b, 35a+1b, 36a+1b, 37a+1b, 38a+1b, 39a+1b, 40a+1b, 41a+1b, 42a+1b, 43a+1b, 44a+1b, 45a+1b, 46a+1b, 47a+1b, 48a+1b, 49a+1b, 50a+1b, 51a+1b, 52a+1b, 53a+1b, 54a+1b, 55a+1b, 56a+1b, 57a+1b, 58a+1b, 59a+1b, 60a+1b, 61a+1b, 62a+1b, 63a+1b, 64a+1b, 65a+1b, 66a+1b, 67a+1b, 68a+1b, 69a+1b, 70a+1b, 71a+1b, 72a+1b, 73a+1b, 74a+1b, 75a+1b, 76a+1b, 77a+1b, 78a+1b, 79a+1b, 80a+1b, 81a+1b, 82a+1b, 83a+1b, 84a+1b, 85a+1b, 86a+1b, 87a+1b, 88a+1b, 89a+1b, 90a+1b, 91a+1b, 92a+1b, 93a+1b, 94a+1b, 95a+1b, 96a+1b, 97a+1b, 98a+1b, 99a+1b, 100a+1b, 101a+1b, 102a+1b, 103a+1b, 104a+1b, 105a+1b, 106a+1b, 107a+1b, 108a+1b, 109a+1b, 110a+1b, 111a+1b, 112a+1b, 113a+1b, 114a+1b, 115a+1b, 116a+1b, 117a+1b, 118a+1b, 119a+1b, 120a+1b, 121a+1b, 122a+1b, 123a+1b, 124a+1b, 125a+1b, 126a+1b, 127a+1b, 128a+1b, 129a+1b, 130a+1b, 131a+1b, 132a+1b, 133a+1b, 134a+1b, 135a+1b, 136a+1b, 137a+1b, 138a+1b, 139a+1b, 140a+1b, 141a+1b, 142a+1b, 143a+1b, 144a+1b, 145a+1b, 146a+1b, 147a+1b, 148a+1b, 149a+1b, 150a+1b, 151a+1b, 152a+1b, 153a+1b, 154a+1b, 155a+1b, 156a+1b, 157a+1b, 158a+1b, 159a+1b, 160a+1b, 161a+1b, 162a+1b, 163a+1b, 164a+1b, 165a+1b, 166a+1b, 167a+1b, 168a+1b, 169a+1b, 170a+1b, 171a+1b, 172a+1b, 173a+1b, 174a+1b, 175a+1b, 176a+1b, 177a+1b, 178a+1b, 179a+1b, 180a+1b, 181a+1b, 182a+1b, 183a+1b, 184a+1b, 185a+1b, 186a+1b, 187a+1b, 188a+1b, 189a+1b, 190a+1b, 191a+1b, 192a+1b, 193a+1b, 194a+1b, 95a+1b, 96a+1b, 97a+1b, 98a+1b, 99a+1b, 200a+1b, 201a+1b, 202a+1b, 203a+1b, 204a+1b, 205a+1b, 206a+1b, 207a+1b, 208a+1b, 209a+1b, 210a+1b, 211a+1b, 212a+1b, 213a+1b, 214a+1b, 215a+1b, 216a+1b, 217a+1b, 218a+1b, 219a+1b, 220a+1b, 221a+1b, 222a+1b, 223a+1b, 224a+1b, 225a+1b, 226a+1b, 227a+1b, 228a+1b, 229a+1b, 1a+2b, 2a+2b, 3a+2b, 4a+2b, 5a+2b, 6a+2b, 7a+2b, 8a+2b, 9a+2b, 10a+2b, 11a+2b, 12a+2b, 13a+2b, 14a+2b, 15a+2b, 16a+2b, 17a+2b, 18a+2b, 19a+2b, 20a+2b, 21a+2b, 22a+2b, 23a+2b, 24a+2b, 25a+2b, 26a+2b, 27a+2b, 28a+2b, 29a+2b, 30a+2b, 31a+2b, 32a+2b, 33a+2b, 34a+2b, 35a+2b, 36a+2b, 37a+2b, 38a+2b, 39a+2b, 40a+2b, 41a+2b, 42a+2b, 43a+2b, 44a+2b, 45a+2b, 46a+2b, 47a+2b, 48a+2b, 49a+2b, 50a+2b, 51a+2b, 52a+2b, 53a+2b, 54a+2b, 55a+2b, 56a+2b, 57a+2b, 58a+2b, 59a+2b, 60a+2b, 61a+2b, 62a+2b, 63a+2b, 64a+2b, 65a+2b, 66a+2b, 67a+2b, 68a+2b, 69a+2b, 70a+2b, 71a+2b, 72a+2b, 73a+2b, 74a+2b, 75a+2b, 76a+2b, 77a+2b, 78a+2b, 79a+2b, 80a+2b, 81a+2b, 82a+2b, 83a+2b, 84a+2b, 85a+2b, 86a+2b, 87a+2b, 88a+2b, 89a+2b, 90a+2b, 91a+2b, 92a+2b, 93a+2b, 94a+2b, 95a+2b, 96a+2b, 97a+2b, 98a+2b, 99a+2b, 100a+2b, 101a+2b, 102a+2b, 103a+2b, 104a+2b, 105a+2b, 106a+2b, 107a+2b, 108a+2b, 109a+2b, 110a+2b, 111a+2b, 112a+2b, 113a+2b, 114a+2b, 115a+2b, 116a+2b, 117a+2b, 118a+2b, 119a+2b, 120a+2b, 121a+2b, 122a+2b, 123a+2b, 124a+2b, 125a+2b, 126a+2b, 127a+2b, 128a+2b, 129a+2b, 130a+2b, 131a+2b, 132a+2b, 133a+2b, 134a+2b, 135a+2b, 136a+2b, 137a+2b, 138a+2b, 139a+2b, 140a+2b, 141a+2b, 142a+2b, 143a+2b, 144a+2b, 145a+2b, 146a+2b, 147a+2b, 148a+2b, 149a+2b, 150a+2b, 151a+2b, 152a+2b, 153a+2b, 154a+2b, 155a+2b, 156a+2b, 157a+2b, 158a+2b, 159a+2b, 160a+2b, 161a+2b, 162a+2b, 163a+2b, 164a+2b, 165a+2b, 166a+2b, 167a+2b, 168a+2b, 169a+2b, 170a+2b, 171a+2b, 172a+2b, 173a+2b, 174a+2b, 175a+2b, 176a+2b, 177a+2b, 178a+2b, 179a+2b, 180a+2b, 181a+2b, 182a+2b, 183a+2b, 184a+2b, 185a+2b, 186a+2b, 187a+2b, 188a+2b, 189a+2b, 190a+2b, 191a+2b, 192a+2b, 193a+2b, 194a+2b, 95a+2b, 96a+2b, 97a+2b, 98a+2b, 99a+2b, 200a+2b, 201a+2b, 202a+2b, 203a+2b, 204a+2b, 205a+2b, 206a+2b, 207a+2b, 208a+2b, 209a+2b, 210a+2b, 211a+2b, 212a+2b, 213a+2b, 214a+2b, 215a+2b, 216a+2b, 217a+2b, 218a+2b, 219a+2b, 220a+2b, 221a+2b, 222a+2b, 223a+2b, 224a+2b, 225a+2b, 226a+2b, 227a+2b, 228a+2b, 229a+2b, etc.
Compositions That Comprise Additional Therapeutic Agents
In addition to incorporation of the above-mentioned therapeutic agents (Ae., anti-infective agents or fibrosis-inhibiting agents), one or more other pharmaceutically active agents can be incorporated into the present compositions to improve or enhance efficacy. In one aspect, the composition may further include a compound which acts to have an inhibitory effect on pathological processes in or around the treatment site. Representative examples of additional therapeutically active agents include, by way of example and not limitation, anti-thrombotic agents, antiproliferative agents, anti-inflammatory agents, neoplastic agents, enzymes, receptor antagonists or agonists, hormones, antibiotics, antimicrobial agents, antibodies, cytokine inhibitors, IMPDH (inosine monophosplate dehydrogenase) inhibitors tyrosine kinase inhibitors, MMP inhibitors, p38 MAP kinase inhibitors, immunosuppressants, apoptosis antagonists, caspase inhibitors, and JNK inhibitors.
In certain embodiments, the composition may further include an antithrombotic agent and/or antiplatelet agent and/or a thrombolytic agent, which reduces the likelihood of thrombotic events upon implantation of a medical implant. Representative examples of anti-thrombotic and/or antiplatelet and/or thrombolytic agents include heparin, heparin fragments, organic salts of heparin, heparin complexes (e.g., benzalkonium heparinate, tridodecylammonium heparinate), dextran, sulfonated carbohydrates such as dextran sulfate, Coumadin, coumarin, heparinoid, danaparoid, argatroban chitosan sulfate, chondroitin sulfate, danaparoid, lepirudin, hirudin, AMP, adenosine, 2-chloroadenosine, acetylsalicylic acid, phenylbutazone, indomethacin, meclofenamate, hydrochloroquine, dipyridamole, iloprost, streptokinase, factor Xa inhibitors, such as DX9065a, magnesium, and tissue plasminogen activator. Further examples include plasminogen, lys- plasminogen, alpha-2-antiplasmin, urokinase, aminocaproic acid, ticlopidine, clopidogrel, trapidil (triazolopyrimidine), naftidrofuryl, auriritricarboxylic acid and glycoprotein llb/llla inhibitors such as abcixamab, eptifibatide, and tirogiban. Other agents capable of affecting the rate of clotting include glycosaminoglycans, danaparoid, 4-hydroxycourmarin, warfarin sodium, dicumarol, phenprocoumon, indan-1 ,3-dione, acenocoumarol, anisindione, and rodenticides including bromadiolone, brodifacoum, diphenadione, chlorophacinone, and pidnone.
The polymeric formulation may further include an agent from one of the following classes of compounds: anti-inflammatory agents (e.g., dexamethasone, cortisone, fludrocortisone, prednisone, prednisolone, 6α- methylprednisolone, triamcinolone, betamethasone, and aspirin); MMP inhibitors (e.g., batimistat, marimistat, TIMP's representative examples of which are included in U.S. Patent Nos. 5,665,777; 5,985,911 ; 6,288,261; 5,952,320; 6,441 ,189; 6,235,786; 6,294,573; 6,294,539; 6,563,002; - 6,071 ;903; 6,358,980; 5,852,213; 6,124,502; 6,160,132; 6,197,791; 6,172,057; 6,288,086; 6,342,508; 6,228,869; 5,977,408; 5,929,097; 6,498,167; 6,534,491; 6,548,524; 5,962,481 ; 6,197,795; 6,162,814; 6,441 ,023; 6,444,704; 6,462,073; 6,162,821 ; 6,444,639; 6,262,080; 6,486,193; 6,329,550; 6,544,980; 6,352,976; 5,968,795; 5,789,434; 5,932,763; 6,500,847; 5,925,637; 6,225,314; 5,804,581; 5,863,915; 5,859,047; 5,861 ,428; 5,886,043; 6,288,063; 5,939,583; 6,166,082; 5,874,473; 5,886,022; 5,932,577; 5,854,277; 5,886,024; 6,495,565; 6,642,255; 6,495,548; 6,479,502; 5,696,082; 5,700,838; 6,444,639; 6,262,080; 6,486,193; 6,329,550; 6,544,980; 6,352,976; 5,968,795; 5,789,434; 5,932,763; 6,500,847; 5,925,637; 6,225,314; 5,804,581; 5,863,915; 5,859,047; 5,861 ,428; 5,886,043; 6,288,063; 5,939,583; 6,166,082; 5,874,473; 5,886,022; 5,932,577; 5,854,277; 5,886,024; 6,495,565; 6,642,255; 6,495,548; 6,479,502; 5,696,082; 5,700,838; 5,861 ,436; 5,691 ,382; 5,763,621 ; 5,866,717; 5,902,791; 5,962,529; 6,017,889; 6,022,873; 6,022,898; 6,103,739; 6,127,427; 6,258,851; 6,310,084; 6,358,987; 5,872,152; 5,917,090; 6,124,329; 6,329,373; 6,344,457; 5,698,706; 5,872,146; 5,853,623; 6,624,144; 6,462,042; 5,981 ,491 ; 5,955,435; 6,090,840; 6,114,372; 6,566,384; 5,994,293; 6,063,786; 6,469,020; 6,118,001 ; 6,187,924; 6,310,088; 5,994,312; 6,180,611 ; 6,110,896; 6,380,253; 5,455,262; 5,470,834; 6,147,114; 6,333,324; 6,489,324; 6,362,183; 6,372,758; 6,448,250; 6,492,367; 6,380,258; 6,583,299; 5,239,078; 5,892,112; 5,773,438; 5,696,147; 6,066,662; 6,600,057; 5,990,158; 5,731 ,293; 6,277,876; 6,521 ,606; 6,168,807; 6,506,414; 6,620,813; 5,684,152; 6,451 ,791 ; 6,476,027; 6,013,649; 6,503,892; 6,420,427; 6,300,514; 6,403,644; 6,177,466; 6,569,899; 5,594,006; 6,417,229; 5,861 ,510; 6,156,798; 6,387,931 ; 6,350,907; 6,090,852; 6,458,822; 6,509,337; 6,147,061 ; 6,114,568; 6,118,016; 5,804,593; 5,847,153; 5,859,061 ; 6,194,451 ; 6,482,827; 6,638,952; 5,677,282; 6,365,630; 6,130,254; 6,455,569; 6,057,369; 6,576,628; 6,110,924; 6,472,396; 6,548,667; 5,618,844; 6,495,578; 6,627,411 ; 5,514,716; 5,256,657; 5,773,428; 6,037,472; 6,579,890; 5,932,595; 6,013,792; 6,420,415; 5,532,265; 5,639,746; 5,672,598; 5,830,915; 6,630,516; 5,324,634; 6,277,061 ; 6,140,099; 6,455,570; 5,595,885; 6,093,398; 6,379,667; 5,641 ,636; 5,698,404; 6,448,058; 6,008,220; 6,265,432; 6,169,103; 6,133,304; 6,541 ,521 ; 6,624,196; 6,307,089; 6,239,288; 5,756,545; 6,020,366; 6,117,869; 6,294,674; 6,037,361; 6,399,612; 6,495,568; 6,624,177; 5,948,780; 6,620,835; 6,284,513; 5,977,141 ; 6,153,612; 6,297,247; 6,559,142; 6,555,535; 6,350,885; 5,627,206; 5,665,764; 5,958,972; 6,420,408; 6,492,422; 6,340,709; 6,022,948; 6,274,703; 6,294,694; 6,531 ,499; 6,465,508; 6,437,177; 6,376,665; 5,268,384; 5,183,900; 5,189,178; 6,511 ,993; 6,617,354; 6,331 ,563; 5,962,466; 5,861 ,427; 5,830,869; and 6,087,359), cytokine inhibitors (chlorpromazine, mycophenolic acid, rapamycin, 1α- hydroxy vitamin D3), IMPDH (inosine monophosplate dehydrogenase) inhibitors (e.g., mycophenolic acid, ribaviran, aminothiadiazole, thiophenfurin, tiazofurin, viramidine) (Representative examples are included in U.S. Patent, Nos. 5,536,747; 5,807,876; 5,932,600; 6,054,472; 6,128,582; 6,344,465; 6,395,763; 6,399,773; 6,420,403; 6,479,628; 6,498,178; 6,514,979; 6,518,291 ; 6,541 ,496; 6,596,747; 6,617,323; and 6,624,184, U.S. Patent Application Nos. 2002/0040022A1 , 2002/0052513A1 , 2002/0055483A1 , 2002/0068346A1 , 2002/0111378A1 , 2002/0111495A1 , 2002/0123520A1 , 2002/0143176A1 , 2002/0147160A1 , 2002/0161038A1 , 2002/0173491 A1 , 2002/0183315A1 , 2002/0193612A1 , 2003/0027845A1 , 2003/0068302A1 , 2003/0105073A1 , 2003/0130254A1, 2003/0143197A1 , 2003/0144300A1 , 2003/0166201 A1 , 2003/0181497A1 , 2003/0186974A1, 2003/0186989A1 , and 2003/0195202A1 , and PCT Publication Nos. WO 00/24725A1 , WO 00/25780A1 , WO 00/26197A1 , WO 00/51615A1 , WO 00/56331 A1 , WO 00/73288A1 , WO 01/00622A1 , WO 01/66706A1 , WO 01/79246A2, WO 01/81340A2, WO 01/85952A2, WO 02/16382A1 , WO 02/18369A2, WO 02/051814A1 , WO 02/057287A2, WO 02/057425A2, WO 02/060875A1 , WO 02/060896A1 , WO 02/060898A1 , WO 02/068058A2, WO 03/020298A1 , WO 03/037349A1 , WO 03/039548A1 , WO 03/045901A2, WO 03/047512A2, WO 03/053958A1 , WO 03/055447A2, WO 03/059269A2, WO 03/063573A2, WO 03/087071 A1, WO 99/001545A1, WO 97/40028A1 , WO 97/41211A1, WO 98/40381A1 , and WO 99/55663A1), p38 MAP kinase inhibitors (MAPK) (e.g., GW-2286, CGP-52411 , BIRB-798, SB220025, RO- 320-1195, RWJ-67657, RWJ-68354, SCIO-469) (Representative examples are included in U.S. Patent Nos. 6,300,347; 6,316,464; 6,316,466; 6,376,527; 6,444,696; 6,479,507; 6,509,361 ; 6,579,874, and 6,630,485, and U.S. Patent Application Publication Nos. 2001/0044538A1, 2002/0013354A1 , 2002/0049220A1 , 2002/0103245A1 , 2002/0151491 A1 , 2002/0156114A1 , 2003/0018051 A1 , 2003/0073832A1 , 2003/0130257A1 , 2003/0130273A1 , 2003/0130319A1 , 2003/0139388A1 , 2003/0139462A1, 2003/0149031A1 , 2003/0166647A1 , and 2003/0181411A1, and PCT Publication Nos. WO 00/63204A2, WO 01/21591A1 , WO 01/35959A1, WO 01/74811A2, WO 02/18379A2, WO 02/064594A2, WO 02/083622A2, WO 02/094842A2,WO 02/096426A1 , WO 02/101015A2, WO 02/103000A2, WO 03/008413A1 , WO 03/016248A2, WO 03/020715A1 , WO 03/024899A2, WO 03/031431 A1, WO 03/040103A1 , WO 03/053940A1 , WO 03/053941 A2, WO 03/063799A2, WO 03/079986A2, WO 03/080024A2, WO 03/082287A1 , WO 97/44467A1 , WO 99/01449A1 , and WO 99/58523A1), and immunomodulatory agents (rapamycin, everolimus, ABT-578, azathioprine azithromycin, analogues of rapamycin, including tacrolimus and derivatives thereof (e.g., EP 0184162B1 and those described in U.S. Patent No. 6,258,823) and everolimus and derivatives thereof (e.g., U.S. Patent No. 5,665,772). Further representative examples of sirolimus analogues and derivatives include ABT-578 and those found in PCT Publication Nos. WO 97/10502, WO 96/41807, WO 96/35423, WO 96/03430, WO 96/00282, WO 95/16691, WO 95/15328, WO 95/07468, WO 95/04738, WO 95/04060, WO 94/25022, WO 94/21644, WO 94/18207, WO 94/10843, WO 94/09010, WO 94/04540, WO 94/02485, WO 94/02137, WO 94/02136, WO 93/25533, WO 93/18043, WO 93/13663, WO 93/11130, WO 93/10122, WO 93/04680, WO 92/14737, and WO 92/05179 and in U.S. Patent Nos. 6,342,507; 5,985,890; 5,604,234; 5,597,715; 5,583,139; 5,563,172; 5,561 ,228; 5,561 ,137; 5,541,193; 5,541 ,189; 5,534,632; 5,527,907; 5,484,799; 5,457,194; 5,457,182; 5,362,735; 5,324,644; 5,318,895; 5,310,903; 5,310,901; 5,258,389; 5,252,732; 5,247,076; 5,225,403; 5,221 ,625; 5,210,030; 5,208,241 ; 5,200,411 ; 5,198,421 ; 5,147,877; 5,140,018; 5,116,756; 5,109,112; 5,093,338; and 5,091 ,389.
Other examples of biologically active agents which may be included in the compositions of the invention include tyrosine kinase inhibitors, such as imantinib, ZK-222584, CGP-52411 , CGP-53716, NVP- AAK980-NX, CP-127374, CP-564959, PD-171026, PD-173956, PD-180970, SU-0879, and SKI-606; MMP inhibitors such as nimesulide, PKF-241-466, PKF-242-484, CGS-27023A, SAR-943, primomastat, SC-77964, PNU- 171829, AG-3433, PNU-142769, SU-5402, and dexlipotam; p38 MAP kinase inhibitors such as include CGH-2466 and PD-98-59; immunosuppressants such as argyrin B, macrocyclic lactone, ADZ-62-826, CCI-779, tilomisole, amcinonide, FK-778, AVE-1726, and MDL-28842; cytokine inhibitors such as TNF-484A, PD-172084, CP-293121 , CP-353164, and PD-168787; NFKB inhibitors, such as, AVE-0547, AVE-0545, and IPL- 576092; HMGCoA reductase inhibitors, such as, pravestatin, atorvastatin, fluvastatin, dalvastatin, glenvastatin, pitavastatin, CP-83101 , U-20685; apoptosis antagonist (e.g., troloxamine, TCH-346 (N-methyl-N-propargyl-10- aminomethyl-dibenzo(b,f)oxepin); and caspase inhibitors (e.g., PF-5901 (benzenemethanol, alpha-pentyl-3-(2-quinolinylmethoxy)-), and JNK inhibitors (e.g., AS-602801).
In another aspect, the composition may further include an antibiotic (e.g., amoxicillin, trimethoprim-sulfamethoxazole, azithromycin, clarithromycin, amoxicillin-clavulanate, cefprozil, cefuroxime, cefpodoxime, or cefdinir).
In certain aspects, a polymeric composition comprising a fibrosis-inhibiting agent is combined with an agent that can modify metabolism of the agent In vivo to enhance efficacy of the fibrosis-inhibiting agent. One class of therapeutic agents that can be used to alter drug metabolism includes agents capable of inhibiting oxidation of the anti- scarring agent by cytochrome P450 (CYP). In one embodiment, compositions are provided that include a fibrosis-inhibiting agent (e.g., ZD- 6474, AP-23573, synthadotin, S-0885, aplidine, Ixabepilone, IDN-5390, SB- 2723005, ABT-518, combretastatin, anecortave acetate, SB-715992, temsirolimus, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, rapamycin, everolimus, isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, dolastatin 15, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17-DMAG, tacrolimus, loteprednol etabonate, juglone, prednisolone, puromycin, 3-BAABE, cladribine, mannose-6-phosphate, 5- azacytidine, Ly333531 (ruboxistaurin), and simvastatin) and a CYP inhibitor, which may be combined (e.g., coated) with any of the devices described herein, including, without limitation, stents, grafts, patches, valves, wraps, and films. Representative examples of CYP inhibitors include flavones, azole antifungals, macrolide antibiotics, HIV protease inhibitors, and anti- sense oligomers. Devices comprising a combination of a fibrosis-inhibiting agent and a CYP inhibitor may be used to treat a variety of proliferative conditions that can lead to undesired scarring of tissue, including intimal hyperplasia, surgical adhesions, and tumor growth.
In another aspect, a composition comprising an anti-infective agent {e.g., anthracyclines {e.g., doxorubicin or mitoxantrone), fluoropyrimidines (e.g., 5-fluorouracil), folic acid antagonists (e.g., methotrexate), quinolone antibacterial agents, and/or podophylotoxins (e.g., etoposide)) can be combined with traditional antibiotic and/or antifungal agents to enhance efficacy. The anti-infective agent may be further combined with anti-thrombotic and/or antiplatelet agents (for example, heparin, dextran sulfate, danaparoid, lepirudin, hirudin, AMP, adenosine, 2- chloroadenosine, aspirin, phenylbutazone, indomethacin, meclofenamate, hydrochloroquine, dipyridamole, iloprost, ticlopidine, clopidogrel, abcixamab, eptifibatide, tirofiban, streptokinase, and/or tissue plasminogen activator) to enhance efficacy.
Although the above therapeutic agents have been provided for the purposes of illustration, it should be understood that the present invention is not so limited. For example, although agents are specifically referred to above, the present invention should be understood to include analogues, derivatives and conjugates of such agents. As an illustration, ZD-6474, AP-23573, synthadotin, S-0885, aplidine, ixabepilone, IDN-5390, SB-2723005, ABT-518, combretastatin, anecortave acetate, SB-715992, temsirolimus, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, dolastatin 15, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17- DMAG, tacrolimus, loteprednol etabonate, juglone, prednisolone, puromycin, 3-BAABE, cladribine, mannose-6-phosphate, 5-azacytidine, Ly333531 (ruboxistaurin), and simvastatinshould be understood to refer to not only the common chemically available form of ZD-6474, AP-23573, synthadotin, S-0885, aplidine, ixabepilone, IDN-5390, SB-2723005, ABT- 518, combretastatin, anecortave acetate, SB-715992, temsirolimus, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, dolastatin 15, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17-DMAG, tacrolimus, loteprednol etabonate, juglone, prednisolone, puromycin, 3-BAABE, cladribine, mannose-6-phosphate, 5-azacytidine, Ly333531 (ruboxistaurin), and simvastatin, but analogues, derivatives or conjugates (e.g., -PEG, - Dextran, -xylos conjugates) of the aforementioned compounds. In addition, as will be evident to one of skill in the art, although the agents set forth above may be noted within the context of one class, many of the agents listed in fact have multiple biological activities. Further, more than one therapeutic agent may be utilized ata time (i.e., in combination), or delivered sequentially.
Compositions That Comprise Additional Components
Within certain embodiments of the invention, the composition can also comprise radio-opaque, echogenic materials and magnetic resonance imaging (MRI) responsive materials (i.e., MRI contrast agents) to aid in visualization of the composition under ultrasound, fluoroscopy and/or MRI. For example, a composition may be echogenic or radiopaque (e.g., made with echogenic or radiopaque with materials such as powdered tantalum, tungsten, barium carbonate, bismuth oxide, barium sulfate, metrazimide, iopamidol, iohexol, iopromide, iobitridol, iomeprol, iopentol, ioversol, ioxilan, iodixanol, iotrolan, acetrizoic acid derivatives, diatrizoic acid derivatives, iothalamic acid derivatives, ioxithalamic acid derivatives, metrizoic acid derivatives, iodamide, lypophylic agents, iodipamide and ioglycamic acid or, by the addition of microspheres or bubbles which present an acoustic interface). For visualization under MRI1 contrast agents (e.g., gadolinium (III) chelates or iron oxide compounds) may be incorporated into the composition.
The compositions may, alternatively, or in addition, be visualized under visible light, using fluorescence, or by other spectroscopic means. Visualization agents that can be included for this purpose include dyes, pigments, and other colored agents. In one aspect, the composition may further include a colorant to improve visualization of the composition in vivo and/or ex vivo. Frequently, compositions can be difficult to visualize upon delivery into a host, especially at the margins of an implant or tissue. A coloring agent can be incorporated into a composition to reduce or eliminate the incidence or severity of this problem. The coloring agent provides a unique color, increased contrast, or unique fluorescence characteristics to the composition. In one aspect, a composition is provided that includes a colorant such that it is readily visible (under visible light or using a fluorescence technique) and easily differentiated from its implant site. In another aspect, a colorant can be included in a liquid or semi-solid composition. For example, a single component of a two component mixture may be colored, such that when combined ex-vivo or in-vivo, the mixture is sufficiently colored.
The coloring agent may be, for example, an endogenous compound (e.g., an amino acid or vitamin) or a nutrient or food material and may be a hydrophobic or a hydrophilic compound. Preferably, the colorant has a very low or no toxicity at the concentration used. Also preferred are colorants that are safe and normally enter the body through absorption such as β-carotene. Representative examples of colored nutrients (under visible light) include fat soluble vitamins such as Vitamin A (yellow); water soluble vitamins such as vitamin B12 (pink-red) and folic acid (yellow-orange); carotenoids such as β-carotene (yellow-purple) and lycopene (red). Other examples of coloring agents include natural product (berry and fruit) extracts such as anthrocyanin (purple) and saffron extract (dark red). The coloring agent may be a fluorescent or phosphorescent compound such as α- tocopherolquinol (a vitamin E derivative) or L-tryptophan.
In one aspect, the compositions of the present invention include one or more coloring agents, also referred to as dyestuffs, which will be present in an effective amount to impart observable coloration to the composition, e.g., the gel. Examples of coloring agents include dyes suitable for food such as those known as F. D. & C. dyes and natural coloring agents such as grape skin extract, beet red powder, beta carotene, annato, carmine, turmeric, paprika, and so forth. Derivatives, analogues, and isomers of any of the above colored compound also may be used. The method for incorporating a colorant into an implant or therapeutic composition may be varied depending on the properties of and the desired location for the colorant. For example, a hydrophobic colorant may be selected for hydrophobic matrices. The colorant may be incorporated into a carrier matrix, such as micelles. Further, the pH of the environment may be ^controlled to further control the color and intensity.
In one aspect, the compositions of the present invention include one or more preservatives or bacteriostatic agents present in an effective amount to preserve the composition and/or inhibit bacterial growth in the composition, for example, bismuth tribromophenate, methyl hydroxybenzoate, bacitracin, ethyl hydroxybenzoate, propyl hydroxybenzoate, erythromycin, chlorocresol, benzalkonium chlorides, and the like. Examples of the preservative include paraoxybenzoic acid esters, chlorobutanol, benzylalcohol, phenethyl alcohol, dehydroacetic acid, sorbic acid, etc. In one aspect, the compositions of the present invention include one or more bactericidal (also known as bacteriacidal) agents.
In one aspect, the compositions of the present invention include one or more antioxidants, present in an effective amount. Examples of the antioxidant include sulfites, alpha-tocopherol, beta-carotene and ascorbic acid. Characteristics of Certain Compositions
In certain embodiments, compositions of the present invention may have a stable shelf-life of at least several months and capable of being produced and maintained under sterile conditions. The composition may be sterile either by preparing them under aseptic environment and/or they may be terminally sterilized using methods known in the art. A combination of both of these methods may also be used to prepare the composition in the sterile form. Sterilization may also occur by terminally using gamma radiation or electron beam sterilization methods.
In one aspect, the compounds and compositions of the present invention are sterile. Many pharmaceuticals are manufactured to be sterile and this criterion is defined by the USP XXII <1211>. The term "USP" refers to U.S. Pharmacopeia (see www.usp.org, Rockville, MD). Sterilization in this embodiment may be accomplished by a number of means accepted in the industry and listed in the USP XXII <1211>, including gas sterilization, ionizing radiation or, when appropriate, filtration. Sterilization may be maintained by what is termed asceptic processing, defined also in USP XXII <1211>. Acceptable gases used for gas sterilization include ethylene oxide. Acceptable radiation types used for ionizing radiation methods include gamma, for instance from a cobalt 60 source and electron beam. A typical dose of gamma radiation is 2.5 MRad. Filtration may be accomplished using a filter with suitable pore size, for example 0.22 μm and of a suitable material, for instance polytetrafluoroethylene (e.g., TEFLON from E. I. DuPont De Nemours and Company, Wilmington, DE).
In another aspect, the compositions of the present invention are contained in a container that allows them to be used for their intended purpose, i.e., as a pharmaceutical composition. Properties of the container that are important are a volume of empty space to allow for the addition of a constitution medium, such as water or other aqueous medium, e.g., saline, acceptable light transmission characteristics in order to prevent light energy from damaging the composition in the container (refer to USP XXII <661>), an acceptable limit of extractables within the container material (refer to USP XXII), an acceptable barrier capacity for moisture (refer to USP XXII <671>) or oxygen. In the case of oxygen penetration, this may be controlled by including in the container, a positive pressure of an inert gas, such as high purity nitrogen, or a noble gas, such as argon.
Typical materials used to make containers for pharmaceuticals include USP Type I through III and Type NP glass (refer to USP XXII <661>), polyethylene, TEFLON, silicone, and gray-butyl rubber. For parenterals, USP Types I to III glass and polyethylene are preferred.
Within certain aspects of the present invention, the therapeutic composition should be biocompatible, and release one or more fibrosis- inhibiting agents and/or anti-infective agents over a period of several hours, days, or, months. As described above, "release of an agent" refers to any statistically significant presence of the agent, or a subcomponent thereof, which has disassociated from the compositions and/or remains active on the surface of (or within) the composition. The compositions of the present invention may release the anti-scarring agent at one or more phases, the one or more phases having similar or different performance (e.g., release) profiles. The therapeutic agent may be made available to the tissue at amounts which may be sustainable, intermittent, or continuous; in one or more phases; and/or rates of delivery; effective to reduce or inhibit any one or more components of fibrosis (or scarring), including: formation of new blood vessels (angiogenesis), migration and proliferation of connective tissue cells (such as fibroblasts or smooth muscle cells), deposition of extracellular matrix (ECM), and remodeling (maturation and organization of the fibrous tissue).
Thus, release rate may be programmed to impact fibrosis (or scarring) by releasing an anti-scarring agent at a time such that at least one of the components of fibrosis is inhibited or reduced. Moreover, the predetermined release rate may reduce agent loading and/or concentration as well as potentially providing minimal drug washout and thus, increases efficiency of drug effect. Any one of the at least one anti-scarring agents may perform one or more functions, including inhibiting the formation of new blood vessels (angiogenesis), inhibiting the migration and proliferation of connective tissue cells (such as fibroblasts or smooth muscle cells), inhibiting the deposition of extracellular matrix (ECM), and inhibiting remodeling (maturation and organization of the fibrous tissue). In one embodiment, the rate of release may provide a sustainable level of the anti- scarring agent to the susceptible tissue site. In another embodiment, the rate of release is substantially constant. The rate may decrease and/or increase over time, and it may optionally include a substantially non-release period. The release rate may comprise a plurality of rates. In an embodiment, the plurality of release rates may include rates selected from the group consisting of substantially constant, decreasing, increasing, substantially non-releasing.
The total amount of anti-scarring agent made available on, in or near the device may be in an amount ranging from about 0.01 μg (micrograms) to about 2500 mg (milligrams). Generally, the anti-scarring agent may be in the amount ranging from 0.01 μg to about 10 μg; or from 10 μg to about 1 mg; or from 1 mg to about 10 mg; or from 10 mg to about 100 mg; or from 100 mg to about 500 mg; or from 500 mg to about 2500 mg.
The total surface amount of anti-scarring agent on, in or near the device may be in an amount ranging from less than 0.01 μg to about 2500 μg per mm2 of device surface area. Generally, the anti-scarring agent may be in the amount ranging from less than 0.01 μg; or from 0.01 μg to about 10 μg; or from 10 μg to about 250 μg; or from 250 μg to about 2500 μg,
The anti-scarring agent that is on, in or near the device may be released from the composition in a time period that may be measured from the time of implantation, which ranges from about less than 1 day to about 180 days. Generally, the release time may also be from about less than 1 day to about 7 days; from 7 days to about 14 days; from 14 days to about 28 days; from 28 days to about 56 days; from 56 days to about 90 days; from 90 days to about 180 days.
The amount of anti-scarring agent released from the composition as a function of time may be determined based on the in vitro release characteristics of the agent from the composition. The in vitro release rate may be determined by placing the anti-scarring agent within the composition or device in an appropriate buffer such as 0.1 M phosphate buffer (pH 7.4)) at 370C. Samples of the buffer solution are then periodically removed for analysis by HPLC, and the buffer is replaced to avoid any saturation effects.
Based on the in vitro release rates, the release of anti-scarring agent per day may range from an amount ranging from about 0.01 μg (micrograms) to about 2500 mg (milligrams). Generally, the antwscarring agent that may be released in a day may be in the amount ranging from 0.01 μg to about 10 μg; or from -10 μg to about 1 mg; or from 1 mg to about 10 mg; or from 10 mg to about 100 mg; or from 100 mg to about 500 mg; or from 500 mg to about 2500 mg.
In one embodiment, the anti-scarring agent is made available to the susceptible tissue site in a programmed, sustained, and/or controlled manner which results in increased efficiency and/or efficacy. Further, the release rates may vary during either or both of the initial and subsequent release phases. There may also be additional phase(s) for release of the same substance(s) and/or different substance(s).
Delivery of Therapeutic Agents or Compositions
The present invention provides various compositions which can be used to inhibit fibrosis and/or infection of tissue in the vicinity of a treatment site (e.g., a surgical site). Within various embodiments, fibrosis and/or infection is inhibited by local or systemic release of specific pharmacological agents that become localized at the site or intervention. Within other embodiments, fibrosis and/or infection can be inhibited by local or systemic release of specific pharmacological agents that become localized adjacent to a device or implant that has been introduced into a host. In certain embodiments, compositions are provided which inhibit fibrosis in and around an implanted device, or prevent "stenosis" of a device/implant in situ, thus enhancing the efficacy. In other embodiments, anti-infective compositions are provided which inhibit or prevent infection in and around an implanted device.
There are numerous methods available for optimizing delivery of the therapeutic agent to the site of the intervention. Several of these are described below.
Systemic, Regional and Local Delivery of Therapeutic Agents A variety of drug-delivery technologies are available for systemic, regional and local delivery of anti-infective and/or anti-fibrosis therapeutic agents.
For systemic delivery of therapeutic agents, several routes of administration would be suitable to provide systemic exposure of the therapeutic agent, including: (a) intravenous, (b) oral, (c) subcutaneous, (d) intraperitoneal, (e) intrathecal, [T) inhaled and intranasal, (g) sublingual or transbuccal, (h) rectal, (i) intravaginal, (j) intra-arterial, (k) intracardiac, (I) transdermal, (m) intraocular and (n) intramuscular. The therapeutic agent may be administered as a sustained low dose therapy to prevent disease progression, prolong disease remission, or decrease symptoms in active disease. Alternatively, the therapeutic agent may be administered in higher doses as a "pulse" therapy to induce remission in acutely active disease. The minimum dose capable of achieving these endpoints can be used and can vary according to patient, severity of disease, formulation of the administered agent, potency and tolerability of the therapeutic agent, and route of administration. For regional and local delivery of therapeutic agents, several techniques would be suitable to achieve preferentially elevated levels of therapeutic agents in the vicinity of the area to be treated. These include: (a) using drug-delivery catheters and/or a syringe and needle for local, regional or systemic delivery of ftbrosis-inhibiting agents to the tissue surrounding the device or implant (typically, drug delivery catheters are advanced through the circulation or inserted directly into tissues under radiological guidance until they reach the desired anatomical location; the fibrosis-inhibiting agent can then be released from the catheter lumen in high local concentrations in order to deliver therapeutic doses of the drug to the tissue surrounding the device or implant); (b) drug localization techniques such as magnetic, ultrasonic or MRI-guided drug delivery; (c) chemical modification of the therapeutic drug or formulation designed to increase uptake of the agent into damaged tissues (e.g., antibodies directed against damaged or healing tissue components such as macrophages, neutrophils, smooth muscle cells, fibroblasts, extracellular matrix components, neovascular tissue); (d) chemical modification of the therapeutic drug or formulation designed to localize the drug to areas of bleeding or disrupted vasculature; and/or (e) direct injection, for example subcutaneous, intramuscular, intra-articular, etc, of the therapeutic agent, for example, under normal or endoscopic vision.
Infiltration of Therapeutic Agents into the Tissue Surrounding a
Device or Implant
Alternatively, the tissue cavity or surgical pocket into which a device or implant is placed can be treated with an anti-infective and/or fibrosis-inhibiting therapeutic agent or a composition that comprises an anti- infective and/or fibrosis-inhibiting therapeutic agent prior to, during, or after the procedure. This can be accomplished in several ways including: (a) topical application of the agent into the anatomical space or surface where the device will be placed (particularly useful for this embodiment is the use of polymeric carriers which release the agent over a period ranging from several hours to several weeks. Compositions that can be used for this application include, e.g., fluids, microspheres, pastes, gels, microparticulates, sprays, aerosols, solid implants and other formulations which release a therapeutic agent into the region where the device or implant will be implanted); (b) microparticulate forms of the therapeutic agent are also useful for directed delivery into the implantation site; (c) sprayable collagen-containing formulations such as COSTASIS and crosslinked derivatized poly(ethylene glycol) -collagen compositions (described, e.g., in U.S. Patent Nos. 5,874,500 and 5,565,519 and referred to herein as "CT3" (both from Angiotech Pharmaceuticals, Inc., Canada), either alone, or loaded with a therapeutic agent, applied to the implantation site (or the implant/device surface); (d) sprayable PEG-containing formulations such as COSEAL or ADHIBIT (Angiotech Pharmaceuticals, Inc.), SPRAYGEL or DURASEAL (both from Confluent Surgical, Inc., Boston, MA), either alone, or loaded with a. therapeutic agent, applied to the implantation site (or the implant/device surface); (e) fibrin-containing formulations such as FLOSEAL or TISSEEL (both from Baxter Healthcare Corporation, Fremont, CA), applied to the implantation site (or the implant/device surface); (f) hyaluronic acid-containing formulations such as RESTYLANE or PERLANE (both from Q-Med AB, Sweden), HYLAFORM (Inamed Corporation (Santa Barbara, CA)), SYNVISC (Biomatrix, Inc., Ridgefield, NJ), SEPRAFILM or SEPRACOAT (both from Genzyme Corporation, Cambridge, MA) loaded with a therapeutic agent applied to the implantation site (or the implant/device surface); (g) polymeric gels for surgical implantation such as REPEL (Life Medical Sciences, Inc., Princeton, NJ) or FLOGEL (Baxter Healthcare Corporation) loaded with a therapeutic agent applied to the implantation site (or the implant/device surface); (h) orthopedic "cements" used to hold prostheses and tissues in place with a therapeutic agent applied to the implantation site (or the implant/device surface); (i) surgical adhesives containing cyanoacrylates such as DERMABOND (Johnson & Johnson, Inc., New Brunswick, NJ), INDERMIL (U.S. Surgical Company, Norwalk, CT), GLUSTITCH (Blacklock Medical Products Inc., Canada), TISSUMEND Il (Veterniary Products Laboratories,, Phoenix, AZ), VETBOND (3M Company, St. Paul, MN), HISTOACRYL BLUE (Davis & Geek, St. Louis, MO) and ORABASE SMOOTHE-N-SEAL Liquid Protectant (Colgate-Palmolive Company, New York, NY) loaded with a therapeutic agent, applied to the implantation site (or the implant/device surface); and/or (j) protein-based sealants or adhesives such as BIOGLUE (Cryolife, Inc.) and TISSUEBOND (TissueMed, Ltd.) loaded with a therapeutic agent, applied to the implantation site (or the implant/device surface).
An exemplary polymeric matrix which can be used to help prevent the formation of fibrous tissue, either alone or in combination with a fibrosis inhibiting agent/composition, is formed from reactants comprising either one or both of pentaerythritol poly(ethylene glycol)ether tetra- sulfhydryl] (4-armed thiol PEG, which includes structures having a linking group(s) between a sulfhydryl group(s) and the terminus of the polyethylene glycol backbone) and pentaerythritol poly(ethylene glycol)ether tetra- succinimidyl glutarate] (4-armed NHS PEG, which again includes structures having a linking group(s) between a NHS group(s) and the terminus of the polyethylene glycol backbone) as reactive reagents. Another preferred composition comprises either one or both of pentaerythritol poly(ethylene glycol)ether tetra-amino] (4-armed amino PEG, which includes structures having a linking group(s) between an amino group(s) and the terminus of the polyethylene glycol backbone) and pentaerythritol poly(ethylene glycol)ether tetra-succinimidyl glutarate] (4-armed NHS PEG, which again includes structures having a linking group(s) between a NHS group(s) and the terminus of the polyethylene glycol backbone) as reactive reagents. Chemical structures for these reactants are shown in, e.g., U.S. Patent 5,874,500. Optionally, collagen or a collagen derivative (e.g., methylated collagen) is added to the poly(ethylene glycol)-containing reactant(s) to form a preferred crosslinked matrix that can serve as a polymeric carrier for a therapeutic agent or a stand-alone composition to help prevent the formation of fibrous tissue.
Additional descriptions of infiltrating tissues around medical devices or implants with the therapeutic agents of the present invention are provided below in connection of using therapeutic agents or pharmaceutical compositions of the present invention.
Delivery of Theraputic Agents via Medical Devices or Implants
In certain embodiments, the therapeutic agents or compositions of the present invention may be delivered via medical devices or implants, for example, as a coating or otherwise a component of the devices or implants. The therapeutic agents may, or may not, be released from the devices or implants.
A medical device or implants useful in delivering the therapeutic agents may be made by (a) directly affixing to the implant or device a desired therapeutic agent or composition containing the therapeutic agent (e.g., by either spraying or electrospraying the medical implant with a drug and/or carrier (polymeric or non-polymeric)-drug composition to create a film and/or coating on all, or parts of the internal or external surface of the device; by dipping the implant or device into a drug and/or carrier (polymeric or non-polymeric)-drug solution to coat all or parts of the device or implant; or by other covalent or noncovalent attachment of the therapeutic agent to the device or implant surface); (b) by coating the medical device or implant with a substance such as a hydrogel which either contains or which will in turn absorb the desired fibrosis-inhibiting agent or composition; (c) by interweaving a "thread" composed of, or coated with, the fibrosis-inhibiting agent into the medical implant or device {e.g., a polymeric strand composed of materials that inhibit fibrosis (e.g., paclitaxel, mitoxantrone, doxorubicin, epithilone B, etoposide, TAXOTERE, Tubercidin, vinblastine, geldanamycin, simvastatin, halifuginone, sirolimus, everolimus, mycophenolic acid, 1- alpha-25 dihydroxy vitamin D3, bay 11-7082, SB202190, sulconizole polymerized drug compositions) or polymers which release a fibrosis- inhibiting agent from the thread}; (d) by covering all, or portions of the device or implant with a sleeve, cover, electrospun fabric or mesh containing a fibrosis-inhibiting agent (i.e., a covering comprised of a fibrosis-inhibiting agent - paclitaxel, mitoxantrone, doxorubicin, epithilone B, etoposide, TAXOTERE, tubercidin, vinblastine, geldanamycin, simvastatin, halifuginone, sirolimus, everolimus, mycophenolic acid, 1-alpha-25 dihydroxy vitamin D3, Bay 11-7082, SB202190, sulconizole or polymerized compositions containing fibrosis-inhibiting agents); (e) constructing all, or parts of the device or implant itself with the desired agent or composition (e.g., paclitaxel, mitoxantrone, doxorubicin, epithilone B, etoposide, TAXOTERE, tubercidin, vinblastine, geldanamycin, simvastatin, halifuginone, sirolimus, everolimus, mycophenolic acid, 1-alpha-25 dihydroxy vitamin D3, bay 11-7082, SB202190, sulconizole or polymerized compositions olfibrosis-inhibiting agents); (f) otherwise impregnating the device or implant with the desired fibrosis-inhibiting agent or composition; (g) composing all, or parts, of the device or implant from metal alloys that inhibit fibrosis; (h) constructing all, or parts of the device or implant itself from a degradable or non-degradable polymer that releases one or more fibrosis-inhibiting agents; (i) utilizing specialized multi-drug releasing medical device systems (for example, U.S. Patent. Nos. 6,527,799; 6,293,967; 6,290,673; 6241762, U.S. Application Publication Nos. 2003/0199970A1 and 2003/0167085A1 , and PCT Publication WO 03/015664) to deliver fibrosis-inhibiting agents alone or in combination.
Additional descriptions of making or using various medical devices or implants that comprise the therapeutic agents of the present invention are provided below in connection with using the therapeutic agents and pharmaceutical compositions of the present invention. Uses of Therapeutic Agents and Pharmaceutical Compositions
The compositions of the present invention can be used in a variety of different applications. For example, the compositions may be used for (a) preventing tissue adhesions; (b) treating or preventing inflammatory arthritis; (c) prevention of cartilage loss; (d) treating or preventing hypertrophic scars and keloids; (e) treating or preventing vascular disease; (f) combining with medical implants or devices, and (g) infiltrating tissues around medical devices or implants. A more detailed description of several specific applications is given below.
(a) Adhesion Preventions
The present invention provides compositions for use in the prevention of adhesions (e.g., surgical adhesions). The compositions may include one or more therapeutically active agents (e.g., anti-scarring agents), which provide pharmacological alteration of cellular and/ or non- cellular processes involved in the development and/or progression of surgical adhesions. Therapeutically active agents are described that can reduce surgical adhesions by inhibiting the formation of fibrous or scar tissue. In another aspect, the present invention provides surgical adhesion barriers that include an anti-scarring agent or a composition that includes an anti-scarring agent.
Surgical adhesions are abnormal, fibrous bands of scar tissue that can form inside the body as a result of the healing process that follows any open or minimally invasive surgical procedure including abdominal, gynecologic, cardiothoracic, spinal, plastic, vascular, ENT, ophthalmologic, urologic, neuro, or orthopedic surgery. Surgical adhesions are typically connective tissue structures that form between adjacent injured areas within the body. Briefly, localized areas of injury trigger an inflammatory and healing response that culminates in healing and scar tissue formation. If scarring results in the formation of fibrous tissue bands or adherence of adjacent anatomical structures (that should be separate), surgical adhesion formation is said to have occurred. Adhesions can range from flimsy, easily separable structures to dense, tenacious fibrous structures that can only be separated by surgical dissection. While many adhesions are benign, some can cause significant clinical problems and are a leading cause of repeat surgical intervention. Surgery to breakdown adhesions (adhesiolysis) often results in failure and recurrence because the surgical trauma involved in breaking down the adhesion triggers the entire process to repeat itself. Surgical breakdown of adhesions is a significant clinical problem and it is estimated that there were 473,000 adhesiolysis procedures in the US in 2002. According to the Diagnosis-Related Groups (DRGs), the total hospital charges for these procedures is likely to be at least US $10 billion annually.
Since all interventions involve a certain degree of trauma to the operative tissues, virtually any procedure (no matter how well executed) has the potential to result in the formation of clinically significant adhesion formation. Adhesions can be triggered by surgical trauma such as cutting, manipulation,, retraction or suturing, as well as from inflammation, infection (e.g., fungal or mycobacterium), bleeding or the presence of a foreign body. Surgical trauma may also result from tissue drying, ischemia, or thermal injury. Due to the diverse etiology of surgical adhesions, the potential for formation exists regardless of whether the surgery is done in a so-called minimally invasive fashion (e.g., catheter-based therapies, laparoscopy) or in a standard open technique involving one or more relatively large incisions. Although a potential complication of any surgical intervention, surgical adhesions are particularly problematic in Gl surgery (causing bowel obstruction), gynecological surgery (causing pain and/or infertility), tendon repairs (causing shortening and flexion deformities), joint capsule procedures (causing capsular contractures), and nerve and muscle repair procedures (causing diminished or lost function).
Surgical adhesions may cause various, often serious and unpredictable clinical complications; some of which manifest themselves only years after the original procedure was completed. Complications from surgical adhesions are a major cause of failed surgical therapy and are the leading cause of bowel obstruction and infertility. Other adhesion-related complications include chronic back or pelvic pain, intestinal obstruction, urethral obstruction and voiding dysfunction. Relieving the post-surgical complications caused by adhesions generally requires another surgery. However, the subsequent surgery is further complicated by adhesions formed as a result of the previous surgery. In addition, the second surgery is likely to result in further adhesions and a continuing cycle of additional surgical complications.
The placement of medical devices and implants also increases the risk that surgical adhesions will occur. In addition to the above mechanisms, an implanted device can trigger a "foreign body" response where the immune system recognizes the implant as foreign and triggers an inflammatory reaction that ultimately leads to scar tissue formation. A specific form of foreign body reaction in response to medical device placement is complete enclosure ("walling off') of the implant in a capsule of scar tissue (encapsulation). Fibrous encapsulation of implanted devices and implants can complicate any procedure, but breast augmentation and reconstruction surgery, joint replacement surgery, hernia repair surgery, artificial vascular graft surgery, stent placement, and neurosurgery are particularly prone to this complication. In each case, the implant becomes encapsulated by a fibrous connective tissue capsule which compromises or impairs the function of the surgical implant (e.g., breast implant, artificial joint, surgical mesh, vascular graft, stent or dural patch).
Adhesions generally begin to form within the first several days after surgery. Generally, adhesion formation is an inflammatory reaction in which factors are released, increasing vascular permeability and resulting in fibrinogen influx and fibrin deposition. This deposition forms a matrix that bridges the abutting tissues. Fibroblasts accumulate, attach to the matrix, deposit collagen and induce angiogenesis. If this cascade of events can be prevented within 4 to 5 days following surgery, then adhesion formation may be inhibited.
Various modes of adhesion prevention have been examined, including (1) prevention of fibrin deposition, (2) reduction of local tissue inflammation and (3) removal of fibrin deposits. Fibrin deposition is prevented through the use of physical barriers that are either mechanical or comprised of viscous solutions. Barriers have the added advantage of physically preventing adjacent tissues from contacting each other and thereby reducing the probability that they will scar together. Although many investigators and commercial products utilize adhesion prevention barriers, a number of technical difficulties exist and significant failure rates have been reported. Inflammation is reduced by the administration of drugs such as corticosteroids and non-steroidal anti-inflammatory drugs. However, the results from the use of these drugs in animal models have not been encouraging due to the extent of the inflammatory response and dose restriction due to systemic side effects. Finally, the removal of fibrin deposits has been investigated using proteolytic and fibrinolytic enzymes. A potential complication to the clinical use of these enzymes is the possibility for post-surgical excessive bleeding (surgical hemostasis is critical for procedural success).
Numerous polymeric compositions for use in the prevention of surgical adhesions (e.g., surgical adhesion barriers) may be used in the practice of the invention, either alone, or in combination with one or more anti-scarring agents. It should be noted that certain polymeric compositions can themselves help prevent the formation of fibrous tissue at a surgical site. In certain embodiments, the polymer composition can form a barrier between the tissue surfaces or organs.
For example, the surgical adhesion barrier may be coated onto tissue surfaces and may be composed of an aqueous solution of a hydrophilic, polymeric material (e.g., polypeptides or polysaccharide) having greater than 50,000 molecular weight and a concentration range of 0.01% to 15% by weight. See e.g., U.S. Patent No. 6,464,970. The surgical adhesion barrier may be a crosslinkable system with at least three reactive compounds each having a polymeric molecular core with at least one functional group. See e.g., U.S. Patent No. 6,458,889. The surgical adhesions barrier may be composed of a non-gelling polyoxyalkylene composition with or without a therapeutic agent. See e.g., U.S. Patent No. 6,436,425. The surgical adhesions barrier may be composed of an anionic polymer having an acid sulfate and sulfur content greater than 5% which acts to inhibit monocyte or macrophage invasion. See e.g., U.S. Patent No. 6,417,173. The surgical adhesions barrier may be an aqueous composition including a surfactant, pentoxifylline and a polyoxyalkylene polyether. See e.g., U.S. Patent No. 6,399,624. The surgical adhesions barrier may be composed by crosslinking two synthetic polymers, one having nucleophilic groups and the other having electrophilic groups, such that they form a matrix that may be used to incorporate a biologically active compound. See e.g., U. S. Patent Nos. 6,323,278; 6,166,130; 6,051 ,648 and 5,874,500. The surgical adhesion barrier may be composed of hyaluronic acid compositions such as those described in U.S. Patents Nos. 6,723,709; 6,531 ,147; and 6,464,970. The surgical adhesions barrier may be a polymeric tissue coating which is formed by applying a polymerization initiator to the tissue and then covering it with a water-soluble macromer that is polymerizable using free radical initiators under the influence of UV light. See e.g., U.S. Patent Nos. 6,177,095 and 6,083,524. The surgical adhesions barrier may be composed of fluent prepolymeric material that is emitted to the tissue surface and then exposed to activating energy in situ to initiate conversion of the applied material to non-fluent polymeric form. See e.g., U.S. Patent Nos. 6,004,547 and 5,612,050. The surgical adhesions barrier may be a hydrogel-forming, self-solvating, absorbable polyester copolymers capable of selective, segmental association into compliant hydrogels mass upon contact with an aqueous environment. See e.g., U.S. Patent No. 5,612,052. The surgical adhesions barrier may be an anionic polymer effective to inhibit cell invasion or fibrosis (e.g., dermatan sulfate, dextran sulfate, pentosan polysulfate, or alginate), and a pharmaceutically effective carrier, in which the carrier may be semi-solid. See e.g., U.S. Patent Nos. 6,756,362; 6,127,348 and 5,994,325. The surgical adhesions barrier may be an acidified hydrogel comprising a carboxypolysaccharide and a polyether having a pH in the range of about 2.0 to about 6.0. See e.g., U.S. Patent No. 6,017,301. The surgical adhesions barrier may be composed of dextran sulfate having a molecular weight about 40,000 to 500,000 Daltons which is used to inhibit neurite outgrowth. See e.g., U.S. Patent No. 5,705,178. The surgical adhesions barrier may be a fragmented biocompatible hydrogel which is at least partially hydrated and is substantially free from an aqueous phase, wherein said hydrogel comprises gelatin and will absorb water when delivered to a moist tissue target site. See e.g., U.S. Patent No. 6,066,325. The surgical adhesions barrier may be a water-soluble, degradable macromer that is composed of at least two-crosslinkable substituents that may -crosslink to other macromers at a localized site when under the influence of a polymerization initiator. See e.g., U.S. Patent No. 6,465,001. The surgical adhesions barrier may be a biocompatible adhesive composition comprising at least one alkyl ester cyanoacrylate monomer and a polymerization initiator or accelerator. See e.g., U.S. Patent No. 6,620,846.
In one embodiment, the polymers that can form a covalent bond with the tissue to which it is applied may be used. Polymers containing and/or terminated with electrophilic groups such as succinimidyl, aldehyde, epoxide, isocyanate, vinyl, vinyl sulfone, maleimide, -S-S-(C5H4N) or activated esters, such as are used in peptide synthesis may be used as the reagents. For example, a 4 armed NHS-derivatized polyethylene glycol (e.g., pentaerythritol poly(ethylene glycol)ether tetra-succinimidyl glutarate) may be applied to the tissue in the solid form or in a solution form. In this embodiment, the 4 armed NHS-derivatized polyethylene glycol is dissolved in an acidic solution (pH about 2-3) and is then co-applied to the tissue using a basic buffer (pH > about 8). The fibrosis-inhibiting agent(s) may be incorporated directly into either the 4 armed NHS-derivatized polyethylene glycol, the acidic solution or the basic buffer. In another embodiment, the fibrosis-inhibiting agent may be incorporated into a secondary carrier that may then be incorporated into the 4 armed NHS-derivatized polyethylene glycol, the acidic solution and/or the basic buffer. Secondary carriers may include microparticles and/or microspheres which are made from degradable polymers. Degradable polymers may include polyesters, where the polyester may comprise the residues of one or more of the monomers selected from lactide, lactic acid, glycolide, glycolic acid, e-caprolactone, gamma-caprolactone, hydroxyvaleric acid, hydroxybutyric acid, beta- butyrolactone, gamma-butyrolactone, gamma-valerolactone, y- decanolactone, δ-decanolactone, trimethylene carbonate, 1 ,4-dioxane-2- one or 1,5-dioxepan-2one, and block copolymers of the form X-Y, Y-X-Y, R- (Y-X)n, R-(X-Y)n and X-Y-X (where X in a polyalkylene oxide (e.g., poly(ethylene glycol, poly(propylene glycol) and block copolymers of poly(ethylene oxide) and poly(propylene oxide) (e.g., PLURONIC and PLURONIC R series of polymers from BASF Corporation, Mount Olive, NJ) and Y is a biodegradable polyester, where the polyester may comprise the residues of one or more of the monomers selected from lactide, lactic acid, glycolide, glycolic acid, e-caprolactone, gamma-caprolactone, hydroxyvaleric acid, hydroxybutyric acid, beta-butyrolactone, gamma- butyrolactone, gamma-valerolactone, γ-decanolactone, δ-decanolactone, trimethylene carbonate, 1 ,4-dioxane-2-one or 1,5-dioxepan-2one (e.g., PLG- PEG-PLG) and R is a multifunctional initiator).
In another embodiment, the tissue reactive polymer may be applied initially and then the fibrosis-inhibiting agent may then be applied to the coated tissue. The fibrosis-inhibiting agent may be applied directly to the tissue or it may be incorporated into a secondary carrier. Secondary carriers may include microspheres (as described above), microparticles (as described above), gels (e.g., hyaluronic acid, carboxymethyl cellulose, dextran, poly(ethylene oxide) - poly(propylene oxide) block copolymers as well as blends, association complexes and crosslinked compositions thereof) and films (degradable polyesters, where the polyester may comprise the residues of one or more of the monomers selected from lactide, lactic acid, glycolide, glycolic acid, e-caprolactone, gamma- caprolactone, hydroxyvaleric acid, hydroxy butyric acid, beta-butyrolactone, gamma-butyrolactone, gamma-valerolactone, γ-decanolactone, δ- decanolactone, trimethylene carbonate, 1 ,4-dioxane-2-one or 1,5-dioxepan- 2one, and block copolymers of the form X-Y, Y-X-Y, R-(Y-X)n, R-(X-Y)n and X-Y-X (where X in a polyalkylene oxide (e.g., poly(ethylene glycol, poly(propylene glycol) and block copolymers of poly(ethylene oxide) and polypropylene oxide) (e.g., PLURONIC and PLURONIC R series of polymers from BASF Corporation, Mount Olive, NJ) and Y is a biodegradable polyester, where the polyester may comprise the residues of one or more of the monomers selected from lactide, lactic acid, glycolide, glycolic acid, e-caprolactone, gamma-caprolactone, hydroxyvaleric acid, hydroxybutyric acid, beta-butyrolactone, gamma-butyrolactone, gamma- valerolactone, γ-decanolactone, δ-decanolactone, trimethylene carbonate, 1 ,4-dioxane-2-one or 1,5-dioxepan-2one (e.g., PLG-PEG-PLG) and R is a multifunctional initiator, hyaluronic acid, carboxymethyl cellulose, dextran, poly(ethylene oxide) - poly(propylene oxide) block copolymers as well as blends, association complexes and crosslinked compositions thereof. A preferred polymeric matrix which can be used to help prevent the formation of fibrous tissue, either alone or in combination with a fibrosis inhibiting agent/composition, is formed from reactants comprising either one or both of pentaerythritol poly(ethylene glycol)ether tetra- sulfhydryl] (4-armed thiol PEG, which includes structures having a linking group(s) between a sulfhydryl group(s) and the terminus of the polyethylene glycol backbone) and pentaerythritol poly(ethylene glycol)ether tetra- succinimidyl glutarate] (4-armed NHS PEG, which again includes structures having a linking group(s) between a NHS group(s) and the terminus of the polyethylene glycol backbone) as reactive reagents. Another preferred composition comprises either one or both of pentaerythritol poly(ethylene glycol)ether tetra-amino] (4-armed amino PEG, which includes structures having a linking group(s) between an amino group(s) and the terminus of the polyethylene glycol backbone) and pentaerythritol poly(ethylene glycol)ether tetra-succinimidyl glutarate] (4-armed NHS PEG, which again includes structures having a linking group(s) between a NHS group(s) and the terminus of the polyethylene glycol backbone) as reactive reagents. Chemical structures for these reactants are shown in, e.g., U.S. Patent 5,874,500. Optionally, collagen or a collagen derivative (e.g., methylated collagen) is added to the poly(ethylene glycol)-containing reactant(s) to form a preferred crosslinked matrix that can serve as a polymeric carrier for a therapeutic agent or a stand-alone composition to help prevent the formation of fibrous tissue.
Surgical adhesion barriers, which may be combined with one or more anti-scarring agents according to the present invention, also include commercially available products. Examples of surgical adhesion barrier compositions into which a fibrosis agent can be incorporated include: (a) sprayable collagen-containing formulations such as COSTASIS or CT3 (Angiotech Pharmaceuticals, Inc., Canada); (b) sprayable PEG-containing formulations such as COSEAL or ADHIBIT (Angiotech Pharmaceuticals, Inc.), SPRAYGEL or DURASEAL (both from Confluent Surgical, Inc., Boston, MA) or FOCALSEAL (Genzyme Corporation, Cambridge, MA); (c) hyaluronic acid-containing formulations such as RESTYLANE or PERLANE (both from Q-Med AB, Sweden), HYLAFORM (Inamed Corporation, Santa Barbara, CA), SYNVISC (Biomatrix, Inc., Ridgefield, NJ), SEPRAFILM or SEPRACOAT (both from Genzyme Corporation), (d) fibrinogen-containing formulations such as FLOSEAL or TISSEAL (both from Baxter Healthcare Corporation, Fremont, CA); (e) polymeric gels such as REPEL (Life Medical Sciences, Inc., Princeton, NJ) or FLOWGEL (Baxter Healthcare Corporation, Deerfield, IL), (f) surgical adhesives containing cyanoacrylates such as DERMABOND (Johnson & Johnson, Inc., New Brunswick, NJ), INDERMIL (U.S. Surgical Company, Norwalk, CT), GLUSTITCH (Blacklock Medical Products Inc., Canada), TISSUMEND (Veterinary Products Laboratories, Phoenix, AZ), VETBOND (3M Company, St. Paul, MN), HISTOACRYL BLUE (Davis & Geek, St. Louis, MO) and ORABASE SOOTHE-N-SEAL LIQUID PROTECTANT (Colgate-Palmolive Company, New York, NY); (g) dextran sulfate gels such as the ADCON range of products (available from Wright Medical Technology, Inc. Arlington, TN), (h) lipid based compositions such as ADSURF (Britannia Pharmaceuticals Ltd., United Kingdom) and (j) film compositions such as INTERCEED (Ethicon, Inc., Somerville, NJ) and HYDROSORB (MacroPore Biosurgery, Inc., San Diego, CA /Medtronic Sofamor Danek, Memphis, TN).
For greater clarity, several specific applications and treatments will be described in greater detail including:
i) Adhesion Prevention in Spinal and Neurosurgical Procedures
Back pain is the number one cause of healthcare expenditures in the United States and accounts for over $50 billion in costs annually ($100 billion worldwide). Over 12 million people in the U.S. have some form of degenerative disc disease (DDD) and 10% of them (1.2 million) will require surgery to correct their problem.
In healthy individuals, the vertebral column is composed of vertebral bone plates separated by intervertebral discs that form strong joints and absorb spinal compression during movement. The intervertebral disc is comprised of an inner gel-like substance called the nucleus pulposus which is surrounded by a tough fibrocartilagenous capsule called the annulus fibrosis. The nucleus pulposus is composed of a loose framework of collagen fibrils and connective tissue cells (resembling fibroblasts and chondrocytes) embedded in a gelatinous matrix of glycosaminoglycans and water. The annulus fibrosus is composed of numerous concentric rings of fibrocartilage that anchor into the vertebral bodies. The most common cause of DDD occurs when tears in the annulus fibrosis create an area of localized weakness that allow bulging, herniation or sequestration of the nucleus pulposis and annulus fibrosis into the spinal canal and/or spinal foramena. The bulging or herniated disc often compresses nerve tissue such as spinal cord fibers or spinal cord nerve root fibers. Pressure on the spinal cord or nerve roots from the damaged intervertebral disc results in neuronal dysfunction (numbness, weakness, tingling), crippling pain, bowel or bladder disturbances and can frequently cause long-term disability. Although many cases of DDD will spontaneously resolve, a significant number of patients will require surgical intervention in the form of minimally invasive procedures, microdiscectomy, major surgical resection of the disc, spinal fusion (fusion of adjacent vertebral bone plates using various techniques and devices), and/or implantation of an artificial disc. The present invention provides for the application of an anti-adhesion or anti- fibrosis agent inihe surgical management of DDD.
Spinal disc removal is mandatory and urgent in cauda equine syndrome when there is a significant neurological deficit; particularly bowel or bladder dysfunction. It is also performed electively to relieve pain and eliminate lesser neurological symptoms. The spinal nerve roots exit the spinal canal through bony spinal foramena (a bony opening between the vertebra above and the vertebra below) that is a common site of nerve entrapment. To gain access to the spinal foramen during back surgeries, vertebral bone tissue is often resected; a process known as laminectomy.
In open surgical resection of a ruptured lumbar disc or entrapped spinal nerve root (laminectomy) the patient is placed in a modified kneeling position under general anesthesia. An incision is made in the posterior midline and the tissue is dissected away to expose the appropriate interspace; the ligamentum flavum is dissected and in some cases portions of the bony lamina are removed to allow adequate visualization. The nerve root is carefully retracted away to expose the herniated fragment and the defect in the annulus. Typically, the cavity of the disc is entered from the tear in the annulus and the loose fragments of the nucleus pulposus are removed with pituitary forceps. Any additional fragments of disc sequestered inside or outside of the disc space are also carefully removed and the disc space is forcefully irrigated to remove to remove any residual fragments. If tears are present in the dura, the dura is closed with sutures that are often augmented with fibrin glue. The tissue is then closed with absorbable sutures.
Microlumbar disc excision (microdiscectomy) can be performed as an outpatient procedure and has largely replaced laminectomy as the intervention of choice for herniated discs or root entrapment. A one inch incision is made from the spinous process above the disc affected to the spinous process below. Using an operating microscope, the tissue is dissected down to the ligamentum flavum and bone is removed from the lamina until the nerve root can be clearly identified. The nerve root is carefully retracted and the tears in the annulus are visualized under magnification. Microdisc forceps are used to remove disc fragments through the annular tear and any sequestered disc fragments are also removed. As with laminectomy, the disc space is irrigated to remove any disc fragments, any dural tears are repaired and the tissue is closed with absorbable sutures. It should be noted that anterior (abdominal) approaches can also be used for both open and endoscopic lumbar disc excision. Cervical and thoracic disc excisions are similar to lumbar procedures and can also be performed from a posterior approach (with laminectomy) or as an anterior discectomy with fusion.
Back surgeries, such as laminectomies, discectomies and microdiscectomies, often leave the spinal dura exposed and unprotected. As a result, scar tissue frequently forms between the dura and the surrounding tissue. This scar is formed from the damaged erector spinae muscles that overlay the laminectomy site. The result is adhesion development between the muscle tissue and the fragile dura, thereby, reducing mobility of the spine and the nerve roots that exit from it, leading to pain, persistent neurological symptoms and slow post-operative recovery. Similarly, adhesions that occur in the epidural and dural tissue cause complications in spinal injury (e.g., compression and crush injuries) cases. In addition, scar and adhesion formation within the dura and around nerve roots has been implicated in rendering subsequent (revision and repeat) spine operations technically more difficult to perform.
To circumvent adhesion development, a scar-reducing barrier may be inserted between the dural sleeve and the paravertebral musculature post-laminectomy. Alternatively (or in addition to this), the adhesion barrier, either alone or containing a fibrosis-inhibiting agent, can be coated on (or infiltrated into the tissues around) the spinal nerve as it exits the spinal canal and traverses the space between the bony vertebra (Ae., the laminectomy site). This reduces cellular and vascular invasion into the epidural space from the overlying muscle and exposed cancellous bone and j.hus,_reduc.es the complications associated with scarring of the canal housing, spinal chord and/or nerve roots. In microdiscectomy procedures it is important that the barrier be deliverable as a spray, gel or fluid material that can be administered via the delivery port of an endoscope. Once again, the adhesion barrier, either alone or containing a fibrosis-inhibiting agent, can be sprayed onto the spinal nerve (or infiltrated into the tissues around it) as it exits the spinal canal and traverses the space between the bony vertebra (i.e., the laminectomy site). The present invention discloses barrier compositions, used either alone or combined with a fibrosis-inhibiting agent, that can be delivered during surgical disc resection and microdiscectomy either directly, using specialized delivery catheters, via an endoscope, or through a needle or other applicator. When dural defects are present, the fibrosis-inhibiting agent will assist in the healing of the dura and prevent complications such as blockage of CSF flow.
In another aspect, adhesion formation may be associated with a neurosurgical (brain) procedure. Neurosurgical procedures are fraught with potentially severe post-operative complications that are often attributed to surgical trauma and unwanted fibrosis or gliosis (gliosis is scar tissue formation in the brain as a result of glial cell activity). Increased intracranial bleeding, infection, cerebrospinal fluid leakage and pain are but some complications resulting from adhesions following neurosurgery. For example, if scar tissue interrupts the normal circulation of cerebrospinal fluid (CSF) following brain or spinal surgery, the fluid can accumulate and exert pressure on surrounding tissues (causing increased intracranial pressure) leading to severe complications (such as uncal herneation, brain damage and/or death). Here the adhesion barrier alone, or combined with a fibrosis- inhibiting agent, can be used to prevent excessive dural scarring and adhesion formation in a variety of neurosurgical procedures.
There are numerous compositions that may be used alone or loaded with a therapeutic agent (e.g., a fibrosis-inhibiting agent or an anti- infective agent), applied to a spinal or neurosurgical site (or to an implant surface placed in the spine - such as an artificial disc, rods, screws, spinal cages, drug-delivery pumps, neurostimulation devices; or to an implant placed in the brain - such as drains, shunts, drug-delivery pumps, neurostimulation devices) for the prevention of surgical adhesions in neurosurgical procedures. It should be noted that certain polymeric compositions can themselves help prevent the formation of fibrous tissue at a spinal or neurosurgical site. These compositions are particularly useful for the practice of this embodiment, either alone, or in combination with a fibrosis-inhibiting composition.
Various polymeric compositions can be infiltrated into the spinal or neurosurgical site (e.g., onto tissue at the surgical site or in the vicinity of the implant-tissue interface) with or without an additional therapeutic agent for the prevention of surgical adhesions.
In one embodiment, the polymers that can form a covalent bond with the tissue to which it is applied may be used. Polymers containing and/or terminated with electrophilic groups such as succinimidyl, aldehyde, epoxide, isocyanate, vinyl, vinyl sulfone, maleimide, -S-S-(C5H4N) or activated esters, such as are used in peptide synthesis may be used as the reagents. For example, a 4 armed NHS-derivatized polyethylene glycol (e.g., pentaerythritol poly(ethylene glycol)ether tetra-succinimidyl glutarate) may be applied to the tissue in the solid form or in a solution form. In this embodiment, the 4 armed NHS-derivatized polyethylene glycol is dissolved in an acidic solution (pH about 2-3) and is then co-applied to the tissue using a basic buffer (pH > about 8). The antifibrosisfibrosis-inhibiting agent(s) may be incorporated directly into either the 4 armed NHS- derivatized polyethylene glycol, the acidic solution or the basic buffer.
In another embodiment, the fibrosis-inhibiting agent may be incorporated into a secondary carrier that may then be incorporated into the 4 armed NHS-derivatized polyethylene glycol, the acidic solution and/or the basic buffer. The secondary carriers may include microparticles and/or microspheres which are made from degradable polymers. The degradable polymers may include polyesters, where the polyester may comprise the residues of one or more of the monomers selected from lactide, lactic acid, glycolide, glycolic acid, e-caprolactone, gamma-caprolactone, hydroxyvaleric acid, hydroxybutyric acid, beta-butyrolactone, gamma- butyrolactone, gamma-valerolactone, γ-decanolactone, δ-decanolactone, trimethylene carbonate, 1 ,4-dioxane-2-one or 1 ,5-dioxepan-2one, and block copolymers of the form X-Y, Y-X-Y, R-(Y-X)n, R-(X-Y)n and X-Y-X (where X in a polyalkylene oxide (e.g., poly(ethylene glycol, polypropylene glycol) and block copolymers of poly(ethylene oxide) and poly(propylene oxide) {e.g., PLURONIC and PLURONIC R series of polymers from BASF Corporation, Mount Olive, NJ) and Y is a biodegradable polyester, where the polyester may comprise the residues of one or more of the monomers selected from lactide, lactic acid, glycolide, glycolic acid, e-caprolactone, gamma-caprolactone, hydroxyvaleric acid, hydroxybutyric acid, beta- butyrolactone, gamma-butyrolactone, gamma-valerolactone, y- decanolactone, δ-decanolactone, trimethylene carbonate, 1 ,4-dioxane-2- one or 1 ,5-dioxepan-2one (e.g., PLG-PEG-PLG) and R is a multifunctional initiator).
In another embodiment, the tissue reactive polymer may be applied initially and then the fibrosis-inhibiting agent may then be applied to the coated tissue. The fibrosis-inhibiting agent may be applied directly to the tissue or it may be incorporated into a secondary carrier. The secondary carriers may include microspheres (as described above), microparticles (as described above), gels (e.g., hyaluronic acid, carboxymethyl cellulose, dextran, poly(ethylene oxide) - polypropylene oxide) block copolymers as well as blends, association complexes and crosslinked compositions thereof) and films (degradable polyesters, where the polyester may comprise the residues of one or more of the monomers selected from lactide, lactic acid, glycolide, glycolic acid, e-caprolactone, gamma- caprolactone, hydroxyvaleric acid, hydroxybutyric acid, beta-butyrolactone, gamma-butyrolactone, gamma-valerolactone, γ-decanolactone, δ- decanolactone, trimethylene carbonate, 1 ,4-dioxane-2-one or 1,5-dioxepan- 2one, and block copolymers of the form X-Y, Y-X-Y, R-(Y-X)n, R-(X-Y)n and X-Y-X where X in a polyalkylene oxide (e.g., poly(ethylene glycol, poly(propylene glycol) and block copolymers of poly(ethylene oxide) and polypropylene oxide) (e.g., PLURONIC and PLURONIC R series of polymers from BASF Corporation, Mount Olive, NJ) and Y is a biodegradable polyester, where the polyester may comprise the residues of one or more of the monomers selected from lactide, lactic acid, glycolide, glycolic acid, e-caprolactone, gamma-caprolactone, hydroxyvaleric acid, hydroxybutyric acid, beta-butyrolactone, gamma-butyrolactone, gamma- valerolactone, Y-decanolactone, δ-decanolactone, trimethylene carbonate, 1 ,4-dioxane-2-one or 1,5-dioxepan-2one (e.g., PLG-PEG-PLG) and R is a multifunctional initiator, hyaluronic acid, carboxymethyl cellulose, dextran, poly(ethylene oxide) - poly(propylene oxide) block copolymers as well as blends, association complexes and crosslinked compositions thereof. A preferred polymeric matrix which can be used to help prevent the formation of fibrous tissue that leads to surgical adhesions, either alone or in combination with a fibrosis inhibiting agent/composition, is formed from reactants comprising either one or both of pentaerythritol poly(ethylene glycol)ether tetra-sulfhydryl] (4-armed thiol PEG, which includes structures having a linking group(s) between a sulfhydryl group(s) and the terminus of the polyethylene glycol backbone) and pentaerythritol poly(ethylene glycol)ether tetra-succinimidyl glutarate] (4-armed NHS PEG, which again includes structures having a linking group(s) between a NHS group(s) and the terminus of the polyethylene glycol backbone) as reactive reagents. Another preferred composition comprises either one or both of pentaerythritol poly(ethylene glycol)ether tetra-amino] (4-armed amino PEG, which includes structures having a linking group(s) between an amino group(s) and the terminus of the polyethylene glycol backbone) and pentaerythritol poly(ethylene glycol)ether tetra-succinimidyl glutarate] (4- armed NHS PEG, which again includes structures having a linking group(s) between a NHS group(s) and the terminus of the polyethylene glycol backbone) as reactive reagents. Chemical structures for these reactants are shown in, e.g., U.S. Patent 5,874,500. Optionally, collagen or a collagen derivative (e.g., methylated collagen) is added to the poly(ethylene glycol)-containing reactant(s) to form a preferred crosslinked matrix that can serve as a polymeric carrier for a therapeutic agent or a stand-alone composition to help prevent the formation of fibrous tissue.
Other examples of polymeric compositions that can be infiltrated into the spinal or neurosurgical site (e.g., onto tissue at the surgical site or in the vicinity of the implant-tissue interface) with or without an additional fibrosis-inhibiting (and/or an anti-infective) therapeutic agent for the prevention of surgical adhesions, include a variety of commercial products. For example, Confluent Surgical, Inc. makes their DURASEAL which is a synthetic hydrogel designed to augment sutured dura closures following cranial surgical procedures. Products that are being developed by Confluent Surgical, Inc. are described in, for example, U.S. Patent No. 6,379,373. FzioMed, Inc. (San Luis Obispo, CA) makes OXIPLEX/SP Gel which is being sold as an adhesion barrier for spine surgery. OXIPLEX/SP Gel is being used for the reduction of pain and radiculopathy in laminectomy, laminotomy and discectomy surgeries. Products being developed by FzioMed, Inc. are described in, for example, U.S. Patent Nos. 6,566,345 and 6,017,301. Anika Therapeutics, Inc. (Woburn, MA) is developing INCERT-S for the prevention of internal adhesions or scarring following spinal surgery. INCERT-S is part of a potential family of bioabsorbable, chemically modified hyaluronic acid therapies. Products being developed by Anika Therapeutics, Inc. are described in, for example, U.S. Patent Nos. 6,548,081; 6,537,979; 6,096,727; 6,013,679; 5,502,081 and 5,356,883. Life Medical Sciences, Inc. (Little Silver, NJ) is developing RELIEVE as a bio-resorbable polymer designed to prevent or reduce the formation of adhesions that can follow spinal surgery. Products being developed by Life Medical Sciences, Inc. are described in, for example, U.S. Patent Nos. 6,696,499; 6,399,624; 6,211 ,249; 6,136,333 and 5,711 ,958. Wright Medical Technology, Inc. is selling the ADCON range of products which are dextran sulfate gels originally developed by Gliatech, Inc. (Beachwood, OH) to inhibit postsurgical peridural fibrosis that occurs in posterior lumbar laminectomy or laminotomy procedures where nerve routes are exposed. ADCON provides a barrier between the spinal cord and nerve roots and the surrounding muscle and bone following lumbar spine surgeries. The ADCON range of products may be described in, for example, U.S. Patent Nos. 6,417,173; 6,127,348; 6,083,930; 5,994,325 and 5,705,178.
Other commercially available materials that may be used alone or loaded with a therapeutic agent (e.g., a fibrosis-inhibiting agent and/or an anti-infective agent), applied to or infiltrated into a spinal or neurosurgical site (or to an implant surface) for the prevention of adhesions include: (a) sprayable collagen-containing formulations such as COSTASIS or CT3; (b) sprayable PEG-containing formulations such as COSEAL, ADHIBIT, FOCALSEAL, or SPRAYGEL; (c) fibrinogen-containing formulations such as FLOSEAL or TISSEAL (both from Baxter Healthcare Corporation, Fremont, CA); (d) hyaluronic acid-containing formulations such as RESTYLANE, PERLANE, HYLAFORM, SYNVISC, SEPRAFILM or SEPRACOAT; (e) polymeric gels for surgical implantation such as REPEL or FLOWGEL; (f) surgical adhesives containing cyanoacrylates such as DERMABOND, INDERMIL, GLUSTITCH, TISSUMEND, VETBOND, HISTOACRYL BLUE and ORABASE SOOTHE-N-SEAL LIQUID PROTECTANT; (h) lipid based compositions such as ADSURF, and G) film compositions such as INTERCEED (Ethicon, Inc., Somerville, NJ) and HYDROSORB (MacroPore Biosurgery, Inc., San Diego, CA /Medtronic Sofamor Danek, Memphis, TN). It should be obvious to one of skill in the art that commercial compositions not specifically cited above as well as next- generation and/or subsequently-developed commercial products are to be anticipated and are suitable for use under the present invention.
As described above, the compositions for the prevention of surgical adhesions can be applied directly or indirectly to the tissue in a spinal or neurosurgical site. The polymeric compositions (either with or without a therapeutic agent) can be administered in any manner described herein. Exemplary methods include either direct application at the time of surgery, with endoscopic, ultrasound, CT, MRI, or fluoroscopic guidance, and/or in conjunction with the placement of a device or implant at the surgical site. Representative examples of devices or implants for use in spinal and neurosurgical procedures includes, without limitation, dural patches, spinal prostheses (e.g., artificial discs, injectable filling or bulking agents for discs, spinal grafts, spinal nucleus implants, intervertebral disc spacers), fusion cages, neurostimulation devices, implantable drug-delivery pumps, shunts, drains, electrodes, and bone fixation devices (e.g., anchoring plates and bone screws). The polymeric composition, with or without a fibrosis-inhibiting agent, may be applied during open or endoscopic procedures: (a) to the surface of the operative site (e.g., as an injectable, solution, paste, gel, in situ forming gel or mesh) before, during, or after the surgical procedure; (b) to the surface of the tissue surrounding the operative site (e.g., as an injectable, solution, paste, gel, in situ forming gel or mesh) before, during or after the surgical procedure; (c) by topical application of the composition into an anatomical space (such as the subdural space or intrathecally) at the surgical site (particularly useful for this embodiment is the use of polymeric carriers which release the fibrosis-inhibiting agent over a period ranging from several hours to several weeks - fluids, suspensions, emulsions, microemulsions, microspheres, pastes, gels, microparticulates, sprays, aerosols, solid implants and other formulations which release the agent and can be delivered into the region where the device will be inserted); (d) via percutaneous injection into the tissue in and around the operative site as a solution, as an infusate, or as a sustained release preparation; and/or (e) by any combination of the aforementioned methods. Combination therapies (i.e., combinations of therapeutic agents and combinations with antithrombotic, anti-infective, and/or antiplatelet agents) can also be used.
In certain applications involving the placement of a medical device or implant, it may be desirable to apply the anti-fibrosis (and/or anti- infective) composition at a site that is adjacent to an implant (preferably near the implant-tissue interface). This can be accomplished during open or endoscopic procedures by applying the polymeric composition, with or without a fibrosis-inhibiting agent: (a) to the implant surface (e.g., as an injectable, solution, paste, gel, in situ forming gel, or mesh) before, during, or after the implantation procedure; (b) to the surface of the adjacent tissue (e.g., as an injectable, solution, paste, gel, in situ forming gel, or mesh) immediately prior to, during, or after implantation of the implant; (c) to the surface of the implant and the tissue surrounding the implant (e.g., as an injectable, solution, paste, gel, in situ forming gel or mesh) before, during, or after implantation of the implant; (d) by topical application of the composition into the anatomical space (such as the sudural space or intrathecally) where the implant will be placed (particularly useful for this embodiment is the use of polymeric carriers which release the fibrosis-inhibiting agent over a period ranging from several hours to several weeks - fluids, suspensions, emulsions, microemulsions, microspheres, pastes, gels, microparticulates, sprays, aerosols, solid implants and other formulations which release the agent and can be delivered into the region where the device will be inserted); (e) via percutaneous injection into the tissue surrounding the implant as a solution, as an infusate, or as a sustained release preparation; and/or (f) by any combination of the aforementioned methods. Combination therapies (i.e., combinations of therapeutic agents and combinations with antithrombotic, anti-infective, and/or antiplatelet agents) can also be used.
In one aspect, the polymeric composition may be delivered to the tissue (or device/tissue interface) in the form of a spray or gel during open, endoscopic or catheter-based procedures. The fibrosis-inhibiting agent can be incorporated directly into the surgical adhesion barrier or it can be incorporated into a secondary carrier (polymeric or non-polymeric), as described above, that is then incorporated into the adhesion barrier. Examples of polymer compositions that may be in the form of a spray or gel include poly(ethylene glycol)-based systems, hyaluronic acid and crosslinked hyaluronic acid compositions. These compositions can be applied as the final composition or they can be applied as materials that form a crosslinked gel in situ.
In another aspect, an activated polymer is dissolved in a biologically acceptable buffer that has a pH lower that 6.8. The resultant solution is then applied to the desired tissue surface in the presence of a second biologically acceptable buffer that has a pH greater than 7.5. Application of the reaction mixture to the tissue site may be by extrusion, brushing, spraying or by any other convenient means. Following application of the composition to the surgical site, any excess solution may be removed from the surgical site if deemed necessary. At this point in time, the surgical site can be closed using conventional means (e.g., sutures, staples, or a bioadhesive). In one embodiment, the activated polymer can form a covalent bond with the tissue to which it is applied may be used. Polymers containing and/or terminated with electrophilic groups such as succinimidyl, aldehyde, epoxide, isocyanate, vinyl, vinyl sulfone, maleimide, -S-S-(C5H4N) or activated esters, such as are used in peptide synthesis may be used as the reagents. For example, a 4 armed NHS-derivatized polyethylene glycol (e.g., pentaerythritol poly(ethylene glycol)ether tetra-succinimidyl glutarate) may be applied to the tissue in the solid form or in a solution form. In this embodiment, the 4 armed NHS-derivatized polyethylene glycol is dissolved in an acidic solution (pH about 2-3) and is then co-applied to the tissue using a basic buffer (pH > about 8). The antifibrosisfibrosis-inhibiting agent(s) may be incorporated directly into either the 4 armed NHS- derivatized polyethylene glycol, the acidic solution or the basic buffer. In another embodiment, the fibrosis-inhibiting agent may be incorporated into a secondary carrier that may then be incorporated into the 4 armed NHS- derivatized polyethylene glycol, the acidic solution and/or the basic buffer. The secondary carriers may include microparticles and/or microspheres which are made from degradable polymers. The degradable polymers may include polyesters, where the polyester may comprise the residues of one or more of the monomers selected from lactide, lactic acid, glycolide, glycolic acid, e-caprolactone, gamma-caprolactone, hydroxyvaleric acid, hydroxybutyric acid, beta-butyrolactone, gamma-butyrolactone, gamma- valerolactone, γ-decanolactone, δ-decanolactone, trimethylene carbonate, 1 ,4-dioxane-2-one or 1,5-dioxepan-2one, and block copolymers of the form X-Y1 Y-X-Y, R-(Y-X)n, R-(X-Y)n and X-Y-X where X in a polyalkylene oxide (e.g., poly(ethylene glycol, poiyφropylene glycol) and block copolymers of poly(ethylene oxide) and poly(propylene oxide) (e.g., PLURONIC and PLURONIC R series of polymers from BASF Corporation, Mount Olive, NJ) and Y is a biodegradable polyester, where the polyester may comprise the residues of one or more of the monomers selected from lactide, lactic acid, glycolide, glycolic acid, e-caprolactone, gamma-caprolactone, hydroxy valeric acid, hydroxybutyric acid, beta-butyrolactone, gamma- butyrolactone, gamma-valerolactone, γ-decano!actone, δ-decanolactone, trimethylene carbonate, 1 ,4-dioxane-2-one or 1,5-dioxepan-2one (e.g., PLG- PEG-PLG) and R is a multifunctional initiator. In another embodiment, the tissue reactive polymer may be applied initially and then the fibrosis- inhibiting agent may then be applied to the coated tissue. The fibrosis- inhibiting agent may be applied directly to the tissue or it may be incorporated into a secondary carrier. The secondary carriers may include microspheres (as described above), microparticles (as described above), gels (e.g., hyaluronic acid, carboxymethyl cellulose, dextran, poly(ethylene oxide) - poly(propylene oxide) block copolymers as well as blends, association complexes and crosslinked compositions thereof) and films (degradable polyesters, where the polyester may comprise the residues of one or more of the monomers selected from lactide, lactic acid, glycolide, glycolic acid, e-caprolactone, gamma-caprolactone, hydroxyvaleric acid, hydroxybutyric acid, beta-butyrolactone, gamma-butyrolactone, gamma- valerolactone, γ-decanolactone, δ-decanolactone, trimethylene carbonate, 1 ,4-dioxane-2-one or 1,5-dioxepan-2one, and block copolymers of the form X-Y, Y-X-Y, R-(Y-X)n, R-(X-Y)n and X-Y-X where X in a polyalkylene oxide (e.g., poly(ethylene glycol, poly(propylene glycol) and block copolymers of poly(ethylene oxide) and poly(propylene oxide) {e.g., PLURONIC and PLURONIC R series of polymers from BASF Corporation, Mount Olive, NJ) and Y is a biodegradable polyester, where the polyester may comprise the residues of one or more of the monomers selected from lactide, lactic acid, glycolide, glycolic acid, e-caprolactone, gamma-caprolactone, hydroxyvaleric acid, hydroxybutyric acid, beta-butyrolactone, gamma- butyrolactone, gamma-valerolactone, γ-decanolactone, δ-decanolactone, trimethylene carbonate, 1 ,4-dioxane-2-one or 1 ,5-dioxepan-2one {e.g., PLG- PEG-PLG) and R is a multifunctional initiator, hyaluronic acid, carboxymethyl cellulose, dextran, poly(ethylene oxide) - poly(propylene oxide) block copolymers as well as blends, association complexes and crosslinked compositions thereof.
In yet another aspect, an activated polymer can be applied to the surgical site in the solid state. The activated polymer can react with the tissue surface to which it was applied as the polymer hydrates. A biologically acceptable buffer, with a pH greater than 7.5 can be applied to the tissue before and/or after the solid activated polymer has been applied. In one embodiment, the activated polymer can form a covalent bond with the tissue to which it is applied may be used. Polymers containing and/or terminated with electrophilic groups such as succinimidyl, aldehyde, epoxide, isocyanate, vinyl, vinyl sulfone, maleimide, -S-S-(C5H4N) or activated esters, such as are used in peptide synthesis may be used as the reagents. For example, a 4 armed NHS-derivatized polyethylene glycol (e.g., pentaerythritol poly(ethylene glycol)ether tetra-succinimidyl glutarate) may be applied to the tissue in the solid form. The antifibrosisfibrosis- inhibiting agent(s) may be incorporated directly into either the 4 armed NHS- derivatized polyethylene glycol, or the basic buffer. In another embodiment, the fibrosis-inhibiting agent may be incorporated into a secondary carrier that may then be incorporated into the 4 armed NHS-derivatized polyethylene glycol, and/or the basic buffer. The secondary carriers may include microparticles and/or microspheres which are made from degradable polymers. The degradable polymers may include polyesters, where the polyester may comprise the residues of one or more of the monomers selected from lactide, lactic acid, glycolide, glycolic acid, e- caprolactone, gamma-caprolactone, hydroxyvaleric acid, hydroxybutyric acid, beta-butyrolactone, gamma-butyrolactone, gamma-valerolactone, γ~ decanolactone, δ-decanolactone, trimethylene carbonate, 1 ,4-dioxane-2- one or 1,5-dioxepan-2one, and block copolymers of the form X-Y, Y-X-Y, R- (Y-X)n, R-(X-Y)n and X-Y-X where X in a polyalkylene oxide (e.g., poly(ethylene glycol, poly(propylene glycol) and block copolymers of poly(ethylene oxide) and poly(propylene oxide) (e.g., PLURONIC and PLURONIC R series of polymers from BASF Corporation, Mount Olive, NJ) and Y is a biodegradable polyester, where the polyester may comprise the residues of one or more of the monomers selected from lactide, lactic acid, glycolide, glycolic acid, e-caprolactone, gamma-caprolactone, hydroxy valeric acid, hydroxybutyric acid, beta-butyrolactone, gamma- butyrolactone, gamma-valerolactone, γ-decanolactone, δ-decanolactone, trimethylene carbonate, 1 ,4-dioxane-2-one or 1 ,5-dioxepan-2one (e.g., PLG- PEG-PLG) and R is a multifunctional initiator. In another embodiment, the tissue reactive polymer may be applied initially and then the fibrosis- inhibiting agent may then be applied to the coated tissue. The fibrosis- inhibiting agent may be applied directly to the tissue or it may be incorporated into a secondary carrier. The secondary carriers may include microspheres (as described above), microparticles (as described above), gels (e.g., hyaluronic acid, carboxymethyl cellulose, dextran, poly(ethylene oxide) - poly(propylene oxide) block copolymers as well as blends, association complexes and crosslinked compositions thereof) and films (degradable polyesters, where the polyester may comprise the residues of one or more of the monomers selected from lactide, lactic acid, glycolide, glycolic acid, e-caprolactone, gamma-caprolactone, hydroxyvaleric acid, hydroxybutyric acid, beta-butyrolactone, gamma-butyrolactone, gamma- valerolactone, γ-decanolactone, δ-decanolactone, trimethylene carbonate, 1 ,4-dioxane-2-one or 1 ,5-dioxepan-2one, and block copolymers of the form X-Y, Y-X-Y, R-(Y-X)n, R-(X-Y)n and X-Y-X where X in a polyalkylene oxide (e.g., poly(ethylene glycol, poly(propylene glycol) and block copolymers of poly(ethylene oxide) and poly(propylene oxide) (e.g., PLURONIC and PLURONIC R series of polymers from BASF Corporation, Mount Olive, NJ) and Y is a biodegradable polyester, where the polyester may comprise the residues of one or more of the monomers selected from lactide, lactic acid, glycolide, glycolic acid, e-caprolactone, gamma-caprolactone, hydroxyvaleric acid, hydroxybutyric acid, beta-butyrolactone, gamma- butyrolactone, gamma-valerolactone, γ-decanolactone, δ-decanolactone, trimethylene carbonate, 1 ,4-dioxane-2-one or 1,5-dioxepan-2one (e.g., PLG- PEG-PLG) and R is a multifunctional initiator, hyaluronic acid, carboxymethyl cellulose, dextran, poly(ethylene oxide) - poly(propylene oxide) block copolymers as well as blends, association complexes and crosslinked compositions thereof
ii) Adhesion Prevention in Gynecological Procedures In one aspect, adhesion formation may be associated with a gynecological surgical procedure. The post-operative adhesions occur in 60 to 90% of patients undergoing major gynecologic surgery and represent one of the most common causes of infertility in the industrialized world. Adhesions can form between the ovaries, the fallopian tubes, the bowel or the walls of the pelvis. Fibrous bands can connect to the normally mobile adnexal structures (ovaries and fallopian tubes) to other tissues, causing them to lose mobility, kink or twist. If the adhesions tighten around, constrict or twist the fallopian tubes themselves, they can block the passage of an ovum from the ovaries into and through the fallopian tube leading to infertility. Adhesions around the fallopian tubes can also interfere with sperm transport to the ovum and also cause infertility. Other adhesion- related complications include chronic pelvic pain, dysparunia, urethral obstruction and voiding dysfunction.
Several products are available commercially or under development for the management of gynecological adhesions. Life Medical Sciences, Inc. is producing the products, REPEL, REPEL-CV, RESOLVE and RELIEVE that are in various stages of development and may be used to prevent surgical adhesions in gynecological and other surgeries. Products being developed by Life Medical Sciences, Inc. are described in, for example, U.S. Patent Nos. 6,696,499; 6,399,624; 6,211,249; 6,136,333 and 5,711 ,958. Confluent Surgical, Inc. makes their SPRAYGEL which is a unique sprayable adhesion barrier that is being developed for use in pelvic and intrauterine surgical procedures. Products that are being developed by Confluent Surgical, Inc. are described in, for example, U.S. Patent No. 6,379,373. Closure Medical Corp. (Raleigh, NC) is developing a cyanoacrylate-based internal adhesives that may be used to seal internal surgical incisions or grafts which may be compatible in gynecology and general surgical specialties. Products that are being developed by Closure Medical, Corp. are described in, for example, U.S. Patent Nos. 6,620,846; 6,579,469; 6,565,840; 6,547,467 and 5,981,621.
Other commercially available materials that may be used alone, or loaded with a therapeutic agent (e.g., a fibrosis-inhibiting agent and/or an anti-infective agent), applied to or infiltrated into a gynecological surgical site (or to the surface of a device or implant) for the prevention of adhesions in open or endoscopic gynecologic surgery include: (a) sprayable collagen-containing formulations such as COSTASIS or CT3; (b) sprayable PEG-containing formulations such as COSEAL, ADHIBIT, FOCALSEAL or DURASEAL; (c) fibrinogen-containing formulations such as FLOSEAL or TISSEAL; (d) hyaluronic acid-containing formulations such as RESTYLANE or PERLANE, HYLAFORM, SYNVlSC, SEPRAFILM or SEPRACOAT; (e) polymeric gels for surgical implantation such as FLOWGEL; (f) surgical adhesives containing cyanoacrylates such as DERMABOND, INDERMIL, GLUSTITCH, TISSUMEND, VETBOND, HISTOACRYL BLUE and ORABASE SOOTHE-N-SEAL LIQUID PROTECTANT; (g) dextran sulfate gels such as the ADCON series of gels; and (h) lipid based compositions such as ADSURF. It should be obvious to one of skill in the art that commercial compositions not specifically cited above as well as next-generation and/or subsequently-developed commercial products are to be anticipated and are suitable for use under the present invention.
Gynecological procedures are performed for a variety of medical conditions including hysterectomy (removal of the uterus), myomectomy (removal of uterine fibroids), endometriosis (ablation procedures), infertility (in vitro fertilization, adhesiolysis), birth control (tubal ligation), reversal of sterilization, pain, dysmennorrhea, dysfunctional uterine bleeding, ectopic pregnancy, ovarian cysts, gynecologic malignancies and numerous other conditions. Although many procedures are still performed through open surgical techniques, increasingly, gynecologic surgery is performed via an endoscope inserted through the umbilicus (belly button). Virtually any manipulation of the pelvic organs or pelvic sidewall can trigger a cascade that ultimately results in the formation of pelvic adhesions. In many instances, the adhesions must be broken down during a repeat surgical intervention for the treatment of pain or infertility. An adhesion barrier, either alone or containing a fibrosis-inhibiting agent (and/or an anti- infective agent), is best applied directly to the affected areas (as a solid, a film, a paste, a gel, a liquid or another such formulation) during the open or endoscopic procedure. In a preferred embodiment, the barrier (alone or containing an anti-fibrotic and/or anti-infective agent) is sprayed under direct endoscopic vision during the procedure onto the pelvic organs (and bowel, pelvic and abdominal sidewall) that are operated on, or manipulated, during the intervention. Since adhesions often occur in areas at a distance from the tissues actually instrumented during a surgical intervention, it is recommended that the barrier (with or without a therapeutic agent) be applied to a wide area in the pelvis (potentially even the entire adnexa, pelvic sidewall and pelvic surface of the uterus). Preferred barriers include liquids, gels, pastes, sprays or other formulations that can be delivered through an endoscope, adhere to the tissues treated, and remain in place long enough to deliver the therapeutic agent and/or prevent adhesion formation. As an alternative, the therapeutic agent can be delivered directly into the peritoneal cavity as an injectable (either before, during or after the procedure) such that the drug is delivered in doses high enough and long enough (multiple dosing and/or sustained release preparations are preferred) to prevent adhesions and the complications arising from them. An ideal adhesion therapy will reduce the incidence, number and tenacity of adhesions and improve patient outcome by reducing pain, improving fertility and limiting the need for repeat interventions.
As described above, the compositions for the prevention of surgical adhesions can be applied directly or indirectly to the tissue in a gynecological site. The polymeric compositions (either with or without an anti-fibrotic or anti-infective therapeutic agent) can be administered in any manner described herein. Exemplary methods include either direct application at the time of surgery or with endoscopic, ultrasound, CT, MRI, or fluoroscopic guidance. If an implanted device is being placed, the composition for the prevention of adhesions can be applied to the surface of the implant, or to the surrounding tissues, in conjunction with placement of a medical device or implant at the surgical site. Representative examples of implants for use in gynecological procedures includes, without limitation, genital-urinary stents, bulking agents, sterilization devices (e.g., valves, clips and clamps), and tubal occlusion implants and plugs.
The polymeric composition,, with or without a fibrosis-inhibiting agent, may be applied during open or endoscopic gynecological surgery: (a) to the tissue surface of the pelvic side wall, adnexa, uterus and any adjacent affected tissues (e.g., as an injectable, solution, paste, gel, in situ forming gel or mesh) during the surgical procedure; (b) to the surface of an implanted device or implant and/or the tissue surrounding the implant (e.g., as an injectable, solution, paste, gel, in situ forming gel or mesh) before, during, or after the surgical procedure; (c) by intraperitoneal or endoscopic injection of the composition into the anatomical space (i.e., the peritoneal or pelvic cavity) at the surgical site (particularly useful for this embodiment is the use of injectable compositions containing polymeric carriers which release the fibrosis-inhibiting agent over a period ranging from several hours to several weeks - fluids, suspensions, emulsions, microemulsions, microspheres, pastes, gels, microparticulates, sprays, aerosols, solid implants and other formulations which release the agent and can be delivered into the region where there is a risk of adhesion formation); (d) via percutaneous injection into the tissue as a solution as an infusate or as a sustained release preparation; (e) by guided catheter or hysteroscopic injection of the composition into the lumen of the fallopian tubes (i.e., inserting a catheter or an endoscope via the vagina, cervix and uterus until it can be advanced into the lumen of the fallopian tube) at the desired tubal location (particularly useful for this embodiment is the use of injectable compositions containing polymeric carriers which release the fibrosis- inhibiting agent over a period ranging from several hours to several weeks - fluids, suspensions, emulsions, microemulsions, microspheres, pastes, gels, microparticulates, sprays, aerosols, solid implants and other formulations which release the agent can be delivered into the areas of the fallopian tube where there is a risk of adhesion formation); and/or (f) by any combination of the aforementioned methods. Combination therapies (Ae., combinations of therapeutic agents and combinations with antithrombotic, anti-infective, and/or antiplatelet agents) can also be used in the manner described above.
In. certain applications involving the placement of a gynecological medical device or implant, it may be desirable to apply the anti-fibrosis (and/or anti-infective) composition at a site that is adjacent to an implant (preferably near the implant-tissue interface). This can be accomplished during open or endoscopic procedures by applying the polymeric composition, with or without a fibrosis-inhibiting agent: (a) to the implant surface (e.g., as an injectable, solution, paste, gel, in situ forming gel, or mesh) before, during, or after the implantation procedure; (b) to the surface of the adjacent tissue (e.g., as an injectable, solution, paste, gel, in situ forming gel, or mesh) immediately prior to, during, or after implantation of the implant; (c) to the surface of the implant and the tissue surrounding the implant (e.g., as an injectable, solution, paste, gel, in situ forming gel or mesh) before, during, or after implantation of the implant; (d) by topical application of the composition into the anatomical space (such as the lumen of the fallopian tube, the uterine cavity, the peritoneal cavity, or the pelvic cavity) where the implant will be placed (particularly useful for this embodiment is the use of polymeric carriers which release the fibrosis- inhibiting agent over a period ranging from several hours to several weeks - fluids, suspensions, emulsions, microemulsions, microspheres, pastes, gels, microparticulates, sprays, aerosols, solid implants and other formulations which release the agent and can be delivered into the region where the device will be inserted); (e) via percutaneous injection into the tissue surrounding the implant as a solution, as an infusate, or as a sustained release preparation; and/or (f) by any combination of the aforementioned methods. Combination therapies (i.e., combinations of therapeutic agents and combinations with antithrombotic, anti-infective, and/or antiplatelet agents) can also be used.
In one aspect, the polymeric composition may be delivered to the female pelvic tissue (or device/tissue interface) in the form of a spray or gel during open, endoscopic or catheter-based procedures. The fibrosis- inhibiting agent can be incorporated directly into the surgical adhesion barrier or it can be incorporated into a secondary carrier (polymeric or non- polymeric), as described above, that is then incorporated into the adhesion barrier. Examples of polymer compositions that may be in the form of a spray or gel include poly(ethylene glycol)-based systems, hyaluronic acid and crosslinked hyaluronic acid compositions. These compositions can be applied as the final composition or they can be applied as materials that form a crosslinked gel in situ.
In another aspect, an activated polymer is dissolved in a biologically acceptable buffer that has a pH lower that 6.8. The resultant solution is then applied to the desired tissue surface in the presence of a second biologically acceptable buffer that has a pH greater than 7.5. Application of the reaction mixture to the tissue site may be by extrusion, brushing, spraying or by any other convenient means. Following application of the composition to the surgical site, any excess solution may be removed from the surgical site if deemed necessary. At this point in time, the surgical site can be closed using conventional means (e.g., sutures, staples, or a bioadhesive). In one embodiment, the activated polymer can form a covalent bond with the tissue to which it is applied may be used. Polymers containing and/or terminated with electrophilic groups such as succinimidyl, aldehyde, epoxide, isocyanate, vinyl, vinyl sulfone, maleimide, -S-S-(CsH4N) or activated esters, such as are used in peptide synthesis may be used as the reagents. For example, a 4 armed NHS-derivatized polyethylene glycol {e.g., pentaerythritol poly(ethylene glycol)ether tetra-succinimidyl glutarate) may be applied to the tissue in the solid form or in a solution form. In this embodiment, the 4 armed NHS-derivatized polyethylene glycol is dissolved in an acidic solution (pH about 2-3) and is then co-applied to the tissue using a basic buffer (pH > about 8). The antifibrosisfibrosis-inhibiting agent(s) may be incorporated directly into either the 4 armed NHS- derivatized polyethylene glycol, the acidic solution or the basic buffer. In another embodiment, the fibrosis-inhibiting agent may be incorporated into a secondary carrier that may then be incorporated into the 4 armed NHS- derivatized polyethylene glycol, the acidic solution and/or the basic buffer. The secondary carriers may include microparticles and/or microspheres which are made from degradable polymers. The degradable polymers may include polyesters, where the polyester may comprise the residues of one or more of the monomers selected from lactide, lactic acid, glycolide, glycolic acid, e-caprolactone, gamma-caprolactone, hydroxyvaleric acid, hydroxybutyric acid, beta-butyrolactone, gamma-butyrolactone, gamma- valerolactone, γ-decanolactone, δ-decanolactone, trimethylene carbonate, 1 ,4-dioxane-2-one or 1,5-dioxepan-2one, and block copolymers of the form X-Y, Y-X-Y, R-(Y-X)n, R-(X-Y)n and X-Y-X where X in a polyalkylene oxide (e.g., poly(ethylene glycol, polypropylene glycol) and block copolymers of poly(ethylene oxide) and polypropylene oxide) (e.g., PLURONIC and PLURONIC R series of polymers from BASF Corporation, Mount Olive, NJ) and Y is a biodegradable polyester, where the polyester may comprise the residues of one or more of the monomers selected from lactide, lactic acid, glycolide, glycolic acid, e-caprolactone, gamma-caprolactone, hydroxyvaleric acid, hydroxybutyric acid, beta-butyrolactone, gamma- butyrolactone, gamma-valerolactone, γ-decanolactone, δ-decanolactone, trimethylene carbonate, 1 ,4-dioxane-2~one or 1 ,5-dioxepan-2one (e.g., PLG- PEG-PLG) and R is a multifunctional initiator. In another embodiment, the tissue reactive polymer may be applied initially and then the fibrosis- inhibiting agent may then be applied to the coated tissue. The fibrosis- inhibiting agent may be applied directly to the tissue or it may be incorporated into a secondary carrier. The secondary carriers may include microspheres (as described above), microparticles (as described above), gels (e.g., hyaluronic acid, carboxymethyl cellulose, dextran, poly(ethylene oxide) - poly(propylene oxide) block copolymers as well as blends, association complexes and crosslinked compositions thereof) and films (degradable polyesters, where the polyester may comprise the residues of one or more of the monomers selected from lactide, lactic acid, glycolide, glycolic acid, e-caprolactone, gamma-caprolactone, hydroxyvaleric acid, hydroxybutyric acid, beta-butyrolactone, gamma-butyrolactone, gamma- valerolactone, γ-decanolactone, δ-decanolactone, trimethylene carbonate, 1 ,4-dioxane-2-one or 1 ,5-dioxepan-2one, and block copolymers of the form X-Y, Y-X-Y, R-(Y-X)n, R-(X-Y)n and X-Y-X where X in a polyalkylene oxide (e.g., poly(ethylene glycol, poly(propylene glycol) and block copolymers of poly(ethylene oxide) and poly(propylene oxide) (e.g., PLURONIC and PLURONIC R series of polymers from BASF Corporation, Mount Olive, NJ) and Y is a biodegradable polyester, where the polyester may comprise the residues of one or more of the monomers selected from lactide, lactic acid, glycolide, glycolic acid, e-caprolactone, gamma-caprolactone, hydroxyvaleric acid, hydroxybutyric acid, beta-butyrolactone, gamma- butyrolactone, gamma-valerolactone, γ-decanolactone, δ-decanolactone, trimethylene carbonate, 1,4-dioxane-2-one or 1,5-dioxepan-2one (e.g., PLG- PEG-PLG) and R is a multifunctional initiator, hyaluronic acid, carboxymethyl cellulose, dextran, poly(ethylene oxide) - poly(propylene oxide) block copolymers as well as blends, association complexes and crosslinked compositions thereof.
In yet another aspect, an activated polymer can be applied to the surgical site in the solid state. The activated polymer can react with the tissue surface to which it was applied as the polymer hydrates. A biologically acceptable buffer, with a pH greater than 7.5 can be applied to the tissue before and/or after the solid activated polymer has been applied. In one embodiment, the activated polymer can form a covalent bond with the tissue to which it is applied may be used. Polymers containing and/or terminated with electrophilic groups such as succinimidyl, aldehyde, epoxide, isocyanate, vinyl, vinyl sulfone, maleimide, -S-S-(C5H4N) or activated esters, such as are used in peptide synthesis may be used as the reagents. For example, a 4 armed NHS-derivatized polyethylene glycol (e.g., pentaerythritol poly(ethylene glycol)ether tetra-succinimidyl glutarate) may be applied to the tissue in the solid form. The antifibrosisfibrosis- inhibiting agent(s) may be incorporated directly into either the 4 armed NHS- derivatized polyethylene glycol, or the basic buffer. In another embodiment, the fibrosis-inhibiting agent may be incorporated into a secondary carrier that may then be incorporated into the 4 armed NHS-derivatized polyethylene glycol, and/or the basic buffer. The secondary carriers may include microparticles and/or microspheres which are made from degradable polymers. The degradable polymers may include polyesters, where the polyester may comprise the residues of one or more of the monomers selected from lactide, lactic acid, glycolide, glycolic acid, e- caprolactone, gamma-caprolactone, hydroxyvaleric acid, hydroxybutyric acid, beta-butyrolactone, gamma-butyrolactone, gamma-valerolactone, y- decanolactone, δ-decanolactone, trimethylene carbonate, 1,4-dioxane-2- one or 1 ,5-dioxepan-2one, and block copolymers of the form X-Y, Y-X-Y, R- (Y-X)n, R-(X-Y)n and X-Y-X where X in a polyalkylene oxide (e.g., poly(ethylene glycol, poly(propylene glycol) and block copolymers of poly(ethylene oxide) and polypropylene oxide) (e.g., PLURONIC and PLURONIC R series of polymers from BASF Corporation, Mount Olive, NJ) and Y is a biodegradable polyester, where the polyester may comprise the residues of one or more of the monomers selected from lactide, lactic acid, glycolide, glycolic acid, e-caprolactone, gamma-caprolactone, hydroxyvaleric acid, hydroxy butyric acid, beta-butyrolactone, gamma- butyrolactone, gamma-valerolactone, γ-decanolactone, δ-decanolactone, trimethylene carbonate, 1 ,4-dioxane-2-one or 1 ,5-dioxepan-2one (e.g., PLG- PEG-PLG) and R is a multifunctional initiator. In another embodiment, the tissue reactive polymer may be applied initially and then the fibrosis- inhibiting agent may then be applied to the coated tissue. The fibrosis- inhibiting agent may be applied directly to the tissue or it may be incorporated into a secondary carrier. The secondary carriers may include microspheres (as described above), microparticles (as described above), gels (e.g., hyaluronic acid, carboxymethyl cellulose, dextran, poly(ethylene oxide) - poly(propylene oxide) block copolymers as well as blends, association complexes and crosslinked compositions thereof) and films (degradable polyesters, where the polyester may comprise the residues of one or more of the monomers selected from lactide, lactic acid, glycolide, glycolic acid, e-caprolactone, gamma-caprolactone, hydroxyvaleric acid, hydroxybutyric acid, beta-butyrolactone, gamma-butyrolactone, gamma- valerolactone, Y-decanolactone, δ-decanolactone, trimethylene carbonate, 1 ,4-dioxane-2-one or 1,5-dioxepan-2one, and block copolymers of the form X-Y1 Y-X-Y, R-(Y-X)n, R-(X-Y)n and X-Y-X where X in a polyalkylene oxide (e.g., poly(ethylene glycol, poly(propylene glycol) and block copolymers of poly(ethylene oxide) and poly(propylene oxide) (e.g., PLURONIC and PLURONIC R series of polymers from BASF Corporation, Mount Olive, NJ) and Y is a biodegradable polyester, where the polyester may comprise the residues of one or more of the monomers selected from lactide, lactic acid, glycolide, glycolic acid, e-caprolactone, gamma-caprolactone, hydroxyvaleric acid, hydroxybutyric acid, beta-butyrolactone, gamma- butyrolactone, gamma-valerolactone, γ-decanolactone, δ-decanolactone, trimethylene carbonate, 1 ,4-dioxane-2-one or 1,5-dioxepan-2one (e.g., PLG- PEG-PLG) and R is a multifunctional initiator, hyaluronic acid, carboxymethyl cellulose, dextran, poly(ethylene oxide) - poly(propylene oxide) block copolymers as well as blends, association complexes and crosslinked compositions thereof.
iii) Adhesion Prevention in Abdominal Procedures In one aspect, adhesions may be associated with an abdominal surgical procedure. Following abdominal surgery, the formation of adhesions may cause loops of intestines become entangled or twisted about fibrous bands of tissue that impair the normal fluid movement of the bowel. The entanglements can cause partial or total flow obstruction through the bowel, scar can constrict around the bowel, volvulus (twisting) can occur, or blood flow to and from the bowel can be compromised. With entanglement, volvulus or fibrous banding the result is typically partial or complete bowel obstruction; a condition that requires immediate decompression, may require surgery and can cause death. Infarction (interruption of blood flow to the bowel) from adhesions or volvulus is a medical emergency that usually requires surgical removal of the affected bowel and can also lead to death if not treated aggressively. Peritoneal adhesions (adhesions between the abdominal wall and the underlying organs) represent another major health care problem causing pain, bowel obstruction and other potentially serious post-operative complications and they are associated with all types of abdominal surgery (incidence of 50- 90% for laparotomies).
As described previously, adhesion barriers are frequently used in the management of abdominal adhesions following open or endoscopic procedures. A variety of commercially available adhesion barriers are suitable for combining with a fibrosis-inhibitor (and/or an anti-infective agent) in the management of abdominal adhesions. Confluent Surgical, Inc. makes their SPRAYGEL which is a unique sprayable adhesion barrier that is being developed for use in abdominal and pelvic surgical procedures. Products that are being developed by Confluent Surgical, Inc. are described in, for example, U.S. Patent No. 6,379,373. Closure Medical Corp. (Raleigh, NC) is developing a cyanoacrylate-based internal adhesives that may be used to seal internal surgical incisions or grafts which may be compatible in gastrointestinal, oncology and general surgical specialties. Products that are being developed by Closure Medical, Corp. are described in, for example, U.S. Patent Nos. 6,620,846; 6,579,469; 6,565,840; 6,547,467 and 5,981 ,621. Genzyme Corporation has developed hyaluronic acid-containing biomaterials, such as SEPRAFILM and SEPRACOAT, to reduce the incidence of adhesions following abdominal and pelvic surgeries (see, e.g., U.S. Patent Nos. 6,780,427; 6,531 ,147; 6,521 ,223 and 6,010,692.
Other commercially available materials that may be used alone, or loaded with a therapeutic agent (e.g., a fibrosis-inhibiting agent or an anti-infective agent), applied to or infiltrated into an abdominal site (or to the surface of an implanted device or implant) for the prevention of adhesions during open or endoscopic abdominal procedures include: (a) sprayable collagen-containing formulations such as COSTASIS or CT3; (b) sprayable PEG-containing formulations such as COSEAL, ADHIBIT, FOCALSEAL or DURASEAL; (c) fibrinogen-containing formulations such as FLOSEAL or TISSEAL; (d) hyaluronic acid-containing formulations such as RESTYLANE or PERLANE, HYLAFORM, or SYNVISC; (e) polymeric gels for surgical implantation such as REPEL or FLOWGEL; (f) surgical adhesives containing cyanoacrylates such as DERMABOND, INDERMIL, GLUSTITCH, TISSUMEND, VETBOND, HISTOACRYL BLUE and ORABASE SOOTHE-N-SEAL LIQUID PROTECTANT; (g) dextran sulfate gels such as the ADCON series of gels; and (h) lipid based compositions such as ADSURF. It should be obvious to one of skill in the art that commercial compositions not specifically cited above as well as next- generation and/or subsequently-developed commercial products are to be anticipated and are suitable for use under the present invention. Abdominal surgical procedures are performed for a variety of medical conditions including hernia repair (abdominal, ventral, inguinal, incisional), bowel obstruction, inflammatory bowel disease (ulcerative colitis, Crohn's disease), appendectomy, trauma (penetrating wounds, blunt tauma), tumor resection, infections (abscesses, peritonitis), cholecystectomy, gastroplasty (bariatric surgery), esophageal and pyloric strictures, colostomy, diversion iliostomy, anal-rectal fistulas, hemorrhoidectomies, splenectomy, hepatic tumor resection, pancreatitis, bowel perforation, upper and lower Gl bleeding, and ischemic bowel. Although many procedures are still performed through open surgical techniques, increasingly, abdominal surgery is performed via an endoscope inserted through the umbilicus (belly button). Virtually any manipulation of the abdominal viscera or peritoneum can trigger a cascade that ultimately results in the formation of abdominal adhesions. In many instances, the adhesions must be broken down during a repeat surgical intervention for the treatment of pain or bowel obstruction. An adhesion barrier, either alone or containing a fibrosis-inhibiting agent (and/or an anti-infective agent), is best applied directly to the affected areas (as a solid, a film, a paste, a gel, a liquid or another such formulation) during the open or endoscopic procedure. In a preferred embodiment, the barrier (alone or containing an anti-fibrotic and/or anti-infective agent) is sprayed under direct or endoscopic vision during the procedure onto the abdominal organs (such as the large and small bowel, stomach, liver, spleen, gall bladder etc.), visceral peritoneum and abdominal (wall) peritoneum that are operated on, or manipulated, during the intervention. Since adhesions often occur in areas at a distance from the tissues actually instrumented during a surgical intervention, it is recommended that the barrier (with or without a therapeutic agent) be applied to a wide area in the abdomen (potentially even the entire viscera and abdominal wall). Preferred barriers include films, liquids, gels, pastes, sprays or other formulations that can be delivered during open procedures or through an endoscope, adhere to the tissues treated, and remain in place long enough to deliver the therapeutic agent and/or prevent adhesion formation. As an alternative, the therapeutic agent can be delivered directly into the peritoneal cavity as an injectable (either before, during or after the procedure) such that the drug is delivered in doses high enough and long enough (multiple dosing and/or sustained release preparations are preferred) to prevent adhesions and the complications arising from them. An ideal adhesion therapy will reduce the incidence, number and tenacity of adhesions and improve patient outcome by reducing pain, preventing bowel obstruction and limiting the need for repeat interventions.
As described above, the compositions for the prevention of surgical adhesions can be applied directly or indirectly to the tissue in an abdominal procedure. The polymeric compositions (either with or without an anti-fibrotic or anti-infective therapeutic agent) can be administered in any manner described herein. Exemplary methods include either direct application at the time of surgery or with endoscopic, ultrasound, CT, MRI, or fluoroscopic guidance. If an implanted device is being placed, the composition for the prevention of adhesions can be applied to the surface of the implant, or to the surrounding tissues, in conjunction with placement of a medical device or implant at the surgical site. Representative examples of implants for use in abdominal procedures includes, without limitation, hernia meshes, restriction devices for obesity, implantable sensors, implantable pumps, peritoneal dialysis catheters, peritoneal drug-delivery catheters, Gl tubes for drainage or feeding, portosystemic shunts, shunts for ascites, gastrostomy or percutaneous feeding tubes, jejunostomy endoscopic tubes, colostomy devices, drainage tubes, biliary T-tubes, hemostatic implants, enteral feeding devices, colonic and biliary stents, low profile devices, gastric banding implants, capsule endoscopes, anti-reflux devices, and esophageal stents.
The polymeric composition, with or without a fibrosis-inhibiting agent, may be applied during open or endoscopic abdominal surgery: (a) to the tissue surface of the peritoneal cavity, visceral peritneum, abdominal organs, abdominal wall and any adjacent affected tissues (e.g., as an injectable, solution, paste, gel, in situ forming gel or mesh) during the surgical procedure; (b) to the surface of an implanted device or implant and/or the tissue surrounding the implant (e.g., as an injectable, solution, paste, gel, in situ forming gel or mesh) before, during, or after the surgical procedure; (c) by intraperitoneal or endoscopic injection of the composition into the anatomical space (i.e., the peritoneal cavity) at the surgical site (particularly useful for this embodiment is the use of injectable compositions containing polymeric carriers which release the fibrosis-inhibiting agent over a period ranging from several hours to several weeks - fluids, suspensions, emulsions, microemulsions, microspheres, pastes, gels, microparticulates, sprays, aerosols, solid implants and other formulations which release the agent and can be delivered into the region where there is a risk of adhesion formation); (d) via percutaneous injection into the tissue as a solution as an infusate or as a sustained release preparation; (e) by guided catheter or endoscopic (gastroscope, ERCP, colonoscope) injection of the composition into the lumen of the Gl tract at the desired location (particularly useful for this embodiment is the use of injectable compositions containing polymeric carriers which release the fibrosis-inhibiting agent over a period ranging from several hours to several weeks - fluids, suspensions, emulsions, microemulsions, microspheres, pastes, gels, microparticulates, sprays, aerosols, solid implants and other formulations which release the agent can be delivered into the areas of the Gl tract where there is a risk of adhesion formation); and/or (f) by any combination of the aforementioned methods. Combination therapies (i.e., combinations of therapeutic agents and combinations with antithrombotic, anti-infective, and/or antiplatelet agents) can also be used in the manner described above.
In certain applications involving the placement of an abdominal or gastrointestinal medical device or implant, it may be desirable to apply the anti-fibrosis (and/or anti-infective) composition at a site that is adjacent to an implant (preferably near the implant-tissue interface). This can be accomplished during open or endoscopic procedures by applying the polymeric composition, with or without a fibrosis-inhibiting agent: (a) to the implant surface (e.g., as an injectable, solution, paste, gel, in situ forming gel, or mesh) before, during, or after the implantation procedure; (b) to the surface of the adjacent tissue {e.g., as an injectable, solution, paste, gel, in situ forming gel, or mesh) immediately prior to, during, or after implantation of the implant; (c) to the surface of the implant and the tissue surrounding the implant (e.g., as an injectable, solution, paste, gel, in situ forming gel or mesh) before, during, or after implantation of the implant; (d) by topical application of the composition into the anatomical space (such as the lumen of the Gl tract or the peritoneal cavity) where the implant will be placed (particularly useful for this embodiment is the use of polymeric carriers which release the fibrosis-inhibiting agent over a period ranging from several hours to several weeks - fluids, suspensions, emulsions, microemulsions, microspheres, pastes, gels, microparticulates, sprays, aerosols, solid implants and other formulations which release the agent and can be delivered into the region where the device will be inserted); (e) via percutaneous injection into the tissue surrounding the implant as a solution, as an infusate, or as a sustained release preparation; and/or (f) by any combination of the aforementioned methods. Combination therapies (i.e., combinations of therapeutic agents and combinations with antithrombotic, anti-infective, and/or antiplatelet agents) can also be used.
In one aspect, the polymeric composition may be delivered to the abdomen (or device/tissue interface) in the form of a spray or gel during open, endoscopic or catheter-based procedures. The fibrosis-inhibiting agent can be incorporated directly into the surgical adhesion barrier or it can be incorporated into a secondary carrier (polymeric or non-polymeric), as described above, that is then incorporated into the adhesion barrier. Examples of polymer compositions that may be in the form of a spray or gel include poly(ethylene glycol)-based systems, hyaluronic acid and crosslinked hyaluronic acid compositions. These compositions can be applied as the final composition or they can be applied as materials that form a crosslinked gel in situ.
In another aspect, an activated polymer is dissolved in a biologically acceptable buffer that has a pH lower that 6.8. The resultant solution is then applied to the desired tissue surface in the presence of a second biologically acceptable buffer that has a pH greater than 7.5. Application of the reaction mixture to the tissue site may be by extrusion, brushing, spraying or by any other convenient means. Following application of the composition to the surgical site, any excess solution may be removed from the surgical site if deemed necessary. At this point in time, the surgical site can be closed using conventional means (e.g., sutures, staples, or a bioadhesive). In one embodiment, the activated polymer can form a covalent bond with the tissue to which it is applied may be used. Polymers containing and/or terminated with electrophilic groups such as succinimidyl, aldehyde, epoxide, isocyanate, vinyl, vinyl sulfone, maleimide; -S-S-(C5H4N) or activated esters, such as are used in peptide synthesis may be used as the reagents. For example, a 4 armed NHS-derivatized polyethylene glycol (e.g., pentaerythritol poly(ethylene glycol)ether tetra-succinimidyl glutarate) may be applied to the tissue in the solid form or in a solution form. In this embodiment, the 4 armed NHS-derivatized polyethylene glycol is dissolved in an acidic solution (pH about 2-3) and is then co-applied to the tissue using a basic buffer (pH > about 8). The antifibrosisfibrosis-inhibiting agent(s) may be incorporated directly into either the 4 armed NHS- derivatized polyethylene glycol, the acidic solution or the basic buffer. In another embodiment, the fibrosis-inhibiting agent may be incorporated into a secondary carrier that may then be incorporated into the 4 armed NHS- derivatized polyethylene glycol, the acidic solution and/or the basic buffer. The secondary carriers may include microparticles and/or microspheres which are made from degradable polymers. The degradable polymers may include polyesters, where the polyester may comprise the residues of one or more of the monomers selected from lactide, lactic acid, glycolide, glycolic acid, e-caprolactone, gamma-caprolactone, hydroxy valeric acid, hydroxy butyric acid, beta-butyrolactone, gamma-butyrolactone, gamma- valerolactone, γ-decanolactone, δ-decanolactone, trimethylene carbonate, 1 ,4-dioxane-2-one or 1 ,5-dioxepan-2one, and block copolymers of the form X-Y, Y-X-Y, R-(Y-X)n, R-(X-Y)n and X-Y-X where X in a polyalkylene oxide (e.g., poly(ethylene glycol, poly(propylene glycol) and block copolymers of poly(ethylene oxide) and polypropylene oxide) (e.g., PLURONIC and PLURONIC R series of polymers from BASF Corporation, Mount Olive, NJ) and Y is a biodegradable polyester, where the polyester may comprise the residues of one or more of the monomers selected from lactide, lactic acid, glycolide, glycolic acid, e-caprolactone, gamma-caprolactone, hydroxyvaleric acid, hydroxybutyric acid, beta-butyrolactone, gamma- butyrolactone, gamma-valerolactone, γ-decanolactone, δ-decanolactone, trimethylene carbonate, 1 ,4-dioxane-2-one or 1 ,5-dioxepan-2one (e.g., PLG- PEG-PLG) and R is a multifunctional initiator.. In another embodiment, the tissue reactive polymer may be applied initially and then the fibrosis- inhibiting agent may then be applied to the coated tissue. The fibrosis- inhibiting agent may be applied directly to the tissue or it may be incorporated into a secondary carrier. The secondary carriers may include microspheres (as described above), microparticles (as described above), gels (e.g., hyaluronic acid, carboxymethyl cellulose, dextran, poly(ethylene oxide) - poly(propylene oxide) block copolymers as well as blends, association complexes and crosslinked compositions thereof) and films (degradable polyesters, where the polyester may comprise the residues of one or more of the monomers selected from lactide, lactic acid, glycolide, glycolic acid, e-caprolactone, gamma-caprolactone, hydroxyvaleric acid, hydroxybutyric acid, beta-butyrolactone, gamma-butyrolactone, gamma- valerolactone, γ-decanolactone, δ-decanolactone, trimethylene carbonate, 1 ,4-dioxane-2-one or 1,5-dioxepan-2one, and block copolymers of the form X-Y, Y-X-Y, R-(Y-X)n, R-(X-Y)n and X-Y-X where X in a polyalkylene oxide (e.g., poly(ethylene glycol, poly(propylene glycol) and block copolymers of poly(ethylene oxide) and polypropylene oxide) (e.g., PLURONIC and PLURONIC R series of polymers from BASF Corporation, Mount Olive, NJ) and Y is a biodegradable polyester, where the polyester may comprise the residues of one or more of the monomers selected from lactide, lactic acid, glycolide, glycolic acid, e-caprolactone, gamma-caprolactone, hydroxyvaleric acid, hydroxybutyric acid, beta-butyrolactone, gamma- butyrolactone, gamma-valerolactone, γ-decanolactone, δ-decanolactone, trimethylene carbonate, 1 ,4-dioxane-2-one or 1 ,5-dioxepan-2one (e.g., PLG- PEG-PLG) and R is a multifunctional initiator, hyaluronic acid, carboxymethyl cellulose, dextran, poly(ethylene oxide) - polyφropylene oxide) block copolymers as well as blends, association complexes and crosslinked compositions thereof.
In yet another aspect, an activated polymer can be applied to the surgical site in the solid state. The activated polymer can react with the tissue surfacelo which it was. applied as the polymer hydrates. A biologically acceptable buffer, with a pH greater than 7.5 can be applied to the tissue before and/or after the solid activated polymer has been applied. In one embodiment, the activated polymer can form a covalent bond with the tissue to which it is applied may be used. Polymers containing and/or terminated with electrophilic groups such as succinimidyl, aldehyde, epoxide, isocyanate, vinyl, vinyl sulfone, maleimide, -S-S-(C5H4N) or activated esters, such as are used in peptide synthesis may be used as the reagents. For example, a 4 armed NHS-derivatized polyethylene glycol (e.g., pentaerythritol poly(ethylene glycol)ether tetra-succinimidyl glutarate) may be applied to the tissue in the solid form. The antifibrosisfibrosis- inhibiting agent(s) may be incorporated directly into either the 4 armed NHS- derivatized polyethylene glycol, or the basic buffer. In another embodiment, the fibrosis-inhibiting agent may be incorporated into a secondary carrier that may then be incorporated into the 4 armed NHS-derivatized polyethylene glycol, and/or the basic buffer. The secondary carriers may include microparticles and/or microspheres which are made from degradable polymers. The degradable polymers may include polyesters, where the polyester may comprise the residues of one or more of the monomers selected from lactide, lactic acid, glycolide, glycolic acid, e- caprolactone, gamma-caprolactone, hydroxy valeric acid, hydroxybutyric acid, beta-butyrolactone, gamma-butyrolactone, gamma-valerolactone, γ~ decanolactone, δ-decanolactone, trimethylene carbonate, 1 ,4-dioxane-2- one or 1 ,5-dioxepan-2one, and block copolymers of the form X-Y, Y-X-Y, R- (Y-X)n, R-(X-Y)n and X-Y-X where X in a polyalkylene oxide (e.g., poly(ethylene glycol, poly(propylene glycol) and block copolymers of poly(ethylene oxide) and polypropylene oxide) (e.g., PLURONIC and PLURONIC R series of polymers from BASF Corporation, Mount Olive, NJ) and Y is a biodegradable polyester, where the polyester may comprise the residues of one or more of the monomers selected from lactide, lactic acid, glycolide, glycolic acid, e-caprolactone, gamma-caprolactone, hydroxyvaleric acid, hydroxybutyric acid, beta-butyrolactone, gamma- butyrolactone, gamma-valerolactone, γ-decanolactone, δ-decanolactone, trimethylene carbonate, 1 ,4-dioxane-2-one or 1,5-dioxepan-2one (e.g., PLG- PEG-PLG) and R is a multifunctional initiator. In another embodiment, the tissue reactive polymer may be applied initially and then the fibrosis- inhibiting agent may then be applied to the coated tissue. The fibrosis- inhibiting agent may be applied directly to the tissue or it may be incorporated into a secondary carrier. The secondary carriers may include microspheres (as described above), microparticles (as described above), gels (e.g., hyaluronic acid, carboxymethyl cellulose, dextran, poly(ethylene oxide) - polyφropylene oxide) block copolymers as well as blends, association complexes and crosslinked compositions thereof) and films (degradable polyesters, where the polyester may comprise the residues of one or more of the monomers selected from lactide, lactic acid, glycolide, glycolic acid, e-caprolactone, gamma-caprolactone, hydroxyvaleric acid, hydroxybutyric acid, beta-butyrolactone, gamma-butyrolactone, gamma- valerolactone, γ-decanolactone, δ-decanolactone, trimethylene carbonate, 1 ,4-dioxane-2-one or 1,5-dioxepan-2one, and block copolymers of the form X-Y, Y-X-Y, R-(Y-X)n, R-(X-Y)n and X-Y-X where X in a polyalkylene oxide (e.g., poly(ethy!ene glycol, polypropylene glycol) and block copolymers of poly(ethylene oxide) and polypropylene oxide) (e.g., PLURONIC and PLURONIC R series of polymers from BASF Corporation, Mount Olive, NJ) and Y is a biodegradable polyester, where the polyester may comprise the residues of one or more of the monomers selected from lactide, lactic acid, glycolide, glycolic acid, e-caprolactone, gamma-caprolactone, hydroxyvaleric acid, hydroxybutyric acid, beta-butyrolactone, gamma- butyrolactone, gamma-valerolactone, γ-decanolactone, δ-decanolactone, trimethylene carbonate, 1 ,4-dioxane-2-one or 1 ,5-dioxepan-2one (e.g., PLG- PEG-PLG) and R is a multifunctional initiator, hyaluronic acid, carboxymethyl cellulose, dextran, poly(ethylene oxide) - polyφropylene oxide) block copolymers as well as blends, association complexes and crosslinked compositions thereof. . - . -
iv) Adhesion Prevention in Cardiac Procedures In one aspect, adhesions may be associated with a cardiac surgical procedure. In the case of cardiac surgery involving transplants, vascular repair, coronary artery bypass grafting (CABG), congenital heart defects, and valve replacements, staged procedures and reoperations (particularly repeat CABG surgery) are very common. As such, cardiac surgeons frequently must operate on tissues that have been surgically traumatized previously and have thick fibrous adhesions present which make dissection difficult. Post-operative pericardial adhesions (adhesions between the two surfaces of the pericardial sac) from initial surgery are common. Pericardial adhesions can cause symptoms by restricting the normal movement and filling of the heart during the cardiac cycle and can subject patients undergoing repeat cardiac surgery to elevated procedural risks. Restemotomy (re-opening the chest wall incision and surgical exposure of the heart) and dissection of the adhesions that accompany it, increases the risk of potential injury to the heart, great vessels and extracardiac grafts, increases operative time (including increasing the time the patient is on heart-lung bypass), and can increase procedural morbidity and mortality. Resternotomy is associated with as much as a 6% incidence of major vascular injury and a greater than 35% mortality has been reported for patients experiencing major hemorrhage during resternotomy. A 50% mortality has been reported for associated injuries to aortocoronary grafts. Staged pediatric open-heart surgery (repeat procedures required as the heart grows) is also associated with a very high incidence of complications due to reoperations.
As described previously, adhesion barriers are frequently used in the management of adhesions following open-heart procedures. A variety of commercially available adhesion barriers are suitable for combining with a fibrosis-inhibitor (and/or an anti-infective agent) in the management of cardiac surgery adhesions. Life Medical Sciences, Inc: is developing the products, REPEL, REPEL-CV, RESOLVE and RELIEVE that are in various stages of development and may be used to prevent surgical adhesions of open heart and other surgeries. Products being developed by Life Medical Sciences, Inc. are described in, for example, U.S. Patent Nos. 6,696,499; 6,399,624; 6,211 ,249; 6,136,333 and 5,711,958. Closure Medical Corp. (Raleigh, NC) is developing a cyanoacrylate-based internal adhesives that may be used to seal internal surgical incisions or grafts which may be compatible in pulmonary and general surgical specialties. Products that are being developed by Closure Medical, Corp. are described in, for example, U.S. Patent Nos. 6,620,846; 6,579,469; 6,565,840; 6,547,467 and 5,981 ,621. Genzyme Corporation has developed hyaluronic acid-containing biomaterials, such as SEPRAFILM and SEPRACOAT, to reduce the incidence of adhesions following cardiothoracic surgeries (see, e.g., U.S. Patent Nos. 6,780,427; 6,531 ,147; 6,521 ,223 and 6,010,692. Other commercially available materials that may be used alone, or loaded with a therapeutic agent (e.g., a fibrosis-inhibiting agent or an anti-infective agent), applied to or infiltrated into cardiac surgery site (or to the surface of an implanted device or implant) for the prevention of adhesions during open or endoscopic heart surgery include: (a) sprayable collagen-containing formulations such as COSTASIS or CT3; (b) sprayable PEG-containing formulations such as COSEAL, ADHIBIT, FOCALSEAL or DURASEAL; (c) fibrinogen-containing formulations such as FLOSEAL or TISSEAL; (d) hyaluronic acid-containing formulations such as RESTYLANE or PERLANE, HYLAFORM, or SYNVISC; (e) polymeric gels for surgical implantation such as REPEL or FLOWGEL; (f) surgical adhesives containing cyanoacrylates such as DERMABOND, INDERMIL, GLUSTITCH, TISSUMEND, VETBOND, HISTOACRYL BLUE and ORABASE SOOTHE-N-SEAL LIQUID PROTECTANT; (g) dextran sulfate gels such as the ADCON series of gels; and (h) lipid based compositions such as ADSURF. It should be obvious to one of skill in the art that commercial compositions not specifically cited above as well as next- generation and/or subsequently-developed commercial products are to be anticipated and are suitable for use under the present invention.
Virtually any manipulation of the chest wall, pericardium and heart can trigger a cascade that ultimately results in the formation of adhesions. In many instances, the adhesions must be broken down during repeat open-heart interventions. An adhesion barrier, either alone or containing a fibrosis-inhibiting agent (and/or an anti-infective agent), is best applied directly to the affected areas (as a solid, a film, a paste, a gel, a liquid or another such formulation) during open or endoscopic cardiac procedures. In a preferred embodiment, the barrier (alone or containing an anti-fibrotic and/or anti-infective agent) is sprayed under direct or endoscopic vision during the procedure onto the heart, pericardium, pleura and chest wall that are operated on, or manipulated, during the intervention. Since adhesions often occur in areas at a distance from the tissues actually instrumented during a surgical intervention, it is recommended that the barrier (with or without a therapeutic agent) be applied to a wide area in the chest (potentially even the entire cardiopulmonary viscera and infiltrated throughout the pericardial sac). Preferred barriers include films, liquids, gels, pastes, sprays or other formulations that can be delivered during open procedures or through an endoscope, adhere to the tissues treated, and remain in place long enough to deliver the therapeutic agent and/or prevent adhesion formation. As an alternative, the therapeutic agent can be delivered directly into the pericardial sac as an injectable (either before, during or after the procedure) such that the drug is delivered in doses high enough and long enough (multiple dosing and/or sustained release preparations are preferred) to prevent adhesions and the complications arising from them. An ideal adhesion therapy will reduce the incidence, number and tenacity of adhesions and improve patient outcome by reducing the complications of repeat interventions.
- As described above, the compositions for the prevention of surgical adhesions can be applied directly or indirectly to the tissue in a cardiac surgery procedure. The polymeric compositions (either with or without an anti-fibrotic or anti-infective therapeutic agent) can be administered in any manner described herein. Exemplary methods include either direct application at the time of surgery or with endoscopic, ultrasound, CT, MRI, or fluoroscopic guidance. If an implanted device is being placed, the composition for the prevention of adhesions can be applied to the surface of the implant, or to the surrounding tissues, in conjunction with placement of a medical device or implant at the surgical site. Representative examples of implants for use in cardiac procedures includes, without limitation, heart valves (porcine, artificial), ventricular assist devices, cardiac pumps, artificial hearts, stents, bypass grafts (artificial and endogenous), patches, cardiac electrical leads, defibrillators and pacemakers. The polymeric composition, with or without a fibrosis-inhibiting agent, may be applied during open or endoscopic heart surgery: (a) to the tissue surface of the pericardium (or infiltrated into the pericardial sac), heart, great vessels, pleura, lungs, chest wall and any adjacent affected tissues (e.g., as an injectable, solution, paste, gel, in situ forming gel or mesh) during the surgical procedure; (b) to the surface of an implanted device or implant and/or the tissue surrounding the implant (e.g., as an injectable, solution, paste, gel, in situ forming gel or mesh) before, during, or after the surgical procedure; (c) by intraperitoneal or endoscopic injection of the composition into the anatomical space (i.e., the pericardial sac) at the surgical site (particularly useful for this embodiment is the use of injectable compositions containing polymeric carriers which release the fibrosis- inhibiting agent over a period ranging from several hours to several weeks - fluids, suspensions, emulsions, microemulsions, microspheres, pastes, gels, microparticulates, sprays, aerosols, solid implants and other formulations which release the agent and can be delivered into the region where there is - a risk of adhesion formation); (d) via percutaneous injection into the tissue as a solution as an infusate or as a sustained release preparation (intrapericardial injection); (e) by guided catheter or endoscopic injection of the composition into the lumen or the walls of the atria, ventricles, great vessels, coronary arteries or the pericardial sac (particularly useful for this embodiment is the use of injectable compositions containing polymeric carriers which release the fibrosis-inhibiting agent over a period ranging from several hours to several weeks - fluids, suspensions, emulsions, microemulsions, microspheres, pastes, gels, microparticulates, sprays, aerosols, solid implants and other formulations which release the agent can be delivered into the areas of the heart where there is a risk of adhesion formation); and/or (f) by any combination of the aforementioned methods. Combination therapies (i.e., combinations of therapeutic agents and combinations with antithrombotic, anti-infective, and/or antiplatelet agents) can also be used in the manner described above. In certain applications involving the placement of a cardiac medical device or implant, it may be desirable to apply the anti-fibrosis (and/or anti-infective) composition at a site that is adjacent to an implant (preferably near the implant-tissue interface). This can be accomplished during open, endoscopic or catheter-based procedures by applying the polymeric composition, with or without a fibrosis-inhibiting agent: (a) to the implant surface (e.g., as an injectable, solution, paste, gel, in situ forming gel, or mesh) before, during, or after the implantation procedure; (b) to the surface of the adjacent tissue (e.g., as an injectable, solution, paste, gel, in situ forming gel, or mesh) immediately prior to, during, or after implantation of the implant; (c) to the surface of the implant and the tissue surrounding the implant (e.g., as an injectable, solution, paste, gel, in situ forming gel or mesh) before, during, or after implantation of the implant; (d) by topical application of the composition into the anatomical space (pericardial sac, intracardiac, intra-arterial) where the implant will be placed (particularly useful for this embodiment is the use of polymeric carriers which release the fibrosis-inhibiting agent over a period ranging from several hours to several weeks - fluids, suspensions, emulsions, microemulsions, microspheres, pastes, gels, microparticulates, sprays, aerosols, solid implants and other formulations which release the agent can be delivered into the region where the device will be inserted); (e) via percutaneous injection into the tissue surrounding the implant as a solution, as an infusate, or as a sustained release preparation; and/or (f) by any combination of the aforementioned methods. Combination therapies (Ae., combinations of therapeutic agents and combinations with antithrombotic, anti-infective, and/or antiplatelet agents) can also be used.
In one aspect, the polymeric composition may be delivered to the heart (or device/tissue interface) in the form of a spray or gel during open, endoscopic or catheter-based procedures. The fibrosis-inhibiting agent can be incorporated directly into the surgical adhesion barrier or it can be incorporated into a secondary carrier (polymeric or non-polymeric), as described above, that is then incorporated into the adhesion barrier. Examples of polymer compositions that may be in the form of a spray or gel include poly(ethylene glycol)-based systems, hyaluronic acid and crosslinked hyaluronic acid compositions. These compositions can be applied as the final composition or they can be applied as materials that form a crosslinked gel in situ.
In another aspect, an activated polymer is dissolved in a biologically acceptable buffer that has a pH lower that 6.8. The resultant solution is then applied to the desired tissue surface in the presence of a second biologically acceptable buffer that has a pH greater than 7.5. Application of the reaction mixture to the tissue site may be by extrusion, brushing, spraying or by any other convenient means. Following application of the composition to the surgical site, any excess solution may be removed from the surgical site if deemed necessary. At this point in time, the surgical site can be closed using conventional means (e.g., sutures, staples, or a bioadhesive). In one embodiment, the activated polymer can form a covalent bond with the tissue to which it is applied may be used. Polymers containing and/or terminated with electrophilic groups such as succinimidyl, aldehyde, epoxide, isocyanate, vinyl, vinyl sulfone, maleimide, -S-S-(C5H4N) or activated esters, such as are used in peptide synthesis may be used as the reagents. For example, a 4 armed NHS-derivatized polyethylene glycol (e.g., pentaerythritol poly(ethylene glycol)ether tetra-succinimidyl glutarate) may be applied to the tissue in the solid form or in a solution form. In this embodiment, the 4 armed NHS-derivatized polyethylene glycol is dissolved in an acidic solution (pH about 2-3) and is then co-applied to the tissue using a basic buffer (pH > about 8). The fibrosis-inhibiting agent(s) may be incorporated directly into either the 4 armed NHS-derivatized polyethylene glycol, the acidic solution or the basic buffer. In another embodiment, the fibrosis-inhibiting agent may be incorporated into a secondary carrier that may then be incorporated into the 4 armed NHS-derivatized polyethylene glycol, the acidic solution and/or the basic buffer. The secondary carriers may include microparticles and/or microspheres which are made from degradable polymers. The degradable polymers may include polyesters, where the polyester may comprise the residues of one or more of the monomers selected from lactide, lactic acid, glycolide, glycolic acid, e- caprolactone, gamma-caprolactone, hydroxy valeric acid, hydroxybutyric acid, beta-butyrolactone, gamma-butyrolactone, gamma-valerolactone, γ~ decanolactone, δ-decanolactone, trimethylene carbonate, 1 ,4-dioxane-2- one or 1 ,5-dioxepan-2one, and block copolymers of the form X-Y, Y-X-Y, R- (Y-X)n, R-(X-Y)n and X-Y-X where X in a polyalkylene oxide (e.g., poly(ethylene glycol, poly(propylene glycol) and block copolymers of poly(ethylene oxide) and poly(propylene oxide) (e.g., PLURONIC and PLURONIC R series of polymers from BASF Corporation, Mount Olive, NJ) and Y is a biodegradable polyester, where the polyester may comprise the residues of one or more of the monomers selected from lactide, lactic acid, glycolide, glycolic acid, e-caprolactone, gamma-caprolactone, hydroxyvaleric acid, hydroxybutyric acid, beta-butyrolactone, gamma- butyrolactone, gamma-valerolactone, γ-decanolactone, δ-decanolactone, trimethylene carbonate, 1 ,4-dioxane-2-one or 1 ,5-dioxepan-2one (e.g., PLG- PEG-PLG) and R is a multifunctional initiator. In another embodiment, the tissue reactive polymer may be applied initially and then the fibrosis- inhibiting agent may then be applied to the coated tissue. The fibrosis- inhibiting agent may be applied directly to the tissue or it may be incorporated into a secondary carrier. The secondary carriers may include microspheres (as described above), microparticles (as described above), gels (e.g., hyaluronic acid, carboxymethyl cellulose, dextran, poly(ethylene oxide) - polyφropylene oxide) block copolymers as well as blends, association complexes and crosslinked compositions thereof) and films (degradable polyesters, where the polyester may comprise the residues of one or more of the monomers selected from lactide, lactic acid, glycolide, glycolic acid, e-caprolactone, gamma-caprolactone, hydroxyvaleric acid, hydroxybutyric acid, beta-butyrolactone, gamma-butyrolactone, gamma- valerolactone, γ-decanolactone, δ-decanolactone, trimethylene carbonate, 1 ,4-dioxane-2-one or 1,5-dioxepan-2one, and block copolymers of the form X-Y, Y-X-Y, R-(Y-X)n, R-(X-Y)n and X-Y-X where X in a polyalkylene oxide (e.g., poly(ethylene glycol, poly(propylene glycol) and block copolymers of poly(ethylene oxide) and poly(propylene oxide) (e.g., PLURONIC and PLURONIC R series of polymers from BASF Corporation, Mount Olive, NJ) and Y is a biodegradable polyester, where the polyester may comprise the residues of one or more of the monomers selected from lactide, lactic acid, glycolide, glycolic acid, e-caprolactone, gamma-caprolactone, hydroxyvaleric acid, hydroxybutyric acid, beta-butyrolactone, gamma- butyrolactone, gamma-valerolactone, γ-decanolactone, δ-decanolactone, trimethylene carbonate, 1 ,4-dioxane-2-one or 1 ,5-dioxepan-2one (e.g., PLG- PEG-PLG) and R is a multifunctional initiator, hyaluronic acid, carboxymethyl cellulose, dextran, poly(ethylene oxide) - poly(propylene oxide) block copolymers as well as blends, association complexes and crosslinked compositions thereof.
In yet another aspect, an activated polymer can be applied to the surgical site in the solid state. The activated polymer can react with the tissue surface to which it was applied as the polymer hydrates. A biologically acceptable buffer, with a pH greater than 7.5 can be applied to the tissue before and/or after the solid activated polymer has been applied. In one embodiment, the activated polymer can form a covalent bond with the tissue to which it is applied may be used. Polymers containing and/or terminated with electrophilic groups such as succinimidyl, aldehyde, epoxide, isocyanate, vinyl, vinyl sulfone, maleimide, -S-S-(C5H4N) or activated esters, such as are used in peptide synthesis may be used as the reagents. For example, a 4 armed NHS-derivatized polyethylene glycol (e.g., pentaerythritol poly(ethylene glycol)ether tetra-succinimidyl glutarate) may be applied to the tissue in the solid form. The fibrosis-inhibiting agent(s) may be incorporated directly into either the 4 armed NHS-derivatized polyethylene glycol, or the basic buffer. In another embodiment, the fibrosis- inhibiting agent may be incorporated into a secondary carrier that may then be incorporated into the 4 armed NHS-derivatized polyethylene glycol, and/or the basic buffer. The secondary carriers may include microparticles and/or microspheres which are made from degradable polymers. The degradable polymers may include polyesters, where the polyester may comprise the residues of one or more of the monomers selected from lactide, lactic acid, glycolide, glycolic acid, e-caprolactone, gamma- caprolactone, hydroxyvaleric acid, hydroxybutyric acid, beta-butyrolactone, gamma-butyrolactone, gamma-valerolactone, γ-decanolactone, δ- decanolactone, trimethylene carbonate, 1 ,4-dioxane-2-one or 1,5-dioxepan- 2one, and block copolymers of the form X-Y, Y-X-Y, R-(Y-X)n, R-(X-Y)n and X-Y-X where X in a polyalkylene oxide (e.g., poly(ethylene glycol, poly(propylene glycol) and block copolymers of poly(ethylene oxide) and polypropylene oxide) (e.g., PLURONIC and PLURONIC R series of polymers from BASF Corporation, Mount Olive, NJ) and Y is a biodegradable polyester, where the polyester may comprise the residues of one or more of the monomers selected from lactide, lactic acid, glycolide, glycolic acid, e-caprolactone, gamma-caprolactone, hydroxyvaleric acid, hydroxybutyric acid, beta-butyrolactone, gamma-butyrolactone, gamma- valerolactone, γ-decanolactone, δ-decanolactone, trimethylene carbonate, 1 ,4-dioxane-2-one or 1,5-dioxepan-2one (e.g., PLG-PEG-PLG) and R is a multifunctional initiator. In another embodiment, the tissue reactive polymer may be applied initially and then the fibrosis-inhibiting agent may then be applied to the coated tissue. The fibrosis-inhibiting agent may be applied directly to the tissue or it may be incorporated into a secondary carrier. The secondary carriers may include microspheres (as described above), microparticles (as described above), gels (e.g., hyaluronic acid, carboxymethyl cellulose, dextran, poly(ethylene oxide) - polyφropylene oxide) block copolymers as well as blends, association complexes and crosslinked compositions thereof) and films (degradable polyesters, where the polyester may comprise the residues of one or more of the monomers selected from lactide, lactic acid, glycolide, glycolic acid, e-caprolactone, gamma-caprolactone, hydroxy valeric acid, hydroxybutyric acid, beta- butyrolactone, gamma-butyrolactone, gamma-valerolactone, y- decanolactone, δ-decanolactone, trimethylene carbonate, 1 ,4-dioxane-2- one or 1 ,5-dioxepan-2one, and block copolymers of the form X-Y, Y-X-Y, R- (Y-X)n, R-(X-Y)n and X-Y-X where X in a polyalkylene oxide (e.g., poly(ethylene glycol, poly(propylene glycol) and block copolymers of poly(ethylene oxide) and polyφropylene oxide) (e.g., PLURONIC and PLURONIC R series of polymers from BASF Corporation, Mount Olive, NJ) and Y is a biodegradable polyester, where the polyester may comprise the residues of one or more of the monomers selected from lactide, lactic acid, glycolide, glycolic acid, e-caprolactone, gamma-caprolactone, hydroxy valeric acid, hydroxybutyric acid, beta-butyrolactone, gamma- butyrolactone, gamma-valerolactone, γ-decanolactone, δ-decanolactone, trimethylene carbonate, 1 ,4-dioxane-2-one or 1 ,5-dioxepan-2one (e.g., PLG- PEG-PLG) and R is a multifunctional initiator, hyaluronic acid, carboxymethyl cellulose, dextran, poly(ethylene oxide) - poly(propylene oxide) block copolymers as well as blends, association complexes and crosslinked compositions thereof.
v) Adhesion Prevention in Orthopedic Procedures In one aspect, adhesions may be associated with an orthopedic surgical procedure. Many orthopedic surgical interventions are performed as a result of injury or trauma (fractures; torn ligaments, cartilage, tendons or muscles) that cause significant tissue damage that can lead to excessive scarring and adhesion formation. As a result, orthopedic procedures often result in potentially severe post-operative complications which may be attributed to the trauma which caused the injury or to the trauma from the surgery itself. In general, excessive scarring and adhesion formation in orthopedic conditions follows certain patterns: (a) in joint injuries, it can result in a deformity such that the joint cannot fully extend, flex, or rotate (contractures); (b) in tendon injuries, it can prevent normal movement and lead to shortening; (c) in cartilage injuries, it can lead to the conversion of hyaline cartilage to fibrocartilage with a resultant loss of function and joint instability; (d) in muscle injuries, it can cause adhesion to adjacent tissues, loss of strength and loss of function; (e) in nerve injuries, it can result in loss of conduction and function; if the nerve becomes entrapped (encircled and constricted) by scar, it can cause pain, sensory impairment and loss of motor function; and (f) in tendons and ligaments, it can cause shortening, loss of range of motion and impaired function. The complications of adhesions can be wide spread; for example, adhesions formed after spinal surgery may produce low back pain, leg pain and sphincter disturbance (bladder and bowel). For this reason strategies designed to reduce adhesion formation in musculoskeletal surgery is a significant clinical problem. The local administration of anti-adhesive compositions, alone or loaded with a fibrosis-inhibiting agent, can be utilized in a wide array of clinical situations and conditions to improve patient outcomes following emergency or elective orthopedic interventions.
As described previously, adhesion barriers are frequently used in the management of adhesions following orthopedic procedures. A variety of commercially available adhesion barriers are suitable for combining with a fibrosis-inhibitor (and/or an anti-infective agent) in the management of orthopedic surgery adhesions. Closure Medical Corp. (Raleigh, NC) is developing a cyanoacrylate-based internal adhesives that may be used to seal internal surgical incisions or grafts which may be compatible in orthopedic and general surgical specialties. Products that are being developed by Closure Medical, Corp. are described in, for example, U.S. Patent Nos. 6,620,846; 6,579,469; 6,565,840; 6,547,467 and 5,981 ,621. Life Medical Sciences, Inc. is developing the products, REPEL, REPEL-CV, RESOLVE and RELIEVE that are in various stages of development and may be used to prevent surgical adhesions in orthopedic and spinal surgeries. Products being developed by Life Medical Sciences, Inc. are described in, for example, U.S. Patent Nos. 6,696,499; 6,399,624; 6,211 ,249; 6,136,333 and 5,711 ,958.
Other commercially available materials that may be used alone, or loaded with a therapeutic agent (e.g., a fibrosis-inhibiting agent or an anti-infective agent), applied to or infiltrated into an orthopedic site (or to the surface of an implanted device or implant) for the prevention of adhesions in open or endoscopic orthopedic surgery include: (a) sprayable collagen-containing formulations such as COSTASIS or CT3; (b) sprayable PEG-containing formulations such as COSEAL, ADHIBIT, FOCALSEAL, SPRAYGEL or DURASEAL; (c) fibrinogen-containing formulations such as FLOSEAL or TISSEAL; (d) hyaluronic acid-containing formulations such as RESTYLANE1 HYLAFORM1 PERLANE, SYNVISC, SEPRAFILM, SEPRACOAT, INTERGEL, or LUBRICOAT; (e) polymeric gels for surgical implantation such as REPEL or FLOWGEL; (f) orthopedic "cements" used to hold prostheses and tissues in place, such as OSTEOBOND (Zimmer), LVC (Wright Medical Technology), SIMPLEX P (Stryker), PALACOS (Smith & Nephew), and ENDURANCE (Johnson & Johnson, Inc.); (g) surgical adhesives containing cyanoacrylates such as DERMABOND, INDERMIL, GLUSTITCH, TISSUMEND, VETBOND, HISTOACRYL BLUE and ORABASE SOOTHE-N-SEAL LIQUID PROTECTANT; (g) implants containing hydroxyapatite (or synthetic bone material such as calcium sulfate, VITOSS (Orthovita) and CORTOSS (Orthovita)); (h) other biocompatible tissue fillers, such as those made by BioCure, 3M Company and Neomend; (i) polysacharride gels such as the ADCON series of gels; (j) films, sponges or meshes such as INTERCEED, VICRYL mesh, and GELFOAM; (o) lipid based compositions such as ADSURF; and (p) OSSIGEL, a viscous formulation of hyaluronic acid (HA) and basic fibroblast growth factor (bFGF) designed to accelerate bone fracture healing (Orquest, Inc.). It should be obvious to one of skill in the art that commercial compositions not specifically cited above as well as next-generation and/or subsequently-developed commercial products are to be anticipated and are suitable for use under the present invention.
Orthopedic surgical procedures are performed for a variety of conditions including fractures (open and closed), sprains, joint dislocations, crush injuries, ligament and muscle tears, tendon injuries, nerve injuries, congenital deformities and malformations, total joint or partial joint replacement, and cartilage injuries. Although many procedures are still performed through open surgical techniques, increasingly, numerous orthopedic procedures are being performed via an arthroscope inserted into the joint. Virtually any musculoskeletal (muscle, tendon, joint, bone, cartilage) injury, traumatic injury, or orthopedic surgical intervention can trigger a cascade that ultimately results in the formation of adhesions. In many instances, the adhesions must be broken down during repeat surgical interventions (e.g., capsulotomies, tendon releases, nerve entrapment releases, frozen joints, etc.). An adhesion barrier, either alone or containing a fibrosis-inhibiting agent (and/or an anti-infective agent), is best applied directly to the affected areas (as a solid, a film, a paste, a gel, a liquid or another such formulation) during open or arthroscopic orthopedic procedures. In a preferred embodiment, the barrier (alone or containing an anti-fibrotic and/or anti-infective agent) is sprayed under direct or arthrocopic vision onto the affected musculoskeletal tissue during the intervention. Since adhesions often occur in areas at a distance from the tissues actually instrumented during a surgical intervention, it is recommended that the barrier (with or without a therapeutic agent) be applied to a wide area around the injured or repaired tissues. Preferred barriers include films, liquids, gels, pastes, sprays or other formulations that can be delivered during open procedures or through an endoscope, adhere to the tissues treated, and remain in place long enough to deliver the therapeutic agent and/or prevent adhesion formation. An ideal adhesion therapy will reduce the incidence, number and tenacity of adhesions and improve patient outcome by reducing pain, weakness and sensory abnormalities, preventing contractures, increasing range of motion, improving function, limiting physical deformity and disability, and reducing the need for repeat interventions.
As described above, the compositions for the prevention of surgical adhesions can be applied directly or indirectly to the tissue in an orthopedic surgery procedure. The polymeric compositions (either with or without an anti-fibrotic or anti-infective therapeutic agent) can be administered in any manner described herein. Exemplary methods include either direct application at the time of surgery or with arthroscopic, ultrasound, CT, MRI, or fluoroscopic guidance. If an implanted device is being placed, the composition for the prevention of adhesions can be applied to the surface of the implant, or to the surrounding tissues, in conjunction with placement of a medical device or implant at the surgical site. Representative examples of implants for use in orthopedic procedures include plates, rods, screws, pins, wires, total and partial joint prostheses (artificial hips, knees, shoulders, phalangeal joints), reinforcement patches, tissue fillers, synthetic bone fillers, bone cement, synthetic graft material, allograft material, autograft material, artificial discs, spinal cages, and intermedulary rods.
The polymeric composition, with or without a fibrosis-inhibiting agent, may be applied during open or arthroscopic orthopedic surgery: (a) to the tissue surface of the bone, joint, muscle, tendon, ligament, cartilage and any adjacent affected tissues (e.g., as an injectable, solution, paste, gel, in situ forming gel or mesh) during the surgical procedure; (b) to the surface of an implanted orthopedic device or implant and/or the tissue surrounding the implant (e.g., as an injectable, solution, paste, gel, in situ forming gel or mesh) before, during, or after the surgical procedure; (c) by intra-articular or endoscopic administration of the composition into the anatomical space (e.g., the joint space, tendon sheath, nerve root, spinal canal) at the surgical site (particularly useful for this embodiment is the use of injectable compositions containing polymeric carriers which release the fibrosis- inhibiting agent over a period ranging from several hours to several weeks - fluids, suspensions, emulsions, microemulsions, microspheres, pastes, gels, microparticulates, sprays, aerosols, solid implants and other formulations which release the agent and can be delivered into the region where there is a risk of adhesion formation); (d) via percutaneous injection into the tissue as a solution as an infusate or as a sustained release preparation (intramuscular or intra-articular injection); (e) by guided catheter injection of the composition into the tissues and/or (f) by any combination of the aforementioned methods. Combination therapies (i.e., combinations of therapeutic agents and combinations with antithrombotic, anti-infective, and/or antiplatelet agents) can also be used in the manner described above.
In certain applications involving the placement of an orthopedic medical device or implant, it may be desirable to apply the anti- fibrosis (or anti-infective) composition at a site that is adjacent to an implant (preferably near the implant-tissue interface). This can be accomplished during open, endoscopic or catheter-based orthopedic procedures by applying the polymeric composition, with or without a fibrosis-inhibiting agent: (a) to the implant surface (e.g., as an injectable, solution, paste, gel, in situ forming gel, or mesh) before, during, or after the implantation procedure; (b) to the surface of the adjacent tissue (e.g., as an injectable, solution, paste, gel, in situ forming gel, or mesh) immediately prior to, during, or after implantation of the orthopedic implant; (c) to the surface of the implant and the tissue surrounding the implant (e.g., as an injectable, solution, paste, gel, in situ forming gel or mesh) before, during, or after implantation of the implant; (d) by topical application of the composition into the anatomical space (joint capsule, spinal canal, marrow, tendon sheath etc.) where the implant will be placed (particularly useful for this embodiment is the use of polymeric carriers which release the fibrosis- inhibiting agent over a period ranging from several hours to several weeks - fluids, suspensions, emulsions, microemulsions, microspheres, pastes, gels, microparticulates, sprays, aerosols, solid implants and other formulations which release the agent can be delivered into the region where the device will be inserted); (e) via percutaneous injection into the tissue surrounding the orthopedic implant as a solution, as an infusate, or as a sustained release preparation; and/or (f) by any combination of the aforementioned methods. Combination therapies (i.e., combinations of therapeutic agents and combinations with antithrombotic, anti-infective, and/or antiplatelet agents) can also be used.
In one aspect, the polymeric composition may be delivered to the musculoskeletal tissue (or device/tissue interface) in the form of a spray or gel during open, endoscopic or catheter-based procedures. The fibrosis- inhibiting (and/or anti-infective) agent can be incorporated directly into the surgical adhesion barrier or it can be incorporated into a secondary carrier (polymeric or non-polymeric), as described above, that is then incorporated into the adhesion barrier. Examples of polymer compositions that may be in the form of a spray or gel include poly(ethylene glycol)-based systems, hyaluronic acid and crosslinked hyaluronic acid compositions. These compositions can be applied as the final composition or they can be applied as materials that form a crosslinked gel in situ.
In another aspect, an activated polymer is dissolved in a biologically acceptable buffer that has a pH lower that 6.8. The resultant solution is then applied to the desired tissue surface in the presence of a second biologically acceptable buffer that has a pH greater than 7.5. Application of the reaction mixture to the tissue site may be by extrusion, brushing, spraying or by any other convenient means. Following application of the composition to the surgical site, any excess solution may be removed from the surgical site if deemed necessary. At this point in time, the surgical site can be closed using conventional means (e.g., sutures, staples, or a bioadhesive). In one embodiment, the activated polymer can form a covalent bond with the tissue to which it is applied may be used. Polymers containing and/or terminated with electrophilic groups such as succinimidyl, aldehyde, epoxide, isocyanate, vinyl, vinyl sulfone, maleimide, -S-S-(CsH4N) or activated esters, such as are used in peptide synthesis may be used as the reagents. For example, a 4 armed NHS-derivatized polyethylene glycol (e.g., pentaerythritol poly(ethylene glycol)ether tetra-succinimidyl glutarate) may be applied to the tissue in the solid form or in a solution form. In this embodiment, the 4 armed NHS-derivatized polyethylene glycol is dissolved in an acidic solution (pH about 2-3) and is then co-applied to the tissue using a basic buffer (pH > about 8). The fibrosis-inhibiting agent(s) may be incorporated directly into either the 4 armed NHS-derivatized polyethylene glycol, the acidic solution or the basic buffer. In another embodiment, the fibrosis-inhibiting agent may be incorporated into a secondary carrier that may then be incorporated into the 4 armed NHS-derivatized polyethylene glycol, the acidic solution and/or the basic buffer. The secondary carriers may include microparticles and/or microspheres which are made from degradable polymers. The degradable polymers may include polyesters, where the polyester may comprise the residues of one or more of the monomers selected from lactide, lactic acid, glyeolide, glycolic acid, e- caprolactone, gamma-caprolactone, hydroxy valeric acid, hydroxybutyric acid, beta-butyrolactone, gamma-butyrolactone, gamma-valerolactone, y- decanolactone, δ-decanolactone, trimethylene carbonate, 1 ,4-dioxane-2- one or 1 ,5-dioxepan-2one, and block copolymers of the form X-Y, Y-X-Y, R- (Y-X)n, R-(X-Y)n and X-Y-X where X in a polyalkylene oxide (e.g., poly(ethylene glycol, polyφropylene glycol) and block copolymers of poly(ethylene oxide) and poly(propylene oxide) (e.g., PLURONIC and PLURONIC R series of polymers from BASF Corporation, Mount Olive, NJ) and Y is a biodegradable polyester, where the polyester may comprise the residues of one or more of the monomers selected from lactide, lactic acid, glycolide, glycolic acid, e-caprolactone, gamma-caprolactone, hydroxyvaleric acid, hydroxybutyric acid, beta-butyrolactone, gamma- butyrolactone, gamma-valerolactone, γ-decanolactone, δ-decanolactone, trimethylene carbonate, 1 ,4-dioxane-2-one or 1 ,5-dioxepan-2one (e.g., PLG- PEG-PLG) and R is a multifunctional initiator. In another embodiment, the tissue reactive polymer may be applied initially and then the fibrosis- inhibiting agent may then be applied to the coated tissue. The fibrosis- inhibiting agent may be applied directly to the tissue or it may be incorporated into a secondary carrier. The secondary carriers may include microspheres (as described above), microparticles (as described above), gels (e.g., hyaluronic acid, carboxymethyl cellulose, dextran, poly(ethylene oxide) - poly(propylene oxide) block copolymers as well as blends, association complexes and crosslinked compositions thereof) and films (degradable polyesters, where the polyester may comprise the residues of one or more of the monomers selected from lactide, lactic acid, glycolide, glycolic acid, e-caprolactone, gamma-caprolactone, hydroxyvaleric acid, hydroxybutyric acid, beta-butyrolactone, gamma-butyrolactone, gamma- valerolactone, γ-decanolactone, δ-decanolactone, trimethylene carbonate, 1 ,4-dioxane-2-one or 1 ,5-dioxepan-2one, and block copolymers of the form X-Y, Y-X-Y, R-(Y-X)n, R-(X-Y)n and X-Y-X where X in a polyalkylene oxide (e.g., poly(ethylene glycol, poly(propylene glycol) and block copolymers of poly(ethylene oxide) and poly(propylene oxide) (e.g., PLURONIC and PLURONIC R series of polymers from BASF Corporation, Mount Olive, NJ) and Y is a biodegradable polyester, where the polyester may comprise the residues of one or more of the monomers selected from lactide, lactic acid, glycolide, glycolic acid, e-caprolactone, gamma-caprolactone, hydroxyvaleric acid, hydroxybutyric acid, beta-butyrolactone, gamma- butyrolactone, gamma-valerolactone, γ-decanolactone, δ-decanolactone, trimethylene carbonate, 1 ,4-dioxane-2-one or 1,5-dioxepan-2one (e.g., PLG- PEG-PLG) and R is a multifunctional initiator, hyaluronic acid, carboxymethyl cellulose, dextran, poly(ethylene oxide) - poly(propylene oxide) block copolymers as well as blends, association complexes and crosslinked compositions thereof.
In yet another aspect, an activated polymer can be applied to the surgical site in the solid state. The activated polymer can react with the tissue surface to which it was applied as the polymer hydrates. A biologically acceptable buffer, with a pH greater than 7.5 can be applied to the tissue before and/or after the solid activated polymer has been applied. In one embodiment, the activated polymer can form a covalent bond with the tissue to which it is applied may be used. Polymers containing and/or terminated with electrophilic groups such as succinimidyl, aldehyde, epoxide, isocyanate, vinyl, vinyl sulfone, maleimide, -S-S-(C5H4N) or activated esters, such as are used in peptide synthesis may be used as the reagents. For example, a 4 armed NHS-derivatized polyethylene glycol (e.g., pentaerythritol poly(ethylene glycol)ether tetra-succinimidyl glutarate) may be applied to the tissue in the solid form. The antifibrosisfibrosis- inhibiting agent(s) may be incorporated directly into either the 4 armed NHS- derivatized polyethylene glycol, or the basic buffer. In another embodiment, the fibrosis-inhibiting agent may be incorporated into a secondary carrier that may then be incorporated into the 4 armed NHS-derivatized polyethylene glycol, and/or the basic buffer. The secondary carriers may include microparticles and/or microspheres which are made from degradable polymers. The degradable polymers may include polyesters, where the polyester may comprise the residues of one or more of the monomers selected from lactide, lactic acid, glycolide, glycolic acid, e- caprolactone, gamma-caprolactone, hydroxyvaleric acid, hydroxybutyric acid, beta-butyrolactone, gamma-butyrolactone, gamma-valerolactone, γ~ decanolactone, δ-decanolactone, trimethylene carbonate, 1 ,4-dioxane-2- one or 1 ,5-dioxepan-2one, and block copolymers of the form X-Y, Y-X-Y, R- (Y-X)n, R-(X-Y)n and X-Y-X where X in a polyalkylene oxide (e.g., poly(ethylene glycol, poly(propylene glycol) and block copolymers of poly(ethylene oxide) and polypropylene oxide) (e.g., PLURONIC and PLURONIC R series of polymers from BASF Corporation, Mount Olive, NJ) and Y is a biodegradable polyester, where the polyester may comprise the residues of one or more of the monomers selected from lactide, lactic acid, glycolide, glycolic acid, e-caprolactone, gamma-caprolactone, hydroxyvaleric acid, hydroxybutyric acid, beta-butyrolactone, gamma- butyrolactone, gamma-valerolactone, γ-decanolactone, δ-decanolactone, trimethylene carbonate, 1 ,4-dioxane-2-one or 1,5-dioxepan-2one (e.g., PLG- PEG-PLG) and R is a multifunctional initiator. In another embodiment, the tissue reactive polymer may be applied initially and then the fibrosis- inhibiting agent may then be applied to the coated tissue. The fibrosis- inhibiting agent may be applied directly to the tissue or it may be incorporated into a secondary carrier. The secondary carriers may include microspheres (as described above), microparticles (as described above), gels (e.g., hyaluronic acid, carboxymethyl cellulose, dextran, poly(ethylene oxide) - poly(propylene oxide) block copolymers as well as blends, association complexes and crosslinked compositions thereof) and films (degradable polyesters, where the polyester may comprise the residues of one or more of the monomers selected from lactide, lactic acid, glycolide, glycolic acid, e-caprolactone, gamma-caprolactone, hydroxyvaleric acid, hydroxybutyric acid, beta-butyrolactone, gamma-butyrolactone, gamma- valerolactone, γ-decanolactone, δ-decanolactone, trimethylene. carbonate, 1 ,4-dioxane-2-one or 1 ,5-dioxepan-2one, and block copolymers of the form X-Y, Y-X-Y, R-(Y-X)n, R-(X-Y)n and X-Y-X where X in a polyalkylene oxide (e.g., poly(ethylene glycol, poly(propylene glycol) and block copolymers of poly(ethylene oxide) and poly(propylene oxide) (e.g., PLURONIC and PLURONIC R series of polymers from BASF Corporation, Mount Olive, NJ) and Y is a biodegradable polyester, where the polyester may comprise the residues of one or more of the monomers selected from lactide, lactic acid, glycolide, glycolic acid, e-caprolactone, gamma-caprolactone, hydroxyvaleric acid, hydroxybutyric acid, beta-butyrolactone, gamma- butyrolactone, gamma-valerolactone, γ-decanolactone, δ-decanolactone, trimethylene carbonate, 1,4-dioxane-2-one or 1 ,5-dioxepan-2one (e.g., PLG- PEG-PLG) and R is a multifunctional initiator, hyaluronic acid, carboxymethyl cellulose, dextran, poly(ethylene oxide) - poly(propylene oxide) block copolymers as well as blends, association complexes and crosslinked compositions thereof. vi) Adhesion Prevention in Reconstructive and Cosmetic Procedures
In one aspect, adhesions may be associated with a cosmetic or reconstructive surgical procedure. The use of soft tissue implants for cosmetic applications (aesthetic and reconstructive) is common in breast augmentation, breast reconstruction after cancer surgery, craniofacial procedures, reconstruction after trauma, congenital craniofacial reconstruction and oculoplastic surgical procedures to name a few.
The clinical function of a soft tissue implant depends upon the implant being able to effectively maintain its shape over time. In many instances, when these devices are implanted in the body, they are subject to a "foreign body" response from the surrounding host tissues. The body recognizes the implanted device as foreign, which triggers an inflammatory response followed by encapsulation of the implant with fibrous connective tissue (adhesion formation). Encapsulation of surgical implants complicates a variety of reconstructive and cosmetic surgeries, but is particularly problematic in the case of breast reconstruction surgery where the breast implant becomes surrounded by a fibrous capsule that alters anatomy and function. Scar capsules that harden and contract (known as "capsular contractures") are the most common complication of breast implant or reconstructive surgery. Capsular (fibrous) contractures can result in hardening of the breast, loss of the normal anatomy and contour of the breast, discomfort, weakening and rupture of the implant shell, asymmetry, infection, and patient dissatisfaction. Further, fibrous encapsulation of any soft tissue implant can occur even after a successful implantation if the device is manipulated or irritated by the daily activities of the patient. Bleeding in and around the implant can also trigger a biological cascade that ultimately leads to excess scar tissue formation. Furthermore, certain types of implantable prostheses (such as breast implants) include gel fillers (e.g., silicone) that tend to leak through the membrane envelope of the implant and can potentially cause a chronic inflammatory response in the surrounding tissue (which encourages tissue encapsulation and contracture formation). The effects of unwanted scarring in the vicinity of the implant are the leading cause of additional surgeries to correct defects, break down scar tissue (capsulotomy or capsulaectomy), to replace the implant, or remove the implant. The local administration of anti-adhesive compositions, alone or loaded with a fibrosis-inhibiting agent, can be utilized in a wide array of cosmetic and reconstructive procedures to improve patient outcomes.
Soft tissue implants are used in a variety of cosmetic, plastic, and reconstructive surgical procedures and may be delivered to many different parts of the body, including, without limitation, the face, nose, breast, chin, buttocks, chest, lip and cheek. Soft tissue implants are used for the reconstruction of surgically or traumatically created tissue voids, augmentation of tissues or organs, contouring of tissues, the restoration of bulk to aging tissues, and to correct soft tissue folds or wrinkles (rhytides). Of all soft tissue implantation procedures, breast implant placement for augmentation or breast reconstruction after mastectomy is the most frequently performed cosmetic surgery implant procedure. For example, in 2002 alone, over 300,000 women had breast implant surgery. Of these, approximately 80,000 were breast reconstructions following a mastectomy due to cancer.
The process for failure of all soft tissue implants is similar regardless of anatomical placement. However, since breast implants have been the most widely studied soft tissue implant, they will be used to illustrate the present invention. In general, breast augmentation or reconstructive surgery involves the placement of a commercially available breast implant, consisting of a capsule filled with either saline or silicone, into the tissues underneath the mammary gland. Four different incision sites have historically been used for breast implantation: axillary (armpit), periareolar (around the underside of the nipple), inframamary (at the base of the breast where it meets the chest wall) and transumbilical (around the belly button). The tissue is dissected away through the small incision, often with the aid of an endoscope (particularly for axillary and transumbilical procedures where tunneling from the incision site to the breast is required). A pocket for placement of the breast implant is created in either the subglandular or the subpectorial region. For subglandular implants, the tissue is dissected to create a space between the glandular tissue and the pectoralis major muscle that extends down to the inframammary crease. For subpectoral implants, the fibers of the pectoralis major muscle are carefully dissected to create a space beneath the pectoralis major muscle and superficial to the rib cage. Careful hemostasis is essential (since it can contribute to complications such as capsular contractures), so much so that minimally invasive procedures (axillary, transumbilical approaches) must be converted to more open procedures (such as periareolar) if bleeding control is inadequate. Depending upon the type of surgical approach selected, the breast implant is often deflated and rolled up for placement in the patient. After accurate positioning is_achieved, the implant can then be filled or expanded to the desired size.
Although many patients are satisfied with the initial procedure, significant percentages suffer from complications that frequently require a repeat intervention to correct. Encapsulation of a breast prosthesis that creates a periprosthetic shell (called capsular contracture) is the most common complication reported after breast enlargement, with up to 50% of patients reporting some dissatisfaction. Calcification can occur within the fibrous capsule adding to its firmness and complicating the interpretation of mammograms. Multiple causes of capsular contracture have identified including: foreign body reaction, migration of silicone gel molecules across the capsule and into the tissue, autoimmune disorders, genetic predisposition, infection, hematoma, and the surface characteristics of the prosthesis. Although no specific etiology has been repeatedly identified, at the cellular level, abnormal fibroblast activity stimulated by a foreign body is a consistent finding. Periprosthetic capsular tissues contain macrophages and occasional T- and B-lymphocytes, suggesting an inflammatory component to the process. Implant surfaces have been made both smooth and textured in an attempt to reduce encapsulation, however, neither has been proven to produce consistently superior results. Animal models suggest that there is an increased tendency for increased capsular thickness and contracture with textured surfaces that encourage fibrous tissue ingrowth on the surface. Placement of the implant in the subpectoral location appears to decrease the rate of encapsulation in both smooth and textured implants.
From a patient's perspective, the biological processes described above lead to a series of commonly described complaints. Implant malposition, hardness and unfavorable shape are the most frequently sited complications and are most often attributed to capsular contracture. When the surrounding scar capsule begins to harden and contract, it results in discomfort, weakening of the shell, asymmetry, skin dimpling and malpositioning. True capsular contractures will occur in approximately 10% of patients after augmentation, and in 25% to 30% of reconstruction cases, with most patients reporting dissatisfaction with the asthetic outcome. Scarring leading to asymmetries occurs in 10% of augmentations and 30% of reconstructions and is the leading cause of revision surgery. Skin wrinkling (due to the contracture pulling the skin in towards the implant) is a complication reported by 10% to 20% of patients. Scarring has even been implicated in implant deflation (1-6% of patients; saline leaking out of the implant and "deflating" it), when fibrous tissue ingrowth into the diaphragmatic valve (the access site used to inflate the implant) causes it to become incontinent and leak. In addition, over 15% of patients undergoing augmentation will suffer from chronic pain and many of these cases are ultimately attributable to scar tissue formation. Other complications of breast augmentation surgery include late leaks, hematoma (approximately 1-6% of patients), seroma (2.5%), hypertrophic scarring (2- 5%) and infections (about 1-4% of cases). Correction can involve several options including removal of the implant, capsulotomy (cutting or surgically releasing the capsule), capsulectomy (surgical removal of the fibrous capsule), or placing the implant in a different location (i.e., from subglandular to subpectoral). Ultimately, additional surgery (revisions, capsulotomy, removal, reimplantation) is required in over 20% of augmentation patients and in over 40% of reconstruction patients, with scar formation and capsular contracture being far and away the most common cause. Procedures to break down the scar may not be sufficient, and approximately 8% of augmentations and 25% of reconstructions ultimately have the implant surgically removed.
A fibrosis-inhibiting agent or composition delivered locally from the soft tissue implant or administered locally into the tissue surrounding the soft tissue implant can minimize fibrous tissue formation, encapsulation and capsular contracture. Application of a fibrosis-inhibiting composition onto the surface of a soft tissue implant or incorporated into a soft tissue implant (e.g., the agent is incorporated into the saline, gel or silicone within the implant and passively diffuses across the capsule into the surrounding tissue) may minimize or prevent fibrous contracture. Infiltration of a fibrosis- inhibiting agent or composition into the tissue surrounding the soft tissue implant, or into the surgical pocket where the implant will be placed, is another strategy for preventing the formation of scar and capsular contracture in augmentation and reconstructive surgery.
As described previously, adhesions and fibrous encapsulation of cosmetic implants is a common complication of asthetic and reconstructive surgery. A variety of commercially available adhesion barriers are suitable for combining with a fibrosis-inhibitor (and/or an anti- infective agent) in the management of this complication. Commercially available materials that may be used alone or loaded with a therapeutic agent (e.g., a fibrosis-inhibiting agent or an anti-infective agent), applied to the surface of a soft tissue implant, contained within the "filler" (typically saline, silicone or gel) of a soft tissue implant, or infiltrated into the tissue surrounding the implantation site for the prevention of adhesions in cosmetic surgery include: (a) sprayable collagen-containing formulations such as COSTASIS or CT3; (b) sprayable PEG-containing formulations such as COSEAL, ADHIBIT, FOCALSEAL, SPRAYGEL or DURASEAL; (c) fibrinogen-containing formulations such as FLOSEAL or TISSEAL; (d) hyaluronic acid-containing formulations such as RESTYLANE or PERLANE, HYLAFORM, SYNVISC, SEPRAFILM or SEPRACOAT; (e) polymeric gels for surgical implantation such as REPEL or FLOWGEL; (f) surgical adhesives containing cyanoacrylates such as DERMABOND, INDERMIL, GLUSTITCH, TISSUMEND, VETBOND, HISTOACRYL BLUE and ORABASE SOOTHE-N-SEAL LIQUID PROTECTANT; (g) dextran sulfate gels such as the ADCON series of gels; and (h) lipid based compositions such as ADSURF. Several of the above agents (e.g., formulations containing PEG, collagen, or fibrinogen such as COSEAL, CT3, ADHIBIT, COSTASIS, FOCALSEAL, SPRAYGEL, DURASEAL, TISSEAL AND FLOSEAL) have the added benefit of being hemostats and vascular sealants, which given the suspected role of inadequate hemostasis in the development of capsular contracture, should also be of benefit in the practice of this invention. It should be obvious to one of skill in the art that commercial compositions not specifically cited above as well as next- generation and/or subsequently-developed commercial products are to be anticipated and are suitable for use under the present invention.
As described above, the compositions for the prevention of surgical adhesions can be applied directly or indirectly to the tissue around the cosmetic implant site. The polymeric compositions (either with or without a therapeutic agent) can be administered in any manner described herein. Exemplary methods include either direct application at the time of surgery or with endoscopic, ultrasound, CT, MRI, or fluoroscopic guidance and in conjunction with placement of a cosmetic implant at the surgical site. Representative examples of implants for use in cosmetic procedures include, without limitation, saline breast implants, silicone breast implants, chin and mandibular implants, nasal implants, cheek implants, lip implants, other facial implants, pectoral and chest implants, malar and submalar implants, tissue fillers, and buttocks implants.
The polymeric composition, with or without a fibrosis-inhibiting agent, may be applied during open or endoscopic cosmetic surgery: (a) to the soft tissue implant surface (e.g., as an injectable, solution, paste, gel, in situ forming gel, or mesh) before, during, or after the implantation procedure; (b) to the surface of the tissue (e.g., as an injectable, solution, paste, gel, in situ forming gel or mesh) of the implantation pocket immediately prior to, or during implantation of the soft tissue implant; (c) to the surface of the soft tissue implant and/or the tissue surrounding the implant (e.g., as an injectable, solution, paste, gel, in situ forming gel or mesh) before, during, or after implantation of the soft tissue implant; (d) by topical application of the anti-fibrosis agent into the anatomical space where the soft tissue implant will be placed (particularly useful for this embodiment is the use of polymeric carriers which release the fibrosis-inhibiting agent over a period ranging from several hours to several weeks - fluids, suspensions, emulsions, microemulsions, microspheres, pastes, gels, microparticulates, sprays, aerosols, solid implants and other formulations which release the agent and can be delivered into the region where the implant will be inserted); (e) via percutaneous injection into the tissue surrounding the implant as a solution, as an infusate, or as a sustained release preparation; and/or (f) by any combination of the aforementioned methods. Combination therapies (i.e., combinations of therapeutic agents and combinations with antithrombotic, anti-infective, and/or antiplatelet agents) can also be used.
A composition that includes an anti-scarring agent can be infiltrated into the space (surgically created pocket) where the soft tissue implant will be implanted. In certain applications involving the placement of a cosmetic soft tissue implant, it may be desirable to apply the anti-fibrosis (or anti-infective) composition at a site that is adjacent to an implant (preferably near the implant-tissue interface). This can be accomplished during open, endoscopic or catheter-based cosmetic procedures by applying the polymeric composition, with or without a fibrosis-inhibiting agent: (a) to the implant surface (e.g., as an injectable, solution, paste, gel, in situ forming gel, or mesh) before, during, or after the implantation procedure; (b) to the surface of the adjacent tissue (e.g., as an injectable, solution, paste, gel, in situ forming gel, or mesh) immediately prior to, during, or after implantation of the soft tissue implant; (c) to the surface of the soft tissue implant and the tissue surrounding the implant (e.g., as an injectable, solution, paste, gel, in situ forming gel or mesh) before, during, or after implantation of the implant; (d) by topical application of the composition into the anatomical space (surgical pocket; for example, in breast implants this is the subglandular or subpectoral space) where the soft tissue implant will be placed (particularly useful for this embodiment is the use of polymeric carriers which release the fibrosis-inhibiting agent over a period ranging from several hours to several weeks -fluids, suspensions, emulsions, microemulsions, microspheres, pastes, gels, microparticulates, sprays, aerosols, solid implants and other formulations which release the agent can be delivered into the region where the device will be inserted); (e) via percutaneous injection into the tissue surrounding the soft tissue implant as a solution, as an infusate, or as a sustained release preparation; and/or (f) by any combination of the aforementioned methods. Combination therapies (i.e., combinations of therapeutic agents and combinations with antithrombotic, anti-infective, and/or antiplatelet agents) can also be used. In one aspect, the polymeric composition may be delivered to the soft tissue implant (or implant/tissue interface) in the form of a spray or gel during open, endoscopic or catheter-based procedures. The fibrosis- inhibiting (and/or anti-infective) agent can be incorporated directly into the surgical adhesion barrier or it can be incorporated into a secondary carrier (polymeric or non-polymeric), as described above, that is then incorporated into the adhesion barrier. Examples of polymer compositions that may be in the form of a spray or gel include poly(ethylene glycol)-based systems, fibrinogen-containing systems, hyaluronic acid and crosslinked hyaluronic acid compositions. These compositions can be applied as the final composition or they can be applied as materials that form a crosslinked gel in situ.
In another aspect, an activated polymer is dissolved in a biologically acceptable buffer that has a pH lower that 6.8. The resultant solution is then applied to the desired tissue surface in the presence of a second biologically acceptable buffer that has a pH greater than 7.5. Application of the reaction mixture to the tissue site may be by extrusion, brushing, spraying or by any other convenient means. Following application of the composition to the surgical site, any excess solution may be removed from the surgical site if deemed necessary. At this point in time, the surgical site can be closed using conventional means (e.g., sutures, staples, or a bioadhesive). In one embodiment, the activated polymer can form a covalent bond with the tissue to which it is applied may be used. Polymers containing and/or terminated with electrophilic groups such as succinimidyl, aldehyde, epoxide, isocyanate, vinyl, vinyl sulfone, maleimide, -S-S-(C5H4N) or activated esters, such as are used in peptide synthesis may be used as the reagents. For example, a 4 armed NHS-derivatized polyethylene glycol (e.g., pentaerythritol poly(ethylene glycol)ether tetra-succinimidyl glutarate) may be applied to the tissue in the solid form or in a solution form. In this embodiment, the 4 armed NHS-derivatized polyethylene glycol is dissolved in an acidic solution (pH about 2-3) and is then co-applied to the tissue using a basic buffer (pH > about 8). The antifibrosisfibrosis-inhibiting agent(s) may be incorporated directly into either the 4 armed NHS- derivatized polyethylene glycol, the acidic solution or the basic buffer. In another embodiment, the fibrosis-inhibiting agent may be incorporated into a secondary carrier that may then be incorporated into the 4 armed NHS- derivatized polyethylene glycol, the acidic solution and/or the basic buffer. The secondary carriers may include microparticles and/or microspheres which are made from degradable polymers. The degradable polymers may include polyesters, where the polyester may comprise the residues of one or more of the monomers selected from lactide, lactic acid, glycolide, glycolic acid, e-caprolactone, gamma-caprolactone, hydroxyvaleric acid, hydroxybutyric acid, beta-butyrolactone, gamma-butyrolactone, gamma- valerolactone, γ-decanolactone, δ-decanolactone, trimethylene carbonate, 1 ,4-dioxane-2-one or 1 ,5-dioxepan-2one, and block copolymers of the form X-Y, Y-X-Y, R-(Y-X)n, R-(X-Y)n and X-Y-X where X in a polyalkylene oxide (e.g., poly(ethylene glycol, polypropylene glycol) and block copolymers of poly(ethylene oxide) and polyφropylene oxide) (e.g., PLURONIC and PLURONIC R series of polymers from BASF Corporation, Mount Olive, NJ) and Y is a biodegradable polyester, where the polyester may comprise the residues of one or more of the monomers selected from lactide, lactic acid, glycolide, glycolic acid, e-caprolactone, gamma-caprolactone, hydroxyvaleric acid, hydroxybutyric acid, beta-butyrolactone, gamma- butyrolactone, gamma-valerolactone, γ-decanolactone, δ-decanolactone, trimethylene carbonate, 1 ,4-dioxane-2-one or 1 ,5-dioxepan-2one (e.g., PLG- PEG-PLG) and R is a multifunctional initiator. In another embodiment, the tissue reactive polymer may be applied initially and then the fibrosis- inhibiting agent may then be applied to the coated tissue. The fibrosis- inhibiting agent may be applied directly to the tissue or it may be incorporated into a secondary carrier. The secondary carriers may include microspheres (as described above), microparticles (as described above), gels (e.g., hyaluronic acid, carboxymethyl cellulose, dextran, poly(ethylene oxide) - poly(propylene oxide) block copolymers as well as blends, association complexes and crosslinked compositions thereof) and films (degradable polyesters, where the polyester may comprise the residues of one or more of the monomers selected from lactide, lactic acid, glycolide, glycolic acid, e-caprolactone, gamma-caprolactone, hydroxyvaleric acid, hydroxybutyric acid, beta-butyrolactone, gamma-butyrolactone, gamma- valerolactone, γ-decanolactone, δ-decanolactone, trimethylene carbonate, 1,4-dioxane-2-one or 1 ,5-dioxepan-2one, and block copolymers of the form X-Y, Y-X-Y, R-(Y-X)n, R-(X-Y)n and X-Y-X where X in a polyalkylene oxide (e.g., poly(ethylene glycol, poly(propylene glycol) and block copolymers of poly(ethylene oxide) and poly(propylene oxide) (e.g., PLURONIC and PLURONIC R series of polymers from BASF Corporation, Mount Olive, NJ) and Y is a biodegradable polyester, where the polyester may comprise the residues of one or more of the monomers selected from lactide, lactic acid, glycolide, glycolic acid, e-caprolactone, gamma-caprolactone, hydroxyvaleric acid, hydroxybutyric acid, beta-butyrolactone, gamma- butyrolactone, gamma-valerolactone, γ-decanolactone, δ-decanolactone, trimethylene carbonate, 1 ,4-dioxane-2-one or 1 ,5-dioxepan-2one (e.g., PLG- PEG-PLG) and R is a multifunctional initiator, hyaluronic acid, carboxymethyl cellulose, dextran, poly(ethylene oxide) - poly(propylene oxide) block copolymers as well as blends, association complexes and crosslinked compositions thereof.
In yet another aspect, an activated polymer can be applied to the surgical site in the solid state. The activated polymer can react with the tissue surface to which it was applied as the polymer hydrates. A biologically acceptable buffer, with a pH greater than 7.5 can be applied to the tissue before and/or after the solid activated polymer has been applied. In one embodiment, the activated polymer can form a covalent bond with the tissue to which it is applied may be used. Polymers containing and/or terminated with electrophilic groups such as succinimidyl, aldehyde, epoxide, isocyanate, vinyl, vinyl sulfone, maleimide, -S-S-(C5H4N) or activated esters, such as are used in peptide synthesis may be used as the reagents. For example, a 4 armed NHS-derivatized polyethylene glycol (e.g., pentaerythritol poly(ethylene glycol)ether tetra-succinimidyl glutarate) may be applied to the tissue in the solid form. The antifibrosisfibrosis- inhibiting agent(s) may be incorporated directly into either the 4 armed NHS- derivatized polyethylene glycol, or the basic buffer. In another embodiment, the fibrosis-inhibiting agent may be incorporated into a secondary carrier that may then be incorporated into the 4 armed NHS-derivatized polyethylene glycol, and/or the basic buffer. The secondary carriers may include microparticles and/or microspheres which are made from degradable polymers. The degradable polymers may include polyesters, where the polyester may comprise the residues of one or more of the monomers selected from lactide, lactic acid, glycolide, glycolic acid, e- caprolactone, gamma-caprolactone, hydroxyvaleric acid, hydroxybutyric acid, beta-butyrolactone, gamma-butyrolactone, gamma-valerolactone, v- decanolactone, δ-decanolactone, trimethylene carbonate, 1,4-dioxane-2- one or 1 ,5-dioxepan-2one, and block copolymers of the form X-Y, Y-X-Y, R- (Y-X)n, R-(X-Y)n and X-Y-X where X in a polyalkylene oxide (e.g., poly(ethylene glycol, polyφropylene glycol) and block copolymers of poly(ethylene oxide) and poly(propylene oxide) (e.g., PLURONIC and PLURONIC R series of polymers from BASF Corporation, Mount Olive, NJ) and Y is a biodegradable polyester, where the polyester may comprise the residues of one or more of the monomers selected from lactide, lactic acid, glycolide, glycolic acid, e-caprolactone, gamma-caprolactone, hydroxyvaleric acid, hydroxybutyric acid, beta-butyrolactone, gamma- butyrolactone, gamma-valerolactone, γ-decanolactone, δ-decanolactone, trimethylene carbonate, 1 ,4-dioxane-2-one or 1 ,5-dioxepan-2one (e.g., PLG- PEG-PLG) and R is a multifunctional initiator. In another embodiment, the tissue reactive polymer may be applied initially and then the fibrosis- inhibiting agent may then be applied to the coated tissue. The fibrosis- inhibiting agent may be applied directly to the tissue or it may be incorporated into a secondary carrier. The secondary carriers may include microspheres (as described above), microparticles (as described above), gels (e.g., hyaluronic acid, carboxymethyl cellulose, dextran, poly(ethylene oxide) - polyφropylene oxide) block copolymers as well as blends, association complexes and crosslinked compositions thereof) and films (degradable polyesters, where the polyester may comprise the residues of one or more of the monomers selected from lactide, lactic acid, glycolide, glycolic acid, e-caprolactone, gamma-caprolactone, hydroxyvaleric acid, hydroxybutyric acid, beta-butyrolactone, gamma-butyrolactone, gamma- valerolactone, γ-decanolactone, δ-decanolactone, trimethylene carbonate, 1 ,4-dioxane-2-one or 1,5-dioxepan-2one, and block copolymers of the form X-Y, Y-X-Y, R-(Y-X)n, R-(X-Y)n and X-Y-X where X in a polyalkylene oxide (e.g., poly(ethylene glycol, poly(propylene glycol) and block copolymers of poly(ethylene oxide) and poly(propylene oxide) (e.g., PLURONIC and PLURONIC R series of polymers from BASF Corporation, Mount Olive, NJ) and Y is a biodegradable polyester, where the polyester may comprise the residues of one or more of the monomers selected from lactide, lactic acid, glycolide, glycolic acid, e-caprolactone, gamma-caprolactone, hydroxyvaleric acid, hydroxybutyric acid, beta-butyrolactone, gamma- butyrolactone, gamma-valerolactone, γ-decanolactone, δ-decanolactone, trimethylene carbonate, 1 ,4-dioxane-2-one or 1 ,5-dioxepan-2one (e.g., PLG- PEG-PLG) and R is a multifunctional initiator, hyaluronic acid, carboxymethyl cellulose, dextranτ poly(ethylene oxide) - polypropylene oxide) block copolymers as well as blends, association complexes and crosslinked compositions thereof.
vii) Agents and Dosages of Fibrosis-lnhibitors In certain aspects of the invention, compositions are provided that can release a therapeutic agent able to reduce scarring (i.e., a fibrosis- inhibiting agent) at a surgical site. Within one embodiment of the invention, surgical adhesion barriers may include or be adapted to release an agent that inhibits one or more of the five general components of the process of fibrosis (or scarring), including: inflammatory response and inflammation, migration and proliferation of connective tissue cells (such as fibroblasts or smooth muscle cells), formation of new blood vessels (angiogenesis), deposition of extracellular matrix (ECM), and remodeling (maturation and organization of the fibrous tissue). By inhibiting one or more of the components of fibrosis (or scarring), the overgrowth of scar tissue may be inhibited or reduced.
Examples of fibrosis-inhibiting agents for use in surgical adhesion barriers include the following: ZD-6474, AP-23573, synthadotin, S- 0885, aplidine, ixabepilone, IDN-5390, SB-2723005, ABT-518, combretastatin, anecortave acetate, SB-715992, temsirolimus, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, as well as analogues and derivatives of the aforementioned.
Additional examples of anti-scarring agents which can be used include those having a high potency in the assays described herein (1- 10OnM IC50 range) such as isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, dolastatin 15, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17- DMAG, and tacrolimus; those having a mid-potency in the assays described herein (100-500 nM IC50 range) such as loteprednol etabonate, juglone, prednisolone, puromycin, 3-BAABE, cladribine, and mannose-6-phosphate; and those having a low potency in the assays described herein (500- 1000nm range IC50 range) such as 5-azacytidine, Ly333531 (ruboxistaurin), and simvastatin.
The drug dose administered from the present compositions for surgical adhesion prevention will depend on a variety of factors, including the type of formulation, the location of the treatment site, and the type of condition being treated. However, certain principles can be applied in the application of this art. Drug dose can be calculated as a function of dose per unit area (of the treatment site), total drug dose administered can be measured and appropriate surface concentrations of active drug can be determined. Drugs are to be used at concentrations that range from several times more than to 50%, 20%, 10%, 5%, or even less than 1% of the concentration typically used in a single systemic dose application. In certain aspects, the anti-scarring agent is released from the composition in effective concentrations in a time period that may be measured from the time of infiltration into tissue adjacent to the device, which ranges from about less than 1 day to about 180 days. Generally, the release time may also be from about less than 1 day to about 180 days; from about 7 days to about 14 days; from about 14 days to about 28 days; from about 28 days to about 56 days; from about 56 days to about 90 days; from about 90 days to about 180 days. In one aspect, the drug is released in effective concentrations for a period ranging from 1 - 90 days.
The exemplary anti-fibrosing agents, used alone or in combination, should be administered under the following dosing guidelines. The total amount (dose) of anti-scarring agent in the composition can be in the range of about 0.01 μg-10 μg, or 10 μg-10 mg, or 10 mg-250 mg, or 250 mg-1000 mg, or 1000 mg-2500 mg. The dose (amount) of anti-scarring agent per unit area of surface to which the agent is applied may be in the range of about 0.01 μg/mm2 - 1 μg/mm2, or 1 μg/mm2 - 10 μg/mm2, or 10 μg/mm2 - 250 μg/mm2, 250 μg/mm2 - 1000 μg/mm2, or 1000 μg/mm2 - 2500 μg/mm2. - -
For high potency drugs, the total dose typically should not exceed 200 mg (range of 0.1 μg to 200 mg) and preferably 1 μg to 100 mg; dose per unit area of 0.01 μg - 100 μg per mm2; preferably 0.1 μg/mm2 - 20 μg/mm2; and minimum concentration of 10~8- 10"4 M of agent should be maintained on the implant or barrier surface. For mid-potency drugs, the total dose typically should not to exceed 500 mg (range of 1.0 μg to 500 mg) and preferably 1 μg to 200 mg; dose per unit area of 0.01 μg - 200 μg per mm2, preferably 0.1 μg/mm2 - 40 μg/mm2; and minimum concentration of 10" 8- 10"4 M of agent should be maintained on the implant or barrier surface. For low potency drugs, the total dose typically should not exceed 1000 mg (range of 0.1 μg to 1000 mg), preferably 1 μg to 500 mg; dose per unit area of 0.01 μg - 500 μg per mm2; preferably 0.1 μg/mm2 - 100 μg/mm2; and minimum concentration of 10"8- 10"4 M of agent should to be maintained on the implant or barrier surface. Provided below are exemplary dosage ranges for various anti- scarring agents that can be used in conjunction with compositions for treating or preventing surgical adhesions in accordance with the invention.
(A) Angiogenesis inhibitors including alphastatin, ZD-6474, IDN-5390, SB- 2723005, ABT-518, combretastatin, and anecortane, analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10^- 10"4 M of agent is to be maintained on the implant or barrier surface.
(B) mTOR inhibitors including AP-23573 and temsirolimus, analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10^- 10"4 M of agent is to be maintained on the implant or barrier surface.
(C) Tubulin antagonists including synthadotin, analogues and derivatives thereofhtotal dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 108 - 10"4 M of agent is to be maintained on the implant or barrier surface. (D) Epithilones including ixabepilone and analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10~8- 10"4 M of agent is to be maintained on the implant or barrier surface. (E) Kinesin antagonists including SB-715992 and analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10"8- 10"4 M of agent is to be maintained on the implant or barrier surface. (F) TNF alpha antagonists including etanercept, humicade, adalimumab and analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10^- 10"4 M of agent is to be maintained on the implant or barrier surface. (G) AKT inhibitor including erucylphosphocholine and analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of -JO'8- 10"4 M of agent is to be maintained on the implant or barrier surface. (H) FGF Inhibitors including IDN-5390 and analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10"8- 10"4 M of agent is to be maintained on the implant or barrier surface. (I) Collagenase Antagonists including S-0885 and analogues and derivatives thereof: total dose not to exceed 1000 mg (range of 0.1 μg to 1000 mg); preferred 1 μg to 500 mg. Dose per unit area of 0.01 μg - 500 μg per mm2; preferred dose of 0.1 μg/mm2 - 100 μg/mm2. Minimum concentration of 10"8 - 10"4 M of agent is to be maintained on the implant or barrier surface. (J) NF Kappa B Inhibitors including Bortezomib and analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10"8- 10"4 M of agent is to be maintained on the implant or barrier surface. (K) Elongation factor-1 alpha inhibitors including aplidine and analogues and derivatives thereof: total dose not to exceed 1000 mg (range of 0.1 μg to 1000 mg); preferred 1 μg to 500 mg. Dose per unit area of 0.01 μg - 500 μg per mm2; preferred dose of 0.1 μg/mm2 - 100 μg/mm2. Minimum concentration of 108 - 104 M of agent is to be maintained on the implant or barrier surface. (L) Tyrosine kinase inhibitors including Gefitinib and analogues and derivatives thereof: total dose not to exceed 1000 mg (range of 0.1 μg to 1000 mg); preferred 1 μg to 500 mg. Dose per unit area of 0.01 μg - 500 μg per mm2; preferred dose of 0.1 μg/mm2 - 100 μg/mm2. Minimum concentration of 10^- 10"4 M of agent is to be maintained on the implant or barrier surface.
According to another aspect, any anti-infective agent described above may be used in combination with the present compositions for surgical adhesion prevention. Exemplary anti-infective agents include (A) anthracyclines (e.g., doxorubicin and mitoxantrone), (B) fluoropyrimidines (e.g., 5-FU), (C) folic acid antagonists (e.g., methotrexate), (D) podophylotoxins (e.g., etoposide), (E) camptothecins, (F) hydroxyureas, and (G) platinum complexes (e.g., cisplatin), as well as analogues and derivatives of the aforementioned.
The drug dose administered from the present compositions for prevention or inhibition of infection in accordance with the present invention will depend on a variety of factors, including the type of formulation, the location of the treatment site, and the type of condition being treated. However, certain principles can be applied in the application of this art. Drug dose can be calculated as a function of dose per unit area (of the treatment site), total drug dose administered can be measured and appropriate surface concentrations of active drug can be determined. Drugs are to be used at concentrations that range from several times more than to 50%, 20%, 10%, 5%, or even less than 1% of the concentration typically used in a single anti-infective systemic dose application. In certain aspects, the anti- infective agent is released from the composition in effective concentrations in a time period that may be measured from the time of infiltration into tissue adjacent to the device, which ranges from about less than 1 day to about 180 days. Generally, the release time may also be from about less than 1 day to about 180 days; from about 7 days to about 14 days; from about 14 days to about 28 days; from about 28 days to about 56 days; from about 56 days to about 90 days; from about 90 days to about 180 days. W
The drug dose administered from the present compositions for prevention or inhibition of infection in accordance with the present invention will depend on a variety of factors, including the type of formulation, the location of the treatment site, and the type of condition being treated. However, certain principles can be applied in the application of this art. Drug dose can be calculated as a function of dose per unit area (of the treatment site), total drug dose administered can be measured and appropriate surface concentrations of active drug can be determined. Drugs are to be used at concentrations that range from several times more than to 50%, 20%, 10%, 5%, or even less than 1% of the concentration typically used in a single anti-infective systemic dose application. In certain aspects, the anti-infective agent is released from the composition in effective concentrations in a time period that may be measured from the time of infiltration into tissue adjacent to the device, which ranges from about less than 1 day to about 180 days. Generally, the release time may also be from about less than 1 day to about 180 days; from about 7 days to about .14 days; from about 14 days to about 28 days; from about 28 days to about 56 days; from about 56 days to about 90 days; from about 90 days to about 180 days.
The exemplary anti-infective agents, used alone or in combination, should be administered under the following dosing guidelines. The total amount (dose) of anti-infective agent in the composition can be in the range of about 0.01 μg-1 μg, or about 1 μg-10 μg, or about 10 μg-1 mg, or about 1 mg to 10 mg, or about 10 mg-100 mg, or about 100 mg to 250 mg, or about 250 mg-1000 mg. The dose (amount) of anti-infective agent per unit area of device or tissue surface to which the agent is applied may be in the range of about 0.01 μg/mm2 - 1 μg/mm2, or about 1 μg/mm2 - 10 μg/mm2, or about 10 μg/mm2 - 100 μg/mm2, or about 100 μg/mm2 to 250 μg/mm2, or about 250 μg/mm2 - 1000 μg/mm2. As different compositions will release the anti-infective agent at differing rates, the above dosing parameters should be utilized in combination with the release rate of the drug from the composition such that a minimum concentration of about 10 s M to 1(T7 M, or about 1(T7 M to 1(T6 M about 10"6 M to 1(T5 M or about 10"5 M to 10"4 M of the agent is maintained on the tissue surface.
(b) Inflammatory Arthritis
In one aspect, the present invention provides compositions for the treatment and prevention of inflammatory arthritis. The compositions of the present invention can comprise one or more polymeric carriers and an anti-scarring agent.
Inflammatory arthritis is a serious health problem in developed countries, particularly given the increasing number of aged individuals and includes a variety of conditions including, but not limited to, rheumatoid arthritis, systemic lupus erythematosus, systemic sclerosis (scleroderma), mixed connective tissue disease, Sjogren's syndrome, ankylosing spondylitis, Behcet's syndrome, sarcoidosis, and osteoarthritis - all of which feature inflamed and/or painful joints as a prominent symptom.
In one aspect, the present compositions may be used to treat or prevent osteoarthritis (OA). Osteoarthritis is a common, debilitating, costly, and currently incurable disease. The disease is characterized by abnormal functioning of chondrocytes and their terminal differentiation, leading ultimately to the initiation of OA and the breakdown of the cartilage matrix in the articular cartilage of affected joints. Age is the most powerful risk factor for OA, but major joint trauma, excessive weight, and repetitive joint use are also important risk factors for OA. The pattern of joint involvement in OA is also influenced by prior vocational or avocational overload.
OA can be of primary (idiopathic) and secondary types. Primary OA is most commonly related to age. Repetitive use of the joints, particularly the weight-bearing joints such as hips, knees, feet and back, irritates and inflames the joints and causes joint pain and swelling. Eventually, cartilage begins to degenerate by flaking or forming tiny crevasses. In advanced cases, there is a total loss of the cartilage cushion between the bones of the joints. Loss of the cartilage cushion causes friction between the bones, leading to pain and limitation of joint mobility. Inflammation of the cartilage can also stimulate new bone outgrowths (spurs) to form around the joints.
Secondary OA is pathologically indistinguishable from idiopathic OA but is attributable to another disease or condition. Conditions that can lead to secondary OA include obesity, repeated trauma (e.g., ligament tears, cartilage tears), surgery to the joint structures (ligament repairs, menisectomy, cartilage removal), abnormal joints at birth (congenital abnormalities), gout, diabetes, and other metabolic disorders.
In one aspect, the present compositions may be used to treat or prevent rheumatoid arthritis (RA). Rheumatoid arthritis is a multisystem chronic, relapsing, inflammatory disease of unknown cause. Although many organs can be affected, RA is basically a severe form of chronic synovitis that sometimes leads to destruction and ankylosis of affected joints (Robbins Pathological Basis of Disease, by R. S. Cotran, V. Kumar, and S. L. Robbins, W.B. Saunders Co., 1989). Pathologically the disease is characterized by a marked thickening of the synovial membrane which forms villous projections that extend into the joint space, multilayering of the synoviocyte lining (synoviocyte proliferation), infiltration of the synovial membrane with white blood cells (macrophages, lymphocytes, plasma cells, and lymphoid follicles; called an "inflammatory synovitis"), and deposition of fibrin with cellular necrosis within the synovium. The tissue formed as a result of this process is called pannus and eventually the pannus grows to fill the joint space. The pannus develops an extensive network of new blood vessels through the process of angiogenesis which is essential to the evolution of the synovitis. Digestive enzymes (matrix metalloproteinases such as collagenase and stromelysin) and other mediators of the inflammatory process (e.g., hydrogen peroxide, superoxides, lysosomal enzymes, and products of arachadonic acid metabolism) released from the cells of the pannus tissue break down the cartilage matrix and cause progressive destruction of the cartilage. The pannus invades the articular cartilage leading to erosions and fragmentation of the cartilage tissue. Eventually there is erosion of the subchondral bone with fibrous ankylosis and ultimately bony ankylosis, of the involved joint.
It is generally believed, but not conclusively proven, that RA is an autoimmune disease, and that many different arthrogenic stimuli activate the immune response in the immunogenetically susceptible host. Both exogenous infectious agents (Ebstein-Barr virus, rubella virus, cytomegalovirus, herpes virus, human T-cell lymphotropic virus, mycoplasma, and others) and endogenous proteins (collagen, proteoglycans, altered immunoglobulins) have been implicated as the causative agent which triggers an inappropriate host immune response. Regardless of the inciting agent, autoimmunity plays a role in the progression of the disease. In particular, the relevant antigen is ingested by antigen-presenting cells (macrophages or dendritic cells in the synovial membrane), processed, and presented to T lymphocytes. The T cells initiate a cellular immune response and stimulate the proliferation and differentiation of B lymphocytes into plasma cells. The end result is the production of an excessive, inappropriate immune response directed against the host tissues (e.g., antibodies directed against type Il collagen, antibodies directed against the Fc portion of autologous IgG (called "Rheumatoid Factor")). This further amplifies the immune response and hastens the destruction of the cartilage tissue. Once this cascade is initiated, numerous mediators of cartilage destruction are responsible for the progression of rheumatoid arthritis.
In rheumatoid arthritis, articular cartilage is destroyed when it is invaded by pannus tissue (which is composed of inflammatory cells, blood vessels, and connective tissue). Generally, chronic inflammation in itself is insufficient to result in damage to the joint surface, but a permanent deficit is created once fibrovascular tissue digests the cartilage tissue. The abnormal growth of blood vessels and pannus tissue may be inhibited by treatment with fibrosis-inhibiting compositions, or fibrosis-inhibiting agents. Incorporation of an anti-scarring agent into these compositions or other intra-articular formulations, can provide an approach that can reduce the rate of progression of the disease.
Thus, within one aspect of the present invention, methods are provided for treating or preventing inflammatory arthritis comprising the step of administering to a patient in need thereof a therapeutically effective amount of an anti-scarring agent or a composition comprising an anti- scarring agent. Inflammatory arthritis includes a variety of conditions including, but not limited to, rheumatoid arthritis, systemic lupus erythematosus, systemic sclerosis (scleroderma), mixed connective tissue disease, Sjogren's syndrome, ankylosing spondylitis, Behcet's syndrome, sarcoidosis, and osteoarthritis - all of which feature inflamed and/or painful joints as a prominent symptom.
An effective anti-scarring therapy for inflammatory arthritis will accomplish one or more of the following: (i) decrease the severity of symptoms (pain, swelling and tenderness of affected joints; morning stiffness, weakness, fatigue, anorexia, weight loss); (ii) decrease the severity of clinical signs of the disease (thickening of the joint capsule, synovial hypertrophy, joint effusion, soft tissue contractures, decreased range of motion, ankylosis and fixed joint deformity); (iii) decrease the extraarticular manifestations of the disease (rheumatic nodules, vasculitis, pulmonary nodules, interstitial fibrosis, pericarditis, episcleritis, iritis, Felty's syndrome, osteoporosis); (iv) increase the frequency and duration of disease remission/symptom-free periods; (v) prevent fixed impairment and disability; and/or (vi) prevent/attenuate chronic progression of the disease.
According to the present invention, any anti-scarring agent described above could be utilized in the practice of this invention. Within certain embodiments of the invention, the composition may release an agent that inhibits one or more of the general components of the process of fibrosis (or scarring) associated with inflammatory arthritis, including: (a) formation of new blood vessels (angiogenesis), (b) migration and/or proliferation of connective tissue cells (such as fibroblasts or synoviocytes), (c) destruction of the cartilage matrix by metalloproteinase activity, (d) inflammatory response by cytokines (such as IL-1 , TNFα, FGF, VEGF). By inhibiting one or more of the components of fibrosis (or scarring), cartilage loss may be inhibited or reduced.
In one aspect, the composition includes an anti-scarring agent and a polymeric carrier suitable for application to treat inflammatory arthritis. Numerous polymeric and non-polymeric delivery systems and compositions containing an anti-scarring agent for use in the treatment of inflammatory arthritis have been described above. An anti-scarring agent may be administered systemically (orally, intravenously, or by intramuscular or subcutaneous injection) in the minimum dose to achieve the above mentioned results. For patients with only a small number of joints affected, or with disease more prominent in a limited number of joints, the anti- scarring agent can be directly injected into the affected joint (intra-articular injection) via percutaneous needle insertion into the joint capsule, or as part of an arthroscopic procedure performed on the joint. In a preferred embodiment, the intra-articular formulation containing a fibrosis-inhibitor is administered to a joint following an injury with a high probability of inducing subsequent arthritis (e.g., cruciate ligament tears in the knee, meniscal tears in the knee). The agent is administered for a period sufficient (either through sustained release preparations and/or repeated injections) to protect the cartilage from breakdown as a result of the injury (or the surgical procedure used to treat it).
The anti-scarring agent can be administered in any manner described herein. However, preferred methods of administration include intravenous, oral, subcutaneous injection, or intramuscular injection. A particularly preferred embodiment involves the administration of the fibrosis- inhibiting compound as an intra-articular injection (directly, via arthroscopic or radiologic guidance, or irrigated into the joint as part of an open surgical procedure). The anti-scarring agent can be administered as a chronic low dose therapy to prevent disease progression, prolong disease remission, or decrease symptoms in active disease. Alternatively, the therapeutic agent can be administered in higher doses as a "pulse" therapy to induce remission in acutely active disease; such as the acute inflammation that follows a traumatic joint injury (intra-articular fractures, ligament tears, meniscal tears, as described below). The minimum dose capable of achieving these endpoints can be used and can vary according to patient, severity of disease, formulation of the administered agent, potency and/or tolerability of the agent, clearance of the agent from the joint, and route of administration.
In one preferred embodiment, the fibrosis-inhibiting composition can be an intra-articular injectable hyaluronic acid-based composition. Hyaluronic acid, which is a normal element of joint synovial fluid, lubricates the joint surface during normal activities (resting, walking) and helps prevent mechanical damage and decrease shock on the joint in high impact activities (such as running, jumping). In patients with OA, the elasticity and viscosity of the synovial fluid and the synovial hyaluronic acid concentration are reduced. It is believed that this contributes to the breakdown of the articular cartilage within the joint. Intra-articularly administered HA (typically sodium hyaluronate) penetrates the articular cartilage surface, the synovial tissue, and the capsule of the joint for a period of time after injection. By injecting hyaluronic acid into the joint (known as visco-supplementation), it is possible to partially restore the normal environment of the synovial fluid, reduce pain, and potentially prevent further damage and disability. Representative examples of hyaluronic acid compositions used in visco-supplementation are described in U.S. Patents Nos. 6,654,120, 6,645,945, and 6,635,287. As such, HA- containing materials are administered as an intra-articular injection (as either a single treatment or a course of repeated treatment cycles) for the treatment of painful osteoarthritis of the knee in patients who have insufficient pain relief from conservative therapies. Occasionally other joints such as hips (injected under fluoroscopy), ankles, shoulders and elbow joints, are also injected with HA to relieve the symptoms of the disease in those particular joints. Depending upon the particular commercial product, the HA material is injected into the joint once a week for 5 to 6 consecutive weeks. When effective, patients may report that they receive symptomatic relief for a period of 6 months or more - at which time the cycle may be repeated to prolong the activity of the therapy. Despite the sustained benefit in some patients, the injected HA is rapidly cleared (removed) from the joint by the body over a period of several days. Prolonging the residence time of the HA in the joint by inhibiting its breakdown may be expected to enhance its efficacy and increase the duration of symptomatic relief. By adding a fibrosis-inhibiting agent to the HA, the intra-articular injection has the added benefit of helping to prevent cartilage breakdown (i.e., it is "chondroprotective").
A variety of commercially available HA compositions for the treatment of inflammatory arthritis may be combined with one or more agents according to the present invention including: SYNVISC (Biomatrix, Inc., Ridgefield, NJ) - an elastoviscous fluid containing hylan (a derivative of sodium hyaluronate (hyaluronan)) polymers derived from rooster combs, HYALGAN (Sanofi-Synthelabo Inc. New York, NY), and ORTHOVISC (Ortho Biotech Products, Bridgewater, NJ) - a highly purified, high molecular weight, high viscosity injectable form of HA intended to relieve pain and to improve joint mobility and range of motion in patients suffering from osteoarthritis (OA) of the knee. ORTHOVISC is injected into the knee to restore the elasticity and viscosity of the synovial fluid. HYVISC is a high molecular weight, injectable HA product developed by Anika Therapeutics (Wobum, MA) currently being used to treat osteoarthritis and lameness in racehorses. Other HA-based viscosupplementation products for the treatment of osteoarthritis include SUPARTZ from Seikagaku Corp. (Japan), SUPLASYN from Bioniche Life Sciences, Inc. (Canada), ARTHREASE from DePuy Orthopaedics, Inc. (Warsaw, IN), and DUROLANE from Q-Med AB (Sweden).
In one aspect, the compositions of the present invention may be used for the management of osteoarthritis in animals (e.g., horses). It should be noted that some HA products (notably HYVISC by Boehringer lngelheim Vetmedica, St. Joseph, MO) are used in veterinary applications (typically in horses to treat osteoarthritis and lameness).
Other intra-articular compositions used to treat arthritis include corticosteroids. The most common corticosteroids currently used for inflammatory arthritis are methylprednisolone acetate (DEPO-MEDROL, Pharmacia & Upjohn Company, Kalamazoo, Ml), and triacinolone acetonide (KENALOG, Bristol-Myers Squibb, New York, NY). By adding a fibrosis- inhibiting agent to the intra-articular corticosteroid injection, the intra- articular injection has the added benefit of helping to prevent cartilage breakdown [i.e., it is "chondroprotective").
Formulations that can be used in these applications include solutions, topical formulations (e.g., solution, cream, ointment, gel) emulsions, micellar solutions, gels (crosslinked and non-crosslinked), suspensions and/or pastes. One form of the formulation is as an injectable composition. For compositions that further contain a polymer to increase the viscosity of the formulation, hyaluronic acid (crosslinked, derivatized and/or non-crosslinked) is an exemplary material. These formulations can further comprise additional polymers (e.g., collagen, poly(ethylene glycol) or dextran) as well as biocompatible solvents (e.g., ethanol, DMSO, or NMP). In one embodiment, the fibrosis-inhibiting therapeutic agent can be incorporated directly into the formulation. In another embodiment, the fibrosis-inhibiting therapeutic agent can be incorporated into a secondary carrier (e.g., micelles, liposomes, emulsions, microspheres, nanospheres etc, as described above). The microsphere and nanospheres may be comprised of degradable polymers. Degradable polymers that can be used include poly(hydroxyl esters) {e.g., PLGA, PLA, PCL, and the like), as well as polyanhydrides, polyorthoesters and polysaccharides (e.g., chitosan and alginates).
In one embodiment, the fibrosis-inhibiting agent further comprises a polymer where the polymer is a degradable polymer. The degradable polymers may include polyesters where the polyester may comprise the residues of one or more of the monomers selected from lactide, lactic acid, glycolide, glycolic acid, e-caprolactone, gamma- , caprolactone, hydroxyvaleric acid, hydroxybutyric acid, beta-butyrolactone, gamma-butyrolactone, gamma-valerolactone, γ-decanolactone, δ- decanolactone, trimethylene carbonate, 1 ,4-dioxane-2-one or 1 ,5-dioxepan- 2one, and block copolymers of the form X-Y, Y-X-Y, R-(Y-X)n, R-(X-Y)n and X-Y-X where X in a polyalkylene oxide (e.g., poly(ethylene glycol, poly(propylene glycol) and block copolymers of poly(ethylene oxide) and polypropylene oxide) (e.g., PLURONIC and PLURONIC R series of polymers from BASF Corporation, Mount Olive, NJ) and Y is a biodegradable polyester, where the polyester may comprise the residues of one or more of the monomers selected from lactide, lactic acid, glycolide, glycolic acid, e-caprolactone, gamma-caprolactone, hydroxyvaleric acid, hydroxybutyric acid, beta-butyrolactone, gamma-butyrolactone, gamma- valerolactone, γ-decanolactone, δ-decanolactone, trimethylene carbonate, 1 ,4-dioxane-2-one or 1 ,5-dioxepan-2one (e.g., PLG-PEG-PLG) and R is a multifunctional initiator. In another embodiment, the fibrosis-inhibiting agent/composition may further comprise a solvent, a liquid oligomer or liquid polymer such that the final composition may be passed through a 18G needle. The reagents that may be used include ethanol, NMP, PEG 200, PEG 300 and low molecular weight liquid polymers of the form X-Y, Y-X-Y, R-(Y-X)n, R-(X-Y)n and X-Y-X where X in a polyalkylene oxide (e.g., poly(ethylene glycol, poly(propylene glycol) and block copolymers of poly(ethylene oxide) and poly(propylene oxide) (e.g., PLURONIC and PLURONIC R series of polymers from BASF Corporation, Mount Olive, NJ) and Y is a biodegradable polyester, where the polyester may comprise the residues of one or more of the monomers selected from lactide, lactic acid, glycolide, glycolic acid, e-caprolactone, gamma-caprolactone, hydroxyvaleric acid, hydroxybutyric acid, beta-butyrolactone, gamma- butyrolactone, gamma-valerolactone, γ-decanolactone, δ-decanolactone, trimethylene carbonate, 1 ,4-dioxane-2-one or 1 ,5-dioxepan-2one (e.g., PLG- PEG-PLG) and R is a multifunctional initiator.
In another embodiment, the fibrosis-inhibiting agent may be in the form of a solution or suspension in an organic solvent, a liquid oligomer or a liquid polymer. In this embodiment, reagents such as ethanol, NMP, PEG 200, PEG 300 and low molecular weight liquid polymers of the form X- Y, Y-X-Y, R-(Y-X)n, R-(X-Y)n and X-Y-X where X in a polyalkylene oxide (e.g., poly(ethylene glycol, poly(propylene glycol) and block copolymers of poly(ethylene oxide) and polyφropylene oxide) (e.g., PLURONIC and PLURONIC R series of polymers from BASF Corporation, Mount Olive, NJ) and Y is a biodegradable polyester, where the polyester may comprise the residues of one or more of the monomers selected from lactide, lactic acid, glycolide, glycolic acid, e-caprolactone, gamma-caprolactone, hydroxyvaleric acid, hydroxybutyric acid, beta-butyrolactone, gamma- butyrolactone, gamma-valerolactone, γ-decanolactone, δ-decanolactone, trimethylene carbonate, 1 ,4-dioxane-2-one or 1 ,5-dioxepan-2one (e.g., PLG- PEG-PLG) and R is a multifunctional initiator, may be used.
Examples of fibrosis-inhibiting agents for use in the treatment of inflammatory arthritis include the following: ZD-6474, AP-23573, synthadotin, S-0885, aplidine, ixabepilone, IDN-5390, SB-2723005, ABT- 518, combretastatin, anecortave acetate, SB-715992, temsirolimus, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, as well as analogues and derivatives of the aforementioned.
Additional examples of anti-scarring agents which can be used include those having a high potency in the assays described herein (1- 10OnM IC50 range) such as isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, dolastatin 15, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17- DMAG, and tacrolimus; those having a mid-potency in the assays described herein (100-500 nM IC50 range) such as loteprednol etabonate, juglone, prednisolone, puromycin, 3-BAABE, cladribine, and mannose-6-phosphate; and those having a low potency in the assays described herein (500- 1000nm range IC50 range) such as 5-azacytidine, Ly333531(ruboxistaurin), and simvastatin.
The drug dose administered from the present compositions for the treatment of inflammatory arthritis will depend on a variety of factors, including the type of formulation and treatment site. However, certain principles can be applied in the application of this art. Drug dose can be calculated as a function of dose per unit area (of the treatment site), total drug dose administered can be measured and appropriate surface concentrations of active drug can be determined. For local application, drugs are to be used at concentrations that range from several times more than to 50%, 20%, 10%, 5%, or even less than 1% of the concentration typically used in a single systemic dose application. In certain aspects, the anti-scarring agent is released from the composition in effective concentrations in a time period that may be measured from the time of infiltration into tissue adjacent to the device, which ranges from about less than 1 day to about 180 days. Generally, the release time may also be from about less than 1 day to about 180 days; from about 7 days to about 14 days; from about 14 days to about 28 days; from about 28 days to about 56 days; from about 56 days to about 90 days; from about 90 days to about 180 days. In one aspect, the drug is released in effective concentrations for a period ranging from 1 - 90 days.
The exemplary anti-fibrosing agents, used alone or in combination, should be administered under the following dosing guidelines. The total amount (dose) of anti-scarring agent in the composition can be in the range of about 0.01 μg-10 μg, or 10 μg-10 mg, or 10 mg-250 mg, or 250 mg-1000 mg, or 1000 mg-2500 mg. The dose (amount) of anti-scarring agent per unit area of surface to which the agent is applied may be in the range of about 0.01 μg/mm2 - 1 μg/mm2, or 1 μg/mm2 - 10 μg/mm2, or 10 μg/mm2 - 250 μg/mm2, 250 μg/mm2 - 1000 μg/mm2, or 1000 μg/mm2 - 2500 μg/mm2.
Provided below are exemplary dosage ranges for various anti- scarring agents that can be used in conjunction with compositions for the treatment of inflammatory arthritis in accordance with the invention. The following dosages are particularly useful for intra-articular administration: (A) Angiogenesis inhibitors including Alphastatin, ZD-6474, IDN-5390, SB- 2723005, ABT-518, combretastatin, and anecortane, analogues and derivatives thereof: total single dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit volume of 0.01 μg - 100 mg per ml_; preferred dose of 0.1 μg/mL - 30 mg/mL (B) mTOR inhibitors including AP-23573 and Temsirolimus, analogues and derivatives thereof: total single dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit volume of 0.01 μg - 100 mg per ml_; preferred dose of 0.1 μg/ ml_ - 20 mg/ ml_. (C) Tubulin antagonists including synthadotin, analogues and derivatives thereof: total single dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit volume of 0.01 μg - 100 μg per mL; preferred dose of 0.1 μg/ mL - 20 mg/ mL. (D) Epithilones including ixabepilone and analogues and derivatives thereof: total single dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit volume of 0.01 μg - 100 mg per mL; preferred dose of 0.1 μg/mL - 20 mg/ mL. (E) Kinesin antagonists including SB-715992 and analogues and derivatives thereof: tota single dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit volume of 0.01 μg - 100 mg per mL; preferred dose of 0.1 μg/ mL - 20 mg/ mL. (F) TNF alpha antagonists including Etanercept, humicade, adalimumab and analogues and derivatives thereof: total single dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit volume of 0.01 μg - 100 mg per mL; preferred dose of 0.1 μg/ ml_ - 20 mg/ mL (G) AKT inhibitor including erucylphosphocholine and analogues and derivatives thereof: total single dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit volume of 0.01 μg - 100 mg per mL; preferred dose of 0.1 μg/ mL - 20 mg/ mL. (H) FGF Inhibitors including IDN-5390 and analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit volume of 0.01 μg - 100 mg per mL; preferred dose of 0.1 μg/ mL - 20 mg/ mL. (I) Collagenase Antagonists including S-0885 and analogues and derivatives thereof: total dose not to exceed 1000 mg (range of 0.1 μg to 1000 mg); preferred 1 μg to 500 mg. Dose per unit volume of 0.01 μg - 300 mg per mL; preferred dose of 0.1 μg/ mL - 100 mg/ mL (J) NF KAPPA B Inhibitors including Bortezomib and analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit volume of 0.01 μg - 100 mg per mL; preferred dose of 0.1 μg/ mL - 20 mg/ mL. (K) Elongation factor-1 alpha inhibitors including aplidine and analogues and derivatives thereof: total single dose not to exceed 1000 mg (range of 0.1 μg to 1000 mg); preferred 1 μg to 500 mg. Dose per unit volume of 0.01 μg - 100 mg per mL; preferred dose of 0.1 μg/ mL - 100 mg/ mL. (L) Tyrosine kinase inhibitors including Gefitinib and analogues and derivatives thereof: total dose not to exceed 1000 mg (range of 0.1 μg to 1000 mg); preferred 1 μg to 500 mg. Dose per unit volume of 0.01 μg - 300 mg per mL; preferred dose of 0.1 μg/ mL - 100-μg/ mL. In another aspect, systemic treatment may be administered when severe exacerbations or systemic disease (e.g., RA) are present. Anti- scarring agents that are delivered systemically should be dosed according to the level of drug required to inhibit the pathologies of inflammatory arthritis as described above. These systemic doses may vary according to patient, severity of disease, formulation of the administered agent, potency and/or tolerability of the agent, and route of administration. For example, for ZD-6474, AP-23573, synthadotin, S-0885, aplidine, ixabepilone, 1DN-5390, SB-2723005, ABT-518, combretastatin, anecortave acetate, SB-715992, temsirolimus, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, doiastatin 15, cerivastatin, jaspiakinolide, herbimycin A, pirfenidone, vinorelbine, 17- DMAG, tacrolimus, loteprednol etabonate, juglone, prednisolone, puromycin, 3-BAABE, cladribine, mannose-6-phosphate, 5-azacytidine, Ly333531 (ruboxistaurin), simvastatin, and an analogue or derivatives of the aforementioned, preferred embodiments would be 10 to 175 mg/m2 once every 1 to 4 weeks, 10 to 75 mg/m2 daily, as tolerated, or 10 to 175 mg/m2 weekly, as tolerated or until symptoms subside. To treat severe acute exacerbations, higher doses of 50 to 250 mg/m2 of ZD-6474, AP-23573, synthadotin, S-0885, aplidine, ixabepilone, IDN-5390, SB-2723005, ABT- 518, combretastatin, anecortave acetate, SB-715992, temsirolimus, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, doiastatin 15, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17-DMAG, tacrolimus, loteprednol etabonate, juglone, prednisolone, puromycin, 3-BAABE, cladribine, mannose-6-phosphate, 5-azacytidine, Ly333531 (ruboxistaurin), or simvastatin, may be administered as a "pulse" systemic therapy. Other anti-scarring agents can be administered at equivalent doses adjusted for the potency and tolerability of the agent.
For specific high potency drugs, the total single dose typically should not exceed 200 mg (range of 0.1 μg to 200 mg) and preferably 1 μg to 100 mg; dose per volume of 0.01 μg - 100 mg per ml_; preferably 0.1 μg/mL - 20 mg/mL For mid-potency drugs, the total single dose typically should not to exceed 500 mg (range of 1.0 μg to 500 mg) and preferably 1 μg to 200 mg; dose per volume of 0.01 μg - 200 mg per ml_, preferably 0.1 μg/mL - 40 mg/mL. For low potency drugs, the total dose typically should not exceed 1000 mg (range of 0.1 μg to 1000 mg), preferably 1 μg to 500 mg; dose per unit volume of 0.01 μg - 300 mg per mL; preferably 0.1 μg/mm2 - 100 mg/mL
According to another aspect, any anti-infective agent described above may be used in conjunction with compositions for the treatment of inflammatory arthritis. Exemplary anti-infective agents include (A) anthracyclines (e.g., doxorubicin and mitoxantrone), (B) fluoropyrimidines (e.g., 5-FU), (C) folic acid antagonists (e.g., methotrexate), (D) podophylotoxins (e.g., etoposide), (E) camptothecins, (F) hydroxyureas, and (G) platinum complexes (e.g., cisplatin), as well as analogues and derivatives of the aforementioned.
The drug dose administered from the present compositions for prevention or inhibition of infection in accordance with the present invention will depend on a variety of factors, including the type of formulation, the location of the treatment site, and the type of condition being treated. However, certain principles can be applied in the application of this art. Drug dose can be calculated as a function of dose per unit area (of the treatment site), total drug dose administered can be measured and appropriate surface concentrations of active drug can be determined. Drugs are to be used at concentrations that range from several times more than to 50%, 20%, 10%, 5%, or even less than 1% of the concentration typically used in a single anti-infective systemic dose application. In certain aspects, the anti-infective agent is released from the composition in effective concentrations in a time period that may be measured from the time of infiltration into tissue adjacent to the device, which ranges from about less than 1 day to about 180 days. Generally, the release time may also be from about less than 1 day to about 180 days; from about 7 days to about 14 days; from about 14 days to about 28 days; from about 28 days to about 56 days; from about 56 days to about 90 days; from about 90 days to about 180 days. The exemplary anti-infective agents, used alone or in combination, should be administered under the following dosing guidelines. The total amount (dose) of anti-infective agent in the composition can be in the range of about 0.01 μg-1 μg, or about 1 μg-10 μg, or about 10 μg-1 mg, or about 1 mg to 10 mg, or about 10 mg-100 mg, or about 100 mg to 250 mg, or about 250 mg-1000 mg. The dose (amount) of anti-infective agent per unit area of device or tissue surface to which the agent is applied may be in the range of about 0.01 μg/mm2 - 1 μg/mm2, or about 1 μg/mm2 - 10 μg/mm2, or about 10 μg/mm2 - 100 μg/mm2, or about 100 μg/mm2 to 250 μg/mm2, or about 250 μg/mm2 - 1000 μg/mm2. As different compositions will release the anti-infective agent at differing rates, the above dosing parameters should be utilized in combination with the release rate of the drug from the composition such that a minimum concentration of about 10"8 M to 10"7 M, or about 10"7 M to 10"6 M about 10"6 M to 10"5 M or about 10'5 M to 10"4 M of the agent is maintained on the tissue surface.
(c) Prevention of Cartilage Loss ("Chondroprotection")
In another aspect, polymeric compositions can be used to prevent or reduce the loss of cartilage loss following an injury (e.g., cruciate ligament tear and/or meniscal tear). It has been known for a long time that damage to a joint can predispose a patient to develop osteoarthritis in the joint at a subsequent point in time, but there has been no effective treatment to prevent this occurrence. Instead most of the focus from the medical community and researchers has been on the treatment of the arthritis after it has become established. Treatments for established disease include antiinflammatory drugs (non-steroidal and steroidal), lubricants or synovial fluid replacements, surgery and joint replacement for severe disease.
Trauma to a joint can take many forms, ranging from a simple sprain which can heal spontaneously to a fracture that creates so many bone fragments that it is almost impossible to reconstruct the joint. The focus for treatment of these injuries revolves around restoring the joint to its normal anatomical state and to resume regular motion. Risk factors for developing arthritis are related to the extent of trauma, the extent of the joint disruption, the degree of the fracture or dislocations, whether or not it is a weight bearing joint, and the characteristic of the joint itself. In general, the greater the trauma to the joint, the greater the risk that the patient will develop osteoarthritis later in life. Surgical correction of a joint to its pre- injury anatomy does not guarantee the prevention of arthritis. In the case of an intra-articular fracture, for example a plateau fracture of the tibia, the treatment is to surgically reconstruct the joint so that it reverts back to a congruent, smooth and intact joint surface with no "step defects" or pieces out of place that would interfere with the gliding of the femur on its surface. Despite improved surgical techniques in repairing these fractures, patients with such fractures have a very high probability of developing degenerative arthritis later on in life.
Anterior cruciate ligament (ACL) injuries in the knee represent a classic example of an injury that predisposes patients to potentially severe degenerative arthritis. The ACL is the ligament that joins the anterior tibial plateau to the posterior femoral intercondylar notch. It is composed of multiple non-parallel fibers with variable fiber lengths that function in bundles to provide tension and mechanical stability to the knee throughout its range of motion. The ACL's stabilizing role has four main functions, including (a) restraining anterior translation of the tibia; (b) preventing hyperextension of the knee; (c) acting as a secondary stabilizer to the valgus stress, reinforcing the medial collateral ligament; and (d) controlling rotation of the tibia on the femur during femoral extensions, and thus, controlling movements such as side-stepping and pivoting. Generally, ACL deficiency results in subluxation of the tibia on the femur causing stretching of the enveloping capsular ligaments and abnormal shear forces on the menisci and on the articular cartilage. Delay in diagnosis and treatment gives rise to increased intra-articular damage as well as stretching of the secondary stabilizing capsular structures. Despite the known high risk for developing osteoarthritis, patients generally have no associated fractures and have normal x-rays at the time of presentation post-ACL injury. Yet it is well documented that anyone who suffers an ACL injury has a high probability of developing arthritis: 50% by 10 years and 80% by 20 years post-injury. Generally after an ACL rupture patients suffer from instability since the ligament is critical in stabilizing the joint during pivoting and rotation. For example, it is not only required for demanding pivoting sports such as basketball, it is also required for daily activity such as a mother holding her baby as she pivots to get an item from the fridge.
The typical treatment and management of an ACL tear is reconstruction using a graft to replace the torn ACL. The graft may be taken from elsewhere in the patient's extremity (autograft), harvested from a cadaver (allograft) or may be made from a synthetic material. Autograft is the most widely performed orthopedic ACL reconstruction. The technique involves harvesting the patient's own tissue, which may be the mid-third of the patellar tendon with bone attached at both ends, one or two medial hamstrings, or the quadriceps tendon with bone at one end. Synthetic materials have the advantage of being readily available, however, there is a higher failure rate of synthetic grafts compared to autografts and allografts and they have mechanical properties that do not closely resemble the normal ligament. Successful ACL reconstruction is dependent on a number of factors, including surgical technique, post-operative rehabilitation and associated secondary ligament instability. During the surgical procedure, arthroscopy is used to determine whether there are any other associated injuries, which may be treated at the same time, such as meniscal tears or chondral trauma. The surgical procedure is done through a small accessory incision, whereby a tunnel is drilled through the tibia and femur so that the graft may be inserted and fixed.
Surgical reconstruction was initially thought to provide a permanent solution: re-establish a stable knee and prevent degeneration. But other studies demonstrated that after joint injury, there is a cascade of inflammatory activity that once initiated, can be destructive to the joint. This explains why surgical repair itself would have not impact on the prevention of degeneration in traumatized joints; stabilizing a joint or the macro reconstruction of a joint does not address the fundamental underlying biology. Unfortunately, although long-term data has shown that surgery is indeed successful in stabilizing the knee and getting people back to normal activity; it has no impact on the subsequent rate of development of osteoarthritis. As a result, the standard of care to day is to repair the joint acutely and treat the arthritis when it ultimately develops. It should be noted that all joints (in addition to knees) have the potential to become arthritic after trauma, but joints typically involved include; fingers, thumbs, metacarpal (wrist), elbow, shoulder, spine joints (facets, sacro-iliac), temperomandibular, otic bones, hips, ankles, tarsal and toes, especially the hallux.
Fibrosis-inhibiting agents such as ZD-6474,J\P-23573, synthadotin, S-0885, aplidine, ixabepilone, IDN-5390, SB-2723005, ABT- 518, combretastatin, anecortave acetate, SB-715992, temsirolimus, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, dolastatin 15, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17-DMAG, tacrolimus, loteprednol etabonate, juglone, prednisolone, puromycin, 3-BAABE, cladribine, mannose-6-phosphate, 5-azacytidine, Ly333531 (ruboxistaurin), and simvastatinmay demonstrate in animal experiments an ability to prevent cartilage breakdown following cruciate ligament tears. This effect has been seen with antifibrotic agents both in an inflammatory model and biomechanical model of joint injury. Hartley Guinea pigs subjected to surgical transaction of the anterior cruciate ligament represent a mechanical model for arthritis. Typically after the anterior cruciate is severed, the animals develop arthritis within several weeks. The introduction of the fibrosis-inhibiting agents such as ZD-6474, AP-23573, synthadotin, S-0885, aplidine, ixabepilone, IDN-5390, SB-2723005, ABT-518, combretastatin, anecortave acetate, SB-715992, temsirolimus, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, dolastatin 15, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17-DMAG, tacrolimus, loteprednol etabonate, juglone, prednisolone, puromycin, 3-BAABE, cladribine, mannose-6-phosphate, 5-azacytidine, Ly333531 (ruboxistaurin), and simvastatin, into the joint may greatly retard the arthritic process and protect not only the cartilage, but also the underlying bone, from breakdown.
The present invention addresses a significant unmet medical need: the prevention of progressive joint degeneration after traumatic injury. Introduction of a composition containing a fibrosis-inhibiting agent into a damaged joint shortly after injury, (e.g., through intra-articular injection, peri- articular administration, via arthroscope, as a joint lavage during open surgical procedures) will impact the cascade of events that lead to joint destruction, such as inhibiting inflammation and preventing cartilage matrix destruction. Most ligament injuries are severe enough or painful enough that patients seek immediate medical attention (within the first 24 to 48 hours); long before irreversible changes have occurred in the joint. If at the time of initial presentation to a health care professional, an intra-articular injection of a fibrosis-inhibitor can be administered into the joint to stop or slow down the destructive activity (in the joint and the tissues surrounding the joint), the articular cartilage can be protected from breakdown. Early introduction of the agents of the present invention intervention will slow, decrease or eliminate the cascade of events that lead to osteoarthritis. The invention can be administered immediately after injury, repeated during the period leading up to stabilization surgery, and/or can be administered after surgery is completed. Thus, within one aspect of the present invention, methods are provided for treating or preventing cartilage loss, comprising the step of administering to a patient in need thereof a therapeutically effective amount of an anti-scarring agent or a composition comprising an anti-scarring agent.
An effective anti-scarring therapy for cartilage loss will accomplish one or more of the following: (i) decrease the severity of symptoms (pain, swelling and tenderness of affected joints; (ii) decrease the severity of clinical signs of the disease (thickening of the joint capsule, synovial hypertrophy, joint effusion, soft tissue contractures, decreased range of motion, ankylosis and fixed joint deformity); (iii) increase the frequency and duration of disease remission/symptom-free periods; (iv) delay or prevent the onset of clinically significant arthritis in a joint that has previously been injured; and/or (v) prevent or reduce fixed impairment and disability.
According to the present invention, any anti-scarring agent described above could be utilized in the practice of this invention. Within, certain embodiments of the invention, the composition may release an agent that inhibits one or more of the general components of the process of fibrosis (or scarring) associated with joint damage, including: (a) formation of new blood vessels (angiogenesis), (b) migration and/or proliferation of connective tissue cells (such as fibroblasts or synoviocytes), (c) deposition and remodeling of extracellular matrix (ECM) by matrix metalloproteinase activity, (d) inflammatory response by cytokines (such as IL-1 , TNFα, FGF, VEGF). By inhibiting one or more of the components of fibrosis (or scarring), joint damage and osteoarthritis development may be reduced or prevented in a previously injured joint..
In one aspect, the composition includes an anti-scarring agent and a polymeric carrier suitable for application to treat an injured joint. Numerous polymeric and non-polymeric delivery systems and compositions containing an anti-scarring agent for use in the prevention of cartilage loss have been described above. An anti-scarring agent may be administered systemically (orally, intravenously, or by intramuscular or subcutaneous injection) in the minimum dose to achieve the above mentioned results. For patients with only a small number of joints affected, or with disease more prominent in a limited number of joints, the anti-scarring agent can be applied onto tissue within a joint or directly injected into the affected joint (intraarticular injection).
The anti-scarring agent can be administered in any manner described herein. However, preferred methods of administration include intravenous, oral, or subcutaneous, intramuscular or intra-articular injection. The anti-scarring agent can be directly injected into the affected joint (intra- articular injection) via percutaneous needle insertion into the joint capsule, or as part of an arthroscopic procedure performed on the joint. In a preferred embodiment, the intra-articular formulation containing a fibrosis- inhibitor is administered to a joint following an injury with a high probability of inducing subsequent arthritis (e.g., cruciate ligament tears in the knee, meniscal tears in the knee). The fibrosis-inhibiting agent is administered for a period sufficient (either through sustained release preparations and/or repeated injections) to protect the cartilage from breakdown as a result of the injury (or the surgical procedure used to treat it). The anti-scarring agent can be administered as a chronic low dose therapy to prevent disease progression, prolong disease remission, or decrease symptoms in active disease. Alternatively, the therapeutic agent can be administered in higher doses as a "pulse" therapy to induce remission in acutely active disease (such as in the period immediately following a joint injury). The minimum dose capable of achieving these endpoints can be used and can vary according to patient, severity of disease, formulation of the administered agent, clearance from the joint, potency and/or tolerability of the agent, and route of administration.
A variety of commercially available HA compositions for intra- articular injection may be combined with one or more agents according to the present invention including: SYNVISC (Biomatrix, Inc., Ridgefield, NJ) - an elastoviscous fluid containing hylan (a derivative of sodium hyaluronate (hyaluronan)) polymers derived from rooster combs, HYALGAN (Sanofi- Synthelabo Inc. New York, NY)1 and ORTHOVISC (Ortho Biotech Products, Bridgewater, NJ) - a highly purified, high molecular weight, high viscosity injectable form of HA intended to relieve pain and to improve joint mobility and range of motion in patients suffering from osteoarthritis (OA) of the knee. ORTHOVISC is injected into the knee to restore the elasticity and viscosity of the synovial fluid. HYVISC is a high molecular weight, injectable HA product developed by Anika Therapeutics (Woburn, MA) currently being used to treat osteoarthritis and lameness in racehorses. Other HA-based viscosupplementation products for intra-articular injection include SUPARTZ from Seikagaku Corp. (Japan), SUPLASYN from Bioniche Life Sciences, Inc. (Canada), ARTHREASE from DePuy Orthopaedics, Inc. (Warsaw, IN), and DUROLANE from Q-Med AB (Sweden). By adding a fibrosis-inhibiting agent to the HA, the intra-articular injection has the added benefit of helping to prevent cartilage breakdown (i.e., it is "chondroprotective").
In one aspect, the compositions of the present invention may be used for the management of osteoarthritis in animals (e.g., horses). It should be noted that some HA products (notably HYVISC by Boehringer lngelheim Vetmedica, St. Joseph, MO) are used in veterinary applications (typically in horses to treat osteoarthritis and lameness).
Fibrosis-inhibiting formulations that can be used for the treatment or prevention of cartilage loss may be in the form of solutions, topical formulations (e.g., solution, cream, ointment, gel) emulsions, micellar solutions, gels (crosslinked and non-crosslinked), suspensions and/or pastes. One form for the formulation is as an injectable composition for intra-articular or arthroscopic delivery. For compositions that further contain a polymer to increase the viscosity of the formulation, hyaluronic acid (crosslinked, derivatized and/or non-crosslinked) is an exemplary material. These formulations can further comprise additional polymers (e.g., collagen, poly(ethylene glycol) or dextran) as well as biocompatible solvents (e.g., ethanol, DMSO, or NMP). In one embodiment, the fibrosis-inhibiting therapeutic agent can be incorporated directly into the formulation. In another embodiment, the fibrosis-inhibiting therapeutic agent can be incorporated into a secondary carrier {e.g., micelles, liposomes, emulsions, microspheres, nanospheres etc, as described above). The microsphere and nanospheres may be comprised of degradable polymers. Degradable polymers that can be used include poly(hydroxyl esters) (e.g., PLGA, PLA, PCL, and the like), as well as polyanhydrides, polyorthoesters and polysaccharides (e.g., chitosan and alginates).
In one embodiment, the fibrosis-inhibiting agent further comprises a polymer where the polymer is a degradable polymer. The degradable polymers may include polyesters where the polyester may comprise the residues of one or more of the monomers selected from lactide, lactic acid, glycolide, glycolic acid, e-caprolactone, gamma- caprolactone, hydroxyvaleric acid, hydroxybutyric acid, beta-butyrolactone, garηma-butyrolactone, gamma-valerolactone, γ-decanolactone, δ- - decanolactone, trimethylene carbonate, 1 ,4-dioxane-2-one or 1,5-dioxepan- 2one, and block copolymers of the form X-Y, Y-X-Y, R-(Y-X)n, R-(X-Y)n and X-Y-X where X in a polyalkylene oxide (e.g., poly(ethylene glycol, poly(propylene glycol) and block copolymers of poly(ethylene oxide) and polypropylene oxide) (e.g., PLURONIC and PLURONIC R series of polymers from BASF Corporation, Mount Olive, NJ) and Y is a biodegradable polyester, where the polyester may comprise the residues of one or more of the monomers selected from lactide, lactic acid, glycolide, glycolic acid, e-caprolactone, gamma-caprolactone, hydroxyvaleric acid, hydroxybutyric acid, beta-butyrolactone, gamma-butyrolactone, gamma- valerolactone, γ-decanolactone, δ-decanolactone, trimethylene carbonate, 1 ,4-dioxane-2-one or 1,5-dioxepan-2one (e.g., PLG-PEG-PLG) and R is a multifunctional initiator. In another embodiment, the fibrosis-inhibiting agent/composition may further comprise a solvent, a liquid oligomer or liquid polymer such that the final composition may be passed through a 18G needle. The reagents that may be used include ethanol, NMP, PEG 200, PEG 300 and low molecular weight liquid polymers of the form X-Y, Y-X-Y, R-(Y-X)n, R-(X-Y)n and X-Y-X where X in a polyalkylene oxide (e.g., poly(ethylene glycol, polypropylene glycol) and block copolymers of poly(ethylene oxide) and polyφropylene oxide) (e.g., PLURONIC and PLURONIC R series of polymers from BASF Corporation, Mount Olive, NJ) and Y is a biodegradable polyester, where the polyester may comprise the residues of one or more of the monomers selected from lactide, lactic acid, glycolide, glycolic acid, e-caprolactone, gamma-caprolactone, hydroxy valeric acid, hydroxybutyric acid, beta-butyrolactone, gamma- butyrolactone, gamma-valerolactone, γ-decanolactone, δ-decanolactone, trimethylene carbonate, 1 ,4-dioxane-2-one or 1,5-dioxepan-2one (e.g., PLG- PEG-PLG) and R is a multifunctional initiator.
In another embodiment, the fibrosis-inhibiting agent may be in the form of a solution or suspension in an organic solvent, a liquid oligomer or a liquid polymer. In this embodiment, reagents such as ethanol, NMP, PEG 200, PEG 300 and low molecular weight liquid polymers of the form X- Y, Y-X-Y, R-(Y-X)n, R-(X-Y)n and X-Y-X where X in a polyalkylene oxide (e.g., poly(ethylene glycol, poly(propylene glycol) and block copolymers of poly(ethylene oxide) and poly(propylene oxide) (e.g., PLURONIC and PLURONIC R series of polymers from BASF Corporation, Mount Olive, NJ) and Y is a biodegradable polyester, where the polyester may comprise the residues of one or more of the monomers selected from lactide, lactic acid, glycolide, glycolic acid, e-caprolactone, gamma-caprolactone, hydroxyvaleric acid, hydroxybutyric acid, beta-butyrolactone, gamma- butyrolactone, gamma-valerolactone, γ-decanolactone, δ-decanolactone, trimethylene carbonate, 1 ,4-dioxane-2-one or 1 ,5-dioxepan-2one (e.g., PLG- PEG-PLG) and R is a multifunctional initiator, may be used.
Examples of fibrosis-inhibiting agents for use in the treatment of, or prevention of, cartilage loss following traumatic injury include the following: ZD-6474, AP-23573, synthadotin, S-0885, aplidine, ixabepilone, IDN-5390, SB-2723005, ABT-518, combretastatin, anecortave acetate, SB- 715992, temsirolimus, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, dolastatin 15, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17- DMAG, tacrolimus, loteprednol etabonate, juglone, prednisolone, puromycin, 3-BAABE, cladribine, mannose-6-phosphate, 5-azacytidine, Ly333531 (ruboxistaurin), simvastatin, as well as analogues and derivatives of the aforementioned.
Additional examples of anti-scarring agents which can be used include those having a high potency in selected assays described herein (approximately 1-10OnM IC50 range) such as isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, dolastatin 15, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17-DMAG, and tacrolimus; those having a mid- potency in selected assays described herein (approximately 100-500 nM IC50 range) such as loteprednol etabonate, juglone, prednisolone, puromycin, 3-BAABE, cladribine, and mannose-6-phosphate; and those having a low potency in selected assays described herein (approximately 500-1 OOOnm range IC50 range) such as 5-azacytidine, Ly333531 (ruboxistaurin), and simvastatin.
The drug dose administered from the present compositions for the treatment of cartilage loss will depend on a variety of factors, including the type of formulation and treatment site. However, certain principles can be applied in the application of this art. Drug dose can be calculated as a function of dose per unit area (of the treatment site), total drug dose administered can be measured and appropriate surface concentrations of active drug can be determined. For local application (such as intra-articular or endoscopic administration), drugs are to be used at concentrations that range from several times more than to 50%, 20%, 10%, 5%, or even less than 1 % of the concentration typically used in a single systemic dose application. In certain aspects, the anti-scarring agent is released from the composition in effective concentrations in a time period that may be measured from the time of infiltration into tissue adjacent to the device, which ranges from about less than 1 day to about 180 days. Generally, the release time may also be from about less than 1 day to about 180 days; from about 7 days to about 14 days; from about 14 days to about 28 days; from about 28 days to about 56 days; from about 56 days to about 90 days; from about 90 days to about 180 days. In one aspect, the drug is released in effective concentrations for a period ranging from 1 - 90 days.
The exemplary anti-fibrosing agents, used alone or in combination, should be administered under the following dosing guidelines. The total amount (dose) of anti-scarring agent in the composition can be in the range of about 0.01 μg-10 μg, or 10 μg-10 mg, or 10 mg-250 mg, or 250 mg-1000 mg, or 1000 mg-2500 mg.
Provided below are exemplary dosage ranges for various anti- scarring agents that can be used in conjunction with compositions for the treatment of inflammatory arthritis in accordance with the invention. The following dosages are particularly useful for intra-articular administration: (A) Angiogenesis inhibitors including Alphastatin, ZD-6474, IDN-5390, SB- 2723005, ABT-518, combretastatin, and anecortane, analogues and derivatives thereof: total single dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit volume of 0.01 μg - 100 mg per mL; preferred dose of 0.1 μg/mL - 30 mg/mL (B) mTOR inhibitors including AP-23573 and Temsirolimus, analogues and derivatives thereof: total single dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit volume of 0.01 μg - 100 mg per mL; preferred dose of 0.1 μg/ mL - 20 mg/ mL. (C) Tubulin antagonists including synthadotin, analogues and derivatives thereof: total single dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit volume of 0.01 μg - 100 μg per mL; preferred dose of 0.1 μg/ mL - 20 mg/ mL. (D) Epithilones including ixabepilone and analogues and derivatives thereof: total single dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit volume of 0.01 μg - 100 mg per ml_; preferred dose of 0.1 μg/mL - 20 mg/ mL (E) Kinesin antagonists including SB-715992 and analogues and derivatives thereof: tota single dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit volume of 0.01 μg - 100 mg per mL; preferred dose of 0.1 μg/ mL - 20 mg/ mL. (F) TNF alpha antagonists including Etanercept, humicade, adalimumab and analogues and derivatives thereof: total single dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit volume of 0.01 μg - 100 mg per mL; preferred dose of 0.1 μg/ mL - 20 mg/ mL. (G) AKT inhibitor including erucylphosphocholine and analogues and derivatives thereof: total single dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit volume of 0.01 μg - 100 mg per mL; preferred dose of 0.1 μg/ mL - 20 mg/ mL. (H) FGF Inhibitors including IDN-5390 and analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit volume of 0.01 μg - 100 mg per mL; preferred dose of 0.1 μg/ mL - 20 mg/ mL. (I) Collagenase Antagonists including S-0885 and analogues and derivatives thereof: total dose not to exceed 1000 mg (range of 0.1 μg to 1000 mg); preferred 1 μg to 500 mg. Dose per unit volume of 0.01 μg - 300 mg per mL; preferred dose of 0.1 μg/ mL - 100 mg/ mL (J) NF KAPPA B Inhibitors including Bortezomib and analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit volume of 0.01 μg - 100 mg per mL; preferred dose of 0.1 μg/ mL - 20 mg/ mL. (K) Elongation factor-1 alpha inhibitors including aplidine and analogues and derivatives thereof: total single dose not to exceed 1000 mg (range of 0.1 μg to 1000 mg); preferred 1 μg to 500 mg. Dose per unit volume of 0.01 μg - 100 mg per mL; preferred dose of 0.1 μg/ mL - 100 mg/ mL. (L) Tyrosine kinase inhibitors including Gefitinib and analogues and derivatives thereof: total dose not to exceed 1000 mg (range of 0.1 μg to 1000 mg); preferred 1 μg to 500 mg. Dose per unit volume of 0.01 μg - 300 mg per mL; preferred dose of 0.1 μg/ ml_ - 100 μg/ mL In another aspect, systemic treatment may be administered when severe exacerbations or systemic disease (e.g., RA) are present. Anti- scarring agents that are delivered systemically should be dosed according to the level of drug required to inhibit the pathologies of inflammatory arthritis as described above. These systemic doses may vary according to patient, severity of disease, formulation of the administered agent, potency and/or tolerability of the agent, and route of administration. For example, for ZD-6474, AP-23573, synthadotin, S-0885, aplidine, ixabepilone, lDN-5390, SB-2723005, ABT-518, combretastatin, anecortave acetate, SB-715992, temsirolimus, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, dolastatin 15, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17- DMAG, tacrolimus, loteprednol etabonate, juglone, prednisolone, . - . puromycin, 3-BAABE, cladribine, mannose-6-phosphate, 5-azacytidine, Ly333531 (ruboxistaurin), simvastatin, and an analogue or derivatives of the aforementioned, preferred embodiments would be 10 to 175 mg/m2 once every 1 to 4 weeks, 10 to 75 mg/m2 daily, as tolerated, or 10 to 175 mg/m2 weekly, as tolerated or until symptoms subside. To treat severe acute exacerbations, higher doses of 50 to 250 mg/m2 of ZD-6474, AP-23573, synthadotin, S-0885, aplidine, ixabepilone, IDN-5390, SB-2723005, ABT- 518, combretastatin, anecortave acetate, SB-715992, temsirolimus, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, dolastatin 15, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17-DMAG, tacrolimus, loteprednol etabonate, juglone, prednisolone, puromycin, 3-BAABE, cladribine, mannose-6-phosphate, 5-azacytidine, Ly333531 (ruboxistaurin), simvastatin, may be administered as a "pulse" systemic therapy. Other anti-scarring agents can be administered at equivalent doses adjusted for the potency and tolerability of the agent.
For specific high potency drugs, the total single dose typically should not exceed 200 mg (range of 0.1 μg to 200 mg) and preferably 1 μg to 100 mg; dose per volume of 0.01 μg - 100 mg per mL; preferably 0.1 μg/mL - 20 mg/mL For mid-potency drugs, the total single dose typically should not to exceed 500 mg (range of 1.0 μg to 500 mg) and preferably 1 μg to 200 mg; dose per volume of 0.01 μg - 200 mg per mL, preferably 0.1 μg/mL - 40 mg/mL. For low potency drugs, the total dose typically should not exceed 1000 mg (range of 0.1 μg to 1000 mg), preferably 1 μg to 500 mg; dose per unit volume of 0.01 μg - 300 mg per mL; preferably 0.1 μg/mm2 - 100 mg/mL.
According to another aspect, any anti-infective agent described above may be used in conjunction with formulations for the treatment or prevention of cartilage loss. Exemplary antynfective agents - include (A) anthracyclines (e.g., doxorubicin and mitoxantrone), (B) fluoropyrimidines (e.g., 5-FU), (C) folic acid antagonists (e.g., methotrexate), (D) podophylotoxins (e.g., etoposide), (E) camptothecins, (F) hydroxyureas, and (G) platinum complexes (e.g., cisplatin), as well as analogues and derivatives of the aforementioned.
The drug dose administered from the present compositions for prevention or inhibition of infection in accordance with the present invention will depend on a variety of factors, including the type of formulation, the location of the treatment site, and the type of condition being treated. However, certain principles can be applied in the application of this art. Drug dose can be calculated as a function of dose per unit area (of the treatment site), total drug dose administered can be measured and appropriate surface concentrations of active drug can be determined. Drugs are to be used at concentrations that range from several times more than to 50%, 20%, 10%, 5%, or even less than 1% of the concentration typically used in a single anti-Infective systemic dose application. In certain aspects, the anti-infective agent is released from the composition in effective concentrations in a time period that may be measured from the time of infiltration into tissue adjacent to the device, which ranges from about less than 1 day to about 180 days. Generally, the release time may also be from about less than 1 day to about 180 days; from about 7 days to about 14 days; from about 14 days to about 28 days; from about 28 days to about 56 days; from about 56 days to about 90 days; from about 90 days to about 180 days.
The exemplary anti-infective agents, used alone or in combination, should be administered under the following dosing guidelines. The total amount (dose) of anti-infective agent in the composition can be in the range of about 0.01 μg-1 μg, or about 1 μg-10 μg, or about 10 μg-1 mg, or about 1 mg to 10 mg, or about 10 mg-100 mg, or about 100 mg to 250 mg, or about 250 mg-1000 mg. The dose (amount) of anti-infective agent per unit area of device or tissue surface to which the agent is applied may ' be in the range of about 0.01 μg/mm2 - 1 μg/mm2, or about 1 μg/mm2 - 10 μg/mm2, or about 10 μg/mm2 - 100 μg/mm2, or about 100 μg/mm2 to 250 μg/mm2, or about 250 μg/mm2 - 1000 μg/mm2. As different compositions will release the anti-infective agent at differing rates, the above dosing parameters should be utilized in combination with the release rate of the drug from the composition such that a minimum concentration of about 10"8 M to 10-7 M, or about 10 M to 10"6 M about 10"6 M to 10"5 M or about lO"5 M to 10"4 M of the agent is maintained on the tissue surface.
(d) Hypertrophic Sears/Keloids
In another aspect of the invention, compositions containing a therapeutically active agent (e.g., a fibrosis-inhibiting agent) and methods are provided for treating hypertrophic scars and keloids.
Hypertrophic scars and keloids are an overgrowth of dense fibrous tissue that is the result of an excessive fibroproliferative wound healing process. Hypertrophic scars and keloids usually develop after healing of a skin injury. Briefly, healing of wounds and scar formation occurs in three phases: inflammation, proliferation, and maturation. The first phase, inflammation, occurs in response to an injury which is severe enough to break the skin. During this phase, which lasts 3 to 4 days, blood and tissue fluid form an adhesive coagulum and fibrinous network which serves to bind the wound surfaces together. This is then followed by a proliferative phase in which there is ingrowth of capillaries and connective tissue from the wound edges, and closure of the skin defect. Finally, once capillary and fibroblastic proliferation has ceased, the maturation process begins wherein the scar contracts and becomes less cellular, less vascular, and appears flat and white. This final phase may take between 6 and 12 months.
If too much connective tissue is produced and the wound remains persistently cellular, the scar may become red and raised. If the scar remains within the boundaries of the original wound it is referred to as a hypertrophic scar, but if it extends beyond the original scar and into the - surrounding tissue, the lesion is referred to as a keloid. Hypertrophic scars and keloids are produced during the second and third phases of scar formation. Several wounds are particularly prone to excessive endothelial and fibroblastic proliferation, including burns, open wounds, and infected wounds. With hypertrophic scars, some degree of maturation occurs and gradual improvement occurs. In the case of keloids however, an actual tumor is produced which can become quite large. Spontaneous improvement in such cases rarely occurs.
Keloids and hypertrophic scars located at most sites are primarily of cosmetic concern; however, some keloids or hypertrophic scars can cause contractures, which may result in a loss of function if overlying a joint, or they can cause significant disfigurement if located on the face. Both keloids and hypertrophic scars can be painful or pruritic.
Within one embodiment of the present invention the compositions are directly injected into a hypertrophic scar or keloid, in order to prevent the progression of these lesions. The frequency of injections will depend upon the release kinetics of the polymer used, and the clinical response. This therapy is of particular value in the prophylactic treatment of conditions which are known to result in the development of hypertrophic scars and keloids (e.g., burns, the excision site of a keloid or hypertrophic scar, wounds on the chest and back of predisposed patients, etc.), and is preferably initiated prior to, or during the proliferative phase (from day 1 forward), but before hypertrophic scar or keloid development (i.e., within the first 3 months post-injury).
In one aspect, the present invention provides topical and injectable compositions that include an anti-scarring agent and a polymeric carrier suitable for application on or into hypertrophic scars or keloids. Numerous polymeric and non-polymeric delivery systems for use in treating hypertrophic scars or keloids have been described above.
Incorporation of a fibrosis-inhibiting agent into a topical formulation or an injectable formulation is one approach to treat this condition. The topical formulation can be in the form of a solution, a suspension, an emulsion, a gel, an ointment, a cream, film or mesh. The injectable formulation can be in the form of a solution, a suspension, an emulsion or a gel. Polymeric and non-polymeric components that can be used to prepare these topical or injectable compositions are described above.
In another embodiment, the fibrosis-inhibiting therapeutic agent can be incorporated into a secondary carrier (e.g., micelles, liposomes, emulsions, microspheres, nanospheres etc, as described above). Microsphere and nanospheres may include degradable polymers. Degradable polymers that can be used include poly(hydroxyl esters) (e.g., PLGA, PLA, PCL, and the like) as well as polyanhydrides, polyorthoesters and polysaccharides (e.g., chitosan and alginates).
In addition, a variety of other compositions and approaches for treating hypertrophic scars and keloids may be used in accordance with the invention. For example, treatment may include the administration of an effective amount of angiogenesis inhibitor (e.g., fumagillol, thalidomide) as a systemic or local treatment to decrease excessive scarring. See, e.g., U.S. Patent No. 6,638,949. The treatment may be a copolymer composed of a hydrophilic polymer, such as polyethylene glycol, that is bound to a polymer that adsorbs readily to the surfaces of body tissues, such as phenylboronic acid. See, e.g., U.S. Patent No. 6,596,267. The treatment may include a cryoprobe containing cryogen whereby it is positioned within the hypertrophic scar or keloid to freeze the tissue. See, e.g., U.S. Patent No. 6,503,246. The treatment may be a method of locally administering an amount of botulinum toxin in or in close proximity to the skin wound, such that the healing is enhanced. See, e.g., U.S. Patent No. 6,447,787. The treatment may be a liquid composition composed of a film-forming carrier such as a collodion which contains one or more active ingredients such as a topical steroid, silicone gel and vitamin E. See, e.g., U.S. Patent No. 6,337,076. The treatment may be a method of administering an antifibrotic amount of fluoroquinolone to prevent or treat scar tissue formation. See, e.g., U.S. Patent No. 6,060,474. The treatment may be a composition of an effective amount of calcium antagonist and protein synthesis inhibitor sufficient to cause matrix degradation at a scar site so as to control scar formation. See, e.g., U.S. Patent No. 5,902,609. The treatment may be a composition of non-biodegradable microspheres with a substantial surface charge in a pharmaceutically acceptable carrier. See, e.g., U.S. Patent No. 5,861 ,149. The treatment may be a composition of endothelial cell growth factor and heparin which may be administered topically or by intralesional injection. See, e.g., U.S. Patent No. 5,500,409.
Treatments and compositions for hypertrophic scars and keloids, which may be combined with one or more fibrosis-inhibiting agents according to the present invention, include commercially available products. Representative products include, for example, PROXIDERM External Tissue Expansion product for wound healing from Progressive Surgical Products (Westbury, NY)1 CICA-CARE Gel Sheet dressing product from Smith & Nephew Healthcare Ltd (India), and MEPIFORM Self-Adherent Silicone Dressing from Molnlycke Health Care (Eddystone, PA).
In one aspect, the present invention provides topical and injectable compositions that include an anti-scarring agent and a polymeric carrier suitable for application on or into hypertrophic scars or keloids or sites that are prone to forming hypertrophic scars or keloids.
Within one embodiment of the present invention either anti- scarring agents alone, or anti-scarring compositions as described above, are directly injected into a hypertrophic scar or keloid, in order to prevent the progression of these lesions. The frequency of injections will depend upon the release kinetics of the polymer used (if present), and the clinical response. This therapy is of particular value in the prophylactic treatment of conditions which are known to result in the development of hypertrophic scars and keloids (e.g., burns, the excision site of a keloid or hypertrophic scar, wounds on the chest and back of predisposed patients, etc.), and is preferably initiated prior to, or during the proliferative phase (from day 1 forward), but before hypertrophic scar or keloid development (i.e., within the first 3 months post-injury).
According to the present invention, any fibrosis-inhibiting agent described above could be utilized alone or in combination in the practice of this embodiment. Within one embodiment of the invention, compositions for treating hypertrophic scars or keloids may release an agent that inhibits one or more of the four general components of the process of fibrosis (or scarring), including: formation of new blood vessels (angiogenesis), migration and proliferation of connective tissue cells (such as fibroblasts or smooth muscle cells), deposition of extracellular matrix (ECM), and remodeling (maturation and organization of the fibrous tissue).
Examples of fibrosis-inhibiting agents for use in composition for treating hypertrophic scars and keloids include the following: ZD-6474, AP-23573, synthadotin, S-0885, aplidine, ixabepilone, IDN-5390, SB- 2723005, ABT-518, combretastatin, anecortave acetate, SB-715992, temsirolimus, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, as well as analogues and derivatives of the aforementioned.
Additional examples of anti-scarring agents which can be used include those having a high potency in the assays described herein (1- 10OnM IC5O range) such as isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, dolastatin 15, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17- DMAG, and tacrolimus; those having a mid-potency in the assays described herein (100-500 nM IC50 range) such as loteprednol etabonate, juglone, prednisolone, puromycin, 3-BAABE, cladribine, and mannose-6-phosphate; and those having a low potency in the assays described herein (500- 1000nm range IC50 range) such as 5-azacytidine, Ly333531(ruboxistaurin), and simvastatin.
The drug dose administered from the present compositions for the treatment of hypertrophic scars and keloids will depend on a variety of factors, including the type of formulation and the type of condition being treated. However, certain principles can be applied in the application of this art. Drug dose can be calculated as a function of dose per unit area (of the treatment site), total drug dose administered can be measured and appropriate surface concentrations of active drug can be determined. Drugs are to be used at concentrations that range from several times more than to 50%, 20%, 10%, 5%, or even less than 1% of the concentration typically used in a single systemic dose application. In certain aspects, the anti- scarring agent is released from the composition in effective concentrations in a time period that may be measured from the time of infiltration into tissue adjacent to the device, which ranges from about less than 1 day to about 180 days. Generally, the release time may also be from about less than 1 day to about 180 days; from about 7 days to about 14 days; from about 14 days to about 28 days; from about 28 days to about 56 days; from about 56 days to about 90 days; from about 90 days to about 180 days. In one aspect, the drug is released in effective concentrations for a period ranging from 1 - 90 days.
The exemplary anti-fibrosing agents, used alone or in combination, should be administered under the following dosing guidelines. The total amount (dose) of anti-scarring agent in the composition can be in the range of about 0.01 μg-10 μg, or 10 μg-10 mg, or 10 mg-250 mg, or 250 mg-1000 mg, or 1000 mg-2500 mg. The dose (amount) of anti-scarring agent per unit area of surface to which the agent is applied may be in the range of about 0.01 μg/mm2 - 1 μg/mm2, or 1 μg/mm2 - 10 μg/mm2, or 10 μg/mm2 - 250 μg/mm2, 250 μg/mm2 - 1000 μg/mm2, or 1000 μg/mm2 - 2500 μg/mm2.
Provided below are exemplary dosage ranges for various anti- scarring agents that can be used in conjunction with compositions for the treatment of inflammatory arthritis in accordance with the invention. The following dosages are particularly useful for intra-articular administration: . , (A) Angiogenesis inhibitors including Alphastatin, ZD-6474, IDN-5390, SB- 2723005, ABT-518, combretastatin, and anecortane, analogues and derivatives thereof: total single dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit volume of 0.01 μg - 100 mg per ml_; preferred dose of 0.1 μg/mL - 30 mg/mL. (B) mTOR inhibitors including AP-23573 and Temsirolimus, analogues and derivatives thereof: total single dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit volume of 0.01 μg - 100 mg per ml_; preferred dose of 0.1 μg/ ml_ - 20 mg/ ml_. (C) Tubulin antagonists including synthadotin, analogues and derivatives thereof: total single dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit volume of 0.01 μg - 100 μg per ml_; preferred dose of 0.1 μg/ ml_ - 20 mg/ ml_. (D) Epithilones including ixabepilone and analogues and derivatives thereof: total single dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit volume of 0.01 μg - 100 mg per mL; preferred dose of 0.1 μg/mL - 20 mg/ mL (E) Kinesin antagonists including SB-715992 and analogues and derivatives thereof: tota single dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit volume of 0.01 μg - 100 mg per mL; preferred dose of 0.1 μg/ mL - 20 mg/ mL. (F) TNF alpha antagonists including Etanercept, humicade, adalimumab and analogues and derivatives thereof: total single dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit volume of 0.01 μg - 100 mg per mL; preferred dose of 0.1 μg/ mL - 20 mg/ mL. (G) AKT inhibitor including erucylphosphocholine and analogues and derivatives thereof: total single dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit volume of 0.01 μg - 100 mg per mL; preferred dose of 0.1 μg/ mL - 20 mg/ mL. (H) FGF Inhibitors including IDN-5390 and analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit volume of 0.01 μg - 100 mg per mL; preferred dose of 0.1 μg/ mL - 20 mg/ mL. (I) Collagenase Antagonists including S-0885 and analogues and derivatives thereof: total dose not to exceed 1000 mg (range of 0.1 μg to 1000 mg); preferred 1 μg to 500 mg. Dose per unit volume of 0.01 μg - 300 mg per mL; preferred dose of 0.1 μg/ mL - 100 mg/ mL (J) NF KAPPA B Inhibitors including Bortezomib and analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit volume of 0.01 μg - 100 mg per mL; preferred dose of 0.1 μg/ mL - 20 mg/ mL. (K) Elongation factor-1 alpha inhibitors including aplidine and analogues and derivatives thereof: total single dose not to exceed 1000 mg (range of 0.1 μg to 1000 mg); preferred 1 μg to 500 mg. Dose per unit volume of 0.01 μg - 100 mg per mL; preferred dose of 0.1 μg/ mL - 100 mg/ mL. (L) Tyrosine kinase inhibitors including Gefitinib and analogues and derivatives thereof: total dose not to exceed 1000 mg (range of 0.1 μg to 1000 mg); preferred 1 μg to 500 mg. Dose per unit volume of 0.01 μg - 300 mg per mL; preferred dose of 0.1 μg/ mL - 100 μg/ ml_. In another aspect, systemic treatment may be administered when severe exacerbations or systemic disease (e.g., RA) are present. Anti- scarring agents that are delivered systemically should be dosed according to the level of drug required to inhibit the pathologies of inflammatory arthritis as described above. These systemic doses may vary according to patient, severity of disease, formulation of the administered agent, potency and/or tolerability of the agent, and route of administration. For example, for ZD-6474, AP-23573, synthadotin, S-0885, aplidine, ixabepilone, IDN-5390, SB-2723005, ABT-518, combretastatin, anecortave acetate, SB-715992, temsirolimus, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, dolastatin 15, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17- DMAG, tacrolimus, loteprednol etabonate, juglone, prednisolone, puromycin, 3-BAABE, cladribine, mannose-6-phosphate, 5-azacytidine, Ly333531 (ruboxistaurin), simvastatin, and an analogue or derivatives of the aforementioned, preferred embodiments would be 10 to 175 mg/m2 once every 1 to 4 weeks, 10 to 75 mg/m2 daily, as tolerated, or 10 to 175 mg/m2 weekly, as tolerated or until symptoms subside. To treat severe acute exacerbations, higher doses of 50 to 250 mg/m2 of ZD-6474, AP-23573, synthadotin, S-0885, aplidine, ixabepilone, IDN-5390, SB-2723005, ABT- 518, combretastatin, anecortave acetate, SB-715992, temsirolimus, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, dolastatin 15, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17-DMAG, tacrolimus, loteprednol etabonate, juglone, prednisolone, puromycin, 3-BAABE, cladribine, mannose-6-phosphate, 5-azacytidine, Ly333531 (ruboxistaurin), or simvastatin may be administered as a "pulse" systemic therapy. Other anti-scarring agents can be administered at equivalent doses adjusted for the potency and tolerability of the agent. For specific high potency drugs, the total single dose typically should not exceed 200 mg (range of 0.1 μg to 200 mg) and preferably 1 μg to 100 mg; dose per volume of 0.01 μg - 100 mg per ml_; preferably 0.1 μg/mL - 20 mg/mL For mid-potency drugs, the total single dose typically should not to exceed 500 mg (range of 1.0 μg to 500 mg) and preferably 1 μg to 200 mg; dose per volume of 0.01 μg - 200 mg per ml_, preferably 0.1 μg/mL - 40 mg/mL. For low potency drugs, the total dose typically should not exceed 1000 mg (range of 0.1 μg to 1000 mg), preferably 1 μg to 500 mg; dose per unit volume of 0.01 μg - 300 mg per mL; preferably 0.1 μg/mm2 - 100 mg/mL.
According to another aspect, any anti-infective agent described above may be used in conjunction with formulations for the treatment or prevention of hypertrophic scars and keloids. Exemplary anti- infective agents include (A) anthracyclines (e.g., doxorubicin and mitoxantrone), (B) fluoropyrimidines (e.g., 5-FU), (C) folic acid antagonists (e.g., methotrexate), (D) podophylotoxins (e.g., etoposide), (E) camptothecins, (F) hydroxyureas, and (G) platinum complexes (e.g., cisplatin), as well as analogues and derivatives of the aforementioned.
The drug dose administered from the present compositions for prevention or inhibition of infection in accordance with the present invention will depend on a variety of factors, including the type of formulation, the location of the treatment site, and the type of condition being treated. However, certain principles can be applied in the application of this art. Drug dose can be calculated as a function of dose per unit area (of the treatment site), total drug dose administered can be measured and appropriate surface concentrations of active drug can be determined. Drugs are to be used at concentrations that range from several times more than to 50%, 20%, 10%, 5%, or even less than 1% of the concentration typically used in a single anti-infective systemic dose application. In certain aspects, the anti-infective agent is released from the composition in effective concentrations in a time period that may be measured from the time of infiltration into tissue adjacent to the device, which ranges from about less than 1 day to about 180 days. Generally, the release time may also be from about less than 1 day to about 180 days; from about 7 days to about 14 days; from about 14 days to about 28 days; from about 28 days to about 56 days; from about 56 days to about 90 days; from about 90 days to about 180 days.
The exemplary anti-infective agents, used alone or in combination, should be administered under the following dosing guidelines. The total amount (dose) of anti-infective agent in the composition can be in the range of about 0.01 μg-1 μg, or about 1 μg-10 μg, or about 10 μg-1 mg, or about 1 mg to 10 mg, or about 10 mg-100 mg, or about 100 mg to 250 mg, or about 250 mg-1000 mg. The dose (amount) of anti-infective agent per unit area of device or tissue surface to which the agent is applied may be in the range of about 0.01 μg/mm2 - 1 μg/mm2, or about 1 μg/mm2 - 10 μg/mm2, or about 10 μg/mm2 - 100 μg/mm2, or about 100 μg/mm2 to 250 μg/mm2, or about 250 μg/mm2 - 1000 μg/mm2. As different compositions will release the anti-infective agent at differing rates, the above dosing parameters should be utilized in combination with the release rate of the drug from the composition such that a minimum concentration of about 10~8 M to 10~7 M, or about 10"7 M to 10"6 M about 10"6 M to 10'5 M or about 10"5 M to 10"4 M of the agent is maintained on the tissue surface.
(e) Vascular Diseases
In one aspect, the present invention provides for the use of a polymer composition comprising a polymeric carrier and one or more fibrosis-inhibiting agents for the treatment of vascular disease (e.g., stenosis, restenosis, or atherosclerosis).
Perivascular Delivery
A further aspect of the invention provides therapeutic compositions which may be delivered perivascularly (e.g., to an external portion of a blood vessel or directly into the adventitia of a blood vessel) for the treatment or prevention of a vascular disease (e.g., stenosis, restenosis, or atherosclerosis).
Perivascular drug delivery involves percutaneous administration of localized (often sustained release) therapeutic formulations using a needle or catheter directed via ultrasound, CT, fluoroscopic, MRI or endoscopic guidance to the adventitial surface of a targeted blood vessel (arteries, veins, autologous bypass grafts, synthetic bypass grafts, AV fistulas). Alternatively the procedure can be performed intra-operatively (e.g., during bypass surgery, hemodialysis access surgery) under direct vision or with additional imaging guidance. Such a procedure can also be performed in conjunction with endovascular procedures such as angioplasty, atherectomy, or stenting or in association with an operative arterial procedure such as endarterectomy, vessel or graft repair or graft insertion. For example, within one embodiment, polymeric formulations of for example, ZD-6474, AP-23573, synthadotin, S-0885, aplidine, ixabepilone, IDN-5390, SB-2723005, ABT-518, combretastatin, anecortave acetate, SB-715992, temsirolimus, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, dolastatin 15, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17-DMAG, tacrolimus, loteprednol etabonate, juglone, prednisolone, puromycin, 3-BAABE, cladribine, mannose-6-phosphate, 5- azacytidine, Ly333531 (ruboxistaurin), and simvastatin, can be injected into the vascular wall or applied to the adventitial surface of a blood vessel allowing drug concentrations to remain highest in regions where biological activity is most needed. This has the potential to reduce local "washout" of the drug that can be accentuated by continuous blood flow over the surface of an endovascular drug delivery device (such as a drug-coated stent). Administration of effective fibrosis-inhibiting agents to the external surface of the vessel can reduce obstruction of the artery, vein or graft and reduce the risk of complications associated with intravascular manipulations (such as restenosis, embolization, thrombosis, plaque rupture, and systemic drug toxicity).
For example, in a patient with narrowing of the superficial femoral artery, balloon angioplasty would be performed in the usual manner (i.e., passing a balloon angioplasty catheter down the artery over a guide wire and inflating the balloon across the lesion). Prior to, at the time of, or after angioplasty, a needle would be inserted through the skin under ultrasound, fluoroscopic, or CT guidance and a fibrosis-inhibiting agent or composition (e.g., ZD-6474, AP-23573, synthadotin, S-0885, aplidine, ixabepilone, IDN-5390, SB-2723005, ABT-518, combretastatin, anecortave acetate, SB-715992, temsirolimus, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, dolastatin 15, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17-DMAG, tacrolimus, loteprednol etabonate, juglone, - prednisolone, puromycin, 3-BAABE, cladribine, mannose-6-phosphate, 5- azacytidine, Ly333531 (ruboxistaurin), and simvastatin) impregnated into a slow release polymer) would be infiltrated through the needle or catheter in a circumferential manner directly around the area of narrowing in the artery. This could be performed around any artery, vein or graft, but ideal candidates for this intervention include diseases of the carotid, coronary, iliac, common femoral, superficial femoral and popliteal arteries and at the site of graft anastomosis. Logical venous sites include infiltration around veins in which indwelling catheters are inserted. Similarly at the time of endoscopic or open coronary bypass surgery, peripheral bypass surgery or hemodialysis access surgery, a fibrosis-inhibiting agent or composition (e.g., ZD-6474, AP-23573, synthadotin, S-0885, aplidine, ixabepilone, IDN-5390, SB-2723005, ABT-518, combretastatin, anecortave acetate, SB-715992, temsirolimus, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, dolastatin 15, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17- DMAG, tacrolimus, loteprednol etabonate, juglone, prednisolone, puromycin, 3-BAABE, cladribine, mannose-6-phosphate, 5-azacytidine, Ly333531 (ruboxistaurin), and simvastatin) impregnated into a slow release polymer) would be infiltrated, sprayed or wrapped in a circumferential manner in the region of the anastomosis where there is an increased incidence of restenosis. This could be performed around any artery, vein or graft, but ideal candidates for this intervention include diseases of the carotid, coronary, iliac, common femoral, superficial femoral and popliteal arteries and at the site of AV graft anastomosis.
According to the present invention, any anti-scarring agent described above can be utilized in the practice of this invention. Within one embodiment, compositions for perivascular drug delivery may be adapted to release an agent that inhibits one or more of the five general components of the process of fibrosis (or scarring), including: inflammatory response and" inflammation, migration and proliferation of connective tissue cells (such as fibroblasts or smooth muscle cells), formation of new blood vessels (angiogenesis), deposition of extracellular matrix (ECM), and remodeling (maturation and organization of the fibrous tissue). By inhibiting one or more of the components of fibrosis (or scarring), the overgrowth of neointimal tissue may be inhibited or reduced.
The drug dose of the fibrosis-inhibiting agent administered from the present compositions for perivascular delivery will depend on a variety of factors, including the type of formulation and the type of condition being treated. However, certain principles can be applied in the application of this art. Drug dose can be calculated as a function of dose per unit area (of the treatment site), total drug dose administered can be measured and appropriate surface concentrations of active drug can be determined. Drugs are to be used at concentrations that range from several times more than to 50%, 20%, 10%, 5%, or even less than 1% of the concentration typically used in a single systemic dose application. In certain aspects, the anti- scarring agent is released from the composition in effective concentrations in a time period that may be measured from the time of infiltration into tissue adjacent to the device, which ranges from about less than 1 day to about 180 days. Generally, the release time may also be from about less than 1 day to about 180 days; from about 7 days to about 14 days; from about 14 days to about 28 days; from about 28 days to about 56 days; from about 56 days to about 90 days; from about 90 days to about 180 days. In one aspect, the drug is released in effective concentrations for a period ranging from 1 - 90 days.
Several examples of fibrosis-inhibiting agents for use with compositions for perivascular drug delivery include the following: ZD-6474, AP-23573, synthadotin, S-0885, aplidine, ixabepilone, IDN-5390, SB- 2723005, ABT-518, combretastatin, anecortave acetate, SB-715992, temsirolimus, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, as well as analogues and derivatives of the aforementioned.
Additional specific examples of anti-scarring agents which can be used include those having a high potency in the assays described herein (1-10OnM IC50 range) such as isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, dolastatin 15, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17- DMAG, and tacrolimus; those having a mid-potency in the assays described herein (100-500 nM IC50 range) such as loteprednol etabonate, juglone, prednisolone, puromycin, 3-BAABE, cladribine, and mannose-6-phosphate; and those having a low potency in the assays described herein (500- 1000nm range IC5O range) such as 5-azacytidine, Ly333531(ruboxistaurin), and simvastatin.
The exemplary anti-fibrosing agents, used alone or in combination, should be administered under the following dosing guidelines. The total amount (dose) of anti-scarring agent in the composition can be in the range of about 0.01 μg-10 μg, or 10 μg-10 mg, or 10 mg-250 mg, or 250 mg-1000 mg, or 1000 mg-2500 mg. The dose (amount) of anti-scarring agent per unit area of surface to which the agent is applied may be in the range of about 0.01 μg/mm2 - 1 μg/mm2, or 1 μg/mm2 - 10 μg/mm2, or 10 μg/mm2 - 250 μg/mm2, 250 μg/mm2 - 1000 μg/mm2, or 1000 μg/mm2 - 2500 μg/mm2.
Provided below are exemplary dosage ranges for various anti- scarring agents that can be used in conjunction with perivascular administration in accordance with the invention. (A) Angiogenesis inhibitors including Alphastatin, ZD-6474, IDN-5390, SB-2723005, ABT-518, combretastatin, and anecortane, analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10^- 10"4 M of agent is to be maintained on the implant or barrier surface. (B) mTOR inhibitors including AP-23573 and Temsirplimus, analogues and derivatives thereof." total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10~8- 10"4 M of agent is to be maintained on the implant or barrier surface. (C) Tubulin antagonists including synthadotin, analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10 s - 10"4 M of agent is to be maintained on the implant or barrier surface. (D) Epithilones including ixabepilone and analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10"8- 10"4 M of agent is to be maintained on the implant or barrier surface. (E) Kinesin antagonists including SB- 715992 and analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10"8- 10"4 M of agent is to be maintained on the implant or barrier surface. (F) TNF alpha antagonists including etanercept, humicade, adalimumab and analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10'8- 10'4 M of agent is to be maintained on the implant or barrier surface. (G) AKT inhibitor including erucylphosphocholine and analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10^- 10"4 M of agent is to be maintained on the implant or barrier surface. (H) FGF Inhibitors including IDN-5390 and analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10"8 - 10"4 M of agent is to be maintained on the implant or barrier surface. (I) Collagenase Antagonists including S-0885 and analogues and derivatives thereof: total dose not to exceed 1000 mg (range of 0.1 μg to 1000 mg); preferred 1 μg to 500 mg. Dose per unit area of 0.01 μg - 500 μg per mm2; preferred dose of 0.1 μg/mm2 - 100 μg/mm2. Minimum concentration of 10^- 10"4 M of agent is to be maintained on the implant or barrier surface. (J) NF KAPPA B Inhibitors including Bortezomib and analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10^- 10"4 M of agent is to be maintained on the implant or barrier surface. (K) Elongation factor- 1 alpha inhibitors including aplidine and analogues and derivatives thereof: total dose not to exceed 1000 mg (range of 0.1 μg to 1000 mg); preferred 1 μg to 500 mg. Dose per unit area of 0.01 μg - 500 μg per mm2; preferred dose of 0.1 μg/mm2 - 100 μg/mm2. Minimum concentration of 10'8- 10"4 M of agent is to be maintained on the implant or barrier surface. (L) Tyrosine kinase inhibitors including Gefitinib and analogues and derivatives thereof: total dose not to exceed 1000 mg (range of 0.1 μg to 1000 mg); preferred 1 μg to 500 mg. Dose per unit area of 0.01 μg - 500 μg per mm2; preferred dose of 0.1 μg/mm2 - 100 μg/mm2. Minimum concentration of 10"8- 10"4 M of agent is to be maintained on the implant or barrier surface.
For high potency drugs, the total dose typically should not exceed 200 mg (range of 0.1 μg to 200 mg) and preferably 1 μg to 100 mg; dose per unit area of 0.01 μg - 100 μg per mm2; preferably 0.1 μg/mm2 - 20 μg/mm2; and minimum concentration of 108 - 10'4 M of agent should be maintained on the implant or barrier surface. For mid-potency drugs, the total dose typically should not to exceed 500 mg (range of 1.0 μg to 500 mg) and preferably 1 μg to 200 mg; dose per unit area of 0.01 μg - 200 μg per mm2, preferably 0.1 μg/mm2 - 40 μg/mm2; and minimum concentration of 10" 8 - 10"4 M of agent should be maintained on the implant or barrier surface. For low potency drugs, the total dose typically should not exceed 1000 mg (range of 0.1 μg to 1000 mg), preferably 1 μg to 500 mg; dose per unit area of 0.01 μg - 500 μg per mm2; preferably 0.1 μg/mm2 - 100 μg/mm2; and minimum concentration of 10^ - 10"4 M of agent should to be maintained on the implant or barrier surface.
According to another aspect, any anti-infective agent described above may be used alone or in conjunction with an anti-fibrosing agent in the practice of the present embodiment. Exemplary anti-infective agents include (A) anthracyclines (e.g., doxorubicin and mitoxantrone), (B) fluoropyrimidines (e.g., 5-FU), (C) folic acid antagonists (e.g., methotrexate), (D) podophylotoxins (e.g., etoposide), (E) camptothecins, (F) hydroxyureas, and (G) platinum complexes (e.g., cisplatin), as well as analogues and derivatives of the aforementioned. The drug dose administered from the present compositions for prevention or inhibition of infection in accordance with the present invention will depend on a variety of factors, including the type of formulation, the location of the treatment site, and the type of condition being treated. However, certain principles can be applied in the application of this art. Drug dose can be calculated as a function of dose per unit area (of the treatment site), total drug dose administered can be measured and appropriate surface concentrations of active drug can be determined. Drugs are to be used at concentrations that range from several times more than to 50%, 20%, 10%, 5%, or even less than 1% of the concentration typically used in a single anti-infective systemic dose application. In certain aspects, the anti-infective agent is released from the composition in effective concentrations in a time period that may be measured from the time of infiltration into tissue adjacent to the device, which ranges from about less than 1 day to about 180 days. Generally, the release time may also be from about less than 1 day to about 180 days; from about 7 days to about 14 days; from about 14 days to about 28 days; from about 28 days to about 56 days; from about 56 days to about 90 days; from about 90 days to about 180 days.
The exemplary anti-infective agents, used alone or in combination, should be administered under the following dosing guidelines. The total amount (dose) of anti-infective agent in the composition can be in the range of about 0.01 μg-1 μg, or about 1 μg-10 μg, or about 10 μg-1 mg, or about 1 mg to 10 mg, or about 10 mg-100 mg, or about 100 mg to 250 mg, or about 250 mg-1000 mg. The dose (amount) of anti-infective agent per unit area of device or tissue surface to which the agent is applied may be in the range of about 0.01 μg/mm2 - 1 μg/mm2, or about 1 μg/mm2 - 10 μg/mm2, or about 10 μg/mm2 - 100 μg/mm2, or about 100 μg/mm2 to 250 μg/mm2, or about 250 μg/mm2 - 1000 μg/mm2. As different compositions will release the anti-infective agent at differing rates, the above dosing parameters should be utilized in combination with the release rate of the drug from the composition such that a minimum concentration of about 10'8 M to 10"7 M, or about 10"7 M to 10'6 M about 10"6 M to 1(T5 M or about 10'5 M to 10"4 M of the agent is maintained on the tissue surface.
(f) Combining With Medical Devices or Implants
The fibrosis-inhibiting agents and/or anti-infective agents and compositions of the present invention can also be combined with an implant or an implantable medical device, (e.g., artificial joints, retaining pins, cranial plates, and the like, of metal, plastic and/or other materials), breast implants (e.g., silicone gel envelopes, foam forms, and the like), implanted catheters and cannulas intended for long-term use (beyond about three days), artificial organs and vessels (e.g., artificial hearts, pancreases, kidneys, blood vessels, and the like), drug delivery devices (including monolithic implants, pumps and controlled release devices such as ALZET minipumps (DURECT Corporation, Cupertino, California), steroid pellets for anabolic growth or contraception, and the like, sutures for dermal or internal use, periodontal membranes, ophthalmic shields, corneal lenticules, and the like.
A range of polymeric and non-polymeric materials can be used to incorporate the fibrosis-inhibiting agent onto or into a device. The anti- fibrosing agent composition can be incorporated into or onto the device in a variety of ways. Coating of the device with the fibrosis-inhibiting agent containing composition or with the fibrosis-inhibiting agent only is one process that can be used to incorporate the fibrosis-inhibiting agent into or onto the device. The anti-fibrosing agent or anti-fibrosing composition may be coated onto the entire device or a portion of the device using a method, such as by dipping, spraying, painting or vacuum deposition, that is appropriate for the particular type of device. B. Dip coating
Dip coating is one coating process that can be used. In one embodiment, the fibrosis-inhibiting agent is dissolved in a solvent for the fibrosis agent and is then coated onto the device.
Fibrosis-inhibiting agent with an inert-solvent In one embodiment, the solvent is an inert solvent for the device such that the solvent does not dissolve the medical device to any great extent and is not absorbed by the device to any great extent. The device can be immersed, either partially or completely, in the fibrosis- inhibiting agent/solvent solution for a specific period of time. The rate of immersion into the fibrosis-inhibiting agent/solvent solution can be altered (e.g., 0.001 cm per sec to 50 cm per sec). The device can then be removed from the solution. The rate at which the device can be withdrawn from the solution can be altered (e.g., 0.001 cm per sec to 50 cm per sec). The coated device can be air-dried. The dipping process can be repeated one or more times depending on the specific application. The device can be dried under vacuum to reduce residual solvent levels. This process will result in the fibrosis-inhibiting agent being coated on the surface of the device.
Fibrosis-inhibiting agent with a swelling solvent In one embodiment, the solvent is one that will not dissolve the device but will be absorbed by the device. These solvents can thus swell the device to some extent. The device can be immersed, either partially or completely, in the fibrosis-inhibiting agent/solvent solution for a specific period of time (seconds to days). The rate of immersion into the fibrosis- inhibiting agent/solvent solution can be altered (e.g., 0.001 cm per sec to 50 cm per sec). The device can then be removed from the solution. The rate at which the device can be withdrawn from the solution can be altered (e.g., 0.001 cm per sec to 50 cm per sec). The coated device can be air-dried. The clipping process can be repeated one or more times depending on the specific application. The device can be dried under vacuum to reduce residual solvent levels. This process will result in the fibrosis-inhibiting agent being adsorbed into the medical device. The fibrosis-inhibiting agent may also be present on the surface of the device. The amount of surface associated fibrosis-inhibiting agent may be reduced by dipping the coated device into a solvent for the fibrosis-inhibiting agent or by spraying the coated device with a solvent for the fibrosis-inhibiting agent.
Fibrosis-inhibiting agent with a solvent
In one embodiment, the solvent is one that will be absorbed by the device and that will dissolve the device. The device can be immersed, either partially or completely, in the fibrosis-inhibiting agent/solvent solution for a specific period of time (seconds to hours). The rate of immersion into the fibrosis-inhibiting agent/solvent solution can be altered (e.g., 0.001 cm per sec to 50 cm per sec). The device can then be removed from the solution. The rate at which the device can be withdrawn from the solution can be altered (e.g., 0.001 cm per sec to 50 cm per sec). The coated device can be air-dried. The dipping process can be repeated one or more times depending on the specific application. The device can be dried under vacuum to reduce residual solvent levels. This process will result in the fibrosis-inhibiting agent being adsorbed into the medical device as well as being surface associated. In the preferred embodiment, the exposure time of the device to the solvent can be such that there are no significant permanent dimensional changes to the device. The fibrosis-inhibiting agent may also be present on the surface of the device. The amount of surface associated fibrosis-inhibiting agent may be reduced by dipping the coated device into a solvent for the fibrosis-inhibiting agent or by spraying the coated device with a solvent for the fibrosis-inhibiting agent.
In the above description the device can be a device that has not been modified as well as a device that has been further modified by coating with a polymer, surface treated by plasma treatment, flame treatment, corona treatment, surface oxidation or reduction, surface etching, mechanical smoothing or roughening, or grafting prior to the coating process.
In one embodiment, the fibrosis-inhibiting agent and a polymer are dissolved in a solvent, for both the polymer and the fibrosis -inhibiting agent, and are then coated onto the device.
In any one the above dip coating methods, the surface of the device can be treated with a plasma polymerization method prior to coating of the scarring agent or scarring agent containing composition, such that a thin polymeric layer is deposited onto the device surface. Examples of such methods include parylene coating of devices and the use of various monomers such hydrocyclosiloxane monomers. Parylene coating may be especially advantageous if the device, or portions of the device, is composed of materials (e.g., stainless steel, nitinol) that do not allow incorporation of the therapeutic agent(s) into the surface layer using one of the above methods. A parylene primer layer may be deposited onto the device using a parylene coater (e.g., PDS 2010 LABCOTER2 from Cookson Electronics) and a suitable reagent (e.g., di-p-xylylene or dichloro-di-p- xylylene) as the coating feed material. Paryiene compounds are commercially available, for example, from Specialty Coating Systems, Indianapolis, IN), including PARYLENE N (di-p-xylylene), PARYLENE C (a monchlorinated derivative of PARYLENE N, and PARYLENE D, a dichlorinated derivative of PARYLENE N).
Fibrosis-inhibiting agent/polymer with an inert-solvent In one embodiment, the solvent is an inert solvent for the device such that the solvent does not dissolve the medical device to any great extent and is not absorbed by the device to any great extent. The device can be immersed, either partially or completely, in the fibrosis- inhibiting agent/polymer/solvent solution for a specific period of time. The rate of immersion into the fibrosis-inhibiting agent/polymer/solvent solution can be altered (e.g., 0.001 cm per sec to 50 cm per sec). The device can then be removed from the solution. The rate at which the device can be withdrawn from the solution can be altered (e.g., 0.001 cm per sec to 50 cm per sec). The coated device can be air-dried. The dipping process can be repeated one or more times depending on the specific application. The device can be dried under vacuum to reduce residual solvent levels. This process will result in the fibrosis-inhibiting agent/polymer being coated on the surface of the device.
Fibrosis-inhibiting agent/polymer with a swelling solvent In one embodiment, the solvent is one that will not dissolve the device but will be absorbed by the device. These solvents can thus swell the device to some extent. The device can be immersed, either partially or completely, in the fibrosis-inhibiting agent/polymer/solvent solution for a specific period of time (seconds to days). The rate of immersion into the fibrosis-inhibiting agent/polymer/solvent solution can be altered (e.g., 0.001 cm per sec to 50 cm per sec). The device can then be removed from the solution. The rate at which the device can be withdrawn from the solution can be altered (e.g., 0.001 cm per sec to 50 cm per sec). The coated device can be air-dried. The dipping process can be repeated one or more times depending on the specific application. The device can be dried under vacuum to reduce residual solvent levels. This process will result in the fibrosis-inhibiting agent/polymer being coated onto the surface of the device as well as the potential for the fibrosis-inhibiting agent being adsorbed into the medical device. The fibrosis-inhibiting agent may also be present on the surface of the device. The amount of surface associated fibrosis-inhibiting agent may be reduced by dipping the coated device into a solvent for the fibrosis-inhibiting agent or by spraying the coated device with a solvent for the fibrosis-inhibiting agent. Fibrosis-inhibiting agent/polymer with a solvent
In one embodiment, the solvent is one that will be absorbed by the device and that will dissolve the device. The device can be immersed, either partially or completely, in the fibrosis-inhibiting agent/solvent solution for a specific period of time (seconds to hours). The rate of immersion into the fibrosis-inhibiting agent/solvent solution can be altered (e.g., 0.001 cm per sec to 50 cm per sec). The device can then be removed from the solution. The rate at which the device can be withdrawn from the solution can be altered (e.g., 0.001 cm per sec to 50 cm per sec). The coated device can be air-dried. The dipping process can be repeated one or more times depending on the specific application. The device can be dried under vacuum to reduce residual solvent levels. In the preferred embodiment, the exposure time of the device to the solvent can be such that there are not significant permanent dimensional changes to the device (other than those associated with the coating itself). The fibrosis-inhibiting agent may also be present on the surface of the device. The amount of surface associated fibrosis-inhibiting agent may be reduced by dipping the coated device into a solvent for the fibrosis-inhibiting agent or by spraying the coated device with a solvent for the fibrosis-inhibiting agent.
In the above description the device can be a device that has not been modified as well as a device that has been further modified by coating with a polymer (e.g., parylene), surface treated by plasma treatment, flame treatment, corona treatment, surface oxidation or reduction, surface etching, mechanical smoothing or roughening, or grafting prior to the coating process.
In another embodiment, a suspension of the fibrosis-inhibiting agent in a polymer solution can be prepared. The suspension can be prepared by choosing a solvent that can dissolve the polymer but not the fibrosis-inhibiting agent or a solvent that can dissolve the polymer and in which the fibrosis-inhibiting agent is above its solubility limit. In similar processes described above, a device can be dipped into the suspension of the fibrosis-inhibiting and polymer solution such that the device is coated with a polymer that has a fibrosis-inhibiting agent suspended within it.
C. Spray coating
Spray coating is another coating process that can be used. In the spray coating process, a solution or suspension of the fibrosis-inhibiting agent, with or without a polymeric or non-polymeric carrier, is nebulized and directed to the device to be coated by a stream of gas. One can use spray devices such as an air-brush (for example models 2020, 360, 175, 100, 200, 150, 350, 250, 400, 3000, 4000, 5000, 6000 from Badger Air-brush Company, Franklin Park, IL), spray painting equipment, TLC reagent sprayers (for example Part # 14545 and 14654, Alltech Associates, Inc. Deerfield, IL, and ultrasonic spray devices (for example those available from Sono-Tek, Milton, NY). One can also use powder sprayers and electrostatic sprayers.
In one embodiment, the fibrosis-inhibiting agent is dissolved in a solvent for the fibrosis agent and is then sprayed onto the device.
Fibrosis-inhibiting agent with an inert-solvent In one embodiment, the solvent is an inert solvent for the device such that the solvent does not dissolve the medical device to any great extent and is not absorbed by the device to any great extent. The device can be held in place or the device can be mounted onto a mandrel or rod that has the ability to move in an X, Y or Z plane or a combination of these planes. Using one of the above described spray devices, the device can be spray coated such that the device is either partially or completely coated with the fibrosis-inhibiting agent/solvent solution. The rate of spraying of the fibrosis-inhibiting agent/solvent solution can be altered (e.g., 0.001 mL per sec to 10 mL per sec) to ensure that a good coating of the fibrosis-inhibiting agent is obtained. The coated device can be air-dried. The spray coating process can be repeated one or more times depending on the specific application. The device can be dried under vacuum to reduce residual solvent levels. This process will result in the fibrosis- inhibiting agent being coated on the surface of the device.
Fibrosis-inhibiting agent with a swelling solvent In one embodiment, the solvent is one that will not dissolve the device but will be absorbed by the device. These solvents can thus swell the device to some extent. The device can be spray coated, either partially or completely, in the fibrosis-inhibiting agent/solvent solution. The rate of spraying of the fibrosis-inhibiting agent/solvent solution can be altered (e.g., 0.001 mL per sec to 10 ml_ per sec) to ensure that a good coating of the fibrosis-inhibiting agent is obtained. The coated device can be air-dried. The spray coating process can be repeated one or more times depending on the specific application. The device can be dried under vacuum to reduce residual solvent levels. This process will result in the fibrosis- inhibiting agent being adsorbed into the medical device. The fibrosis- inhibiting agent may also be present on the surface of the device. The amount of surface associated fibrosis-inhibiting agent may be reduced by dipping the coated device into a solvent for the fibrosis-inhibiting agent or by spraying the coated device with a solvent for the fibrosis-inhibiting agent.
Fibrosis-inhibiting agent with a solvent
In one embodiment, the solvent is one that will be absorbed by the device and that will dissolve the device. The device can be spray coated, either partially or completely, in the fibrosis-inhibiting agent/solvent solution. The rate of spraying of the fibrosis-inhibiting agent/solvent solution can be altered (e.g., 0.001 mL per sec to 10 mL per sec) to ensure that a good coating of the fibrosis-inhibiting agent is obtained. The coated device can be air-dried. The spray coating process can be repeated one or more times depending on the specific application. The device can be dried under vacuum to reduce residual solvent levels. This process will result in the fibrosis-inhibiting agent being adsorbed into the medical device as well as being surface associated. In the preferred embodiment, the exposure time of the device to the solvent can be such that there are not significant permanent dimensional changes to the device. The fibrosis-inhibiting agent may also be present on the surface of the device. The amount of surface associated fibrosis-inhibiting agent may be reduced by dipping the coated device into a solvent for the fibrosis-inhibiting agent or by spraying the coated device with a solvent for the fibrosis-inhibiting agent.
In the above description the device can be a device that has not been modified as well as a device that has been further modified by coating with a polymer (e.g., parylene), surface treated by plasma treatment, flame treatment, corona treatment, surface oxidation or reduction, surface etching, mechanical smoothing or roughening, or grafting prior to the coating process.
In one embodiment, the fibrosis-inhibiting agent and a polymer are dissolved in a solvent, for both the polymer and the anti-fibrosing agent, and are then spray coated onto the device.
Fibrosis-inhibiting agent/polymer with an inert-solvent In one embodiment, the solvent is an inert solvent for the device such that the solvent does not dissolve the medical device to any great extent and is not absorbed by the device to any great extent. The device can be spray coated, either partially or completely, in the fibrosis- inhibiting agent/polymer/solvent solution for a specific period of time. The rate of spraying of the fibrosis-inhibiting agent/solvent solution can be altered (e.g., 0.001 ml. per sec to 10 ml_ per sec) to ensure that a good coating of the fibrosis-inhibiting agent is obtained. The coated device can be air-dried. The spray coating process can be repeated one or more times depending on the specific application. The device can be dried under vacuum to reduce residual solvent levels. This process will result in the fibrosis-inhibiting agent/polymer being coated on the surface of the device. Fibrosis-inhibitinq agent/polymer with a swelling solvent In one embodiment, the solvent is one that will not dissolve the device but will be absorbed by the device. These solvents can thus swell the device to some extent. The device can be spray coated, either partially or completely, in the fibrosis-inhibiting agent/polymer/solvent solution. The rate of spraying of the fibrosis-inhibiting agent/solvent solution can be altered (e.g., 0.001 ml_ per sec to 10 mL per sec) to ensure that a good coating of the fibrosis-inhibiting agent is obtained. The coated device can be air-dried. The spray coating process can be repeated one or more times depending on the specific application. The device can be dried under vacuum to reduce residual solvent levels. This process will result in the fibrosis-inhibiting agent/polymer being coated onto the surface of the device as well as the potential for the fibrosis-inhibiting agent being adsorbed into the medical device. The fibrosis-inhibiting agent may also be present on the surface of the device. The amount of surface associated fibrosis-inhibiting agent may be reduced by dipping the coated device into a solvent for the fibrosis-inhibiting agent or by spraying the coated device with a solvent for the fibrosis-inhibiting agent.
Fibrosis-inhibiting agent/polymer with a solvent In one embodiment, the solvent is one that will be absorbed by the device and that will dissolve the device. The device can be spray coated, either partially or completely, in the fibrosis-inhibiting agent/solvent solution. The rate of spraying of the fibrosis-inhibiting agent/solvent solution can be altered (e.g., 0.001 mL per sec to 10 mL per sec) to ensure that a good coating of the fibrosis-inhibiting agent is obtained. The coated device can be air-dried. The spray coating process can be repeated one or more times depending on the specific application. The device can be dried under vacuum to reduce residual solvent levels. In the preferred embodiment, the exposure time of the device to the solvent can be such that there are not significant permanent dimensional changes to the device (other than those associated with the coating itself). The fibrosis-inhibiting agent may also be present on the surface of the device. The amount of surface associated fibrosis-inhibiting agent may be reduced by dipping the coated device into a solvent for the fibrosis-inhibiting agent or by spraying the coated device with a solvent for the fibrosis-inhibiting agent.
In the above description the device can be a device that has not been modified as well as a device that has been further modified by coating with a polymer (e.g., parylene), surface treated by plasma treatment, flame treatment, corona treatment, surface oxidation or reduction, surface etching, mechanical smoothing or roughening, or grafting prior to the coating process.
In another embodiment, a suspension of the fibrosis-inhibiting agent in a polymer solution can be prepared. The suspension can be prepared by choosing a solvent that can dissolve the polymer but not the fibrosis-inhibiting agent or a solvent that can dissolve the polymer and in which the fibrosis-inhibiting agent is above its solubility limit. In similar processes described above, the suspension of the fibrosis-inhibiting and polymer solution can be sprayed onto the device such that the device is coated with a polymer that has a fibrosis-inhibiting agent suspended within it.
In a general method for coating a surface of a synthetic implant, the multifunctional compounds are exposed to the modified environment, and a thin layer of the composition is then applied to a surface of the implant before substantial inter-reaction has occurred. In one embodiment, in order to minimize cellular and fibrous reaction to the coated implant, the compounds are selected so as to result in a matrix that has a net neutral charge. Application of the compounds to the implant surface may be by extrusion, brushing, spraying, or by any other convenient means. Following application of the compounds to the implant surface, inter-reaction is allowed to continue until complete and the three-dimensional matrix is formed. Although this method can be used to coat the surface of any type of synthetic implant, it is particularly useful for implants where reduced thrombogenicity is an important consideration, such as artificial blood vessels and heart valves, vascular grafts, vascular stents, anastomotic connector devices, and stent/graft combinations. The method may also be used to coat implantable surgical membranes (e.g., monofilament polypropylene) or meshes (e.g., for use in hernia repair). Breast implants may also be coated using the above method in order to minimize capsular contracture.
The fibrosis-inhibiting compounds and compositions can also be coated on a suitable fibrous material, which can then be wrapped around a bone to provide structural integrity to the bone. The term "suitable fibrous material" as used herein, refers to a fibrous material which is substantially insoluble in water, non-immunogenic, biocompatible, and immiscible with the crosslinkable compositions of the invention. The fibrous material may comprise any of a variety of materials having these characteristics and may be combined with crosslinkable compositions herein in order to form and/or provide structural integrity to various implants or devices used in connection with medical and pharmaceutical uses.
The fibrosis-inhibiting compounds and compositions of the present invention may also be used to coat lenticules, which are made from either naturally occurring or synthetic polymers.
In yet another example, the device can be coated with a polyurethane and then allowed to partially dry such that the surface is still tacky. A particulate form of the fibrosis-inhibiting agent or fibrosis-inhibiting agent/secondary carrier can then be applied to all or a portion of the tacky coating after which the device is dried.
In yet another example, the device can be coated with one of the coatings described above. A thermal treatment process can then be used to soften the coating, afterwhich the fibrosis-inhibiting agent or the fibrosis-inhibiting agent/secondary carrier is applied to the entire device or to a portion of the device (e.g., outer surface).
In one embodiment, all or a portion of the device is coated with a primer (bonding) layer and a drug release layer, as described in U.S. Patent application entitled, "Stent with Medicated Multi-Layer Hybrid Polymer Coating," filed September 16, 2003 (U.S. Serial No. 10/662,877).
In order to develop a hybrid polymer delivery system for targeted therapy, it is desirable to be able to control and manipulate the properties of the system both in terms of physical and drug release characteristics. The active agents can be imbibed into a surface hybrid polymer layer, or incorporated directly into the hybrid polymer coating solutions. Imbibing drugs into surface polymer layers is an efficient method for evaluating polymer-drug performance in the laboratory, but for commercial production it may be preferred for the polymer and drug to be premixed in the casting mixture. Greater efficacy can be achieved by combining the two elements in the coating mixtures in order to control the ratio of active agent to polymer in the coatings. Such ratios are important parameters to the final properties of the medicated layers, i.e., they allow for better control of active agent concentration and duration of pharmacological activity.
Typical polymers used in the drug-release system can include water-insoluble cellulose esters, various polyurethane polymers including hydrophilic and hydrophobic versions, hydrophilic polymers such as polyethylene glycol (PEG), polyethylene oxide (PEO), polyvinylpyrrolidone (PVP), PVP copolymers such as vinyl acetate, hydroxyethyl methacrylate (HEMA) and copolymers such as methylmethacrylate (PMMA-HEMA), and other hydrophilic and hydrophobic acrylate polymers and copolymers containing functional groups such as carboxyl and/or hydroxyl.
Cellulose esters such as cellulose acetate, cellulose acetate propionate, cellulose acetate butyrate, cellulose acetate phthalate, and cellulose nitrate may be used. In one aspect of the invention, the therapeutic agent is formulated with a cellulose ester. Cellulose nitrate is a preferred cellulose ester because of its compatibility with the active agents and its ability to impart non-tackiness and cohesiveness to the coatings. Cellulose nitrate has been shown to stabilize entrapped drugs in ambient and processing conditions. Various grades of cellulose nitrate are available and may be used in a coating on a device, including cellulose nitrate having a nitrogen content = 11.8-12.2%. Various viscosity grades, including 3.5, 0.5 or 0.25 seconds, may be used in order to provide proper rheological properties when combined with the coating solids used in these formulations. Higher or lower viscosity grades can be used. However, the higher viscosity grades can be more difficult to use because of their higher viscosities. Thus, the lower viscosity grades, such as 3.5, 0.5 or 0.25 seconds, are generally preferred. Physical properties such as tensile strength, elongation, flexibility, and softening point are related to viscosity (molecular weight) and can decrease with the lower molecular weight species, especially below the 0,25 second grades.
The cellulose derivatives comprise hydroglucose structures. Cellulose nitrate is a hydrophobic, water-insoluble polymer, and has high water resistance properties. This structure leads to high compatibility with many active agents, accounting for the high degree of stabilization provided to drugs entrapped in cellulose nitrate. The structure of nitrocellulose is given below:
Figure imgf000413_0001
nitrocellulose
Cellulose nitrate is a hard, relatively inflexible polymer, and has limited adhesion to many polymers that are typically used to make medical devices. Also, control of drug elution dynamics is limited if only one polymer is used in the binding matrix. Accordingly, in one embodiment of the invention, the therapeutic agent is formulated with two or more polymers before being associated with the device. In one aspect, the agent is formulated with both polyurethane ((e.g., CHRONOFLEX AR, CHRONOFLEX AL, and BIONATE, PELLETHANE) and cellulose nitrate to provide a hybrid polymer drug loaded matrix. Polyurethanes provide the hybrid polymer matrix with greater flexibility and adhesion to the device, particularly when the device has been pre-coated with a primer. Polyurethanes can also be used to slow or hasten the drug elution from coatings. Aliphatic, aromatic, polytetramethylene ether glycol, and polycarbonate are among the types of polyurethanes, which can be used in the coatings. In one aspect, an anti-scarring agent (e.g., ZD-6474, AP- 23573, synthadotin, S-0885, aplidine, ixabepilone, IDN-5390, SB-2723005, ABT-518, combretastatin, anecortave acetate, SB-715992, temsirolimus, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib. as well as^analogues and derivatives of the aforementioned) may be incorporated into a carrier that includes a polyurethane and a cellulose derivative. Additional examples of anti-scarring agents which can be used include those having a high potency in the assays described herein (1- 10OnM IC5O range) such as isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, dolastatin 15, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17- DMAG, and tacrolimus; those having a mid-potency in the assays described herein (100-500 nM IC50 range) such as loteprednol etabonate, juglone, prednisolone, puromycin, 3-BAABE, cladribine, and mannose-6-phosphate; and those having a low potency in the assays described herein (500- 1000nm range IC50 range) such as 5-azacytidine, Ly333531(ruboxistaurin), and simvastatin.
A heparin complex, such as benzalkonium heparinate or tridodecylammonium heparinate), may optionally be included in the formulation. From the structure below, it is possible to see how more or less hydrophilic polyurethane polymers may be created based on the number of hydrophilic groups contained in the polymer structures. In one aspect of the invention, the device is associated with a formulation that includes therapeutic agent, cellulose ester, and a polyurethane that is water- insoluble, flexible, and compatible with the cellulose ester.
Figure imgf000415_0001
Polyvinylpyrrolidone (PVP) is a polyamide that possesses unusual complexing and colloidal properties and is essentially physiologically inert. PVP and other hydrophilic polymers are typically biocompatible. PVP may be incorporated into drug loaded hybrid polymer compositions in order to increase drug release rates. In one embodiment, the concentration of PVP that is used in drug loaded hybrid polymer compositions can be less than 20%. This concentration can not make the layers bioerodable or lubricious. In general, PVP concentrations from <1% to greater than 80% are deemed workable. In one aspect of the invention, the therapeutic agent that is associated with an device is formulated with a PVP polymer.
Figure imgf000415_0002
polyvinylpyrrolidone
Acrylate polymers and copolymers including polymethylmethacrylate (PMMA) and polymethylmethacrylate hydroxyethyl
Figure imgf000415_0003
methacrylate (PMMA/HEMA) are known for their biocompatibility as a result of their widespread use in contact and intraocular lens applications. This class of polymer generally provokes very little smooth muscle and endothelial cell growth, and very low inflammatory response (Bar). These polymers/copolymers are compatible with drugs and the other polymers and layers of the instant invention. Thus, in one aspect, the device is associated with a composition that comprises an anti-scarring agent as described above, and an acrylate polymer or copolymer.
Methylmethacrylate Hydroxyethylmethacrylate Copolymer
Within another aspect of the invention, the coated device which inhibits or reduces an in vivo fibrotic reaction is further coated with a compound or compositions which delay the release of and/or activity of the fibrosis-inhibiting agent. Representative examples of such agents include biologically inert materials such as gelatin, PLGA/MePEG film, PLA, polyurethanes, silicone rubbers, surfactants, lipids, or polyethylene glycol, as well as biologically active materials such as heparin (e.g., to induce coagulation).
For example, in one embodiment of the invention, the active agent on the device is top-coated with a physical barrier. Such barriers can include non-degradable materials or biodegradable materials such as gelatin, PLGA/MePEG film, PLA, or polyethylene glycol among others. In one embodiment, the rate of diffusion of the therapeutic agent in the barrier coat is slower that the rate of diffusion of the therapeutic agent in the coating layer. In the case of PLGA/ MePEG, once the PLGA/ MePEG becomes exposed to the bloodstream, the MePEG can dissolve out of the PLGA, leaving channels through the PLGA layer to an underlying layer containing the fibrosis-inhibiting agent, which then can then diffuse into the vessel wall and initiate its biological activity. In another embodiment of the invention, a particulate form of the active agent may be coated onto any of the devices described below) using a polymer (e.g., PLG, PLA, aor a polyurethane). A second polymer, that dissolves slowly or degrades (e.g., MePEG-PLGA or PLG) and that does not contain the active agent, may be coated over the first layer. Once the top layer dissolves or degrades, it exposes the under coating which allows the active agent to be exposed to the treatment site or to be released from the coating.
Within another aspect of the invention, the outer layer of the coating of a coated device, which inhibits an in vivo fibrotic response, is further treated to crosslink the outer layer of the coating. This can be accomplished by subjecting the coated device to a plasma treatment process. The degree of crosslinking and nature of the surface modification can be altered by changing the RF power setting, the location with respect to the plasma, the duration of treatment as well as the gas composition introduced into the plasma chamber.
Protection of a biologically active surface can also be utilized by coating the device surface with an inert molecule that prevents access to the active site through steric hindrance, or by coating the surface with an inactive form of the fibrosis-inhibiting agent, which is later activated. For example, the device can be coated with an enzyme, which causes either release of the fibrosis-inhibiting agent or activates the fibrosis-inhibiting agent.
In another embodiment, the device is coated with a fibrosis- inhibiting agent and then further coated with a composition that comprises an anticoagulant such as heparin. As the anticoagulant dissolves away, the anticoagulant activity slows or stops, and the newly exposed fibrosis- inhibiting agent is available to inhibit or reduce fibrosis from occurring in the adjacent tissue.
The device can be coated with an inactive form of the fibrosis- inhibiting agent, which is then activated once the device is deployed. Such activation can be achieved by injecting another material into the treatment area after the device (as desribed below) is deployed or after the fibrosis- inhibiting agent has been administered to the treatment area (via, e.g., injections, spray, wash, drug delivery catheters or balloons). For example, the device can be coated with an inactive form of the fibrosis-inhibiting agent. Once the device is deployed, the activating substance is injected or applied into or onto the treatment site where the inactive form of the fibrosis- inhibiting agent has been applied. For example, a device can be coated with a biologically active fibrosis-inhibiting agent and a first substance having moieties that capable of forming an ester bond with another material. The coating can be covered with a second substance such as polyethylene glycol. The first and second substances can react to form an ester bond via, e.g., a condensation reaction. Prior to the deployment of the device, an esterase is injected into the treatment site around the outside of the device, which can cleave the bond between the ester and the fibrosis-inhibiting agent, allowing the agent to initiate fibrosis-inhibition.
In another aspect, a medical device may include a plurality of reservoirs within its structure, each reservoir configured to house and protect a therapeutic drug. The reservoirs may be formed from divets in the device surface or micropores or channels in the device body. In one aspect, the reservoirs are formed from voids in the structure of the device. The reservoirs may house a single type of drug or more than one type of drug. The drug(s) may be formulated with a carrier (e.g., a polymeric or non- polymeric material) that is loaded into the reservoirs. The filled reservoir can function as a drug delivery depot which can release drug over a period of time dependent on the release kinetics of the drug from the carrier. In certain embodiments, the reservoir may be loaded with a plurality of layers. Each layer may include a different drug having a particular amount (dose) of drug, and each layer may have a different composition to further tailor the amount of drug that is released from the substrate. The multi-layered carrier may further include a barrier layer that prevents release of the drug(s). The barrier layer can be used, for example, to control the direction that the drug elutes from the void.
As described above, the anti-fibrosing agent can be associated with a medical device using the polymeric carriers or coatings described above. In addition to the compositions and methods described above, there are various other compositions and methods that are known in the art. Representative examples of these compositions and methods for applying (e.g., coating) these compositons to devices are described in U.S. Patent. Nos. 6,610,016; 6,358,557; 6,306,176; 6,110,483; 6,106,473; 5,997,517; 5,800,412; 5,525,348; 5,331 ,027; 5,001,009; 6,562,136; 6,406,754; 6,344,035; 6,254,921 ; 6,214,901; 6,077,698; 6,603,040; 6,278,018; 6,238,799; 6,096,726, 5,766,158, 5,599,576, 4,119,094; 4,100,309; 6,599,558; 6,369,168; 6,521 ,283; 6,497,916; 6,251 ,964; 6,225,431 ; 6,087,462; 6,083,257; 5,739,237; 5,739,236; 5,705,583; 5,648,442; 5,645,883; 5,556,710; 5,496,581; 4,689,386; 6,214,115; 6,090,901 ; 6,599,448; 6,054,504; 4,987,182; 4,847,324; and 4,642,267; U.S. Patent Application Publication Nos. 2002/0146581, 2003/0129130, 2003/0129130, 2001/0026834; 2003/0190420; 2001/0000785; 2003/0059631 ; 2003/0190405; 2002/0146581 ; 2003/020399; 2001/0026834; 2003/0190420; 2001/0000785; 2003/0059631; 2003/0190405; and 2003/020399; and PCT Publication Nos. WO 02/055121; WO 01/57048; WO 01/52915; and WO 01/01957.
Representative examples of medical devices which may be coated using the compositions of the invention and are described in more detail below include vascular stents, gastrointestinal stents, tracheal/bronchial stents, genital-urinary stents, ENT stents, intra-articular implants, intraocular lenses, implants for hypertrophic scars and keloids, vascular grafts, anastomotic connector devices, implantable sensors, implantable pumps, implantable electrical devices, such as implantable neurostimulators, implantable electrical leads, surgical adhesion barriers, glaucoma drainage devices, film or mesh, prosthetic heart valves, tympanostomy tubes, penile implants, endotracheal and tracheostomy tubes, peritoneal dialysis catheters, intracranial pressure monitors, vena cava filters, central venous cathethers (CVCs), ventricular assist devices (e.g., LVAD's), spinal prostheses, urinary (Foley) catheters, prosthetic bladder sphincters, orthopedic implants, and gastrointestinal drainage tubes. There are numerous medical devices where the occurrence of a fibrotic reaction will adversely affect the functioning of the device or the biological problem for which the device was implanted or used. Representative examples of implants or devices that can be coated with or otherwise constructed to contain and/or release the therapeutic agents provided herein include cardiovascular devices (e.g., implantable venous catheters, venous ports, tunneled venous catheters, chronic infusion lines or ports, including hepatic artery infusion catheters, pacemakers and pacemaker leads, implantable defibrillators; neurologic/neurosurgical devices (e.g., ventricular peritoneal shunts, ventricular atrial shunts, dural patches and implants to prevent epidural fibrosis post-laminectomy, devices for continuous subarachnoid infusions); gastrointestinal devices (e.g., chronic indwelling catheters, feeding tubes, portosystemic shunts, shunts for ascites, peritoneal implants for drug delivery, peritoneal dialysis catheters, and suspensions or solid implants to prevent surgical adhesions); genitourinary devices (e.g., uterine implants, including intrauterine devices (IUDs) and devices to prevent endometrial hyperplasia, fallopian tubal implants, including reversible sterilization devices, fallopian tubal stents, ureteric stents, chronic indwelling catheters, bladder augmentations, or wraps or splints for vasovasostomy, central venous catheters, urinary catheters; prosthetic heart valves, vascular grafts, ophthalmologic implants (e.g., multino implants and other implants for neovascular glaucoma, drug eluting contact lenses for pterygiums, splints for failed dacrocystalrhinostomy, drug eluting contact lenses for corneal neovascularity, implants for diabetic retinopathy, drug eluting contact lenses for high risk corneal transplants); otolaryngology devices (e.g., ossicular implants, Eustachian tube splints or stents for glue ear or chronic otitis as an alternative to transtempanic drains); catheter cuffs and orthopedic implants (e.g., cemented orthopedic prostheses).
Other examples of implants include drainage tubes, biliary T- tubes, clips, sutures, braids, meshes (e.g., hernia meshes, tissue support meshes), barriers (for the prevention of adhesions), anastomotic devices, anastomotic connectors, ventrical assist devices (e.g., LVAD's), artificial hearts, artificial joints, conduits, irrigation fluids, packing agents, stents, staples, inferior vena cava filters, pumps (e.g., for the delivery of therapeutics), hemostatic implants (e.g., sponges), tissue fillers, surgical adhesion barriers (e.g., INTERCEED, degradable polyester films (e.g., PLLA/PDLLA), CMC/PEO association complexes (e.g., OXIPLEX from Fziomed), hyaluronic acid/CMC films (e.g., SEPRAFILM from Genzyme Corporation), bone grafts, skin grafts, tissue sealants, intrauterine devices (IUD), ligatures, titanium implants (particularly for use in dental applications), chest tubes, nasogastric tubes, percutaneous feeding tubes, colostomy devices, bone wax, and Penrose drains, hair plugs, ear rings, nose rings, and other piercing-associated implants, as well as anaesthetic solutions.
The coating of fibrosis-inhibiting agent(s) onto or incorporation of a fibrosis-inhibiting agent(s) into medical devices provides a solution to the clinical problems that can be encountered with these devices. Alternatively, or additional, compositions that comprise anti-scarring agents can be infiltrated in to the space or onto tissue surrounding the area where medical devices are implanted either before, during or after implantation of the devices.
Described below are examples of medical devices whose functioning can be improved by the use of a fibrosis-inhibiting agent as well as methods for incorporating fibrosis-inhibiting agents into or onto these devices and methods for using such devices. Intravascular Devices
The present invention provides for the combination of an anti- scarring agent and an intravascular device. "Intravascular devices" refers to devices that are implanted at least partially within the vasculature (e.g., blood vessels). Examples of intravascular devices that can be used to deliver anti-scarring agents to the desired location include, e.g., catheters, balloon catheters, balloons, stents, covered stents, stent grafts, anastomotic connectors, and guidewires.
In one aspect, the present invention provides for the combination of an anti-scarring agent or a composition comprising an anti- scarring agent and an intravascular stent.
"Stent" refers to devices comprising a cylindrical tube (composed of a metal, textile, non-degradable or degradable polymer, and/or other suitable material (such as biological tissue) which maintains the flow of blood from one portion of a blood vessel to another. In one aspect, a stent is an endovascular scaffolding which maintains the lumen of a body passageway (e.g., an artery) and allows bloodflow. Representative examples of stents that can benefit from being coated with or having incorporated therein, a fibrosis-inhibiting agent include vascular stents, such as coronary stents, peripheral stents, and covered stents.
Stents that can be used in the present invention include metallic stents, polymeric stents, biodegradable stents and covered stents. Stents may be self-expandable or balloon-expandable, composed of a variety of metal compounds and/or polymeric materials, fabricated in innumerable designs, used in coronary or peripheral vessels, composed of degradable and/or nondegradable components, fully or partially covered with vascular graft materials (so called "covered stents") or "sleeves", and can be bare metal or drug-eluting.
Stents may be comprise a metal or metal alloy such as stainless steel, spring tempered stainless steel, stainless steel alloys, gold, platinum, super elastic alloys, cobalt-chromium alloys and other cobalt- containing alloys (including ELGILOY (Combined Metals of Chicago, Grove Village, IL), PHYNOX (Alloy Wire International, United Kingdom) and CONICHROME (Carpenter Technology Corporation, Wyomissing, PA)), titanium-containing alloys, platinum-tungsten alloys, nickel-containing alloys, nickel-titanium alloys (including nitinol), malleable metals (including tantalum); a composite material or a clad composite material and/or other functionally equivalent materials; and/or a polymeric (non-biodegradable or biodegradable) material. Representative examples of polymers that may be included in the stent construction include polyethylene, polypropylene, polyurethanes, polyesters, such as polyethylene terephthalate (e.g., DACRON or MYLAR (E. I. DuPont De Nemours and Company, Wilmington, DE)), polyamides, polyaramids (e.g., KEVLAR from E.I. DuPont De Nemours and Company), polyfluorocarbons such as poly(tetrafluoroethylene with and without copolymerized hexafluoropropylene) (available, e.g., under the trade name TEFLON (E. I. DuPont De Nemours and Company), silk, as well as the mixtures, blends and copolymers of these polymers. Stents also may be made with engineering plastics, such as thermotropic liquid crystal polymers (LCP), such as those formed from p,p'-dihydroxy-polynuclear- aromatics or dicarboxy-polynuclear-aromatics.
Further types of stents that can be used with the described therapeutic agents are described, e.g., in PCT Publication No. WO 01/01957 and U.S. Patent Nos. 6,165, 210; 6,099,561; 6,071 ,305; 6,063,101 ; 5,997,468; 5,980,551; 5,980,566; 5,972,027; 5,968,092; 5,951 ,586; 5,893,840; 5,891,108; 5,851 ,231; 5,843,172; 5,837,008; 5,766,237; 5,769,883; 5,735,811; 5,700,286; 5,683,448; 5,679,400; 5,665,115; 5,649,977; 5,637,113; 5,591 ,227; 5,551 ,954; 5,545,208; 5,500,013; 5,464,450; 5,419,760; 5,411 ,550; 5,342,348; 5,286,254; and 5,163,952. Removable drug-eluting stents are described, e.g., in Lambert, T. (1993) J. Am. Coll. Cardiol.: 21: 483A. Moreover, the stent may be adapted to release the desired agent at only the distal ends, or along the entire body of the stent. Balloon over stent devices, such as are described in Wilensky, R.L (1993) J. Am. Coll. Cardiol.: 21: 185A, also are suitable for local delivery of a fibrosing agent to a treatment site.
In addition to using the more traditional stents, stents that are specifically designed for drug delivery can be used. Examples of these specialized drug delivery stents as well as traditional stents include those from Conor Medsystems (Palo Alto, CA) (e.g., U.S. Patent. Nos. 6,527,799; 6,293,967; 6,290,673; 6,241,762; U.S. Patent Application Publication Nos. 2003/0199970 and 2003/0167085; and PCT Publication No. WO 03/015664).
Examples of intravascular stents, which may be combined with one or more therapeutic agents according to the present invention, include commercially available products. The stent may be self-expanding or balloon expandable (e.g., STRECKER stent by Medi-Tech/Boston Scientific Corporation), or implanted by a change in temperature (e.g., nitinol stent). Self-expanding stents that can be used include the coronary WALLSTENT and the SCIMED RADIUS stent from Boston Scientific Corporation (Natick, MA) and the GIANTURCO stents from Cook Group, Inc. (Bloomington, IN). Examples of balloon expandable stents that can be used include the CROSSFLEX stent, BX-VELOCITY stent and the PALMAZ-SCHATZ crown and spiral stents from Cordis Corporation (Miami Lakes, FL), the V-FLEX PLUS stent by Cook Group, Inc., the NIR, EXPRESS and LIBRERTE stents from Boston Scientific Corporation, the ACS MULTILINK, MULTILINK PENTA, SPIRIT, and CHAMPION stents from Guidant Corporation, and the Coronary Stent S670 and S7 by Medtronic, Inc. (Minneapolis, MN).
Other examples of stents that can be combined with a fibrosing agent in accordance with the invention include those from Boston Scientific Corporation, (e.g., the drug-eluting TAXUS EXPRESS2 drug- Eluting Coronary Stent System; over the wire stent stents such as the Express2 Coronary Stent System and NIR Elite OTW Stent System; rapid exchange stents such as the EXPRESS2 Coronary Stent System and the NIR ELITE MONORAIL Stent System; and self-expanding stents such as the MAGIC WALLSTENT Stent System and RADIUS Self Expanding Stent); Medtronic, Inc. (Minneapolis, MN) (e.g., DRIVER ABT578-eluting stent, DRIVER ZIPPER MX Multi-Exchange Coronary Stent System and the DRIVER Over-the-Wire Coronary Stent System; the S7 ZIPPER MX Multi- Exchange Coronary Stent System; S7, S670, S660, and BESTENT2 with Discrete Technology Over-the-Wire Coronary Stent System); Guidant Corporation (e.g., cobalt chromium stents such as the MULTI-LINK VISION Coronary Stent System; MULTI-LINK ZETA Coronary Stent System; MULTI- LINK PIXEL Coronary Stent System; MULTI-LINK ULTRA Coronary Stent System; and the MULTI-LINK FRONTIER); Johnson & Johnson/Cordis Corporation (e.g., CYPHER sirolimus-eluting Stent; PALMAZ-SCHATZ Balloon Expandable Stent; and S.M.A.R.T. Stents); Abbott Vascular (Redwood City, California) (e.g., MATRIX LO Stent; TRIMAXX Stent; and DEXAMET stent); Conor Medsystems (Menlo Park, California) (e.g., MEDSTENT and COSTAR stent); AMG GmbH (Germany) (e.g., PICO Elite stent); Biosensors International (Singapore) (e.g., MATRIX stent, CHAMPION Stent (formerly the S-STENT), and CHALLENGE Stent); Biotronik (Switzerland) (e.g., MAGIC AMS stent); Clearstream Technologies (Ireland) (e.g., CLEARFLEX stent); Cook Inc. (Bloomington, Indiana) (e.g., V-FLEX PLUS stent, ZILVER PTX self-expanding vascular stent coating, LOGIX PTX stent (in development); Devax (e.g., AXXESS stent) (Irvine, CA); DISA Vascular (Pty) Ltd (South Africa) (e.g., CHROMOFLEX Stent, S- FLEX Stent, S-FLEX Micro Stent, and TAXOCHROME DES); lntek Technology (Baar, Switzerland) (e.g., APOLLO stent); Orbus Medical Technologies (Hoevelaken, The Netherlands) (e.g., GENOUS); Sorin Biomedica (Saluggia, Italy) (e.g., JANUS and CARBOSTENT); and stents from Bard/Angiomed GmbH Medizintechnik KG (Murray Hill, NJ), and Blue Medical Supply & Equipment (Mariettta, GA), Aachen Resonance GmbH (Germany); Eucatech AG (Germany), Eurocor GmbH (Bonn, Gemany), Prot, Goodman, Terumo (Japan), Translumina GmbH (Germany), MIV Therapeutics (Canada), Occam International B.V. (Eindhoven, The Netherlands), Sahajanand Medical Technologies PVT LTD. (India); AVI Biopharma/Medtronic/ Interventional Technologies (Portland, OR) (e.g., RESTEN NG-coated stent); and Jomed (e.g., FLEXMASTER drug-eluting stent) (Sweden).
Generally, stents are inserted in a similar fashion regardless of the site or the disease being treated. Briefly, a preinsertion examination, usually a diagnostic imaging procedure, endoscopy, or direct visualization at the time of surgery, is generally first performed in order to determine the appropriate positioning for stent insertion. A guidewire is then advanced through the lesion or proposed site of insertion, and over this is passed a delivery catheter which allows a stent in its collapsed form to be inserted. Intravascular stents may be inserted into an artery such as the femoral artery in the groin and advanced through the circulation under radiological guidance until they reach the anatomical location of the plaque in the coronary or peripheral circulation. Typically, stents are capable of being compressed, so that they can be inserted through tiny cavities via small catheters, and then expanded to a larger diameter once they are at the desired location. The delivery catheter then is removed, leaving the stent standing on its own as a scaffold. Once expanded, the stent physically forces the walls of the passageway apart and holds them open. A post insertion examination, usually an x-ray, is often utilized to confirm appropriate positioning.
Stents are typically maneuvered into place under, radiologic or direct visual control, taking particular care to place the stent precisely within the vessel being treated. In certain aspects, the stent can further include a radio-opaque, echogenic material, or MRI responsive material (e.g., MRI contrast agent) to aid in visualization of the device under ultrasound, fluoroscopy and/or magnetic resonance imaging. The radio-opaque or MRI visible material may be in the form of one or more markers (e.g., bands of material that are disposed on either end of the stent) that may be used to orient and guide the device during the implantation procedure.
In another aspect, the present invention provides for the combination of an anti-scarring agent or a composition comprising an anti- scarring agent and an intravascular catheter.
"Intravascular Catheter" refers to any intravascular catheter containing one or more lumens suitable for the delivery of aqueous, microparticulate, fluid, or gel formulations into the bloodstream or into the vascular wall. These formulations may contain a biologically active agent (e.g., an anti-scarring agent). Numerous intravascular catheters have been described for direct, site-specific drug delivery (e.g., microinjector catheters, catheters placed within or immediately adjacent to the target tissue), regional drug delivery (i.e., catheters placed in an artery that supplies the target organ or tissue), or systemic drug delivery (i.e., intra-arterial and intravenous catheters placed in the peripheral circulation). For example, catheters and balloon catheters can deliver anti-fibrosing agents from an end orifice, through one or more side ports, through a microporous outer structure, or through direct injection into the desired tissue or vascular location.
A variety of catheters are available for regional or localized arterial drug-delivery. Intravascular balloon and non-balloon catheters for delivering drugs are described, for example, in U.S. Patent Nos. 5,180,366; 5,171,217; 5,049,132; 5,021,044; 6,592,568; 5,304,121; 5,295,962; 5,286,254; 5,254,089; 5,112,305; PCT Publication Nos WO 93/08866, WO 92/11890, and WO 92/11895; and Riessen et at. (1994) JACC 23: 1234- 1244, Kandarpa K. (2000) J. Vase. Interv. Radio. 11 (suppl.): 419-423, and Yang, X. (2003) Imaging of Vascular Gene Therapy 228(1): 36-49.
Representative examples of drug delivery catheters include balloon catheters, such as the CHANNEL and TRANSPORT balloon catheters from Boston Scientific Corporation (Natick, MA) and Stack Perfusion Coronary Dilitation catheters from Advanced Cardiovascular Systems, Inc. (Santa Clara, CA). Other examples of drug delivery catheters include infusion catheters, such as the CRESCENDO coronary infusion catheter available from Cordis Corporation (Miami Lakes, FL), the Cragg- McNamara Valved Infusion Catheter available from Microtherapeutics, Inc. (San Clemente, CA), the DISPATCH catheter from Boston Scientific Corporation, the GALILEO Centering Catheter from Guidant Corporation (Houston, TX), and infusion sleeve catheters, such as the INFUSASLEEVE catheter from LocalMed, Inc. (Sunnyvale, CA). Infusion sleeve catheters are described in, e.g., U.S. Patent Nos. 5,318,531 ; 5,336,178; 5,279,565; 5,364,356; 5,772,629; 5,810,767; and 5,941 ,868. Catheters that mechanically or electrically enhance drug delivery include, for example, pressure driven catheters (e.g., needle injection catheters having injector ports, such as the INFILTRATOR catheter available from Interventional Technologies, Inc. (San Diego, CA)) (see, e.g., U.S. Patent No. 5,354,279) and ultrasonically assisted (phonophoresis) and iontophoresis catheters (see, e.g., Singh, J., et al. (1989) Drug Des. Deliv.: 4: 1-12 and U.S. Patent . Nos. 5,362,309; 5,318,014; 5,315,998; 5,304,120; 5,282,785; and 5,267,985).
In one aspect, the present invention provides for the combination of an anti-scarring agent or a composition comprising an anti- scarring agent and a drug delivery balloon.
"Drug-Delivery Balloon" refers to an intra-arterial balloon (typically based upon percutaneous angioplasty balloons) suitable for insertion into a peripheral artery (typically the femoral artery) and manipulated via a catheter to the treatment site (either in the coronary or peripheral circulation). Numerous drug delivery balloons have been developed for local delivery of therapeutic agents to the arterial wall such as "sweaty balloons," "channel balloons," "microinjector balloons," "double balloons," "spiral balloons" and other specialized drug-delivery balloons. Other examples of balloons include BHP balloons and Transurethral and Radiofrequency Needle Ablation (TUNA or RFNA)) balloons for prostate applications.
In addition, numerous drug delivery balloons have been developed for local delivery of therapeutic agents to the arterial wall. Representative examples of drug delivery balloons include porous (WOLINSKY) balloons, available from Advanced Polymers (Salem, NH), described in, e.g., U.S. Patent No. 5,087,244. Microporous and macroporous balloons (Ae., "sweaty balloons") for use in infusion catheters are described in, e.g., Lambert, CR. etal. (1992) Circ. Res. 71: 27-33. Other types of specialized drug delivery balloons include hydrogel coated balloons (e.g., ULTRATHIN GLIDES from Boston Scientific Corporation) (see, e.g., Fram, D.B. et al. (1992) Circulation: 86 Suppl. I: 1-380), "channel balloons" (see, e.g., U.S. Patent Nos. 5,860,954; 5,843,033; and 5,254,089, and Hong, M.K., et al. (1992) Circulation: 86 Suppl. 1: 1-380), "microinjector balloons" (see, e.g., U.S. Patent Nos. 5,681,281 and 5,746,716), "double balloons," described in, e.g., U.S. Patent No. 6,544,221 , and double-layer channeled perfusion balloons (such as the REMEDY balloon from Boston Scientific Corportion), and "spiral balloons" (see, e.g., U.S. Patent Nos. 6,527,739 and 6,605,056). Drug delivery catheters that include helical (i.e., spiral) balloons are described in, e.g., U.S. Patent Nos. 6,190,356; 5,279,546; 5236424, 5,226,888; 5,181,911 ; 4,824,436; and 4,636,195.
The balloon catheter systems that can be used include systems in which the balloon can be inflated at the desired location the desired fibrosis-inducing agents can be delivered through holes that are located in the balloon wall. Other balloon catheters that can be used include systems that have a plurality of holes that are located between two balloons. The system can be guided into the desired location such that the inflatable balloon components are located on either side of the specific site that is to be treated. The balloons can then be inflated to isolate the treatment area. The compositions containing the fibrosing agent are then injected into the isolated area through the plurality of holes between the two balloons. Representative examples of these types of drug delivery balloons are described in U.S. Patent. Nos. 5,087,244, 6,623,452, 5,397,307, 4,636,195 and 4,994,033.
The compositions of the invention can be delivered using a catheter that has the ability to enhance uptake or efficacy of the compositions of the invention. The stimulus for enhanced uptake can include the use of heat, the use of cooling, the use of electrical fields or the use of radiation (e.g., ultraviolet light, visible light, infrared, microwaves, ultrasound or X-rays). Further Representative examples of catheter systems that can be used are described in U.S. Patent. Nos. 5,362,309 and 6,623,444; U.S. Patent Application Publication Nos. 2002/0138036 and 2002/0068869; and PCT Publication Nos. WO 01/15771; WO 94/05361 ; WO 96/04955 and WO 96/22111.
In another aspect of the invention, the compositions of the inventions can be delivered into the treatment site and/or into the tissue surrounding the treatment site by using catheter systems that have one or more injectors that can penetrate the surrounding tissue. Following insertion into the appropriate vessel, the catheter can be maneuvered into the desired position such that the injectors are aligned with or adjacent to the tissue. The injector(s) are inserted into the desired location, for example by direct insertion into the tissue, by inflating the balloon or mechanical rotation of the injector, and the composition of the invention is injected into the desired location. Representative examples of catheters that can be used for this application are described in and U.S. Patent Application Publication No. 2002/0082594 and U.S. Patent. Nos. 6,443,949; 6,488,659; 6,569,144; 5,609,151; 5,385,148; 5,551,427; 5,746,716; 5,681,281 ; and 5,713,863.
In another aspect of the invention, the catheter may be adapted to deliver a thermoreversible polymer composition. For the site- specific delivery of these materials, a catheter delivery system has the ability to either heat the composition to above body temperature or to cool the composition to below body temperature such that the composition remains in a fluent state within the catheter delivery system. The catheter delivery system can be guided to the desired location and the composition of the invention can be delivered to the surface of the surrounding tissue or can be injected directly into the surrounding tissue. A representative example of a catheter delivery system for direct injection of a thermoreversible material is described in U.S. Patent. No. 6,488,659. Representative examples of catheter delivery systems that can deliver the thermoreversible compositions to the surface of the vessel are described in U.S. Patent. Nos. 6,443,941 ; 6,290,729; 5,947,977; 5,800,538; and 5,749,922.
In another aspect, the present invention provides for the combination of an anti-scarring agent or a composition comprising an anti- scarring agent and an anastomotic connector device.
"Anasomotic connector device" refers to any vascular device that mechanizes the creation of a vascular anastomosis (i.e., artery-to- artery, vein-to-artery, artery-to-vein, artery-to-synthetic graft, synthetic graft- to-artery, vein-to-synthetic graft or synthetic graft-to-vein anastomosis) without the manual suturing that is typically done in the creation of an anastomosis. The term also refers to anastomotic connector devices (described below), designed to produce a facilitated semiautomatic vascular anastomosis without the use of suture and reduce connection time substantially (often to several seconds), where there are numerous types and designs of such devices. The term also refers to devices which facilitate attachment of a vascular graft to an aperture or orifice (e.g., in the side or at the end of a vessel) in a target vessel. Anastomotic connector devices may be anchored to the outside of a blood vessel, and/or into the wall of a blood vessel (e.g., into the adventitial, intramural, or intimal layer of the tissue), and/or a portion of the device may reside within the lumen of the vessel.
Anastomotic connector devices also may be used to create new flow from one structure to another through a channel or diversionary shunt. Accordingly, such devices (also referred to herein as "bypass devices") typically include at least one tubular structure, wherein a tubular structure defines a lumen. Anastomotic connector devices may include one tubular structure or a plurality of tubular structures through which blood can flow. At least a portion of the tubular structure resides external to a blood vessel (i.e., extravascular) to provide a diversionary passageway. A portion of the device also may reside within the lumen and/or within the tissue of the blood vessel.
Examples of anastomotic connector devices are described in co-pending application entitled, "Anastomotic Connector Devices", filed May 23, 2003 (U.S. Ser. No. 60/473,185). Representative examples of anastomotic connector devices include, without limitation, vascular clips, vascular sutures, vascular staples, vascular clamps, suturing devices, anastomotic coupling devices (i.e., anastomotic couplers), including couplers that include tubular segments for carrying blood, anastomotic rings, and percutaneous in situ coronary artery bypass (PISCAB and PICVA) devices. Broadly, anastomotic connector devices may be classified into three categories: (1) automated and modified suturing methods and devices, (2) micromechanical devices, and (3) anastomotic coupling devices.
(1) Automated and Modified Suturing Methods and Devices Automated sutures and modified suturing methods generally facilitate the rapid deployment of multiple sutures, usually in a single step, and eliminate the need for knot tying or the use of aortic side-biting clamps. Suturing devices include those devices that are adapted to be minimally invasive such that anastomoses are formed between vascular conduits and hollow organ structures by applying sutures or other surgical fasteners through device ports or other small openings. With these devices, sutures and other fasteners are applied in a relatively quick and automated manner within bodily areas that have limited access. By using minimally invasive means for establishing anastomoses, there is less blood loss and there is no need to temporarily stop the flow of blood distal to the operating site. For example, the suturing device may be composed of a shaft-supported vascular conduit that is adapted for anastomosis and a collar that is slideable on the shaft configured to hold a plurality of needles and sutures that passes through the vascular conduit. See, e.g., U.S. Patent No. 6,709,441. The suturing device may be composed of a carrier portion for inserting Λgraft, arm portions that extend to support the graft into position, and a needle assembly adapted to retain and advance coil fasteners into engagement with the vessel wall and the graft flange to complete the anastomosis. See, e.g., U.S. Patent No. 6,709,442. The suturing device may include two oblong interlinked members that include a split bush adapted for suturing (e.g., U.S. Patent No. 4,350,160).
One representative example of a suturing device is the HEARTFLOW device, made by Perclose-Abbott Labs, Redwood City, CA (see generally, U.S. Patent Nos. 6,358,258, 6,355,050, 6,190,396, and 6,036,699, and PCT Publication No. WO 01/19257)
The nitinol U-CLIP suture clip device by Coalescent Surgical (Sunnyvale, CA) consists of a self-closing nitinol wire loop attached to a flexible member and a needle with a quick release mechanism. This device facilitates the construction of anastomosis by simplifying suture management and eliminating knot tying (see generally, U.S. Patent Nos. 6,074,401 and 6,149,658, and PCT Publication Nos. WO 99/62406, WO 99/62409, WO 00/59380, WO 01/17441).
The ENCLOSE Anastomotic Assist Device (Novare Surgical Systems, Cupertino, CA) allows a surgeon to create a sutured anastomosis using standard suturing techniques but without the use of a partial occluding side-biting aortic clamp, avoiding aortic wall distortion (see U.S. Patent Nos. 6,312,445 and 6,165,186).
In one aspect, automated and modified suturing methods and devices can deliver a surgical fastener (e.g., a suture or suture clip) that comprises an anti-scarring agent. In another aspect, automated and modified suturing methods and devices can deliver a vascular graft that comprises an anti-scarring agent to complete an anastomosis.
(2) Micromechanical devices
Micromechanical devices are used to create an anastomosis and/or secure a graft vessel to the site of an anastomosis. Representative examples of micromechanical devices include staples (either penetrating or non-penetrating) and clips.
Anastomotic staple and clip devices may take a variety of forms and may be made from different types of materials. For example, staples and clips may be formed of a metal or metal alloy, such as titanium, nickel-titanium alloy, or stainless steel, or a polymeric material, such as silicone, poly(urethane), rubber, or a thermoplastic elastomer.
The polymeric material may be an absorbable or biodegradable material designed to dissolve after completion of the anastomosis. Biodegradable polymers include, for example, homopolymers and copolymers that comprise one or more of the monomers selected from lactide, lactic acid, glycolide, glycolic acid, ε-caprolactone, gamma- caprolactone, hydroxyvaleric acid, hydroxybutyric acid, beta-butyrolactone, gamma-butyrolactone, gamma-valerolactone, γ-decanolactone, δ- decanolactone, trimethylene carbonate, 1 ,4-dioxane-2-one or 1 ,5-dioxepan- 2one.
A variety of devices for guiding staples and clips into position also have been described.
One manufacturer of non-penetrating staples for use in the creation of anastomosis is United States Surgical Corp. (Norwalk, CT). The VCS system (Autosuture) is an automatic stapling device that applies nonpenetrating, titanium vascular clips which are usually used in an interrupted fashion to evert tissue edges with high compressive forces. (See, e.g., U.S. Patent Nos. 6,440,146, 6,391 ,039, 6,024,748, 5,833,698, 5,799,857, 5,779,718, 5,725,538, 5,725,537, 5,720,756, 5,360,154, 5,193,731 , and 5,005,749 for the description of anastomotic connector devices made by U.S. Surgical).
An anastomotic clip may be composed of a shape memory material, such as nitinol, which is self-closing between an open U-shaped configuration and a closed configuration. See, e.g., U.S. Patent No. 6,641,593. The anastomotic clip may be composed of a wire having a shape memory that defines a closed configuration which may be substantially spiral-shaped and having a needle that may be releasably attached to the clip. See, e.g., U.S. Patent No. 6,551 ,332. Other anastomotic clips are described in, e.g., U.S. Patent Nos. 6,461 ,365; and 6,514,265.
Automatic stapling devices are also made by Bypass/Ethicon, Inc. (Somerville, NJ) and are described in, e.g., U.S. Patent Nos. 6,193,129; 5,632,433; 5,609,285; 5,533,661 ; 5,439,156; 5,350,104; 5,333,773; 5,312,024; 5,292,053; 5,285,945; 5,275,322; 5,271,544; 5,271,543 and 5,205,459 and WO 03/02016. Resorbable surgical staples that include a polymer blend that is rich in glycolide (i.e., 65 to 85 weight % polymerized glycolide) are described in, e.g., U.S. Patent No. 4,741 ,337 and 4,889,119. Surgical staples made from a blend of lactide/glycolide-copolymer and poly(p-dioxanone) are described in U.S. Patent No. 4,646,741. Other types of stapling devices are described in, e.g., U.S. Patent Nos. 5,234,447; 5,904,697 and 6,565,582; and U.S. Publication No. 2002/0185517A1.
In another aspect, the micromechanical device may be an anastomotic clip. For example, an anastomotic clip may be composed of a shape memory material, such as nitinol, which is self-closing between an open U-shaped configuration and a closed configuration. See, e.g., U.S. Patent No. 6,641,593. The anastomotic clip may be composed of a wire having a shape memory that defines a closed configuration which may be substantially spiral-shaped and having a needle that may be releasably attached to the clip. See, e.g., U.S. Patent No. 6,551 ,332. Other anastomotic clips are described in, e.g., U.S. Patent Nos. 6,461,365; 6,187,019; and 6,514,265.
In one aspect, the present invention provides for the combination of a micromechanical anastomotic device (e.g., a staple or a clip) and an anti-scarring agent.
(3) Anastomotic Coupling Devices
Anastomotic coupling devices may be used to connect a first blood vessel to a second vessel, either with or without a graft vessel, for completion of an anastomosis. In one aspect, anastomotic coupling devices facilitate automated attachment of a graft or vessel to an aperture or orifice (e.g., in the side or at the end of a vessel) in a target vessel without the use of sutures or staples. In another aspect, the anastomotic coupling device comprises a tubular structure defining a lumen through which blood may flow (described below).
Anastomotic coupling devices that facilitate automated attachment of a graft or vessel to an aperture or orifice in a target vessel may take a variety of forms and may be made from a variety of materials. Typically, such devices are made of a biocompatible material, such as a polymer or a metal or metal alloy. For example, the device may be formed from a synthetic material, such as a fluoropolymer, such as expanded poly(tetrafluoroethylene) (ePTFE) (ePTFE) sold under the trade name GORE-TEX available from W.L. Gore & Associates, Inc. or fluorinated ethylene propylene (FEP), a polyurethane, polyethylene, polyamide (nylon), silicone, polypropylene, polysulfone, or a polyester.
Anastomotic coupling devices may include an absorbable or biodegradable material designed to dissolve after completion of the anastomosis. Biodegradable polymers include, for example, homopolymers and copolymers that comprise one or more of the monomers selected from lactide, lactic acid, glycolide, glycolic acid, ε-caprolactone, gamma- caprolactone, hydroxyvaleric acid, hydroxybutyric acid, beta-butyrolactone, gamma-butyrolactone, gamma-valerolactone, γ-decanolactone, δ- decanolactone, trimethylene carbonate, 1 ,4-dioxane-2-one or 1 ,5-dioxepan- 2one.
The device may include a metal or metal alloy (e.g., nitinol, stainless steel, titanium, iron, nickel, nickel-titanium, cobalt, platinum, tungsten, tantalum, silver, gold, molybdenum, chromium, and chrome), or a combination of a metal and a polymer.
The device may be anchored to the outside of a vessel, within the tissue that surrounds the lumen of a blood vessel, and/or a portion of the device may reside within the lumen of the vessel.
In one aspect, the anastomotic coupler may be an artificially formed aperture connector that is placed in the side wall of the target vessel so that the tubular graft conduit may be extended from the target vessel. The connector may include a plurality of tissue-piercing members and retention fingers disposed in a concentric annular array which may be passed through the side wall of the tubular graft conduit for securing and retaining the graft to the connector in a fluid-tight configuration. See, e.g., U.S. Patent No. 6,702,829 and 6,699,256.
In another aspect, the anastomotic coupler may be in the form of a frame. For example, the frame may be configured to be deformable and scissor-shaped such that spreading members are moveable to secure a graft vessel upon insertion into a target vessel. See, e.g., U.S. Patent No. 6,179,849.
In another aspect, the anastomotic coupler may be a ring-like device that is used as an anastomotic interface between a lumen of a graft and an opening in a lumen of a target vessel. For example, the anastomotic ring may be composed of stainless steel alloy, titanium alloy, or cobalt alloy and have a flange with an expandable diameter. See, e.g., U.S. Patent No. 6,699,257. Anastomosis rings are also described in, e.g., U.S. Patent No. 6,248,117.
In another aspect, the anastomotic coupler is resorbable. Resorbable anastomotic coupling devices may include, for example, a polymeric blend that is rich in glycolide (i.e., 65 to 85 weight % polymerized glycolide) (see, e.g., U.S. Patent No. 4,741 ,337 and 4,889,119) or a blend of lactide/glycolide-copolymer and poly(p-dioxanone) (see, e.g., U.S. Patent No. 4,646,741).
In another aspect, the anastomotic coupler includes a bioabsorbable, elastomeric material. Representative examples of elastomeric materials for use in resorbable devices are described in, e.g., U.S. Patent No. 5,468,253.
In another aspect, the anastomotic coupler may be used to connect a first blood vessel to a second vessel, either with or without a graft vessel. For example, the anastomotic coupler may be a device that serves to interconnect two vessels in a side-to-side anastomosis, such as when grafting two juxtaposed cardiac vessels. The anastomotic coupler may be configured as two partially opened cylindrical segments that are interconnected along the periphery by a flow opening whereby the device may be inserted in a minimally-invasive manner which then conforms to provide pressure against the interior wall when in the original configuration such that leakage is prevented. See, e.g., U.S. Patent Nos. 6,464,709; 6,458,140 and 6,251,116 and U.S. Application Publication No. 2003/0100920A1.
In another aspect, the anastomotic coupler may also be incorporated in the design of a vascular graft to eliminate the step of attaching the interface prior to deployment. For example, the anastomotic coupler may have a leading and rear petal for dilating the vessel opening during advancement, and a base which is configured for attachment to a graft while forming a seal with the opening of the vessel. See, e.g., U.S. Patent No. 6,702,828.
In another aspect, the anastomotic coupler may be in the form of a frame. For example, the anastomotic coupler may be composed of a deformable, scissor-shaped frame with spreading members that is inserted into a target vessel. See, e.g., U.S. Patent No. 6,179,849. In another aspect, the anastomotic coupling device may include a graft that incorporates fixation mechanisms (e.g., a collet or a grommet) at its opposite ends and a heating element to create a thermal bond between the graft and a blood vessel (see, e.g., U.S. Patent Nos. 6,652,544 and 6,293,955).
In another aspect, the anastomotic coupling device includes a compressible, expandable fitting for securing the ends of a bypass graft to two vessels. The fitting may be incorporated in the bypass graft design to eliminate the step of attaching the graft to the fitting prior to deployment (see, e.g., U.S. Patent No. 6,494,889).
In another aspect, the anastomotic coupling device includes a pair of coupling disc members for joining two vessels in an end-to-end or end-to-side fashion. One of the members includes hook members, while the other member has receptor cavities aligned with the hooks for locking everted tissue of the vessels together (see, e.g., U.S. Patent No. 4,523,592).
Representative examples of anastomotic connector devices of Bypass/Ethicon, Inc. are described in U.S. Application Publication Nos. US2002/0082625A1 and 2003/0100910A1 and U.S. Patent Nos. 6,036,703, 6,036,700, 6,015,416, and 5,346,501.
Other anastomotic coupling devices are those described in e.g., U.S. Patent No. 6,036,702; 6,508,822; 6,599,303; 6,673,084, 5,695,504; 6,569,173; 4,931 ,057; 5,868,763; 4,624,257; 4,917,090; 4,917,091 ; 5,697,943; 5,562,690; 5,454,825; 5,447,514; 5,437,684; 5,376,098; 6,652,542; 6,551 ,334; and 6,726,694 and U.S. Application Publication Nos. 2003/0120293A1 and 2004/0030348A1.
Anastomotic coupling devices may include proximal aortic connectors and distal coronary connectors. For example, aortic anastomotic connectors include devices such as the SYMMETRY Bypass Aortic Connector device made by St. Jude Medical, Inc. (Maple Grove, MN), which consists of an aortic cutter or hole punch assembly and a graft delivery system. The aortic hole punch is a cylindrical cutter with a barbed needle that provides an anchor and back pressure for the rotating cutter to core a round hole in the wall of the aorta. The graft delivery system is a radially expandable nitinol device that holds the vein graft with small hooks which pierce through vein graft wall. The graft is fixed to the aorta through use of an inner and outer ring of struts or flanges. This and other anastomotic connector devices by St. Jude are described in U.S. Patent Nos. 6,309,416, 6,302,905, 6,152,937, and PCT Publication Nos. WO 00/27312 and WO 00/27311.
The CORLINK Automated Anastomotic connector device, which is produced by the CardioVations division of Ethicon, Inc. (Johnson & Johnson, Somerville, NJ), uses a nitinol metal alloy fastener to connect the grafted vessel to the aorta. It consists of a central cylindrical body made of interconnected elliptical arches and two sets of several pins radiating from each end. The graft is loaded into a CORLINK insertion instrument and deployed to create an anastomosis in one step.
Further examples of anastomotic coupling devices include those made by Cardica (see, U.S. Patent Nos. 6,719,769; 6,419,681 and 6,537,287), Converge Medical (formerly Advanced Bypass Technologies), Onux Medical (see, e.g., PCT Publication No. WO 01/34037) and Ventrica, Menlo Park, CA (VENTRICA Magnetic Vascular Positioner) (see, e.g., U.S. Patent Nos. 6,719,768; 6,517,558 and 6,352,543).
As described above, an anastomotic coupling device may comprise a tubular structure defining a lumen through which blood may flow. These types of devices (also referred to herein as "bypass devices") can function as an artificial passageway or conduit for fluid communication between blood vessels and can be used to divert {i.e., shunt) blood from one part of a blood vessel (e.g., an artery) to another part of the same vessel, or to a second vessel (e.g., an artery or a vein) or to multiple vessels (e.g., a vein and an artery). In one aspect of the invention, the anastomotic device is a bypass device. Bypass devices may be used in a variety of end-to-end and end-to-side anastomotic procedures. The bypass device may be placed into a patient where it is desired to create a pathway between two or more vascular structures, or between two different parts of the same vascular structure. For example, bypass devices may be used to create a passageway which allows blood to flow around a blood vessel, such as an artery (e.g., coronary artery, carotid artery, or artery supplying the lower limb), which has become damaged or completely or partially obstructed. Bypass devices may be used in coronary artery bypass surgery to shunt blood from an artery, such as the aorta, to a portion of a coronary artery downstream from an occlusion in the artery.
Certain types of anastomotic coupling devices are configured to join two abutting vessels. The device can further include a tubular segment to shunt blood to another vessel. These types of connectors are often used for end-to-end anastomosis if a vessel is severed or injured.
Bypass devices include at least one tubular structure having a first end and a second end, which defines a single lumen through which blood can flow, or may include more than one tubular structure, defining multiple lumens through which blood can flow. The tubular structure includes an extravascular portion and may, optionally, include an intravascular portion. The extravascular portion resides external to the adventitial tissue of a blood vessel, whereas the intravascular portion may reside within the vessel lumen or within the intimal, medial, and/or adventitial tissue.
The configuration of the tubular segment may take a variety of forms. For example, the tubular portion may be generally straight, bent or curved (e.g., L-shaped or helical), tapered, branched (e.g., bifurcated or trifurcated), or may include a network of conduits through which blood may flow. Generally, straight or bent devices have a single lumen through which blood may flow, while branched conduits (e.g., generally T-shaped and Y- shaped devices) and conduit networks (described below) have two or more lumens through which blood may flow. A tubular structure may be in the form, for example, of a hollow cylinder and may or may not include a support structure, such as a mesh or porous framework. Depending on the procedure, the device may be biodegradable or non-biodegradable; expandable or rigid; metal and/or polymeric; and/or may include a shape- memory material (e.g., nitinol). In certain embodiments, the device may include a self-expanding stent structure.
Bypass devices typically are made of a biocompatible material. Any of the materials described above for other types of connectors may be used to make a bypass device, such as a synthetic or naturally-derived polymer, or a metal or metal alloy. For example, the device may be formed from a synthetic material, such as a fluoropolymer, such as expanded poly(tetrafluoroethylene) (ePTFE) or fluorinated ethylene propylene (FEP), a polyurethane, polyethylene, polyamide (nylon), silicone, polypropylene, polysulfone, or a polyester and/or a naturally derived material, such as collagen or a polysaccharide. The device may include a metal or metal alloy (e.g., nitinol, stainless steel, titanium, nickel, nickel-titanium, cobalt, platinum, iron, tungsten, tantalum, silver, gold, molybdenum, chromium and chrome), or a combination of a metal and a polymer. Other types of devices include a natural graft material (e.g., autologous vessel, homologous vessel, or xenograft), or a combination of a synthetic and a natural graft material. In another aspect, the bypass device may be formed of an absorbable or biodegradable material designed to dissolve after completion of the anastomosis (e.g., polylactide, polyglycolide, and copolymers of lactide and glycolide). In yet another aspect, demineralized bone may be used to provide a pliable tubular conduit (see, e.g., U.S. Patent No. 6,290,718).
The tubular structure(s) include a proximal end that may be configured for attachment to a proximal blood vessel and a distal end configured for attachment to a distal blood vessel. As described above, an anastomosis may be described as being either "proximal" or "distal" depending on its location relative to the vascular obstruction. The "proximal" anastomosis may be formed in a proximal blood vessel, and the "distal" anastomosis may be formed in a distal blood vessel, which may the same vessel or a different vessel than the proximal vessel. The terms "distal" and "proximal" may also be used to describe the direction that blood flows through a tubular structure from one vessel into another vessel. For example, blood may flow from a proximal vessel (e.g., the aorta) into a distal vessel, such as a coronary artery to bypass an obstruction in the coronary artery.
The tubular structure may be attached directly to a proximal or distal blood vessel. Alternatively, the bypass device may further include a graft vessel or be configured to receive a graft vessel, which can be connected to the same or a different blood vessel for completion of the anastomosis. Representative examples of graft vessels include, for example, vascular grafts or grafts used in hemodialysis applications (e.g., AV graft, AV shunt, or AV graft).
In one aspect, a tubular anastomotic coupler includes a proximal end that is attached to a proximal vessel and a distal end that is used to attach a bypass graft. The bypass graft can be secured to the distal vessel to complete the anastomosis. The direction of blood flow can be from the proximal blood vessel and into the proximal end of the tubular structure. Blood can exit through the distal end of the tubular structure and into the graft vessel.
In another aspect, the tubular anastomotic coupler includes a proximal end that is attached to a graft vessel, which is secured to the proximal blood vessel, and a distal end that is configured for attachment to a distal blood vessel. The direction of blood flow can be from the proximal vessel into the graft vessel and into the proximal end of the tubular structure. Blood can exit through the distal end of the tubular structure and into the distal vessel.
Anastomotic bypass devices may be anchored to a blood vessel in a variety of ways and may be attached to a blood vessel for the formation of an anastomosis with or without the use of sutures. Bypass devices may be attached to the outside of a blood vessel, and/or a portion of the device may be implanted into a vessel. For example, a portion of the implanted device may reside within the lumen of the vessel (i.e., endoluminally), and/or a portion of the implanted device may reside intravascularly (i.e., within the intimal, intramural, and/or adventitial tissue of the blood vessel). In one aspect, at least one of the tubular structures, or a portion thereof, may be inserted into the end of a vessel or into the side of a blood vessel. The device may be secured directly to the vessel using, for example, a fastener, such as sutures, staples, or clips and/or an adhesive. Bypass devices may include an interface to secure the conduit to a target vessel without the use of sutures. The interface may include means, such as, for example, hooks, barbs, pins, clamps, or a flange or lip for coupling the device to the site of an anastomosis.
Representative examples of anastomotic coupling devices that include at least one tubular portion include, without limitation, devices used for end-to-end anastomosis procedures (e.g., anastomotic stents and anastomotic sleeves) and end-to-side anastomosis procedures (e.g., single- lumen and multi-lumen bypass devices).
In one aspect of the invention, the anastomotic coupling device comprises a single tubular portion that may by used as a shunt to divert blood from a source vessel to a graft vessel (e.g., in an end-to-side anastomosis procedure). In one aspect, an end of the tubular portion may be connected directly or indirectly to a target vessel, as described above. The opposite end of the tubular portion may be attached to a graft vessel, where the graft vessel may be secured to a target vessel to complete the anastomosis.
The tubular portion(s) may be straight or may have a curved or bent shape (e.g., L-shaped or helical) and may be oriented orthogonally or at an angle relative to the vessel to which it is connected. In one aspect, the conduit may be secured into the site by, for example, a fastener, such as staples, clamps, or hooks, or by adhesives, radiofrequency sealing, or by other methods known to those skilled in the art.
In one aspect, the anastomotic coupling device may be, for example, a tubular metal braided graft with suture rings welded at the distal end to provide a means for securing in place to the target vessel. See, e.g., U.S. Patent No. 6,235,054. Other types of conduits that are secured into the site include, e.g., U.S. Patent Nos. 4,368,736 and 4,366,819.
In certain types of single-lumen coupling devices, the conduit terminates in a flange that resides within the lumen of the vessel. For example, the conduit may have a tubular body with a connector which has a plurality of extensions and is configured for disposition annularly within the inside of a tubular vessel. See, e.g., U.S. Patent No. 6,660,015. In other devices, the flange may be attached into or onto the surface of the adventitial tissue of the blood vessel.
Other types of single-lumen bypass devices are described, for example, in U.S. Patent Nos, 6,241 ,743; 6,428,550; 6,241 ,743; 6,428,550; 5,904,697; 5,290,298; 6,007,576; 6,361,559; 6,648,901, 4,931,057 and U.S. Application Publication Nos. 2004/0015180A1, 2003/0065344A1 , and 2002/0116018A1.
In one aspect of the invention, the anastomotic coupling device comprises more than one lumen through which blood may travel. Multi-lumen bypass devices may include two or more tubular portions configured to interconnect multiple (two or more) blood vessels. Multi-lumen coupling devices may be used in a variety of anastomosis procedures. For example, such devices may be used in coronary artery bypass graft (CABG) surgery to divert blood from an occluded proximal vessel (e.g., an artery) into one or more target (i.e., distal) vessels (e.g., an artery or vein).
In one aspect, at least one tubular portion may by used as a shunt for diverting blood between a source vessel and a target vessel. In another aspect, the device may be configured as an interface for securing a graft vessel to a target vessel for completion of an anastomosis. Depending on the procedure, the tubular arms may be of equal length and diameter or of unequal length and diameter and may include a tubular portion(s) that is expandable and/or includes a shape-memory material (e.g., nitinol). Furthermore, the tubular portions may be made of the same material or a different material.
In one aspect, one or more ends of a tubular portion may be inserted into the end or into the side of one or more blood vessels. In other embodiments, one or more tubular portions of the device may reside within the lumen of a blood or graft vessel. The device, optionally, may be secured to the blood vessel using a fastener or an adhesive, or another approach known to those skilled in the art.
At least one arm of the multi-lumen connector may be attached to a graft vessel. The graft vessel may be a synthetic graft, such as an ePTFE or polyester graft, or natural graft material (e.g., autologous vessel, homologous vessel, or xenograft), or a combination of a synthetic and a natural graft material. In certain embodiments, a graft vessel may be attached to an end of a tubular portion of the device, and a second graft vessel may be attached to the opposite end of the same tubular portion or to the end of another tubular portion. The graft vessel(s) may be further attached to a target vessel(s) for the completion of the anastomosis.
In one aspect, the device may include three or more tubular arms that extend from a junction site. For example, the multi-lumen device may be generally T-shaped or Y-shaped (i.e., having two or three lumens, respectively). For example, the multi-lumen device may be a T-shaped tubular graft connector having a longitudinal member that extends into the target vessel and a second section that is exterior to the vessel which provides a connection to an alternate tubular structure. See, e.g., U.S. Patent Nos. 6,152,945 and 5,972,017. Other multi-lumen devices are described in, (see, e.g., U.S. Patent Nos. 6,152,945; 6,451,033; 5,755,778; 5,922,022; 6,293,965; 6,517,558 and 6,626,914 and U.S. Publication No. 2004/0015180A1). In another aspect, the device may be a tube for bypassing blood flow directly from a portion of the heart (e.g., left ventricle) to a coronary artery. For example, the device may be a hollow tube that may be partially closable by a one-way valve in response to movement of the cardiac tissue during diastole while permitting blood flow during systole (see, e.g., U.S. Patent No. 6,641,610). The device may be an elongated rigid shunt body composed of a diversion tube having two apertures in which one may be disposed within the cyocardium of the left ventricle and the other may be disposed within the coronary artery (see, e.g., WO 00/15146 and U.S. Application Publication No. 2003/0055371A1). The device may be a valved, tubular apparatus that is L- or T-shaped which is adapted for insertion into the wall of the heart to provide blood communication from the heart to a coronary vessel (see, e.g., U.S. Patent No. 6,123,682).
In another aspect, the device may include a network of interconnected tubular conduits.. For example, the device may include two tubular portions that may be oriented generally axially or orthogonally relative to each other. See U.S. Patent No. 6,241 ,761 and 6,241 ,764. Communication between the two tubular structures may be achieved through a flow channel which facilitates blood to flow between the bores of each tube.
In another aspect, the anastomotic coupling device is a resorbable device that may be configured with two or three termini which provide a vessel interface without the need for sutures and provides a fluid communication through an intersecting lumen, such as a bypass graft or alternate vessel. See, e.g., U.S. Application Publication Nos. 2002/0052572A1 and PCT Publication No. WO 02/24114A2. An anastomotic connector may also be formed of a resorbable tubular structure configured to include snap-connectors or other components for securing it to the tissue as well as hemostasis inducing sealing rings to prevent blood leakage. See, e.g., U.S. Patent Nos. 6,056,762. The anastomotic connector may be designed with three legs whereby two legs are adapted to be inserted within the continuous blood vessel in a contracted state and then enlarged to form a tight fit and the third leg is adapted for connecting and sealing with a third conduit. See, e.g., U.S. Patent No. 6,019,788.
An example of a commercially available multi-lumen anastomotic coupling device is the SOLEM graft connector (made by Jomed, Sweden). This device, which is described in more detail in PCT Publication No. WO 01/13820, and U.S. Patent Nos. 6,179,848, D438618 and D429334, includes a T-shaped connector composed of nitinol and an ePTFE graft for completion of a distal anastomosis.
Another example of an anastomotic connector is the HOLLY GRAFT System (in development) for use in bypass surgery from CABG Medical, Inc. (Minneapolis, MN), which is described, e.g., in U.S. Patent Nos. 6,241,761 and 6,241 ,764.
In one aspect, the present invention provides for the combination of an anastomotic coupling device and an anti-scarring agent or a composition comprising an anti-scarring device. In one aspect, the anastomotic coupling device may be attached to a blood vessel for the formation of an anastomosis without the use of sutures or staples. In certain aspects, the anastomotic coupling device may comprise a tubular structure defining a lumen through which blood may flow, and an anti-scarring agent. The device may include one, two, three, or more lumens defined by one, two, three, or more tubular structures, depending on the number of vessels to be connected.
Introduction of an anastomotic connector into or onto an intramural, luminal, or adventitial portion of a blood vessel may irritate or damage the endothelial tissue of the blood vessel and/or may alter the natural hemodynamic flow through the vessel. This irritation or damage may stimulate a cascade of biological events resulting in a fibrotic response which can lead to the formation of scar tissue in the vessel. Incorporation of a therapeutic agent in accordance with the invention into or onto a portion of the device that is in direct contact with the blood vessel (e.g., a terminal portion or edge of the device) may inhibit one or more of the scarring processes described above (e.g., smooth muscle cell proliferation, cell migration, inflammation), making the vessel less prone to the formation of intimal hyperplasia and stenosis.
Thus, in one aspect, the therapeutic agent may be associated only with the portion of the device that is in contact with the blood or endothelial tissue. For example, the anti-scarring agent may be incorporated into only an intravascular portion (i.e., that portion that resides within the lumen of the vessel or in the vessel tissue) of the device. The anti-scarring agent may be incorporated onto all or a portion of the intravascular portion of the device. In other embodiments, the coating may reside on all or a portion of an extravascular portion of the device.
The anti-scarring agent or a composition that includes an anti- scarring agent may be coated onto a portion of or onto the entire surface of the device or may be incorporated into a portion of, or into the entire the structure of, the device (e.g., either within voids, reservoirs, or divets in the device or within the material used to construct the device). In other aspects, the agent or a composition comprising the agent is impregnated into or affixed onto the device surface.
As described above, the device may include a tubular portion that is disposed within the lumen of a blood vessel. The entire tubular portion may, for example, be coated with an anti-scarring agent or a composition comprising an anti-scarring. Alternatively, only a portion of the tubular portion may include the anti-scarring agent. For example, only an external (abluminal) surface or only the interior (endoluminal) surface of the tubular portion may be coated. In other embodiments, one or both termini of the tubular portion may be coated. For example, the endoluminal and/or abluminal surface of the tubular section through which blood enters into the device (Ae., proximal end) may be coated with the anti-scarring agent or composition comprising the anti-scarring agent. In another aspect, the endoluminal and/or abluminal surface of the tubular section through which blood exits (i.e., distal end) from the device may be coated with the anti- scarring agent or composition comprising the anti-scarring agent.
In another embodiment, the anti-scarring agent or composition comprising the anti-scarring agent is associated (e.g., coated onto or incorporated into) with an anchoring member (e.g., a fastener, such as a staple or clip) that secures the device to a blood vessel.
As described above, anastomotic connector devices can include a fibrosis-inhibiting agent as a means to improve the clinical efficacy of the device. In another approach, the fibrosis-inhibiting agent can be incorporated into or onto a film or mesh (described in further detail below) that is applied in a perivascular manner to an anastomotic site (e.g., at the junction of a graft vessel and the blood vessel). These films or wraps can be used with any of the anastomotic connector devices described above and, typically, are placed around the outside of the anastomosis at the time of surgery. In other embodiments, the agent may be delivered to the anastomotic site in the form of a spray, paste, gel, or the like. In yet another approach, the fibrosis-inhibiting agent can be incorporated into or onto the graft vessel that is secured to the blood vessel with the connector device.
In yet another aspect, other specialized intravascular devices, such as coronary drug infusion guidewires, such as those available from TherOx, Inc., grafts and balloon over stent devices, such as are described in Wilensky, R.L. (1993) J. Am. Coll. Cardiol.: 21 : 185A can also be utilized for local delivery of an anti-fibrosing agent.
As described above, the present invention provides intravascular devices (e.g., anastomotic connectors, stents, drug-delivery balloons, intravascular catheters) that include an anti-scarring agent or a composition that includes an anti-scarring agent. Numerous polymeric and non-polymeric delivery systems for use with intravascular devices have been described above. Methods for incorporating coating fibrosis-inhibiting agents and compositions onto or into intravascular devices include: (a) directly affixing to the intravascular device a fibrosis-inhibiting composition (e.g., by either a spraying process or dipping process as described above, with or without a carrier), (b) directly incorporating into the device a fibrosis- inhibiting composition (e.g., by either a spraying process or dipping process as described above, with or without a carrier (c) by coating the device with a substance such as a hydrogel which will in turn absorb the fibrosis-inhibiting composition), (d) by interweaving fibrosis-inhibiting composition coated thread (or the polymer itself formed into a thread) into the device structure, (e) by inserting the device into a sleeve or mesh which contains or is coated with a fibrosis-inhibiting composition, (T) constructing the device itself or a portion of the device with a fibrosis-inhibiting composition, or (g) by covalently binding the fibrosis-inhibiting agent directly to the device surface or to a linker (small molecule or polymer) that is coated or attached to the device surface. For these devices, the coating process can be performed in such a manner as to (a) coat the external surface of the stent, (b) coat the internal (luminal) surface of the stent or (c) coat all or parts of both the internal and external surfaces of the stent.
The intravascular device (e.g., a stent) may be adapted to release the desired agent at only the distal ends, or along the entire body of the device.
According to the present invention, any fibrosis-inhibiting agent described above can be utilized in the practice of this embodiment. Within one embodiment of the invention, intravascular devices may be adapted to release an agent that inhibits one or more of the four general components of the process of fibrosis (or scarring), including: formation of new blood vessels (angiogenesis), migration and proliferation of connective tissue cells (such as fibroblasts or smooth muscle cells), deposition of extracellular matrix (ECM), and remodeling (maturation and organization of the fibrous tissue). By inhibiting one or more of the components of fibrosis (or scarring), the overgrowth of granulation tissue may be inhibited or reduced. As intravascular devices are made in a variety of configurations and sizes, the exact dose administered will vary with device size, surface area and design. However, certain principles can be applied in the application of this art. Drug dose can be calculated as a function of dose per unit area (of the portion of the device being coated), total drug dose administered, and appropriate surface concentrations of active drug can be determined. Drugs are to be used at concentrations that range from several times more than to 10%, 5%, or even less than 1% of the concentration typically used in a single chemotherapeutic systemic dose application. Preferably, the drug is released in effective concentrations for a period ranging from 1 - 90 days.
Several examples of agents for use in intravascular devices include the following: ZD-6474, AP-23573, synthadotin, S-0885, aplidine, ixabepilone, IDN-5390, SB-2723005, ABT-518, combretastatin, anecortave acetate, SB-715992, temsirolimus, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, as well as analogues and derivatives of the aforementioned.
Additional examples of anti-scarring agents which can be used include those having a high potency in the assays described herein (1- 10OnM IC50 range) such as isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, dolastatin 15, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17- DMAG, and tacrolimus; those having a mid-potency in the assays described herein (100-500 nM IC50 range) such as loteprednol etabonate, juglone, prednisolone, puromycin, 3-BAABE, cladribine, and mannose-6-phosphate; and those having a low potency in the assays described herein (500- 1000nm range IC50 range) such as 5-azacytidine, Ly333531(ruboxistaurin), and simvastatin.
Regardless of the method of application of the drug to the intravascular device, the exemplary anti-fibrosing agents, used alone or in combination, should be administered under the following dosing guidelines. The total amount (dose) of anti-scarring agent in or on the device may be in the range of about 0.01 μg-10 μg, or 10 μg-10 mg, or 10 mg-250 mg, or 250 mg-1000 mg, or 1000 mg-2500 mg. The dose (amount) of anti-scarring agent per unit area of device surface to which the agent is applied may be in the range of about 0.01 μg/mm2 - 1 μg/mm2, or 1 μg/mm2 - 10 μg/mm2, or 10 μg/mm2 - 250 μg/mm2, 250 μg/mm2 - 1000 μg/mm2, or 1000 μg/mm2 - 2500 μg/mm2.
Provided below are exemplary dosage ranges for various anti- scarring agents that can be used in conjunction with intravascular devices in accordance with the invention. (A) Angiogenesis inhibitors including Alphastatin, ZD-6474, IDN-5390, SB-2723005, ABT-518, combretastatin, and anecortane, analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10^- 10"4 M of agent is to be maintained on the implant or barrier surface. (B) mTOR inhibitors including AP-23573 and temsirolimus, analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10^- 10"4 M of agent is to be maintained on the implant or barrier surface. (C) Tubulin antagonists including synthadotin, analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10^- 10"4 M of agent is to be maintained on the implant or barrier surface. (D) Epithilones including ixabepilone and analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10^- 10"4 M of agent is to be maintained on the implant or barrier surface. (E) Kinesin antagonists including SB- 715992 and analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10"8- 10"4 M of agent is to be maintained on the implant or barrier surface. (F) TNF alpha antagonists including etanercept, humicade, adalimumab and analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10'8- 10"4 M of agent is to be maintained on the implant or barrier surface. (G) AKT inhibitor including erucylphosphocholine and analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10^- 10"4 M of agent is to be maintained on the implant or barrier surface. (H) FGF Inhibitors including IDN-5390 and analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10'8- 10"4 M of agent is to be maintained on the implant or barrier surface. (I) Collagenase Antagonists including S-0885 and analogues and derivatives thereof: total dose not to exceed 1000 mg (range of 0.1 μg to 1000 mg); preferred 1 μg to 500 mg. Dose per unit area of 0.01 μg - 500 μg per mm2; preferred dose of 0.1 μg/mm2 - 100 μg/mm2. Minimum concentration of 10'8- 10"4 M of agent is to be maintained on the implant or barrier surface. (J) NF Kappa B Inhibitors including Bortesomib and analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 108 - 104 M of agent is to be maintained on the implant or barrier surface. (K) elongation factor- 1 alpha inhibitors including aplidine and analogues and derivatives thereof: total dose not to exceed 1000 mg (range of 0.1 μg to 1000 mg); preferred 1 μg to 500 mg. Dose per unit area of 0.01 μg - 500 μg per mm2; preferred dose of 0.1 μg/mm2 - 100 μg/mm2. Minimum concentration of 10"8- 10'4 M of agent is to be maintained on the implant or barrier surface. (L) Tyrosine kinase inhibitors including gefitinib and analogues and derivatives thereof: total dose not to exceed 1000 mg (range of 0.1 μg to 1000 mg); preferred 1 μg to 500 mg. Dose per unit area of 0.01 μg - 500 μg per mm2; preferred dose of 0.1 μg/mm2 - 100 μg/mm2. Minimum concentration of 10"8- 10"4 M of agent is to be maintained on the implant or barrier surface.
For high potency drugs, the total dose typically should not exceed 200 mg (range of 0.1 μg to 200 mg) and preferably 1 μg to 100 mg; dose per unit area of 0.01 μg - 100 μg per mm2; preferably 0.1 μg/mm2 - 20 μg/mm2; and minimum concentration of 10"8- 10"4 M of agent should be maintained on the implant or barrier surface. For mid-potency drugs, the total dose typically should not to exceed 500 mg (range of 1.0 μg to 500 mg) and preferably 1 μg to 200 mg; dose per unit area of 0.01 μg - 200 μg per mm2, preferably 0.1 μg/mm2 - 40 μg/mm2; and minimum concentration of 10" 8 - 10"4 M of agent should be maintained on the implant or barrier surface. For low potency drugs, the total dose typically should not exceed 1000 mg (range of 0.1 μg to 1000 mg), preferably 1 μg to 500 mg; dose per unit area of 0.01 μg - 500 μg per mm2; preferably 0.1 μg/mm2 - 100 μg/mm2; and minimum concentration of 10"8- 10"4 M of agent should to be maintained on the implant or barrier surface.
Gastrointestinal Stents
The present invention provides for the combination of a drug and a gastrointestinal (Gl) stent. The term Gl stent refers to devices that are located in the gastrointestinal tract including the biliary duct, pancreatic duct, colon, and the esophagus. Gl stents are or comprise scaffoldings that are used to treat endoluminal body passageways that have become blocked due to disease or damage, including malignancy or benign disease. In one aspect, the Gl stent may be an esophageal stent used to keep the esophagus open whereby food is able to travel from the mouth to the stomach. For example, the esophageal stent may be composed of a cylindrical supporting mesh inner layer, retaining mesh outer layer and a semi-permeable membrane sandwiched between. See, e.g., U.S. Patent No. 6,146,416. The esophageal stent may be a radially, self-expanding stent of open weave construction with an elastomeric film formed along the stent to prevent tissue ingrowth and distal cuffs that resist stent migration. See, e.g., U.S. Patent No. 5,876,448. The esophageal stent may be composed of a flexible wire configuration to form a cylindrical tube with a deformed end portion increased to a larger diameter for anchoring pressure. See, e.g., U.S. Patent No. 5,876,445. The esophageal stent may be a flexible, self-expandable tubular wall incorporating at least one truncated conical segment along the longitudinal axis. See, e.g., U.S. Patent No. 6,533,810.
In another aspect, the Gl stent may be a biliary stent used to keep the biliary duct open whereby bile is able to drain into the small intestines. For example, the biliary stent may be composed of shape memory alloy. See, e.g., U.S. Patent No. 5,466,242. The biliary stent may be a plurality of radially extending wings with grooves which project from a helical core. See, e.g., U.S. Patent Nos. 5,776,160 and 5,486,191.
In another aspect, the Gl stent may be a colonic stent. For example, the colonic stent may be a hollow tubular body that may expand radially and be secured to the inner wall of the organ in a release fitting. See, e.g., European Patent Application No. EP1092400A2.
In another aspect, the Gl stent may be a pancreatic stent used to keep the pancreatic duct open to facilitate secretion into the small intestines. For example, the pancreatic stent may be composed of a soft biocompatible material which is resiliently compliant which conforms to the duct's curvature and contains perforations that facilitates drainage. See, e.g., U.S. Patent No. 6,132,471. Gl stents, which may be combined with one or more drugs according to the present invention, include commercially available products, such as the NIR Biliary Stent System and the WALLSTENT Endoprostheses from Boston Scientific Corporation.
In one aspect, the present invention provides Gl stents that include an anti-scarring agent or a composition that includes an anti-scarring agent. Numerous polymeric and non-polymeric delivery systems for use in Gl stents have been described above.
Methods for incorporating fibrosis-inhibiting agents or fibrosis- inhibiting compositions onto or into the Gl stents include: (a) directly affixing to the stent a fibrosis-inhibiting composition (e.g., by either a spraying process or dipping process as described above, with or without a carrier), (b) directly incorporating into the stent a fibrosis-inhibiting composition (e.g., by either a spraying process or dipping process as described above, with or without a carrier), (c) by coating the stent with a substance such as a hydrogel which will in turn absorb the fibrosis-inhibiting composition, (d) by interweaving fibrosis-inhibiting composition coated thread (or the polymer itself formed into a thread) into the stent structure, (e) by inserting the stent into a sleeve or mesh which is comprised of or coated with a fibrosis- inhibiting composition, (f) constructing the stent itself or a portion of the stent with a fibrosis-inhibiting composition, or (g) by covalently binding the fibrosis-inhibiting agent directly to the stent surface or to a linker (small molecule or polymer) that is coated or attached to the stent surface. For these devices, the coating process can be performed in such a manner as to (a) coat the external surface of the stent, (b) coat the internal (luminal) surface of the stent or (c) coat all or parts of both the internal and external surfaces of the stent.
In addition to coating the Gl stent with the fibrosis-inhibiting composition, the fibrosis-inhibiting agent can be mixed with the materials that are used to make the device such that the fibrosis-inhibiting agent is incorporated into the final device. This can include the Gl stent structure itself, the outer covering or sleeve, if applicable, or both the stent structure and the outer covering or sleeve.
According to the present invention, any fibrosis-inhibiting agent described above can be utilized in the practice of this embodiment. Within one embodiment of the invention, Gl stents may be adapted to release an agent that inhibits one or more of the four general components of the process of fibrosis (or scarring), including: formation of new blood vessels (angiogenesis), migration and proliferation of connective tissue cells (such as fibroblasts or smooth muscle cells), deposition of extracellular matrix (ECM), and remodeling (maturation and organization of the fibrous tissue). By inhibiting one or more of the components of fibrosis (or scarring), the overgrowth of granulation tissue may be inhibited or reduced.
As Gl stents are made in a variety of configurations and sizes, the exact dose administered will vary with device size, surface area and design. However, certain principles can be applied in the application of this art. Drug dose can be calculated as a function of dose per unit area (of the portion of the device being coated), total dose administered, and appropriate surface concentrations of active drug can be determined. Drugs are to be used at concentrations that range from several times more than to 10%, 5%, or even less than 1 % of the concentration typically used in a single chemotherapeutic systemic dose application. Preferably, the drug is released in effective concentrations for a period ranging from 1 - 90 days.
Several examples of scarring agents for use in Gl stents include the following: ZD-6474, AP-23573, synthadotin, S-0885, aplidine, ixabepilone, IDN-5390, SB-2723005, ABT-518, combretastatin, anecortave acetate, SB-715992, temsirolimus, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, as well as analogues and derivatives of the aforementioned.
Additional examples of anti-scarring agents which can be used include those having a high potency in the assays described herein (1- 10OnM IC50 range) such as isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, dolastatin 15, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17- DMAG, and tacrolimus; those having a mid-potency in the assays described herein (100-500 nM IC50 range) such as loteprednol etabonate, juglone, prednisolone, puromycin, 3-BAABE, cladribine, and mannose-6-phosphate; and those having a low potency in the assays described herein (500- 1000nm range IC50 range) such as 5-azacytidine, Ly333531(ruboxistaurin), and simvastatin.
Regardless of the method of application of the drug to the Gl stent, the exemplary anti-fibrosing agents, used alone or in combination, should be administered under the following dosing guidelines. The total amount (dose) of anti-scarring agent in or on the device may be in the range of about 0.01 μg-10 μg, or 10 μg-10 mg, or 10 mg-250 mg, or 250 mg-1000 mg, or 1000 mg-2500 mg. The dose (amount) of anti-scarring agent per unit area of device surface to which the agent is applied may be in the range of about 0.01 μg/mm2 - 1 μg/mm2, or 1 μg/mm2 - 10 μg/mm2, or 10 μg/mm2 - 250 μg/mm2, 250 μg/mm2 - 1000 μg/mm2, or 1000 μg/mm2 - 2500 μg/mm2.
Provided below are exemplary dosage ranges for various anti- scarring agents that can be used in conjunction with Gl stent devices in accordance with the invention. (A) Angiogenesis inhibitors including Alphastatin, ZD-6474, IDN-5390, SB-2723005, ABT-518, combretastatin, and anecortane, analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10^- 10"4 M of agent is to be maintained on the implant or barrier surface. (B) mTOR inhibitors including AP-23573 and temsirolimus, analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10^- 10"4 M of agent is to be maintained on the implant or barrier surface. (C) Tubulin antagonists including synthadotin, analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10'8- 10"4 M of agent is to be maintained on the implant or barrier surface. (D) Epithilones including ixabepilone and analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10^- 10"4 M of agent is to be maintained on the implant or barrier surface. (E) Kinesin antagonists including SB- 715992 and analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10"8- 10"4 M of agent is to be maintained on the implant or barrier surface. (F) TNF alpha antagonists including etanercept, humicadβj adalimumab and analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10^- 10"4 M of agent is to be maintained on the implant or barrier surface. (G) AKT inhibitor including erucylphosphocholine and analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10^- 10"4 M of agent is to be maintained on the implant or barrier surface. (H) FGF Inhibitors including IDN-5390 and analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10^- 10"4 M of agent is to be maintained on the implant or barrier surface. (I) Collagenase Antagonists including S-0885 and analogues and derivatives thereof: total dose not to exceed 1000 mg (range of 0.1 μg to 1000 mg); preferred 1 μg to 500 mg. Dose per unit area of 0.01 μg - 500 μg per mm2; preferred dose of 0.1 μg/mm2 - 100 μg/mm2. Minimum concentration of 10"8- 10"4 M of agent is to be maintained on the implant or barrier surface. (J) NF KAPPA B Inhibitors including Bortezomib and analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10"8- 10"4 M of agent is to be maintained on the implant or barrier surface. (K) Elongation factor-1 alpha inhibitors including aplidine and analogues and derivatives thereof: total dose not to exceed 1000 mg (range of 0.1 μg to 1000 mg); preferred 1 μg to 500 mg. Dose per unit area of 0.01 μg - 500 μg per mm2; preferred dose of 0.1 μg/mm2 - 100 μg/mm2. Minimum concentration of 10'8- 10"4 M of agent is to be maintained on the implant or barrier surface. (L) Tyrosine kinase inhibitors including Gefitinib and analogues and derivatives thereof: total dose not to exceed 1000 mg (range of 0.1 μg to 1000 mg); preferred 1 μg to 500 mg. Dose per unit area of 0.01 μg - 500 μg per mm2; preferred dose of 0.1 μg/mm2 - 100 μg/mm2. Minimum concentration of 10"8- 10~4 M of agent is to be maintained on the implant or barrier surface.
For high potency drugs, the total dose typically should not exceed 200 mg (range of 0.1 μg to 200 mg) and preferably 1 μg to 100 mg; dose per unit area of 0.01 μg - 100 μg per mm2; preferably 0.1 μg/mm2 - 20 μg/mm2; and minimum concentration of 10^ - 10"4 M of agent should be maintained on the implant or barrier surface. For mid-potency drugs, the total dose typically should not to exceed 500 mg (range of 1.0 μg to 500 mg) and preferably 1 μg to 200 mg; dose per unit area of 0.01 μg - 200 μg per mm2, preferably 0.1 μg/mm2 - 40 μg/mm2; and minimum concentration of 10' 8- 10"4 M of agent should be maintained on the implant or barrier surface. For low potency drugs, the total dose typically should not exceed 1000 mg (range of 0.1 μg to 1000 mg), preferably 1 μg to 500 mg; dose per unit area of 0.01 μg - 500 μg per mm2; preferably 0.1 μg/mm2 - 100 μg/mm2; and minimum concentration of 10"8- 10"4 M of agent should to be maintained on the implant or barrier surface.
Tracheal and Bronchial Stents
The present invention provides for the combination of an anti- scarring agent and a tracheal or bronchial stent device.
Representative examples of tracheal or bronchial stents that can benefit from being coated with or having incorporated therein, a fibrosis- inhibiting agent include tracheal stents or bronchial stents, including metallic and polymeric tracheal or bronchial stents and tracheal or bronchial stents that have an external covering (e.g., polyurethane, poly(ethylene terephthalate), PTFE, or silicone rubber).
Tracheal and bronchial stents may be, for example, composed of an elastic plastic shaft with metal clasps that expands to form a lumen along the axis for opening the diseased portion of the trachea and having three sections to emulate the natural shape of the trachea. See, e.g., U.S. Patent No. 5,480,431. The tracheal/bronchial stent may be a T-shaped tube having a tracheotomy tubular portion that projects outwardly through a tracheotomy orifice which is configured to close and form a fluid seal. See, e.g., U.S. Patent Nos. 5,184,610 and 3,721,233. The tracheal/bronchial stent may be composed of a flexible, synthetic polymeric resin with a tracheotomy tube mounted on the wall with a bifurcated bronchial end that is configured in a T-Y shape with specific curves at the intersections to minimize tissue damage. See, e.g., U.S. Patent No. 4,795,465. The tracheal/bronchial stent may be a scaffolding configured to be substantially cylindrical with a shape-memory frame having geometrical patterns and having a coating of sufficient thickness to prevent epithelialization. See, e.g., U.S. Patent Application Publication No. 2003/0024534A1.
Tracheal/bronchial stents, which may be combined with one or more agents according to the present invention, include commercially available products, such as the WALLSTENT Tracheobronchial Endoprostheses and ULTRAFLEX Tracheobronchial Stent Systems from Boston Scientific Corporation and the DUMON Tracheobronchial Silicone Stents from Bryan Corporation (Woburn, MA).
In one aspect, the present invention provides tracheal and bronchial stents that include an anti-scarring agent or a composition that includes an anti-scarring agent. Numerous polymeric and non-polymeric delivery systems for use in tracheal and bronchial stents have been described above. Methods for incorporating fibrosis-inhibiting agents or fibrosis-inhibiting compositions onto or into the tracheal or bronchial stents include: (a) directly affixing to the stent a fibrosis-inhibiting composition (e.g., by either a spraying process or dipping process as described above, with or without a carrier), (b) directly incorporating into the stent a fibrosis- inhibiting composition (e.g., by either a spraying process or dipping process as described above, with or without a carrier (c) by coating the stent with a substance such as a hydrogel which will in turn absorb the fibrosis-inhibiting composition), (d) by interweaving fibrosis-inhibiting composition coated thread (or the polymer itself formed into a thread) into the stent structure, (e) by inserting the stent into a sleeve or mesh which is comprised of or coated with a fibrosis-inhibiting composition, (f) constructing the stent itself or a portion of the stent with a fibrosis-inhibiting composition, or (g) by covalently binding the fibrosis-inhibiting agent directly to the stent surface or to a linker (small molecule or polymer) that is coated or attached to the stent surface. For these devices, the coating process can be performed in such a manner as to (a) coat the external surface of the stent, (b) coat the internal (luminal) surface of the stent or (c) coat all or parts of both the internal and external surfaces of the stent.
In addition to coating the device with the fibrosis-inhibiting composition, the fibrosis-inhibiting agent can be mixed with the materials that are used to make the device such that the fibrosis-inhibiting agent is incorporated into the final device. This can include the stent structure itself, the outer covering or sleeve, if applicable, or both the stent structure and the outer covering or sleeve.
According to the present invention, any fibrosis-inhibiting agent described above can be utilized in the practice of this embodiment. Within one embodiment of the invention, tracheal and bronchial stents may be adapted to release an agent that inhibits one or more of the four general components of the process of fibrosis (or scarring), including: formation of new blood vessels (angiogenesis), migration and proliferation of connective tissue cells (such as fibroblasts or smooth muscle cells), deposition of extracellular matrix (ECM), and remodeling (maturation and organization of the fibrous tissue). By inhibiting one or more of the components of fibrosis (or scarring), the overgrowth of granulation tissue may be inhibited or reduced.
As tracheal and bronchial stents are made in a variety of configurations and sizes, the exact dose administered will vary with device size, surface area and design. However, certain principles can be applied in . the application of this art. Drug dose can be calculated as a function of dose per unit area (of the portion of the device being coated), total dose administered, and appropriate surface concentrations of active drug can be determined. Drugs are to be used at concentrations that range from several times more than to 10%, 5%, or even less than 1 % of the concentration typically used in a single chemotherapeutic systemic dose application. Preferably, the drug is released in effective concentrations for a period ranging from 1 - 90 days.
Several fibrosis-inhibiting agents for use in tracheal and bronchial stents include the following: ZD-6474, AP-23573, synthadotin, S- 0885, aplidine, ixabepilone, IDN-5390, SB-2723005, ABT-518, combretastatin, anecortave acetate, SB-715992, temsirolimus, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, as well as analogues and derivatives of the aforementioned. Additional examples of anti-scarring agents which can be used include those having a high potency in the assays described herein (1- 10OnM IC50 range) such as isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, dolastatin 15, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17- DMAG, and tacrolimus; those having a mid-potency in the assays described herein (100-500 nM IC50 range) such as loteprednol etabonate, juglone, prednisolone, puromycin, 3-BAABE, cladribine, and mannose-6-phosphate; and those having a low potency in the assays described herein (500- 1000nm range IC50 range) such as 5-azacytidine, Ly333531(ruboxistaurin), and simvastatin.
Regardless of the method of application of the drug to the tracheal or bronchial stent, the exemplary anti-fibrosing agents, used alone or in combination, should be administered under the following dosing guidelines. The total amount (dose) of anti-scarring agent in or on the device may-be in the range of about 0.01 μg-10 μg, or 10 μg-10 mg, or 10 mg-250 mg, or 250 mg-1000 mg, or 1000 mg-2500 mg. The dose (amount) of anti-scarring agent per unit area of device surface to which the agent is applied may be in the range of about 0.01 μg/mm2 - 1 μg/mm2, or 1 μg/mm2 - 10 μg/mm2, or 10 μg/mm2 - 250 μg/mm2, 250 μg/mm2 - 1000 μg/mm2, or 1000 μg/mm2 - 2500 μg/mm2.
Provided below are exemplary dosage ranges for various anti- scarring agents that can be used in conjunction with tracheal and bronchial stent devices in accordance with the invention. (A) Angiogenesis inhibitors including alphastatin, ZD-6474, IDN-5390, SB-2723005, ABT-518, combretastatin, and anecortane, analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10^- 10"4 M of agent is to be maintained on the implant or barrier surface. (B) mTOR inhibitors including AP-23573 and temsirolimus, analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10"8- 10"4 M of agent is to be maintained on the implant or barrier surface. (C) Tubulin antagonists including synthadotin, analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10^- 10"4 M of agent is to be maintained on the implant or barrier surface. (D) Epithilones including ixabepilone and analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10^- 10"4 M of agent is to be maintained on the implant or barrier surface. (E) Kinesin antagonists including SB- 715992 and analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0,1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10~s- 10"4 M of agent is to be maintained on the implant or barrier surface. (F) TNF alpha antagonists including Etanercept, humicade, adalimumab and analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10^- 10"4 M of agent is to be maintained on the implant or barrier surface. (G) AKT inhibitor including erucylphosphocholine and analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10^- 10"4 M of agent is to be maintained on the implant or barrier surface. (H) FGF Inhibitors including IDN-5390 and analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10"8- 10"4 M of agent is to be maintained on the implant or barrier surface. (I) Collagenase Antagonists including S-0885 and analogues and derivatives thereof: total dose not to exceed 1000 mg (range of 0.1 μg to 1000 mg); preferred 1 μg to 500 mg. Dose per unit area of 0.01 μg - 500 μg per mm2; preferred dose of 0.1 μg/mm2 - 100 μg/mm2. Minimum concentration of 10^- 10"4 M of agent is to be maintained on the implant or barrier surface. (J) NF KAPPA B Inhibitors including Bortezomib and analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10^- 10"4 M of agent is to be maintained on the implant or barrier surface. (K) Elongation factor-1 alpha inhibitors including aplidine and analogues and derivatives thereof: total dose not to exceed 1000 mg (range of 0.1 μg to 1000 mg); preferred 1 μg to 500 mg. Dose per unit area of 0.01 μg - 500 μg per mm2; preferred dose of 0.1 μg/mm2 - 100 μg/mm2. Minimum concentration of 10'8- 10'4 M of agent is to be maintained on the implant or barrier surface. (L) Tyrosine kinase inhibitors including gefitinib and analogues and derivatives thereof: total dose not to exceed 1000 mg (range of 0.1 μg to 1000 mg); preferred 1 μg to 500 mg. Dose per unit area of 0.01 μg - 500 μg per mm2; preferred dose of 0.1 μg/mm2 - 100 μg/mm2. Minimum concentration of 10"s- 10"4 M of agent is to be maintained on the implant or barrier surface.
For high potency drugs, the total dose typically should not exceed 200 mg (range of 0.1 μg to 200 mg) and preferably 1 μg to 100 mg; dose per unit area of 0.01 μg - 100 μg per mm2; preferably 0.1 μg/mm2 - 20 μg/mm2; and minimum concentration of 10^- 10"4 M of agent should be maintained on the implant or barrier surface. For mid-potency drugs, the total dose typically should not to exceed 500 mg (range of 1.0 μg to 500 mg) and preferably 1 μg to 200 mg; dose per unit area of 0.01 μg - 200 μg per mm2, preferably 0.1 μg/mm2 - 40 μg/mm2; and minimum concentration of 10' 8- 10'4 M of agent should be maintained on the implant or barrier surface. For low potency drugs, the total dose typically should not exceed 1000 mg (range of 0.1 μg to 1000 mg), preferably 1 μg to 500 mg; dose per unit area of 0.01 μg - 500 μg per mm2; preferably 0.1 μg/mm2 - 100 μg/mm2; and minimum concentration of 10~8- 10'4 M of agent should to be maintained on the implant or barrier surface.
Genital-Urinary Stents
The present invention provides for the combination of an anti- scarring agent and genital-urinary (GU) stent device.
Representative examples genital-urinary (GU) stents that can benefit from being coated with or having incorporated therein, a fibrosis- inhibiting agent include ureteric and urethral stents, fallopian tube stents, prostate stents, including metallic and polymeric GU stents and GU stents that have an external covering (e.g., polyurethane, poly(ethylene terephthalate), PTFE or silicone rubber).
In one aspect, genital-urinary stents include ureteric and urethral stents. Ureteral stents are hollow tubes with holes along the sides and coils at either end to prevent migration. Ureteral stents are used to relieve obstructions (caused by stones or malignancy), to facilitate the passage of stones, or to allow healing of ureteral anastomoses or leaks following surgery or trauma. They are placed endoscopically via the bladder or percutaneously via the kidney.
Urethral stents are used for the treatment of recurrent urethral strictures, detruso-external sphincter dyssynergia and bladder outlet obstruction due to benign prostatic hypertrophy. In addition, procedures that are conducted for the prostate, such as external radiation or brachytherapy, may lead to fibrosis due to tissue insult resulting from these procedures. The incidence of urethral stricture in prostate cancer patients treated with external beam radiation is about 2%. Development of urethral stricture may also occur in other conditions such as following urinary catheterization or surgery, which results in damage to the epithelium of the urethra. The clinical manifestation of urinary tract obstruction includes decreased force and caliber of the urinary stream, intermittency, postvoid dribbling, hesitance and nocturia. Complete closure of the urethra can result in numerous problems including eventual kidney failure. To maintain patency in the urethra, urethral stents may be used. The stents are typically self- expanding and composed of metal superalloy, titanium, stainless steel or polyurethane.
For example, the ureteric/urethral stent may be composed of a main catheter body of flexible polymeric material having an enlarged entry end with a hydrophilic tip that dissolves when contacted with body fluids. See, e.g., U.S. Patent No. 5,401,257. The ureteric/urethral stent may be composed of a multi-sections including a closed section at that the bladder end which does not contain any fluid passageways such that it acts as an anti-reflux device to prevent reflux of urine back into the kidney. See, e.g., U.S. Patent No. 5,647,843. The ureteric/urethral stent may be composed of . a central catheter tube made of shape memory material that forms a stent with a retention coil for anchoring to the ureter. See, e.g., U.S. Patent No. 5,681 ,274. The ureteric/urethral stent may be a composed of an elongated flexible tubular stent with preformed set curls at both ends and an elongated tubular rigid extension attached to the distal end which allows the combination function as an externalized ureteral catheter. See, e.g., U.S. Patent Nos. 5,221,253 and 5,116,309. The ureteric/urethral stent may be composed of an elongated member, a proximal retention structure, and a resilient portion connecting them together, whereby they are all in fluid communication with each other with a slideable portion providing a retracted and expanded position. See, e.g., U.S. Patent No. 6,685,744. The ureteric/urethral stent may be a hollow cylindrical tube that has a flexible connecting means and locating means that expands and selectively contracts. See, e.g., U.S. Patent No. 5,322,501. The ureteric/urethral stent may be composed of a stiff polymeric body that affords superior columnar and axial strength for advancement into the ureter, and a softer bladder coil portion for reducing the risk of irritation. See, e.g., U.S. Patent No. 5,141 ,502. The ureteric/urethral stent may be composed of an elongated tubular segment that has a pliable wall at the proximal region and a plurality of members that prevent blockage of fluid drainage upon compression. See, e.g., U.S. Patent No. 6,676,623. The ureteric/urethral stent may be a catheter composed of a conduit which is part of an assembly that allows for non-contaminated insertion into a urinary canal by providing a sealing member that surrounds the catheter during dismantling. See, e.g., U.S. Patent Application Publication No. 2003/0060807A1.
In another aspect, genital-urinary stents include prostatic stents. For example, the prostatic stent may be composed of two polymeric rings constructed of tubing with a plurality of connecting arm members connecting the rings in a parallel manner. See, e.g., U.S. Patent No. 5,269,802. The prostatic stent may be composed of thermoplastic material and a circumferential reinforcing helical spring, which provides rigid mechanical support while being flexible to accommodate the natural anatomical bend of the prostatic urethra. See, e.g., U.S. Patent No. 5,069,169.
In another aspect, genital-urinary stents include fallopian stents and other female genital-urinary devices. For example, the genital- urinary device may be a female urinary incontinence device composed of a vaginal-insertable supporting portion that is resilient and flexible, which is capable of self-support by expansion against the vaginal wall and extending about the urethral orifice. See, e.g., U.S. Patent No. 3,661,155. The genital-urinary device may be a urinary evacuation device composed of a ovular bulbous concave wall having an opening to a body engaging perimetal edge integral with the wall and an attached tubular member with a pleated body. See, e.g., U.S. Patent No. 6,041,448.
Genital-urinary stents, which may be combined with one or more agents according to the present invention, include commercially available products, such as the UROLUME Endoprosthesis Stents from American Medical Systems, Inc. (Minnetonka, MN), the RELIEVE Prostatic/Urethral Endoscopic Device from InjecTx, Inc. (San Jose, CA), the PERCUFLEX Ureteral Stents from Boston Scientific Corporation, and the TARKINGTON Urethral Stents and FIRLIT-KLUGE Urethral Stents from Cook Group lnc (Bloomington, IN).
In one aspect, the present invention provides GU stents that include an anti-scarring agent or a composition that includes an anti-scarring agent. Numerous polymeric and non-polymeric delivery systems for use in GU stents have been described above. Methods for incorporating fibrosing agents or fibrosis-inhibiting compositions onto or into the GU stents include: (a) directly affixing to the stent a fibrosis-inhibiting composition (e.g., by either a spraying process or dipping process as described above, with or without a carrier), (b) directly incorporating into the stent a fibrosis-inhibiting composition (e.g., by either a spraying process or dipping process as described above, with or without a carrier), (c) by coating the stent with a substance such as a hydrogel which will in turn absorb the fibrosis-inhibiting composition, (d) by interweaving fibrosis-inhibiting composition coated thread (or the polymer itself formed into a thread) into the stent structure, (e) by inserting the stent into a sleeve or mesh which is comprised of or coated with a fibrosis-inhibiting composition, (f) constructing the stent itself or a portion of the stent with a fibrosis-inhibiting composition, or (g) by covalently binding the fibrosis-inhibiting agent directly to the stent surface or to a linker (small molecule or polymer) that is coated or attached to the stent surface. For these devices, the coating process can be performed in such a manner as to (a) coat the external surface of the stent, (b) coat the internal (luminal) surface of the stent or (c) coat all or parts of both the internal and external surfaces of the stent.
In addition to coating the device with the fibrosis-inhibiting composition, the fibrosis-inhibiting agent can be mixed with the materials that are used to make the device such that the fibrosis-inhibiting agent is incorporated into the final device.
According to the present invention, any fibrosis-inhibiting agent described above can be utilized in the practice of this embodiment. Within one embodiment of the invention, GU stents may be adapted to release an agent that inhibits one or more of the four general components of the process of fibrosis (or scarring), including: formation of new blood vessels (angiogenesis), migration and proliferation of connective tissue cells (such as fibroblasts or smooth muscle cells), deposition of extracellular matrix (ECM), and remodeling (maturation and organization of the fibrous tissue). By inhibiting one or more of the components of fibrosis (or scarring), the overgrowth of granulation tissue may be inhibited or reduced.
As GU stents are made in a variety of configurations and sizes, the exact dose administered will vary with device size, surface area and design. However, certain principles can be applied in the application of this art. Drug dose can be calculated as a function of dose per unit area (of . the portion of the device being coated), total dose administered, and appropriate surface concentrations of active drug can be determined. Drugs are to be used at concentrations that range from several times more than to 10%, 5%, or even less than 1% of the concentration typically used in a single chemotherapeutic systemic dose application. Preferably, the drug is released in effective concentrations for a period ranging from 1 - 90 days.
Several examples of scarring agents for use in GU stents include the following: ZD-6474, AP-23573, synthadotin, S-0885, aplidine, ixabepilone, IDN-5390, SB-2723005, ABT-518, combretastatin, anecortave acetate, SB-715992, temsirolimus, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, as well as analogues and derivatives of the aforementioned.
Additional examples of anti-scarring agents which can be used include those having a high potency in the assays described herein (1- 10OnM IC50 range) such as isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, dolastatin 15, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17- DMAG, and tacrolimus; those having a mid-potency in the assays described herein (100-500 nM IC50 range) such as loteprednol etabonate, juglone, prednisolone, puromycin, 3-BAABE, cladribine, and mannose-6-phosphate; and those having a low potency in the assays described herein (500- 1000nm range IC50 range) such as 5-azacytidine, Ly333531(ruboxistaurin), and simvastatin.
Regardless of the method of application of the drug to the GU stent, the exemplary anti-fibrosing agents, used alone or in combination, should be administered under the following dosing guidelines. The total amount (dose) of anti-scarring agent in or on the device may be in the range of about 0.01 μg-10 μg, or 10 μg-10 mg, or 10 mg-250 mg, or 250 mg-1000 mg, or 1000 mg-2500 mg. The dose (amount) of anti-scarring agent per unit area of device surface to which the agent is applied may be in the range of i about 0.01 μg/mm2 - 1 μg/mm2, or 1 μg/mm2 - 10 μg/mm2, or 10 μg/mm2 - . 250 μg/mm2, 250 μg/mm2 - 1000 μg/mm2, or 1000 μg/mm2 - 2500 μg/mm2.
Provided below are exemplary dosage ranges for various anti- scarring agents that can be used in conjunction with GU stent devices in accordance with the invention. (A) Angiogenesis inhibitors including Alphastatin, ZD-6474, IDN-5390, SB-2723005, ABT-518, combretastatin, and anecortane, analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10^- 10"4 M of agent is to be maintained on the implant or barrier surface. (B) mTOR inhibitors including AP-23573 and Temsirolimus, analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10^- 10"4 M of agent is to be maintained on the implant or barrier surface. (C) Tubulin antagonists including synthadotin, analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10"8 - 10"4 M of agent is to be maintained on the implant or barrier surface. (D) Epithilones including ixabepilone and analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10^- 10"4 M of agent is to be maintained on the implant or barrier surface. (E) Kinesin antagonists including SB- 715992 and analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10^- 10"4 M of agent is to be maintained on the implant or barrier surface. (F) TNF alpha antagonists including Etanercept, humicade, adalimumab and analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10'8- 10"4 M of agent is to be maintained on the implant or barrier surface. (G) AKT inhibitor including erucylphosphocholine and analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10^ - 10"4 M of agent is to be maintained on the implant or barrier surface. (H) FGF Inhibitors including IDN-5390 and analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10^- 10"4 M of agent is to be maintained on the implant or barrier surface. (I) Collagenase Antagonists including S-0885 and analogues and derivatives thereof: total dose not to exceed 1000 mg (range of 0.1 μg to 1000 mg); preferred 1 μg to 500 mg. Dose per unit area of 0.01 μg - 500 μg per mm2; preferred dose of 0.1 μg/mm2 - 100 μg/mm2. Minimum concentration of 10'8- 10"4 M of agent is to be maintained on the implant or barrier surface. (J) NF KAPPA B Inhibitors including Bortezomib and analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10"8- 10"4 M of agent is to be maintained on the implant or barrier surface. (K) Elongation factor-1 alpha inhibitors including aplidine and analogues and derivatives thereof: total dose not to exceed 1000 mg (range of 0.1 μg to 1000 mg); preferred 1 μg to 500 mg. Dose per unit area of 0.01 μg - 500 μg per mm2; preferred dose of 0.1 μg/mm2 - 100 μg/mm2. Minimum concentration of 10'8- 10'4 M of agent is to be maintained on the implant or barrier surface. (L) Tyrosine kinase inhibitors including Gefitinib and analogues and derivatives thereof: total dose not to exceed 1000 mg (range of 0.1 μg to 1000 mg); preferred 1 μg to 500 mg. Dose per unit area of 0.01 μg - 500 μg per mm2; preferred dose of 0.1 μg/mm2 - 100 μg/mm2. Minimum concentration of 10'8- 10'4 M of agent is to be maintained on the implant or barrier surface.
For high potency drugs, the total dose typically should not exceed 200 mg (range of 0.1 μg to 200 mg) and preferably 1 μg to 100 mg; dose per unit area of 0.01 μg - 100 μg per mm2; preferably 0.1 μg/mm2 - 20 μg/mm2; and minimum concentration of 10'8- 10"4 M of agent should be maintained on the implant or barrier surface. For mid-potency drugs, the total dose typically should not to exceed 500 mg (range of 1.0 μg to 500 mg) and preferably 1 μg to 200 mg; dose per unit area of 0.01 μg - 200 μg per mm2, preferably 0.1 μg/mm2 - 40 μg/mm2; and minimum concentration of 10' 8- 10"4 M of agent should be maintained on the implant or barrier surface. For low potency drugs, the total dose typically should not exceed 1000 mg (range of 0.1 μg to 1000 mg), preferably 1 μg to 500 mg; dose per unit area of 0.01 μg - 500 μg per mm2; preferably 0.1 μg/mm2 - 100 μg/mm2; and minimum concentration of 10'8- 10"4 M of agent should to be maintained on the implant or barrier surface.
Ear and Nose Stents
The present invention provides for the combination of an anti- scarring agent and an ear-nose-throat (ENT) stent device {e.g., a lacrimal duct stent, Eustachian tube stent, nasal stent, or sinus stent).
The sinuses are four pairs of hollow regions contained in the bones of the skull named after the bones in which they are located (ethmoid, maxillary, frontal and sphenoid). All are lined by respiratory mucosa which is directly attached to the bone. Following an inflammatory insult such as an upper respiratory tract infection or allergic rhinitis, a purulent form of sinusitis can develop. Occasionally secretions can be retained in the sinus due to altered ciliary function or obstruction of the opening (ostea) that drains the sinus. Incomplete drainage makes the sinus prone to infection typically with Haemophilus influenza, Streptococcus pneumoniae, Moraxella catarrhalis, Veillonella, Peptococcus, Corynebacterium acnes and certain species of fungi.
When initial treatment such as antibiotics, intranasal steroid sprays and decongestants are ineffective, it may become necessary to perform surgical drainage of the infected sinus. Surgical therapy often involves debridement of the ostea to remove anatomic obstructions and removal of parts of the mucosa. Occasionally a stent (a cylindrical tube which physically holds the lumen of the ostea open) is left in the osta to ensure drainage is maintained even in the presence of postoperative swelling. ENT stents, typically made of stainless steel or plastic, remain in place for several days or several weeks before being removed.
Representative examples of ENT stents that can benefit from being coated with or having incorporated therein a fibrosis-inhibiting agent include lacrimal duct stents, Eustachian tube stents, nasal stents, and sinus stents. In one aspect, the present invention provides for the combination of a lacrimal duct stent and a fibrosis-inhibiting agent or a composition comprising a fibrosis-inhibiting agent.
In another aspect, the present invention provides for the combination of a Eustachian tube stent and a fibrosis-inhibiting agent or a composition comprising a fibrosis-inhibiting agent.
In yet another aspect, the present invention provides for the combination of a sinus stent and a fibrosis-inhibiting agent or a composition comprising a fibrosis-inhibiting agent.
In yet another aspect, the present invention provides for the combination of a nasal stent and a fibrosis-inhibiting agent or a composition comprising a fibrosis-inhibiting agent.
The ENT stent may be a choanal atresia stent composed of two long hollow tubes that are bridged by a flexible transverse tube. See, e.g., U.S. Patent No. 6,606,995. The ENT stent may be an expandable nasal stent for postoperative nasal packing composed of a highly porous, pliable and absorbent foam material capable of expanding outwardly, which has a nonadherent surface. See, e.g., U.S. Patent No. 5,336,163. The ENT stent may be a nasal stent composed of a deformable cylinder with a breathing passageway that has a smooth outer non-absorbent surface used for packing the nasal cavity following surgery. See, e.g., U.S. Patent No. 5,601 ,594. The ENT stent may be a ventilation tube composed of a flexible, plastic, tubular vent with a rectangular flexible flange which is used for the nasal sinuses following endoscopic antrostomy. See, e.g., U.S. Patent No. 5,246,455. The ENT stent may be a ventilating ear tube composed of a shaft and an extended tab which is used for equalizing the pressure between the middle ear and outer ear. See, e.g., U.S. Patent No. 6,042,574. The ENT stent may be a middle ear vent tube composed of a non-compressible, tubular base and an eccentric flange. See, e.g., U.S. Patent No. 5,047,053. ENT stents, which may be combined with one or more agents according to the present invention, include commercially available products such as Genzyme Corporation (Ridgefield, NJ) SEPRAGEL Sinus Stents and MEROGEL Nasal Dressing and Sinus Stents from Medtronic Xomed Surgical Products, Inc. (Jacksonville, FL).
In one aspect, the present invention provides ENT stents that include an anti-scarring agent or a composition that includes an anti-scarring agent. Numerous polymeric and non-polymeric delivery systems for use in ENT stents have been described above. Methods for incorporating fibrosis- inhibiting compositions onto or into the ENT stents include: (a) directly affixing to the stent a fibrosis-inhibiting composition (e.g., by either a spraying process or dipping process as described above, with or without a carrier), (b) directly incorporating into the stent a fibrosis-inhibiting composition (e.g., by either a spraying process or dipping process as described above, with or without a carrier), (c) by coating the stent with a substance such as a hydrogel which will in turn absorb the fibrosis-inhibiting composition, (d) by interweaving fibrosis-inhibiting composition coated thread (or the polymer itself formed into a thread) into the stent structure, (e) by inserting the stent into a sleeve or mesh which is comprised of or coated with a fibrosis-inhibiting composition, (f) constructing the stent itself or a portion of the stent with a fibrosis-inhibiting composition, or (g) by covalently binding the fibrosis-inhibiting agent directly to the stent surface or to a linker (small molecule or polymer) that is coated or attached to the stent surface. For these devices, the coating process can be performed in such a manner as to (a) coat the external surface of the specific stent, (b) coat the internal (luminal) surface of the stent, or (c) coat all or parts of both the interna) and external surfaces of the device.
In addition to coating the device with the fibrosis-inhibiting composition, the fibrosis-inhibiting agent can be mixed with the materials that are used to make the device such that the fibrosis-inhibiting agent is incorporated into the final device. According to the present invention, any fibrosis-inhibiting agent described above can be utilized in the practice of this embodiment. Within one embodiment of the invention, ENT stents may be adapted to release an agent that inhibits one or more of the four general components of the process of fibrosis (or scarring), including: formation of new blood vessels (angiogenesis), migration and proliferation of connective tissue cells (such as fibroblasts or smooth muscle cells), deposition of extracellular matrix (ECM), and remodeling (maturation and organization of the fibrous tissue). By inhibiting one or more of the components of fibrosis (or scarring), the overgrowth of granulation tissue may be inhibited or reduced.
As ENT stents are made in a variety of configurations and sizes, the exact dose administered will vary with device size, surface area and design. However, certain principles can be applied in the application of this art. Drug dose can be calculated as a function of dose per unit area (of the portion of the device being coated), total dose administered, and appropriate surface concentrations of active drug can be determined. Drugs are to be used at concentrations that range from several times more than to 10%, 5%, or even less than 1% of the concentration typically used in a single chemotherapeutic systemic dose application. Preferably, the drug is released in effective concentrations for a period ranging from 1 - 90 days.
Several examples of fibrosis-inhibiting agents for use in ENT stents include the following: ZD-6474, AP-23573, synthadotin, S-0885, aplidine, ixabepilone, IDN-5390, SB-2723005, ABT-518, combretastatin, anecortave acetate, SB-715992, temsirolimus, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, as well as analogues and derivatives of the aforementioned.
Additional examples of anti-scarring agents which can be used include those having a high potency in the assays described herein (1- 10OnM IC5O range) such as isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, dolastatin 15, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17- DMAG, and tacrolimus; those having a mid-potency in the assays described herein (100-500 nM IC50 range) such as loteprednol etabonate, juglone, prednisolone, puromycin, 3-BAABE, cladribine, and mannose-6-phosphate; and those having a low potency in the assays described herein (500- 1000nm range IC50 range) such as 5-azacytidine, Ly333531(ruboxistaurin), and simvastatin.
Regardless of the method of application of the drug to the ENT stent, the exemplary anti-fibrosing agents, used alone or in combination, should be administered under the following dosing guidelines. The total amount (dose) of anti-scarring agent in or on the device may be in the range of about 0.01 μg-10 μg, or 10 μg-10 mg, or 10 mg-250 mg, or 250 mg-1000 mg, or 1000 mg-2500 mg. The dose (amount) of anti-scarring agent per unit area of device surface to which the agent is applied may be in the range of about 0.01 μg/mm2 - 1 μg/mm2, or 1 μg/mm2 - 10 μg/mm2, or 10 μg/mm2 - 250 μg/mm2, 250 μg/mm2 - 1000 μg/mm2, or 1000 μg/mm2 - 2500 μg/mm2.
Provided below are exemplary dosage ranges for various anti- scarring agents that can be used in conjunction with ENT stent devices in accordance with the invention. (A) Angiogenesis inhibitors including Alphastatin, ZD-6474, IDN-5390, SB-2723005, ABT-518, combretastatin, and anecortane, analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10^- 10"4 M of agent is to be maintained on the implant or barrier surface. (B) mTOR inhibitors including AP-23573 and Temsirolimus, analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10^- 10"4 M of agent is to be maintained on the implant or barrier surface. (C) Tubulin antagonists including synthadotin, analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10~8- 10'4 M of agent is to be maintained on the implant or barrier surface. (D) Epithilones including ixabepilone and analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10^- 10"4 M of agent is to be maintained on the implant or barrier surface. (E) Kinesin antagonists including SB- 715992 and analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10~8~ 10"4 M of agent is to be maintained on the implant or barrier surface. (F) TNF alpha antagonists including Etanercept, humicade, adalimumab and analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg o 100 mg. Dose perunit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10^- 10"4 M of agent is to be maintained on the implant or barrier surface. (G) AKT inhibitor including erucylphosphocholine and analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10^- 10"4 M of agent is to be maintained on the implant or barrier surface. (H) FGF Inhibitors including IDN-5390 and analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10^- 10"4 M of agent is to be maintained on the implant or barrier surface. (I) Collagenase Antagonists including S-0885 and analogues and derivatives thereof: total dose not to exceed 1000 mg (range of 0.1 μg to 1000 mg); preferred 1 μg to 500 mg. Dose per unit area of 0.01 μg - 500 μg per mm2; preferred dose of 0.1 μg/mm2 - 100 μg/mm2. Minimum concentration of 10"8- 10'4 M of agent is to be maintained on the implant or barrier surface. (J) NF KAPPA B Inhibitors including Bortezomib and analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10"8- 10"4 M of agent is to be maintained on the implant or barrier surface. (K) Elongation factor-1 alpha inhibitors including aplidine and analogues and derivatives thereof: total dose not to exceed 1000 mg (range of 0.1 μg to 1000 mg); preferred 1 μg to 500 mg. Dose per unit area of 0.01 μg - 500 μg per mm2; preferred dose of 0.1 μg/mm2 - 100 μg/mm2. Minimum concentration of 10"8- 10"4 M of agent is to be maintained on the implant or barrier surface. (L) Tyrosine kinase inhibitors including Gefitinib and analogues and derivatives thereof: total dose not to exceed 1000 mg (range of 0.1 μg to 1000 mg); preferred 1 μg to 500 mg. Dose per unit area of 0.01 μg - 500 μg per mm2; preferred dose of 0.1 μg/mm2 - 100 μg/mm2. Minimum_concentration of 10^- 10"4 M of agent is to be maintained on the implant or barrier surface.
For high potency drugs, the total dose typically should not exceed 200 mg (range of 0.1 μg to 200 mg) and preferably 1 μg to 100 mg; dose per unit area of 0.01 μg - 100 μg per mm2; preferably 0.1 μg/mm2 - 20 μg/mm2; and minimum concentration of 10^- 10"4 M of agent should be maintained on the implant or barrier surface. For mid-potency drugs, the total dose typically should not to exceed 500 mg (range of 1.0 μg to 500 mg) and preferably 1 μg to 200 mg; dose per unit area of 0.01 μg - 200 μg per mm2, preferably 0.1 μg/mm2 - 40 μg/mm2; and minimum concentration of 10" 8 - 10"4 M of agent should be maintained on the implant or barrier surface. For low potency drugs, the total dose typically should not exceed 1000 mg (range of 0.1 μg to 1000 mg), preferably 1 μg to 500 mg; dose per unit area of 0.01 μg - 500 μg per mm2; preferably 0.1 μg/mm2 - 100 μg/mm2; and minimum concentration of 10 s- 10"4 M of agent should to be maintained on the implant or barrier surface. Ear Ventilation Tubes
In another aspect, the present invention provides for the combination of an anti-scarring agent and an ear ventilation tube (also referred to as a tympanostomy tube).
Acute otitis media is the most common bacterial infection, the most frequent indication for surgical therapy, the leading cause of hearing loss and a common cause of impaired language development in children. The cost of treating this condition in children under the age of five is estimated at $5 billion annually in the United States alone. In fact, 85% of all children will have at least one episode of otitis media and 600,000 will require surgical therapy annually. The prevalence of otitis media is increasing and for severe cases surgical therapy is more cost effective than conservative management.
Acute otitis media (bacterial infection of the middle ear) is characterized by Eustachian tube dysfunction leading to failure of the middle ear clearance mechanism. The most common causes of otitis media are Streptococcus pneumoniae (30%), Haemophilus influenza (20%), Branhamella catarrhalis (12%), Streptococcus pyogenes (3%), and Staphylococcus aureus (1.5%). The end result is the accumulation of bacteria, white blood cells and fluid which, in the absence of an ability to drain through the Eustachian tube, results in increased pressure in the middle ear. For many cases antibiotic therapy is sufficient treatment and the condition resolves. However, for a significant number of patients the condition becomes frequently recurrent or does not resolve completely. In recurrent otitis media or chronic otitis media with effusion, there is a continuous build-up of fluid and bacteria that creates a pressure gradient across the tympanic membrane causing pain and impaired hearing. Fenestration of the tympanic membrane (typically with placement of a tympanostomy tube) relieves the pressure gradient and facilitates drainage of the middle ear (through the outer ear instead of through the Eustachian tube - a form of "Eustachian tube bypass"). Recurrent otitis media or otitis media with effusion may be treated with tympanostomy tubes or artificial Eustachian tubes/stents, such as described above. These ventilation tubes are indicated for chronic otitis media with effusion, recurrent acute otitis media, tympanic membrane atelectasis, and complications of acute otitis media in children. The excessive formation of granulation tissue around these devices can result in a decreased functioning of these devices. This can then result in a second procedure to either clear the obstruction or to insert a new device. The incorporation of a fibrosis-inhibiting agent into or onto the ventilation tubes may prevent the overgrowth of this granulation tissue.
Surgical placement of tympanostomy tubes is the most widely used treatment for chronic otitis media because, although not curative, it improves hearing (which in turn improves language development) and reduces the incidence of acute otitis media. Tympanostomy tube placement is one of the most common surgical procedures in the United States with 1.3 million surgical placements per year.
Representative examples of ear ventilation tubes that can benefit from being coated with or having incorporated therein a fibrosis- inhibiting agent include, without limitation, grommet-shaped tubes, T-tubes, tympanostomy tubes, drain tubes, tympanic tubes, otological tubes, myringotomy tubes, artificial Eustachian tubes, Eustachian tube prostheses, and Eustachian stents. Ear ventilation tubes have been made out of, e.g., polytetrafluoroethylene (e.g., TEFLON), silicone, nylon, polyethylene and other polymers, stainless steel, titanium, and gold plated steel.
In one aspect, the ear ventilation tube may be a tympanostomy tube that is used to provide an alternative conduit for ventilation of the middle ear cavity via the external ear canal. Typically, ventilation of the middle ear is performed by conducting a myringotomy, in which a slit or opening in the tympanic membrane is surgically made to alleviate a buildup or reduction of pressure in the middle ear cavity and to drain accumulated fluids. Tympanostomy tubes may be inserted into the surgical slit of the tympanic membrane to serve as a bypass for the normal Eustachian tube, which drains the middle ear cavity under normal conditions. For example, the tympanostomy tube may be an elongated uniform tubular member composed of pure titanium or titanium alloy that has a concavity inwardly spaced from one end that forms a flange. See, e.g., U.S. Patent No. 5,645,584. The tympanostomy tube may be composed of a micro-pitted titanium exterior flangeless surface used to ventilate the middle ear. See, e.g., U.S. Patent No. 4,971 ,076. The tympanostomy tube may be composed of a shaft with a tab that extends outwardly perpendicular from the bottom of the shaft. See, e.g., U.S. Patent No. 6,042,574. The tympanostomy tube may be a permanent ear ventilation device composed of an elongated tubular base having a flange eccentrically connected made of a non-compressible material. See, e.g., U.S. Patent No. 5,047,053. The tympanostomy tube may be composed of a cap-plug, central body and end cap, which together form a plurality of lumens within the tube. See, e.g., U.S. Patent No. 5,851 ,199. The tympanostomy tube may be composed of a microporous resin cured to form a gas-permeable matrix containing a homogenous dispersion of silver particles capable of migrating to the surface of the tube sidewalls to provide antimicrobial activity. See, e.g., U.S. Patent No. 6,361 ,526. The tympanostomy tube may be composed of tubular body and a rib structure that projects outwardly to define a channel spiraling around the tubular body. See, e.g., U.S. Patent No. 5,775,336. The tympanostomy tube may be composed of an integral cutting tang extending from one of two flanges of a grommet for incising the tympanic membrane. See, e.g., U.S. Patent Nos. 5,827,295 and 5,643,280. The tympanostomy tube may be composed of a tubular member having two opposed flanges in which the insertion of the tube is facilitated by a cutting edge on the flange which induces an incision of the tympanic membrane. See, e.g., U.S. Patent Nos. 5,489,286; 5,466,239; 5,254,120 and 5,207,685. Other tympanostomy tubes are described in, e.g., U.S. Patent Nos. 6,406,453; 5,178,623; 4,808,171 and 4,744,792. In another aspect, the ear ventilation tube may be used to establish the normal function of the Eustachian tube and thus, attempt to resolve the stenosis that prevents its normal function. Fluid in the middle ear cavity normally secretes away from the tympanic membrane and thus, restoring the normal function of the Eustachian tube may provide optimal ventilation and drainage. For example, the ventilation tube may be an Eustachian stent composed of a hollow tubular body having a compressible core with two connected parallel arms and a radially-oriented flange, which is placed in the Eustachian tube to maintain patency. See, e.g., U.S. Patent No. 6,589,286. The ventilation tube may be an Eustachian tube prosthesis composed of a flexible tube having a flange that extends radially for positioning within the Eustachian tube passageway. See, e.g., U.S. Patent No. 4,015,607.
Tympanostomy tubes, which may be combined with one or more agents according to the present invention, include commercially available products. For example, Medtronic Xomed, Inc. (Jackonsville, FL) sells a variety of ear ventilation tubes, including Long-Term Ventilation Tubes and Grommet Style Ventilation Tubes, including ARMSTRONG Grommets, GOODE T-Grommets, VENTURI Style Ventilation Tubes, SHEEHY Type Collar Buttons, REUTER Bobbins, COHEN T-Grommets, and SOILEAU TYTAN Titanium Tubes. Micromedics, Inc. (Eagan, MN) also sells a variety of ear ventilation tubes, including BAXTER Bevel Buttons, TINY TOUMA, SPOONER, TOUMA T-Tubes, SHOEHORN Bobbins, SHAH, and SILVERSTEIN MICROWICK Eustachian Tubes. Gyrus ENT LLC (Bartlett, TN) also sells a variety of ear ventilation tubes, including ULTRASIL Ventilation Tubes, RICHARDS COLLAR Bobbins, BALDWIN BUTTERFLY Ventilation Tubes and PAPARELLA 2000 Tubes.
In one aspect, the present invention provides ear ventilation tube devices that include an anti-scarring agent or a composition that includes an anti-scarring agent. Numerous polymeric and non-polymeric delivery systems for use in ear ventilation tubes have been described above. These compositions can further include one or more fibrosis- inhibiting agents such that the overgrowth of granulation tissue is inhibited or reduced.
Numerous polymeric and non-polymeric delivery systems for use in ear ventilation tubes have been described above. Methods for incorporating the fibrosis-inhibiting agent or a composition comprising the fibrosis-inhibiting agent into or onto the device includes: (a) directly affixing to the device a fibrosis-inhibiting composition (e.g., by either a spraying process or dipping process as described above, with or without a carrier), (b) directly incorporating into the device a fibrosis-inhibiting composition (e.g., by either a spraying process or dipping process as described above, with or without a carrier, (c) by coating the device with a substance such as a hydrogel which will in turn absorb the fibrosis-inhibiting composition, (d) by interweaving fibrosis-inhibiting composition coated thread (or the polymer itself formed into a thread) into the device structure, (e) constructing the device itself or a portion of the device with a fibrosis-inhibiting composition, or (f) by covalently binding the fibrosis-inhibiting agent directly to the device surface or to a linker (small molecule or polymer) that is coated or attached to the device surface. The coatings can be applied to different portions of the device. For example, the coating can be (a) a coating applied to the external surface of the ear ventilation tube; (b) a coating applied to the internal (luminal) surface of the ear ventilation tube; or (c) a coating applied to all or parts of both surfaces.
In addition to coating the device with the fibrosis-inhibiting composition, the fibrosis-inhibiting agent can be mixed with the materials that are used to make the device such that the fibrosis-inhibiting agent is incorporated into the final device.
In addition to incorporation of a fibrosis-inhibiting agent into or onto the device, another biologically active agent can be incorporated into or onto the device, for example an anti-inflammatory (e.g., dexamethazone or aspirin) and/or an antibiotic (e.g., amoxicillin, trimethoprim- sulfamethoxazole, azithromycin, clarithromycin, amoxicillin-clavulanate, cefprozil, cefuroxime, cefpodoxime, or cefdinir).
According to the present invention, any fibrosis-inhibiting agent described above can be utilized in the practice of this embodiment. Within one embodiment of the invention, ear ventilation tubes may be adapted to release an agent that inhibits one or more of the four general components of the process of fibrosis (or scarring), including: formation of new blood vessels (angiogenesis), migration and proliferation of connective tissue cells (such as fibroblasts or smooth muscle cells), deposition of extracellular matrix (ECM), and remodeling (maturation and organization of the fibrous tissue). By inhibiting one or more of the components of fibrosis (or scarring), the overgrowth of granulation tissue may be inhibited or reduced.
As ear ventilation tube devices are made in a variety of configurations and sizes, the exact dose administered will vary with device size, surface area and design. However, certain principles can be applied in the application of this art. Drug dose can be calculated as a function of dose per unit area (of the portion of the device being coated), total dose administered, and appropriate surface concentrations of active drug can be determined. Drugs are to be used at concentrations that range from several times more than to 10%, 5%, or even less than 1% of the concentration typically used in a single chemotherapeutic systemic dose application. Preferably, the drug is released in effective concentrations for a period ranging from 1 - 90 days.
Several examples of fibrosis-inhibiting agents for use in ear ventilation tubes include the following: ZD-6474, AP-23573, synthadotin, S- 0885, aplidine, ixabepilone, IDN-5390, SB-2723005, ABT-518, combretastatin, anecortave acetate, SB-715992, temsirolimus, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, as well as analogues and derivatives of the aforementioned.
Additional examples of anti-scarring agents which can be used include those having a high potency in the assays described herein (1- 10OnM IC5O range) such as isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, dolastatin 15, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17- DMAG, and tacrolimus; those having a mid-potency in the assays described herein (100-500 nM IC5O range) such as loteprednol etabonate, juglone, prednisolone, puromycin, 3-BAABE, cladribine, and mannose-6-phosphate; and those having a low potency in the assays described herein (500- 1000nm range IC50 range) such as 5-azacytidine, Ly333531(ruboxistaurin), and simvastatin.
Regardless of the method of application of the drug to the ear ventilation tube device, the exemplary anti-fibrosing agents, used alone or in combination, should be administered under the following dosing guidelines. The total amount (dose) of anti-scarring agent in or on the device may be in the range of about 0.01 μg-10 μg, or 10 μg-10 mg, or 10 mg-250 mg, or 250 mg-1000 mg, or 1000 mg-2500 mg. The dose (amount) of anti-scarring agent per unit area of device surface to which the agent is applied may be in the range of about 0.01 μg/mm2 - 1 μg/mm2, or 1 μg/mm2 - 10 μg/mm2, or 10 μg/mm2 - 250 μg/mm2, 250 μg/mm2 - 1000 μg/mm2, or 1000 μg/mm2 - 2500 μg/mm2.
Provided below are exemplary dosage ranges for various anti- scarring agents that can be used in conjunction with ear ventilation tube devices in accordance with the invention. (A) Angiogenesis inhibitors including Alphastatin, ZD-6474, IDN-5390, SB-2723005, ABT-518, combretastatin, and anecortane, analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10^- 10"4 M of agent is to be maintained on the implant or barrier surface. (B) mTOR inhibitors including AP-23573 and Temsirolimus, analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10"8- 10"4 M of agent is to be maintained on the implant or barrier surface. (C) Tubulin antagonists including synthadotin, analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10'8- 10"4 M of agent is to be maintained on the implant or barrier surface. (D) Epithilones including ixabepilone and analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10^- 10"4 M of agent is to be maintained on the implant or barrier surface. (E) Kinesin antagonists including SB- 715992 and analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10"8 - 10"4 M of agent is to be maintained on the implant or barrier surface. (F) TNF alpha antagonists including Etanercept, humicade, adalimumab and analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10"8- 10'4 M of agent is to be maintained on the implant or barrier surface. (G) AKT inhibitor including erucylphosphocholine and analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10^- 10"4 M of agent is to be maintained on the implant or barrier surface. (H) FGF Inhibitors including IDN-5390 and analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10^ - 10"4 M of agent is to be maintained on the implant or barrier surface. (I) Collagenase Antagonists including S-0885 and analogues and derivatives thereof: total dose not to exceed 1000 mg (range of 0.1 μg to 1000 mg); preferred 1 μg to 500 mg. Dose per unit area of 0.01 μg - 500 μg per mm2; preferred dose of 0.1 μg/mm2 - 100 μg/mm2. Minimum concentration of 10"8- 10~4 M of agent is to be maintained on the implant or barrier surface. (J) NF KAPPA B Inhibitors including Bortezomib and analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10'8- 10"4 M of agent is to be maintained on the implant or barrier surface. (K) Elongation factor-1 alpha inhibitors including aplidine and analogues and derivatives thereof: total dose not to exceed 1000 mg (range of 0.1 μg to 1000 mg); preferred 1 μg to 500 mg. Dose per unit area of 0.01 μg - 500 μg per mm2; preferred dose of 0.1 μg/mm2 - 100 μg/mm2. Minimum concentration of 10"8- 10'4 M of agent is to be maintained on the implant or barrier surface. (L) Tyrosine kinase inhibitors including Gefitinib and analogues and derivatives thereof: total dose not to exceed 1000 mg (range of 0.1 μg to 1000 mg); preferred 1 μg to 500 mg. Dose per unit area of 0.01 μg - 500 μg per mm2; preferred dose of 0.1 μg/mm2 - 100 μg/mm2. Minimum concentration of 10"8- 10'4 M of agent is to be maintained on the implant or barrier surface.
For high potency drugs, the total dose typically should not exceed 200 mg (range of 0.1 μg to 200 mg) and preferably 1 μg to 100 mg; dose per unit area of 0.01 μg - 100 μg per mm2; preferably 0.1 μg/mm2 - 20 μg/mm2; and minimum concentration of 10~8- 10"4 M of agent should be maintained on the implant or barrier surface. For mid-potency drugs, the total dose typically should not to exceed 500 mg (range of 1.0 μg to 500 mg) and preferably 1 μg to 200 mg; dose per unit area of 0.01 μg - 200 μg per mm2, preferably 0.1 μg/mm2 - 40 μg/mm2; and minimum concentration of 10" 8- 10"4 M of agent should be maintained on the implant or barrier surface. For low potency drugs, the total dose typically should not exceed 1000 mg (range of 0.1 μg to 1000 mg), preferably 1 μg to 500 mg; dose per unit area of 0.01 μg - 500 μg per mm2; preferably 0.1 μg/mm2 - 100 μg/mm2; and minimum concentration of 10'8- 10"4 M of agent should to be maintained on the implant or barrier surface.
Intraocular Implants
In another aspect, the present invention provides for the combination of an anti-scarring agent and an intraocular implant.
In one embodiment, the intraocular implant is an intraocular lens device for the prevention of lens (e.g., anterior or posterior lens) opacification. Eyesight deficiencies that may be treated with intraocular lenses include, without limitation, cataracts, myopia, hyperopia, astigmatism and other eye diseases. Intraocular lenses are most commonly used to replace the natural crystalline lens which is removed during cataract surgery. A cataract results from a change in the transparency of the normal crystalline lens in the eye. When the lens becomes opaque from calcification (e.g., yellow and/or cloudy), the light cannot enter the eye properly and vision is impaired.
Implantation of intraocular lenses into the eye is a standard technique to restore useful vision in diseased or damaged eyes. The number of intraocular lenses implanted in the United States has grown exponentially over the last decade. Currently, over 1 million intraocular lenses are implanted annually, with the vast majority (90%) being placed in the posterior chamber of the eye. The intent of intraocular lenses is to replace the natural crystalline lens {i.e., aphakic eye) or to supplement and correct refractive errors {i.e., phakic eye, natural crystalline lens is not removed).
Implanted intraocular lenses may develop complications caused by mechanical trauma, inflammation, infection or optical problems. Mechanical and inflammatory injury may lead to reduced vision, chronic pain, secondary cataracts, corneal decompensation, cystoid macular edema, hyphema, uveitis or glaucoma. One common problem that occurs with cataract extraction is opacification which results from the tissue's reaction to the surgical procedure or to the artificial lens. Opacification leads to clouding of the intraocular lens, thus reducing the long-term benefits. Opacification typically results when proliferation and migration of epithelial cells occur along the posterior capsule behind the intraocular lens. Subsequent surgery may be required to correct this reaction; however, it involves a complex technical process and may lead to further serious, sight- threatening complications. Therefore, coating or incorporating the intraocular lens with a fibrosis-inhibiting agent may reduce these complications.
Representative examples of intraocular lenses that can benefit from being coated with or having incorporated therein a fibrosis-inhibiting agent include, without limitation, polymethylmethacrylate (PMMA) intraocular lenses, silicone intraocular lenses, achromatic lenses, pseudophakos, phakic lenses, aphakic lenses, multi-focal intraocular lenses, hydrophilic and hydrophobic acrylic intraocular lenses, intraocular implants, optic lenses and rigid gas permeable (RGP) lenses.
In one aspect, intraocular lenses may be foldable or rigid. The foldable lenses may be inserted in a small incision site using a tiny tube whereas the hard lenses are inserted through a larger incision site. Foldable lenses may be composed of silicone, acrylic or hydrogel whereas rigid lenses may be composed of hard polymeric compositions (PMMA).
In one aspect, the intraocular lens may be used as an implant for the treatment of cataracts, where the natural crystalline lens of the eye has been removed (i.e., aphakic lens). For example, the intraocular lens may be composed of two lenses having distinct refractive indices and distinct optical powers being joined together as an achromatic lens that may be connected within a posterior or anterior chamber of the eye. See, e.g., U.S. Patent No. 5,201 ,762. The intraocular lens may be secured in the posterior chamber by a system of posts that protrude through the iris attached to retaining rings. See, e.g., U.S. Patent No. 4,053,953. The intraocular lens may be hard with a shape memory which is capable of deforming for insertion into the eye but will harden at normal body temperature. See, e.g., U.S. Patent No. 4,946,470. The intraocular lens may be coated with proteins, polypeptides, polyamino acids, polyamines or carbohydrates bound to the surface of the implant See, e.g., U.S. Patent Nos. 6,454,802 and 6,106,554. Other examples of aphakic intraocular lenses are described in, e.g., U.S. Patent Nos. 6,599,317; 6,585,768; 6,558,419; 6,533,813; 6,210,438; 5,266,074; 4,753,654; 4,718,904 and 4,704,123.
In another aspect, the intraocular lens may be used as a corrective implant for vision impairment, where the natural crystalline lens of the eye has not been removed (i.e., phakic lens). For example, the intraocular lens may be a narrow profile, glare reducing, phakic anterior chamber lens that may be composed of an optic zone and a transition zone that has a curvature shaped to minimize direct glare. See, e.g., U.S. Patent No. 6,596,025. The intraocular lens may be a self-centering phakic lens inserted in the posterior chamber lens in which arms (i.e., haptic bodies) extend outwardly and protrude into the pupil such that the iris provides centering force to keep lens in place. See, e.g., U.S. Patent No. 6,015,435. The intraocular lens may be composed of a circumferential edge and two haptics extending from the edge to a transverse member which is substantially straight or bowed inward toward the lens. See, e.g., U.S. Patent No. 6,241,777. Other examples of phakic intraocular lenses are described in, e.g., U.S. Patent Nos. 6,228,115; 5,480,428 and 5,222,981.
In another aspect, the intraocular lens may be a multi-focal lens capable of variable accommodation to enable the user to look through different portions of the lens to achieve different levels of focusing power. For example, the intraocular lens may be a variable focus lens composed of two lens portions with an optical zone between the lenses which may contain a fluid reservoir and channel containing charged solution. See, e.g., U.S. Patent No. 5,443,506.
In another aspect, intraocular lenses may be deformable such that the lens may be folded for insertion through a tunnel incision. For example, the intraocular lens may be composed of a lens with fixation members for retaining the lens in the eye which may be configured for folding or rolling from a normal optical condition into an insertion condition to permit the lens to be passed through an incision into the eye. See, e.g., U.S. Patent No. 5,476,513. The intraocular lens may be composed of a resilient, deformable silicone based optic with a fixation means coupled to the optic for retaining the optic in the eye. See, e.g., U.S. Patent No. 5,201,763. The intraocular lens may be composed of a copolymer of three constituents which may be deformable from its original shape. See, e.g., U.S. Patent No. 5,359,021. The intraocular lens may be composed of a transparent, flexible membrane with an interior sac and an attached bladder, in which optical fluid medium is shunted from the optical element to the bladder to aid in its deformity during insertion. See, e.g., U.S. Patent No. 6,048,364. The intraocular lens may be a biocomposite composed of an optic portion made of high water content hydrogel capable of being folded and a haptic portion of low water content hydrogel having strength and rigidity. See, e.g., U.S. Patent No. 5,211 ,662. Other deformable intraocular lenses are described in, e.g., U.S. Patent Nos. 6,267,784; 5,507,806 and U.S. Patent Application Publication No. 2003/0114928A1.
Other related devices and/or compositions (e.g., insertion devices) that may be used in conjunction with intraocular lenses are described in, e.g., U.S. Patent Nos. 6,629,979; 6,187,042; 6,113,633; 4,740,282 and U.S. Patent Application Publication Nos. 2003/0212409A1 and 2003/0187455A1
Intraocular lenses, which may be combined with one or more agents according to the present invention, include commercially available products. For example, Alcon Laboratories, Inc. (Fort Worth, TX) sells the foldable ACRYSOF Intraocular Lens. Bausch & Lomb Surgical, Inc. (San Dimas, CA) sells the foldable SOFLEX SE Intraocular Lens. Advanced Medical Optics, lnc (Santa Ana, CA) sells the CLARIFLEX Foldable Intraocular Lens, SENSAR Acrylic Intraocular Lens, and PHACOFLEX Il SI40NB and SI30NB.
The intraocular implant may comprise the fibrosis-inhibiting agent or a composition that includes the fibrosis-inhibiting agent directly. Alternatively, or in addition, the agent may be coated, absorbed into, or bound onto the lens surface (e.g., to the haptics), or may be released from a hole (pore) or cavity outside the optical part of the lens surface.
The intraocular implants of the invention may be used in various surgical procedures. For example, the intraocular implant may be used in conjunction with a transplant for the cornea. Synthetic corneas can be used in patients loosing vision due to a degenarative cornea. Implanted synthetic corneas can restore patient vision, however, they often induce a fibrous foreign body response that limits their use. The intraocular implant of the present invention can prevent the foreign body response to the synthetic cornea and extend the cornea longevity. In another example, the synthetic cornea itself is coated with the agents of the invention, thus minimizing tissue reaction to corneal implantation.
In another aspect, the intraocular lens may be used in conjunction with treatment of secondary cataract after extracapsular cataract extraction.
As described above, the present invention provides intraocular lenses and other implants that include an anti-scarring agent or a composition that includes an anti-scarring agent.
Numerous polymeric and non-polymeric delivery systems for use in intraocular implants have been described above.
Methods for coating fibrosis-inhibiting compositions onto or into the implants include: (a) directly affixing to the implants a fibrosis- inhibiting composition (e.g., by either a spraying process or dipping process as described above, with or without a carrier), (b) directly incorporating into the implant a fibrosis-inhibiting composition (e.g., by either a spraying process or dipping process as described above, with or without a carrier (c) by coating the implant with a substance such as a hydrogel which will in turn absorb the fibrosis-inhibiting composition, (d) constructing the implant itself or a portion of the lens with a fibrosis-inhibiting composition, or (e) by covalently binding the fibrosis-inhibiting agent directly to the lens surface or to a linker (small molecule or polymer) that is coated or attached to the implant surface. For these devices, the coating process can be performed in such a manner as to (a) coat the posterior surface of the specific implant, (b) coat the anterior surface of the implant or (c) coat all or parts of both the posterior and anterior surfaces of the device. The protruding arms of the implant can also be coated with the fibrosis-inhibiting agent.
In addition to coating the device with the fibrosis-inhibiting composition, the fibrosis-inhibiting agent can be mixed with the materials that are used to make the device such that the fibrosis-inhibiting agent is incorporated into the final device.
The process of coating these implants with a fibrosis-inhibiting agent or incorporating the fibrosis-inhibiting agent into the implant and the materials selected for these processes are such that they do not significantly alter the refractive index of the intraocular implant or the visible light transmission of the implant or lens.
According to the present invention, any scarring agent described above can be utilized in the practice of this embodiment. Within one embodiment of the invention, intraocular implants may be adapted to release an agent that inhibits one or more of the four general components of the process of fibrosis (or scarring), including: formation of new blood vessels (angiogenesis), migration and proliferation of connective tissue cells (such as fibroblasts or smooth muscle cells), deposition of extracellular matrix (ECM), and remodeling (maturation and organization of the fibrous tissue).. By inhibiting one or more of the components of fibrosis (or scarring), the overgrowth of granulation tissue may be inhibited or reduced.
As intraocular implants are made in a variety of configurations and sizes, the exact dose administered will vary with device size, surface area and design. However, certain principles can be applied in the application of this art. Drug dose can be calculated as a function of dose per unit area (of the portion of the device being coated), total dose administered, and appropriate surface concentrations of active drug can be determined. Drugs are to be used at concentrations that range from several times more than to 10%, 5%, or even less than 1% of the concentration typically used in a single chemotherapeutic systemic dose application. Preferably, the drug is released in effective concentrations for a period ranging from 1 - 90 days.
Several examples of fibrosis-inhibiting agents for use in intraocular implants include the following: ZD-6474, AP-23573, synthadotin, S-0885, aplidine, ixabepilone, IDN-5390, SB-2723005, ABT-518, combretastatin, anecortave acetate, SB-715992, temsirolimus, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, as well as analogues and derivatives of the aforementioned.
Additional examples of anti-scarring agents which can be used include those having a high potency in the assays described herein (1- 10OnM IC50 range) such as isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, dolastatin 15, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17- DMAG, and tacrolimus; those having a mid-potency in the assays described herein (100-500 nM IC50 range) such as loteprednol etabonate, juglone, prednisolone, puromycin, 3-BAABE, cladribine, and mannose-6-phosphate; and those having a low potency in the assays described herein (500- 1000nm range IC50 range) such as 5-azacytidine, Ly333531(ruboxistaurin), and simvastatin. Regardless of the method of application of the drug to the intraocular implant, the exemplary anti-fibrosing agents, used alone or in combination, should be administered under the following dosing guidelines. The total amount (dose) of anti-scarring agent in or on the device may be in the range of about 0.01 μg-10 μg, or 10 μg-10 mg, or 10 mg-250 mg, or 250 mg-1000 mg, or 1000 mg-2500 mg. The dose (amount) of anti-scarring agent per unit area of device surface to which the agent is applied may be in the range of about 0.01 μg/mm2 - 1 μg/mm2, or 1 μg/mm2 - 10 μg/mm2, or 10 μg/mm2 - 250 μg/mm2, 250 μg/mm2 - 1000 μg/mm2, or 1000 μg/mm2 - 2500 μg/mm2.
Provided below are exemplary dosage ranges for various anti- scarring agents that can be used in conjunction with intraocular implants in accordance with the invention. (A) Angiogenesis inhibitors including Alphastatin, ZD-6474, IDN-5390, SB-2723005, ABT-518, combretastatin, and anecortane, analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10^- 10"4 M of agent is to be maintained on the implant or barrier surface. (B) mTOR inhibitors including AP-23573 and Temsirolimus, analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10"8- 10"4 M of agent is to be maintained on the implant or barrier surface. (C) Tubulin antagonists including synthadotin, analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10^- 10"4 M of agent is to be maintained on the implant or barrier surface. (D) Epithilones including ixabepilone and analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10^- 10"4 M of agent is to be maintained on the implant or barrier surface. (E) Kinesin antagonists including SB- 715992 and analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10^- 10"4 M of agent is to be maintained on the implant or barrier surface. (F) TNF alpha antagonists including Etanercept, humicade, adalimumab and analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10 s- 10"4 M of agent is to be maintained on the implant or barrier surface. (G) AKT inhibitor including erucylphosphocholine and analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10"8- 10"4 M of agent is to be maintained on the implant or barrier surface. (H) FGF Inhibitors including IDN-5390 and analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10^- 10"4 M of agent is to be maintained on the implant or barrier surface. (I) Collagenase Antagonists including S-0885 and analogues and derivatives thereof: total dose not to exceed 1000 mg (range of 0.1 μg to 1000 mg); preferred 1 μg to 500 mg. Dose per unit area of 0.01 μg - 500 μg per mm2; preferred dose of 0.1 μg/mm2 - 100 μg/mm2. Minimum concentration of 10^- 10"4 M of agent is to be maintained on the implant or barrier surface. (J) NF KAPPA B Inhibitors including Bortezomib and analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10"8- 10"4 M of agent is to be maintained on the implant or barrier surface. (K) Elongation factor-1 alpha inhibitors including aplidine and analogues and derivatives thereof: total dose not to exceed 1000 mg (range of 0.1 μg to 1000 mg); preferred 1 μg to 500 mg. Dose per unit area of 0.01 μg - 500 μg per mm2; preferred dose of 0.1 μg/mm2 - 100 μg/mm2. Minimum concentration of 10'8- 10"4 M of agent is to be maintained on the implant or barrier surface. (L) Tyrosine kinase inhibitors including Gefitinib and analogues and derivatives thereof: total dose not to exceed 1000 mg (range of 0.1 μg to 1000 mg); preferred 1 μg to 500 mg. Dose per unit area of 0.01 μg - 500 μg per mm2; preferred dose of 0.1 μg/mm2 - 100 μg/mm2. Minimum concentration of 10'8- 10"4 M of agent is to be maintained on the implant or barrier surface.
For high potency drugs, the total dose typically should not exceed 200 mg (range of 0.1 μg to 200 mg) and preferably 1 μg to 100 mg; dose per unit area of 0.01 μg - 100 μg per mm2; preferably 0.1 μg/mm2 - 20 μg/mm2; and minimum concentration of 10"- 10"4 M of agent should be maintained on the implant or barrier surface. For mid-potency drugs, the total dose typically should not to exceed 500 mg (range of 1.0 μg to 500 mg) and preferably 1 μg to 200 mg; dose per unit area of 0.01 μg - 200 μg per mm2, preferably 0.1 μg/mm2 - 40 μg/mm2; and minimum concentration of 10" 8- 10"4 M of agent should be maintained on the implant or barrier surface. For low potency drugs, the total dose typically should not exceed 1000 mg (range of 0.1 μg to 1000 mg), preferably 1 μg to 500 mg; dose per unit area of 0.01 μg - 500 μg per mm2; preferably 0.1 μg/mm2 - 100 μg/mm2; and minimum concentration of 10^- 10"4 M of agent should to be maintained on the implant or barrier surface.
Hypertrophic Scars and Keloids
In another aspect, the present invention provides for the combination of an anti-scarring agent and a device for use in treating hypertrophic scars and keloids. Hypertrophic scars and keloids are the result of an excessive fibroproliferative wound healing response. Briefly, healing of wounds and scar formation occurs in three phases: inflammation, proliferation, and maturation. The first phase, inflammation, occurs in response to an injury which is severe enough to break the skin. During this phase, which lasts 3 to 4 days, blood and tissue fluid form an adhesive coagulum and fibrinous network which serves to bind the wound surfaces together. This is then followed by a proliferative phase in which there is ingrowth of capillaries and connective tissue from the wound edges, and closure of the skin defect. Finally, once capillary and fibroblastic proliferation has ceased, the maturation process begins wherein the scar contracts and becomes less cellular, less vascular, and appears flat and white. This final phase may take between 6 and 12 months. If too much connective tissue is produced and the wound remains persistently cellular, the scar may become red and raised. If the scar remains within the boundaries of the original wound it is referred to as a hypertrophic scar, but if it extends beyond the original scar and into the surrounding tissue, the lesion is referred to as a keloid. Hypertrophic scars and keloids are produced during the second and third phases of scar formation. Several wounds are particularly prone to excessive endothelial and fibroblastic proliferation, including burns, open wounds, and infected wounds. With hypertrophic scars, some degree of maturation occurs and gradual improvement occurs. In the case of keloids however, an actual tumor is produced which can become quite large. Spontaneous improvement in such cases rarely occurs.
A variety of devices for treating hypertrophic scars and keloids have been described. For example, the device may be an external tissue expansion device composed of two suture steel plates with adhesive attached foam cushions which apply constant continuous low grade force to skin and tissue to provide removal of hypertrophic scars and keloids. See, e.g., U.S. Patent No. 6,254,624. The device may be a masking element which is pressed onto the scar tissue with an adjustable force by means of a pressure control unit and is connected with inflatable or suction members in the masking element. See, e.g., U.S. Patent No. 6,013,094. The treatment may be a device having locking elements and grasping structures such that the dermal and epidermal layers of a skin wound can be pushed together such that the tissue edges are abutting, such that a wound may be closed with minimal scarring. See, e.g., U.S. Patent No. 5,591 ,206.
In another aspect, the hypertrophic scar or keloid may be treated by using a device in conjunction with a coating or sheet that may be used to deliver either anti-scarring agents alone, or anti-scarring compositions as described above. For example, the coating or sheet may be a copolymer composed of a hydrophilic polymer, such as polyethylene glycol, that is bound to a polymer that adsorbs readily to the surfaces of body tissues, such as phenylboronic acid. See, e.g., U.S. Patent No. 6,596,267. The coating or sheet may be a self-adhering silicone sheet which is impregnated with an antioxidant and/or antimicrobial. See, e.g., U.S. PatentJsJo. 6,572,878. The coating or sheet may be a wound dressing garment composed of an outer pliable layer and a self-adhesive inner gel lining which serves as a dressing for contacting wounds. See, e.g., U.S. Patent No. 6,548,728. The coating or sheet may be a liquid composition composed of a film-forming carrier such as a collodion which contains one or more active ingredients such as a topical steroid, silicone gel and vitamin E. See, e.g., U.S. Patent No. 6,337,076. The coating or sheet may be a bandage with a scar treatment pad with a layer of silicone elastomer or silicone gel. See, e.g., U.S. Patent Nos. 6,284,941 and 5,891,076.
In another aspect, a medical device may be used in conjunction with an injectable composition that may be directly injected into a hypertrophic scar or keloid, in order to prevent the progression of these lesions. The frequency of injections will depend upon the release kinetics of the polymer used (if present), and the clinical response. This therapy is of particular value in the prophylactic treatment of conditions which are known to result in the development of hypertrophic scars and keloids (e.g., burns), and is preferably initiated after the proliferative phase has had time to progress (approximately 14 days after the initial injury), but before hypertrophic scar or keloid development. For example, an injectable treatment for hypertrophic scars and keloids may include the administration of an effective amount of angiogenesis inhibitor (e.g., fumagillol, thalidomide) as a systemic or local treatment to decrease excessive scarring. See, e.g., U.S. Patent No. 6,638,949. The injectable treatment may be a cryoprobe containing cryogen whereby it is positioned within the hypertrophic scar or keloid to freeze the tissue. See, e.g., U.S. Patent No. 6,503,246. The injectable treatment may be a method of locally administering an amount of botulinum toxin in or in close proximity to the skin wound, such that the healing is enhanced. See, e.g., U.S. Patent No. 6,447,787. The injectable treatment may be a method of administering an antifibrotic amount of fluoroquinolone to prevent or treat scar tissue formation. See, e.g., U.S. Patent No. 6,060,474. The injectable treatment may be a composition of an effective amount of calcium antagonist and protein synthesis inhibitor sufficient to cause matrix degradation at a scar site so as to control scar formation. See, e.g., U.S. Patent No. 5,902,609. The injectable treatment may be a composition of non-biodegradable microspheres with a substantial surface charge in a pharmaceutically acceptable carrier. See, e.g., U.S. Patent No. 5,861 ,149. The injectable treatment may be a composition of endothelial cell growth factor and heparin which may be administered topically or by intralesional injection. See, e.g., U.S. Patent No. 5,500,409.
Treatments and devices used for hypertrophic scars and keloids, which may be combined with one or more agents according to the present invention, include commercially available products. Representative products include, for example, PROXIDERM External Tissue Expansion product for wound healing from Progressive Surgical Products (Westbury, NY), CICA-CARE Gel Sheet dressing product from Smith & Nephew Healthcare Ltd. (India), and MEPIFORM Self-Adherent Silicone Dressing from Molnlycke Health Care (Eddystone, PA).
In one aspect, devices for the treatment of hypertrophic scars and keloids may be combined with a topical or injectable composition that includes an anti-scarring agent and a polymeric carrier suitable for application on or into hypertrophic scars or keloids. Incorporation of a fibrosis-inhibiting agent into a topical formulation or an injectable formulation is one approach to treat this condition. The topical formulation can be in the form of a solution, a suspension, an emulsion, a gel, an ointment, a cream, film or mesh. The injectable formulation can be in the form of a solution, a suspension, an emulsion or a gel. Polymeric and non-polymeric components that can be used to prepare these topical or injectable compositions are described above.
In another embodiment, the therapeutic agent can be incorporated into a secondary carrier (e.g., micelles, liposomes, emulsions, microspheres, nanospheres etc, as described above). Microsphere and nanospheres may include degradable polymers. Degradable polymers that can be used include poly(hydroxyl esters) (e.g., PLGA, PLA, PCL, and the like) as well as polyanhydrides, polyorthoesters and polysaccharides (e.g., chitosan and alginates).
Within another aspect of the invention, these fibrosis-inhibiting agent/secondary carrier compositions can be a) incorporated directly into or onto the device, b) incorporated into a solution, c) incorporated into a gel or viscous solution, d) incorporated into the composition used for coating the device or e) incorporated into or onto the device following coating of the device with a coating composition. For example, fibrosis-inhibiting agent loaded PLGA microspheres can be incorporated into a polyurethane coating solution which is then coated onto the device.
According to the present invention, any fibrosis-inhibiting agent described above can be utilized in the practice of this embodiment. Within one embodiment of the invention, devices for the treatment of hypertrophic scars and keloids may be adapted to release an agent that inhibits one or more of the four general components of the process of fibrosis (or scarring), including: formation of new blood vessels (angiogenesis), migration and proliferation of connective tissue cells (such as fibroblasts or smooth muscle cells), deposition of extracellular matrix (ECM), and remodeling (maturation and organization of the fibrous tissue). By inhibiting one or more of the components of fibrosis (or scarring), the overgrowth of granulation tissue may be inhibited or reduced.
As devices for preventing hypertrophic scarring or keloids are made in a variety of configurations and sizes, the exact dose administered will vary with device size, surface area and design. However, certain principles can be applied in the application of this art. Drug dose can be calculated as a function of dose per unit area (of the portion of the device being coated), total dose administered, and appropriate surface concentrations of active drug can be determined. Drugs are to be used at concentrations that range from. several times more than to.10%, 5%, or even less than 1 % of the concentration typically used in a single chemotherapeutic systemic dose application. Preferably, the drug is released in effective concentrations for a period ranging from 1 - 90 days.
Several examples of fibrosis-inhibiting agents for use devices for treating hypertrophic scars and keloids include the following: ZD-6474, AP-23573, synthadotin, S-0885, aplidine, ixabepilone, IDN-5390, SB- 2723005, ABT-518, combretastatin, anecortave acetate, SB-715992, temsirolimus, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, as well as analogues and derivatives of the aforementioned.
Additional examples of anti-scarring agents which can be used include those having a high potency in the assays described herein (1- 10OnM IC50 range) such as isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, dolastatin 15, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17- DMAG, and tacrolimus; those having a mid-potency in the assays described herein (100-500 nM IC50 range) such as loteprednol etabonate, juglone, prednisolone, puromycin, 3-BAABE, cladribine, and mannose-6-phosphate; and those having a low potency in the assays described herein (500- 1000nm range IC50 range) such as 5-azacytidine, Ly333531(ruboxistaurin), and simvastatin.
Regardless of the method of application of the drug to the device, the exemplary anti-fibrosing agents, used alone or in combination, should be administered under the following dosing guidelines. The total amount (dose) of anti-scarring agent in or on the device may be in the range of about 0.01 μg-10 μg, or 10 μg-10 mg, or 10 mg-250 mg, or 250 mg-1000 mg, or 1000 mg-2500 mg. The dose (amount) of anti-scarring agent per unit area of device surface to which the agent is applied may be in the range of about 0.01 μg/mm2 - 1 μg/mm2, or 1 μg/mm2 - 10 μg/mm2, or 10 μg/mm2 - 250 μg/mm2, 250 μg/mm2 - 1000 μg/mm2, or 1000 μg/mm2 - 2500 μg/mm2.
Provided below are exemplary dosage ranges for various anti- scarring agents that can be used in conjunction with devices for treating hypertrophic scars and keloids in accordance with the invention. (A) Angiogenesis inhibitors including Alphastatin, ZD-6474, IDN-5390, SB- 2723005, ABT-518, combretastatin, and anecortane, analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10^- 10"4 M of agent is to be maintained on the implant or barrier surface.
(B) mTOR inhibitors including AP-23573 and Temsirolimus, analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10^- 10"4 M of agent is to be maintained on the implant or barrier surface.
(C) Tubulin antagonists including synthadotin, analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10"8- 10"4 M of agent is to be maintained on the implant or barrier surface. (D) Epithilones including ixabepilone and analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10"8- 10"4 M of agent is to be maintained on the implant or barrier surface. (E) Kinesin antagonists including SB-715992 and analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10^- 10"4 M of agent is to be maintained on the implant or barrier surface. (F) TNF alpha antagonists including Etanercept, humicade, adalimumab and analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10^- 10"4 M of agent is to be maintained on the implant or barrier surface. (G) AKT inhibitor including erucylphosphocholine and analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10^- 10"4 M of agent is to be maintained on the implant or barrier surface. (H) FGF Inhibitors including IDN-5390 and analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10"8 - 10"4 M of agent is to be maintained on the implant or barrier surface. (I) Collagenase Antagonists including S-0885 and analogues and derivatives thereof: total dose not to exceed 1000 mg (range of 0.1 μg to 1000 mg); preferred 1 μg to 500 mg. Dose per unit area of 0.01 μg - 500 μg per mm2; preferred dose of 0.1 μg/mm2 - 100 μg/mm2. Minimum concentration of 10"8 - 10"4 M of agent is to be maintained on the implant or barrier surface. (J) NF KAPPA B Inhibitors including Bortezomib and analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10'8- 10"4 M of agent is to be maintained on the implant or barrier surface. (K) Elongation factor-1 alpha inhibitors including aplidine and analogues and derivatives thereof: total dose not to exceed 1000 mg (range of 0.1 μg to 1000 mg); preferred 1 μg to 500 mg. Dose per unit area of 0.01 μg - 500 μg per mm2; preferred dose of 0.1 μg/mm2 - 100 μg/mm2. Minimum concentration of 10'8 - 104 M of agent is to be maintained on the implant or barrier surface. (L) Tyrosine kinase inhibitors including Gefitinib and analogues and derivatives thereof: total dose not to exceed 1000 mg (range of 0.1 μg to 1000 mg); preferred 1 μg to 500 mg. Dose per unit area of 0.01 μg - 500 μg per mm2; preferred dose of 0.1 μg/mm2 - 100 μg/mm2. Minimum concentration of 10"8- 10"4 M of agent is to be maintained on the implant or barrier surface.
For high potency drugs, the total dose typically should not exceed 200 mg (range of 0.1 μg to 200 mg) and preferably 1 μg to 100 mg; dose per unit area of 0.01 μg - 100 μg per mm2; preferably 0.1 μg/mm2 - 20 μg/mm2; and minimum concentration of 10^- 10"4 M of agent should be maintained on the implant or barrier surface. For mid-potency drugs, the total dose typically should not to exceed 500 mg (range of 1.0 μg to 500 mg) and preferably 1 μg to 200 mg; dose per unit area of 0.01 μg - 200 μg per mm2, preferably 0.1 μg/mm2 - 40 μg/mm2; and minimum concentration of 10" 8 - 10"4 M of agent should be maintained on the implant or barrier surface. For low potency drugs, the total dose typically should not exceed 1000 mg (range of 0.1 μg to 1000 mg), preferably 1 μg to 500 mg; dose per unit area of 0.01 μg - 500 μg per mm2; preferably 0.1 μg/mm2 - 100 μg/mm2; and minimum concentration of 10"8 - 10"4 M of agent should to be maintained on the implant or barrier surface.
Vascular Grafts
In one aspect, the present invention provides for the combination of an anti-scarring agent and a vascular graft. Vascular graft devices that include a fibrosis-inhibiting agent are capable of inhibiting or reducing the overgrowth of granulation tissue, which can improve the clinical efficacy of these devices.
The vascular graft may be an extravascular graft or an intravascular (i.e., endoluminal) graft. The vascular graft may be, without limitation, in the form of a peripheral bypass application or a coronary bypass application. Vascular grafts may be used to replace or substitute damaged or diseased veins and arteries, including, without limitation, blood vessels damaged by aneurysms, intimal hyperplasia and thrombosis. Vascular grafts may also be used to provide access to blood vessels, for - - example, for hemodialysis access. Vascular grafts are implanted, for example, to provide an alternative conduit for blood flow through damaged or diseased areas in veins and arteries, including, without limitation, blood vessels damaged by aneurysms, intimal hyperplasia and thrombosis, however, the graft may lead to further complications, including, without limitation, infections, inflammation, thrombosis and intimal hyperplasia. The lack of long-term patency with vascular grafts may be due, for example, to surgical injury and abnormal hemodynamics and material mismatch at the suture line. Typically, further disease (e.g., restenosis) of the vessel occurs along the bed of the artery.
Some forms of improvements to vascular grafts have been made in an attempt to reduce the restenosis that occurs at the anastomosis site. Improvements include: (a) using a Miller cuff, which is a small piece of natural vein to make a short cuff that is joined by stitching it to the artery opening and the prosthetic graft; (b) using a flanged graft whereby the graft has a terminal skirt or cuff that facilitates an end-to-side anastomosis; (c) using a graft with an enlarged chamber having a large diameter for suture at the anastomosis site; and (d) using a graft that dispensing an agent that prevents thrombosis and/or intimal hyperplasia.
Representative examples of vascular grafts include, without limitation, synthetic bypass grafts (e.g., femoral-popliteal, femoral-femoral, axillary-femoral, and the like), vein grafts (e.g., peripheral and coronary), and internal mammary (e.g., coronary) grafts, bifurcated vascular grafts, intraluminal grafts, endovascular grafts and prosthetic grafts. Synthetic grafts can be made from a variety of polymeric materials, such as, for example, polytetrafluoroethylene (e.g., ePTFE), polyesters such as DACRON, polyurethanes, and combinations of polymeric materials.
Endoluminal vascular grafts may be used to treat aneurysms. For example, the vascular graft may be composed of a tubular graft with two tubular self-expanding stents that may be implanted for the treatment of aneurysms by means of minimally invasive procedures. See, e.g., U.S. Patent No. 6,168,620. The vascular graft may be composed of a flexible tubular body and a compressible frame positioned against the tubular body for support which has pores on the surface to promote ingrowth. See, e.g., U.S. Patent No. 5,693,088. The vascular graft may be bifurcated endovascular graft having a tubular trunk and two tubular limbs. See, e.g., U.S. Patent No. 6,454,796. The vascular graft may be a kink-resistant endoluminal bifurcated graft having two separate lumens contacted by a single lumen section. See, e.g., U.S. Patent No. 6,551 ,350. The vascular graft may be an intraluminal tube composed of ePTFE that has a seamline formed by overlapping the edges such that the microstructure fibrils are oriented in perpendicular directions. See, e.g., U.S. Patent No. 5,718,973.
In another aspect, the vascular graft may be used as a conduit to bypass vascular stenosis or other vascular abnormalities. For example, the vascular graft may be composed of a porous material having a layer of porous hollow fibers positioned along the inner surface which allows for tissue growth while inhibiting bleeding during the healing process. See, e.g., U.S. Patent No. 5,024,671. The vascular graft may be a flexible, monolithic, reinforced polymer tube having a microporous ePTFE tubular member and external ePTFE rib members projecting outwardly from the outer wall. See, e.g., U.S. Patent No. 5,609,624. The vascular graft may be composed of a tubular wall having longitudinally extending pleats that respond flexurally to changes in blood pressure while maintaining high compliance with reduced kinking. See, e.g., U.S. Patent No. 5,653,745. The vascular graft may be a radially supported ePTFE tube that is reinforced with greater density ring-shaped regions. See, e.g., U.S. Patent No. 5,747,128. The vascular graft may be porous PTFE tubing composed of a microstructure of nodes interconnected by fibrils which has a coating of elastomer on the outer wall. See, e.g., U.S. Patent Nos. 5,152,782 and 4,955,899. The vascular graft may be a plurality of polymeric fibers knitted together composed of at least three different fibers in which two fibers are absorbable and one is non-absorbable. See, e.g., U.S. Patent Nos. 4,997,440; 4,871 ,365 and 4,652,264.
In another aspect, the vascular graft may be modified to reduce thrombus formation or intimal hyperplasia at the anastomotic site. For example, the vascular graft may have an enlarged chamber having a first diameter parallel to the axis of the tubular wall and a second diameter transverse to the axis of the tube. See, e.g., U.S. Patent No. 6,589,278. The vascular graft may have a flanged skirt or cuff section with facilitates an end-to-side anastomosis directly between the artery and the end of the flanged bypass graft. See, e.g., U.S. Patent No. 6,273,912. The vascular graft may be composed of a tubular wall having a non-thrombogenic agent within the luminal layer and a thrombogenic layer forming the exterior of the vascular graft. See, e.g., U.S. Patent No. 6,440,166. The vascular graft may be composed of a smooth luminal surface made of ePTFE with a small pore size to reduce adherence of occlusive blood components. See, e.g., U.S. Patent No. 6,517,571. The vascular graft may be composed of hollow tubing that contains drug that is helically wrapped around the outer wall of a porous ePTFE graft whereby drug is dispensed by infusion through the porous interstices of the graft wall. See, e.g., U.S. Patent No. 6,355,063.
In another aspect, the vascular graft may be a harvested blood vessel that is used for bypass grafting. For example, vascular grafts may be composed of harvested arterial vessels from a host, such as the internal mammary arteries or inferior epigastric arteries. See, e.g., U.S. Patent No. 5,797,946. Vascular grafts may also be composed of saphenous veins which may be harvested from the host and used for coronary bypass or peripheral bypass procedures. See, e.g., U.S. Patent No. 6,558,313.
Other examples of vascular grafts are described in U.S. Patent Nos. 3,096,560, 3,805,301 , 3,945,052, 4,140,126, 4,323,525, 4,355,426, 4,475,972, 4,530,113, 4,550,447, 4,562,596, 4,601 ,718, 4,647,416, 4,878,908, 5,024,671, 5,104,399, 5,116,360, 5,151 ,105, 5,197,977, 5,282,824, 5,405,379, 5,609,624, 5,693,088, and 5,910,168.
Vascular grafts, which may be combined with one or more agents according to the present invention, include commercially available products. GORE-TEX Vascular Grafts and GORE-TEX INTERING Vascular Grafts are sold by Gore Medical Division (W. L. Gore & Associates, Inc. Newark, DE). CR. Bard, Inc. (Murray Hill, NJ) sells the DISTAFLO Bypass Grafts and IMPRA CARBOFLO Vascular Grafts.
In one aspect, the anti-scarring agent or a composition containing the anti-scarring agent is combined with a vascular graft.
Numerous polymeric and non-polymeric delivery systems for use in vascular grafts have been described above. Methods for incorporating fibrosis-inhibiting agents or fibrosis-inhibiting compositions onto or into the graft include: (a) affixing (directly or indirectly) to the graft a fibrosis-inhibiting composition (e.g., by either a spraying process or dipping process as described above, with or without a carrier), (b) incorporating or impregnating into the graft a fibrosis-inhibiting composition (e.g., by either a spraying process or dipping process as described above, with or without a carrier), (c) by coating the graft with a substance such as a hydrogel which will in turn absorb the fibrosis-inhibiting composition, (d) constructing the graft itself or a portion of the graft with a fibrosis-inhibiting composition, or (e) by covalently binding the fibrosis-inhibiting agent directly to the graft surface or to a linker (small molecule or polymer) that is coated or attached to the graft surface. For these grafts, the coating process can be performed in such a manner as to (a) coat the external surface of the graft, (b) coat the interior (luminal) surface of the graft, or (c) coat all or parts of both the external and internal surfaces of the graft, or (d) coat at least one end of the graft.
The fibrosis-inhibiting agent can be incorporated directly into the coating composition or into a secondary carrier (e.g., micelles, liposomes, emulsions, microspheres, nanospheres etc, as described above). Microsphere and nanospheres may include degradable polymers. Degradable polymers that can be used include poly(hydroxyl esters) {e.g., PLGA, PLA, PCL, and the like) as well as polyanhydrides, polyorthoesters and polysaccharides (e.g., chitosan and alginates).
In yet another embodiment, a gel, paste, thermogel or in situ forming gel that includes a fibrosis-inhibiting agent can be applied in a perivascular manner to the anastomosis produced during implantation of the graft device. Numerous polymeric and non-polymeric delivery systems for use in paste and gel formulations have been described above. The fibrosis- inhibiting agent can be incorporated directly into the gel or paste composition, or the therapeutic agent can be incorporated into a secondary carrier (e.g., micelles, liposomes, emulsions, microspheres, nanospheres etc, as described above).
In another aspect, the fibrosis-inhibiting agent can be incorporated into or onto an implant (e.g., a film or mesh material), which can be used in conjunction with a vascular graft to inhibit scarring at an anastomotic site. For example, a film or mesh material may be placed or wrapped in a perivascular (periadventitial) manner around the outside of the anastomosis at the time of surgery. Film and mesh implants may be used with a various types of vascular grafts, including synthetic bypass grafts (femoral-popliteal, femoral-femoral, axillary-femoral etc.), vein grafts (peripheral and coronary), internal mammary (coronary) grafts or hemodialysis grafts (AV fistulas, AV access grafts). Representative examples of films and meshes are described in further detail below.
In addition to the fibrosis-inhibiting agent, the vascular graft devices compositions for use with vascular graft devices can also further contain an anti-inflammatory agent (e.g., dexamethazone or aspirin) and/or an anti-thrombotic agent (e.g., heparin, heparin complexes, hydrophobic heparin derivatives, dipyridamole, or aspirin). The combination of agents may be coated onto the entire or portions of the vascular graft such that the thrombogenicity and/or fibrosis is reduced or inhibited. In certain embodiments, these agents may be coated onto the vascular graft using biodegradable polymers. For example, polymeric material that forms a gel in the pores and/or on the surface of the graft may be used, such as alginates, chitosan and chitosan sulfate, hyaluronic acid, dextran sulfate, PLURONIC polymers, chain extended PLURONIC polymers, polyester- polyether block copolymers of the various configurations (e.g., MePEG-PLA, PLA-PEG-PLA, and the like).
In one aspect, synthetic vascular grafts are provided that comprise, in addition to the anti-fibrosing agent, a composition in the form of a biodegradable gel. The gel composition can have anti-thrombogenic properties or include an agent having anti-thrombogenic properties, which may or may not be released from the gel composition. Gel coated grafts may reduce or prevent early thrombotic events commonly associated with implantation of synthetic grafts.
Polymeric biodegradable gels may comprise, for example, a chain extended PLURONIC polymer. Chain extended polymers may include a PLURONIC polymer (e.g., F127, F87, or the like) that has been reacted with a difunctional molecule such as succinyl chloride to increase the molecular weight of the polymer and thereby increase the viscosity of the PLURONIC polymer. Chain extended polymers can be dissolved in a solvent and then coated onto the synthetic vascular graft.
Gel compositions may be formed from a combination of small and/or polymeric molecules having two or more electrophilic groups and two or more nucleophilic groups. For example, the formulations may include a combination of a multi-armed PEG molecule in which the terminal hydroxyl groups are activated with succinimidyl moieties and a multi-armed PEG molecule having terminal amino and/or sulfhydryl groups. The multi-armed PEG reagents may be dissolved separately in an appropriate solvent (e.g., aqueous buffer, IPA, dichloromethane, or a combination of solvents) and then sprayed sequentially or simultaneously onto the desired surface of the graft, such that the two components react to produce a crosslinked gel. The solvent may then be removed by air or vacuum drying.
In another embodiment, the composition may be formed from a polymer having two or more succinimidyl groups and a small molecule - having two or more amino or sulfhydryl groups (e.g., dilysine). Alternatively, the polymer components can include two or more sulfhydryl groups or amino groups, and the small molecule contains two or more succinimidyl groups.
In yet another embodiment, gel coatings may be produced from polyester-polyether block copolymers of various configurations (e.g., X- Y, X-Y-X or Y-X-Y, R-(Y-X)n, R-(X-Y)n where X is a polyalkylene oxide and Y is a polyester (e.g., polyester can comprise the residues of one or more of the monomers selected from lactide, lactic acid, glycolide, glycolic acid, e- caprolactone, gamma-caprolactone, hydroxyvaleric acid, hydroxybutyric acid, beta-butyrolactone, gamma-butyrolactone, gamma-valerolactone, γ~ decanolactone, δ-decanolactone, trimethylene carbonate, 1,4-dioxane-2- one or 1,5-dioxepan-2one.), R is a multifunctional initiator and copolymers as well as blends and copolymers thereof.) may be used to form the gel coating. In one embodiment, the synthetic vascular graft is formed of a porous synthetic material such as expanded PTFE (ePTFE). A coating comprising a gel composition, such as described above, may be applied onto the entire graft or a portion of the graft surface (e.g., the interior surface of the graft or the ends of the graft). Further, the pores of the graft may be either partially or fully filled with the coating composition. The extent to which the coating occupies the pores of the device can be altered by changing the solvent used to dissolve the polymer. For example, a surface coating may be achieved by using a hydrophilic solvent such as water which will not wet the hydrophobic surface of an ePTFE graft. Coating from a solvent such as dichloromethane, which wets an ePTFE surface, can be used to coat the polymer composition onto the inner pore structure of the graft.
The gel formulations may have anti-thrombogenic properties due to the hydrophilicity. Hydrophilic coatings may be physically removed from the surface of the graft over time which. may reduce the adhesion of platelets to the graft surface. Additionally, an anti-thrombogenic agent (e.g., heparin, fragments of heparin, organic soluble salts of heparin, sulfonated carbohydrates, warfarin, Coumadin, coumarin, heparinoid, danaparoid, argatroban chitosan sulfate, or chondroitin sulfate) may be included into the formulation. In one embodiment, the anti-thrombotic agent(s) may be incorporated into microspheres. Other additives which may be added into gel compositions for use with vascular grafts include buffers, osmolality modifiers, viscosity modifiers, and hydrating agents (e.g., PEG, MePEG, and various sugars).
According to the present invention, any scarring agent described above can be utilized in the practice of this embodiment. Within one embodiment of the invention, vascular grafts may be adapted to release an agent that inhibits one or more of the four general components of the process of fibrosis (or scarring), including: formation of new blood vessels (angiogenesis), migration and proliferation of connective tissue cells (such as fibroblasts or smooth muscle cells), deposition of extracellular matrix (ECM), and remodeling (maturation and organization of the fibrous tissue). By inhibiting one or more of the components of fibrosis (or scarring), the overgrowth of granulation tissue may be inhibited or reduced.
As vascular grafts are made in a variety of configurations and sizes, the exact dose administered will vary with device size, surface area and design. However, certain principles can be applied in the application of this art. Drug dose can be calculated as a function of dose per unit area (of the portion of the device being coated), total dose administered, and appropriate surface concentrations of active drug can be determined. Drugs are to be used at concentrations that range from several times more than to 10%, 5%, or even less than 1% of the concentration typically used in a single chemotherapeutic systemic dose application. Preferably, the drug is released in effective concentrations for a period ranging from 1 - 90 days.
Several examples of fibrosis-inhibiting agents for use with vascular grafts include the following: ZD-6474, AP-23573, synthadotin, S- 0885, aplidine, ixabepilone, IDN-5390, SB-2723005, ABT-518, combretastatin, anecortave acetate, SB-715992, temsirolimus, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, as well as analogues and derivatives of the aforementioned.
Additional examples of anti-scarring agents which can be used include those having a high potency in the assays described herein (1- 10OnM IC50 range) such as isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, dolastatin 15, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17- DMAG, and tacrolimus; those having a mid-potency in the assays described herein (100-500 nM IC50 range) such as loteprednol etabonate, juglone, prednisolone, puromycin, 3-BAABE, cladribine, and mannose-6-phosphate; and those having a low potency in the assays described herein (500- 1000nm range IC50 range) such as 5-azacytidine, Ly333531(ruboxistaurin), and simvastatin. Regardless of the method of application of the drug to the vascular graft, the exemplary anti-fibrosing agents, used alone or in combination, should be administered under the following dosing guidelines. The total amount (dose) of anti-scarring agent in or on the device may be in the range of about 0.01 μg-10 μg, or 10 μg-10 mg, or 10 mg-250 mg, or 250 mg-1000 mg, or 1000 mg-2500 mg. The dose (amount) of anti-scarring agent per unit area of device surface to which the agent is applied may be in the range of about 0.01 μg/mm2 - 1 μg/mm2, or 1 μg/mm2 - 10 μg/mm2, or 10 μg/mm2 - 250 μg/mm2, 250 μg/mm2 - 1000 μg/mm2, or 1000 μg/mm2 - 2500 μg/mm2.
Provided below are exemplary dosage ranges for various anti- scarring agents that can be used in conjunction with vascular graft devices in accordance with the invention. (A) Angiogenesis inhibitors including Alphastatin, ZD-6474, IDN-5390, SB-2723005, ABT-518, combretastatin, and anecortane, analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10^- 10"4 M of agent is to be maintained on the implant or barrier surface. (B) mTOR inhibitors including AP-23573 and Temsirolimus, analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10^- 10"4 M of agent is to be maintained on the implant or barrier surface. (C) Tubulin antagonists including synthadotin, analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10^- 10"4 M of agent is to be maintained on the implant or barrier surface. (D) Epithilones including ixabepilone and analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10'8 - 10'4 M of agent is to be maintained on the implant or barrier surface. (E) Kinesin antagonists including SB- 715992 and analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10^ - 10"4 M of agent is to be maintained on the implant or barrier surface. (F) TNF alpha antagonists including Etanercept, humicade, adalimumab and analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10^- 10"4 M of agent is to be maintained on the implant or barrier surface. (G) AKT inhibitor including erucylphosphocholine and analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of O.Olμg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10^- 10"4 M of agent is to be maintained on the implant or barrier surface. (H) FGF Inhibitors including IDN-5390 and analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 108 - 10"4 M of agent is to be maintained on the implant or barrier surface. (I) Collagenase Antagonists including S-0885 and analogues and derivatives thereof: total dose not to exceed 1000 mg (range of 0.1 μg to 1000 mg); preferred 1 μg to 500 mg. Dose per unit area of 0.01 μg - 500 μg per mm2; preferred dose of 0.1 μg/mm2 - 100 μg/mm2. Minimum concentration of 10^- 10"4 M of agent is to be maintained on the implant or barrier surface. (J) NF KAPPA B Inhibitors including Bortezomib and analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10"8- 10"4 M of agent is to be maintained on the implant or barrier surface. (K) Elongation factor-1 alpha inhibitors including aplidine and analogues and derivatives thereof: total dose not to exceed 1000 mg (range of 0.1 μg to 1000 mg); preferred 1 μg to 500 mg. Dose per unit area of 0.01 μg - 500 μg per mm2; preferred dose of 0.1 μg/mm2 - 100 μg/mm2. Minimum concentration of 10"8- 10"4 M of agent is to be maintained on the implant or barrier surface. (L) Tyrosine kinase inhibitors including Gefitinib and analogues and derivatives thereof: total dose not to exceed 1000 mg (range of 0.1 μg to 1000 mg); preferred 1 μg to 500 mg. Dose per unit area of 0.01 μg - 500 μg per mm2; preferred dose of 0.1 μg/mm2 - 100 μg/mm2. Minimum concentration of 10"8- 10"4 M of agent is to be maintained on the implant or barrier surface.
For high potency drugs, the total dose typically should not exceed 200 mg (range of 0.1 μg to 200 mg) and preferably 1 μg to 100 mg; dose per unit area of 0.01 μg - 100 μg per mm2; preferably 0.1 μg/mm2 - 20 μg/mm2; and minimum concentration of 10"8- 10"4 M of agent should be maintained on the implant or barrier surface. For mid-potency drugs, the total dose typically should not to exceed 500 mg (range of 1.0 μg to 500 mg) and preferably 1 μg to 200 mg; dose per unit area of 0.01 μg - 200 μg per mm2, preferably 0.1 μg/mm2 - 40 μg/mm2; and minimum concentration of 10" 8- 10"4 M of agent should be maintained on the implant or barrier surface. For low potency drugs, the total dose typically should not exceed 1000 mg (range of 0.1 μg to 1000 mg), preferably 1 μg to 500 mg; dose per unit area of 0.01 μg - 500 μg per mm2; preferably 0.1 μg/mm2 - 100 μg/mm2; and minimum concentration of 10^- 10"4 M of agent should to be maintained on the implant or barrier surface.
Hemodialysis Access Devices
In one aspect, the present invention provides for the combination of an anti-scarring agent and a hemodialysis access device. Hemodialysis dialysis access devices that include a fibrosis-inhibiting agent are capable of inhibiting or reducing the overgrowth of granulation tissue, which can improve the clinical efficacy of these devices.
Hemodialysis access devices may be used when blood needs to be removed, cleansed and then returned to the body. Hemodialysis regulates the body's fluid and chemical balances as well as removes waste from the blood stream that cannot be cleansed by a normally functioning kidney due to disease or injury. For hemodialysis to occur, the blood may be obtained through a hemodialysis access or vascular access, in which minor surgery is performed to provide access through an AV fistula or AV access graft. These hemodialysis access devices may develop complications, including infections, inflammation, thrombosis and intimal hyperplasia of the associated blood vessels. The lack of long-term patency with hemodialysis access may be due to surgical injury, abnormal hemodynamics and material mismatch at the suture line. Typically, further disease (e.g., restenosis) of the vessel occurs along the bed of the artery and/or at the site of anastomosis.
In addition to the AV fistulas and AV access grafts described above, implantable subcutaneous hemodialysis access systems such as the commercially available catheters, ports, and shunts, may also be used for hemodialysis patients. These access systems may consist of a small metallic or polymeric device or devices implanted underneath the skin. These devices may be connected to flexible tubes, which are inserted into a vessel to allow for blood access.
Representative examples of hemodialysis access devices include, without limitation, AV access grafts, venous catheters, vascular grafts, implantable ports, and AV shunts. Synthetic hemodialysis access devices can be made from metals or polymers, such as polytetrafluoroethylene (e.g., ePTFE), polyesters such as DACRON, polyurethanes, or combinations of these materials.
In one aspect, the hemodialysis access device may be an AV access graft. For example, the AV access graft may be composed of an implantable self-expanding flexible percutaneous stent-graft of open weave construction with ends being compressible and having an elastic layer arranged along a portion of its length. See, e.g., U.S. Patent Nos. 5,755,775 and 5,591 ,226. The AV access graft may be composed of a tubular section with a generally constant diameter which tapers towards the venous end. See, e.g., U.S. Patent No. 6,585,762. The AV access graft may be composed of a two microporous ePTFE tubes that are circumferentially disposed over each other with a polymeric layer interposed between such that the graft is self-sealing and exhibits superior radial tensile strength and suture hole elongation resistance. See, e.g., U.S. Patent No. 6,428,571. The AV access graft may be composed of a coaxial double lumen tube with an inner and outer tube having a self-sealing, nonbiodegradable, polymeric adhesive between the tubes. See, e.g., U.S. Patent No. 4,619,641. The AV access graft may be composed of a synthetic fabric having a high external velour profile which is woven or knitted to form a tubular prosthesis which has elastic fibers that allows self- sealing following a punctured state. See, e.g., U.S. Patent No. 6,547,820. The AV access graft may be of tubular form having a base tube with the ablumenal surface covered with a deflectable material, such as a porous film, which is arranged adjacently to allow movement. See, e.g., U.S. Patent No. 5,910,168.
In another aspect, the hemodialysis access device may be a catheter system. For example, the catheter system may be composed of a suction and return line that are adapted for disposition in the vascular system of the body and are connected to a subcutaneous connector port. See, e.g., U.S. Patent Nos. 6,620,118 and 5,989,206. The catheter system may be an apparatus that is used to arterialize a vein by creating an AV fistula by inserting a catheter into a vein and a catheter into an adjacent artery. See, e.g., U.S. Patent No. 6,464,665. The catheter system may be composed of a hollow sheath that provides percutaneous introduction of fistula-generating vascular catheters through a perforation in a vessel wall, such that the catheters generate an intervascular fistula on-demand between adjacent vessels. See, e.g., U.S. Patent Nos. 6,099,542 and 5,830,224.
In another aspect, the hemodialysis access device may be used for an AV fistula. For example, the hemodialysis access device may be an AV fistula assembly composed of a synthetic coiled stent graft with helically-extending turns with gaps used to enhance the function of an AV fistula. See, e.g., U.S. Patent No. 6,585,760.
In another aspect, the hemodialysis access device may be an implantable access port, shunt or valve. These devices may be implanted subcutaneously with communication to the blood supply and accessed using a percutaneous puncture. For example, the hemodialysis access device may be composed of housing having an entry port and an exit port to a passageway which has an elastomeric sealing valve that provides access into the exit port for a needle. See, e.g., U.S. Patent No. 5,741,228. The hemodialysis, access device may be a shunt composed of a slideable valve and flexible lid that has a fluid communication tube between the arterial and venous ends. See, e.g., U.S. Patent No. 5,879,320. The hemodialysis access device may be a shunt in the form of a junction that has a connector with two legs that are inserted into the native blood vessel and one leg that is adapted for sealing to another blood vessel without punctures. See, e.g., U.S. Patent No. 6,019,788. The hemodialysis access device may be a surface access double hemostatic valve that may be mounted on the wall of an AV graft for hemodialysis access. See, e.g., U.S. Patent Nos. 6,004,301 and 6,090,067.
Hemodialysis access devices, which may be combined with one or more agents according to the present invention, include commercially available products. For example, hemodialysis access devices include products, such as the LIFESITE (Vasca Inc., Tewksbury, MA) and the DIALOCK catheters from Biolink Corp. (Middleboro, MA), VECTRA Vascular Access Grafts and VENAFLO Vascular Grafts from CR. Bard, Inc. (Murray Hill, NJ), and GORE-TEX Vascular Grafts and Stretch Vascular Grafts from Gore Medical Division (W. L. Gore & Associates, Inc. Newark, DE).
In one aspect, the anti-scarring agent or a composition containing the anti-scarring agent is combined with a hemodialysis access device. Numerous polymeric and non-polymeric delivery systems for use in hemodialysis access devices have been described above. Methods for incorporating fibrosis-inhibiting agents or compositions comprising fibrosis- inhibiting agents onto or into the hemodialysis access device include: (a) directly affixing to the hemodialysis access device a fibrosis-inhibiting composition (e.g., by either a spraying process or dipping process as described above, with or without a carrier), (b) directly incorporating into the hemodialysis access device a fibrosis-inhibiting composition (e.g., by either a spraying process or dipping process as described above, with or without a carrier), (c) by coating the hemodialysis access device with a substance such as a hydrogel which will in turn absorb the fibrosis-inhibiting composition, (d) constructing the hemodialysis access device itself or a portion of the graft with a fibrosis-inhibiting composition, or (e) by covalently binding the fibrosis-inhibiting agent directly to the hemodialysis access device surface or to a linker (small molecule or polymer) that is coated or attached to the hemodialysis access device surface. For devices that are coated, the coating process can be performed in such a manner as to (a) coat only the external surface of the device; (b) coat the internal (luminal) surface of the device; or (c) coat all or parts of both the external and internal surfaces.
In another aspect, the fibrosis-inhibiting agent or a composition containing the fibrosis-inhibiting agent can be incorporated into an implant, such as a film or mesh, which can be used in conjunction with a hemodialysis access device to inhibit scarring at the site of an anastomosis or fistula. These films or meshes may be placed or wrapped in a perivascular (periadventitial) manner around the outside of the fistula or anastomosis at the time of surgery. Representative examples of implants (i.e., meshes and films) for use with hemodialysis access devices are described below.
In yet another aspect, a composition in the form of, for example, a gel, paste, thermogel, or in situ forming gel, which includes a fibrosis-inhibiting agent can be applied in a perivascular manner to the fistula or anastomosis produced during implantation of the hemodialysis access device.
The fibrosis-inhibiting agent can be incorporated directly into the gel or paste composition, or the therapeutic agent can be incorporated into a secondary carrier (e.g., micelles, liposomes, emulsions, microspheres, nanospheres etc, as described above) that is then incorporated into the composition that is to be delivered. Microsphere and nanospheres may include degradable polymers. Degradable polymers that can be used include poly (hydroxyl esters) (e.g., PLGA, PLA, PCL, and the like) as well as polyanhydrides, polyorthoesters and polysaccharides (e.g., chitosan and alginates).
In addition to the fibrosis-inhibiting agent, hemodialysis access devices and compositions for use with hemodialysis access devices can also further contain an anti-inflammatory agent (e.g., dexamethazone or aspirin) and/or an anti-thrombotic agent (e.g., heparin, heparin complexes, hydrophobic heparin derivatives, dipyridamole, or aspirin).
According to the present invention, any scarring agent described above can be utilized in the practice of this embodiment. Within one embodiment of the invention, hemodialysis access devices may be adapted to release an agent that inhibits one or more of the four general components of the process of fibrosis (or scarring), including: formation of new blood vessels (angiogenesis), migration and proliferation of connective tissue cells (such as fibroblasts or smooth muscle cells), deposition of extracellular matrix (ECM), and remodeling (maturation and organization of the fibrous tissue). By inhibiting one or more of the components of fibrosis (or scarring), the overgrowth of granulation tissue may be inhibited or reduced.
Several examples of fibrosis-inhibiting agents for use with hemodialysis access devices include the following: ZD-6474, AP-23573, synthadotin, S-0885, aplidine, ixabepilone, IDN-5390, SB-2723005, ABT- 518, combretastatin, anecortave acetate, SB-715992, temsirolimus, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, as well as analogues and derivatives of the aforementioned.
Additional examples of anti-scarring agents which can be used include those having a high potency in the assays described herein (1- 10OnM IC50 range) such as isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, dolastatin 15, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17- DMAG, and tacrolimus; those having a mid-potency in the assays described herein (100-500 nM IC50 range) such as loteprednol etabonate, juglone, prednisolone, puromycin, 3-BAABE, cladribine, and mannose-6-phosphate; and those having a low potency in the assays described herein (500- 1000nm range IC50 range) such as 5-azacytidine, Ly333531(ruboxistaurin), and simvastatin.
As hemodialysis access devices are made in a variety of configurations and sizes, the exact dose administered will vary with device size, surface area and design. However, certain principles can be applied in the application of this art. Drug dose can be calculated as a function of dose per unit area (of the portion of the device being coated), and total amount of drug on the device can be measured and appropriate surface concentrations of active drug can be determined. Drugs are to be used at concentrations that range from several times more than to 10%, 5%, or even less than 1 % of the concentration typically used in a single chemotherapeutic systemic dose application. Preferably, the drug is released in effective concentrations for a period ranging from 1 - 90 days. Regardless of the method of application of the drug to the device, the exemplary anti-fibrosing agents, used alone or in combination, should be administered under the following dosing guidelines. The total amount (dose) of anti-scarring agent in or on the device may be in the range of about 0.01 μg-10 μg, or 10 μg-10 mg, or 10 mg-250 mg, or 250 mg-1000 mg, or 1000 mg-2500 mg. The dose (amount) of anti-scarring agent per unit area of device surface to which the agent is applied may be in the range of about 0.01 μg/mm2 - 1 μg/mm2, or 1 μg/mm2 - 10 μg/mm2, or 10 μg/mm2 - 250 μg/mm2, 250 μg/mm2 - 1000 μg/mm2, or 1000 μg/mm2 - 2500 μg/mm2.
Provided below are exemplary dosage ranges for various anti- scarring agents that can be used in conjunction with hemodialysis access devices in accordance with the invention. (A) Angiogenesis inhibitors including Alphastatin, 2D-6474, IDN-5390, SB-2723005, ABT-518, combretastatin, and anecortane, analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit jarea of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10^- 10"4 M of agent is to be maintained on the implant or barrier surface. (B) mTOR inhibitors including AP-23573 and Temsirolimus, analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10^- 10"4 M of agent is to be maintained on the implant or barrier surface. (C) Tubulin antagonists including synthadotin, analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10"8- 10"4 M of agent is to be maintained on the implant or barrier surface. (D) Epithilones including ixabepilone and analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10'8- 10"4 M of agent is to be maintained on the implant or barrier surface. (E) Kinesin antagonists including SB- 715992 and analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10^- 10"4 M of agent is to be maintained on the implant or barrier surface. (F) TNF alpha antagonists including Etanercept, humicade, adalimumab and analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10"8 - 10"4 M of agent is to be maintained on the implant or barrier surface. (G) AKT inhibitor including erucylphosphocholine and analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10^- 10"4 M of agent is to be maintained on the implant or barrier surface. (H) FGF Inhibitors including IDN-5390 and analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10"8- 10'4 M of agent is to be maintained on the implant or barrier surface. (I) Collagenase Antagonists including S-0885 and analogues and derivatives thereof: total dose not to exceed 1000 mg (range of 0.1 μg to 1000 mg); preferred 1 μg to 500 mg. Dose per unit area of 0.01 μg - 500 μg per mm2; preferred dose of 0.1 μg/mm2 - 100 μg/mm2. Minimum concentration of 10"8 - 10"4 M of agent is to be maintained on the implant or barrier surface. (J) NF KAPPA B Inhibitors including Bortezomib and analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10^- 10"4 M of agent is to be maintained on the implant or barrier surface. (K) Elongation factor-1 alpha inhibitors including aplidine and analogues and derivatives thereof: total dose not to exceed 1000 mg (range of 0.1 μg to 1000 mg); preferred 1 μg to 500 mg. Dose per unit area of 0.01 μg - 500 μg per mm2; preferred dose of 0.1 μg/mm2 - 100 μg/mm2. Minimum concentration of 10^- 10"4 M of agent is to be maintained on the implant or barrier surface. (L) Tyrosine kinase inhibitors including Gefitinib and analogues and derivatives thereof: total dose not to exceed 1000 mg (range of 0.1 μg to 1000 mg); preferred 1 μg to 500 mg. Dose per unit area of 0.01 μg - 500 μg per mm2; preferred dose of 0.1 μg/mm2 - 100 μg/mm2. Minimum concentration of 10"8- 10"4 M of agent is to be maintained on the implant or barrier surface.
For high potency drugs, the total dose typically should not exceed 200 mg (range of 0.1 μg to 200 mg) and preferably 1 μg to 100 mg; dose per unit area of 0.01 μg - 100 μg per mm2; preferably 0.1 μg/mm2 - 20 μg/mm2; and minimum concentration of 10"8- 10"4 M of agent should be maintained on the implant or barrier surface. For. mid-potency drugs, the total dose typically should not to exceed 500 mg (range of 1.0 μg to 500 mg) and preferably 1 μg to 200 mg; dose per unit area of 0.01 μg - 200 μg per mm2, preferably 0.1 μg/mm2 - 40 μg/mm2; and minimum concentration of 10" 8- 10"4 M of agent should be maintained on the implant or barrier surface. For low potency drugs, the total dose typically should not exceed 1000 mg (range of 0.1 μg to 1000 mg), preferably 1 μg to 500 mg; dose per unit area of 0.01 μg - 500 μg per mm2; preferably 0.1 μg/mm2 - 100 μg/mm2; and minimum concentration of 10^- 10"4 M of agent should to be maintained on the implant or barrier surface.
Films and Meshes
In one aspect, the present invention provides for the combination of an anti-scarring agent and a film or mesh. Incorporation of a fibrosis-inhibiting agent into or onto a film or mesh can minimize fibrosis (or scarring) in the vicinity of the implant and may reduce or prevent the formation of adhesions between the implant and the surrounding tissue. In certain aspects, the film or mesh may be used as a drug-delivery vehicle (e.g., as a perivascular delivery device for the prevention of neointimal hyperplasia at an anastomotic site).
Films or meshes may take a variety of forms including, but not limited to, surgical barriers, surgical adhesion barriers, membranes (e.g., barrier membranes), surgical sheets, surgical patches (e.g., dural patches), surgical wraps (e.g., vascular, perivascular, adventitial, periadventitital wraps, and adventitial sheets), meshes (e.g., perivascular meshes), bandages, liquid bandages, surgical dressings, gauze, fabrics, tapes, surgical membranes, polymer matrices, shells, envelopes, tissue coverings, and other types of surgical matrices, scaffolds, and coatings.
In one aspect, the device comprises or may be in the form of a film. The film may be formed into one of many geometric shapes. Depending on the application, the film may be formed into the shape of a tube or may be a thin, elastic sheet of polymer. Generally, films are less" than 5, 4, 3, 2, or 1 mm thick, more preferably less than 0.75 mm, 0.5 mm, 0.25 mm, or, 0.10 mm thick. Films can also be generated of thicknesses less than 50 μm, 25 μm or 10 μm. Films generally are flexible with a good tensile strength (e.g., greater than 50, preferably greater than 100, and more preferably greater than 150 or 200 N/cm2), good adhesive properties (i.e., adheres to moist or wet surfaces), and have controlled permeability. Polymeric films (which may be porous or non-porous) are particularly useful for application to the surface of a device or implant as well as to the surface of tissue, cavity or an organ.
Films may be made by several processes, including for example, by casting, and by spraying, or may be formed at the treatment site in situ. For example, a sprayable formulation may be applied onto the treatment site which then forms into a solid film.
In another aspect, the device may comprise or be in the form of a polymer, wherein at least some of the polymer is in the form of a mesh. A mesh, as used herein, is a material composed of a plurality of fibers or filaments (i.e., a fibrous material), where the fibers or filaments are arranged in such a manner (e.g., interwoven, knotted, braided, overlapping, looped, knitted, interlaced, intertwined, webbed, felted, and the like) so as to form a porous structure. Typically, a mesh is a pliable material, such that it has sufficient flexibility to be wrapped around the external surface of a body passageway or cavity, or a portion thereof. The mesh may be capable of providing support to the structure (e.g., the vessel or cavity wall) and may be adapted to release an amount of the therapeutic agent.
Mesh materials may take a variety of forms. For example, the mesh may be in a woven, knit, or non-woven form and may include fibers or filaments that are randomly oriented relative to each other or that are arranged in an ordered array or pattern. In one embodiment, for example, a mesh may be in the form of a fabric, such as, for example, a knitted, braided, crocheted, woven, non-woven (e.g., a melt-blown or wet-laid) or webbed fabric. In one embodiment, a mesh may include a natural or synthetic biodegradable polymer that may be formed into a knit mesh, a weave mesh, a sprayed mesh, a web mesh, a braided mesh, a looped mesh, and the like. Preferably, a mesh or wrap has intertwined threads that form a porous structure, which may be, for example, knitted, woven, or webbed.
The structure and properties of the mesh used in a device depend on the application and the desired mechanical (i.e., flexibility, tensile strength, and elasticity), degradation properties, and the desired loading and release characteristics for the selected therapeutic agent(s). The mesh should have mechanical properties, such that the device will remain sufficiently strong until the surrounding tissue has healed. Factors that affect the flexibility and mechanical strength of the mesh include, for example, the porosity, fabric thickness, fiber diameter, polymer composition (e.g., type of monomers and initiators), process conditions, and the additives that are used to prepare the material. Typically, the mesh possesses sufficient porosity to permit the flow of fluids through the pores of the fiber network and to facilitate tissue ingrowth. Generally, the interstices of the mesh should be sufficiently wide apart to allow light visible by eye, or fluids, to pass through the pores. However, materials having a more compact structure also may be used. The flow of fluid through the interstices of the mesh depends on a variety of factors, including, for example, the stitch count or thread density. The porosity of the mesh may be further tailored by, for example, filling the interstices of the mesh with another material (e.g., particles or polymer) or by processing the mesh (e.g., by heating) in order to reduce the pore size and to create non-fibrous areas. Fluid flow through the mesh of the invention will vary depending on the properties of the fluid, such as viscosity, hydrophilicity/hydrophobicity, ionic concentration, temperature, elasticity, pseudoplasticity, particulate content, and the like. Preferably, the interstices do not prevent the release of impregnated or coated therapeutic agent(s) from the mesh, and the interstices preferably do not prevent the exchange of tissue fluid at the application site.
Mesh materials should be sufficiently flexible so as to be capable of being wrapped around all or a portion of the external surface of a body passageway or cavity. Flexible mesh materials are typically in the form of flexible woven or knitted sheets having a thickness ranging from about 25 microns to about 3000 microns; preferably from about 50 to about 1000 microns. Mesh material suitable for wrapping around arteries and veins typically ranges from about 100 to 400 microns in thickness.
The diameter and length of the fibers or filaments may range in size depending on the form of the material (e.g., knit, woven, or non- woven), and the desired elasticity, porosity, surface area, flexibility, and tensile strength. The fibers may be of any length, ranging from short filaments to long threads (i.e., several microns to hundreds of meters in length). Depending on the application, the fibers may have a monofilament or a multifilament construction.
The mesh may include fibers that are of same dimension or of different dimensions, and the fibers may be formed from the same or different types of biodegradable polymers. Woven materials, for example, may include a regular or irregular array of warp and weft strands and may include one type of polymer in the weft direction and another type (having the same or a different degradation profile from the first polymer) in the warp direction. The degradation profile of the weft polymer may be different than or the same as the degradation profile of the warp polymer. Similarly, knit materials may include one or more types (e.g., monofilament, multi-filament) and sizes of fibers and may include fibers made from the same or from different types of biodegradable polymers.
The structure of the mesh (e.g., fiber density and porosity) may impact the amount of therapeutic agent that may be loaded into or onto the device. For example, a fabric having a loose weave characterized by a low fiber density and high porosity will have a lower thread count, resulting in a reduced total fiber volume and surface area. As a result, the amount of agent that may be loaded into or onto, with a fixed carrier: therapeutic agent ratio, the fibers will be lower than for a fabric having a high fiber density and lower porosity. It is preferable that the mesh also should not invoke biologically detrimental inflammatory or toxic response, should be capable of being fully metabolized in the body, have an acceptable shelf life, and be easily sterilized.
The device may include multiple mesh materials in any combination or arrangement. For example, a portion of the device may be a knitted material and another portion may be a woven material. In another embodiment, the device may more than one layer (e.g., a layer of woven material fused to a layer of knitted material or to another layer of the same type or a different type of woven material). In some embodiments, multilayer constructions (e.g., device having two or more layers of material) may be used, for example, to enhance the performance properties of the device (e.g., for enhancing the rigidity or for altering the porosity, elasticity, or tensile strength of the device) or for increasing the amount of drug loading.
Multi-layer constructions may be useful, for example, in devices containing more than one type of therapeutic agent. For example, a first layer of mesh material may be loaded with one type of agent and a second layer may be loaded with another type of agent. The two layers may be unconnected or connected (e.g., fused together, such as by heat welding or ultrasonic welding) and may be formed of the same type of fabric or from a different type of fabric having a different polymer composition and/or structure.
In certain aspects, a mesh may include portions that are not in the form of a mesh. For example, the device may include the form of a film, sheet, paste, and the like, and combinations thereof. For example, the device may have a multi-layer construction having a film layer that includes the therapeutic agent and one or more layers of mesh material. For example, the film layer may be interposed between two layers of mesh or may be disposed on just one side the mesh material. The film layer may include a first therapeutic agent, whereas one or more of the layers of mesh may include the same or a different agent. In another embodiment, the device includes at least two layers of mesh. In one aspect, at least two of the at least two layers of mesh are fused together.
In one aspect, multilayer devices are provided that may further include a film layer. The film layer may reside between two of the at least two layers of mesh. In yet another embodiment, a delivery device is described that includes a mesh, wherein the mesh includes a biodegradable polymer and a first therapeutic agent. The device may further include a film that includes a second therapeutic agent, which may have the same or a different composition than the first therapeutic agent. For example, in one embodiment, a device suitable for wrapping around a vein or artery includes a layer of mesh and a film layer loaded with a therapeutic agent. The device may be wrapped around a body passageway or cavity, such that the film layer contacts the external surface of the passageway or cavity. Thus, the device may deliver the appropriate dosage of agent and may provide sufficient mechanical strength to improve and maintain the structural integrity of the body passageway or cavity.
In one aspect, the mesh or film includes a polymer. The polymer may be a biodegradable polymer. Biodegradable compositions that may be used to prepare the mesh include polymers that comprise albumin, collagen, hyaluronic acid and derivatives, sodium alginate and derivatives, chitosan and derivatives gelatin, starch, cellulose polymers (for example methylcellulose, hydroxypropylcellulose, hydroxypropylmethylcellulose, carboxymethylcellulose, cellulose acetate phthalate, cellulose acetate succinate, hydroxypropylmethylcellulose phthalate), casein, dextran and derivatives, polysaccharides, poly(caprolactone), fibrinogen, poly(hydroxyl acids), poly(L-lactide) poly(D,L lactide), poly(D,L-lactide-co-glycolide), poly(L-lactide-co-glycolide), copolymers of lactic acid and glycolic acid, copolymers of ε-caprolactone and lactide, copolymers of glycolide and ε- caprolactone, copolymers of lactide and 1 ,4-dioxane-2-one, polymers and copolymers that include one or more of the residue units of the monomers D-lactide, L-lactide, D.L-lactide, glycolide, ε-caprolactone, trimethylene carbonate, 1,4-dioxane-2-one or 1 ,5-dioxepan-2-one, poly(glycolide), poly(hydroxybutyrate), poly(alkylcarbonate) and poly(orthoesters), polyesters, poly(hydroxyvaleric acid), polydioxanone, poly(ethylene terephthalate), poly(malic acid), poly(tartronic acid), polyanhydrides, polyphosphazenes, poly(amino acids). These compositions include copolymers of the above polymers as well as blends and combinations of the above polymers, (see generally, Ilium, L., Davids, S. S. (eds.) "Polymers in Controlled Drug Delivery" Wright, Bristol, 1987; Arshady, J. Controlled Release 77:1-22, 1991; Pitt, Int. J. Phar. 59:173-196, 1990; Holland et al., J. Controlled Release 4:155-0180, 1986).
In one aspect, the mesh or film includes a biodegradable or resorbable polymer that is formed from one or more monomers selected from the group consisting of lactide, glycolide, e-caprolactone, trimethylene carbonate, 1,4-dioxan-2-one, 1 ,5-dioxepan-2-one, 1 ,4-dioxepan-2-one, hydroxyvalerate, and hydroxybutyrate. In one aspect, the polymer may include, for example, a copolymer of a lactide and a glycolide. In another aspect, the polymer includes a poly(caprolactone). In yet another aspect, the polymer includes a poly(lactιc acid), poly(L-lactide)/poly(D,L-Lactide) blends or copolymers of L-lactide and D, L-lactide. In yet another aspect, the polymer includes a copolymer of lactide and e-caprolactone. In yet another aspect, the polymer includes a polyester (e.g., a poly(lactide-co-glycolide). The poly(lactide-co-glycolide) may have a lactide :glycolide ratio ranges from about 20:80 to about 2:98, a lactide.glycolide ratio of about 10:90, or a lactideiglycolide ratio of about 5:95. In one aspect, the poly(lactide-co- glycolide) is poly(L-lactide-co-glycolide). Other examples of biodegradable materials include polyglactin, polyglycolic acid, autogenous, heterogenous, and xenogeneic tissue (e.g., pericardium or small intestine submucosa), and oxidized, regenerated cellulose. These meshes can be knitted, woven or non-woven meshes. Examples of non-woven meshes include electrospun materials.
Meshes and films may be prepared from non-biodegradable polymers. Representative examples of non-biodegradable compositions include ethylene-co-vinyl acetate copolymers, acrylic-based and methacrylic-based polymers (e.g., poly(acrylic acid), poly(methylacrylic acid), poly(methylmethacrylate), poly(hydroxyethylmethacrylate), poly(alkylcynoacrylate), poly(alkyl acrylates), poly(alkyl methacrylates)), polyolefins such as poly(ethylene) or poly(propylene), polyamides (e.g., nylon 6,6), poly(urethanes) (e.g., poly(ester urethanes), poly(ether urethanes)., polycarbonate urethanes), poiy(ester-urea)), polyesters (e.g., PET, polybutyleneterephthalate, and polyhexyleneterephthalate), polyethers (poly(ethylene oxide), poly(propylene oxide), poly(ethylene oxide)- poly(propylene oxide) copolymers, diblock and triblock copolymers, poly(tetramethylene glycol)), silicone containing polymers and vinyl-based polymers (polyvinylpyrrolidone, polyvinyl alcohol), polyvinyl acetate phthalate), poly(styrene-co-isobutylene-co-styrene), fluorine containing polymers (fluoropolymers) such as fluorinated ethylene propylene (FEP) or polytetrafluoroethylene (e.g., expanded PTFE).
The mesh or film material may comprise a combination of the above-mentioned biodegradable and non-degradable polymers. Further examples of polymers that may be used are either anionic (e.g., alginate, carrageenin, hyaluronic acid, dextran sulfate, chondroitin sulfate, carboxymethyl dextran, caboxymethyl cellulose and poly(acrylic acid)], or cationic [e.g., chitosan, poly-l-lysine, polyethylenimine, and poly(allyl amine)) (see generally, Dunn et al., J. Applied Polymer Sci. 50:353, 1993; Cascone et al., J. Materials Sci.: Materials in Medicine 5:770, 1994; Shiraishi et al., Biol. Pharm. Bull. -/6:1164, 1993; Thacharodi and Rao, Int'I J. Pharm. 720:115, 1995; Miyazaki et al., Int'I J. Pharm. 118:257, 1995). Preferred polymers (including copolymers and blends of these polymers) include poly(ethylene-co-vinyl acetate), polycarbonate urethanes), poly(hydroxyl acids) (e.g., poly(D,L-lactic acid) oligomers and polymers, poly(L-lactic acid) oligomers and polymers, poly(D-lactic acid) oligomers and polymers, poly(glycolic acid), copolymers of lactic acid and glycolic acid, copolymers of lactide and glycolide, poly(caprolactone), copolymers of lactide or glycolide and ε-caprolactone), poly(valerolactone), poly(anhydrides), copolymers prepared from caprolactone and/or lactide and/or glycolide and/or polyethylene glycol.
A variety of polymeric and non-polymeric films and meshes have been described which may be combined with an anti-scarring agent. For example, the film or mesh may be a biodegradable polymeric matrix that conforms to the tissue and releases the agent in a controlled release manner. See, e.g., U.S. Patent No. 6,461 ,640. The film or mesh may be a self-adhering silicone sheet which is impregnated with an antioxidantand/or antimicrobial. See, e.g., U.S. Patent No. 6,572,878. The film or mesh may be a pliable shield with attachment ports and fenestrations that is adapted to cover a bony dissection in the spine. See, e.g., U.S. Patent No. 5,868,745 and U.S. Patent Application No. 2003/0078588. The film or mesh may be a resorbable micro-membrane having a single layer of non-porous polymer base material of poly-lactide. See, e.g., U.S. Patent No. 6,531,146 and U.S. Application No. 2004/0137033. The film or mesh may be a flexible neuro decompression device that has an outer surface texturized with microstructures to reduce fibroplasia when it is wrapped around a nerve in a canal. See, e.g., U.S. Patent No. 6,106,558. The film or mesh may be a resorbable collagen membrane that is wrapped around the spinal chord to inhibit cell adhesions. See, e.g., U.S. Patent No. 6,221 ,109. The film or mesh may be a wound dressing garment composed of an outer pliable layer and a self-adhesive inner gel lining which serves as a dressing for contacting wounds. See, e.g., U.S. Patent No. 6,548,728. The film or mesh may be a bandage with a scar treatment pad with a layer of silicone elastomer or silicone gel. See, e.g., U.S. Patent Nos. 6,284,941 and 5,891 ,076. The film or mesh may be a crosslinkable system with at least three reactive compounds each having a polymeric molecular core with at least one functional group. See, e.g., U.S. Patent No. 6,458,889. The film or mesh may be composed of a prosthetic fabric having a 3-dimensional structure separating two surfaces in which one is open to post-surgical cell colonization and one is linked to a film of collagenous material. See, e.g., U.S. Patent No. 6,451 ,032. The film or mesh may be composed by crosslinking two synthetic polymers, one having nucleophilic groups and the other having electrophilic groups, such that they form a matrix that may be used to incorporate a biologically active compound. See, e.g., U.S. Patent Nos. 6,323,278; 6,166,130; 6,051,648 and 5,874,500. The film or mesh may be a film composed of hetero-bifunctional anti-adhesion binding agents that act to covalently link substrate materials, such as collagen, to receptive tissue. See, e.g., U.S. Patent No. 5,580,923. The film or mesh may be a conformable warp-knit fabric of oxidized regenerated cellulose or other bioresorbable material which acts like a physical barrier to prevent postoperative adhesions. See, e.g., U.S. Patent No. 5,007,916. Meshes for use in the practice of the invention also are described in U.S. Patent Nos. 6,575,887, and co-pending application, entitled "Perivascular Wraps," filed September 26, 2003 (U.S. Sen No. (U.S. Ser. No. 10/673,046).
In one aspect, the mesh may be suitable for use in hernia repair surgery or in other types of surgical procedures. Mesh fabrics for use in connection with hernia repairs are disclosed in U.S. Patent Nos. 6,638,284; 5,292,328; 4,769,038 and 2,671,444. Surgical meshes may be produced by knitting, weaving, braiding, or otherwise forming a plurality of yarns (e.g., monofilament or multifilament yarns made of polymeric materials such as polypropylene and polyester) into a support trellis. Knitted and woven fabrics constructed from a variety of synthetic fibers and the use of the fabrics, in surgical repair are also discussed in U.S. Patent Nos. 3,054,406; 3,124,136; 4,193,137; 4,347,847; 4,452,245; 4,520,821 ; 4,633,873; 4,652,264; 4,655,221; 4,838,884 and 5,002,551 and European Patent Application No. 334,046. Implantable hernia meshes are described in U.S. Patent Nos. 6,610,006; 6,368,541 and 6,319,264. Hernia meshes for the repair of hiatal hernias are described in, e.g., U.S. Patent No. 6,436,030. Hernia meshes for the repair of abdominal (e.g., ventral and umbilical) hernias are described in U.S. Patent No. 6,383,201. Infection- resistant hernia meshes are described in, e.g., U.S. Patent No. 6,375,662. Hernia meshes such as those described in the patents listed above are suitable for combining with a fibrosis-inducing agent to create a mesh which promotes the growth of fibrous tissue.
In one aspect, the fibrosis-inhibiting agent can be incorporated into a biodegradable or dissolvable film or mesh that is then applied to the treatment site prior or post implantation of the prosthesis/implant. Exemplary materials for the manufacture of these films or meshes are hyaluronic acid (crosslinked or non-crosslinked), cellulose derivatives (e.g., hydroxypropyl cellulose), PLGA, collagen and crosslinked poly(ethylene glycol).
The film or mesh may be in the form of a tissue graft, which may be an autograft, allograft, biograft, biogenic graft or xenograft. Tissue grafts may be derived from various tissue types. Representative examples of tissues that may be used to prepare biografts include, but are not limited to, rectus sheaths, peritoneum, bladder, pericardium, veins, arteries, diaphragm and pleura. The biograft may be harvested from a host, loaded with an anti-scarring agent and then applied in a perivascular manner at the site where lesions and intimal hyperplasia can develop (e.g. , at an anastomotic site). Once implanted, the agent (e.g., ZD-6474, AP-23573, synthadotin, S-0885, aplidine, ixabepilone, IDN-5390, SB-2723005, ABT- 518, combretastatin, anecortave acetate, SB-715992, temsirolimus, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, dolastatin 15, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17-DMAG, tacrolimus, loteprednol etabonate, juglone, prednisolone, puromycin, 3-BAABE, cladribine, mannose-6-phosphate, 5-azacytidine, Ly333531 (ruboxistaurin), or simvastatin)) is released from the graft and can penetrate the vessel wall to prevent the formation of intimal hyperplasia at the treatment site. In certain embodiments, the biograft may be used as a backing layer to enclose a composition (e.g., a gel or paste loaded with anti-scarring agent). Films and meshes, which may be combined with one or more anti-scarring agents according to the present invention, include commercially available products. Examples of films and meshes into which a fibrosis agent can be incorporated include INTERCEED (Johnson & Johnson, Inc.), PRECLUDE (W.L. Gore), and POLYACTIVE (poly(ether ester) multiblock copolymers (Osteotech, Inc., Shrewsbury, NJ), based on poly(ethylene glycol) and poly(butylene terephthalate), and SURGICAL absorbable hemostat gauze-like sheet from Johnson & Johnson. Another mesh is a prosthetic polypropylene mesh with a bioresorbable coating called SEPRAMESH Biosurgical Composite (Genzyme Corporation, Cambridge, MA). One side of the mesh is coated with a bioresorbable layer of sodium hyaluronate and carboxymethylcellulose, providing a temporary physical barrier that separates the underlying tissue and organ surfaces from the mesh. The other side of the mesh is uncoated, allowing for complete tissue ingrowth similar to bare polypropylene mesh. In one embodiment, the fibrosis-inducing agent may be applied only to the uncoated side of SEPRAMESH and not to the sodium hyaluronate/ carboxymethylcellulose coated side. Other films and meshes include: (a) BARD MARLEX mesh (CR. Bard, Inc.), which is a very dense knitted fabric structure with low porosity; (b) monofilament polypropylene mesh such as PROLENE available from Ethicon, Inc. Somerville, NJ (see, e.g., U.S. Patent Nos. 5,634,931 and 5,824,082)); (c) SURGISIS GOLD and SURGISIS IHM soft tissue graft (both from Cook Surgical, Inc.) which are devices specifically configured for use to reinforce soft tissue in repair of inguinal hernias in open and laparoscopic procedures; (d) thin walled polypropylene surgical meshes such as are available from Atrium Medical Corporation (Hudson, NH) under the trade names PROLITE, PROLITE ULTRA, and LITEMESH; (e) COMPOSIX hernia mesh (CR. Bard, Murray Hill, NJ), which incorporates a mesh patch (the patch includes two layers of an inert synthetic mesh, generally made of polypropylene, and is described in U.S. Patent No. 6,280,453) that includes a filament to stiffen and maintain the device in a flat configuration; (f) VISILEX mesh (from CR. Bard, Inc.), which is a polypropylene mesh that is constructed with monofilament polypropylene; (g) other meshes available from CR. Bard, Inc. which include PERFIX Plug, KUGEL Hernia Patch, 3D MAX mesh, LHI mesh, DULEX mesh, and the VENTRALEX Hernia Patch; and (h) other types of polypropylene monofilament hernia mesh and plug products include HERTRA mesh 1 , 2, and 2A, HERMESH 3, 4 & 5 and HERNIAMESH plugs Tl, T2, and T3 from Hemiamesh USA, Inc. (Great Neck, NY).
Other examples of commercially available meshes which may be combined with fibrosis-inhibiting agents are described below. One example includes a prosthetic polypropylene mesh with a bioresorbable coating sold under the trade name SEPRAMESH Biosurgical Composite (Genzyme Corporation). One side of the mesh is coated with a bioresorbable layer of sodium hyaluronate and carboxymethylcellulose, providing a temporary physical barrier that separates the underlying tissue and organ surfaces from the mesh. The other side of the mesh is uncoated, allowing for complete tissue ingrowth similar to bare polypropylene mesh. In one embodiment, the fibrosis-inducing agent may be applied only to the uncoated side of SEPRAMESH and not to the sodium hyaluronate/ carboxymethylcellulose coated side. Boston Scientific Corporation sells the TRELEX NATURAL Mesh which is composed of a unique knitted polypropylene material. Ethicon, Inc. makes the absorbable VICRYL (polyglactin 910) meshes (knitted and woven) and MERSILENE Polyester Fiber Mesh. Dow Corning Corporation (Midland, MIj sells a mesh material formed from silicone elastomer known as SILASTIC Rx Medical Grade Sheeting (Platinum Cured). United States Surgical / Syneture (Norwalk, CT) sells a mesh made from absorbable polyglycolic acid under the trade name DEXON Mesh Products. Membrana Accurel Systems (Obemburg, Germany) sells the CELGARD microporous polypropylene fiber and membrane. Gynecare Worldwide, a division of Ethicon, Inc. sells a mesh material made from oxidized, regenerated cellulose known as INTERCEED TC7. Integra LifeSciences Corporation (Plainsboro, NJ) makes DURAGEN PLUS Adhesion Barrier Matrix, which can be used as a barrier against adhesions following spinal and cranial surgery and for restoration of the dura mater. HYDROSORB Shield from MacroPore Biosurgery, Inc. (San Diego, CA) is a film for temporary wound support to control the formation of adhesions in specific spinal applications.
Numerous polymeric and non-polymeric carrier systems that can be used with films and meshes have been described above. Methods for incorporating the fibrosis-inhibiting compositions onto or into the film or mesh include: (a) affixing (directly or indirectly) to the film or mesh a fibrosis-inhibiting composition (e.g., by either a spraying process or dipping process as described above, with or without a carrier), (b) incorporating or impregnating into the film or mesh a fibrosis-inhibiting composition (e.g., by either a spraying process or dipping process as described above, with or without a carrier (c) by coating the film or mesh with a substance such as a hydrogel which will in turn absorb the fibrosis-inhibiting composition, (d) constructing the film or mesh itself or a portion of the film or mesh with a fibrosis-inhibiting composition, or (e) by covalently binding the fibrosis- inhibiting agent directly to the film or mesh surface or to a linker (small molecule or polymer) that is coated or attached to the film or mesh surface. For devices that are coated, the coating process can be performed in such a manner as to (a) coat only one surface of the film or mesh or (b) coat all or parts of both sides of the film or mesh.
The therapeutic agent(s) may be an integral part of the film or mesh (i.e., may reside within the fibers of the mesh). The fibrosis inhibiting agent can be incorporated directly into the film or mesh or it can be incorporated into a secondary carrier (polymeric or non-polymeric), as described above, that is then incorporated into the film or mesh.
The film or mesh may be coated with a fibrosis-inhibiting agent or a composition that includes the fibrosis-inhibiting agent. In some embodiments, the composition is a polymer composition can function as a surgical adhesion barrier. The coating may take the form of a surface- adherent coating, mask, film, gel, foam, or mold.
A variety of polymeric compositions have been described that may be used in conjunction with the films and meshes of the invention. Such compositions may be in the form of, for example, gels, sprays, liquids, and pastes, or may be polymerized from monomeric or prepolymeric constituents in situ. For example, the composition may be a polymeric tissue coating which is formed by applying a polymerization initiator to the tissue and then covering it with a water-soluble macromer that is polymerizable using free radical initiators under the influence of UV light. See, e.g., U.S. Patent Nos. 6,177,095 and 6,083,524. The composition may be an aqueous composition including a surfactant, pentoxifylline and a polyoxyalkylene polyether. See, e.g., U.S. Patent No. 6,399,624. The composition may be a hydrogel-forming, self-solvating, absorbable polyester copolymers capable of selective, segmental association into compliant hydrogels mass upon contact with an aqueous environment. See, e.g., U.S. Patent No. 5,612,052. The composition may be composed of fluent prepolymeric material that is emitted to the tissue surface and then exposed to activating energy in situ to initiate conversion of the applied material to non- fluent polymeric form. See, e.g., U.S. Patent Nos. 6,004,547 and 5,612,050. The composition may be composed of a gas mixture of oxygen present in a volume ratio of 1 to 20%. See, e.g., U.S. Patent No. 6,428,500. The composition may be composed of an anionic polymer having an acid sulfate and sulfur content greater than 5% which acts to inhibit monocyte or macrophage invasion. See, e.g., U.S. Patent No. 6,417,173. The composition may be composed of a non-gelling polyoxyalkylene composition with or without a therapeutic agent. See, e.g., U.S. Patent No. 6,436,425. The composition may be coated onto tissue surfaces and may be composed of an aqueous solution of a hydrophilic, polymeric material (e.g., polypeptides or polysaccharide) having greater than 50,000 molecular weight and a concentration range of 0.01% to 15% by weight. See, e.g., U.S. Patent No. 6,464,970.
Other representative examples of polymeric compositions which may be coated onto the film or mesh include poly(ethylene glycol)- based systems, hyaluronic acid and crosslinked hyaluronic acid compositions. These compositions can be applied as the final composition or they can be applied as materials that form crosslinked gel in situ.
Other compositions that can be used in conjunction with films and meshes, include, but are not limited to: (a) sprayable PEG-containing formulations such as COSEAL, SPRAYGEL, FOCALSEAL or DURASEAL; (b) hyaluronic acid-containing formulations such as RESTYLANE, HYLAFORM, PERLANE, SYNVISC, SEPRAFILM, SEPRACOAT, INTERGEL, (c) polymeric gels such as REPEL or FLOWGEL, (d) dextran sulfate gels such as the ADCON range of products, (e) lipid based compositions such as ADSURF (Brittania Pharmaceuticals).
The film or mesh (or device comprising the film or mesh) may be made sterile either by preparing them under aseptic environment and/or they may be terminally sterilized using methods known in the art, such as gamma radiation or electron beam sterilization methods or a combination of both of these methods.
Films and meshes may be applied to any bodily conduit or any tissue that may be prone to the development of fibrosis or intima! hyperplasia. Prior to implantation, the film or mesh may be trimmed or cut from a sheet of bulk material to match the configuration of the widened foramen, canal, or dissection region, or at a minimum, to overlay the exposed tissue area. The film or mesh may be bent or shaped to match the particular configuration of the placement region. The film or mesh may also be rolled in a cuff shape or cylindrical shape and placed around the exterior periphery of the desired tissue. The film or mesh may be provided in a relatively large bulk sheet and then cut into shapes to mold the particular structure and surface topography of the tissue or device to be wrapped. Alternatively, the film or mesh may be pre-shaped into one or more patterns for subsequent use. The films and meshes may be typically rectangular in shape and be placed at the desired location within the surgical site by direct surgical placement or by endoscopic techniques. The film or mesh may be secured into place by wrapping it onto itself (i.e., self-adhesive), or by securing it with sutures, staples, sealant, and the like. Alternatively, the film or mesh may adhere readily to tissue and therefore, additional securing mechanisms may not be required.
The films or meshes of the invention may be used for a variety of indications, including, without limitation: (a) prevention of surgical adhesions between tissues following surgery (e.g., gyneacologic surgery, vasovasostomy, hernia repair, nerve root decompression surgery and laminectomy); (b) prevention of hypertrophic scars or keloids (e.g., resulting from tissue burns or other wounds); (c) prevention of intimal hyperplasia and/or restenosis (e.g., resulting from insertion of vascular grafts or hemodialysis access devices); or (d) may be used in affiliation with devices and implants that lead to scarring as described herein (e.g., as a sleeve or mesh around a breast implant to reduce or inhibit scarring).
In one embodiment, films or meshes may be used to prevent adhesions that occur between tissues following surgery, injury or disease. Adhesion formation, a complex process in which bodily tissues that are normally separate grow together, occurs most commonly as a result of surgical intervention and/or trauma. Generally, adhesion formation is an inflammatory reaction in which factors are released, increasing vascular permeability and resulting in fibrinogen influx and fibrin deposition. This deposition forms a matrix that bridges the abutting tissues. Fibroblasts accumulate, attach to the matrix, deposit collagen and induce angiogenesis. If this cascade of events can be prevented within 4 to 5 days following surgery, then adhesion formation can be inhibited. Adhesion formation or unwanted scar tissue accumulation and encapsulation complicates a variety of surgical procedures and virtually any open or endoscopic surgical procedure in the abdominal or pelvic cavity. Encapsulation of surgical implants also complicates breast reconstruction surgery, joint replacement surgery, hernia repair surgery, artificial vascular graft surgery, and neurosurgery. In each case, the implant becomes encapsulated by a fibrous connective tissue capsule which compromises or impairs the function of the surgical implant (e.g., breast implant, artificial joint, surgical mesh, vascular graft, dural patch). Chronic inflammation and scarring also occurs during surgery to correct chronic sinusitis or removal of other regions of chronic inflammation (e.g., foreign bodies, infections (fungal, mycobacterium). Surgical procedures that may lead to surgical adhesions may include cardiac, spinal, neurologic, pleural, thoracic and gynaecologic surgeries. However, adhesions may also develop as a result of other processes, including, but not limited to, non-surgical mechanical injury, ischemia, hemorrhage, radiation treatment, infection-related inflammation, pelvic inflammatory disease and/or foreign body reaction. This abnormal scarring interferes with normal physiological functioning and, in come cases, can force and/or interfere with follow-up, corrective or other surgical operations. For example, these post-operative surgical adhesions occur in 60 to 90% of patients undergoing major gynaecologic surgery and represent one of the most common causes of intestinal obstruction in the industrialized world. These adhesions are a major cause of failed surgical therapy and are the leading cause of bowel obstruction and infertility. Other adhesion- treated complications include chronic pelvic pain, urethral obstruction and voiding dysfunction.
Currently, preventative therapies, administered 4 to 5 days following surgery, are used to inhibit adhesion formation. Various modes of adhesion prevention have been examined, including (1) prevention of fibrin deposition, (2) reduction of local tissue inflammation, and (3) removal of fibrin deposits. Fibrin deposition is prevented through the use of physical adhesion barriers that are either mechanical or comprised of viscous solutions. Although many investigators are utilizing adhesion prevention barriers, a number of technical difficulties exist.
In one aspect, the present invention provides films and meshes that include an anti-scarring agent or a composition that includes an anti-scarring agent for use as surgical adhesion barriers.
In one aspect, films and meshes may be used to prevent surgical adhesions in the epidural and dural tissue which is a factor contributing to failed back surgeries and complications associated with spinal injuries (e.g., compression and crush injuries). Scar formation within dura and around nerve roots has been implicated in rendering subsequent spine operations technically more difficult. To gain access to the spinal foramen during back surgeries, vertebral bone tissue is often disrupted. Back surgeries, such as laminectomies and diskectomies, often leave the spinal dura exposed and unprotected. As a result, scar tissue frequently forms between the dura and the surrounding tissue. This scar is formed from the damaged erector spinae muscles that overlay the laminectomy site. This results in adhesion development between the muscle tissue and the fragile dura, thereby, reducing mobility of the spine and nerve roots which leads to pain and slow post-operative recovery. To circumvent adhesion development, a scar-reducing barrier may be inserted between the dural sleeve and the paravertebral musculature post-laminotomy. This reduces cellular and vascular invasion into the epidural space from the overlying muscle and exposed cancellous bone and thus, reduces the complications associated with the canal housing the spinal chord and/or nerve roots.
In another aspect, films and meshes comprising an anti- scarring agent may be used to prevent the fibrosis from occurring between a hernia repair mesh and the surrounding tissue. Hernias are abnormal protrusions (outpouchings) of an organ or other body structure through a defect or natural opening in a covering membrane, muscle or bone. Hernias themselves are not dangerous, but can become extremely problematic if they become incarcerated. Surgical prostheses used in hernia repair (referred to herein as "hernia meshes") include prosthetic mesh-or gauze- like materials, which support the repaired hernia or other body structures during the healing process. Hernias are often repaired surgically to prevent complications. Conditions in which a hernia mesh may need to be used include, without limitation, the repair of inguinal (i.e., groin), umbilical, ventral, femoral, abdominal, diaphragmatic, epigastric, gastroesophageal, hiatal, intermuscular, mesenteric, paraperitoneal, rectovaginal, rectocecal, uterine, and vesical hernias. Hernia repair typically involves returning the viscera to its normal location and the defect in the wall is primarily closed with sutures, but for bigger gaps, a mesh is placed over the defect to close the hernia opening. Inclusion of an anti-scarring agent or composition comprising an anti-scarring agent into or onto a hernia repair mesh may reduce or prevent fibrosis proximate to the implanted hernia mesh, thereby minimizing the possibility of adhesions between the abdominal wall or other tissues and the mesh itself, and reducing further complications and abdominal pain.
In yet another aspect, films or meshes may be used to prevent hypertrophic scars or keloids (e.g., resulting from tissue bums or other wounds). Hypertrophic scars and keloids are the result of an excessive fibroproliferative wound healing response. Briefly, healing of wounds and scar formation occurs in three phases: inflammation, proliferation, and maturation. The first phase, inflammation, occurs in response to an injury which is severe enough to break the skin. During this phase, which lasts 3 to 4 days, blood and tissue fluid form an adhesive coagulum and fibrinous network which serves to bind the wound surfaces together. This is then followed by a proliferative phase in which there is ingrowth of capillaries and connective tissue from the wound edges, and closure of the skin defect. Finally, once capillary and fibroblastic proliferation has ceased, the maturation process begins wherein the scar contracts and becomes less cellular, less vascular, and appears flat and white. This final phase may take between 6 and 12 months. If too much connective tissue is produced and the wound remains persistently cellular, the scar may become red and raised. If the scar remains within the boundaries of the original wound it is referred to as a hypertrophic scar, but if it extends beyond the original scar and into the surrounding tissue, the lesion is referred to as a keloid. Hypertrophic scars and keloids are produced during the second and third phases of scar formation. Several wounds are particularly prone to excessive endothelial and fibroblastic proliferation, including burns, open wounds, and infected wounds. With hypertrophic scars, some degree of maturation occurs and gradual improvement occurs. In the case of keloids however, an actual tumor is produced which can become quite large. Spontaneous improvement in such cases rarely occurs. A film or mesh that comprises an anti-scarring agent or a composition that comprises an anti- scarring agent may be placed in contact with a wound or burn site in order to prevent formation of hypertrophic scar or keloids.
In yet another aspect, films and meshes are provided that may be used for delivering an anti-scarring agent to an external portion (surface) of a body passageway or cavity. Examples of body passageways include arteries, veins, the heart, the esophagus, the stomach, the duodenum, the small intestine, the large intestine, biliary tracts, the ureter, the bladder, the urethra, lacrimal ducts, the trachea, bronchi, bronchiole, nasal airways, eustachian tubes, the external auditory mayal, vas deferens and fallopian tubes. Examples of cavities include the abdominal cavity, the buccal cavity, the peritoneal cavity, the pericardial cavity, the pelvic cavity, perivisceral cavity, pleural cavity and uterine cavity.
Examples of conditions that may be treated or prevented with fibrosis-inhibiting films and meshes include iatrogenic complications of arterial and venous catheterization, complications of vascular dissection, complications of gastrointestinal passageway rupture and dissection, restonotic complications associated with vascular surgery (e.g., bypass surgery), and intimal hyperplasia.
In one aspect, an anti-scarring agent may be delivered from a film or mesh to the external walls of body passageways or cavities for the purpose of preventing and/or reducing a proliferative biological response that may obstruct or hinder the optimal functioning of the passageway or cavity, including, for example, iatrogenic complications of arterial and venous catheterization, aortic dissection, cardiac rupture, aneurysm, cardiac valve dehiscence, graft placement (e.g., A-V-bypass, peripheral bypass, CABG), fistula formation, passageway rupture and surgical wound repair.
The films or meshes may be used in the form of a perivascular wrap to prevent restenosis at anastomotic sites resulting from insertion of vascular grafts or hemodialysis access devices. In this case, perivascular wraps may be incorporated with or coated with a fibrosis-inhibiting agent, which can be used in conjunction with a vascular graft to inhibit scarring at an anastomotic site. These films or meshes may be placed or wrapped in a perivascular (periadventitial) manner around the outside of the anastomosis at the time of surgery. Film and mesh implants comprising an anti-scarring agent may be used with synthetic bypass grafts (femoral-popliteal, femoral- femoral, axillary-femoral etc.), vein grafts (peripheral and coronary), internal mammary (coronary) grafts or hemodialysis grafts (AV fistulas, AV access grafts).
In order to further the understanding of such conditions, representative complications leading to compromised body passageway or cavity integrity are discussed in more detail below.
Cardiac Bypass Surgery
Coronary artery bypass graft ("CABG") surgery was introduced in the 1950s, and still remains a highly invasive, open surgical procedure, although less invasive surgical techniques are being developed. CABG surgery is a surgical procedure that is performed to overcome many types of coronary artery blockages. The purpose of bypass surgery is to increase the circulation and nourishment to the heart muscle that has been reduced due to arterial blockage. This procedure involves the surgeon accessing the heart and the diseased arteries, usually through an incision in the middle of the chest. Often, healthy arteries or veins are "harvested" from the patient to create "bypass grafts" that channel the needed blood flow around the blocked portions of the coronary arteries. The arteries or veins are connected from the aorta to the surface of the heart beyond the blockages thereby forming an autologous graft. This allows the blood to flow through these grafts and "bypass" the narrowed or closed vessel. The use of synthetic graft materials to create the "bypass" has been limited due to the lack of the appropriate biocompatibility of these synthetic grafts. CABG has significant short term limitations, including medical complications, such as stroke, multiple organ dysfunction, inflammatory response, respiratory failure and post-operative bleeding, each of which may result in death. Another problem associated with CABG is restenosis. Restenosis is typically defined as a renarrowing of an arterial blood vessel within six months of the CABG procedure. It typically occurs in approximately 25% to 45% of patients, and is the result of an excessive healing response to arterial injury after a revascularization procedure. Restenosis may occur within a short period following a procedure or may develop over the course of months or years. Longer term or " late" restenosis may result from excessive proliferation of scar tissue at the treatment site, the causes of which are not well understood. Thus any product that may reduce the incidence or magnitude of the restenotic process following CABG surgery can greatly enhance the well being of a patient.
In order to prevent the restenotic complications associated with CABG surgery, such as those discussed above, a wide variety of therapeutic agents (with or without a carrier) may be delivered to the external portion of the blood vessel. The carrier (e.g., polymer) or therapeutic agent/composition can be applied to the external portion of the vessel following the interventional or surgical procedure in order to prevent the restenotic complications.
Peripheral Bypass Surgery
Peripheral arterial disease (PAD) refers to diseases of any of the blood vessels outside of the heart. PAD is a range of disorders that may affect the blood vessels in the hands, arms, legs, or feet. The most common form of PAD is atherosclerosis. Atherosclerosis is a gradual process in which cholesterol and scar tissue build up in the arteries to form plaque. This build -up causes a gradual narrowing of the artery, which leads to a decrease in the amount of blood flow through that artery. When the flow of blood decreases, it results in a decrease of oxygen and nutrient supply to the body's tissues, which in turn may result in pain sensation. When the arteries to the legs are affected, the most common symptom is pain in the calf when walking. This is known as intermittent claudication.
Peripheral bypass surgery is a procedure to bypass an area of stenosed (narrowed) or blocked artery that is a result of atherosclerosis. In this surgical procedure, a synthetic graft (artificial blood vessels) or an autologous graft, vein, will be implanted to provide blood flow around the diseased area. First, the surgeon makes an incision in the leg, thigh, calf or ankle skin. The location of the incision may vary based on which vessels need to be bypassed and where there is healthy artery to connect to maintain the blood flow. The bypass graft is sewn into the artery above the stenosis or blockage, and below the stenosis or blockage. This bypass provides a means whereby blood will reach the tissue that has not been receiving enough blood and oxygen. Synthetic bypass grafts used in the legs are usually made of ePTFE.
Restenosis and occlusion of bypass grafts are one of the most important problems in peripheral bypass surgery. This restenosis is caused by neointimal growth (hyperplasia) and is especially pronounced within artificial graft material. This restenosis is usually at the anastomotic site where the graft and artery are connected via a surgical procedure. The Arterio-Venous (AV) Graft Surgery
The AV graft surgical procedure is used for similar application as those for the AV fistula {e.g., hemodialysis patients). For the AV graft surgery, a synthetic graft material is used to connect the artery to the vein rather that the direct connection of the artery to the vein as is the case for the AV fistula. The incidence of intimal hyperplasia, which leads to occlusion of the graft, is one of the main factors that affect the long term patency of these grafts. This intimal hyperplasia may occur at the venous anastomosis and at the floor of the vein. A product that may reduce or prevent this occurrence of intimal hyperplasia will increase the duration of patency of these grafts. In order to reduce the occurrence of intimal hyperplasia at the venous anastomosis of an AV graft, a wide variety of therapeutic agents (with or without a carrier) /compositions may be delivered to the external portion of the blood vessel. The polymer or therapeutic agent/composition can be applied to the external portion of the vessel/anastomotic site following the interventional or surgical procedure in order to prevent the restenotic complications.
Anastomotic Closure Devices
Anastomotic closure devices provide a means for rapidly repairing an anastomosis. The use of some of these devices requires an invasive surgical procedure. In one embodiment of this invention, following the use of an anastomotic closure device, the mesh containing the therapeutic agent may be wrapped around the anastomosis and the anastomotic closure device, if it is left at the surgical site.
In one embodiment, the invention provides a method for treating or preventing intimal hyperplasia that includes delivering to an anastomotic site a delivery device. The device includes a therapeutic agent and a biodegradable polymer, wherein at least some of the biodegradable polymer is in the form of a mesh. Exemplary anastomotic sites include venous anastomosis, arterial anastomosis, arteriovenous fistula, arterial bypass, and arteriovenous graft. Preferably, the device includes a polymer
553 intimal tissue typically grows from the native vessel into the graft. In order to prevent the restenotic complications associated with peripheral bypass surgery, such as those discussed above, a wide variety of therapeutic agents (with or without a carrier) /compositions may be delivered to the external portion of the blood vessel. The polymer or therapeutic agent/composition can be applied to the external portion of the vessel/anastomotic site following the interventional or surgical procedure in order to prevent the restenotic complications.
Arterio-Venous (AV) Fistula
The arterio-venous (AV) fistula is surgically created vascular connection which allows the flow of blood from an artery directly to a vein. The AV fistula was first created by researchers for kidney failure patients who must undergo kidney dialysis.
Hemodialysis requires a viable artery and vein to draw blood from and jeturn it to the body. The repeated puncturing often either causes a vein or artery to fail or causes other complications for the patient. The AV fistula increases the amount of possible puncture sites for hemodialysis and minimizes the damage to the patient's natural blood vessels. The connection that is created between the vein and artery forms a large blood vessel that continuously supplies an increased blood flow for performing hemodialysis.
Restenosis and eventual occlusion are one of the most important problems in the long term patency of the AV fistula. In order to prevent the restenotic complications associated with the surgical formation of an AV fistula, a wide variety of therapeutic agents (with or without a carrier) /compositions may be delivered to the external portion of the blood vessel. The polymer or therapeutic agent/composition can be applied to the external portion of the vessel/anastomotic site following the interventional or surgical procedure in order to prevent the restenotic complications.
552 mesh with a therapeutic agent is delivered to an external portion of an anastomotic site.
Transplant Applications
There are many applications in which various organs in the human body fail to function in a manner to sustain the well being of the patient. When an appropriate donor organ is available, an impaired organ may be replaced by a donor organ {e.g., lung, heart, kidney etc). One of the potential complications following these transplant surgeries is the potential for stenosis to occur in the blood vessels at or near the anastomotic site between the donor and recipient vessels. For example, transplant renal artery stenosis is a complication that may occur following a kidney transplant. Transplant renal artery stenosis is when the artery from the abdominal aorta to the kidney narrows, limiting blood flow to the kidney. This may also make it difficult to keep blood pressure under control. Treatment typically involves expanding the narrowed segment using a small balloon.
One method to treat this stenosis is to apply the composition of this invention around the anastomotic site (junction of the donor and recipient vessels) in a perivascular manner. In a similar manner, the composition of this invention may be applied in a peritubular manner to the exterior surfaces of the trachea and or bronchi following a lung transplant procedure.
According to the present invention, any scarring agent described above can be utilized in the practice of this embodiment. Films and meshes may be adapted to contain and/or release an agent that inhibits one or more of the four general components of the process of fibrosis (or scarring), including: formation of new blood vessels (angiogenesis), migration and proliferation of connective tissue cells (such as fibroblasts or smooth muscle cells), deposition of extracellular matrix (ECM), and remodeling (maturation and organization of the fibrous tissue). As films and meshes are made in a variety of configurations and sizes, the exact dose administered will vary with device size, surface area and design. However, certain principles can be applied in the application of this art. Drug dose can be calculated as a function of dose per unit area (of the portion of the device being coated), total dose administered, and appropriate surface concentrations of active drug can be determined. Drugs are to be used at concentrations that range from several times more than to 10%, 5%, or even less than 1% of the concentration typically used in a single chemotherapeutic systemic dose application. Preferably, the drug is released in effective concentrations for a period ranging from 1 - 90 days.
Several examples of fibrosis-inhibiting agents for use in films or meshes include the following: ZD-6474, AP-23573, synthadotin, S-0885, aplidine, ixabepilone, IDN-5390, SB-2723005, ABT-518, combretastatin, anecortave acetate, SB-715992, temsirolimus, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, as well as analogues and derivatives of the aforementioned.
Additional examples of anti-scarring agents which can be used include those having a high potency in the assays described herein (1- 10OnM IC50 range) such as isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, dolastatin 15, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17- DMAG, and tacrolimus; those having a mid-potency in the assays described herein (100-500 nM IC5O range) such as loteprednol etabonate, juglone, prednisolone, puromycin, 3-BAABE, cladribine, and mannose-6-phosphate; and those having a low potency in the assays described herein (500- 1000nm range IC50 range) such as 5-azacytidine, Ly333531(ruboxistaurin), and simvastatin.
Regardless of the method of application of the drug to the film or mesh, the exemplary anti-fibrosing agents, used alone or in combination, should be administered under the following dosing guidelines. The total amount (dose) of anti-scarring agent in or on the film or mesh may be in the range of about 0.01 μg-10 μg, or 10 μg-10 mg, or 10 mg-250 mg, or 250 mg-1000 mg, or 1000 mg-2500 mg. The dose (amount) of anti-scarring agent per unit area of device surface to which the agent is applied may be in the range of about 0.01 μg/mm2 - 1 μg/mm2, or 1 μg/mm2 - 10 μg/mm2, or 10 μg/mm2 - 250 μg/mm2, 250 μg/mm2 - 1000 μg/mm2, or 1000 μg/mm2 - 2500 μg/mm2.
Provided below are exemplary dosage ranges for various anti- scarring agents that can be used in conjunction with films or meshes in accordance with the invention. (A) Angiogenesis inhibitors including Alphastatin, ZD-6474, IDN-5390, SB-2723005, ABT-518, combretastatin, and anecortane, analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10 s - 10"4 M of agent is to be maintained on the implant or barrier surface. (B) mTOR inhibitors including AP-23573 and Temsirolimus, analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10"8- 10"4 M of agent is to be maintained on the implant or barrier surface. (C) Tubulin antagonists including synthadotin, analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10 s- 10"4 M of agent is to be maintained on the implant or barrier surface. (D) Epithilones including ixabepilone and analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10^- 10"4 M of agent is to be maintained on the implant or barrier surface. (E) Kinesin antagonists including SB- 715992 and analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10^- 10"4 M of agent is to be maintained on the implant or barrier surface. (F) TNF alpha antagonists including Etanercept, humicade, adalimumab and analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10'8- 10~4 M of agent is to be maintained on the implant or barrier surface. (G) AKT inhibitor including erucylphosphocholine and analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10"8- 10"4 M of agent is to be maintained on the implant or barrier surface. (H) FGF Inhibitors including IDN-5390 and analogues and derivatives thereof: total dose not to, exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10"8- 10"4 M of agent is to be maintained on the implant or barrier surface. (I) Collagenase Antagonists including S-0885 and analogues and derivatives thereof: total dose not to exceed 1000 mg (range of 0.1 μg to 1000 mg); preferred 1 μg to 500 mg. Dose per unit area of 0.01 μg - 500 μg per mm2; preferred dose of 0.1 μg/mm2 - 100 μg/mm2. Minimum concentration of 10^- 10"4 M of agent is to be maintained on the implant or barrier surface. (J) NF KAPPA B Inhibitors including Bortezomib dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10^- 10"4 M of agent is to be maintained on the implant or barrier surface. (K) Elongation factor-1 alpha inhibitors including aplidine and analogues and derivatives thereof: total dose not to exceed 1000 mg (range of 0.1 μg to 1000 mg); preferred 1 μg to 500 mg. Dose per unit area of 0.01 μg - 500 μg per mm2; preferred dose of 0.1 μg/mm2 - 100 μg/mm2. Minimum concentration of 10"8- 10'4 M of agent is to be maintained on the implant or barrier surface. (L) Tyrosine kinase inhibitors including Gefitinib and analogues and derivatives thereof: total dose not to exceed 1000 mg (range of 0.1 μg to 1000 mg); preferred 1 μg to 500 mg. Dose per unit area of 0.01 μg - 500 μg per mm2; preferred dose of 0.1 μg/mm2 - 100 μg/mm2. Minimum concentration of 10'8 - 10"4 M of agent is to be maintained on the implant or barrier surface.
For high potency drugs, the total dose typically should not exceed 200 mg (range of 0.1 μg to 200 mg) and preferably 1 μg to 100 mg; dose per unit area of 0.01 μg - 100 μg per mm2; preferably 0.1 μg/mm2 - 20 μg/mm2; and minimum concentration of 10^- 10"4 M of agent should be maintained on the implant or barrier surface. For mid-potency drugs, the total dose typically should not to exceed 500 mg (range of 1.0 μg to 500 mg) and preferably 1 μg to 200 mg; dose per unit area of 0JD1 μg - 200 μg per mm2, preferably 0.1 μg/mm2 - 40 μg/mm2; and minimum concentration of 10' 8- 10"4 M of agent should be maintained on the implant or barrier surface. For low potency drugs, the total dose typically should not exceed 1000 mg (range of 0.1 μg to 1000 mg), preferably 1 μg to 500 mg; dose per unit area of 0.01 μg - 500 μg per mm2; preferably 0.1 μg/mm2 - 100 μg/mm2; and minimum concentration of 10"8- 10"4 M of agent should to be maintained on the implant or barrier surface.
Glaucoma Drainage Devices
In one aspect, the present invention provides for the combination of an anti-scarring agent and a glaucoma drainage device.
Various types of glaucoma drainage devices have been described. Some glaucoma drainage devices include a plate and a tube. The function of the tube is to deliver aqueous from within the eye onto the upper surface of the episcleral plate. The episcleral plate is firmly sutured to the sclera and covered by a thick flap of Tenon's tissue and conjunctiva. The function of the plate is to initiate the formation of a large circular bleb which develops a specialized fibrovascular bleb lining and becomes distended by aqueous. It is this fibrovascular bleb lining which is responsible for regulating the escape of aqueous from the eye and which determines the final level of intraocular pressure (lOP) that is achieved after insertion of the implant. If the fibrovascular response is too great, the drainage capability of the device is reduced. In an embodiment of the present invention, a fibrosis-inhibiting agent is incorporated into or onto all or a portion of the device such that the released fibrosis-inhibiting agent modulates the healing response, thereby enabling the device to function correctly.
Glaucoma drainage devices may be, for example, a conduit attached to an episcleral drainage plate having a porous posterior surface for cellular ingrowth and attachment by the sclera. See, e.g., U.S. Patent No. 5,882,327. The glaucoma drainage device may be composed of a foldable and reliable episcleral plate and a drainage tube whereby the device may be delivered to the implant site through an injection delivery system. See, e.g., U.S. Patent No. 6,589,203. The glaucoma drainage device may be pressure regulator composed of a base plate formed of a thin, flexible rubber material (e.g., silicone rubber) which has a mounted housing chamber that is attached to a tube. See, e.g., U.S. Patent No. 5,752,928. The glaucoma drainage device may be composed of an elastomeric plate having a sealing member that conforms to the sclera to restrict fluid and an attached non-valved elastomeric drainage tube. See, e.g., U.S. Patent No. 5,476,445. The glaucoma drainage device may be composed of ridged plates that extend outwardly that are concave on one side to match the curvature of the sclera and are adapted for side by side attachment to the sclera whereby a tube extends between the ridged plates for communication. See, e.g., U.S. Patent No. 4,457,757. The glaucoma drainage device may be composed of a thin, elliptical, elastomeric plate having a centrally positioned hole for growth of scar tissue and an elastomeric drainage tube attached to the plate for fluid communication with the eye. See, e.g., U.S. Patent No. 5,397,300. The glaucoma drainage device may be composed of a tube with a circumferential hole with a connected disk at the outlet end of the tube for placing on a surface of an eyeball. See, e.g., U.S. Patent No. 5,868,697. The glaucoma drainage device may be a tube with a flow controlling structure that constricts flow passage within the tube and has at least one circumferential hole within the tube that is temporarily occluded with an absorbable material. See, e.g., U.S. Patent No. 6,203,513. The glaucoma drainage device may be composed of a tube with an engagement means and a porous, liquid- absorbing plug with an attached filamentary extension that substantially restricts fluid flow. See, e.g., U.S. Patent No. 5,300,020. The glaucoma drainage device may be a resilient polymeric drain implant with a passage extending between the ends and flanges that project radially from the body. See, e.g., U.S. Patent No. 4,968,296. The glaucoma drainage device may¬ be a shunt to divert aqueous humor in the eye from the anterior chamber into a portion of the device that branches to provide fluid communication in either direction along the Schlemm's canal. See, e.g., U.S. Patent No. 6,626,858.
Glaucoma drainage devices, which may be combined with one or more anti-scarring agents according to the present invention, include commercially available products. For example, cylindrical tubes, such as the AQUAFLOvV Collagen Glaucoma Drainage Device (STAAR Surgical Company, Monrovia, CA) may be used in the practice of the present invention. Other examples of glaucoma drainage devices includes the Molteno Glaucoma Implant (Single Plate Molteno Implant, Pressure Ridge Single Plate Molteno Implant (D1), Microphthalmic Plate Molteno Implant (M1), Double Plate Molteno Implant (R2/L2), and Pressure Ridge Double Plate Molteno Implant (DR2/DL2) from Molteno Opthalmic Limited (New Zealand), BAERVELDT Glaucoma Implants (Models BG-101-350, BG-102- 350, BG-103-250; Pfizer, New York, NY), and the Ahmed Glaucoma Valve (Models FP7, S2, S3, PS2, PS3, B1 from New World Medical, Inc. (Rancho Cucamonga, CA).
In one aspect, the present invention provides a glaucoma drainage device that includes an anti-scarring agent or a composition that includes an anti-scarring agent. Numerous polymeric and non-polymeric delivery systems for use in glaucoma drainage devices have been described above. Methods for incorporating the fibrosis-inhibiting agent into or onto the device includes: (a) directly affixing to the device a fibrosis-inhibiting composition (e.g., by either a spraying process or dipping process as described above, with or without a carrier), (b) directly incorporating into the device a fibrosis-inhibiting composition (e.g., by either a spraying process or dipping process as described above, with or without a carrier), (c) by coating the device with a substance such as a hydrogel which will in turn absorb the fibrosis-inhibiting composition, (d) by interweaving fibrosis-inhibiting composition coated thread (or the polymer itself formed into a thread) into the device structure, (e) by inserting the device into a sleeve or mesh which is comprised of or coated with a fibrosis-inhibiting composition, (f) constructing the device itself or a portion of the device with a fibrosis- inhibiting composition, or (g) by covalently binding the fibrosis-inhibiting agent directly to the device surface or to a linker (small molecule or polymer) that is coated or attached to the device surface.
In addition to coating the device with the fibrosis-inhibiting composition, the fibrosis-inhibiting agent can be mixed with the materials that are used to make the device such that the fibrosis-inhibiting agent is incorporated into the final device.
In one embodiment, the methods above can be used to incorporate the fibrosis-inhibiting agent into or onto all or portions of the plate of the device. In another embodiment, the methods above can be used to incorporate the fibrosis-inhibiting agent into or onto all or portions of the tube of the device.
In yet another embodiment, the methods above can be used to incorporate the fibrosis-inhibiting agent into or onto all or potions of both the plate and the tube of the device.
In addition to incorporation of a fibrosis-inhibiting agent into or onto the device (e.g., as a coating), another biologically active agent can be incorporated into or onto the device, for example an anti-inflammatory (e.g., dexamethazone or aspirin) or a MMP inhibitor.
According to the present invention, any fibrosis-inhibiting agent described above can be utilized in the practice of this embodiment. Within one embodiment of the invention, glaucoma drainage devices may be adapted to release an agent that inhibits one or more of the four general components of the process of fibrosis (or scarring), including: formation of new blood vessels (angiogenesis), migration and proliferation of connective tissue cells (such as fibroblasts or smooth muscle cells), deposition of extracellular matrix (ECM), and remodeling (maturation and organization of the fibrous tissue). By inhibiting one or more of the components of fibrosis (or scarring), the overgrowth of granulation tissue may be inhibited or reduced.
As glaucoma drainage devices are made in a variety of configurations and sizes, the exact dose administered will vary with device size, surface area and design. However, certain principles can be applied in the application of this art. Drug dose can be calculated as a function of dose per unit area (of the portion of the device being coated), total dose administered, and appropriate surface concentrations of active drug όan be determined. Drugs are to be used at concentrations that range from several times more than to 10%, 5%, or even less than 1% of the concentration typically used in a single chemotherapeutic systemic dose application. Preferably, the drug is released in effective concentrations for a period ranging from 1 - 90 days.
Several examples of fibrosis-inhibiting agents for use in glaucoma drainage devices include the following: ZD-6474, AP-23573, synthadotin, S-0885, aplidine, ixabepilone, IDN-5390, SB-2723005, ABT- 518, combretastatin, anecortave acetate, SB-715992, temsirolimus, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, as well as analogues and derivatives of the aforementioned.
Additional examples of anti-scarring agents which can be used include those having a high potency in the assays described herein (1- 10OnM IC5O range) such as isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, dolastatin 15, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17- DMAG, and tacrolimus; those having a mid-potency in the assays described herein (100-500 nM IC50 range) such as loteprednol etabonate, juglone, prednisolone, puromycin, 3-BAABE, cladribine, and mannose-6-phosphate; and those having a low potency in the assays described herein (500- 1000nm range IC50 range) such as 5-azacytidine, Ly333531(ruboxistaurin), and simvastatin.
Regardless of the method of application of the drug to the devices, the exemplary anti-fibrosing agents, used alone or in combination, should be administered under the following dosing guidelines. The total amount (dose) of anti-scarring agent in or on the device may be in the range of about 0.01 μg-10 μg, or 10 μg-10 mg, or 10 mg-250 mg, or 250 mg-1000 mg, or 1000 mg-2500 mg. The dose (amount) of anti-scarring agent per unit area of device surface to which the agent is applied may be in the range of about 0.01 μg/mm2 - 1 μg/mm2, or 1 μg/mm2 - 10 μg/mm2, or 10 μg/mm2 - 250 μg/mm2, 250 μg/mm2 - 1000 μg/mm2, or 1000 μg/mm2 - 2500 μg/mm2.
Provided below are exemplary dosage ranges for various anti- scarring agents that can be used in conjunction with glaucoma drainage devices in accordance with the invention. (A) Angiogenesis inhibitors including Alphastatin, ZD-6474, IDN-5390, SB-2723005, ABT-518, combretastatin, and anecortane, analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10"8 - 10"4 M of agent is to be maintained on the implant or barrier surface. (B) mTOR inhibitors including AP-23573 and Temsirolimus, analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10 s- 10"4 M of agent is to be maintained on the implant or barrier surface. (C) Tubulin antagonists including synthadotin, analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10'8- 10"4 M of agent is to be maintained on the implant or barrier surface. (D) Epithilones including ixabepilone and analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10^- 10"4 M of agent is to be maintained on the implant or barrier surface. (E) Kinesin antagonists including SB- 715992 and analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10^- 10"4 M of agent is to be maintained on the implant or barrier surface. (F) TNF alpha antagonists including Etanercept, humicade, adalimumab and analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10"8- 10"4 M of agent is to be maintained on the implant or barrier surface. (G) AKT inhibitor including erucylphosphochoiine and analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10^- 10"4 M of agent is to be maintained on the implant or barrier surface. (H) FGF Inhibitors including IDN-5390 and analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10"8- 10"4 M of agent is to be maintained on the implant or barrier surface. (I) Collagenase Antagonists including S-0885 and analogues and derivatives thereof: total dose not to exceed 1000 mg (range of 0.1 μg to 1000 mg); preferred 1 μg to 500 mg. Dose per unit area of 0.01 μg - 500 μg per mm2; preferred dose of 0.1 μg/mm2 - 100 μg/mm2. Minimum concentration of 10"8- 10"4 M of agent is to be maintained on the implant or barrier surface. (J) NF KAPPA B Inhibitors including Bortezomib and analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10^- 10"4 M of agent is to be maintained on the implant or barrier surface. (K) Elongation factor-1 alpha inhibitors including aplidine and analogues and derivatives thereof: total dose not to exceed 1000 mg (range of 0.1 μg to 1000 mg); preferred 1 μg to 500 mg. Dose per unit area of 0.01 μg - 500 μg per mm2; preferred dose of 0.1 μg/mm2 - 100 μg/mm2. Minimum concentration of 10^- 10"4 M of agent is to be maintained on the implant or barrier surface. (L) Tyrosine kinase inhibitors including Gefitinib and analogues and derivatives thereof: total dose not to exceed 1000 mg (range of 0.1 μg to 1000 mg); preferred 1 μg to 500 mg. Dose per unit area of 0.01 μg - 500 μg per mm2; preferred dose of 0.1 μg/mm2 - 100 μg/mm2. Minimum concentration of 10^- 10"4 M of agent is to be maintained on the implant or barrier surface. For high potency drugs, the total dose typically should not exceed 200 mg (range of 0.1 μg to 200 mg) and preferably 1 μg to 100 mg; dose per unit area of 0.01 μg - 100 μg per mm2; preferably 0.1 μg/mm2 - 20 μg/mm2; and minimum concentration of 10"8- 10"4 M of agent should be maintained on the implant or barrier surface. For mid-potency drugs, the total dose typically should not to exceed 500 mg (range of 1.0 μg to 500 mg) and preferably 1 μg to 200 mg; dose per unit area of 0.01 μg - 200 μg per mm2, preferably 0.1 μg/mm2 - 40 μg/mm2; and minimum concentration of 10" 8- 10"4 M of agent should be maintained on the implant or barrier surface. For low potency drugs, the total dose typically should not exceed 1000 mg (range of 0.1 μg to 1000 mg), preferably 1 μg to 500 mg; dose per unit area of 0.01 μg - 500 μg per mm2; preferably 0.1 μg/mm2 - 100 μg/mm2; and minimum concentration of 10 s- 10'4 M of agent should to be maintained on the implant or barrier surface.
Prosthetic Heart Valves
The present invention provides for the combination of a drug and a prosthetic heart valve.
Prosthetic heart valves are devices that are used to replace natural heart valves that are defective, due to congenital malformations, infections, partial occlusion, or wearing. Prosthetic heart valves are typically composed of an occluder(s) attached to the occluder base, which is in turn attached to the suture ring that provides anchorage of the device to the heart tissue. The occluder base is annular and provides a passageway for blood flow. There may be one or more occluders which alternate in an opened and closed position to regulate the flow of blood. To secure the prosthetic heart valve to the heart tissue, a suture ring, typically composed of a knit fabric tube, is rolled into a toroidal form and is secured to the periphery of the occluder base of the prosthesis. Affixing the suture ring to the heart tissue typically occurs using sutures, sealants, adhesives, staples, or clamping with metal or polymer wires. Although the design of prosthetic heart valves has been gradually refined, complications continue to occur. Since the suture rings are often made out of synthetic material, thrombus, fibrosis and pannus often occur around the prosthetic heart valve. This scar formation often hinders the function of the valve and over time may require a second surgical procedure and replacement. Suture rings are generally composed of synthetic polymer, including, but not limited to, polyester (e.g., DACRON), polytetrafluoroethylene (e.g., TEFLON), silicone, and polypropylene. Suture rings are often made of a filler material with a woven material stitched over the filler. The surface of the suture ring is often course due to the covering cloth material. This predisposes the suture ring to scarring formation early in the post-operative period with severe pannus/fibrosis developing over several months following implantation. The consequences of fibrosis encroachment onto a prosthetic heart valve can be drastic, and potentially catastrophic. For example, fibrosis may inhibit valve occluder function by limiting its ability to open and close properly. The fibrosis may extend from the suture ring to the leaflets. This fibrosis may fuse the leaflets at their commissure, distort individual leaflets, and/or stiffen leaflets such that they do not open or close properly. The end result of this fibrosis typically is a heart valve that is both stenotic and insufficient.
There are two main types of prosthetic heart valves, mechanical and bioprosthetic. Typically, both mechanical and bioprosthetic heart valves utilize a synthetic suture ring. They differ primarily in the type of occluder that is utilized. The occluders of the mechanical heart valve may be composed of a ball and cage assembly, single leaflet disk valves, or bileaflet disk valves. The occluders of the bioprosthetic heart valve are composed of animal or human tissue that mimic the appearance and function of the natural heart valve it is replacing. The bioprosthetic heart valve leaflets are usually composed of chemically treated tissue. The harvested valves are fixed in glutaraldehyde or similar fixatives in order to make them suitable for human implantation. In one aspect, the prosthetic heart valve may be a mechanical prosthesis which is typically composed of rigid leaflets formed of a biocompatible substance (e.g., pyrolitic carbon, titanium or DACRON). Mechanical prosthetic heart valves may be a ball and cage assembly, bileaflet, trileaflet or tilting disks. The most common is the bileaflet type since the hemodynamics of this valve is better as blood flow is smoother and less turbulent. For example, the mechanical prosthesis may be composed of a base with an external suture ring and an internal rim for blood flow as well as at least two closing leaflets. See, e.g., U.S. Patent No. 6,068,657. The mechanical prosthesis may be composed of annular valve housing with a center orifice and first and second valve leaflets pivotally mounted to the valve housing. See, e.g., U.S. Patent Nos. 4,808,180 and 5,026,391. The mechanical prosthesis may be designed with an annular body with at least one leaflet pivotally mounted such that it is movable between an open and closed position by a magnet that exerts a force on the leaflet at a defined pressure. See, e.g., U.S. Patent No. 6,638,303. The mechanical prosthesis may have an annular body with a plurality of hinges which form an entrance ramp and supports at least one leaflet to the valve body. See, e.g., U.S. Patent Nos. 6,645,244 and 5,919,226. The mechanical prosthesis may be composed of a supporting flexible, cylindrical frame with a cover that forms a cusp supporting stent for the valve trileaflet apparatus and a sewing ring as an attachment surface. See, e.g., U.S. Patent No. 5,258,023. The mechanical prosthesis may have an increased valve lumen composed of a single piece valve orifice housing with at least one movable occluder coupled to the housing and a suture cuff for attaching the housing to the heart tissue. See, e.g., U.S. Patent Nos. 6,007,577 and 6,391,053. The mechanical prosthesis may be composed of a sewing ring and a removable valve assembly which slides in a central core of the sewing ring. See, e.g., U.S. Patent No. 5,032,128. The mechanical prosthesis may be a highly flexible cylindrical stent composed of a plurality of separate adjacent stent members with alternating cusps and commissures that are able to move radially and support a plurality of flexible leaflets. See, e.g., U.S. Patent Nos. 6,558,418 and 6,338,740. Other mechanical heart valve prostheses are described in, e.g., U.S. Patent Nos. 6,395,025; 6,358,278; 6,176,877; 6,139,575 and 5,984,958.
In another aspect, the prosthetic heart valve may be a bioprosthetic device which typically is flexible leaflets formed of a biological material (e.g., porcine valves or bovine pericardial valves). Tissue valves may be supported with a stent frame that provides the leaflets with more structure and durability. Stentless tissue valves may also be implanted by harvesting the porcine valves with the pig's aorta still attached. For example, the bioprosthetic heart valve, which may be obtained from a donor (e.g., porcine), may be treated to reduce antigens to prevent inflammatory response upon transplantation. See, e.g., U.S. Patent No. 6,592,618. The bioprosthetic heart valve may be composed of a biological tissue material disposed around a mechanical annular support to provide at least part of the sewing ring. See, e.g., U.S. Patent No. 6,582,464. The bioprosthetic heart- valve may be composed of a xenograft mitral valve (e.g., porcine) and a sewing tube and cover of flexible material which is attached to the mitral valve. See, e.g., U.S. Patent No. 5,662,704. The bioprosthetic heart valve may be composed of a natural tissue heart valve attached to a prosthetic stent frame that may be covered by a fabric cover. See, e.g., U.S. Patent Nos. 3,983,581; 4,035,849; 5,861,028; 6,350,282 and 6,585,766. The bioprosthetic heart valve may be a self-supporting stentless valve that may be composed of a tubular body of mammalian origin. See, e.g., U.S. Patent Nos. 5,156,621 and 6,342,070.
In another aspect, the prosthetic heart valve may be inserted into place using minimally-invasive techniques. For example, the prosthetic heart valve may be an expandable device adapted for delivery in a collapsed state to an implantation site and then expanded to a plurality of leaflets attached to a stent system. See, e.g., U.S. Patent No. 6,454,799. In another aspect, the device may be a component of the heart valve. For example, the device may be an implantable annular ring for receiving a prosthetic heart valve. See, e.g., U.S. Patent No. 6,106,550. The device may be a suture ring having an outer peripheral tapered thread for attaching a heart valve prosthesis. See, e.g., U.S. Patent No. 6,113,632. The device may be a suture ring for a mechanical heart valve composed of a stiffening ring attachment, a knit fabric sewing cuff and a locking ring. See, e.g., U.S. Patent No. 5,071,431.
Prosthetic heart valves and components thereof (e.g., annular suture rings), which may be combined with one or more drugs according to the present invention, include commercially available products, such as the Carpentier-Edwards PERIMOUNT (CEP) Pericardial Bioprosthesis, Carpentier-Edwards S.A.V. Aortic Bioprosthesis and Edwards PRIMA PLUS STENTLESS BIOPROSTHESIS from Edwards Lifesciences (Irvine, CA), the SJM REGENT Valve from St. Jude Medical (St. Paul, MN), and the MOSAIC Bioprosthetic Heart Valve from Medtronic (Minneapolis, MN).
In one aspect, the present invention provides prosthetic heart valve devices that include an anti-scarring agent or a composition that includes an anti-scarring agent. Numerous polymeric and non-polymeric delivery systems for use in prosthetic heart valves have been described above. Methods for incorporating the fibrosis-inhibiting agent into or onto the device includes: (a) directly affixing to the device a fibrosis-inhibiting composition (e.g., by either a spraying process or dipping process as described above, with or without a carrier), (b) directly incorporating into the device a fibrosis-inhibiting composition (e.g., by either a spraying process or dipping process as described above, with or without a carrier (c) by coating the device with a substance such as a hydrogel which will in turn absorb the fibrosis-inhibiting composition, (d) by interweaving fibrosis-inhibiting composition coated thread (or the polymer itself formed into a thread) into the device structure, (e) constructing the device itself or a portion of the device with a fibrosis-inhibiting composition, or (f) by covalently binding the fibrosis-inhibiting agent directly to the device surface or to a linker (small molecule or polymer) that is coated or attached to the device surface, and/or (g) any combination of the aforementioned.
According to the present invention, any fibrosis-inhibiting agent described above can be utilized in the practice of this embodiment. Within one embodiment of the invention, prosthetic heart valves may be adapted to release an agent that inhibits one or more of the four general components of the process of fibrosis (or scarring), including: formation of new blood vessels (angiogenesis), migration and proliferation of connective tissue cells (such as fibroblasts or smooth muscle cells), deposition of extracellular matrix (ECM), and remodeling (maturation and organization of the fibrous tissue). By inhibiting one or more of the components of fibrosis (or scarring), the overgrowth of granulation tissue may be inhibited or reduced.
As prosthetic heart valve devices are made in a variety of configurations and sizes, the exact dose administered will vary with device size, surface area and design. However, certain principles can be applied-in the application of this art. Drug dose can be calculated as a function of dose per unit area (of the portion of the device being coated), total dose administered, and appropriate surface concentrations of active drug can be determined. Drugs are to be used at concentrations that range from several times more than to 10%, 5%, or even less than 1% of the concentration typically used in a single chemotherapeutic systemic dose application. Preferably, the drug is released in effective concentrations for a period ranging from 1 - 90 days.
Several examples offibrosis-inhibiting agents for use in prosthetic heart valves include the following: ZD-6474, AP-23573, synthadotin, S-0885, aplidine, ixabepilone, IDN-5390, SB-2723005, ABT- 518, combretastatin, anecortave acetate, SB-715992, temsirolimus, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, as well as analogues and derivatives of the aforementioned. Additional examples of anti-scarring agents which can be used include those having a high potency in the assays described herein (1- 10OnM IC50 range) such as isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, dolastatin 15, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17- DMAG, and tacrolimus; those having a mid-potency in the assays described herein (100-500 nM IC50 range) such as loteprednol etabonate, juglone, prednisolone, puromycin, 3-BAABE, cladribine, and mannose-6-phosphate; and those having a low potency in the assays described herein (500- 1000nm range IC50 range) such as 5-azacytidine, Ly333531(ruboxistaurin), and simvastatin.
Regardless of the method of application of the drug to the prosthetic heart valve, the exemplary anti-fibrosing agents, used alone or in combination, should be administered under the following dosing guidelines. The total amount (dose) of anti-scarring agent in or on the prosthetic heart valve may be in the range of about 0.01 μg-10 μg, or 10 μg-10 mg, or 10 mg-250 mg, or 250 mg-1000 mg, or 1000 mg-2500 mg. The dose (amount) of anti-scarring agent per unit area of device surface to which the agent is applied may be in the range of about 0.01 μg/mm2 - 1 μg/mm2, or 1 μg/mm2 - 10 μg/mm2, or 10 μg/mm2 - 250 μg/mm2, 250 μg/mm2 - 1000 μg/mm2, or 1000 μg/mm2 - 2500 μg/mm2.
Provided below are exemplary dosage ranges for various anti- scarring agents that can be used in conjunction with prosthetic heart valve devices in accordance with the invention. (A) Angiogenesis inhibitors including Alphastatin, ZD-6474, IDN-5390, SB-2723005, ABT-518, combretastatin, and anecortane, analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10"8- 10"4 M of agent is to be maintained on the implant or barrier surface. (B) mTOR inhibitors including AP-23573 and Temsirolimus, analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10"8- 10"4 M of agent is to be maintained on the implant or barrier surface. (C) Tubulin antagonists including synthadotin, analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10"8- 10"4 M of agent is to be maintained on the implant or barrier surface. (D) Epithilones including ixabepilone and analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10'8 - 10"4 M of agent is to be maintained on the implant or barrier surface. (E) Kinesin antagonists including SB- 715992 and analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg; Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10^- 10"4 M of agent is to be maintained on the implant or barrier surface. (F) TNF alpha antagonists including Etanercept, humicade, adalimumab and analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10^- 10"4 M of agent is to be maintained on the implant or barrier surface. (G) AKT inhibitor including erucylphosphocholine and analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10^- 10"4 M of agent is to be maintained on the implant or barrier surface. (H) FGF Inhibitors including IDN-5390 and analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10^- 10"4 M of agent is to be maintained on the implant or barrier surface. (I) Collagenase Antagonists including S-0885 and analogues and derivatives thereof: total dose not to exceed 1000 mg (range of 0.1 μg to 1000 mg); preferred 1 μg to 500 mg. Dose per unit area of 0.01 μg - 500 μg per mm2; preferred dose of 0.1 μg/mm2 - 100 μg/mm2. Minimum concentration of 10~8- 10"4 M of agent is to be maintained on the implant or barrier surface. (J) NF KAPPA B Inhibitors including Bortezomib and analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10'8- 10"4 M of agent is to be maintained on the implant or barrier surface. (K) Elongation factor-1 alpha inhibitors including aplidine and analogues and derivatives thereof: total dose not to exceed 1000 mg (range of 0.1 μg to 1000 mg); preferred 1 μg to 500 mg. Dose per unit area of 0.01 μg - 500 μg per mm2; preferred dose of 0.1 μg/mm2 - 100 μg/mm2. Minimum concentration of 10"8- 10~4 M of agent is to be maintained on the implant or barrier surface. (L) Tyrosine kinase inhibitors including Gefitinib and analogues and derivatives thereof: total dose not to exceed 1000 mg (range of 0.1 μg to 1000 mg); preferred 1 μg to 500 mg. Dose per unit area of 0.01 μg - 500 μg per mm2; preferred dose of 0.1 μg/mm2 - 100 μg/mm2. Minimum concentration of 10^- 10"4 M of agent is to be maintained on the implant or barrier surface.
For high potency drugs, the total dose typically should not exceed 200 mg (range of 0.1 μg to 200 mg) and preferably 1 μg to 100 mg; dose per unit area of 0.01 μg - 100 μg per mm2; preferably 0.1 μg/mm2 - 20 μg/mm2; and minimum concentration of 10"8- 10"4 M of agent should be maintained on the implant or barrier surface. For mid-potency drugs, the total dose typically should not to exceed 500 mg (range of 1.0 μg to 500 mg) and preferably 1 μg to 200 mg; dose per unit area of 0.01 μg - 200 μg per mm2, preferably 0.1 μg/mm2 - 40 μg/mm2; and minimum concentration of 10" 8- 10"4 M of agent should be maintained on the implant or barrier surface. For low potency drugs, the total dose typically should not exceed 1000 mg (range of 0.1 μg to 1000 mg), preferably 1 μg to 500 mg; dose per unit area of 0.01 μg - 500 μg per mm2; preferably 0.1 μg/mm2 - 100 μg/mm2; and minimum concentration of 10"8- 10"4 M of agent should to be maintained on the implant or barrier surface.
Penile Implants
In one aspect, the present invention provides for the combination of an anti-scarring agent and a penile implant device. In one aspect, penile implants are loaded with an anti-scarring drug or a composition that includes an anti-scarring drug to prevent fibrous encapsulation.
Penile implants are used to treat erectile dysfunction and are generally flexible rods, hinged rods or inflatable devices with a pump. Penile implants may be composed of rods, coils, inflatable tubes and/or - pressure chambers and may be used to provide erectile function, enlargement or provide shape to a misshapen or damaged penis. For example, the penile implant may be an implantable polymeric material which is injected into the lamina propria mucosae of the glans in order to enlarge the glans of the male genital organ. See, e.g., U.S. Patent No. 6,418,934. The penile implant may be composed of a pair of arced, elongated portions made of silicone rubber that are mirror images of each other, which has a varying circumferential wall thickness. See, e.g., U.S. Patent No. 6,537,204. The penile implant may be used to increase penile volume by being adapted to cover the outer lateral sides of the corpus cavernosum without covering the upper and lower sides thereof. See, e.g., U.S. Patent No. 6,015,380. The penile implant may be an inflatable, self-contained implant composed of a cylindrical body having a pump that transfers fluid from a reservoir to a pressure chamber that has a pressure relief valve. See, e.g., U.S. Patent Nos. 4,898,158 and 4,823,779. The penile implant may be composed of an elongated rod having a relatively short proximal stem portion, which is covered by a layer of hydrophilic material that contains a plurality of openings and swells as it absorbs water. See, e.g., U.S. Patent No. 4,611 ,584. The penile implant may be composed of at least one inflatable tube that has fluid interchange with a mounting base which is controlled by a manual pump implanted in the scrotum. See, e.g., U.S. Patent No. 6,475,137. The penile implant may be a flexible double-walled partial cylindrical sleeve that has bellow-like construction which is suited for penile malformation. See, e.g., U.S. Patent No. 5,669,870. The penile implant may be used for correcting erectile impotence by being composed of at least one flexible portion with a pressure chamber connected by tubing to an accumulator charged with fluid, such that pressurizing fluid flows when the valve is opened. See, e.g., U.S. Patent No. 4,917,110. The penile implant may be composed of a stainless steel pad supported by a plurality of strands which is surrounded by a cylinder with a silicone ring that can move longitudinally in response to the expansion or shrinkage of the penis. See,- e.g., U.S. Patent No. 5,433,694. The penile implant may increase girth and length by being composed of a cylindrical sleeve that has an elastic outer sheet and an inner inelastic sheet that forms a closed sack to receive a fluid under pressure from a fluid source. See, e.g., U.S. Patent No. 5,445,594. The penile implant may be composed of a braided sleeve with an outer elastomeric surface and inner surface having grooves and ribs in a helical arrangement, such that the implant is malleable having both a bendable configuration and an unbent rigid configuration. See, e.g., U.S. Patent No. 5,512,033. The penile implant may be a polymeric matrix having dissociated cartilage-forming cells deposited on and in said matrix whereby a cartilaginous structure is formed upon implantation having controlled biomechanical properties and tensile strength. See, e.g., U.S. Patent No. 6,547,719. The penile implant may be composed of an implantable supply pump, deformable reservoir, and conducting/dispensing catheters, such that a vasodilator agent is delivered to the erectile bodies to treat male impotence. See, e.g., U.S. Patent No. 6,679,832. Other penile implants are described in, e.g., U.S. Patent Nos. 6,579,230; 5,704,895; 5,250,020; 5,048,510 and 4,875,472.
A fibrosis-inhibiting agent may be incorporated into, onto or near the device. Penile implants, which may be combined with one or more agents according to the present invention, include commercially available products, such as, for example, the TITAN Inflatable Penile Prosthesis from Mentor Corporation (Santa Barbara, CA) and the AMS penile prosthesis product line including the AMS 700 CX CXM, AMS AMBICOR, and AMS Malleable 600M Penile Prostheses from American Medical Systems, Inc. (Minnetonka, MN),
In one aspect, the present invention provides penile implant devices that include an anti-scarring agent or a composition that includes an anti-scarring agent. Numerous polymeric and non-polymeric delivery systems for use in penile implants have been described above. Methods for incorporating the fibrosis-inhibiting agent into or onto the device includes: (a) directly affixing to the device a fibrosis-inhibiting composition (e.g., by either a spraying process or dipping process as described above, with or without a carrier), (b) directly incorporating into the device a fibrosis- inhibiting composition (e.g., by either a spraying process or dipping process as described above, with or without a carrier), (c) by coating the device with a substance such as a hydrogel which will in turn absorb the fibrosis- inhibiting composition, (d) by interweaving fibrosis-inhibiting composition coated thread (or the polymer itself formed into a thread) into the device structure, (e) constructing the device itself or a portion of the device with a fibrosis-inhibiting composition, or (f) by covalently binding the fibrosis- inhibiting agent directly to the device surface or to a linker (small molecule or polymer) that is coated or attached to the device surface. The coatings can be applied to different portions of the device. For example, the coating can be (a) a coating applied to the external surface of the portion of the penile implant that is implanted into the penis; (b) a coating applied to the external surfaces of the portions of the penile implant that are implanted in the scrotum, or (c) a coating applied to all or parts of the surfaces of the entire device.
In addition to coating the device with the fibrosis-inhibiting composition, the fibrosis-inhibiting agent can be mixed with the materials that are used to make the device such that the fibrosis-inhibiting agent is incorporated into the final device.
In addition to incorporation of a fibrosis-inhibiting agent into or onto the device, another biologically active agent can be incorporated into or onto the device, for example an anti-inflammatory (e.g., dexamethazone or aspirin).
In another aspect, the device may further comprise an antibiotic or a combination of an antibiotic and an anti-inflammatory agent in order to combat infection associated with implantation of penile implants.
The placement of penile implants can be complicated by infection (usually in the first 6 months after surgery) with Coagulase Negative Staphylococci (including Staphylococcus epidermidis), Staphylococcus aureus, Pseudomonas aeruginosa, Enterococci, Serratia and Candida. Infection is characterized by fever, erythema, induration and purulent drainage from the operative site. The usual route of infection is through the incision at the time of surgery and up to 3% of penile implants become infected despite the best sterile surgical technique. To help combat this, intraoperative irrigation with antibiotic solutions is often employed.
Drug-coating of, or drug incorporation into, the penile implant can allow bacteriocidal drug levels to be achieved locally, thus reducing the incidence of bacterial colonization (and subsequent development of local infection and device failure), while producing negligible systemic exposure to the drugs.
Representative examples of antibiotics include amoxicillin, trimethoprim-sulfamethoxazole, azithromycin, clarithromycin, amoxicillin- clavulanate, cefprozil, cefuroxime, cefpodoxime, or cefdinir). Other examples of anti-infective compounds include doxorubicin, mitoxantrone, 5-fluorouracil and etoposide.
Utilizing the fluoropyrimidine, 5-fluorouracil, as an example, whether applied as a polymer coating, incorporated into the polymers which make up the implant, or applied without a carrier polymer, the total dose of 5-fluorouracil applied should not exceed 250 mg (range of 1.0 μg to 250 mg). In a particularly preferred embodiment, the total amount of drug applied should be in the range of 10 μg to 25 mg. The dose per unit area (i.e., the amount of drug as a function of the surface area of the portion of the implant to which drug is applied and/or incorporated) should fall within the range of 0.1 μg - 1 mg per mm2 of surface area. In a particularly preferred embodiment, 5-fluorouracil should be applied to the implant surface at a dose of 1.0 μg/mm2 - 50 μg/mm2. As different polymer and non-polymer coatings will release 5-fluorouracil at differing rates, the above dosing parameters should be utilized in combination with the release rate of the drug from the implant surface such that a minimum concentration of 10"4 - 10"7 M of 5-fluorouracil is maintained. It is necessary to insure that surface drug concentrations exceed concentrations of 5-fluorouracil known to be lethal to numerous species of bacteria and fungi (i.e., are in excess of 10"4 M; although for some embodiments lower drug levels will be sufficient). In a preferred embodiment, 5-fluorouracil is released from the implant surface such that anti-infective activity is maintained for a period ranging from several hours to several months. In a particularly preferred embodiment the drug is released in effective concentrations for a period ranging from 1 week - 6 months. It should be readily evident based upon the discussions provided herein that analogues and derivatives of 5-fluorouracil (as described previously) with similar functional activity can be utilized for the purposes of this invention; the above dosing parameters are then adjusted according to the relative potency of the analogue or derivative as compared to the parent compound (e.g., a compound twice as potent as 5-fluorouracil is administered at half the above parameters, a compound half as potent as 5-fluorouracil is administered at twice the above parameters, etc.).
Anti-inflammatory and anti-infective agents may be formulated, for example, into a coating applied to the surface of the penile implant. The drug(s) can be applied in several manners: (a) as a coating applied to the external surface of the penile implant; and/or (b) incorporated into the polymers which comprise the penile implant.
According to the present invention, any fibrosis-inhibiting agent described above can be utilized in the practice of this embodiment. Within one embodiment of the invention, penile implants may be adapted to release an agent that inhibits one or more of the four general components of the process of fibrosis (or scarring), including: formation of new blood vessels (angiogenesis), migration and proliferation of connective tissue cells (such as fibroblasts or smooth muscle cells), deposition of extracellular matrix (ECM), and remodeling (maturation and organization of the fibrous tissue). By inhibiting one or more of the components of fibrosis (or scarring), the overgrowth of granulation tissue may be inhibited or reduced.
As penile implant devices are made in a variety of configurations and sizes, the exact dose administered will vary with device size, surface area and design. However, certain principles can be applied in the application of this art. Drug dose can be calculated as a function of dose per unit area (of the portion of the device being coated), total dose administered, and appropriate surface concentrations of active drug can be determined. Drugs are to be used at concentrations that range from several times more than to 10%, 5%, or even less than 1% of the concentration typically used in a single chemotherapeutic systemic dose application. Preferably, the drug is released in effective concentrations for a period ranging from 1 - 90 days.
Several examples of fibrosis-inhibiting agents for use in penile implants include the following: ZD-6474, AP-23573, synthadotin, S-0885, aplidine, ixabepilone, IDN-5390, SB-2723005, ABT-518, combretastatin, anecortave acetate, SB-715992, temsirolimus, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, as well as analogues and derivatives of the aforementioned.
Additional examples of anti-scarring agents which can be used include those having a high potency in the assays described herein (1- 10OnM IC50 range) such as isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, dolastatin 15, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17- DMAG, and tacrolimus; those having a mid-potency in the assays described herein (100-500 nM IC50 range) such as loteprednol etabonate, juglone, prednisolone, puromycin, 3-BAABE, cladribine, and mannose-6-phosphate; and those having a low potency in the assays described herein (500- 1000nm range IC50 range) such as 5-azacytidine, Ly333531(ruboxistaurin), and simvastatin.
Regardless of the method of application of the drug to the penile implant, the exemplary anti-fibrosing agents, used alone or in combination, should be administered under the following dosing guidelines. The total amount (dose) of anti-scarring agent in or on the penile implant may be in the range of about 0.01 μg-10 μg, or 10 μg-10 mg, or 10 mg-250 mg, or 250 mg-1000 mg, or 1000 mg-2500 mg. The dose (amount) of anti- scarring agent per unit area of device surface to which the agent is applied may be in the range of about 0.01 μg/mm2 - 1 μg/mm2, or 1 μg/mm2 - 10 μg/mm2, or 10 μg/mm2 - 250 μg/mm2, 250 μg/mm2 - 1000 μg/mm2, or 1000 μg/mm2 - 2500 μg/mm2.
Provided below are exemplary dosage ranges for various anti- scarring agents that can be used in conjunction with penile implant devices in accordance with the invention. (A) Angiogenesis inhibitors including Alphastatin, ZD-6474, IDN-5390, SB-2723005, ABT-518, combretastatin, and anecortane, analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10"8 - 10"4 M of agent is to be maintained on the implant or barrier surface. (B) mTOR inhibitors including AP-23573 and Temsirolimus, analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10'8- 10'4 M of agent is to be maintained on the implant or barrier surface. (C) Tubulin antagonists including synthadotin, analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10"8- 10"4 M of agent is to be maintained on the implant or barrier surface. (D) Epithilones including ixabepilone and analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg.to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10^- 10"4 M of agent is to be maintained on the implant or barrier surface. (E) Kinesin antagonists including SB- 715992 and analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10"8- 10"4 M of agent is to be maintained on the implant or barrier surface. (F) TNF alpha antagonists including Etanercept, humicade, adalimumab and analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10'8- 10"4 M of agent is to be maintained on the implant or barrier surface. (G) AKT inhibitor including erucylphosphocholine and analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10^- 10"4 M of agent is to be maintained on the implant or barrier surface. (H) FGF Inhibitors including IDN-5390 and analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10"8 - 10"4 M of agent is to be maintained on the implant or barrier surface. (I) Collagenase Antagonists including S-0885 and analogues and derivatives thereof: total dose not to exceed 1000 mg (range of 0.1 μg to 1000 mg); preferred 1 μg to 500 mg. Dose per unit area of 0.01 μg - 500 μg per mm2; preferred dose of 0.1 μg/mm2 - 100 μg/mm2. Minimum concentration of 10^- 10"4 M of agent is to be maintained on the implant or barrier surface. (J) NF KAPPA B Inhibitors including Bortezomib and analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10~8- 10"4 M of agent is to be maintained on the implant or barrier surface. (K) Elongation factor-1- alpha inhibitors including aplidine and analogues and derivatives thereof: total dose not to exceed 1000 mg (range of 0.1 μg to 1000 mg); preferred 1 μg to 500 mg. Dose per unit area of 0.01 μg - 500 μg per mm2; preferred dose of 0.1 μg/mm2 - 100 μg/mm2. Minimum concentration of 10"8- 10"4 M of agent is to be maintained on the implant or barrier surface. (L) Tyrosine kinase inhibitors including Gefitinib and analogues and derivatives thereof: total dose not to exceed 1000 mg (range of 0.1 μg to 1000 mg); preferred 1 μg to 500 mg. Dose per unit area of 0.01 μg - 500 μg per mm2; preferred dose of 0.1 μg/mm2 - 100 μg/mm2. Minimum concentration of 10"8 - 10'4 M of agent is to be maintained on the implant or barrier surface.
For high potency drugs, the total dose typically should not exceed 200 mg (range of 0.1 μg to 200 mg) and preferably 1 μg to 100 mg; dose per unit area of 0.01 μg - 100 μg per mm2; preferably 0.1 μg/mm2 - 20 μg/mm2; and minimum concentration of 10^- 10"4 M of agent should be maintained on the implant or barrier surface. For mid-potency drugs, the total dose typically should not to exceed 500 mg (range of 1.0 μg to 500 mg) and preferably 1 μg to 200 mg; dose per unit area of 0.01 μg - 200 μg per mm2, preferably 0.1 μg/mm2 - 40 μg/mm2; and minimum concentration of 10" 8- 10"4 M of agent should be maintained on the implant or barrier surface. For low potency drugs, the total dose typically should not exceed 1000 mg (range of 0.1 μg to 1000 mg), preferably 1 μg to 500 mg; dose per unit area of 0.01 μg - 500 μg per mm2; preferably 0.1 μg/mm2 - 100 μg/mm2; and minimum concentration of 10"8 ~ 10"4 M of agent should to be maintained on the implant or barrier surface.
Endotracheal and Tracheostomy Tubes
In one aspect, the present invention provides for the combination of an anti-scarring agent and endotracheal and tracheostomy tube devices. Association of an anti-scarring agent with an endotracheal or a tracheostomy tube (e.g., chest tube) may be used to prevent stenosis of the artificial airway.
Endotracheal tubes and tracheostomy tubes are used to maintain the airway when ventilatory assistance is required. Endotracheal tubes tend to be used to establish an airway in the acute setting, while tracheostomy tubes are used when prolonged ventilation is required or when there is a fixed obstruction in the upper airway.
In one aspect, endotracheal tubes may be used to provide a mechanical air passageway, which may be required for ventilation of the lungs during injury or surgery. Endotracheal tubes may have a single lumen or double lumen, and may have a flange or balloon for engaging its position within the trachea. For example, the endotracheal tube may be composed of an inner and outer flexible tube having a radially extending flange that prevents advancement beyond the larynx. See, e.g., U.S. Patent No. 5,259,371. The endotracheal tube may have a double lumen which is removably affixed whereby the first tubular lumen may be removed from the airway while the second tubular lumen remains intact. See, e.g., U.S. Patent No. 6,443,156. The endotracheal tube may have a tracheal portion and a bronchial portion attached at an angle that forms a single lumen, whereby when a balloon that is positioned within the tube is inflated, it blocks the flow of gas through the bronchial portion. See, e.g., U.S. Patent No. 6,609,521. The endotracheal tube may be composed of two cylindrical portions of different diameters which are connected by a non-circularly shaped tapered portion to complement the glottis which has a plurality of sealing gills that are thin and pliable that extends from the tapered portion. See, e.g., U.S. Patent No. 5,429,127. The endotracheal tube may be composed of a tubular portion with a visual indicator to provide guidance of the rotational orientation of the beveled tip at the distal end as it is advanced along the airway. See, e.g., U.S. Patent No. 6,568,393. The endotracheal tube may be composed of a light reflective coated bore to enhance image transmission and a flexible plurality of passages, one adapted to receive a fiber optic bundle, another connected to an inflatable cuff, and another adapted to receive a malleable stylette to aid in insertion and removal. See, e.g., U.S. Patent No. 6,629,924. The endotracheal tube may be composed of a hollow, flexible, cylindrical tube having an annular flange at its tip and a connector with an annular internal ridge that is concentrically mounted upon the outer proximal surface of the tube portion. See, e.g., U.S. Patent No. 5,251 ,617. The endotracheal tube may be composed of a main tube with an inflatable cuff for sealing, which has a double lumen for irrigation and suction for removal of secretions that may pool in the trachea. See, e.g., U.S. Patent No. 5,143,062. Other endotracheal tubes are described in, e.g., U.S. Patent Nos. 6,321,749; 5,765,559; 5,353,787; 5,291 ,882 and 4,977,894.
Tracheostomy tubes can be used to provide a bypass supply of air when the throat is obstructed. Tracheostomy tubes are used with an obturator for percutaneous insertion into a trachea through a stoma in the neck between adjacent cartilages to assist breathing. For example, the tracheostomy tube may be a tubular cannula formed of soft flexible plastic material which has a tapered distal end that is beveled, narrow, angled and curved downwardly for positioning within the trachea. See, e.g., U.S. Patent No. 5,058,580. The tracheostomy tube may be composed of a tube with a removable fitting mounted on the exposed end which may be sealed to the tube. See, e.g., U.S. Patent No. 5,606,966. The tracheostomy tube may be composed of an arcuate cannula with a flange that extends laterally outward and a rotatable tubular elbow that has a fluid connection with the cannula. See, e.g., U.S. Patent Nos. 5,259,376 and 5,054,482. The tracheostomy tube may be composed of two airways with a pneumatic vibrator that generates sonic vibrations to permit audible speech. See, e.g., U.S. Patent No. 4,773,412. The tracheostomy tube may be composed of an inner cannula removably received within an outer cannula with a sealing cuff between the outer cannula and the trachea to substantially prevent air from escaping from the trachea and to allow phonation through a secondary passageway formed between the inner and outer cannula. See, e.g., U.S. Patent No. 4,573,460. The tracheostomy tube may be composed of a first port for orienting outside the neck of the wearer, a second port for orienting within the trachea, and a third connecting port to provide and control gas flow via a valve. See, e.g., U.S. Patent No. 5,957,978. The tracheostomy tube may be composed of a hollow tube, an inflatable balloon having orthogonal projections, and a flange that provides an anchor external to the throat. See, e.g., U.S. Patent No. 6,612,305. The tracheostomy tube may be composed of a highly flexible material having wire reinforcement and a neck plate with a collar portion that may slide along the tube. See, e.g., U.S. Patent No. 5,443,064. Other tracheostomy tubes are described in, e.g., U.S. Patent Nos. 6,662,804; 6,135,110 and 5,983,895.
Endotracheal tubes, which may be combined with one or more agents according to the present invention, include commercially available products, such as the HI-LO Tracheal Tubes, LASER-FLEX Tracheal Tubes, and ENDOTROL Tracheal Tubes from Nellcor Puritan Bennett Inc. (Pleasanton, CA), the SHERIDAN Endotracheal Tubes from Hudson RCI (Temecula, CA)1 and the BARD Endotracheal Tube, Cuffed from CR. Bard, Inc. (Murray Hill, NJ).
Tracheostomy tubes, which may be combined with one or more agents according to the present invention, include commercially available products, such as the SHILEY TRACHEOSOFT XLT Tracheostomy Tubes, PHONATE Speaking Valves, and Reusable Cannula Cuffless Tracheostomy Tubes from Nellcor Puritan Bennett Inc. (Pleasanton, CA), the PER-FIT Percutaneous Dilational Tracheostomy Kits, PORTEX BLUE LINE Cuffed Tracheostomy Tubes, and BIVONA Uncuffed Tracheostomy Tubes from Portex, Inc. (Keene, NH), and the CRYSTALCLEAR Tracheostomy Tubes from Rusch (Germany).
In one aspect, the present invention provides endotracheal and tracheostomy tube devices that include an anti-scarring agent or a composition that includes an anti-scarring agent. Numerous polymeric and non-polymeric delivery systems for use in endotracheal and tracheostomy devices have been described above. Methods for incorporating the fibrosis- inhibiting agent into or onto the device includes: (a) directly affixing to the device a fibrosis-inhibiting composition (e.g., by either a spraying process or dipping process as described above, with or without a carrier), (b) directly incorporating into the device a fibrosis-inhibiting composition (e.g., by either a spraying process or dipping process as described above, with or without a carrier), (c) by coating the device with a substance such as a hydrogel which will in turn absorb the fibrosis-inhibiting composition, (d) by interweaving fibrosis-inhibiting composition coated thread (or the polymer itself formed into a thread) into the device structure, (e) constructing the device itself or a portion of the device with a fibrosis-inhibiting composition, or (f) by covalently binding the fibrosis-inhibiting agent directly to the device surface or to a linker (small molecule or polymer) that is coated or attached to the device surface. The coatings can be applied to different portions of the device. For example, the coating can be (a) as a coating applied to the internal (luminal) surface of the endotracheal tube or tracheostomy tube; (b) as a coating applied to the external surface of the endotracheal tube or tracheostomy tube; or (c) as a coating applied to all or parts of both surfaces.
The fibrosis-inhibiting agent can be mixed with the materials that are used to make the device such that the fibrosis-inhibiting agent is incorporated into the final device.
In addition to incorporation of a fibrosis-inhibiting agent into or onto the device, another biologically active agent can be incorporated into or onto the device, for example an anti-inflammatory (e.g., dexamethazone or aspirin) and/or an antibiotic (e.g., amoxicillin, trimethoprim- sulfamethoxazole, azithromycin, clarithromycin, amoxicillin-clavulanate, cefprozil, cefuroxime, cefpodoxime, or cefdinir).
According to the present invention, any fibrosis-inhibiting agent described above can be utilized in the practice of this embodiment. Within one embodiment of the invention, endotracheal and tracheostomy devices may be adapted to release an agent that inhibits one ormore of the four general components of the process of fibrosis (or scarring), including: formation of new blood vessels (angiogenesis), migration and proliferation of connective tissue cells (such as fibroblasts or smooth muscle cells), deposition of extracellular matrix (ECM), and remodeling (maturation and organization of the fibrous tissue). By inhibiting one or more of the components of fibrosis (or scarring), the overgrowth of granulation tissue may be inhibited or reduced.
As endotracheal and tracheostomy tube devices are made in a variety of configurations and sizes, the exact dose administered will vary with device size, surface area and design. However, certain principles can be applied in the application of this art. Drug dose can be calculated as a function of dose per unit area (of the portion of the device being coated), total dose administered, and appropriate surface concentrations of active drug can be determined. Drugs are to be used at concentrations that range from several times more than to 10%, 5%, or even less than 1% of the concentration typically used in a single chemotherapeutic systemic dose application. Preferably, the drug is released in effective concentrations for a period ranging from 1 - 90 days.
Several examples of fibrosis-inhibiting agents for use in endotracheal and tracheostomy tube devices include the following: ZD- 6474, AP-23573, synthadotin, S-0885, aplidine, ixabepilone, IDN-5390, SB- 2723005, ABT-518, combretastatin, anecortave acetate, SB-715992, temsirolimus, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, as well as analogues and derivatives of the aforementioned.
Additional examples of anti-scarring agents which can be used include those having a high potency in the assays described herein (1- 10OnM IC50 range) such as isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, dolastatin 15, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17- DMAG, and tacrolimus; those having a mid-potency in the assays described herein (100-500 nM IC5O range) such as loteprednol etabonate, juglone, prednisolone, puromycin, 3-BAABE, cladribine, and mannose-6-phosphate; and those having a low potency in the assays described herein (500- 1000nm range IC50 range) such as 5-azacytidine, Ly333531(ruboxistaurin), and simvastatin.
Regardless of the method of application of the drug to the device, the exemplary anti-fibrosing agents, used alone or in combination, should be administered under the following dosing guidelines. The total amount (dose) of anti-scarring agent in or on the device may be in the range of about 0.01 μg-10 μg, or 10 μg-10 mg, or 10 mg-250 mg, or 250 mg-1000 mg, or 1000 mg-2500 mg. The dose (amount) of anti-scarring agent per unit area of device surface to which the agent is applied may be in the range of about 0.01 μg/mm2 - 1 μg/mm2, or 1 μg/mm2 - 10 μg/mm2, or 10 μg/mm2 - 250 μg/mm2, 250 μg/mm2 - 1000 μg/mm2, or 1000 μg/mm2 - 2500 μg/mm2. Provided below are exemplary dosage ranges for various anti- scarring agents that can be used in conjunction with endotracheal and tracheostomy tube devices in accordance with the invention. (A) Angiogenesis inhibitors including Alphastatin, ZD-6474, IDN-5390, SB- 2723005, ABT-518, combretastatin, and anecortane, analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10^- 10"4 M of agent is to be maintained on the implant or barrier surface.
(B) mTOR inhibitors including AP-23573 and Temsirolimus, analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10^- 10"4 M of agent is to be maintained on the implant or barrier surface.
(C) Tubulin antagonists including synthadotin, analogues and derivatives thereof:- total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10~8- 10"4 M of agent is to be maintained on the implant or barrier surface. (D) Epithilones including ixabepilone and analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10"8- 10~4 M of agent is to be maintained on the implant or barrier surface. (E) Kinesin antagonists including SB-715992 and analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10^- 10"4 M of agent is to be maintained on the implant or barrier surface. (F) TNF alpha antagonists including Etanercept, humicade, adalimumab and analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10^- 10"4 M of agent is to be maintained on the implant or barrier surface. (G) AKT inhibitor including erucylphosphocholine and analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10^- 10"4 M of agent is to be maintained on the implant or barrier surface. (H) FGF Inhibitors including IDN-5390 and analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10'8- 10~4 M of agent is to be maintained on the implant or barrier surface. (I) Collagenase Antagonists including S-0885 and analogues and derivatives thereof: total dose not to exceed 1000 mg (range of 0.1 μg to 1000 mg); preferred 1 μg to 500 mg. Dose per unit area of 0.01 μg - 500 μg per mm2; preferred dose of 0.1 μg/mm2 - 100 μg/mm2. Minimum concentration of 10"8 - 10"4 M of agent is to be maintained on the implant or barrier surface. (J) NF KAPPA B Inhibitors including Bortezomib and analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10"8- 10"4 M of agent is to be maintained on the implant or barrier surface. (K) Elongation factor-1 alpha inhibitors including aplidine and analogues and derivatives thereof: total dose not to exceed 1000 mg (range of 0.1 μg to 1000 mg); preferred 1 μg to 500 mg. Dose per unit area of 0.01 μg - 500 μg per mm2; preferred dose of 0.1 μg/mm2 - 100 μg/mm2. Minimum concentration of 10^- 10"4 M of agent is to be maintained on the implant or barrier surface. (L) Tyrosine kinase inhibitors including Gefitinib and analogues and derivatives thereof: total dose not to exceed 1000 mg (range of 0.1 μg to 1000 mg); preferred 1 μg to 500 mg. Dose per unit area of 0.01 μg - 500 μg per mm2; preferred dose of 0.1 μg/mm2 - 100 μg/mm2. Minimum concentration of 10^ - 10"4 M of agent is to be maintained on the implant or barrier surface.
For high potency drugs, the total dose typically should not exceed 200 mg (range of 0.1 μg to 200 mg) and preferably 1 μg to 100 mg; dose per unit area of 0.01 μg - 100 μg per mm2; preferably 0.1 μg/mm2 - 20 μg/mm2; and minimum concentration of 10^- 10"4 M of agent should be maintained on the implant or barrier surface. For mid-potency drugs, the total dose typically should not to exceed 500 mg (range of 1.0 μg to 500 mg) and preferably 1 μg to 200 mg; dose per unit area of 0.01 μg - 200 μg per mm2, preferably 0.1 μg/mm2 - 40 μg/mm2; and minimum concentration of 10' 8- 10"4 M of agent should be maintained on the implant or barrier surface. For low potency drugs, the total dose typically should not exceed 1000 mg (range of 0.1 μg to 1000 mg), preferably 1 μg to 500 mg; dose per unit area of 0.01 μg - 500 μg per mm2; preferably 0.1 μg/mm2 - 100 μg/mm2; and minimum concentration of 10 s - 10"4 M of agent should to be maintained on the implant or barrier surface.
Peritoneal Dialysis Catheters
In one aspect, the present invention provides for the combination of an anti-scarring agent and a peritoneal dialysis catheter or a peritoneal implant for drug delivery.
Peritoneal catheters may be used for peritoneal dialysis. Peritoneal dialysis is a form of dialysis in which the blood is not removed from the body but instead, cleansing fluid is put into the abdominal cavity where the body's peritoneum acts as the dialysis membrane. The dialysate equilibrates with plasma for several hours and then the equilibrated dialysate is drained with the associated toxins. The peritoneal catheter is surgically placed into the peritoneal cavity in order to drain dialysate into and out of the peritoneal cavity. Peritoneal dialysis catheters are typically double-cuffed and tunnelled catheters that provide access to the peritoneum. The most common peritoneal dialysis catheter designs are the Tenckhoff catheter, the Swan Neck Missouri catheter and the Toronto Western catheter. In peritoneal dialysis, the peritoneum acts as a semipermeable membrane across which solutes can be exchanged down a concentration gradient. Continuous peritoneal access catheters are permanently implanted for those that require repeated access to the peritoneum. Implanted peritoneal catheters may be used for peritoneal dialysis or for a means of delivering drug to the peritoneum. These catheters may be composed of synthetic materials, such as silicone, rubber, polyurethane or other polymers that provide flexibility. They may be designed to be configured as a straight tube or may be bent and molded into a variety of shapes to provide different configurations, including helices and coils. The peritoneal catheters may be composed of one continuous element or may be sectioned into parts to provide flanges, cuffs, beads or discs at one of the ends to fix the catheter in position.
For example, the peritoneal catheter may be a resilient, foldable, T-shaped housing chamber with access ports that have elongated, flexible, fluid channels that gather or distribute a liquid such as dialysis fluid. See, e.g., U.S. Patent No. 5,322,519. The peritoneal catheter may be composed of two linearly mated inflow and outflow conduits contoured as a circular cross-section, which join fluted fluid transport branches. See, e.g., U.S. Patent No. 6,659,134. The peritoneal catheter may be composed of a ductwork of multiple tubes with fluid holes enclosed within a fluid permeable envelope structure that has slits to allow fluid flow but not tissue adherence. See, e.g., U.S. Patent No. 5,254,084. The peritoneal catheter may have a one-half helical turn to provide a radial flow and be composed of a plurality of ingress and egress ports positioned about its circumference and length, and have a coating of ultra low temperature isotropic carbon on the intraabdominal section. See, e.g., U.S. Patent No. 5,098,413. The peritoneal catheter may be an elongated flexible tube with one end connected to a pair of spaced apart sheets that extends exteriorly into the body cavity with at least one cuff for preventing catheter infections. See, e.g., U.S. Patent No. 4,368,737. The peritoneal catheter may be composed of two sections which includes a retainer section that permanently ingrows into the abdominal wall and an elongated flexible tube section for delivering and withdrawing dialysate. See, e.g., U.S. Patent No. 4,278,092. The peritoneal catheter may be flexible tube having a natural bent segment between the proximal and distal ends which includes a flange extending circumferentially at a nonperpendicular angle relative to the axis of the catheter tube. See, e.g., U.S. Patent No. 4,687,471. The peritoneal catheter may be a percutaneous access device composed of a cylindrical neck portion for skin protrusion, an annular skirt portion for anchoring into the dermis/subcutaneous tissue, and a catheter tube that may be threaded through the neck and skirt portions that has flexible bellows which can form a 90 degree angle. See, e.g., U.S. Patent No. 4,886,502. The. peritoneal catheter may be a flexible, elongated tube with perforations in the wall to pass fluid with a means for urging the central portion of the tube into a tightly wound cylindrical helix configuration. See, e.g., U.S. Patent No. 4,681,570. Other examples of peritoneal catheters used for dialysis are described in, e.g., U.S. Patent Nos. 6,290,669; 5,752,939 and 5,171,227.
In another aspect, the peritoneal catheter may be used to administer drugs to the peritoneum. For example, the peritoneal catheter may be a subcutaneous injection catheter apparatus having a receiving chamber with a penetrable membrane to accommodate an injection needle, which may be interconnected to the peritoneal cavity by a hollow stem. See, e.g., U.S. Patent No. 4,400,169. The peritoneal catheter may be composed of a porous outer casing defining an inner space with an inlet and outlet catheter of non-porous material which are in communication with an opening of the outer casing to form two passageways. See, e.g., U.S. Patent No. 5,100,392. Long-term use of peritoneal catheters may lead to infections or blockage of the catheter due to fibrin formation. Synthetic peritoneal catheters and delivery devices that include an anti-scarring drug are capable of preventing stenosis.
Peritoneal catheters, which may be combined with one or more agents according to the present invention, include commercially available products. For example, Cook Critical Care (Bloomington, IN) sells the Spiral Chronic Peritoneal Dialysis Catheters and Tenckhoff Chronic Peritoneal Dialysis Catheters. Bard Access Systems (Salt Lake City, UT) sells the Tenckhoff and HEMOSPLIT Peritoneal Dialysis Catheters. CardioMed Supplies, lnc (ON, Canada) sells the Single Cuff and Double Cuff Straight Peritoneal Dialysis Catheters, as well as the Single Cuff and Double Cuff Coiled Peritoneal Dialysis Catheters. Other companies that sell Single and Double Cuff, Straight and Coiled Tenckhoff catheters and other types of peritoneal catheters include Baxter International, Inc. (Deerfield, IL), Fresenius Medical Care (Lexington, MA) and Gambro AB (Sweden).
In one aspect, the present invention provides peritoneal access catheters that include an anti-scarring agent or a composition that includes an anti-scarring agent. Numerous polymeric and non-polymeric delivery systems for use in peritoneal dialysis implants and catheters have been described above.
Methods for incorporating the fibrosis-inhibiting agent into or onto the device includes: (a) directly affixing to the device a fibrosis- inhibiting composition (e.g., by either a spraying process or dipping process as described above, with or without a carrier), (b) directly incorporating into the device a fibrosis-inhibiting composition (e.g., by either a spraying process or dipping process as described above, with or without a carrier), (c) by coating the device with a substance such as a hydrogel which will in turn absorb the fibrosis-inhibiting composition, (d) by interweaving fibrosis- inhibiting composition coated thread (or the polymer itself formed into a thread) into the device structure, (e) constructing the device itself or a portion of the device with a fibrosis-inhibiting composition, or (f) by covalently binding the fibrosis-inhibiting agent directly to the device surface or to a linker (small molecule or polymer) that is coated or attached to the device surface. The coatings can be applied to different portions of the device. For example, the coating can be (a) as a coating applied to the external surface of the graft; (b) as a coating applied to the internal (luminal) surface of the graft; (c) as a coating applied to the superficial cuff; (d) as a coating applied to the deep cuff; or (e) as a coating applied to a combination of these surfaces.
The fibrosis-inhibiting agent can be mixed with the materials that are used to make the device such that the fibrosis-inhibiting agent is incorporated into the final device.
In addition to incorporation of a fibrosis-inhibiting agent into or onto the device, another biologically active agent can be incorporated into or onto the device, for example an anti-inflammatory (e.g., dexamethazone or aspirin), antithrombotic agents (e.g., heparin, heparin complexes, hydrophobic heparin derivatives, aspirin, or dipyridamole) and/or an antibiotic including sulfonamides, penicillins, cephalosporins, aminoglycosides (e.g., amoxicillin, trimethoprim-sulfamethoxazole, azithromycin, clarithromycin, bacitracin, polymixin, chloramphenicol, erythromycin, clindomycin, amoxicillin-clavulanate, cefprozil, cefuroxime, cefpodoxime, or cefdinir).
According to the present invention, any fibrosis-inhibiting agent described above can be utilized in the practice of this embodiment. Within one embodiment of the invention, peritoneal dialysis implants and catheters may be adapted to release an agent that inhibits one or more of the four general components of the process of fibrosis (or scarring), including: formation of new blood vessels (angiogenesis), migration and proliferation of connective tissue cells (such as fibroblasts or smooth muscle cells), deposition of extracellular matrix (ECM), and remodeling (maturation and organization of the fibrous tissue). By inhibiting one or more of the components of fibrosis (or scarring), the overgrowth of granulation tissue may be inhibited or reduced.
As peritoneal access catheters devices are made in a variety of configurations and sizes, the exact dose administered will vary with device size, surface area and design. However, certain principles can be applied in the application of this art. Drug dose can be calculated as a function of dose per unit area (of the portion of the device being coated), total dose administered, and appropriate surface concentrations of active drug can be determined. Drugs are to be used at concentrations that range from several times more than to 10%, 5%, or even less than 1% of the concentration typically used in a single chemotherapeutic systemic dose application. Preferably, the drug is released in effective concentrations for a period ranging from 1 - 90 days.
Preferred fibrosis-inhibiting agents for use in peritoneal access catheters and implants include the following: ZD-6474, AP-23573, synthadotin, S-0885, aplidine, ixabepilone, IDN-5390, SB-2723005, ABT- 518, combretastatin, anecortave acetate, SB-715992, temsirolimus, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, as well as analogues and derivatives of the aforementioned.
Additional examples of anti-scarring agents which can be used include those having a high potency in the assays described herein (1- 10OnM IC50 range) such as isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, dolastatin 15, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17- DMAG, and tacrolimus; those having a mid-potency in the assays described herein (100-500 nM IC50 range) such as loteprednol etabonate, juglone, prednisolone, puromycin, 3-BAABE, cladribine, and mannose-6-phosphate; and those having a low potency in the assays described herein (500- 1000nm range IC50 range) such as 5-azacytidine, Ly333531(ruboxistaurin), and simvastatin. Regardless of the method of application of the drug to the device, the exemplary anti-fibrosing agents, used alone or in combination, should be administered under the following dosing guidelines. The total amount (dose) of anti-scarring agent in or on the device may be in the range of about 0.01 μg-10 μg, or 10 μg-10 mg, or 10 mg-250 mg, or 250 mg-1000 mg, or 1000 mg-2500 mg. The dose (amount) of anti-scarring agent per unit area of device surface to which the agent is applied may be in the range of about 0.01 μg/mm2 - 1 μg/mm2, or 1 μg/mm2 - 10 μg/mm2, or 10 μg/mm2 - 250 μg/mm2, 250 μg/mm2 - 1000 μg/mm2, or 1000 μg/mm2 - 2500 μg/mm2.
Provided below are exemplary dosage ranges for various anti- scarring agents that can be used in conjunction with peritoneal access catheter devices and implants in accordance with the invention. (A) Angiogenesis inhibitors including Alphastatin, ZD-6474, IDN-5390, SB- 2723005, ABT-518, combretastatin, and anecortane, analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10^- 10"4 M of agent is to be maintained on the implant or barrier surface.
(B) mTOR inhibitors including AP-23573 and Temsirolimus, analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10^- 10"4 M of agent is to be maintained on the implant or barrier surface.
(C) Tubulin antagonists including synthadotin, analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10"8- 10"4 M of agent is to be maintained on the implant or barrier surface. (D) Epithilones including ixabepilone and analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10^- 10"4 M of agent is to be maintained on the implant or barrier surface. (E) Kinesin antagonists including SB-715992 and analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10~8- 10"4 M of agent is to be maintained on the implant or barrier surface. (F) TNF alpha antagonists including Etanercept, humicade, adalimumab and analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10^- 10"4 M of agent is to be maintained on the implant or barrier surface. (G) AKT inhibitor including erucylphosphocholine and analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10^- 10"4 M of agent is to be maintained on the implant or barrier surface. (H) FGF Inhibitors including IDN-5390 and analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10"8 - 10"4 M of agent is to be maintained on the implant or barrier surface. (I) Collagenase Antagonists including S-0885 and analogues and derivatives thereof: total dose not to exceed 1000 mg (range of 0.1 μg to 1000 mg); preferred 1 μg to 500 mg. Dose per unit area of 0.01 μg - 500 μg per mm2; preferred dose of 0.1 μg/mm2 - 100 μg/mm2. Minimum concentration of 10~8 - 10"4 M of agent is to be maintained on the implant or barrier surface. (J) NF KAPPA B Inhibitors including Bortezomib and analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10'8- 10"4 M of agent is to be maintained on the implant or barrier surface. (K) Elongation factor-1 alpha inhibitors including aplidine and analogues and derivatives thereof: total dose not to exceed 1000 mg (range of 0.1 μg to 1000 mg); preferred 1 μg to 500 mg. Dose per unit area of 0.01 μg - 500 μg per mm2; preferred dose of 0.1 μg/mm2 - 100 μg/mm2. Minimum concentration of 10"8- 10"4 M of agent is to be maintained on the implant or barrier surface. (L) Tyrosine kinase inhibitors including Gefitinib and analogues and derivatives thereof: total dose not to exceed 1000 mg (range of 0.1 μg to 1000 mg); preferred 1 μg to 500 mg. Dose per unit area of 0.01 μg - 500 μg per mm2; preferred dose of 0.1 μg/mm2 - 100 μg/mm2. Minimum concentration of 10"8~ 10"4 M of agent is to be maintained on the implant or barrier surface.
For high potency drugs, the total dose typically should not exceed 200 mg (range of 0.1 μg to 200 mg) and preferably 1 μg to 100 mg; dose per unit area of 0.01 μg - 100 μg per mm2; preferably 0.1 μg/mm2 - 20 μg/mm2; and minimum concentration of 10"8- 10"4 M of agent should be maintained on the implant or barrier surface. For mid-potency drugs, the total dose typically should not to exceed 500 mg (range of 1.0 μg to 500 mg) and preferably 1 μg to 200 mg; dose per unit area of 0.01 μg - 200 μg per mm2, preferably 0.1 μg/mm2 - 40 μg/mm2; and minimum concentration of 10' 8- 10"4 M of agent should be maintained on the implant or barrier surface. For low potency drugs, the total dose typically should not exceed 1000 mg (range of 0.1 μg to 1000 mg), preferably 1 μg to 500 mg; dose per unit area of 0.01 μg - 500 μg per mm2; preferably 0.1 μg/mm2 - 100 μg/mm2; and minimum concentration of 10~8- 10"4 M of agent should to be maintained on the implant or barrier surface.
Central Nervous System Shunts and Pressure Monitoring Devices
In one aspect, the present invention provides for the combination of an anti-scarring agent and a central nervous system (CNS) device, such as a CNS shunt or a pressure monitoring device. CNS devices that comprise an anti-scarring agent are capable of preventing stenosis and obstruction of the device leading to hydrocephalus and increased intercranial pressure.
Hydocephalus, or accumulation of cerebrospinal fluid (CSF) in the brain, is a frequently encountered neurosurgical condition arising from congenital malformations, infection, hemmorrhage, or malignancy. The incompressible fluid exerts pressure on the brain leading to brain damage or even death if untreated. CNS shunts are conduits placed in the ventricles of the brain to divert the flow of CSF from the brain to other body compartments and relieve the fluid pressure. Ventricular CSF is diverted via a prosthetic shunt to a number of drainage locations including the pleura (ventriculopleural shunt), jugular vein, vena cava (VA shunt), gallbladder and peritoneum (VP shunt; most common).
Representative examples of CNS devices include, e.g., CNS shunts, such as ventriculopleural shunts, jugular vein and vena cava (VA) shunts, and ventriculoperitoneal shunt (VP shunt), such as gallbladder and peritoneum shunts; External Ventricular Drainage (EVD) devices; and Intracranial Pressure (ICP) Monitoring Devices. Other CNS devices include, e.g., dural patches and implants to prevent epidural fibrosis post- laminectomy; and devices for continuous subarachnoid infusions.
In one aspect, the CNS device may be a drainage shunt used to drain fluids in the brain. For example, the CNS device may be a cerebrospinal shunt composed of two tubes whereby an inner tube supplies the fluid from the brain ventricles to the peritoneum region and an outer tube is arranged to exert pressure on the inner tube as the volume of fluid builds in the outer tube. See, e.g., U.S. Patent No. 5,405,316. The CNS device may be a ventricular drainage system adapted for connection to a ventricular drainage catheter for receiving cerebrospinal fluid and having a valve for controlling fluid flow therethrough. See, e.g., U.S. Patent No. 5,772,625. The CNS device may be a brain ventricular shunt system composed of a brain check valve for preventing cerebrospinal fluid backflow and a flow-rate switching mechanism to provide flow of cerebrospinal fluid from the brain ventricle catheter to the peritoneum or auricle catheter. See, e.g., U.S. Patent No. 4,781 ,673. The CNS device may be shunt member with a flow restricting passage that is connected to catheters to provide cerebrospinal fluid drainage from the brain ventricle to the sinus sagittalis. See, e.g., U.S. Patent No. 6,283,934. The CNS device may be a ventricular end of a ventriculo-cardiac shunt that has a closed distal end with lateral passageways adjacent thereto which are porous and expansible for providing an umbrella-like liner to allow passage of fluid while preventing obstruction. See, e.g., U.S. Patent No. 3,690,323. The CNS device may be a hydrocephalus valve composed of a chamber with an inlet and outlet valve for routing cerebrospinal fluid away from the brain at a controlled pressure. See, e.g., U.S. Patent No. 5,069,663. The CNS device may be a hydrocephalus device composed of an external, flexible shell forming a fluid reservoir and housing a non-obstructive, self-regulating valve having a folded membrane which forms a slit-like opening, which has inlet and outlet tubes. See, e.g., U.S. Patent No. 5,728,061. The CNS device may be a cerebral spinal fluid draining shunt composed of an implantable master control unit that interconnects a cerebral spinal space catheter with a catheter that drains the fluid into a body cavity. See, e.g., U.S. Patent No. 6,585,677. The CNS device may be a cerebrospinal fluid shunt composed of a ventricular catheter connected to a flexible drainage tube which has an exterior flexible tubular cover from which the drainage tube may be drawn. See, e.g., U.S. Patent No. 4,950,232. The CNS device may be an intracranial shunting tube composed of a thin film that extends radially and outwardly from the open end of a ventricular tube which has a plurality of side holes to bypass ventricular cerebrospinal fluid to the subdural space on the surface of the brain. See, e.g., U.S. Patent No. 5,000,731. Other CNS shunts are described in, e.g., U.S. Patent Nos. 6,575,928; 5,437,626 and 4,631 ,051. In another aspect, the CNS device may be a pressure monitoring device. For example, the pressure monitoring device may be an intracranial pressure sensor which is mounted within the skull of a body at the situs where the pressure is to be monitored and a means of transmitting the pressure externally from the skull. See, e.g., U.S. Patent No. 4,003,141. The pressure monitoring device may be a telemetric differential pressure sensitive device composed of a thin, planar, closed, conductive loop which moves with a flexible diaphragm upon changes in the difference of two bodily pressures on its opposite sides. See, e.g., U.S. Patent No. 4,593,703. The pressure monitoring device may be composed of a radio- opaque liquid contained within a resiliently compressible vessel of a silastic material in which the volume of liquid is variable as a function of the pressure or force applied to the vessel. See, e.g., U.S. Patent No. 3,877,137. The pressure monitoring device may be a probe composed of a threaded shaft having a lumen and an engaging lock nut, which is inserted through an opening in the scalp and into the subarachnoid space. See, e.g., U.S. Patent No. 4,600,013. The pressure monitoring device may be composed of an external transceiver unit and an implantable cavity resonator unit having a dielectric-filled cavity with a predetermined resonance frequency for high frequency electromagnetic waves. See, e.g., U.S. Patent No. 5,873,840. The pressure monitoring device may be an implantable sensor that detects a physiological parameter (e.g., cerebral spinal fluid flow) and then generates, processes, and transmits the signal to an external receiver. See, e.g., U.S. Patent No. 6,533,733. Other CNS pressure monitoring devices are described in, e.g., U.S. Patent Nos. 6,248,080 and 6,210,346.
CNS shunts, which may be combined with one or more agents according to the present invention, include commercially available products, such as the Codman HAKIM Programmable Valves from Codman & Shurtleff, Inc. (Raynham, MA), a Johnson & Johnson Company. Other examples include the Integra Neuro Sciences (Plainsboro, NJ) HEYER- SCHULTE Neurosurgical Shunts, HERMETIC CSF Drainage Systems, and OSV Il SMART VALVE Systems and the Medtronic, Inc. (Minneapolis, MN) Shunt Assemblies, including the STRATA, DELTA, CSF-Snap and CSF- Flow Control Shunt Assemblies.
Pressure Monitoring CNS devices, which may be combined with one or more agents according to the present invention, include commercially available products such as the VENTRIX Pressure Monitoring Kits and CAMINO Micro Ventricular Bolt ICP Monitoring Catheters from Integra Neuro Sciences (Plainsboro, NJ).
In one aspect, the present invention provides CNS devices that include an anti-scarring agent or a composition that includes an anti- scarring agent. Numerous polymeric and non-polymeric delivery systems for use in CNS devices have been described above. Methods for incorporating the fibrosis-inhibiting agent into or onto the device includes: (a) directly affixing to the device a fibrosis-inhibiting composition (e.g., by either a spraying process or dipping process as described above, with or without a carrier), (b) directly incorporating into the device a fibrosis- inhibiting composition (e.g., by either a spraying process or dipping process as described above, with or without a carrier), (c) by coating the device with a substance such as a hydrogel which will in turn absorb the fibrosis- inhibiting composition, (d) by interweaving fibrosis-inhibiting composition coated thread (or the polymer itself formed into a thread) into the device structure, (e) constructing the device itself or a portion of the device with a fibrosis-inhibiting composition, or (f) by covalently binding the fibrosis- inhibiting agent directly to the device surface or to a linker (small molecule or polymer) that is coated or attached to the device surface. The coatings can be applied to different portions of the device. For example, the coating can be (a) as a coating applied to the external surface of the shunt; (b) as a coating applied to the internal (luminal) surface of the shunt; or (c) as a coating applied to all or parts of both surfaces The fibrosis-inhibiting agent can be mixed with the materials that are used to make the device such that the fibrosis-inhibiting agent is incorporated into the final device.
In addition to incorporation of a fibrosis-inhibiting agent into or onto the device, another biologically active agent can be incorporated into or onto the device, for example an anti-inflammatory (e.g., dexamethazone or aspirin), antithrombotic agents (e.g., heparin, heparin complexes, hydrophobic heparin derivatives, aspirin, or dipyridamole) and/or an antibiotic (e.g., amoxicillin, trimethoprim-sulfamethoxazole, azithromycin, clarithromycin, amoxicillin-clavulanate, cefprozil, cefuroxime, cefpodoxime, or cefdinir).
According to the present invention, any fibrosis-inhibiting agent described above can be utilized in the practice of this embodiment. Within one embodiment of the invention, CNS devices may be adapted to release an agent that inhibits one or more of the four general components of the process of fibrosis (or scarring), including: formation of new blood vessels (angiogenesis), migration and proliferation of connective tissue cells (such as fibroblasts or smooth muscle cells), deposition of extracellular matrix (ECM), and remodeling (maturation and organization of the fibrous tissue). By inhibiting one or more of the components of fibrosis (or scarring), the overgrowth of granulation tissue may be inhibited or reduced.
As CNS devices are made in a variety of configurations and sizes, the exact dose administered will vary with device size, surface area and design. However, certain principles can be applied in the application of this art. Drug dose can be calculated as a function of dose per unit area (of the portion of the device being coated), total dose administered, and appropriate surface concentrations of active drug can be determined. Drugs are to be used at concentrations that range from several times more than to 10%, 5%, or even less than 1% of the concentration typically used in a single chemotherapeutic systemic dose application. Preferably, the drug is released in effective concentrations for a period ranging from 1 - 90 days. Several examples of fibrosis-inhibiting agents for use in CNS devices include the following: ZD-6474, AP-23573, synthadotin, S-0885, aplidine, ixabepilone, IDN-5390, SB-2723005, ABT-518, combretastatin, anecortave acetate, SB-715992, temsirolimus, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, as well as analogues and derivatives of the aforementioned.
Additional examples of anti-scarring agents which can be used include those having a high potency in the assays described herein (1- 10OnM IC50 range) such as isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A1 brefeldin A, thapsigargin, dolastatin 15, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17- DMAG, and tacrolimus; those having a mid-potency in the assays described herein (100-500 nM IC50 range) such as loteprednol etabonate, juglone, prednisolone, puromycin, 3-BAABE, cladribine, and mannose-6-phosphate; and those having a low potency in the assays described herein (500- 1000nm range IC50 range) such as 5-azacytidine, Ly333531(ruboxistaurin), and simvastatin.
Regardless of the method of application of the drug to the device, the exemplary anti-fibrosing agents, used alone or in combination, should be administered under the following dosing guidelines. The total amount (dose) of anti-scarring agent in or on the device may be in the range of about 0.01 μg-10 μg, or 10 μg-10 mg, or 10 mg-250 mg, or 250 mg-1000 mg, or 1000 mg-2500 mg. The dose (amount) of anti-scarring agent per unit area of device surface to which the agent is applied may be in the range of about 0.01 μg/mm2 - 1 μg/mm2, or 1 μg/mm2 - 10 μg/mm2, or 10 μg/mm2 - 250 μg/mm2, 250 μg/mm2 - 1000 μg/mm2, or 1000 μg/mm2 - 2500 μg/mm2.
Provided below are exemplary dosage ranges for various anti- scarring agents that can be used in conjunction with CNS devices in accordance with the invention. (A) Angiogenesis inhibitors including Alphastatin, ZD-6474, IDN-5390, SB-2723005, ABT-518, combretastatin, and anecortane, analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10^- 10"4 M of agent is to be maintained on the implant or barrier surface. (B) mTOR inhibitors including AP-23573 and Temsirolimus, analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10^- 10"4 M of agent is to be maintained on the implant or barrier surface. (C) Tubulin antagonists including synthadotin, analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10"8- 10"4 M of agent is to be maintained on the implant or barrier surface. (D) Epithilones including ixabepilone and analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10"8- 10"4 M of agent is to be maintained on the implant or barrier surface. (E) Kinesin antagonists including SB- 715992 and analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10^- 10"4 M of agent is to be maintained on the implant or barrier surface. (F) TNF alpha antagonists including Etanercept, humicade, adalimumab and analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10^- 10"4 M of agent is to be maintained on the implant or barrier surface. (G) AKT inhibitor including erucylphosphocholine and analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10^- 10"4 M of agent is to be maintained on the implant or barrier surface. (H) FGF Inhibitors including IDN-5390 and analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10"8- 10"4 M of agent is to be maintained on the implant or barrier surface. (I) Collagenase Antagonists including S-0885 and analogues and derivatives thereof: total dose not to exceed 1000 mg (range of 0.1 μg to 1000 mg); preferred 1 μg to 500 mg. Dose per unit area of 0.01 μg - 500 μg per mm2; preferred dose of 0.1 μg/mm2 - 100 μg/mm2. Minimum concentration of 10'8- 10"4 M of agent is to be maintained on the implant or barrier surface. (J) NF KAPPA B Inhibitors including Bortezomib and analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10"8- 10~4 M of agent is to be maintained on the implant or barrier surface. (K) Elongation factor-1 alpha inhibitors including aplidine and analogues and derivatives thereof: total dose not to exceed 1000 mg (range of 0.1 μg to 1000 mg); preferred 1 μg to 500 mg. Dose per unit area of 0.01 μg - 500 μg per mm2; preferred . dose of 0.1 μg/mm2 - 100 μg/mm2. Minimum concentration of 10~8- 10"4 M of agent is to be maintained on the implant or barrier surface. (L) Tyrosine kinase inhibitors including Gefitinib and analogues and derivatives thereof: total dose not to exceed 1000 mg (range of 0.1 μg to 1000 mg); preferred 1 μg to 500 mg. Dose per unit area of 0.01 μg - 500 μg per mm2; preferred dose of 0.1 μg/mm2 - 100 μg/mm2. Minimum concentration of 10~8- 10"4 M of agent is to be maintained on the implant or barrier surface.
For high potency drugs, the total dose typically should not exceed 200 mg (range of 0.1 μg to 200 mg) and preferably 1 μg to 100 mg; dose per unit area of 0.01 μg - 100 μg per mm2; preferably 0.1 μg/mm2 - 20 μg/mm2; and minimum concentration of 10'8- 1CT4 M of agent should be maintained on the implant or barrier surface. For mid-potency drugs, the total dose typically should not to exceed 500 mg (range of 1.0 μg to 500 mg) and preferably 1 μg to 200 mg; dose per unit area of 0.01 μg - 200 μg per mm2, preferably 0.1 μg/mm2 - 40 μg/mm2; and minimum concentration of 10" 8- 10"4 M of agent should be maintained on the implant or barrier surface. For low potency drugs, the total dose typically should not exceed 1000 mg (range of 0.1 μg to 1000 mg), preferably 1 μg to 500 mg; dose per unit area of 0.01 μg - 500 μg per mm2; preferably 0.1 μg/mm2 - 100 μg/mm2; and minimum concentration of 10"8- 10"4 M of agent should to be maintained on the implant or barrier surface.
Inferior Vena Cava Filters
In one aspect, the present invention provides for the combination of an anti-scarring agent and an inferior vena cava filter device. The term inferior vena cava filters are devices that are intended to capture emboli and prevent them from migrating through the blood stream. Examples of inferior vena cava filters include, without limitation, vascular filters, blood filters, implantable blood filters, caval filters, vena cava filters, vena cava filtering devices, thrombosis filters, thrombus filters, antimigration filters, filtering devices, percutaneous filter systems, intravascular traps, intravascular filters, clot filters, vein filters and body vessel filters.
Inferior vena cava filters catch blood clots to prevent them from traveling to other parts of the body to form an embolus. It may be life threatening if plaques or blood clots migrate through the blood stream and travel to the lungs and cause a pulmonary embolism. To prevent such an occurrence, inferior vena cava filters are placed in the large veins of the body to prevent pulmonary emboli in patients with (or at risk of developing) deep vein thrombosis. Most often these filters are composed of synthetic polymers or metals. These filters may be a variety of configurations, including but not limited to, baskets, cones, umbrellas or loops. The shape of the filter must provide adequate trapping ability while allowing sufficient blood flow. Along with the functional shape, filters may also have other design features including peripheral loops for alignment or anchoring features to prevent migration (e.g., ridges, struts or sharp points). Where the filter comes into contact with the vessel wall for anchoring, a fibrotic response may occur. This fibrotic response can result in difficulties in removal of the filter. This is a particular problem for filters that are to be kept in place for a relatively short period of time. Incorporation of a fibrosis- inhibiting agent into or onto the filter may reduce or prevent stenosis or obstruction of the device via a fibroproliferative response.
In one aspect, inferior vena cava filters may be designed in a variety of configurations. For example, the inferior vena cava filter may be composed of a plurality of intraluminal filter elements held by a retainer in a filter configuration that may be released to an open, stent-like configuration. See, e.g., U.S. Patent No. 6,267,776. The inferior vena cava filter may be composed of an embolus capturing portion having a plurality of elongated filter wires diverging in a helical arrangement to form a conical surface and an anchoring portion that has a plurality of struts. See, e.g., U.S. Patent No. 6,391 ,045. The inferior vena cava filter may be composed of a textured echogenic feature so the filter position may be determined by sonographic visualization. See, e.g., U.S. Patent No. 6,436,120. The inferior vena cava filter may be composed of a plurality of core wire struts that are anchored to radiate outwardly which are interconnected by compression material to form a filter basket. See, e.g., U.S. Patent No. 5,370,657. The inferior vena cava filter may be composed of an apical head with a plurality of divergent legs in a conical shaped geometry which have a hook and pad for securing to the vessel. See, e.g., U.S. Patent No. 5, 059,205. The inferior vena cava filter may be composed of a filtering device made of shape memory/superelastic material formed at the distal end of a deployment/retrieval wire section for minimally invasive positioning. See, e.g., U.S. Patent No. 5,893,869. The inferior vena cava filter may be composed of a plurality of intraluminal elements joined by a retainer, whereby upon release of the retainer, the intraluminal filter elements convert to an open configuration in the blood vessel. See, e.g., U.S. Patent Nos. 6,517,559 and 6,267,776. The inferior vena cava filter may be composed of an outer catheter and an inner catheter having a collapsible mesh-like filter basket at the distal end made of spring wires or plastic monofilaments. See, e.g., U.S. Patent No. 5,549,626. The inferior vena cava filter may be composed of a plurality of radiating struts that attach at a body element and has a two layer surface treatment to provide endothelial cell growth and anti-proliferative properties. See, e.g., U.S. Patent No. 6,273,901. The inferior vena cava filter may be composed of a metal fabric that is configured as a particle-trapping screen that may be slideable along a guidewire. See, e.g., U.S. Patent No. 6,605,102. The inferior vena cava filter may be non-permanent with a single high memory coiled wire having a cylindrical and a conical segment. See, e.g., U.S. Patent No. 6,059,825. Other inferior vena cava filters are described in, e.g., U.S. Patent Nos. 6,623,506; 6,391 ,044; 6,231 ,589; 5,984,947; 5,695,518 and 4,817,600.
Vena cava filters, which may be combined with one or more anti-scarring agents according to the present invention, include commercially available products. Examples of vena cava filters that can benefit from the incorporation of a fibrosis-inhibiting agent include, without limitation, the GUNTHER TULIP Vena Cava FILTER and the GIANTURCO- ROEHM BIRD'S NEST Filter which are sold by Cook, Inc. (Bloomington, IN). CR. Bard (Murray Hill, NJ) sells the SIMON-NITINOL FILTER and RECOVERY Filter. Cordis Endovascular which is a subsidiary of Cordis Corporation (Miami Lakes, FL) sells the TRAPEASE Permanent Vena Cava Filter. B. Braun Medical Inc. (Bethlehem, PA) sells the VENA TECH LP Vena Cava Filter and VENA TECH - LGM Vena Cava Filter. Boston Scientific Corporation (Natick, MA) sells the Over-the-Wire GREENFIELD Vena Cava Filter. In one aspect, the present invention provides inferior vena cava filter devices that include an anti-scarring agent or a composition that includes an anti-scarring agent. Numerous polymeric and non-polymeric delivery systems for use in inferior vena cava filters have been described above. These compositions can further comprise one or more fibrosis- inhibiting agents such that the overgrowth of granulation tissue is inhibited or reduced.
Methods for incorporating the fibrosis-inhibiting agent into or onto the device includes: (a) directly affixing to the device a fibrosis- inhibiting composition (e.g., by either a spraying process or dipping process as described above, with or without a carrier), (b) directly incorporating into the device a fibrosis-inhibiting composition (e.g., by either a spraying process or dipping process as described above, with or without a carrier, (c) by coating the device with a substance such as a hydroge! which will in turn absorb the fibrosis-inhibiting composition, (d) by interweaving fibrosis- inhibiting composition coated thread (or the polymer itself formed into a thread) into the device structure, (e) constructing the device itself or a portion of the device with a fibrosis-inhibiting composition, or (T) by covalently binding the fibrosis-inhibiting agent directly to the device surface or to a linker (small molecule or polymer) that is coated or attached to the device surface. The coatings can be applied to different portions of the device. For example, the coating can be (a) as a coating applied to the entire leg of the filter; (b) as a coating applied to the tips of the filter that come into contact with the blood vessel; and/or (c) as a coating applied to all or parts of the entire filter device.
In addition to coating the device with the fibrosis-inhibiting composition, the fibrosis-inhibiting agent can be mixed with the materials that are used to make the device such that the fibrosis-inhibiting agent is incorporated into the final device.
In addition to incorporation of a fibrosis-inhibiting agent into or onto the device, another biologically active agent can be incorporated into or onto the device, for example an anti-inflammatory (e.g., dexamethazone or aspirin), antithrombotic agents (e.g., heparin, heparin complexes, hydrophobic heparin derivatives, aspirin, or dipyridamole), and/or an antibiotic (e.g., amoxicillin, trimethoprim-sulfamethoxazole, azithromycin, clarithromycin, amoxicillin-clavulanate, cefprozil, cefuroxime, cefpodoxime, or cefdinir).
According to the present invention, any fibrosis-inhibiting agent described above can be utilized in the practice of this embodiment. Within one embodiment of the invention, vena cava filters (e.g., inferior vena cava filters) may be adapted to release an agent that inhibits one or more of the four general components of the process of fibrosis (or scarring), including: formation of new blood vessels (angiogenesis), migration and proliferation of connective tissue cells (such as fibroblasts or smooth muscle cells), deposition of extracellular matrix (ECM), and remodeling (maturation and organization of the fibrous tissue). By inhibiting one or more of the components of fibrosis (or scarring), the overgrowth of granulation tissue may be inhibited or reduced.
Several examples of fibrosis-inhibiting agents for use in vena cava filter devices include the following: ZD-6474, AP-23573, synthadotin, S-0885, aplidine, ixabepilone, IDN-5390, SB-2723005, ABT-518, combretastatin, anecortave acetate, SB-715992, temsirolimus, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, as well as analogues and derivatives of the aforementioned.
Additional examples of anti-scarring agents which can be used include those having a high potency in the assays described herein (1- 10OnM IC50 range) such as isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, dolastatin 15, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17- DMAG, and tacrolimus; those having a mid-potency in the assays described herein (100-500 nM IC50 range) such as loteprednol etabonate, juglone, prednisolone, puromycin, 3-BAABE, cladribine, and mannose-6-phosphate; and those having a low potency in the assays described herein (500- 1000nm range IC50 range) such as 5-azacytidine, Ly333531(ruboxistaurin), and simvastatin.
As vena cava filter devices are made in a variety of configurations and sizes, the exact dose administered will vary with device size, surface area and design. However, certain principles can be applied in the application of this art. Drug dose can be calculated as a function of dose per unit area (of the portion of the device being coated), total dose administered, and appropriate surface concentrations of active drug can be determined. Drugs are to be used at concentrations that range from several times more than to 10%, 5%, or even less than 1% of the concentration typically used in a single chemotherapeutic systemic dose application. Preferably, the drug is released in effective concentrations for a period ranging from 1 - 90 days.
Regardless of the method of application of the drug to the device, the exemplary anti-fibrosing agents, used alone or in combination, should be administered under the following dosing guidelines. The total amount (dose) of anti-scarring agent in or on the device may be in the range of about 0.01 μg-10 μg, or 10 μg-10 mg, or 10 mg-250 mg, or 250 mg-1000 mg, or 1000 mg-2500 mg. The dose (amount) of anti-scarring agent per unit area of device surface to which the agent is applied may be in the range of about 0.01 μg/mm2 - 1 μg/mm2, or 1 μg/mm2 - 10 μg/mm2, or 10 μg/mm2 - 250 μg/mm2, 250 μg/mm2 - 1000 μg/mm2, or 1000 μg/mm2 - 2500 μg/mm2.
Provided below are exemplary dosage ranges for various anti- scarring agents that can be used in conjunction with vena cava devices in accordance with the invention. (A) Angiogenesis inhibitors including Alphastatin, ZD-6474, IDN-5390, SB-2723005, ABT-518, combretastatin, and anecortane, analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10^- 10"4 M of agent is to be maintained on the implant or barrier surface. (B) mTOR inhibitors including AP-23573 and Temsirolimus, analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10"8 - 10"4 M of agent is to be maintained on the implant or barrier surface. (C) Tubulin antagonists including synthadotin, analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10'8 - 10"4 M of agent is to be maintained on the implant or barrier surface. (D) Epithilones including ixabepilone and analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10^- 10"4 M of agent is to be maintained on the implant or barrier surface. (E) Kinesin antagonists including SB- 715992 and analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10^- 10"4 M of agent is to be maintained on the implant or barrier surface. (F) TNF alpha antagonists including Etanercept, humicade, adalimumab and analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10^- 10"4 M of agent is to be maintained on the implant or barrier surface. (G) AKT inhibitor including erucylphosphocholine and analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10^- 10"4 M of agent is to be maintained on the implant or barrier surface. (H) FGF Inhibitors including IDN-5390 and analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10"8 - 10'4 M of agent is to be maintained on the implant or barrier surface. (I) Collagenase Antagonists including S-0885 and analogues and derivatives thereof: total dose not to exceed 1000 mg (range of 0.1 μg to 1000 mg); preferred 1 μg to 500 mg. Dose per unit area of 0.01 μg - 500 μg per mm2; preferred dose of 0.1 μg/mm2 - 100 μg/mm2. Minimum concentration of 10"8- 10'4 M of agent is to be maintained on the implant or barrier surface. (J) NF KAPPA B Inhibitors including Bortezomib and analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10"8- 10"4 M of agent is to be maintained on the implant or barrier surface. (K) Elongation factor-1 alpha inhibitors including aplidine and analogues and derivatives thereof: total dose not to exceed 1000 mg (range of 0.1 μg to 1000 mg); preferred 1 μg to 500 mg. Dose per unit area of 0.01 μg - 500 μg per mm2; preferred dose of 0.1 μg/mm2 - 100 μg/mm2. Minimum concentration of 10"s- 10"4 M of agent is to be maintained on the implant or barrier surface. (L) Tyrosine kinase inhibitors including Gefitinib and analogues and derivatives thereof: total dose not to exceed 1000 mg (range of 0.1 μg to 1000 mg); preferred 1 μg to 500 mg. Dose per unit area of 0.01 μg - 500 μg per mm2; preferred dose of 0.1 μg/mm2 - 100 μg/mm2. Minimum concentration of 10"8- 10"4 M of agent is to be maintained on the implant or barrier surface.
For high potency drugs, the total dose typically should not exceed 200 mg (range of 0.1 μg to 200 mg) and preferably 1 μg to 100 mg; dose per unit area of 0.01 μg - 100 μg per mm2; preferably 0.1 μg/mm2 - 20 μg/mm2; and minimum concentration of 10'8- 10"4 M of agent should be maintained on the implant or barrier surface. For mid-potency drugs, the total dose typically should not to exceed 500 mg (range of 1.0 μg to 500 mg) and preferably 1 μg to 200 mg; dose per unit area of 0.01 μg - 200 μg per mm2, preferably 0.1 μg/mm2 - 40 μg/mm2; and minimum concentration of 10" 8- 10"4 M of agent should be maintained on the implant or barrier surface. For low potency drugs, the total dose typically should not exceed 1000 mg (range of 0.1 μg to 1000 mg), preferably 1 μg to 500 mg; dose per unit area of 0.01 μg - 500 μg per mm2; preferably 0.1 μg/mm2 - 100 μg/mm2; and minimum concentration of 10"8- 10"4 M of agent should to be maintained on the implant or barrier surface.
Gastrointestinal Devices
In one aspect, the present invention provides for the combination of an anti-scarring agent and a gastrointestinal (Gl) device. There are many gastrointestinal tube devices that are used for feeding applications and for drainage applications. The functioning of these tubes can be compromised if there is an excessive fibroproliferative response to these devices. The incorporation of a fibrosis-inhibiting agent into or onto the device can modulate this fibroproliferative response (e.g., to prevent stenosis and/or obstruction of the device) thereby maintaining performance of the device.
A variety of Gl tubes for drainage or feeding can be combined with a fibrosis-inhibiting agent to prevent stenosis and/or obstruction of the device. These devices may include, without limitation, Gl tubes for drainage or feeding, portosystemic shunts, shunts for ascites, nasogastric or nasoenteral tubes, gastrostomy or percutaneous feeding tubes, jejunostomy endoscopic tubes, colostomy devices, drainage tubes, biliary T-tubes, biopsy forceps, biliary stone removal devices, endoscopic retrograde cholangiopancretography (ERCP) devices, dilation balloons, enteral feeding devices, stents, low profile devices, virtual colonoscopy (VC) devices, capsule endoscopes, and retrieval devices.
Gl devices may be composed of synthetic materials, including, without limitation, stainless steel, metals, nitinol, glass, resins or polymers. In one aspect, the Gl device may be an instrument used to examine or provide access to the interior of the gastrointestinal tract. This may include optical imaging in the form of still imaging or videoing for diagnosing purposes. Procedures that use these devices include, without limitation, enteroscopy, colonoscopy or esophagogastroduodenoscopy, where an endoscope enters the esophagus or anal canal to assess portions of the Gl tract. For example, the Gl device may be an endoscope having a tubular shaft for receiving a viewing lens and a treatment instrument. See, e.g., U.S. Patent No. 5,421 ,323. The Gl device may be a multi-lumen endoscopic catheter that may be inserted through an endoscope for the practice of endoscopic retrograde cholangiopancreatography, whereby the first lumen has a wire threaded through it, the second lumen provides a conduit to infuse a radio-opaque contrast medium to identify obstructions, and the third lumen provides a conduit to dilate a balloon. See, e.g., U.S. Patent Nos. 5,788,681 and 5,843,028. The Gl device may be a video endoscope system composed of a swallowable capsule, a transmitter and a reception system. See, e.g., U.S. Patent No. 5,604,531. The Gl device may be an endoscope composed of an encapsulated ultrasonic transducer capsule having a self-contained electromechanical sector scanner, which may be used for transesophageal echocardiography. See, e.g., U.S. Patent Nos. 4,977,898 and 4,834,102. The Gl device may be a sterilizable endoscope having an image sensor mounted on a cylindrical capsule and a separable disposable channel. See, e.g., U.S. Patent No. 5,643,175. The Gl device may be a body canal intrusion instrument that may be composed of a bi-directional surface friction for engaging tissue during navigation to decrease the risk of puncture and time associated with the insertion of catheters, guidewires and endoscopes through body cavities and canals. See, e.g., U.S. Patent No. 6,589,213. The Gl device may be a colonic access device composed of flexible tubing with a tether for releasing from a colonoscope, which may be placed in the colon for up to several days to monitor and treat colorectal diseases. See, e.g., U.S. Patent No. 6,149,581. The Gl device may be adapted for the bile or pancreatic duct by being composed of a mother endoscope that is inserted into the duodenum and a daughter endoscope that is inserted via papilla through a forceps channel. See, e.g., U.S. Patent No. 4,979,496.
In another aspect, the Gl device may be used as a conduit for long-term tube feeding. These Gl devices may include, without limitation, percutaneous feeding tubes, enteral feeding devices/catheters, gastrostomy feeding tubes, low profile devices, and nasogastric tubes. These long-term feeding tubes may be advanced through the Gl tract via nasal canal or through the abdominal wall via a gastrostomy. For example, the Gl device may be an enteral feeding catheter adapted to serve as a conduit for passage of sustenance through an abdominal wall into the body and having a retainer and retractable locking means. See, e.g., Patent No. 4,826,481. The Gl device may be an enteral feeding tube having a catheter that allows for easy insertion and removal by having a slim, tapered guide tube and a balloon bolster. See, e.g., U.S. Patent No. 6,582,395. The Gl device may be an enteral feeding device for administering fluids into the stomach, which is composed of a female connector, flexible feeding tube, fluid discharge tube, and probe, which are connected to the male end of the guide wire. See, e.g., U.S. Patent No. 5,242,429. The Gl device may be a hollow, cylindrical elongated body with a spring-biased valve, which is maintained through a surgical opening in the stomach wall by an extended concentric flange that facilitates fixation. See, e.g., U.S. Patent No. 4,344,435. The Gl device may be a nasogastric tube having openings along its distal end with a coupled introducer flexible sheath extending longitudinally along the tube. See, e.g., U.S. Patent No. 5,334,167. Other Gl devices used as feeding tubes or related devices are described in, e.g., U.S. Patent Nos. 6,582,395; 5,989,225; 5,720,734; 5,716,347; 5,503,629; 5,342,321; 4,861, 334; 4,758,219 and 4,057r065.
In another aspect, the Gl device may be used for irrigation or aspiration of the Gl tract. These Gl devices may be used, for example, to remove ingested poisons or blood, to treat absorption-related conditions, to decompress the stomach, pre-operatively to ensure portions of the Gl tract is empty, post-operatively to remove gas, and to treat diseases such as bowel obstructions or paralytic ileus. For example, the Gl tube may be elongated and configured to be inserted in the Gl tract having a slidable treatment device for controlling bleeding and a fluid reservoir coupled to the tube. See, e.g., U.S. Patent No. 5,947,926. The Gl tube may be a nasogastric flexible tube with a curved or bent leading end to anatomically conform and facilitate advancement into the esophagus and stomach. See, e.g., U.S. Patent No. 5,690,620. The Gl tube may be a nasogastric elongated tube fixedly bent to extend from the nostril without affixation to avoid pressure necrosis in the nose due to force exertion. See, e.g., U.S. Patent No. 4,363,323. The Gl device may be composed of aspirating, feeding and inflation lumens, which is surgically inserted through the abdominal and gastric wall. See, e.g., U.S. Patent No. 4,543,089. The Gl device may be composed of drain tube and irrigating tube with a cuffed fluid sealing that is used for unidirectional irrigation of the bowels. See, e.g., U.S. Patent No. 4,637,814. The Gl device may be an open-ended, thin-walled, balloon-like tube shaped to extend through at least part of an alimentary canal for the purpose of passing digested food solids and thereby treating absorption-related diseases. See, e.g., U.S. Patent Nos. 4,315,509 and 4,134,405.
In another aspect, the Gl device may be a colostomy device. For example, the colostomy device may be an artificial anus composed of a hollow tubular support with a cylindrical body having a pair of radially- extending flanges to engage the member See, e.g., U.S. Patent No. 4,781 ,176. The colostomy device may be composed of internal and external balloons connected by a tube and an annular supporting plate for attachment to the stoma or rectum. See, e.g., U.S. Patent No. 5,569,216.
In another aspect, the Gl device may be a mechanical hemostatic device used to control Gl bleeding. Hemostatic devices, which are used to constrict blood flow, may include, without limitation, clamps, clips, staples and sutures. For example, the hemostatic device may be a compression clip composed of an anchor and stem having a transverse hole and a bolster which may be fixed or movable along the stem. See, e.g., U.S. Patent No. 6,387,114. The hemostatic device may be an endoscopic clip composed of deformable material and a tissue-penetrating pair of hollow jaws. See, e.g., U.S. Patent No. 5,989,268.
In another aspect, the Gl device may be a means to clear blocked Gl tracts. For example, the Gl device may be a dilation catheter composed of a shroud tube having a strain relief tube extending from within which is used to alter the configuration of a dilation balloon. See, e.g., U.S. Patent No. 6,537,247.
In another aspect, the Gl device may function to deliver drug to the Gl tract. For example, the Gl device may be orally administered and composed of a two-chambered water-permeable body, in which one chamber has an orifice for expelling a liquid drug when under pressure, and the second chamber contains an electric circuit that generates a gas which compresses the first chamber to expel the drug. See, e.g., U.S. Patent No. 5,925,030. The Gl device may be a collapsible, ellipsoidal gastric anchor with a tether and a long, narrow intestinal payload module, which contains slow release medicaments, bound enzymes or nonpathogenic microorganisms. See, e.g., U.S. Patent No. 4,878,905. The Gl device may be an ingestible device for delivering a substance to a chosen site within the Gl tract, which includes a receiver of electromagnetic radiation for powering an openable part of the device for inserting or dispensing the substance. See, e.g., U.S. Patent No. 6,632,216.
In another aspect, the Gl device may be a shunting device used to provide communication between two bodily systems. Shunting devices may be used to treat abnormal conditions, such as bypassing occlusions in a body passageway or transferring unwanted accumulation of fluids from a body cavity to a site where it can be processed by the body. For example, a shunting device may be used to displace peritoneal cavity fluid into the systemic venous circulation as a treatment for ascites. Shunting devices may include, without limitation, portosystemic shunts and peritoneovenous shunts. For example, the shunt may be an implantable pump composed of a cylindrical chamber and port with pumping means for aspirating fluid and expelling fluids. See, e.g., U.S. Patent No. 4,725,207. The shunt may be an implantable peritoneovenous shunt system composed of a double-chambered ascites collection device, a pump (e.g., magnetically driven or compression driven), and an anti-reflux catheter, that are all connected by flexible tubing. See, e.g., U.S. Patent No. 4,657,530 and 4,610,658. The shunt may be composed of a peritoneal tube connected to a hollow plastic implanted valve assembly that passes fluid when under pressure to a venous tube. See, e.g., U.S. Patent No. 5,520,632. The shunt may be a collapsible, shape-memory metal fabric with a plurality of woven metal strands having a central passageway for fluid and delivered in a collapsed state through a body channel to create a portosystemic shunt. See, e.g., U.S. Patent No. 6,468,303. The Gl device may be a laparoscopic tunneling dissector composed of an inflatable balloon and a hollow blunt tipped obturator which is used to tunnel through tissue to provide an anatomic working space for laparoscopic procedures. See, e.g., U.S. Patent Nos. 5,836,961 and 5,817,123.
Gl devices, which may be combined with one or more agents according to the present invention, include commercially available products.
In one aspect, Gl devices that are used for feeding purposes may include a variety of devices. For example, gastrostomy tubes such as the DURA-G Polyurethane Gastrostomy Tubes and MAGNA-PORT Gastrostomy Tubes are sold by Ross Products (Columbus, OH), a division of Abbott Laboratories. Moss Tubes, Inc. (West Sand Lake, NY) sells the MOSS G-Tube Percutaneous Endoscopic Gastrostomy Kits. Other enteral feeding tubes include, for example, EASY-FEED Enteral Feeding Sets which are sold by Ross Products (Columbus, OH), a division of Abbott Laboratories. COMPAT Enteral Delivery Systems are sold by Novartis AG (Basel, Switzerland). CORFLO Feeding Tubes are sold by VIASYS Healthcare Medsystems Division (Wheeling, IL). ENDOVIVE Enteral Feeding Systems are sold by Boston Scientific Corporation. Nasogastric tubes, such as the Mark IV Nasal (SIL) Tubes are sold by Moss Tubes, Inc. (West Sand Lake, NY). Bard Medical Division (Covington, Georgia) of CR. Bard, Inc. and Andersen Products Limited (England, United Kingdom) also sells a variety of Nasogastric Feeding Tubes. Low profile devices, such as the Low-Profile Replacement Gastrostomy Devices and the Bard Button Replacement Gastrostomy Devices are sold by Bard Endoscopic Technologies (Billerica, MA), a division of CR. Bard, Inc.
In another aspect, Gl devices may include gastrointestinal tubes for irrigation or aspiration, such as the LAVACUATOR Gastro Intestinal Tubes and VENTROL Levine Tubes, which are sold by Nellcor Puritan Bennett Inc. (Pleasanton, CA).
In another aspect, Gl devices may include those used as portosystemic shunts or other shunting devices, such as the VIATORR TIPS Endoprostheses that are sold by W.L. Gore & Associates, Inc. (Newark, DE). Denver Ascites Shunts are sold by Denver Biomedical, Inc. (Golden, CO). LEVEEN Shunts are sold by Becton, Dickinson and Company (Franklin Lakes, NJ).
In another aspect, Gl devices may include colostomy devices, such as ASSURA Pouches and COLOPLAST Pouches, which are sold by Coloplast Corporation (Marietta, GA). ESTEEM SYNERGY Standard Closed-End Pouches and SUR-FIT NATURA Closed-End Pouches are sold by ConvaTec (Princeton, NJ), a Bristol-Myers Squibb Company. Cymed Ostomy Company (Berkeley, CA) sells the MICROSKIN Colostomy Pouching Systems. KARAYA 5 One-Piece Pouching Systems, CONTOUR I One-Piece Ostomy Pouching Systems, and CENTERPOINTLOCK (CPL) Two-Piece Pouching Systems are sold by Hollister Inc. (Libertyville, IL). Bard Medical Division (Covington, Georgia) of CR. Bard, Inc. also sells a variety of Colostomy Pouches.
In another aspect, Gl devices may include dilatation catheters, such as the ELIMINATOR Multi-Stage Balloon Dilators, which are sold by Bard Endoscopic Technologies (Billerica, MA), a division of CR. Bard, Inc. CRE Fixed Wire and Wireguided Balloon Dilators are sold by Boston Scientific Corporation (Natick, MA).
In one aspect, the present invention provides Gl devices that include an anti-scarring agent or a composition that includes an anti-scarring agent. Numerous polymeric and non-polymeric delivery systems have been described above. These compositions can further comprise one or more fibrosis-inhibiting agents such that the overgrowth of granulation tissue is inhibited or reduced.
Methods for incorporating the fibrosis-inhibiting agent into or onto the device includes: (a) directly affixing to the device a fibrosis- inhibiting composition (e.g., by either a spraying process or dipping process as described above, with or without a carrier), (b) directly incorporating into the device a fibrosis-inhibiting composition (e.g., by either a spraying process or dipping process as described above, with or without a carrier, (c) by coating the device with a substance such as a hydrogel which will in turn absorb the fibrosis-inhibiting composition, (d) by interweaving fibrosis- inhibiting composition coated thread (or the polymer itself formed into a thread) into the device structure, (e) constructing the device itself or a portion of the device with a fibrosis-inhibiting composition, or (f) by covalently binding the fibrosis-inhibiting agent directly to the device surface or to a linker (small molecule or polymer) that is coated or attached to the device surface. The coatings can be applied to different portions of the device. For example, the coating can be (a) as a coating applied to the external surface of the tube; (b) as a coating applied to the internal (luminal) surface of the tube; (c) as a coating applied to the ends of the tube; and/or (d) as a coating applied to all or parts of both surfaces of the tube. In addition to coating the device with the fibrosis-inhibiting composition, the fibrosis-inhibiting agent can be mixed with the materials that are used to make the device such that the fibrosis-inhibiting agent is incorporated into the final device.
In addition to incorporation of a fibrosis-inhibiting agent into or onto the device, another biologically active agent can be incorporated into or onto the device, for example an anti-inflammatory (e.g., dexamethazone or aspirin), antithrombotic agents (e.g., heparin, heparin complexes, hydrophobic heparin derivatives, aspirin, or dipyridamole) and/or an antibiotic (e.g., amoxicillin, trimethoprim-sulfamethoxazole, azithromycin, clarithromycin, amoxicillin-clavulanate, cefprozil, cefuroxime, cefpodoxime, or cefdinir).
According to the present invention, any anti-scarring agent described above can be utilized in the practice of this embodiment. Within one embodiment of the invention, Gl devices may be adapted to release an agent that inhibits one or more of the four general components of the process of fibrosis (or scarring), including: formation of new blood vessels (angiogenesis), migration and proliferation of connective tissue cells (such as fibroblasts or smooth muscle cells), deposition of extracellular matrix (ECM), and remodeling (maturation and organization of the fibrous tissue). By inhibiting one or more of the components of fibrosis (or scarring), the overgrowth of granulation tissue may be inhibited or reduced.
Examples of fibrosis-inhibiting agents for use with Gl devices include the following: ZD-6474, AP-23573, synthadotin, S-0885, aplidine, ixabepilone, IDN-5390, SB-2723005, ABT-518, combretastatin, anecortave acetate, SB-715992, temsirolimus, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, as well as analogues and derivatives of the aforementioned.
Additional examples of anti-scarring agents which can be used include those having a high potency in the assays described herein (1- 10OnM IC50 range) such as isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, dolastatin 15, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17- DMAG, and tacrolimus; those having a mid-potency in the assays described herein (100-500 nM IC50 range) such as loteprednol etabonate, juglone, prednisolone, puromycin, 3-BAABE, cladribine, and mannose-6-phosphate; and those having a low potency in the assays described herein (500- 1000nm range IC50 range) such as 5-azacytidine, Ly333531(ruboxistaurin), and simvastatin.
As Gl devices are made in a variety of configurations and sizes, the exact dose administered will vary with device size, surface area and design. However, certain principles can be applied in the application of this art. Drug dose can be calculated as a function of dose per unit area (of the portion of the device being coated), total dose administered, and appropriate surface concentrations of active drug can be determined. Drugs are to be used at concentrations that range from several times more than to 10%, 5%, or even less than 1% of the concentration typically used in a single chemotherapeutic systemic dose application. Preferably, the drug is released in effective concentrations for a period ranging from 1 - 90 days.
Regardless of the method of application of the drug to the device, the exemplary anti-fibrosing agents, used alone or in combination, should be administered under the following dosing guidelines. The total amount (dose) of anti-scarring agent in or on the device may be in the range of about 0.01 μg-10 μg, or 10 μg-10 mg, or 10 mg-250 mg, or 250 mg-1000 mg, or 1000 mg-2500 mg. The dose (amount) of anti-scarring agent per unit area of device surface to which the agent is applied may be in the range of about 0.01 μg/mm2 - 1 μg/mm2, or 1 μg/mm2 - 10 μg/mm2, or 10 μg/mm2 - 250 μg/mm2, 250 μg/mm2 - 1000 μg/mm2, or 1000 μg/mm2 - 2500 μg/mm2.
Provided below are exemplary dosage ranges for various anti- scarring agents that can be used in conjunction with Gl devices in accordance with the invention. (A) Angiogenesis inhibitors including Alphastatin, ZD-6474, IDN-5390, SB-2723005, ABT-518, combretastatin, and anecortane, analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10^- 10"4 M of agent is to be maintained on the implant or barrier surface. (B) mTOR inhibitors including AP-23573 and Temsirolimus, analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10'8 - 10"4 M of agent is to be maintained on the implant or barrier surface. (C) Tubulin antagonists including synthadotin, analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10"8- 10"4 M of agent is to be maintained on the implant or barrier surface. (D) Epithilones including ixabepilone and analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10^- 10"4 M of agent is to be maintained on the implant or barrier surface. (E) Kinesin antagonists including SB- 715992 and analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10^- 10"4 M of agent is to be maintained on the implant or barrier surface. (F) TNF alpha antagonists including Etanercept, humicade, adalimumab and analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10"8- 10"4 M of agent is to be maintained on the implant or barrier surface. (G) AKT inhibitor including erucylphosphocholine and analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10^- 10"4 M of agent is to be maintained on the implant or barrier surface. (H) FGF Inhibitors including IDN-5390 and analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10"8- 10"4 M of agent is to be maintained on the implant or barrier surface. (I) Collagenase Antagonists including S-0885 and analogues and derivatives thereof: total dose not to exceed 1000 mg (range of 0.1 μg to 1000 mg); preferred 1 μg to 500 mg. Dose per unit area of 0.01 μg - 500 μg per mm2; preferred dose of 0.1 μg/mm2 - 100 μg/mm2. Minimum concentration of 10"8- 10~4 M of agent is to be maintained on the implant or barrier surface. (J) NF KAPPA B Inhibitors including Bortezomib and analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10~8- 10"4 M of agent is to be maintained on the implant or barrier surface. (K) Elongation factor-1 alpha inhibitors including aplidine and analogues and derivatives thereof: total dose not to exceed 1000 mg (range of 0.1 μg to 1000 mg); preferred 1 μg to 500 mg. Dose per unit area of 0.01 μg - 500 μg per mm2; preferred dose of 0.1 μg/mm2 - 100 μg/mm2. Minimum concentration of 10"8- 10'4 M of agent is to be maintained on the implant or barrier surface. (L) Tyrosine kinase inhibitors including Gefitinib and analogues and derivatives thereof: total dose not to exceed 1000 mg (range of 0.1 μg to 1000 mg); preferred 1 μg to 500 mg. Dose per unit area of 0.01 μg - 500 μg per mm2; preferred dose of 0.1 μg/mm2 - 100 μg/mm2. Minimum concentration of 10'8- 10"4 M of agent is to be maintained on the implant or barrier surface.
For high potency drugs, the total dose typically should not exceed 200 mg (range of 0.1 μg to 200 mg) and preferably 1 μg to 100 mg; dose per unit area of 0.01 μg - 100 μg per mm2; preferably 0.1 μg/mm2 - 20 μg/mm2; and minimum concentration of 10"8- 10'4 M of agent should be maintained on the implant or barrier surface. For mid-potency drugs, the total dose typically should not to exceed 500 mg (range of 1.0 μg to 500 mg) and preferably 1 μg to 200 mg; dose per unit area of 0.01 μg - 200 μg per mm2, preferably 0.1 μg/mm2 - 40 μg/mm2; and minimum concentration of 10" 8- 10"4 M of agent should be maintained on the implant or barrier surface. For low potency drugs, the total dose typically should not exceed 1000 mg (range of 0.1 μg to 1000 mg), preferably 1 μg to 500 mg; dose per unit area of 0.01 μg - 500 μg per mm2; preferably 0.1 μg/mm2 - 100 μg/mm2; and minimum concentration of 10"8- 10'4 M of agent should to be maintained on the implant or barrier surface.
Central Venous Catheters
In one aspect, the present invention provides for the combination of an anti-scarring agent and a central venous catheter (CVC) device. For the purposes of this invention, the term "Central Venous Catheters" should be understood to include any catheter or line that is used to deliver fluids to the large (central) veins of the body (e.g., jugular, pulmonary, femoral, iliac, inferior vena cava, superior vena cava, axillary etc.). CVC devices are generally hollow, tubular cannulae that are inserted into body passageways to permit injection or withdrawal of bodily fluids. CVCs may be inserted into a large vein, such as the superior vena cava, with a portion of the catheter disposed within the body and a connection port which extends out of the body for access to the circulatory system. CVCs may be used to administer drugs (e.g., chemotherapy or antibiotic therapy) or intravenous feeding, pressure monitoring or periodic blood sampling.
CVCs may be designed with or without a cuff or flange. Cuffs are used to prevent the catheter from slipping or becoming infected. CVCs may have one lumen or multiple lumens and range in many sizes to adapt to the required needs. They may be composed of synthetic materials, including, but not limited to, polyurethane, polyethylene, silicone, copolymers and other polymeric compositions.
CVCs are typically left in the body for a long period of time and thus, may develop infection or inflammation in response to the catheter. CVC access lumens may be blocked by clotted blood or thrombus formation. Some CVCs may also be available with coatings and treated surfaces to minimize the risk of infection and/or inflammation. The incorporation of a fibrosis-inhibiting agent into or onto the device can modulate an excessive fibroproliferative response to the device, which may prevent stenosis and/or obstruction of the device.
In one aspect, the CVC may be designed for specialized access to the circulatory system for specific conditions/purposes. For example, the CVC may be especially made for hemodialysis use by being elongated with a needle-like, dual lumen that may be used as a conduit for administering drugs or additives into the body through an AV access fistula or-graft. See, e.g., U.S. Patent No. 5,876,366. The CVC may be composed of an indwelling cannula adapted for placement within the superior vena cava having an exit port at the distal end whereby fluid medicament may be delivered to essentially the area of subcutaneous tissue surrounding the cannula. See, e.g., U.S. Patent No. 5,817,072.
In another aspect, the CVC may be designed to provide multiple conduits for accessing the circulatory system. For example, the CVC may be an elongated, integral flexible catheter tube with a plurality of independent lumens that may be adapted for attachment to a separate fluid conveying device whereby fluids may be separately infused into the vein without becoming mixed, and blood may be withdrawn and venous pressure monitored simultaneously with fluid infusion. See, e.g., U.S. 4,072,146. The CVC may be a multi-lumen catheter composed of a central flexible lumen with a formed fluid passageway and a plurality of collapsible lumens mounted around the periphery of the central lumen also having formed fluid passageways therein. See, e.g., U.S. Patent No. 4,406,656. In another aspect, the CVC may have a means for preventing infection as a result of long-term use. For example, the CVC may be composed of polyurethane with a thin hydrophilic layer on the surface loaded with an antibiotic of the ramoplanin group to inhibit bacterial colonization on the catheter after insertion. See, e.g., U.S. Patent No. 5,752,941. The CVC may be composed of a polymeric material that has an outer surface embedded by atoms of an antimicrobial metal (e.g., silver) that extend in a subsurface stratum to form a nonleaching surface treatment. See, e.g., U.S. Patent No. 5,520,664.
In another aspect, the CVC may be used with an apparatus that provides a means of controlling the injection or withdrawal of bodily fluids through the CVC. For example, the CVC apparatus may be composed of a syringe body with two barrels that have two separate fluid conduits with independent plungers and a valve body. See, e.g., U.S. Patent No. 5,411,485. The CVC apparatus may be composed of an upper and. lower molded sheets and a plurality of syringe channels and barrels that are individually operated by syringe plungers. See, e.g., U.S. Patent No. 5,417,667. The CVC apparatus may be an integrally molded base sheet which forms opposed slide valve walls that have a plurality of syringes mounted for fluid communication with the inlet ports. See, e.g., U.S. Patent No. 5,454,792. The CVC apparatus may be composed with access apparatus to provide easier accessibility by being composed of a connector that is in bi-directional fluid communication between a manifold and a CVC. See, e.g., U.S. Patent No. 5,308,322. The CVC apparatus may be a valve assembly that is provided for the distal end of a CVC for controlling fluid passage from the catheter to the blood flow passage in which it is inserted. See, e.g., U.S. Patent No. 5,030,210.
Other examples of central venous catheters include total parenteral nutrition catheters, peripherally inserted central venous catheters, flow-directed balloon-tipped pulmonary artery catheters, long-term central venous access catheters (such as Hickman lines and Broviac catheters). Representative examples of such catheters are described in U.S. Patent Nos. 3,995,623, 4,072,1464,096,860, 4,099,528, 4,134,402, 4,180,068, 4,385,631 , 4,406,656, 4,568,329, 4,960,409, 5,176,661 , 5,916,208.
CVCs, which may be combined with one or more agents according to the present invention, include commercially available products. For example, Bard Access Systems (Salt Lake City, UT) which is a division of CR. Bard sells the HICKMAN, BROVIAC and LEONARD Central Venous Catheters which are available with SureCuff tissue in-growth cuff and the VitaCuff Antimicrobial Cuff. Edward Lifesciences (Irvine, CA) sells the VANTEX Catheter as well as the PRESEP CENTRAL VENOUS OXIMETRY Catheter. Cook Critical Care (Bloomington, IN) sells the SPECTRUM Antibiotic Impregnated Catheters as well as other CVC sets and trays. Arrow International (Reading, PA) sells the ARROWGARD BLUE Catheters that have single or multiple lumens.
A variety of central venous catheters are available for use in hemodialysis including, but not restricted to, catheters which are totally implanted such as the Lifesite (Vasca Inc., Tewksbury, Mass.) and the Dialock (Biolink Corp., Middleboro, Mass.). Central venous catheters are prone to infection and embodiments for that purpose are described above.
In one aspect, the present invention provides CVC devices that include an anti-scarring agent or a composition that includes an anti- scarring agent. Numerous polymeric and non-polymeric delivery systems have been described above. These compositions can further comprise one or more fibrosis-inhibiting agents such that the overgrowth of granulation tissue is inhibited or reduced.
Methods for incorporating the fibrosis-inhibiting agent into or onto the device includes: (a) directly affixing to the device a fibrosis- inhibiting composition (e.g., by either a spraying process or dipping process as described above, with or without a carrier), (b) directly incorporating into the device a fibrosis-inhibiting composition (e.g., by either a spraying process or dipping process as described above, with or without a carrier, (c) by coating the device with a substance such as a hydrogel which will in turn absorb the fibrosis-inhibiting composition, (d) by interweaving fibrosis- inhibiting composition coated thread (or the polymer itself formed into a thread) into the device structure, (e) constructing the device itself or a portion of the device with a fibrosis-inhibiting composition, or (f) by covalently binding the fibrosis-inhibiting agent directly to the device surface or to a linker (small molecule or polymer) that is coated or attached to the device surface. The coatings can be applied to different portions of the device. For example, the coating can be (a) as a coating applied to the external surface of the tube; (b) as a coating applied to the internal (luminal) surface of the tube; (c) as a coating applied to the ends of the tube; and/or (d) as a coating applied to all or parts of both surfaces of the tube.
In addition to coating the device with the fibrosis-inhibiting composition, the fibrosis-inhibiting agent can be mixed with the materials that are used to make the device such that the fibrosis-inhibiting agent is incorporated into the final device.
In addition to incorporation of a fibrosis-inhibiting agent into or onto the device, another biologically active agent can be incorporated into or onto the device, for example an anti-inflammatory (e.g., dexamethazone or aspirin), antithrombotic agents (e.g., heparin, heparin complexes, hydrophobic heparin derivatives, aspirin, or dipyridamole) and/or an antibiotic (e.g., amoxicillin, trimethoprim-sulfamethoxazole, azithromycin, clarithromycin, amoxicillin-clavulanate, cefprozil, cefuroxime, cefpodoxime, or cefdinir).
According to the present invention, any anti-scarring agent described above can be utilized in the practice of this embodiment. Within one embodiment of the invention, CVC devices may be adapted to release an agent that inhibits one or more of the four general components of the process of fibrosis (or scarring), including: formation of new blood vessels (angiogenesis), migration and proliferation of connective tissue cells (such as fibroblasts or smooth muscle cells), deposition of extracellular matrix (ECM), and remodeling (maturation and organization of the fibrous tissue). By inhibiting one or more of the components of fibrosis (or scarring), the overgrowth of granulation tissue may be inhibited or reduced.
Examples of fibrosis-inhibiting agents for use in CVC devices include the following: ZD-6474, AP-23573, synthadotin, S-0885, aplidine, ixabepilone, IDN-5390, SB-2723005, ABT-518, combretastatin, anecortave acetate, SB-715992, temsirolimus, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, as well as analogues and derivatives of the aforementioned.
Additional examples of anti-scarring agents which can be used include those having a high potency in the assays described herein (1- 10OnM IC50 range) such as isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, dolastatin 15, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17- DMAG, and tacrolimus; those having a mid-potency in the assays described herein (100-500 nM IC50 range) such as loteprednol etabonate, juglone, prednisolone, puromycin, 3-BAABE, cladribine, and mannose-6-phosphate; and those having a low potency in the assays described herein (500- 1000nm range IC50 range) such as 5-azacytidine, Ly333531(ruboxistaurin), and simvastatin.
As CVC devices are made in a variety of configurations and sizes, the exact dose administered will vary with device size, surface area and design. However, certain principles can be applied in the application of this art. Drug dose can be calculated as a function of dose per unit area (of the portion of the device being coated), total dose administered, and appropriate surface concentrations of active drug can be determined. Drugs are to be used at concentrations that range from several times more than to 10%, 5%, or even less than 1% of the concentration typically used in a single chemotherapeutic systemic dose application. Preferably, the drug is released in effective concentrations for a period ranging from 1 - 90 days. Regardless of the method of application of the drug to the device, the exemplary anti-fibrosing agents, used alone or in combination, should be administered under the following dosing guidelines. The total amount (dose) of anti-scarring agent in or on the device may be in the range of about 0.01 μg-10 μg, or 10 μg-10 mg, or 10 mg-250 mg, or 250 mg-1000 mg, or 1000 mg-2500 mg. The dose (amount) of anti-scarring agent per unit area of device surface to which the agent is applied may be in the range of about 0.01 μg/mm2 - 1 μg/mm2, or 1 μg/mm2 - 10 μg/mm2, or 10 μg/mm2 - 250 μg/mm2, 250 μg/mm2 - 1000 μg/mm2, or 1000 μg/mm2 - 2500 μg/mm2.
Provided below are exemplary dosage ranges for various anti- scarring agents that can be used in conjunction with CVC devices in accordance with the invention. (A) Angiogenesis inhibitors including Alphastatin, ZD-6474, IDN-5390, SB-2723005, ABT-518, combretastatin, and anecortane, analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10^- 10"4 M of agent is to be maintained on the implant or barrier surface. (B) mTOR inhibitors including AP-23573 and Temsirolimus, analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10^- 10"4 M of agent is to be maintained on the implant or barrier surface. (C) Tubulin antagonists including synthadotin, analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10~8- 10"4 M of agent is to be maintained on the implant or barrier surface. (D) Epithilones including ixabepilone and analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 2006/013030
μg/mm . Minimum concentration of 10 - 10 M of agent is to be maintained on the implant or barrier surface. (E) Kinesin antagonists including SB- 715992 and analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10'8- 10'4 M of agent is to be maintained on the implant or barrier surface. (F) TNF alpha antagonists including Etanercept, humicade, adalimumab and analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10'8- 10~4 M of agent is to be maintained on the implant or barrier surface. (G) AKT inhibitor including erucylphosphocholine and analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10'8- 104 M of agent is to be maintained on the implant or barrier surface. (H) FGF Inhibitors including IDN-5390 and analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10"8- 10~4 M of agent is to be maintained on the implant or barrier surface. (I) Collagenase Antagonists including S-0885 and analogues and derivatives thereof: total dose not to exceed 1000 mg (range of 0.1 μg to 1000 mg); preferred 1 μg to 500 mg. Dose per unit area of 0.01 μg - 500 μg per mm2; preferred dose of 0.1 μg/mm2 - 100 μg/mm2. Minimum concentration of 10'8- 10"4 M of agent is to be maintained on the implant or barrier surface. (J) NF KAPPA B Inhibitors including Bortezomib dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10^- 10"4 M of agent is to be maintained on the implant or barrier surface. (K) Elongation factor-1 alpha inhibitors including aplidine and analogues and derivatives thereof: total dose not to exceed 1000 mg (range of 0.1 μg to 1000 mg); preferred 1 μg to 500 mg. Dose per unit area of 0.01 μg - 500 μg per mm2; preferred dose of 0.1 μg/mm2 - 100 μg/mm2. Minimum concentration of 10"8 - 10'4 M of agent is to be maintained on the implant or barrier surface. (L) Tyrosine kinase inhibitors including Gefitinib and analogues and derivatives thereof: total dose not to exceed 1000 mg (range of 0.1 μg to 1000 mg); preferred 1 μg to 500 mg. Dose per unit area of 0.01 μg - 500 μg per mm2; preferred dose of 0.1 μg/mm2 - 100 μg/mm2. Minimum concentration of 10'8 - 10'4 M of agent is to be maintained on the implant or barrier surface.
For high potency drugs, the total dose typically should not exceed 200 mg (range of 0.1 μg to 200 mg) and preferably 1 μg to 100 mg; dose per unit area of 0.01 μg - 100 μg per mm2; preferably 0.1 μg/mm2 - 20 μg/mm2; and minimum concentration of 10"8 - 104 M of agent should be maintained on the implant or barrier surface. For mid-potency drugs, the total dose typically should not to exceed 500 mg (range of 1.0 μg to 500 mg) and preferably 1 μg to 200 mg; dose per unit area of 0.01 μg - 200 μg per mm2, preferably 0.1 μg/mm2 - 40 μg/mm2; and minimum concentration of 10" 8 - 10"4 M of agent should be maintained on the implant or barrier surface. For low potency drugs, the total dose typically should not exceed 1000 mg (range of 0.1 μg to 1000 mg), preferably 1 μg to 500 mg; dose per unit area of 0.01 μg - 500 μg per mm2; preferably 0.1 μg/mm2 - 100 μg/mm2; and minimum concentration of 10"8- 10"4 M of agent should to be maintained on the implant or barrier surface.
Ventricular Assist Devices
In one aspect, the present invention provides for the combination of an anti-scarring agent and a ventricular assist device (VAD).
Ventrical assist devices are intended to assist the native heart in pumping blood throughout the body. Examples of VADs and other related 6 013030
devices include, without limitation, left ventricular assist devices, right ventricular assist devices, biventricular assist devices, cardiac assist devices, mechanical assist devices, artificial cardiac assist devices, implantable heart assist systems, implantable ventricular assist devices, heart assist pumps and intra-ventricular cardiac assist devices.
VADs are used to treat heart failure where the heart is incapable of pumping blood throughout the body at the rate needed to maintain adequate blood flow. Heart failure includes, without limitation, acute myocardial infarction, cardiomyopathy, cardiac valvular dysfunction, extensive cardiac surgery and uncontrolled cardiac arrhythmias. VADs assist the failing heart by increasing its pumping ability and allowing the heart to rest to recover its normal pumping function. In general, VADs are typically composed of a blood pump that is attached between the ventricle and aorta, cannulae that connect the pump to the heart, and a drive console that powers and controls the device. The most common VAD that exists is the left VAD because the left ventricle of the heart becomes diseased more often than the right ventricle; however, VADs may be used to pump blood from the left ventricle, right ventricle or both ventricles. VADs may be categorized by the pumping drives, which may function as either pulsatile (e.g., intra-aortic balloon pumps) or continuous, (e.g., reciprocating piston- type pumps or rotary pumps (centrifugal or axial impellers)).
VADs, however, may have medical complications associated with the implantation or prolonged use, such as, infections, septic emboli, hemorrhaging, inflammation as a reaction to tissue damage, and thrombosis induced by coagulation or blood stasis. These complications may obstruct the utility of the VAD and may lead to life threatening events. Incorporation of an anti-scarring agent into a VAD may prevent stenosis and/or obstruction of the device.
In one aspect, the VAD may be a pulsatile pump. These devices may have flexible sacks or diaphragms which are compressed and released to provide pulsatile pumping action. One type of pulsatile pump is the intra-aortic balloon pumps (IABP) which is a pulsatile sack device that may be implemented using minimally invasive procedures and are most functional when the left ventricle is able to eject blood to maintain a systemic arterial pressure. For example, the VAD may be an IABP that is a temporary, removable support within the aortic arch that descends through the aorta which has both a depressurized and pressurized position which is maintained by a pumping and blocking balloon. See, e.g., U.S. Patent No. 6,228,018. The VAD may be an IABP catheter and a pumping chamber having both a large and small diameter portions that are separated by a flexible diaphragm/membrane. See, e.g., U.S. Patent No. 5,928,132. The VAD may be a pulsatile pump composed of a cannula with an outer sheath and lumen, intake and outlet valves, fluid reservoir, and hydraulic pump that produces a pulsatile pumping action of blood through the cannula. See, e.g., U.S. Patent No. 6,007,479.
In another aspect, the VAD may be a continuous pump providing mostly steady flow of blood which may include an imperceptible pulsatile component. Continuous pumps may include reciprocating piston- type pumps, such as pneumatically powered devices or magnetically operated devices, and rotary pumps, such as centrifugal or axial impellors. For example, the VAD may be an implantable apparatus with a stator member and a magnetically suspended rotor member that act as a centrifugal pump where an impeller draws blood from the left ventricle and delivers it to the aorta thereby reducing the left ventricle pressure. See, e.g., U.S. Patent No. 5,928,131. The VAD may be composed of an implantable reciprocating piston for driving an implanted blood-pumping mechanism which is controlled by external electromagnets. See, e.g., U.S. Patent No. 5,089,017.
In another aspect, the VAD may be a device for assisting the pumping capacity of one of either the left or right ventricle. For example, the VAD may be composed of a housing apparatus with a pair of chambers with an inlet and outlet port, at least one ventricular outflow conduit, and an actuator that contracts one of the chambers while expanding the other to provide a positive displacement pump. See, e.g., U.S. Patent No. 6,264,601. The VAD may be composed of a pump, a chamber above the pump, and a tube that connects the pump and chamber using liquid and gas as a means for communication. See, e.g., U.S. Patent No. 6,146,325.
In another aspect, the VAD may be a device designed specifically for the left ventricle. For example, the VAD may be a blood pump adapted to be joined in flow communication between the left ventricle and the aorta using an inlet flow pressure sensor and a controller that may adjust speed of pump based on sensor feedback. See, e.g., U.S. Patent No. 6,623,420. The VAD may be composed of a bag adapted to expand by being filled with blood and able to contract to expel the blood, and the means for varying the resistance of the bag by using gaseous substance through a duet to a containing casing. See, e.g., U.S. Patent No. 6,569,079. The VAD may be a pump system composed of a deformable sac with inlet and outlet means and a pair of plates on opposite sides of the sac to deform the sac. See, e.g., U.S. Patent No. 5,599,173.
In another aspect, the VAD may be a device designed as a biventricular assist device. For example, the VAD may be a biventricular assist device composed of a self-supporting cup having an annular diaphragm that forms a fluid chamber around the heart cavity whereby it may have a pressure inlet/port that communicates with the fluid chamber to regulate positive and negative pressures. See, e.g., U.S. Patent No. 5,908,378; 5,749,839 and 5,738,627.
In another aspect, the VAD may be an implanted system used to supplement the pumping of blood circulation from a location outside the heart. For example, the VAD may be an extracardiac pumping system composed of an inflow and outflow conduit fluidly coupled to the pump (e.g., pulsatile or rotary pump) and a control circuit to synchronously actuate the pump. See, e.g., U.S. Patent Nos. 6,610,004; 6,428,464 and 6,200,260. 6 013030
In another aspect, the VAD related devices may be a used in conjunction with VADs or as stand alone to treat congestive heart failure victims. For example, a VAD related device may be a reinforcement device composed of a jacket that is applied to the heart to constrain cardiac expansion to a predetermined limit. See, e.g., U.S. Patent Nos. 6,582,355; 6,567,699; 6,241,654 and 6,169,922.
Representative examples of VADs, which may be combined with one or more agents according to the present invention, include commercially available products. For example, Thoratec Corporation (Pleasanton, CA) sells the HEARTMATE Left Ventricular Assist Systems. WorldHeart Corporation (ON, Canada) sells the WORLDHEART NOVACOR Left Ventricular Assist System. Arrow International (Reading, PA) sells the LIONHEART Left Ventricular Assist System.
In one aspect, the present invention provides LVAD devices that include an anti-scarring agent or a composition that includes an anti- scarring agent. Numerous polymeric and non-polymeric delivery systems have been described above. These compositions can further comprise one or more fibrosis-inhibiting agents such that the overgrowth of granulation tissue is inhibited or reduced.
Methods for incorporating the fibrosis-inhibiting agent into or onto the device includes: (a) directly affixing to the device a fibrosis- inhibiting composition (e.g., by either a spraying process or dipping process as described above, with or without a carrier), (b) directly incorporating into the device a fibrosis-inhibiting composition (e.g., by either a spraying process or dipping process as described above, with or without a carrier, (c) by coating the device with a substance such as a hydrogel which will in turn absorb the fibrosis-inhibiting composition, (d) by interweaving fibrosis- inhibiting composition coated thread (or the polymer itself formed into a thread) into the device structure, (e) constructing the device itself or a portion of the device with a fibrosis-inhibiting composition, or (T) by covalently binding the fibrosis-inhibiting agent directly to the device surface or to a linker (small molecule or polymer) that is coated or attached to the device surface. The coatings can be applied to different portions of the device. For example, the coating can be (a) as a coating applied to the external surface of the tube that leads out of the left ventricle; (b) as a coating applied to the internal (luminal) surface of the tube that leads out of the left ventricle; (c) as a coating applied to external surface of the tube that lead to the aorta; (d) as a coating applied to internal (luminal) surface of the tube that lead to the aorta; (e) as a coating that is applied to the ends of the tube where they are in contact with the heart tissue, and/or (f) as a coating applied to all or parts of the entire device.
In addition to coating the device with the fibrosis-inhibiting composition, the fibrosis-inhibiting agent can be mixed with the materials that are used to make the device such that the fibrosis-inhibiting agent is incorporated into the final device.
In addition to incorporation of a fibrosis-inhibiting agent into or onto the device, another biologically active agent can be incorporated into or onto the device, for example an anti-inflammatory (e.g., dexamethazone or aspirin), antithrombotic agents (e.g., heparin, heparin complexes, hydrophobic heparin derivatives, aspirin, or dipyridamole) and/or an antibiotic (e.g., amoxicillin, trimethoprim-sulfamethoxazole, azithromycin, clarithromycin, amoxicillin-clavulanate, cefprozil, cefuroxime, cefpodoxime, or cefdinir).
According to the present invention, any anti-scarring agent described above can be utilized in the practice of this embodiment. Within one embodiment of the invention, VAD devices (e.g., LVAD's) may be adapted to release an agent that inhibits one or more of the four general components of the process of fibrosis (or scarring), including: formation of new blood vessels (angiogenesis), migration and proliferation of connective tissue cells (such as fibroblasts or smooth muscle cells), deposition of extracellular matrix (ECM), and remodeling (maturation and organization of the fibrous tissue). By inhibiting one or more of the components of fibrosis (or scarring), the overgrowth of granulation tissue may be inhibited or reduced.
Examples of fibrosis-inhibiting agents for use in left ventricular assist devices include the following: ZD-6474, AP-23573, synthadotin, S- 0885, aplidine, ixabepilone, IDN-5390, SB-2723005, ABT-518, combretastatin, anecortave acetate, SB-715992, temsirolimus, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, as well as analogues and derivatives of the aforementioned.
Additional examples of anti-scarring agents which can be used include those having a high potency in the assays described herein (1- 10OnM IC50 range) such as isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, dolastatin 15, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17- DMAG, and tacrolimus; those having a mid-potency in the assays described herein (100-500 nM IC5O range) such as loteprednol etabonate, juglone, prednisolone, puromycin, 3-BAABE, cladribine, and mannose-6-phosphate; and those having a low potency in the assays described herein (500- 1000nm range IC50 range) such as 5-azacytidine, Ly333531(ruboxistaurin), and simvastatin.
As ventricular assist devices are made in a variety of configurations and sizes, the exact dose administered will vary with device size, surface area and design. However, certain principles can be applied in the application of this art. Drug dose can be calculated as a function of dose per unit area (of the portion of the device being coated), total dose administered, and appropriate surface concentrations of active drug can be determined. Drugs are to be used at concentrations that range from several times more than to 10%, 5%, or even less than 1% of the concentration typically used in a single chemotherapeutic systemic dose application. Preferably, the drug is released in effective concentrations for a period ranging from 1 - 90 days. Regardless of the method of application of the drug to the device, the exemplary anti-fibrosing agents, used alone or in combination, should be administered under the following dosing guidelines. The total amount (dose) of anti-scarring agent in or on the device may be in the range of about 0.01 μg-10 μg, or 10 μg-10 mg, or 10 mg-250 mg, or 250 mg-1000 mg, or 1000 mg-2500 mg. The dose (amount) of anti-scarring agent per unit area of device surface to which the agent is applied may be in the range of about 0.01 μg/mm2 - 1 μg/mm2, or 1 μg/mm2 - 10 μg/mm2, or 10 μg/mm2 - 250 μg/mm2, 250 μg/mm2 - 1000 μg/mm2, or 1000 μg/mm2 - 2500 μg/mm2.
Provided below are exemplary dosage ranges for various anti- scarring agents that can be used in conjunction with ventricular assist devices in accordance with the invention. (A) Angiogenesis inhibitors including Alphastatin, ZD-6474, IDN-5390, SB-2723005, ABT-518, combretastatin, and anecortane, analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10^- 10"4 M of agent is to be maintained on the implant or barrier surface. (B) mTOR inhibitors including AP-23573 and Temsirolimus, analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10"8- 10"4 M of agent is to be maintained on the implant or barrier surface. (C) Tubulin antagonists including synthadotin, analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10^- 10"4 M of agent is to be maintained on the implant or barrier surface. (D) Epithilones including ixabepilone and analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10"8- 10"4 M of agent is to be maintained on the implant or barrier surface. (E) Kinesin antagonists including SB- 715992 and analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10^- 10"4 M of agent is to be maintained on the implant or barrier surface. (F) TNF alpha antagonists including Etanercept, humicade, adalimumab and analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10"8- 10"4 M of agent is to be maintained on the implant or barrier surface. (G) AKT inhibitor including erucylphosphocholine and analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 -20 μg/mm2. Minimum concentration of 10^ - 10"4 M of agent is to be maintained on the implant or barrier surface. (H) FGF Inhibitors including IDN-5390 and analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10 s- 10"4 M of agent is to be maintained on the implant or barrier surface. (I) Collagenase Antagonists including S-0885 and analogues and derivatives thereof: total dose not to exceed 1000 mg (range of 0.1 μg to 1000 mg); preferred 1 μg to 500 mg. Dose per unit area of 0.01 μg - 500 μg per mm2; preferred dose of 0.1 μg/mm2 - 100 μg/mm2. Minimum concentration of 10~8- 10"4 M of agent is to be maintained on the implant or barrier surface. (J) NF KAPPA B Inhibitors including Bortezomib and analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10^- 10"4 M of agent is to be maintained on the implant or barrier surface. (K) Elongation factor- 1 alpha inhibitors including aplidine and analogues and derivatives thereof: total dose not to exceed 1000 mg (range of 0.1 μg to 1000 mg); preferred 1 μg to 500 mg. Dose per unit area of 0.01 μg - 500 μg per mm2; preferred dose of 0.1 μg/mm2 - 100 μg/mm2. Minimum concentration of 10"8- 10"4 M of agent is to be maintained on the implant or barrier surface. (L) Tyrosine kinase inhibitors including Gefitinib and analogues and derivatives thereof: total dose not to exceed 1000 mg (range of 0.1 μg to 1000 mg); preferred 1 μg to 500 mg. Dose per unit area of 0.01 μg - 500 μg per mm2; preferred dose of 0.1 μg/mm2 - 100 μg/mm2. Minimum concentration of 10"8- 10'4 M of agent is to be maintained on the implant or barrier surface.
For high potency drugs, the total dose typically should not exceed 200 mg (range of 0.1 μg to 200 mg) and preferably 1 μg to 100 mg; dose per unit area of 0.01 μg - 100 μg per mm2; preferably 0.1 μg/mm2 - 20 μg/mm2; and minimum concentration of 10^- 10"4 M of agent should be maintained on the implant or barrier surface. For mid-potency drugs, the total dose typically should not to exceed 500 mg (range of 1.0 μg to 500 mg) and preferably 1 μg to 200 mg; dose per unit area of 0.01 μg - 200 μg per mm2, preferably 0.1 μg/mm2 - 40 μg/mm2; and minimum concentration of 10" 8- 10"4 M of agent should be maintained on the implant or barrier surface. For low potency drugs, the total dose typically should not exceed 1000 mg (range of 0.1 μg to 1000 mg), preferably 1 μg to 500 mg; dose per unit area of 0.01 μg - 500 μg per mm2; preferably 0.1 μg/mm2 - 100 μg/mm2; and minimum concentration of 10"8 - 10"4 M of agent should to be maintained on the implant or barrier surface.
Spinal Implants
In one aspect, the present invention provides for the combination of an anti-scarring agent and a spinal implant (e.g., a spinal prosthesis). As used herein, the term "spinal prostheses" refers to devices that are located in, on, or near the spine and which enhance the ability of the spine to perform its function in the host. Spinal prostheses may be used to treat the vertebral column following degeneration or damage to the spine or a component or portion thereof. In healthy hosts, the vertebral column is composed of vertebral bone plates separated by intervertebral discs that form strong joints and absorb spinal compression. The intervertebral disc is comprised of an inner gel-like substance called the nucleus pulposus with surrounding tough fibrocartilagenous fibers called the annulus fibrosis. When damage occurs to the intervertebral disc, the host can develop spinal dysfunction, crippling pain, as well as long-term disability. Typically, damage to an intervertebral disc requires surgery which often results in the fusion of adjacent vertebral bone plates using various techniques and devices. Fusion of vertebral segments alleviates the pain by restricting vertebral motion at the damaged intervertebral disc. When only one vertebral segment is fused, the host will not have any noticeable motion limitations. However, when two or more segments are fused, the normal motion of the backmay become limited and thus, pain relief may not resolve due to the additional stress that is induced across the remaining vertebral joints.
In one aspect, the damaged vertebral segment may be treated using a spinal prosthesis that induces fusion between the vertebral plates. This may be conducted when only one vertebral segment is damaged. In another aspect, the damaged vertebral segment may be treated using a spinal prosthesis that maintains vertebral movement within the vertebral joint. This may be conducted when damage to more than one vertebral segment occurs.
Examples of spinal prostheses include, without limitation, spinal discs and related devices including vertebral implants, vertebral disc prostheses, lumbar disc implants, cervical disc implants, intervertebral discs, implantable prostheses, spinal prostheses, artificial discs, prosthetic implants, prosthetic spinal discs, spinal disc endoprostheses, spinal implants, artificial spinal discs, intervertebral implants, implantable spinal grafts, implantable bone grafts, artificial lumbar discs, spinal nucleus implants, and intervertebral disc spacers. Also included within the term spinal prostheses are fusion cages and related devices including fusion baskets, fusion cage apparatus, interbody cages, interbody implants, fusion devices, fusion cage anchoring devices, bone fixation apparatus, bone fixation instrumentation, bone fixation devices, fusion stabilization chamber, fusion cage anchoring plates, anchoring bone plates and bone screws.
A spinal prosthesis according to the present invention may be composed of a single material or a variety of materials including, without limitation, allograft bone material (see, e.g., U.S. 6,143,033), metals (see, e.g., U.S. 4,955,908), and/or synthetic materials (see, e.g., U.S. 6,264,695, 6,419,706, 5,824,093 and 4,911 ,718). The prosthesis must be biocompatible. It may consist of biodegradable or non-biodegradable components depending on the intended function of the device. See, e.g., U.S. 4,772,287. The spinal prosthesis may be biologically inert and serve as a mechanical means of stabilizing the. vertebral column (see, e.g., U.S. 4,955,908 and 5,716,415) or it may be biologically active and serve to promote fusion with the adjacent vertebral bone plates (see, e.g., U.S. 5,489,308 and 6,520,993).
In one aspect, the prosthesis may be a fusion cage designed to promote vertebral fusion in order to limit movement between adjacent vertebrae. Fusion cages may be interbody devices that fit within the intervertebral space or they may encompass both the intervertebral space and the anterior region of the vertebral column. Fusion cages may have various shapes. For example, fusion cages may be have a rectangular shape or may be cylindrical in shape and may have a plurality of openings and helical threading. Fusion cages may have an outer body and a hollow cavity that may or may not be used to insert bone growth-promoting material for stimulating bone fusion. For example, the prosthesis may be an interbody fusion cage that has an externally threaded stem projecting from a domed outer end which is fixed using an assembly of a plate, a fastener and bone screws. See, e.g., U.S. 6,156,037. The prosthesis may be a fusion cage with a threaded outer surface adapted for promoting fusion with bone structures when a bone-growth-inducing substance is packed into the cage body. See, e.g., U.S.4,961 ,740, 5,015,247, 4,878,915 and 4,501 ,269. The prosthesis may be a generally tubular shell with a helical thread projecting with a plurality of pillars with holes to facilitate bone ingrowth and mechanical anchoring. See, e.g., U.S. 6,071,310 and 5,489,308. Other U.S. patents that describe the threaded spinal implant include U.S. Patent numbers 5,263,953, 5,458,638 and 5,026,373.
In another aspect, the prosthesis may be a bone fixation device designed to promote vertebral fusion in order to limit movement between adjacent vertebrae. For example, bone dowels, rods, hooks, wires, wedges, plates, screws and other components may be used to fix the vertebra) segments into place. The fixation device may fit within the intervertebral space or it may encompass both the intervertebral space and the anterior region of the vertebral column or it may only encompass the anterior region of the vertebral column. A bone fixation device may be used with a fusion cage to assist in stabilizing the device within the intervertebral area. For example, the prosthesis may be in the form of a solid annular body having a plurality of discrete bone-engaging teeth protruding on the superior and inferior surfaces and having a central opening that may be filled with a bone growth-promoting material. See, e.g., U.S. 6,520,993. The prosthesis may have a disk-like body with weld-like raised parts disposed on opposite surfaces to enhance lateral stability in situ. See, e.g., U.S. 4,917,704. The prosthesis may be composed of opposite end pieces that maintain the height of the intervertebral space with an integral central element that is smaller in diameter wherein osteogenic material is disposed within the annular pocket between the end pieces. See, e.g., U.S. 6,146,420. The prosthesis may be composed of first and second side surfaces extending parallel to each other with upper and lower surfaces that engage the adjacent vertebrae. See, e.g., U.S. 5,716,415. The prosthesis may be a fusion stabilization chamber composed of a hollow intervertebral spacer and an end portion with at least one hole for affixing into the surrounding bone. See, e.g., U.S. 6,066,175. The prosthesis may be composed of a metallic body tapering conically from the ventral to the dorsal end and having a plurality of fishplates extending from opposite sides with openings for bone screws. See, e.g., U.S. 4,955,908. The prosthesis may be composed of a pair of plates which may have protrusions for engaging the adjacent vertebrae and an alignment device disposed between the engaging plates for separating the plates to maintain them in lordotic alignment. See, e.g., U.S. 6,576,016. The prosthesis may be a plurality of implants that are inserted side by side into the disc space that promote bone fusion across an intervertebral space. See, e.g., U.S. 5,522,899. The prosthesis may be an anchoring device composed of an anchoring plate with a central portion configured for attachment to a vertebral implant (e.g., fusion cage) and the end portions adapted to fasten in a fixed manner to a bony segment of the vertebra. See, e.g., U.S. 6,306,170. The prosthesis may be a bone fixation apparatus composed of a bone plate and a fastener apparatus (e.g., bone screws). See, e.g., U.S. 6,342,055, 6,454,769, 6,602,257 and 6,620,163.
In another aspect, the prosthesis may be an alternative to spinal fusion. The prosthesis may be a disc designed to provide normal movement between vertebral bone plates. The disc may be intended to mimic the natural shock absorbent function of the natural disc. The disc may be composed of a center core and end elements that support the disc against the adjacent vertebra or it may be intended to replace only a portion of the natural intervertebral disc (e.g., nucleus pulposus). For example, the disc may be in the form of an elastomeric section sandwiched between two rigid plates. See, e.g., U.S. 6,162,252; 5,534,030, 5,017,437 and 5,031 ,437. The disc may be an elongated prosthetic disc nucleus composed of a hydrogel core and a constraining flexible jacket that allows the core to deform and reform. See, e.g., U.S. 5,824,093. The disc may be composed of a rigid superior and inferior concaval-convex elements and a nuclear body which is located between the concave surfaces to permit movement. See, e.g., U.S. 6,156,067. The disc may be a partial spinal prosthesis composed of a core made of an elastic material such as silicone polymer or an elastomer which is covered by a casing made of a rigid material which is in contact with the adjacent vertebrae. See, e.g., U.S. 6,419,706. The disc may replace only the nucleus pulposus tissue by using a spinal nucleus implant comprised of a swellable, biomimetic plastic with a hydrophobic and hydrophilic phase which can be expanded in situ to conform to the natural size and shape. See, e.g., U.S. 6,264,695. The disc may be composed of a central core formed from a biocompatible elastomer wrapped by multi-layered laminae made from elastomer and fibers. See, e.g., U.S. 4,911 ,718. The disc may be composed of a fluid-filled inner bladder with an outer layer of strong, inert fibers intermingled with a bioresorbable material which promotes tissue ingrowth. See, e.g., U.S. 4,772, 287.
In another aspect, the spinal implant may be a device that reduces spine compression or reduces adhesions that may form as a result to spinal surgery and/or trauma. For example, the device may be a protection device composed of a shield to fit onto at least one lamina on the posterior surface to prevent postoperative formation of adhesions to the spinal dura. See, e.g., U.S. Patent Nos. 5,437,672 and 5,868,745 and U.S. Patent Application No. 2003/0078588. The device may be a prosthesis having a patch flange and a suture flange extending circumferentially around the patch such that the tissue underlying the patch is shielded and effectively nonadhesive to scar growth. See, e.g., U.S. Patent No. 5,634,944. The device may be a protective intervening barrier composed of a biocompatible shield which is used following intraspinal or vertebral surgery to prevent postoperative adhesions from binding onto the spinal nerves. See, e.g., U.S. Patent No. 4,013,078. The device may be used for neuro decompression while reducing fibroplasia proximate to the nerve tissue by having a surface topography texturized with outwardly-extending microstructures. See, e.g., U.S. Patent No. 6,106,558 and U.S. Patent Application No. 2003/0078673.
Spinal prostheses and other spinal implants, which may be combined with one or more drugs according to the present invention, include commercially available products. Medtronic Sofamor Danek (Memphis, TN) sells the fusion cage product INTERFIX Threaded Fusion Device. Centerpulse Spine-Tech (Minneapolis, MN) sells the BAK/C Cervical Interbody Fusion System fusion cage product and the CERVI-LOK Cervical Fixation System fixation device. Spinal Concepts (Austin, TX) sells the SC-ACUFIX Anterior Cervical Plate System. DePuy Spine, Inc. (Raynham, MA) sells the spinal discs, ACROFLEX TDR prostheses and the CHARITE Artificial Disc. Synthes-Stratec (Switzerland) sells the PRODISC system, including the PRODISC Cervical-C IDE disc replacement. Raymedica, Inc. (Minneapolis, MN) sells the PDN (PROSTHETIC DISC NUCLEUS).
Numerous polymeric and non-polymeric carrier systems that can be used in conjunction with spinal implants have been described above. Incorporation of a fibrosis-inhibiting agent into or onto a spinal implant can minimize fibrosis (or scarring) in the vicinity of the implant and may reduce or prevent the formation of adhesions between the implant and the surrounding tissue.
In one aspect, the present invention provides spinal implants that include an anti-scarring agent or a composition that includes an anti- scarring agent to inhibit scarring and adhesion between the device and the surrounding bone.
Methods for incorporating the anti-fibrosing compositions onto or into a spinal implant include: (a) directly affixing to the device an anti- fibrosing composition (e.g., by either a spraying process or dipping process as described above, with or without a carrier, (b) directly incorporating into the device an anti-fibrosing composition (e.g., by either a spraying process or clipping process as described above, with or without a carrier, (c) by coating the device with a substance such as a hydrogel which will in turn absorb the anti-fibrosing composition, (d) by interweaving anti-fibrosing composition coated thread (or the polymer itself formed into a thread) into the device structure, (e) by binding film or mesh which is comprised of or coated with an anti-fibrosing composition to the spinal prosthesis, (f) constructing the device itself or a portion of the device with an anti-fibrosing composition, or (g) by covalently binding the anti-fibrosing agent directly to the device surface or to a linker (small molecule or polymer) that is coated or attached to the device surface. For these devices, the coating process can be performed in such a manner as to a) coat the exterior surfaces of the device, b) coat the interior surfaces of the device or c) coat all or parts of both external and internal surface of the device.
In one aspect, a spinal implant (e.g., an implantable cages or disc) is coated with an anti-scarring agent or a composition that includes the anti-scarring agent. In certain aspects, the spina! implant may be coated with (or adapted to contain) an anti-scarring agent on one part of the device and a fibrosis-inducing agent (e.g., silk or talc) on another part of the device. For example, the outer surface of the implant (e.g., a vertebral implant) may be coated with a fibrosis-inducing agent to improve adhesion between the device and the surrounding tissue, while the interior of the device may be coated with an anti-scarring agent to minimize adhesion of tissue to the interior of the implant. Examples of fibrosis-inducing agents and methods of using fibrosis-inducing agents in combination with spinal implants are described in co-pending application entitled, "Medical Implants and Fibrosis- inducing Agents," filed November 20, 2003 (U.S. Ser. No. 60/524,023) and June 9, 2004 (U.S. Ser. No. 60/578,471).
In addition to coating the device with the anti-fibrosing composition, the anti-fibrosing agent can be mixed with the materials that are used to make the device such that the anti-fibrosing agent is incorporated into the final device. In addition to applying the fibrosis agent to the spinal implant, an in situ forming composition, gel or thermogel composition that further comprises a fibrosis-inhibiting agent can be applied to the placement site of the spinal prosthesis, (a) prior to placement of the prosthesis, (b) after placement of the prosthesis and/or (c) both prior and post placement on the prosthesis.
For the in situ forming, thermogel and gel compositions, the fibrosis-inhibiting agents can be incorporated directly into the formulation to produced a suspension or1 a solution or it can be incorporated into a secondary carrier (e.g., micelles, liposomes, microspheres, microparticles, nanospheres, microparticulates, emulsions and/or microemulations) that is then incorporated into the in situ forming compositions. In another embodiment, the fibrosis-inhibiting agent can be electrostatically or covalently bound to one or more of the polymeric components of the in situ forming composition.
In another embodiment, the fibrosis-inhibiting agent can be incorporated into a biodegradable or dissolvable film or mesh that is then applied to the treatment site prior or post implantation of the prosthesis/implant. Preferred materials for the manufacture of these films or meshes are hyaluronic acid (crosslinked or non-crosslinked), cellulose derivatives (e.g., hydroxypropyl cellulose), PLGA, POLYACTIVE, collagen and crosslinked poly(ethylene glycol).
In another embodiment, a solution or suspension that further comprises a fibrosis-inhibiting agent can be applied to the placement site of the spinal prosthesis, (a) prior to placement of the prosthesis, (b) after placement of the prosthesis and/or (c) both prior and post placement on the prosthesis. The fibrosis-inhibiting agents can be incorporated directly into the formulation to produced a suspension or a solution or it can be incorporated into a secondary carrier (e.g., micelles, liposomes, microspheres, microparticles, nanospheres, microparticulates, emulsions and/or microemulations) that is then incorporated into the in situ forming compositions. This solution or suspension can be applied (sprayed, rubbed, dripped etc) onto the treatment are prior to or post prosthesis placement.
In addition to incorporation of a fibrosis-inhibiting agent into or onto the device, another biologically active agent can be incorporated into or onto the device, for example an anti-inflammatory (e.g., dexamethazone or aspirin), antithrombotic agent (e.g., heparin, heparin complexes, hydrophobic heparin derivatives, aspirin, or dipyridamole) and/or an antibiotic (e.g., amoxicillin, trimethoprim-sulfamethoxazole, azithromycin, clarithromycin, amoxicillin-clavulanate, cefprozil, cefuroxime, cefpodoxime, or cefdinir).
According to the present invention, any adhesion or fibrosis- inducing agent described above can be utilized in the practice of this embodiment. Within one embodiment of the invention, spinal implants may be adapted to release an agent that inhibits one or more of the four general components of the process of fibrosis (or scarring), including: formation of new blood vessels (angiogenesis), migration and proliferation of connective tissue cells (such as fibroblasts or smooth muscle cells), deposition of extracellular matrix (ECM), and remodeling (maturation and organization of the fibrous tissue). By inhibiting one or more of the components of fibrosis (or scarring), the overgrowth of granulation tissue may be inhibited or reduced.
Examples of fibrosis-inhibiting agents for use in spinal implants include the following: ZD-6474, AP-23573, synthadotin, S-0885, aplidine, ixabepilone, IDN-5390, SB-2723005, ABT-518, combretastatin, anecortave acetate, SB-715992, temsirolimus, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, as well as analogues and derivatives of the aforementioned.
Additional examples of anti-scarring agents which can be used include those having a high potency in the assays described herein (1- 10OnM IC50 range) such as isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, dolastatin 15, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17- DMAG, and tacrolimus; those having a mid-potency in the assays described herein (100-500 nM IC50 range) such as loteprednol etabonate, juglone, prednisolone, puromycin, 3-BAABE, cladribine, and mannose-6-phosphate; and those having a low potency in the assays described herein (500- 1000nm range IC5O range) such as 5-azacytidine, Ly333531(ruboxistaurin), and simvastatin.
As spinal implants are made in a variety of configurations and sizes, the exact dose administered will vary with device size, surface area and design. However, certain principles can be applied in the application of this art. Drug dose can be calculated as a function of dose per unit area (of the portion of the device being coated), total dose administered, and appropriate surface concentrations of active drug can be determined. Drugs are to be used at concentrations that range from several times more than to 10%, 5%, or even less than 1% of the concentration typically used in a single chemotherapeutic systemic dose application. Preferably, the drug is released in effective concentrations for a period ranging from 1 - 90 days.
Regardless of the method of application of the drug to the device, the exemplary anti-fibrosing agents, used alone or in combination, should be administered under the following dosing guidelines. The total amount (dose) of anti-scarring agent in or on the device may be in the range of about 0.01 μg-10 μg, or 10 μg-10 mg, or 10 mg-250 mg, or 250 mg-1000 mg, or 1000 mg-2500 mg. The dose (amount) of anti-scarring agent per unit area of device surface to which the agent is applied may be in the range of about 0.01 μg/mm2 - 1 μg/mm2, or 1 μg/mm2 - 10 μg/mm2, or 10 μg/mm2 - 250 μg/mm2, 250 μg/mm2 - 1000 μg/mm2, or 1000 μg/mm2 - 2500 μg/mm2.
Provided below are exemplary dosage ranges for various anti- scarring agents that can be used in conjunction with spinal implants and devices in accordance with the invention. (A) Angiogenesis inhibitors including Alphastatin, ZD-6474, IDN-5390, SB-2723005, ABT-518, combretastatin, and anecortane, analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10^- 10"4 M of agent is to be maintained on the implant or barrier surface. (B) mTOR inhibitors including AP-23573 and Temsirolimus, analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10"8- 10"4 M of agent is to be maintained on the implant or barrier surface. (C) Tubulin antagonists including synthadotin, analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10"8- 10"4 M of agent is to be maintained on the implant or barrier surface. (D) Epithilones including ixabepilone and analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10"8- 10"4 M of agent is to be maintained on the implant or barrier surface. (E) Kinesin antagonists including SB- 715992 and analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10"8- 10"4 M of agent is to be maintained on the implant or barrier surface. (F) TNF alpha antagonists including Etanercept, humicade, adalimumab and analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10^- 10"4 M of agent is to be maintained on the implant or barrier surface. (G) AKT inhibitor including erucylphosphocholine and analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10^- 10"4 M of agent is to be maintained on the implant or barrier surface. (H) FGF Inhibitors including IDN-5390 and analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10"8 - 10'4 M of agent is to be maintained on the implant or barrier surface. (I) Collagenase Antagonists including S-0885 and analogues and derivatives thereof: total dose not to exceed 1000 mg (range of 0.1 μg to 1000 mg); preferred 1 μg to 500 mg. Dose per unit area of 0.01 μg - 500 μg per mm2; preferred dose of 0.1 μg/mm2 - 100 μg/mm2. Minimum concentration of 10"8- 10'4 M of agent is to be maintained on the implant or barrier surface. (J) NF KAPPA B Inhibitors including Bortezomib and analogues and derivatives thereof: total dose not to exceed 200 mg (range of 0.1 μg to 200 mg); preferred 1 μg to 100 mg. Dose per unit area of 0.01 μg - 100 μg per mm2; preferred dose of 0.1 μg/mm2 - 20 μg/mm2. Minimum concentration of 10~8- 10'4 M of agent is to be maintained on the implant or barrier surface. (K) Elongation factor-1 alpha inhibitors including aplidine and analogues and derivatives thereof: total dose not to exceed 1000 mg (range of 0.1 μg to 1000 mg); preferred 1 μg to 500 mg. Dose per unit area of 0.01 μg - 500 μg per mm2; preferred dose of 0.1 μg/mm2 - 100 μg/mm2. Minimum concentration of 10~8- 10"4 M of agent is to be maintained on the implant or barrier surface. (L) Tyrosine kinase inhibitors including Gefitinib and analogues and derivatives thereof: total dose not to exceed 1000 mg (range of 0.1 μg to 1000 mg); preferred 1 μg to 500 mg. Dose per unit area of 0.01 μg - 500 μg per mm2; preferred dose of 0.1 μg/mm2 - 100 μg/mm2. Minimum concentration of 10"8- 10"4 M of agent is to be maintained on the implant or barrier surface.
For high potency drugs, the total dose typically should not exceed 200 mg (range of 0.1 μg to 200 mg) and preferably 1 μg to 100 mg; dose per unit area of 0.01 μg - 100 μg per mm2; preferably 0.1 μg/mm2 - 20 μg/mm2; and minimum concentration of 10~8- 10~4 M of agent should be maintained on the implant or barrier surface. For mid-potency drugs, the total dose typically should not to exceed 500 mg (range of 1.0 μg to 500 mg) and preferably 1 μg to 200 mg; dose per unit area of 0.01 μg - 200 μg per mm2, preferably 0.1 μg/mm2 - 40 μg/mm2; and minimum concentration of 10" 8- 10"4 M of agent should be maintained on the implant or barrier surface. For low potency drugs, the total dose typically should not exceed 1000 mg (range of 0.1 μg to 1000 mg), preferably 1 μg to 500 mg; dose per unit area of 0.01 μg - 500 μg per mm2; preferably 0.1 μg/mm2 - 100 μg/mm2; and minimum concentration of 10"8- 10"4 M of agent should to be maintained on the implant or barrier surface.
(g) Infiltration of Therapeutic Agents or Pharmaceutical Compositions Around Medical Devices or Implants
Another use of the agents and compositions described herein may be to infiltrate the agent or composition into tissue adjacent to a medical device. The subject compositions may contain an anti-fibrotic and/or anti-infective agent.
The agents or compositions of the present invention may be infiltrated around implanted medical devices by applying the composition directly and/or indirectly into and/or onto (a) tissue adjacent to the medical device; (b) the vicinity of the medical device-tissue interface; (c) the region around the medical device; and (d) tissue surrounding the medical device. Methods for infiltrating the subject compositions into tissue adjacent to a medical device include delivering the composition: (a) to the medical device surface (e.g., as an injectable, paste, gel or mesh) during the implantation procedure; (b) to the surface of the tissue (e.g., as an injectable, paste, gel, in situ forming gel or mesh) immediately prior to, or during, implantation of the medical device; (c) to the surface of the medical device and/or the tissue surrounding the implanted medical device (e.g., as an injectable, paste, gel, in situ forming gel or mesh) immediately after the implantation of the medical device; (d) by topical application of the composition into the anatomical space where the medical device may be placed (particularly useful for this embodiment is the use of polymeric carriers which release the therapeutic agent over a period ranging from several hours to several weeks - fluids, suspensions, emulsions, microemulsions, microspheres, pastes, gels, microparticulates, sprays, aerosols, solid implants and other formulations which release the agent may be delivered into the region where the device may be inserted); (e) via percutaneous injection into the tissue surrounding the medical device as a solution as an infusate or as a sustained release preparation; (f) by any combination of the aforementioned methods. Combination therapies (e.g., combinations of therapeutic agents and combinations with antithrombotic and/or antiplatelet agents) may also be used. In all cases it is understood that the subject compositions may be infiltrated into tissue adjacent to all or a portion of the device.
Representative examples of compositions that may be infiltrated into tissue adjacent to a medical device include: (a) sprayable collagen-containing formulations such as COSTASIS (Angiotech Pharmaceuticals, Inc., Canada) and crosslinked poly(ethylene glycol) - methylated collagen compositions (described, e.g., in U.S. Patent Nos. 5,874,500 and 5,565,519), either alone, or loaded with a therapeutic agent (e.g., an anti-scarring and/or anti-infective agent), infiltrated into tissue adjacent to the medical device; (b) sprayable PEG-containing formulations such as COSEAL (Angiotech Pharmaceuticals, Inc.), FOCALSEAL (Genzyme Corporation, Cambridge, MA), SPRAYGEL or DURASEAL (both from Confluent Surgical, Inc., Boston, MA), either alone, or loaded with a therapeutic agent (e.g., an anti-scarring and/or anti-infective agent), infiltrated into tissue adjacent to the medical device; (c) fibrinogen- containing formulations such as FLOSEAL or TISSEAL (both from Baxter Healthcare Corporation, Fremont, CA), either alone, or loaded with a therapeutic agent (e.g., an anti-scarring and/or anti-infective agent), infiltrated into tissue adjacent to the medical device; (d) hyaluronic acid- containing formulations such as RESTYLANE or PERLANE (both from Q- Med AB, Sweden), HYLAFORM (Inamed Corporation, Santa Barbara, CA), SYNVISC (Biomatrix, Inc., Ridgefield, NJ)1 SEPRAFILM or SEPRACOAT (both from Genzyme Corporation), either alone, or loaded with a therapeutic agent (e.g., an anti-scarring and/or anti-infective agent), infiltrated into tissue adjacent to the medical device; (e) polymeric gels for surgical implantation such as REPEL (Life Medical Sciences, Inc., Princeton, NJ) or FLOWGEL (Baxter Healthcare Corporation), either alone, or loaded with a therapeutic agent (e.g., an anti-scarring and/or anti-infective agent), infiltrated into tissue adjacent to the medical device; (f) orthopedic "cements" used to hold prostheses and tissues in place, either alone, or loaded with a therapeutic agent (e.g., an anti-scarring and/or anti-infective agent), infiltrated into tissue adjacent to the medical device, such as OSTEOBOND (Zimmer, Inc., Warsaw, IN), low viscosity cement (LVC); Wright Medical Technology, Inc., Arlington, TN), SIMPLEX P (Stryker Corporation, Kalamazoo, Ml), PALACOS (Smith & Nephew Corporation, United Kingdom), and ENDURANCE (Johnson & Johnson, Inc., New Brunswick, NJ); (g) surgical adhesives containing cyanoacrylates such as DERMABOND (Johnson & Johnson, Inc.), INDERMIL (U.S. Surgical Company, Norwalk, CT), GLUSTITCH (Blacklock Medical Products Inc., Canada), TISSUEMEND (Veterinary Products Laboratories, Phoenix, AZ), VETBOND (3M Company, St. Paul, MN), HISTOACRYL BLUE (Davis & Geek, St. Louis, MO) and ORABASE SOOTHE-N-SEAL LIQUID PROTECTANT (Colgate-Palmolive Company, New York, NY), either alone, or loaded with a therapeutic agent (e.g., an anti-scarring and/or anti-infective agent), infiltrated into tissue adjacent to the medical device; (h) implants containing hydroxyapatite (or synthetic bone material such as calcium sulfate, VITOSS and CORTOSS (both from Orthovita, Inc., Malvern, PA), either alone, or loaded with a therapeutic agent (e.g., an anti-scarring and/or anti-infective agent), infiltrated into tissue adjacent to the medical device; (i) other biocompatible tissue fillers, such as those made by BioCure, Inc. (Norcross, GA), 3M Company (St. Paul, MN) and Neomend, Inc. (Sunnyvale, CA), either alone, or loaded with a therapeutic agent (e.g., an anti-scarring and/or anti-infective agent), infiltrated into tissue adjacent to the medical device; (j) polysacharride gels such as the ADCON series of gels (available from Gliatech, Inc., Cleveland, OH) either alone, or loaded with a therapeutic agent (e.g., an anti-scarring and/or anti-infective agent), infiltrated into tissue adjacent to the medical device; and/or (k) films, sponges or meshes such as INTERCEED (Gynecare Worldwide, a division of Ethicon, Inc., Somerville, NJ), VICRYL mesh (Ethicon, Inc.), and GELFOAM (Pfizer, Inc., New York, NY), either alone, or loaded with a therapeutic agent (e.g., an anti-scarring and/or anti-infective agent), infiltrated into tissue adjacent to the medical device.
Other examples of compositions that may be infiltrated into tissue adjacent to a medical device include compositions formed from reactants comprising either one or both of pentaerythritol poly(ethylene glycol)ether tetra-sulfhydryl] (4-armed thiol PEG, which includes structures having a linking group(s) between a sulfhydryl group(s) and the terminus of the polyethylene glycol backbone) and pentaerythritol poly(ethylene glycol)ether tetra-succinimidyl glutarate] (4-armed NHS PEG, which again includes structures having a linking group(s) between a NHS group(s) and the terminus of the polyethylene glycol backbone) as reactive reagents. Another preferred composition comprises either one or both of pentaerythritol poly(ethylene glycol)ether tetra-amino] (4-armed amino PEG, which includes structures having a linking group(s) between an amino group(s) and the terminus of the polyethylene glycol backbone) and pentaerythritol poly(ethylene glycol)ether tetra-succinimidyl glutarate] (4- armed NHS PEG, which again includes structures having a linking group(s) between a NHS group(s) and the terminus of the polyethylene glycol backbone) as reactive reagents. Chemical structures for these reactants are shown in, e.g., U.S. Patent 5,874,500. Optionally, collagen or a collagen derivative (e.g., methylated collagen) is added to the poly(ethylene glycol)-containing reactant(s) to form a preferred crosslinked matrix.
Representative examples of medical devices for use with the subject compositions are described below.
Intravascular Devices
In one aspect, the present invention provides therapeutic agents or pharmaceutical compositions that may be infiltrated into the tissue adjacent to the intravascular devices (e.g., anastomotic connectors, stents, drug-delivery balloons, intravascular catheters), where the polymeric composition may include a therapeutic agent (e.g., an anti-scarring or anti- infective agent). Examples of intravascular devices are provided above in conjunction with the coating of medical devices. Numerous agents or compositions for use with intravascular devices have been described above which may be infiltrated into the tissue adjacent to the device (preferably near the device-tissue interface).
Therapeutic agents or pharmaceutical compositions may be infiltrated around implanted intravascular devices by applying the composition directly and/or indirectly into and/or onto (a) tissue adjacent to the intravascular device; (b) the vicinity of the intravascular device-tissue interface; (c) the region around the intravascular device; and (d) tissue surrounding the intravascular device. Methods for infiltrating the subject agents or compositions into tissue adjacent to an intravascular device include delivering the agent or composition: (a) to the intravascular device surface (e.g., as an injectable, paste, gel or mesh) during the implantation procedure; (b) to the surface of the tissue (e.g., as an injectable, paste, gel, in situ forming gel or mesh) immediately prior to, or during, implantation of the intravascular device; (c) to the surface of the intravascular device and/or the tissue surrounding the implanted intravascular device (e.g., as an injectable, paste, gel, in situ forming gel or mesh) immediately after the implantation of the intravascular device; (d) by topical application of the composition into the anatomical space where the intravascular device may be placed (particularly useful for this embodiment is the use of polymeric carriers which release the therapeutic agent over a period ranging from several hours to several weeks - fluids, suspensions, emulsions, microemulsions, microspheres, pastes, gels, microparticulates, sprays, aerosols, solid implants and other formulations which release the agent may be delivered into the region where the intravascular device may be inserted); (e) via percutaneous injection into the tissue surrounding the intravascular device as a solution as an infusate or as a sustained release preparation; (f) by any combination of the aforementioned methods. Combination therapies (Ae., combinations of therapeutic agents and combinations with antithrombotic and/or antiplatelet agents) may also be used. In all cases it is understood that the subject agents or compositions may be infiltrated into tissue adjacent to all or a portion of the device.
According to one aspect, any fibrosis-inhibiting and/or anti- infective agent described above may be utilized in the practice of the present invention. In one aspect of the invention, the subject agents or compositions infiltrated into tissue adjacent to intravascular devices inhibit one or more of the four general components of the process of fibrosis (or scarring), including: formation of new blood vessels (angiogenesis), migration and proliferation of connective tissue cells (such as fibroblasts or smooth muscle cells), deposition of extracellular matrix (ECM), and remodeling (maturation and organization of the fibrous tissue). By inhibiting one or more of the components of fibrosis (or scarring), the overgrowth of granulation tissue may be inhibited or reduced.
Examples of fibrosis-inhibiting agents for use in the present invention include the following: ZD-6474, AP-23573, synthadotin, S-0885, aplidine, ixabepilone, IDN-5390, SB-2723005, ABT-518, combretastatin, anecortave acetate, SB-715992, temsirolimus, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, as well as analogues and derivatives of the aforementioned. Additional examples of anti-scarring agents which can be used include those having a high potency in the assays described herein (1- 10OnM IC5O range) such as isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, dolastatin 15, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17- DMAG1 and tacrolimus; those having a mid-potency in the assays described herein (100-500 nM IC50 range) such as loteprednol etabonate, juglone, prednisolone, puromycin, 3-BAABE, cladribine, and mannose-6-phosphate; and those having a low potency in the assays described herein (500- 1000nm range IC50 range) such as 5-azacytidine, Ly333531(ruboxistaurin), and simvastatin.
The drug dose administered from the present agents or compositions for prevention or inhibition of fibrosis in accordance with the present invention will depend on a variety of factors, including the type of formulation, the location of the treatment site, and the type of condition being treated. As intravascular devices are made in a variety of configurations and sizes, the exact dose administered will also vary with device size, surface area and design. However, certain principles can be applied in the application of this art. Drug dose can be calculated as a function of dose per unit area (of the treatment site), total drug dose administered can be measured and appropriate surface concentrations of active drug can be determined. Drugs are to be used at concentrations that range from several times more than to 50%, 20%, 10%, 5%, or even less than 1 % of the concentration typically used in a single chemotherapeutic systemic dose application. In certain aspects, the anti-scarring agent is released from the composition in effective concentrations in a time period that may be measured from the time of infiltration into tissue adjacent to the device, which ranges from about less than 1 day to about 180 days. Generally, the release time may also be from about less than 1 day to about 180 days; from about 7 days to about 14 days; from about 14 days to about 28 days; from about 28 days to about 56 days; from about 56 days to about 90 days; from about 90 days to about 180 days.
The exemplary anti-fibrosing agents, used alone or in combination, should be administered under the following dosing guidelines. The total amount (dose) of anti-scarring agent in the composition can be in the range of about 0.01 μg-10 μg, or about 10 μg-10 mg, or about 10 mg- 250 mg, or about 250 mg-1000 mg, or about 1000 mg-2500 mg. The dose (amount) of anti-scarring agent per unit area of device or tissue surface to which the agent is applied may be in the range of about 0.01 μg/mm2 - 1 μg/mm2, or about 1 μg/mm2 - 10 μg/mm2, or about 10 μg/mm2 - 250 μg/mm2, or about 250 μg/mm2 - 1000 μg/mm2, or about 1000 μg/mm2 - 2500 μg/mm2.
According to another aspect, any anti-infective agent described above may be used in the practice of the present invention. Exemplary anti-infective agents include (A) anthracyclines (e.g., doxorubicin and mitoxantrone), (B) fluoropyrimidines (e.g., 5-FU), (C) folic acid antagonists (e.g., methotrexate), (D) podophylotoxins (e.g., etoposide), (E) camptothecins, (F) hydroxyureas, and (G) platinum complexes (e.g., cisplatin), as well as analogues and derivatives of the aforementioned.
The drug dose administered from the present agents or compositions for prevention or inhibition of infection in accordance with the present invention will depend on a variety of factors, including the type of formulation, the location of the treatment site, and the type of condition being treated. However, certain principles can be applied in the application of this art. Drug dose can be calculated as a function of dose per unit area (of the treatment site), total drug dose administered can be measured and appropriate surface concentrations of active drug can be determined. Drugs are to be used at concentrations that range from several times more than to 50%, 20%, 10%, 5%, or even less than 1% of the concentration typically used in a single anti-infective systemic dose application. In certain aspects, the anti-infective agent is released from the composition in effective concentrations in a time period that may be measured from the time of infiltration into tissue adjacent to the device, which ranges from about less than 1 day to about 180 days. Generally, the release time may also be from about less than 1 day to about 180 days; from about 7 days to about 14 days; from about 14 days to about 28 days; from about 28 days to about 56 days; from about 56 days to about 90 days; from about 90 days to about 180 days.
The exemplary anti-infective agents, used alone or in combination, should be administered under the following dosing guidelines. The total amount (dose) of anti-infective agent in the composition can be in the range of about 0.01 μg-1 μg, or about 1 μg-10 μg, or about 10 μg-1 mg, or about 1 mg to 10 mg, or about 10 mg-100 mg, or about 100 mg to 250 mg, or about 250 mg-1000 mg. The dose (amount) of anti-infective agent per unit area of device or tissue surface to which the agent is applied may be in the range of about 0.01 μg/mm2 - 1 μg/mm2, or about 1 μg/mm2 - 10 μg/mm2, or about 10 μg/mm2 - 100 μg/mm?, or about 100 μg/mm2 to 250 μg/mm2, or about 250 μg/mm2 - 1000 μg/mm2. As different compositions will release the anti-infective agent at differing rates, the above dosing parameters should be utilized in combination with the release rate of the drug from the composition such that a minimum concentration of about 10"8 to 10"7, or about 10'7 to 10"6 about 10"6 to 10"5or about 10'5 to 10'4 Of the agent is maintained on the tissue surface.
It should be readily evident based upon the discussions provided herein that combinations of anthracyclines (e.g., doxorubicin or mitoxantrone), fluoropyrimidines (e.g., 5-fluorouracil), folic acid antagonists (e.g., methotrexate), quinolones, and/or podophylotoxins (e.g., etoposide) may be utilized to enhance the antibacterial activity of the composition.
Gastrointestinal Stents
In one aspect, the therapeutic agent or pharmaceutical compositions of the present invention may be infiltrated into tissue adjacent to a gastrointestinal (Gl) stent. The subject compositions may contain a therapeutic agent (e.g., an anti-scarring and/or anti-infective agent). Examples of gastrointestinal stents are provided above in conjunction with the coating of medical devices. Numerous agents or compositions for use with intravascular devices have been described above which may be infiltrated into the tissue adjacent to the device (preferably near the device- tissue interface).
Therapeutic agents or pharmaceutical compositions may be infiltrated around implanted Gl stents by applying the composition directly and/or indirectly into and/or onto (a) tissue adjacent to the Gl stent; (b) the vicinity of the Gl stent-tissue interface; (c) the region around the Gl stent; and (d) tissue surrounding the Gl stent. Methods for infiltrating the subject agents or compositions into tissue adjacent to a Gl stent include delivering the composition: (a) to the Gl stent surface (e.g., as an injectable, paste, gel or mesh) during the implantation procedure; (b) to the surface of the tissue (e.g., as an injectable, paste, gel, in situ forming gel or mesh) immediately prior to, or during, implantation of the Gl stent; (c) to the surface of the Gl stent and/or the tissue surrounding the implanted Gl stent (e.g., as an injectable, paste, gel, in situ forming gel or mesh) immediately after the implantation of the Gl stent; (d) by topical application of the composition into the anatomical space where the Gl stent may be placed (particularly useful for this embodiment is the use of polymeric carriers which release the therapeutic agent over a period ranging from several hours to several weeks - fluids, suspensions, emulsions, microemulsions, microspheres, pastes, gels, microparticulates, sprays, aerosols, solid implants and other formulations which release the agent may be delivered into the region where the device may be inserted); (e) via percutaneous injection into the tissue surrounding the Gl stent as a solution as an infusate or as a sustained release preparation; (f) by any combination of the aforementioned methods. Combination therapies (i.e., combinations of therapeutic agents and combinations with antithrombotic and/or antiplatelet agents) may also be used. In all cases it is understood that the subject agents or compositions may be infiltrated into tissue adjacent to all or a portion of the device.
According to one aspect, any fibrosis-inhibiting and/or anti- infective agent described above may be utilized in the practice of the present invention. In one aspect of the invention, the subject agents or compositions infiltrated into tissue adjacent to Gl stents may be adapted to release an agent that inhibits one or more of the four general components of the process of fibrosis (or scarring), including: formation of new blood vessels (angiogenesis), migration and proliferation of connective tissue cells (such as fibroblasts or smooth muscle cells), deposition of extracellular matrix (ECM), and remodeling (maturation and organization of the fibrous tissue). By inhibiting one or more of the components of fibrosis (or scarring), the overgrowth of granulation tissue may be inhibited or reduced.
Examples of fibrosis-inhibiting agents for use in the present invention include the following: ZD-6474, AP-23573, synthadotin, S-0885, aplidine, ixabepilone, IDN-5390, SB-2723005, ABT-518, cembretastatin, anecortave acetate, SB-715992, temsirolimus, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, as well as analogues and derivatives of the aforementioned.
Additional examples of anti-scarring agents which can be used include those having a high potency in the assays described herein (1- 10OnM IC50 range) such as isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, dolastatin 15, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17- DMAG, and tacrolimus; those having a mid-potency in the assays described herein (100-500 nM IC50 range) such as loteprednol etabonate, juglone, prednisolone, puromycin, 3-BAABE, cladribine, and mannose-6-phosphate; and those having a low potency in the assays described herein (500- 1000nm range IC50 range) such as 5-azacytidine, Ly333531(ruboxistaurin), and simvastatin. The drug dose administered from the present agents or compositions for prevention or inhibition of fibrosis in accordance with the present invention will depend on a variety of factors, including the type of formulation, the location of the treatment site, and the type of condition being treated. As Gl stents are made in a variety of configurations and sizes, the exact dose administered will also vary with device size, surface area and design. However, certain principles can be applied in the application of this art. Drug dose can be calculated as a function of dose per unit area (of the treatment site), total drug dose administered can be measured and appropriate surface concentrations of active drug can be determined. Drugs are to be used at concentrations that range from several times more than to 50%, 20%, 10%, 5%, or even less than 1% of the concentration typically used in a single chemotherapeutic systemic dose application. In certain aspects, the anti-scarring agent is released from the composition in effective concentrations in a time period that may be measured from the time of infiltration into tissue adjacent to the device, which ranges from about less than 1 day to about 180 days. Generally, the release time may also be from about less than 1 day to about 180 days; from about 7 days to about 14 days; from about 14 days to about 28 days; from about 28 days to about 56 days; from about 56 days to about 90 days; from about 90 days to about 180 days.
The exemplary anti-fibrosing agents, used alone or in combination, should be administered under the following dosing guidelines. The total amount (dose) of anti-scarring agent in the composition can be in the range of about 0.01 μg-10 μg, or about 10 μg-10 mg, or about 10 mg- 250 mg, or about 250 mg-1000 mg, or about 1000 mg-2500 mg. The dose (amount) of anti-scarring agent per unit area of device or tissue surface to which the agent is applied may be in the range of about 0.01 μg/mm2 - 1 μg/mm2, or about 1 μg/mm2 - 10 μg/mm2, or about 10 μg/mm2 - 250 μg/mm2, or about 250 μg/mm2 - 1000 μg/mm2, or about 1000 μg/mm2 - 2500 μg/mm2. According to another aspect, any anti-infective agent described above may be used in the practice of the present invention. Exemplary anti-infective agents include (A) anthracyclines (e.g., doxorubicin and mitoxantrone), (B) fluoropyrimidines (e.g., 5-FU), (C) folic acid antagonists (e.g., methotrexate), (D) podophylotoxins (e.g., etoposide), (E) camptothecins, (F) hydroxyureas, and (G) platinum complexes (e.g., cisplatin), as well as analogues and derivatives of the aforementioned.
The drug dose administered from the present agents or compositions for prevention or inhibition of infection in accordance with the present invention will depend on a variety of factors, including the type of formulation, the location of the treatment site, and the type of condition being treated. However, certain principles can be applied in the application of this art. Drug dose can be calculated as a function of dose per unit area (of the treatment site), total drug dose administered can be measured and appropriate surface concentrations of active drug can be determined. Drugs are to be used at concentrations that range from several times more than to 50%, 20%, 10%, 5%, or even less than 1% of the concentration typically used in a single anti-infective systemic dose application. In certain aspects, the anti-infective agent is released from the composition in effective concentrations in a time period that may be measured from the time of infiltration into tissue adjacent to the device, which ranges from about less than 1 day to about 180 days. Generally, the release time may also be from about less than 1 day to about 180 days; from about 7 days to about 14 days; from about 14 days to about 28 days; from about 28 days to about 56 days; from about 56 days to about 90 days; from about 90 days to about 180 days.
The exemplary anti-infective agents, used alone or in combination, should be administered under the following dosing guidelines. The total amount (dose) of anti-infective agent in the composition can be in the range of about 0.01 μg-1 μg, or about 1 μg-10 μg, or about 10 μg-1 mg, or about 1 mg to 10 mg, or about 10 mg-100 mg, or about 100 mg to 250 mg, or about 250 mg-1000 mg. The dose (amount) of anti-infective agent per unit area of device or tissue surface to which the agent is applied may be in the range of about 0.01 μg/mm2 - 1 μg/mm2, or about 1 μg/mm2 - 10 μg/mm2, or about 10 μg/mm2 - 100 μg/mm2, or about 100 μg/mm2 to 250 μg/mm2, or about 250 μg/mm2 - 1000 μg/mm2. As different compositions will release the anti-infective agent at differing rates, the above dosing parameters should be utilized in combination with the release rate of the drug from the composition such that a minimum concentration of about 10"8 to 10"7, or about 1O-7 to 106 about 10"6 to 10"5 or about 10"5 to 10"4 of the agent is maintained on the tissue surface.
It should be readily evident based upon the discussions provided herein that combinations of anthracyclines (e.g., doxorubicin or mitoxantrone), fluoropyrimidines (e.g., 5-fluorouracil), folic acid antagonists (e.g., methotrexate), quinolones, and/or podophylotoxins (e.g., etoposide) may be utilized to enhance the antibacterial activity of the composition.
Tracheal and Bronchial Stents
The present invention provides for infiltration of the subject agents or compositions into tissue adjacent to a tracheal or bronchial stent device. The subject compositions may contain a therapeutic agent (e.g., an anti-scarring and/or anti-infective agent). Representative examples of tracheal or bronchial stents that may benefit from having the subject compositions infiltrated into adjacent tissue tracheal and bronchial stents are provided above in conjunction with the coating of medical devices. Numerous agents or compositions for use with tracheal and bronchial have been described above which may be infiltrated into the tissue adjacent to the device (preferably near the device-tissue interface).
Therapeutic agents or pharmaceutical compositionsmay be infiltrated around implanted tracheal and bronchial stents by applying the composition directly and/or indirectly into and/or onto (a) tissue adjacent to the tracheal/bronchial stent; (b) the vicinity of the tracheal/bronchial stent- tissue interface; (c) the region around the tracheal/bronchial stent; and (d) tissue surrounding the tracheal/bronchial stent. Methods for infiltrating the subject agents or compositions into tissue adjacent to a tracheal/bronchial stent include delivering the composition: (a) to the tracheal/bronchial stent surface (e.g., as an injectable, paste, gel or mesh) during the implantation procedure; (b) to the surface of the tissue (e.g., as an injectable, paste, gel, in situ forming gel or mesh) immediately prior to, or during, implantation of the tracheal/bronchial stent; (c) to the surface of the tracheal/bronchial stent and/or the tissue surrounding the implanted tracheal/bronchial stent (e.g., as an injectable, paste, gel, in situ forming gel or mesh) immediately after the implantation of the tracheal/bronchial stent; (d) by topical application of the composition into the anatomical space where the tracheal/bronchial stent may be placed (particularly useful for this embodiment is the use of polymeric carriers which release the therapeutic agent over a period ranging from several hours to several weeks - fluids, suspensions, emulsions, microemulsions, microspheres, pastes, gels, microparticulates, sprays, aerosols, solid implants and other formulations which release the agent may be delivered into the region where the device may be inserted); (e) via percutaneous injection into the tissue surrounding the tracheal/bronchial stent as a solution as an infusate or as a sustained release preparation; (f) by any combination of the aforementioned methods. Combination therapies (Ae., combinations of therapeutic agents and combinations with antithrombotic and/or antiplatelet agents) may also be used. In all cases it is understood that the subject agents or compositions may be infiltrated into tissue adjacent to all or a portion of the device.
According to one aspect, any fibrosis-inhibiting and/or anti- infective agent described above may be utilized in the practice of the present invention. In one aspect of the invention, the subject compositions infiltrated into tissue adjacent to tracheal and bronchial stents may be adapted to release an agent that inhibits one or more of the four general components of the process of fibrosis (or scarring), including: formation of new blood vessels (angiogenesis), migration and proliferation of connective tissue cells (such as fibroblasts or smooth muscle cells), deposition of extracellular matrix (ECM), and remodeling (maturation and organization of the fibrous tissue). By inhibiting one or more of the components of fibrosis (or scarring), the overgrowth of granulation tissue may be inhibited or reduced.
Examples of fibrosis-inhibiting agents for use in the present invention include the following: ZD-6474, AP-23573, synthadotin, S-0885, aplidine, ixabepilone, IDN-5390, SB-2723005, ABT-518, combretastatin, anecortave acetate, SB-715992, temsirolimus, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, as well as analogues and derivatives of the aforementioned.
Additional examples of anti-scarring agents which can be used include those having a high potency in the assays described herein (1- 10OnM IC50 range) such as isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, dolastatin 15, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17- DMAG, and tacrolimus; those having a mid-potency in the assays described herein (100-500 nM IC50 range) such as loteprednol etabonate, juglone, prednisolone, puromycin, 3-BAABE, cladribine, and mannose-6-phosphate; and those having a low potency in the assays described herein (500- 1000nm range IC50 range) such as 5-azacytidine, Ly333531(ruboxistaurin), and simvastatin.
The drug dose administered from the present compositions for prevention or inhibition of fibrosis in accordance with the present invention will depend on a variety of factors, including the type of formulation, the location of the treatment site, and the type of condition being treated. As tracheal and bronchial stents are made in a variety of configurations and sizes, the exact dose administered will also vary with device size, surface area and design. However, certain principles can be applied in the application of this art. Drug dose can be calculated as a function of dose per unit area (of the treatment site), total drug dose administered can be measured and appropriate surface concentrations of active drug can be determined. Drugs are to be used at concentrations that range from several times more than to 50%, 20%, 10%, 5%, or even less than 1% of the concentration typically used in a single chemotherapeutic systemic dose application. In certain aspects, the anti-scarring agent is released from the composition in effective concentrations in a time period that may be measured from the time of infiltration into tissue adjacent to the device, which ranges from about less than 1 day to about 180 days. Generally, the release time may also be from about less than 1 day to about 180 days; from about 7 days to about 14 days; from about 14 days to about 28 days; from about 28 days to about 56 days; from about 56 days to about 90 days; from about 90 days to about 180 days.
The exemplary anti-fibrosing agents, used alone or in combination, should be administered under the following dosing guidelines. The total amount (dose) of anti-scarring agent in the composition can be in the range of about 0.01 μg-10 μg, or about 10 μg-10 mg, or about 10 mg- 250 mg, or about 250 mg-1000 mg, or about 1000 mg-2500 mg. The dose (amount) of anti-scarring agent per unit area of device or tissue surface to which the agent is applied may be in the range of about 0.01 μg/mm2 - 1 μg/mm2, or about 1 μg/mm2 - 10 μg/mm2, or about 10 μg/mm2 - 250 μg/mm2, or about 250 μg/mm2 - 1000 μg/mm2, or about 1000 μg/mm2 - 2500 μg/mm2.
According to another aspect, any anti-infective agent described above may be used in the practice of the present invention. Exemplary anti-infective agents include (A) anthracyclines (e.g., doxorubicin and mitoxantrone), (B) fluoropyrimidines (e.g., 5-FU), (C) folic acid antagonists (e.g., methotrexate), (D) podophylotoxins (e.g., etoposide), (E) camptothecins, (F) hydroxyureas, and (G) platinum complexes (e.g., cisplatin), as well as analogues and derivatives of the aforementioned. The drug dose administered from the present compositions for prevention or inhibition of infection in accordance with the present invention will depend on a variety of factors, including the type of formulation, the location of the treatment site, and the type of condition being treated. However, certain principles can be applied in the application of this art. Drug dose can be calculated as a function of dose per unit area (of the treatment site), total drug dose administered can be measured and appropriate surface concentrations of active drug can be determined. Drugs are to be used at concentrations that range from several times more than to 50%, 20%, 10%, 5%, or even less than 1% of the concentration typically used in a single anti-infective systemic dose application. In certain aspects, the anti-infective agent is released from the composition in effective concentrations in a time period that may be measured from the time of infiltration into tissue adjacent to the device, which ranges from about less than 1 day to about 180 days. Generally, the release time may also be from about less than 1 day to about 180 days; from about 7 days to about 14 days; from about 14 days to about 28 days; from about 28 days to about 56 days; from about 56 days to about 90 days; from about 90 days to about 180 days.
The exemplary anti-infective agents, used alone or in combination, should be administered under the following dosing guidelines. The total amount (dose) of anti-infective agent in the composition can be in the range of about 0.01 μg-1 μg, or about 1 μg-10 μg, or about 10 μg-1 mg, or about 1 mg to 10 mg, or about 10 mg-100 mg, or about 100 mg to 250 mg, or about 250 mg-1000 mg. The dose (amount) of anti-infective agent per unit area of device or tissue surface to which the agent is applied may be in the range of about 0.01 μg/mm2 - 1 μg/mm2, or about 1 μg/mm2 - 10 μg/mm2, or about 10 μg/mm2 - 100 μg/mm2, or about 100 μg/mm2 to 250 μg/mm2, or about 250 μg/mm2 - 1000 μg/mm2. As different compositions will release the anti-infective agent at differing rates, the above dosing parameters should be utilized in combination with the release rate of the drug from the composition such that a minimum concentration of about 10"8 M to 10"7 M, or about 10'7 M to 10"6 M about 1CT6 M to 10"5 M or about 10'5 M to 10"4 M of the agent is maintained on the tissue surface.
It should be readily evident based upon the discussions provided herein that combinations of anthracyclines (e.g., doxorubicin or mitoxantrone), fluoropyrimidines (e.g., 5-fluorouracil), folic acid antagonists (e.g., methotrexate), quinolones, and/or podophylotoxins (e.g., etoposide) may be utilized to enhance the antibacterial activity of the composition.
Genital-Urinary Stents
In one aspect, the subject compositions may be infiltrated into tissue adjacent to a genital-urinary (GU) stent device. The subject compositions may contain a therapeutic agent (e.g., an anti-scarring and/or anti-infective agent).
Representative examples genital-urinary (GU) stents that may benefit from having the subject compositions infiltrated into adjacent tissue are provided above in conjunction with the coating of medical devices. Numerous polymeric compositions for use with tracheal and bronchial have been described above which may be infiltrated into the tissue adjacent to the device (preferably near the device-tissue interface).
Therapeutic agents or pharmaceutical compositionsmay be infiltrated around implanted GU stents by applying the composition directly and/or indirectly into and/or onto (a) tissue adjacent to the GU stent; (b) the vicinity of the GU stent-tissue interface; (c) the region around the GU stent; and (d) tissue surrounding the GU stent. Methods for infiltrating the subject compositions into tissue adjacent to a GU stent include delivering the composition: (a) to the GU stent surface (e.g., as an injectable, paste, gel or mesh) during the implantation procedure; (b) to the surface of the tissue (e.g., as an injectable, paste, gel, in situ forming gel or mesh) immediately prior to, or during, implantation of the GU stent; (c) to the surface of the GU stent and/or the tissue surrounding the implanted GU stent (e.g., as an injectable, paste, gel, in situ forming gel or mesh) immediately after the implantation of the GU stent; (d) by topical application of the composition into the anatomical space where the GU stent may be placed (particularly useful for this embodiment is the use of polymeric carriers which release the therapeutic agent over a period ranging from several hours to several weeks - fluids, suspensions, emulsions, microemulsions, microspheres, pastes, gels, microparticulates, sprays, aerosols, solid implants and other formulations which release the agent may be delivered into the region where the device may be inserted); (e) via percutaneous injection into the tissue surrounding the GU stent as a solution as an infusate or as a sustained release preparation; (f) by any combination of the aforementioned methods. Combination therapies {i.e., combinations of therapeutic agents and combinations with antithrombotic and/or antiplatelet agents) may also be used. In all cases it is understood that the subject compositions may be infiltrated into tissue adjacent to all or a portion of the device.
According to one aspect, any fibrosis-inhibiting and/or anti- infective agent described above may be utilized in the practice of the present invention. In one aspect of the invention, the subject compositions infiltrated into tissue adjacent to GU stents may be adapted to release an agent that inhibits one or more of the four general components of the process of fibrosis (or scarring), including: formation of new blood vessels (angiogenesis), migration and proliferation of connective tissue cells (such as fibroblasts or smooth muscle cells), deposition of extracellular matrix (ECM), and remodeling (maturation and organization of the fibrous tissue). By inhibiting one or more of the components of fibrosis (or scarring), the overgrowth of granulation tissue may be inhibited or reduced. Examples of fibrosis-inhibiting agents for use in the present invention include the following: ZD-6474, AP-23573, synthadotin, S-0885, aplidine, ixabepilone, IDN-5390, SB-2723005, ABT-518, combretastatin, anecortave acetate, SB- 715992, temsirolimus, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, as well as analogues and derivatives of the aforementioned.
Additional examples of anti-scarring agents which can be used include those having a high potency in the assays described herein (1- 10OnM IC50 range) such as isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, dolastatin 15, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17- DMAG, and tacrolimus; those having a mid-potency in the assays described herein (100-500 nM IC50 range) such as loteprednol etabonate, juglone, prednisolone, puromycin, 3-BAABE, cladribine, and mannose-6-phosphate; and those having a low potency in the assays described herein (500- 1000nm range IC50 range) such as 5-azacytidine, Ly333531(ruboxistaurin), and simvastatin.
The drug dose administered from the present compositions for prevention or inhibition of fibrosis in accordance with the present invention will depend on a variety of factors, including the type of formulation, the location of the treatment site, and the type of condition being treated. As GU stents are made in a variety of configurations and sizes, the exact dose administered will also vary with device size, surface area and design. However, certain principles can be applied in the application of this art. Drug dose can be calculated as a function of dose per unit area (of the treatment site), total drug dose administered can be measured and appropriate surface concentrations of active drug can be determined. Drugs are to be used at concentrations that range from several times more than to 50%, 20%, 10%, 5%, or even less than 1% of the concentration typically used in a single chemotherapeutic systemic dose application. In certain aspects, the anti-scarring agent is released from the composition in effective concentrations in a time period that may be measured from the time of infiltration into tissue adjacent to the device, which ranges from about less than 1 day to about 180 days. Generally, the release time may also be from about less than 1 day to about 180 days; from about 7 days to about 14 days; from about 14 days to about 28 days; from about 28 days to about 56 days; from about 56 days to about 90 days; from about 90 days to about 180 days.
The exemplary anti-fibrosing agents, used alone or in combination, should be administered under the following dosing guidelines. The total amount (dose) of anti-scarring agent in the composition can be in the range of about 0.01 μg-10 μg, or about 10 μg-10 mg, or about 10 mg- 250 mg, or about 250 mg-1000 mg, or about 1000 mg-2500 mg. The dose (amount) of anti-scarring agent per unit area of device or tissue surface to which the agent is applied may be in the range of about 0.01 μg/mm2 - 1 μg/mm2, or about 1 μg/mm2 - 10 μg/mm2, or about 10 μg/mm2 - 250 μg/mm2, or about 250 μg/mm2 - 1000 μg/mm2, or about 1000 μg/mm2 - 2500 μg/mm2.
According to another aspect, any anti-infective agent described above may be used in the practice of the present invention. Exemplary anti-infective agents include (A) anthracyclines (e.g., doxorubicin and mitoxantrone), (B) fluoropyrimidines (e.g., 5-FU), (C) folic acid antagonists (e.g., methotrexate), (D) podophylotoxins (e.g., etoposide), (E) camptothecins, (F) hydroxyureas, and (G) platinum complexes (e.g., cisplatin), as well as analogues and derivatives of the aforementioned.
The drug dose administered from the present compositions for prevention or inhibition of infection in accordance with the present invention will depend on a variety of factors, including the type of formulation, the location of the treatment site, and the type of condition being treated. However, certain principles can be applied in the application of this art. Drug dose can be calculated as a function of dose per unit area (of the treatment site), total drug dose administered can be measured and appropriate surface concentrations of active drug can be determined. Drugs are to be used at concentrations that range from several times more than to 50%, 20%, 10%, 5%, or even less than 1% of the concentration typically used in a single anti-infective systemic dose application. In certain aspects, the anti- infective agent is released from the composition in effective concentrations in a time period that may be measured from the time of infiltration into tissue adjacent to the device, which ranges from about less than 1 day to about 180 days. Generally, the release time may also be from about less than 1 day to about 180 days; from about 7 days to about 14 days; from about 14 days to about 28 days; from about 28 days to about 56 days; from about 56 days to about 90 days; from about 90 days to about 180 days.
The exemplary anti-infective agents, used alone or in combination, should be administered under the following dosing guidelines. The total amount (dose) of anti-infective agent in the composition can be in the range of about 0.01 μg-1 μg, or about 1 μg-10 μg, or about 10 μg-1 mg, or about 1 mg to 10 mg, or about 10 mg-100 mg, or about 100 mg to 250 mg, or about 250 mg-1000 mg. The dose (amount) of anti-infective agent per unit area of device or tissue surface to which the agent is applied may be in the range of about 0.01 μg/mm2 - 1 μg/mm2, or about 1 μg/mm2 - 10 μg/mm2, or about 10 μg/mm2 - 100 μg/mm2, or about 100 μg/mm2 to 250 μg/mm2, or about 250 μg/mm2 - 1000 μg/mm2. As different compositions will release the anti-infective agent at differing rates, the above dosing parameters should be utilized in combination with the release rate of the drug from the composition such that a minimum concentration of about 10"8 to 10'7, or about 1O-7 to 10"6 about lO"6 to 10'5or about lO'5 to 104 of the agent is maintained on the tissue surface.
It should be readily evident based upon the discussions provided herein that combinations of anthracyclines (e.g., doxorubicin or mitoxantrone), fluoropyrimidines (e.g., 5-fluorouracil), folic acid antagonists (e.g., methotrexate), quinolones, and/or podophylotoxins (e.g., etoposide) may be utilized to enhance the antibacterial activity of the composition. Ear and Nose and Throat Stents
In one aspect, the present subject compositions may be infiltrated into tissue adjacent to an ear-nose-throat (ENT) stent device (e.g., a lacrimal duct stent, Eustachian tube stent, nasal stent, or sinus stent). The subject compositions may contain a therapeutic agent (e.g., an anti- scarring and/or anti-infective agent).
Representative examples ear and nose stents that may benefit from having the subject compositions infiltrated into adjacent tissue are provided above in conjunction with the coating of medical devices. Numerous polymeric compositions for use with tracheal and bronchial have been described above which may be infiltrated into the tissue adjacent to the device (preferably near the device-tissue interface).
Therapeutic agents or pharmaceutical compositionsmay be infiltrated around implanted ENT stents by applying the composition directly and/or indirectly into and/or onto (a) tissue adjacent to the ENT stent; (b) the vicinity of the ENT stent-tissue interface; (c) the region around the ENT stent; and (d) tissue surrounding the ENT stent. Methods for infiltrating the subject compositions into tissue adjacent to a ENT stent include delivering the composition: (a) to the ENT stent surface (e.g., as an injectable, paste, gel or mesh) during the implantation procedure; (b) to the surface of the tissue (e.g., as an injectable, paste, gel, in situ forming gel or mesh) immediately prior to, or during, implantation of the ENT stent; (c) to the surface of the ENT stent and/or the tissue surrounding the implanted ENT stent (e.g., as an injectable, paste, gel, in situ forming gel or mesh) immediately after the implantation of the ENT stent; (d) by topical application of the composition into the anatomical space where the ENT stent may be placed (particularly useful for this embodiment is the use of polymeric carriers which release the therapeutic agent over a period ranging from several hours to several weeks - fluids, suspensions, emulsions, microemulsions, microspheres, pastes, gels, microparticulates, sprays, aerosols, solid implants and other formulations which release the agent may be delivered into the region where the device may be inserted); (e) via percutaneous injection into the tissue surrounding the ENT stent as a solution as an infusate or as a sustained release preparation; (f) by any combination of the aforementioned methods. Combination therapies (Ae., combinations of therapeutic agents and combinations with antithrombotic and/or antiplatelet agents) may also be used. In all cases it is understood that the subject compositions may be infiltrated into tissue adjacent to all or a portion of the device.
According to one aspect, any fibrosis-inhibiting and/or anti- infective agent described above may be utilized in the practice of the present invention. In one aspect of the invention, the subject compositions infiltrated into tissue adjacent to ENT stents may be adapted to release an agent that inhibits one or more of the four general components of the process of fibrosis (or scarring), including: formation of new blood vessels (angiogenesis), migration and proliferation of connective tissue cells (such as fibroblasts or smooth muscle cells), deposition of extracellular matrix (ECM), and remodeling (maturation and organization of the fibrous tissue). By inhibiting one or more of the components of fibrosis (or scarring), the overgrowth of granulation tissue may be inhibited or reduced.
Examples of fibrosis-inhibiting agents for use in the present invention include the following: ZD-6474, AP-23573, synthadotin, S-0885, aplidine, ixabepilone, IDN-5390, SB-2723005, ABT-518, combretastatin, anecortave acetate, SB-715992, temsirolimus, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, as well as analogues and derivatives of the aforementioned.
Additional examples of anti-scarring agents which can be used include those having a high potency in the assays described herein (1- 10OnM IC50 range) such as isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, dolastatin 15, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17- DMAG, and tacrolimus; those having a mid-potency in the assays described herein (100-500 nM IC50 range) such as loteprednol etabonate, juglone, prednisolone, puromycin, 3-BAABE, cladribine, and mannose-6-phosphate; and those having a low potency in the assays described herein (500- 1000nm range IC5O range) such as 5-azacytidine, Ly333531(ruboxistaurin), and simvastatin.
The drug dose administered from the present compositions for prevention or inhibition of fibrosis in accordance with the present invention will depend on a variety of factors, including the type of formulation, the location of the treatment site, and the type of condition being treated. As ENT stents are made in a variety of configurations and sizes, the exact dose administered will also vary with device size, surface area and design. However, certain principles can be applied in the application of this art. Drug dose can be calculated as a function of dose per unit area (of the treatment site), total drug dose administered can be measured and appropriate surface concentrations of active drug can be determined. Drugs are to be used at concentrations that range from several times more than to 50%, 20%, 10%, 5%, or even less than 1% of the concentration typically used in a single chemotherapeutic systemic dose application. In certain aspects, the anti-scarring agent is released from the composition in effective concentrations in a time period that may be measured from the time of infiltration into tissue adjacent to the device, which ranges from about less than 1 day to about 180 days. Generally, the release time may also be from about less than 1 day to about 180 days; from about 7 days to about 14 days; from about 14 days to about 28 days; from about 28 days to about 56 days; from about 56 days to about 90 days; from about 90 days to about 180 days.
The exemplary anti-fibrosing agents, used alone or in combination, should be administered under the following dosing guidelines. The total amount (dose) of anti-scarring agent in the composition can be in the range of about 0.01 μg-10 μg, or about 10 μg-10 mg, or about 10 mg- 250 mg, or about 250 mg-1000 mg, or about 1000 mg-2500 mg. The dose (amount) of anti-scarring agent per unit area of device or tissue surface to which the agent is applied may be in the range of about 0.01 μg/mm2 - 1 μg/mm2, or about 1 μg/mm2 - 10 μg/mm2, or about 10 μg/mm2 - 250 μg/mm2, or about 250 μg/mm2 - 1000 μg/mm2, or about 1000 μg/mm2 - 2500 μg/mm2.
According to another aspect, any anti-infective agent described above may be used in the practice of the present invention. Exemplary anti-infective agents include (A) anthracyclines (e.g., doxorubicin and mitoxantrone), (B) fluoropyrimidines (e.g., 5-FU), (C) folic acid antagonists (e.g., methotrexate), (D) podophylotoxins {e.g., etoposide), (E) camptothecins, (F) hydroxyureas, and (G) platinum complexes (e.g., cisplatin), as well as analogues and derivatives of the aforementioned.
The drug dose administered from the present compositions for prevention or inhibition of infection in accordance with the present invention will depend on a variety of factors, including the type of formulation, the location of the treatment site, and the type of condition being treated. However, certain principles can be applied in the application of this art. Drug dose can be calculated as a function of dose per unit area (of the treatment site), total drug dose administered can be measured and appropriate surface concentrations of active drug can be determined. Drugs are to be used at concentrations that range from several times more than to 50%, 20%, 10%, 5%, or even less than 1% of the concentration typically used in a single anti-infective systemic dose application. In certain aspects, the anti- infective agent is released from the composition in effective concentrations in a time period that may be measured from the time of infiltration into tissue adjacent to the device, which ranges from about less than 1 day to about 180 days. Generally, the release time may also be from about less than 1 day to about 180 days; from about 7 days to about 14 days; from about 14 days to about 28 days; from about 28 days to about 56 days; from about 56 days to about 90 days; from about 90 days to about 180 days. The exemplary anti-infective agents, used alone or in combination, should be administered under the following dosing guidelines. The total amount (dose) of anti-infective agent in the composition can be in the range of about 0.01 μg-1 μg, or about 1 μg-10 μg, or about 10 μg-1 mg, or about 1 mg to 10 mg, or about 10 mg-100 mg, or about 100 mg to 250 mg, or about 250 mg-1000 mg. The dose (amount) of anti-infective agent per unit area of device or tissue surface to which the agent is applied may be in the range of about 0.01 μg/mm2 - 1 μg/mm2, or about 1 μg/mm2 - 10 μg/mm2, or about 10 μg/mm2 - 100 μg/mm2, or about 100 μg/mm2 to 250 μg/mm2, or about 250 μg/mm2 - 1000 μg/mm2. As different compositions will release the anti-infective agent at differing rates, the above dosing parameters should be utilized in combination with the release rate of the drug from the composition such that a minimum concentration of about 10'8 to 10"7, or about 1O-7 to lO"6 about 10"6 to 10'5 or about 10"5 to 10"4 of the agent is maintained on the tissue surface.
It should be readily evident based upon the discussions provided herein that combinations of anthracyclines (e.g., doxorubicin or mitoxantrone), fluoropyrimidines (e.g., 5-fluorouracil), folic acid antagonists (e.g., methotrexate), quinolones, and/or podophylotoxins (e.g., etoposide) may be utilized to enhance the antibacterial activity of the composition.
Ear Ventilation Tubes
In another aspect, the subject compositions may be infiltrated into tissue adjacent to an ear ventilation tube (also referred to as a tympanostomy tube). The subject compositions may contain a therapeutic agent (e.g., an anti-scarring and/or anti-infective agent).
Representative examples ear ventilation tubes that may benefit from having the subject compositions infiltrated into adjacent tissue are provided above in conjunction with the coating of medical devices. Numerous agents or compositions for use with ear ventilation tubes have been described above which may be infiltrated into the tissue adjacent to the device (preferably near the device-tissue interface).
Therapeutic agents or pharmaceutical compositionsmay be infiltrated around implanted ear ventilation tube devices by applying the composition directly and/or indirectly into and/or onto (a) tissue adjacent to the ear ventilation tube devices; (b) the vicinity of the ear ventilation tube device-tissue interface; (c) the region around the ear ventilation tube device; and (d) tissue surrounding the ear ventilation tube device. Methods for infiltrating the subject compositions into tissue adjacent to an ear ventilation tube device include delivering the composition: (a) to the ear ventilation tube device surface (e.g., as an injectable, paste, gel or mesh) during the implantation procedure; (b) to the surface of the tissue (e.g., as an injectable, paste, gel, in situ forming gel or mesh) immediately prior to, or during, implantation of the ear ventilation tube device; (c) to the surface of the ear ventilation tube device and/or the tissue surrounding the implanted ear ventilation tube device (e.g., as an injectable, paste, gel; in situ forming gel or mesh) immediately after the implantation of the ear ventilation tube device; (d) by topical application of the composition into the anatomical space where the ear ventilation tube device may be placed (particularly useful for this embodiment is the use of polymeric carriers which release the therapeutic agent over a period ranging from several hours to several weeks - fluids, suspensions, emulsions, microemulsions, microspheres, pastes, gels, microparticulates, sprays, aerosols, solid implants and other formulations which release the agent may be delivered into the region where the device may be inserted); (e) via percutaneous injection into the tissue surrounding the ear ventilation tube device as a solution as an infusate or as a sustained release preparation; (f) by any combination of the aforementioned methods. Combination therapies (i.e., combinations of therapeutic agents and combinations with antithrombotic and/or antiplatelet agents) may also be used. In all cases it is understood that the subject compositions may be infiltrated into tissue adjacent to all or a portion of the device.
According to one aspect, any fibrosis-inhibiting and/or anti- infective agent described above can be utilized in the practice of the present invention. In one aspect of the invention, the subject compositions infiltrated into tissue adjacent to ear ventilation tubes may be adapted to release an agent that inhibits one or more of the four general components of the process of fibrosis (or scarring), including: formation of new blood vessels (angiogenesis), migration and proliferation of connective tissue cells (such as fibroblasts or smooth muscle cells), deposition of extracellular matrix (ECM), and remodeling (maturation and organization of the fibrous tissue). By inhibiting one or more of the components of fibrosis (or scarring), the overgrowth of granulation tissue may be inhibited or reduced.
Examples of fibrosis-inhibiting agents for use in the present invention include the following: ZD-6474, AP-23573, synthadotin, S-0885, aplidine, ixabepilone, IDN-5390, SB-2723005, ABT-518, combretastatin, anecortave acetate, SB-715992, temsirolimus, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, as well as analogues and derivatives of the aforementioned.
Additional examples of anti-scarring agents which can be used include those having a high potency in the assays described herein (1- 10OnM IC50 range) such as isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, dolastatin 15, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17- DMAG, and tacrolimus; those having a mid-potency in the assays described herein (100-500 nM ICs0 range) such as loteprednol etabonate, juglone, prednisolone, puromycin, 3-BAABE, cladribine, and mannose-6-phosphate; and those having a low potency in the assays described herein (500- 1000nm range IC50 range) such as 5-azacytidine, Ly333531(ruboxistaurin), and simvastatin. The drug dose administered from the present compositions for prevention or inhibition of fibrosis in accordance with the present invention will depend on a variety of factors, including the type of formulation, the location of the treatment site, and the type of condition being treated. As ear ventilation tubes are made in a variety of configurations and sizes, the exact dose administered will also vary with device size, surface area and design. However, certain principles can be applied in the application of this art. Drug dose can be calculated as a function of dose per unit area (of the treatment site), total drug dose administered can be measured and appropriate surface concentrations of active drug can be determined. Drugs are to be used at concentrations that range from several times more than to 50%, 20%, 10%, 5%, or even less than 1% of the concentration typically used in a single chemotherapeutic systemic dose application. In certain aspects, the anti-scarring agent is released from the composition in effective concentrations in a time period that may be measured from the time of infiltration into tissue adjacent to the device, which ranges from about less than 1 day to about 180 days. Generally, the release time may also be from about less than 1 day to about 180 days; from about 7 days to about 14 days; from about 14 days to about 28 days; from about 28 days to about 56 days; from about 56 days to about 90 days; from about 90 days to about 180 days.
The exemplary anti-fibrosing agents, used alone or in combination, should be administered under the following dosing guidelines. The total amount (dose) of anti-scarring agent in the composition can be in the range of about 0.01 μg-10 μg, or about 10 μg-10 mg, or about 10 mg- 250 mg, or about 250 mg-1000 mg, or about 1000 mg-2500 mg. The dose (amount) of anti-scarring agent per unit area of device or tissue surface to which the agent is applied may be in the range of about 0.01 μg/mm2 - 1 μg/mm2, or about 1 μg/mm2 - 10 μg/mm2, or about 10 μg/mm2 - 250 μg/mm2, or about 250 μg/mm2 - 1000 μg/mm2, or about 1000 μg/mm2 - 2500 μg/mm2. According to another aspect, any anti-infective agent described above may be used in the practice of the present invention. Exemplary anti-infective agents include (A) anthracyclines (e.g., doxorubicin and mitoxantrone), (B) fluoropyrimidines (e.g., 5-FU), (C) folic acid antagonists (e.g., methotrexate), (D) podophylotoxins (e.g., etoposide), (E) camptothecins, (F) hydroxyureas, and (G) platinum complexes (e.g., cisplatin), as well as analogues and derivatives of the aforementioned.
The drug dose administered from the present compositions for prevention or inhibition of infection in accordance with the present invention will depend on a variety of factors, including the type of formulation, the location of the treatment site, and the type of condition being treated. However, certain principles can be applied in the application of this art. Drug dose can be calculated as a function of dose per unit area (of the treatment site), total drug dose administered can be measured and appropriate surface concentrations of active drug can be determined. Drugs are to be used at concentrations that range from several times more than to 50%, 20%, 10%, 5%, or even less than 1% of the concentration typically used in a single anti-infective systemic dose application. In certain aspects, the anti- infective agent is released from the composition in effective concentrations in a time period that may be measured from the time of infiltration into tissue adjacent to the device, which ranges from about less than 1 day to about 180 days. Generally, the release time may also be from about less than 1 day to about 180 days; from about 7 days to about 14 days; from about 14 days to about 28 days; from about 28 days to about 56 days; from about 56 days to about 90 days; from about 90 days to about 180 days.
The exemplary anti-infective agents, used alone or in combination, should be administered under the following dosing guidelines. The total amount (dose) of anti-infective agent in the composition can be in the range of about 0.01 μg-1 μg, or about 1 μg-10 μg, or about 10 μg-1 mg, or about 1 mg to 10 mg, or about 10 mg-100 mg, or about 100 mg to 250 mg, or about 250 mg-1000 mg. The dose (amount) of anti-infective agent per unit area of device or tissue surface to which the agent is applied may be in the range of about 0.01 μg/mm2 - 1 μg/mm2, or about 1 μg/mm2 - 10 μg/mm2, or about 10 μg/mm2 - 100 μg/mm2, or about 100 μg/mm2 to 250 μg/mm2, or about 250 μg/mm2 - 1000 μg/mm2. As different compositions will release the anti-infective agent at differing rates, the above dosing parameters should be utilized in combination with the release rate of the drug from the composition such that a minimum concentration of about 10"8 to 10"7, or about 10"7 to 10"6 about 10'6 to 10"5or about 10~5 to 10"4 of the agent is maintained on the tissue surface.
It should be readily evident based upon the discussions provided herein that combinations of anthracyclines (e.g., doxorubicin or mitoxantrone), fluoropyrimidines (e.g., 5-fluorouracil), folic acid antagonists (e.g., methotrexate), quinolones, and/or podophylotoxins (e.g., etoposide) may be utilized to enhance the antibacterial activity of the composition.
Intraocular Implants
In another aspect, the subject compositions may be infiltrated into tissue adjacent to an intraocular implant. The subject compositions may contain a therapeutic agent (e.g., an anti-scarring and/or anti-infective agent).
Representative examples of intraocular implants that may benefit from having the subject compositions infiltrated into adjacent tissue are provided above in conjunction with the coating of medical devices. Numerous agents or compositions for use with intraocular implants have been described above which may be infiltrated into the tissue adjacent to the device (preferably near the device-tissue interface).
Therapeutic agents or pharmaceutical compositionsmay be infiltrated around implanted intraocular implants by applying the composition directly and/or indirectly into and/or onto (a) tissue adjacent to the intraocular implant; (b) the vicinity of the intraocular implant-tissue interface; (c) the region around the intraocular implant; and (d) tissue surrounding the intraocular implant. Methods for infiltrating the subject compositions into tissue adjacent to an intraocular implant include delivering the composition: (a) to the intraocular implant surface (e.g., as an injectable, paste, gel or mesh) during the implantation procedure; (b) to the surface of the tissue (e.g., as an injectable, paste, gel, in situ forming gel or mesh) immediately prior to, or during, implantation of the intraocular implant; (c) to the surface of the intraocular implant and/or the tissue surrounding the implanted intraocular implant (e.g., as an injectable, paste, gel, in situ forming gel or mesh) immediately after the implantation of the intraocular implant; (d) by topical application of the composition into the anatomical space where the intraocular implant may be placed (particularly useful for this embodiment is the use of polymeric carriers which release the therapeutic agent over a period ranging from several hours to several weeks - fluids, suspensions, emulsions, microemulsions, microspheres, pastes, gels, microparticulates, sprays, aerosols, solid implants and other formulations which release the agent may be delivered into the region where the device may be inserted);
(e) via percutaneous injection into the tissue surrounding the intraocular implant as a solution as an infusate or as a sustained release preparation;
(f) by any combination of the aforementioned methods. Combination therapies (i.e., combinations of therapeutic agents and combinations with antithrombotic and/or antiplatelet agents) may also be used. In all cases it is understood that the subject compositions may be infiltrated into tissue adjacent to all or a portion of the device.
The process of infiltrating the subject compositions into tissue adjacent to these implants and the materials selected for these processes are such that they do not significantly alter the refractive index of the intraocular implant or the visible light transmission of the implant or lens.
According to one aspect, any fibrosis-inhibiting and/or anti- infective agent described above may be utilized in the practice of the present invention. In one aspect of the invention, the subject compositions infiltrated into tissue adjacent to intraocular implants may be adapted to release an agent that inhibits one or more of the four general components of the process of fibrosis (or scarring), including: formation of new blood vessels (angiogenesis), migration and proliferation of connective tissue cells (such as fibroblasts or smooth muscle cells), deposition of extracellular matrix (ECM), and remodeling (maturation and organization of the fibrous tissue). By inhibiting one or more of the components of fibrosis (or scarring), the overgrowth of granulation tissue may be inhibited or reduced.
Examples of fibrosis-inhibiting agents for use in the present invention include the following: ZD-6474, AP-23573, synthadotin, S-0885, aplidine, ixabepilone, IDN-5390, SB-2723005, ABT-518, combretastatin, anecortave acetate, SB-715992, temsirolimus, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, as well as analogues and derivatives of the aforementioned.
Additional examples of anti-scarring agents which can be used include those having a high potency in the assays described herein (1- 10OnM IC50 range) such as isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, dolastatin 15, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17- DMAG, and tacrolimus; those having a mid-potency in the assays described herein (100-500 nM IC50 range) such as loteprednol etabonate, juglone, prednisolone, puromycin, 3-BAABE, cladribine, and mannose-6-phosphate; and those having a low potency in the assays described herein (500- 1000nm range IC50 range) such as 5-azacytidine, Ly333531(ruboxistaurin), and simvastatin.
The drug dose administered from the present compositions for prevention or inhibition of fibrosis in accordance with the present invention will depend on a variety of factors, including the type of formulation, the location of the treatment site, and the type of condition being treated. As intraocular implants are made in a variety of configurations and sizes, the exact dose administered will also vary with device size, surface area and design. However, certain principles can be applied in the application of this art. Drug dose can be calculated as a function of dose per unit area (of the treatment site), total drug dose administered can be measured and appropriate surface concentrations of active drug can be determined. Drugs are to be used at concentrations that range from several times more than to 50%, 20%, 10%, 5%, or even less than 1% of the concentration typically used in a single chemotherapeutic systemic dose application. In certain aspects, the anti-scarring agent is released from the composition in effective concentrations in a time period that may be measured from the time of infiltration into tissue adjacent to the device, which ranges from about less than 1 day to about 180 days. Generally, the release time may also be from about less than 1 day to about 180 days; from about 7 days to about 14 days; from about 14 days to about 28 days; from about 28 days to about 56 days; from about 56 days to about 90 days; from about 90 days to about 180 days.
The exemplary anti-fibrosing agents, used alone or in combination, should be administered under the following dosing guidelines. The total amount (dose) of anti-scarring agent in the composition can be in the range of about 0.01 μg-10 μg, or about 10 μg-10 mg, or about 10 mg- 250 mg, or about 250 mg-1000 mg, or about 1000 mg-2500 mg. The dose (amount) of anti-scarring agent per unit area of device or tissue surface to which the agent is applied may be in the range of about 0.01 μg/mm2 - 1 μg/mm2, or about 1 μg/mm2 - 10 μg/mm2, or about 10 μg/mm2 - 250 μg/mm2, or about 250 μg/mm2 - 1000 μg/mm2, or about 1000 μg/mm2 - 2500 μg/mm2.
According to another aspect, any anti-infective agent described above may be used in the practice of the present invention. Exemplary anti-infective agents include (A) anthracyclines (e.g., doxorubicin and mitoxantrone), (B) fluoropyrimidines (e.g., 5-FU), (C) folic acid antagonists (e.g., methotrexate), (D) podophylotoxins (e.g., etoposide), (E) camptothecins, (F) hydroxyureas, and (G) platinum complexes {e.g., cisplatin), as well as analogues and derivatives of the aforementioned.
The drug dose administered from the present compositions for prevention or inhibition of infection in accordance with the present invention will depend on a variety of factors, including the type of formulation, the location of the treatment site, and the type of condition being treated. However, certain principles can be applied in the application of this art. Drug dose can be calculated as a function of dose per unit area (of the treatment site), total drug dose administered can be measured and appropriate surface concentrations of active drug can be determined. Drugs are to be used at concentrations that range from several times more than to 50%, 20%, 10%, 5%, or even less than 1% of the concentration typically used in a single anti-infective systemic dose application. In certain aspects, the anti- infective agent is released from the composition in effective concentrations in a time period that may be measured from the time of infiltration into tissue adjacent to the device, which ranges from about less than 1 day to about 180 days. Generally, the release time may also be from about less than 1 day to about 180 days; from about 7 days to about 14 days; from about 14 days to about 28 days; from about 28 days to about 56 days; from about 56 days to about 90 days; from about 90 days to about 180 days.
The exemplary anti-infective agents, used alone or in combination, should be administered under the following dosing guidelines. The total amount (dose) of anti-infective agent in the composition can be in the range of about 0.01 μg-1 μg, or about 1 μg-10 μg, or about 10 μg-1 mg, or about 1 mg to 10 mg, or about 10 mg-100 mg, or about 100 mg to 250 mg, or about 250 mg-1000 mg. The dose (amount) of anti-infective agent per unit area of device or tissue surface to which the agent is applied may be in the range of about 0.01 μg/mm2 - 1 μg/mm2, or about 1 μg/mm2 - 10 μg/mm2, or about 10 μg/mm2 - 100 μg/mm2, or about 100 μg/mm2 to 250 μg/mm2, or about 250 μg/mm2 - 1000 μg/mm2. As different compositions will release the anti-infective agent at differing rates, the above dosing parameters should be utilized in combination with the release rate of the drug from the composition such that a minimum concentration of about 10"8 to 10"7, or about 10"7 to 1(T6 about 10"6 to 10"5 or about 1(T5 to 10'4of the agent is maintained on the tissue surface.
It should be readily evident based upon the discussions provided herein that combinations of anthracyclines (e.g., doxorubicin or mitoxantrone), fluoropyrimidines (e.g., 5-fluorouracil), folic acid antagonists (e.g., methotrexate), quinolones, and/or podophylotoxins (e.g., etoposide) may be utilized to enhance the antibacterial activity of the composition.
Hypertrophic Scars and Keloids
In another aspect, the subject compositions may be infiltrated into tissue adjacent to a device for use in treating hypertrophic scars and keloids. The subject compositions may contain a therapeutic agent (e.g., an anti-scarring and/or anti-infective agent).
Representative examples of implants for use in treating hypertrophic scars and keloids that may benefit from having the subject compositions infiltrated into adjacent tissue are provided above in conjunction with the coating of medical devices. Numerous agents or compositions for use with hypertrophic scar and keloid implants have been described above which may be infiltrated into the tissue adjacent to the device (preferably near the device-tissue interface).
The compositions may be a topical or injectable composition that includes an anti-scarring and/or anti-infective agent and a polymeric carrier suitable for application on or into hypertrophic scars or keloids. Incorporation of a fibrosis-inhibiting and/or anti-infective agent into a topical formulation or an injectable formulation is one approach to treat this condition. The topical formulation can be in the form of a solution, a suspension, an emulsion, a gel, an ointment, a cream, film or mesh. The injectable formulation can be in the form of a solution, a suspension, an emulsion or a gel. Polymeric and non-polymeric components that can be used to prepare these topical or injectable compositions are described above.
Therapeutic agents or pharmaceutical compositionsmay be infiltrated around devices used for hypertrophic scars and keloids by applying the composition directly and/or indirectly into and/or onto (a) tissue adjacent to the device used for hypertrophic scars and keloids; (b) the vicinity of the tissue interface with the device used for hypertrophic scars and keloids; (c) the region around the device used for hypertrophic scars and keloids; and (d) tissue surrounding the device used for hypertrophic scars and keloids. Methods for infiltrating the subject compositions into tissue adjacent to a device used for hypertrophic scars and keloids include delivering the composition: (a) to the surface of the device used for hypertrophic scars and keloids (e.g., as an injectable, paste, gel or mesh) during the implantation procedure; (b) to the surface of the tissue (e.g., as an injectable, paste, gel, in situ forming-gel or mesh) immediately prior to, or during, implantation of the device used for hypertrophic scars and keloids; (c) to the surface of the device used for hypertrophic scars and keloids and/or the tissue surrounding the implanted device used for hypertrophic scars and keloids (e.g., as an injectable, paste, gel, in situ forming gel or mesh) immediately after the implantation of the device used for hypertrophic scars and keloids; (d) by topical application of the composition into the anatomical space where the device used for hypertrophic scars and keloids may be placed (particularly useful for this embodiment is the use of polymeric carriers which release the therapeutic agent over a period ranging from several hours to several weeks - fluids, suspensions, emulsions, microemulsions, microspheres, pastes, gels, microparticulates, sprays, aerosols, solid implants and other formulations which release the agent may be delivered into the region where the device may be inserted); (e) via percutaneous injection into the tissue surrounding the device used for hypertrophic scars and keloids as a solution as an infusate or as a sustained release preparation; (f) by any combination of the aforementioned methods. Combination therapies (i.e., combinations of therapeutic agents and combinations with antithrombotic and/or antiplatelet agents) may also be used. In all cases it is understood that the subject compositions may be infiltrated into tissue adjacent to all or a portion of the device.
According to one aspect, any fibrosis-inhibiting and/or anti- infective agent described above may be utilized in the practice of the present invention. In one aspect of the invention, the subject compositions infiltrated into tissue adjacent to devices for the treatment of hypertrophic scars and keloids may be adapted to release an agent that inhibits one or more of the four general components of the process of fibrosis (or scarring), including: formation of new blood vessels (angiogenesis), migration and proliferation of connective tissue cells (such as fibroblasts or smooth muscle cells), deposition of extracellular matrix (ECM), and remodeling (maturation and organization of the fibrous tissue). By inhibiting one or more of the components of fibrosis (or scarring), the overgrowth of granulation tissue may be inhibited or reduced.
Examples of fibrosis-inhibiting agents for use in the present invention include the following: ZD-6474, AP-23573, synthadotin, S-0885, aplidine, ixabepilone, IDN-5390, SB-2723005, ABT-518, combretastatin, anecortave acetate, SB-715992, temsirolimus, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, as well as analogues and derivatives of the aforementioned.
Additional examples of anti-scarring agents which can be used include those having a high potency in the assays described herein (1- 10OnM IC50 range) such as isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, dolastatin 15, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17- DMAG, and tacrolimus; those having a mid-potency in the assays described herein (100-500 nM IC50 range) such as loteprednol etabonate, juglone, prednisolone, puromycin, 3-BAABE, cladribine, and mannose-6-phosphate; and those having a low potency in the assays described herein (500- 1000nm range IC50 range) such as 5-azacytidine, Ly333531(ruboxistaurin), and simvastatin.
The drug dose administered from the present compositions for prevention or inhibition of fibrosis in accordance with the present invention will depend on a variety of factors, including the type of formulation, the location of the treatment site, and the type of condition being treated. As devices for the treatment of hypertrophic scars and keloids are made in a variety of configurations and sizes, the exact dose administered will also vary with device size, surface area and design. However, certain principles can be applied in the application of this art. Drug dose can be calculated as a function of dose per unit area (of the treatment site), total drug dose administered can be measured and appropriate surface concentrations of active drug can be determined. Drugs are to be used at concentrations that range from several times more than to 50%, 20%, 10%, 5%, or even less than 1% of the concentration typically used in a single chemotherapeutic systemic dose application. In certain aspects, the anti-scarring agent is released from the composition in effective concentrations in a time period that may be measured from the time of infiltration into tissue adjacent to the device, which ranges from about less than 1 day to about 180 days. Generally, the release time may also be from about less than 1 day to about 180 days; from about 7 days to about 14 days; from about 14 days to about 28 days; from about 28 days to about 56 days; from about 56 days to about 90 days; from about 90 days to about 180 days.
The exemplary anti-fibrosing agents, used alone or in combination, should be administered under the following dosing guidelines. The total amount (dose) of anti-scarring agent in the composition can be in the range of about 0.01 μg-10 μg, or about 10 μg-10 mg, or about 10 mg- 250 mg, or about 250 mg-1000 mg, or about 1000 mg-2500 mg. The dose (amount) of anti-scarring agent per unit area of device or tissue surface to which the agent is applied may be in the range of about 0.01 μg/mm2 - 1 μg/mm2, or about 1 μg/mm2 - 10 μg/mm2, or about 10 μg/mm2 - 250 μg/mm2, or about 250 μg/mm2 - 1000 μg/mm2, or about 1000 μg/mm2 - 2500 μg/mm2.
According to another aspect, any anti-infective agent described above may be used in the practice of the present invention. Exemplary anti-infective agents include (A) anthracyclines (e.g., doxorubicin and mitoxantrone), (B) fluoropyrimidines (e.g., 5-FU), (C) folic acid antagonists (e.g., methotrexate), (D) podophylotoxins (e.g., etoposide), (E) camptothecins, (F) hydroxyureas, and (G) platinum complexes (e.g., cisplatin), as well as analogues and derivatives of the aforementioned.
The drug dose administered from the present compositions for prevention or inhibition of infection in accordance with the present invention will depend on a variety of factors, including the type of formulation, the location of the treatment site, and the type of condition being treated. However, certain principles can be applied in the application of this art. Drug dose can be calculated as a function of dose per unit area (of the treatment site), total drug dose administered can be measured and appropriate surface concentrations of active drug can be determined. Drugs are to be used at concentrations that range from several times more than to 50%, 20%, 10%, 5%, or even less than 1% of the concentration typically used in a single anti-infective systemic dose application. In certain aspects, the anti-infective agent is released from the composition in effective concentrations in a time period that may be measured from the time of infiltration into tissue adjacent to the device, which ranges from about less than 1 day to about 180 days. Generally, the release time may also be from about less than 1 day to about 180 days; from about 7 days to about 14 days; from about 14 days to about 28 days; from about 28 days to about 56 days; from about 56 days to about 90 days; from about 90 days to about 180 days.
The exemplary anti-infective agents, used alone or in combination, should be administered under the following dosing guidelines. The total amount (dose) of anti-infective agent in the composition can be in the range of about 0.01 μg-1 μg, or about 1 μg-10 μg, or about 10 μg-1 mg, or about 1 mg to 10 mg, or about 10 mg-100 mg, or about 100 mg to 250 mg, or about 250 mg-1000 mg. The dose (amount) of anti-infective agent per unit area of device or tissue surface to which the agent is applied may be in the range of about 0.01 μg/mm2 - 1 μg/mm2, or about 1 μg/mm2 - 10 μg/mm2, or about 10 μg/mm2 - 100 μg/mm2, or about 100 μg/mm2 to 250 μg/mm2, or about 250 μg/mm2 - 1000 μg/mm2. As different compositions will release the anti-infective agent at differing rates, the above dosing parameters should be utilized in combination with the release rate of the drug from the composition such that a minimum concentration of about 10"8 to 10"7, or about 10"7 to 10"6 about 10"6 to 10"5 or about 10"5 to 10"4 of the agent is maintained on the tissue surface.
It should be readily evident based upon the discussions provided herein that combinations of anthracyclines (e.g., doxorubicin or mitoxantrone), fluoropyrimidines (e.g., 5-fluorouracii), folic acid antagonists (e.g., methotrexate), quinolones, and/or podophylotoxins (e.g., etoposide) may be utilized to enhance the antibacterial activity of the composition.
Vascular Grafts
In one aspect, The present invention provides for infiltration of the subject agents or compositions into tissue adjacent to a vascular graft. Vascular graft devices having a composition containing a fibrosis-inhibiting and/or anti-infective agent infiltrated into adjacent tissue are capable of inhibiting or reducing the overgrowth of granulation tissue and/or inhibiting or preventing infection, which can improve the clinical efficacy of these devices.
Representative examples of vascular grafts that may benefit from having the subject compositions infiltrated into adjacent tissue are provided above in conjunction with the coating of medical devices. Numerous agents or compositions for use with vascular grafts have been described above which may be infiltrated into the tissue adjacent to the device (preferably near the device-tissue interface).
Therapeutic agents or pharmaceutical compositionsmay be infiltrated around implanted vascular grafts by applying the composition directly and/or indirectly into and/or onto (a) tissue adjacent to the vascular graft; (b) the vicinity of the vascular graft-tissue interface; (c) the region around the vascular graft; and (d) tissue surrounding the vascular graft. Methods for infiltrating the subject compositions into tissue adjacent to a vascular graft include delivering the composition: (a) to the vascular graft surface (e.g., as an injectable, paste, gel or mesh) during the implantation procedure; (b) to the surface of the tissue (e.g., as an injectable, paste, gel, in situ forming gel or mesh) immediately prior to, or during, implantation of the vascular graft; (c) to the surface of the vascular graft and/or the tissue surrounding the implanted vascular graft (e.g., as an injectable, paste, gel, in situ forming gel or mesh) immediately after the implantation of the vascular graft; (d) by topical application of the composition into the anatomical space where the vascular graft may be placed (particularly useful for this embodiment is the use of polymeric carriers which release the therapeutic agent over a period ranging from several hours to several weeks - fluids, suspensions, emulsions, microemulsions, microspheres, pastes, gels, microparticulates, sprays, aerosols, solid implants and other formulations which release the agent may be delivered into the region where the device may be inserted); (e) via percutaneous injection into the tissue surrounding the vascular graft as a solution as an infusate or as a sustained release preparation; (f) by any combination of the aforementioned methods. Combination therapies (i.e., combinations of therapeutic agents and combinations with antithrombotic and/or antiplatelet agents) may also be used. In ail cases it is understood that the subject compositions may be infiltrated into tissue adjacent to all or a portion of the device.
In addition to the fibrosis-inhibiting and/or anti-infective agent, the subject compositions infiltrated into tissue adjacent to vascular graft devices can also further contain an anti-inflammatory agent (e.g., dexamethazone or aspirin) and/or an anti-thrombotic agent (e.g., heparin, heparin complexes, hydrophobic heparin derivatives, dipyridamole, or aspirin). The combination of agents may be contained in the composition infiltrated into tissue adjacent to the vascular graft such that the thrombogenicity and/or fibrosis is reduced or inhibited. In certain embodiments, these agents may be contained in biodegradable polymers. For example, polymeric material that forms a gel in the pores and/or on the surface of the graft may be used, such as alginates, chitosan and chitosan sulfate, hyaluronic acid, dextran sulfate, PLURONIC polymers, chain extended PLURONIC polymers, polyester-polyether block copolymers of the various configurations (e.g., MePEG-PLA, PLA-PEG-PLA, and the like).
According to one aspect, any anti-scarring and/or anti-infective agent described above may be utilized in the practice of the present invention. In one aspect of the invention, the subject compositions infiltrated into tissue adjacent to vascular grafts may be adapted to release an agent that inhibits one or more of the four general components of the process of fibrosis (or scarring), including: formation of new blood vessels (angiogenesis), migration and proliferation of connective tissue cells (such as fibroblasts or smooth muscle cells), deposition of extracellular matrix (ECM), and remodeling (maturation and organization of the fibrous tissue). By inhibiting one or more of the components of fibrosis (or scarring), the overgrowth of granulation tissue may be inhibited or reduced.
Examples of fibrosis-inhibiting agents for use in the present invention include the following: ZD-6474, AP-23573, synthadotin, S-0885, aplidine, ixabepilone, IDN-5390, SB-2723005, ABT-518, combretastatin, anecortave acetate, SB-715992, temsirolimus, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, as well as analogues and derivatives of the aforementioned.
Additional examples of anti-scarring agents which can be used include those having a high potency in the assays described herein (1- 10OnM IC5O range) such as isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, dolastatin 15, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17- DMAG, and tacrolimus; those having a mid-potency in the assays described herein (100-500 nM IC5O range) such as loteprednol etabonate, juglone, prednisolone, puromycin, 3-BAABE, cladribine, and mannose-6-phosphate; and those having a low potency in the assays described herein (500- 1000nm range IC50 range) such as 5-azacytidine, Ly333531(ruboxistaurin), and simvastatin.
The drug dose administered from the present compositions for prevention or inhibition of fibrosis in accordance with the present invention will depend on a variety of factors, including the type of formulation, the location of the treatment site, and the type of condition being treated. As vascular grafts are made in a variety of configurations and sizes, the exact dose administered will also vary with device size, surface area and design. However, certain principles can be applied in the application of this art. Drug dose can be calculated as a function of dose per unit area (of the treatment site), total drug dose administered can be measured and appropriate surface concentrations of active drug can be determined. Drugs are to be used at concentrations that range from several times more than to 50%, 20%, 10%, 5%, or even less than 1% of the concentration typically used in a single chemotherapeutic systemic dose application. In certain aspects, the anti-scarring agent is released from the composition in effective concentrations in a time period that may be measured from the time of infiltration into tissue adjacent to the device, which ranges from about less than 1 day to about 180 days. Generally, the release time may also be from about less than 1 day to about 180 days; from about 7 days to about 14 days; from about 14 days to about 28 days; from about 28 days to about 56 days; from about 56 days to about 90 days; from about 90 days to about 180 days. The exemplary anti-fibrosing agents, used alone or in combination, should be administered under the following dosing guidelines. The total amount (dose) of anti-scarring agent in the composition can be in the range of about 0.01 μg-10 μg, or about 10 μg-10 mg, or about 10 mg- 250 mg, or about 250 mg-1000 mg, or about 1000 mg-2500 mg. The dose (amount) of anti-scarring agent per unit area of device or tissue surface to which the agent is applied may be in the range of about 0.01 μg/mm2 - 1 μg/mm2, or about 1 μg/mm2 - 10 μg/mm2, or about 10 μg/mm2 - 250 μg/mm2, or about 250 μg/mm2 - 1000 μg/mm2, or about 1000 μg/mm2 - 2500 μg/mm2.
According to another aspect, any anti-infective agent described above may be used in the practice of the present invention. Exemplary anti-infective agents include (A) anthracyclines (e.g., doxorubicin and mitoxantrone), (B) fluoropyrimidines (e.g., 5-FU), (C) folic acid antagonists (e.g., methotrexate), (D) podophylotoxins (e.g., etoposide), (E) camptbthecins, (F) hydroxyureas, and (G) platinum complexes (e.g., cisplatin), as well as analogues and derivatives of the aforementioned.
The drug dose administered from the present compositions for prevention or inhibition of infection in accordance with the present invention will depend on a variety of factors, including the type of formulation, the location of the treatment site, and the type of condition being treated. However, certain principles can be applied in the application of this art. Drug dose can be calculated as a function of dose per unit area (of the treatment site), total drug dose administered can be measured and appropriate surface concentrations of active drug can be determined. Drugs are to be used at concentrations that range from several times more than to 50%, 20%, 10%, 5%, or even less than 1% of the concentration typically used in a single anti-infective systemic dose application. In certain aspects, the anti- infective agent is released from the composition ϊn effective concentrations in a time period that may be measured from the time of infiltration into tissue adjacent to the device, which ranges from about less than 1 day to about 180 days. Generally, the release time may also be from about less than 1 day to about 180 days; from about 7 days to about 14 days; from about 14 days to about 28 days; from about 28 days to about 56 days; from about 56 days to about 90 days; from about 90 days to about 180 days.
The exemplary anti-infective agents, used alone or in combination, should be administered under the following dosing guidelines. The total amount (dose) of anti-infective agent in the composition can be in the range of about 0.01 μg-1 μg, or about 1 μg-10 μg, or about 10 μg-1 mg, or about 1 mg to 10 mg, or about 10 mg-100 mg, or about 100 mg to 250 mg, or about 250 mg-1000 mg. The dose (amount) of anti-infective agent per unit area of device or tissue surface to which the agent is applied may be in the range of about 0.01 μg/mm2 - 1 μg/mm2, or about 1 μg/mm2 - 10 μg/mm2, or about 10 μg/mm2 - 100 μg/mm2, or about 100 μg/mm2 to 250 μg/mm2, or about 250 μg/mm2 - 1000 μg/mm2. As different compositions will release the anti-infective agent at differing rates, the above dosing parameters should be utilized in combination with the release rate of the drug from the composition such that a minimum concentration of about 10"8 to 10"7, or about 10"7 to 10"6 about 10~6 to 10'5 or about 10'5 to 10"4 of the agent is maintained on the tissue surface.
It should be readily evident based upon the discussions provided herein that combinations of anthracyclines (e.g., doxorubicin or mitoxantrone), fluoropyrimidines (e.g., 5-fluorouracil), folic acid antagonists (e.g., methotrexate), quinolones, and/or podophylotoxins (e.g., etoposide) may be utilized to enhance the antibacterial activity of the composition.
Hemodialysis Access Devices
In one aspect, the subject compositions may be infiltrated into tissue adjacent to a hemodialysis access device. The subject compositions may contain a therapeutic agent (e.g., an anti-scarring and/or anti-infective agent). Hemodialysis dialysis access devices that include a fibrosis- inhibiting and/or anti-infective agent are capable of inhibiting or reducing the overgrowth of granulation tissue and/or inhibiting or preventing infection, which can improve the clinical efficacy of these devices.
Representative examples of hemodialysis access devices that may benefit from having the subject compositions infiltrated into adjacent tissue are provided above in conjunction with the coating of medical devices. Numerous agents or compositions for use with hemodialysis access devices have been described above which may be infiltrated into the tissue adjacent to the device (preferably near the device-tissue interface).
Therapeutic agents or pharmaceutical compositionsmay be infiltrated around implanted hemodialysis access devices by applying the composition directly and/or indirectly into and/or onto (a) tissue adjacent to the hemodialysis access device; (b) the vicinity of the hemodialysis access device -tissue interface; (c) the region around the hemodialysis access device; and (d) tissue surrounding the hemodialysis access device. Methods for infiltrating the subject compositions into tissue adjacent to a hemodialysis access device include delivering the composition: (a) to the hemodialysis access device surface (e.g., as an injectable, paste, gel or mesh) during the implantation procedure; (b) to the surface of the tissue (e.g., as an injectable, paste, gel, in situ forming gel or mesh) immediately prior to, or during, implantation of the hemodialysis access device; (c) to the surface of the hemodialysis access device and/or the tissue surrounding the implanted hemodialysis access device (e.g., as an injectable, paste, gel, in situ forming gel or mesh) immediately after the implantation of the hemodialysis access device; (d) by topical application of the composition into the anatomical space where the hemodialysis access device may be placed (particularly useful for this embodiment is the use of polymeric carriers which release the therapeutic agent over a period ranging from several hours to several weeks - fluids, suspensions, emulsions, microemulsions, microspheres, pastes, gels, microparticulates, sprays, aerosols, solid implants and other formulations which release the agent may be delivered into the region where the device may be inserted); (e) via percutaneous injection into the tissue surrounding the hemodialysis access device as a solution as an infusate or as a sustained release preparation; (f) by any combination of the aforementioned methods. Combination therapies (Ae., combinations of therapeutic agents and combinations with antithrombotic and/or antiplatelet agents) may also be used. In all cases it is understood that the subject compositions may be infiltrated into tissue adjacent to all or a portion of the device.
In addition to the fibrosis-inhibiting and/or anti-infective agent, subject compositions infiltrated into tissue adjacent to hemodialysis access devices can also further contain an anti-inflammatory agent (e.g., dexamethazone or aspirin) and/or an antithrombotic agent (e.g., heparin, heparin complexes, hydrophobic heparin derivatives, dipyridamole, or aspirin).
According to the one aspect, any anti-scarring and/or anti- infective agent described above may be utilized in the practice of the present invention. In one aspect of the invention, the subject compositions infiltrated into tissue adjacent to hemodialysis access devices may be adapted to release an agent that inhibits one or more of the four general components of the process of fibrosis (or scarring), including: formation of new blood vessels (angiogenesis), migration and proliferation of connective tissue cells (such as fibroblasts or smooth muscle cells), deposition of extracellular matrix (ECM)1 and remodeling (maturation and organization of the fibrous tissue). By inhibiting one or more of the components of fibrosis (or scarring), the overgrowth of granulation tissue may be inhibited or reduced.
Examples of fibrosis-inhibiting agents for use in the present invention include the following: ZD-6474, AP-23573, synthadotin, S-0885, aplidine, ixabepilone, IDN-5390, SB-2723005, ABT-518, combretastatin, anecortave acetate, SB-715992, temsirolimus, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, as well as analogues and derivatives of the aforementioned.
Additional examples of anti-scarring agents which can be used include those having a high potency in the assays described herein (1- 10OnM IC50 range) such as isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, dolastatin 15, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17- DMAG, and tacrolimus; those having a mid-potency in the assays described herein (100-500 nM IC50 range) such as loteprednol etabonate, juglone, prednisolone, puromycin, 3-BAABE, cladribine, and mannose-6-phosphate; and those having a low potency in the assays described herein (500- 1000nm range IC50 range) such as 5-azacytidine, Ly333531(ruboxistaurin), and simvastatin.
The drug dose administered from the present compositions for prevention or inhibition of fibrosis in accordance with the present invention will depend on a variety of factors, including the type of formulation, the location of the treatment site, and the type of condition being treated. As hemodialysis access devices are made in a variety of configurations and sizes, the exact dose administered will also vary with device size, surface area and design. However, certain principles can be applied in the application of this art. Drug dose can be calculated as a function of dose per unit area (of the treatment site), total drug dose administered can be measured and appropriate surface concentrations of active drug can be determined. Drugs are to be used at concentrations that range from several times more than to 50%, 20%, 10%, 5%, or even less than 1% of the concentration typically used in a single chemotherapeutic systemic dose application. In certain aspects, the anti-scarring agent is released from the composition in effective concentrations in a time period that may be measured from the time of infiltration into tissue adjacent to the device, which ranges from about less than 1 day to about 180 days. Generally, the release time may also be from about less than 1 day to about 180 days; from about 7 days to about 14 days; from about 14 days to about 28 days; from about 28 days to about 56 days; from about 56 days to about 90 days; from about 90 days to about 180 days.
The exemplary anti-fibrosing agents, used alone or in combination, should be administered under the following dosing guidelines. The total amount (dose) of anti-scarring agent in the composition can be in the range of about 0.01 μg-10 μg, or about 10 μg-10 mg, or about 10 mg- 250 mg, or about 250 mg-1000 mg, or about 1000 mg-2500 mg. The dose (amount) of anti-scarring agent per unit area of device or tissue surface to which the agent is applied may be in the range of about 0.01 μg/mm2 - 1 μg/mm2, or about 1 μg/mm2 - 10 μg/mm2, or about 10 μg/mm2 - 250 μg/mm2, or about 250 μg/mm2 - 1000 μg/mm2, or about 1000 μg/mm2 - 2500 μg/mm2.
According to another aspect, any anti-infective agent described above may be used in the practice of the present invention. Exemplary anti-infective agents include (A) anthracyclines (e.g., doxorubicin and mitoxantrone), (B) fluoropyrimidines (e.g., 5-FU), (C) folic acid antagonists (e.g., methotrexate), (D) podophylotoxins (e.g., etoposide), (E) camptothecins, (F) hydroxyureas, and (G) platinum complexes (e.g., cisplatin), as well as analogues and derivatives of the aforementioned.
The drug dose administered from the present compositions for prevention or inhibition of infection in accordance with the present invention will depend on a variety of factors, including the type of formulation, the location of the treatment site, and the type of condition being treated. However, certain principles can be applied in the application of this art. Drug dose can be calculated as a function of dose per unit area (of the treatment site), total drug dose administered can be measured and appropriate surface concentrations of active drug can be determined. Drugs are to be used at concentrations that range from several times more than to 50%, 20%, 10%, 5%, or even less than 1% of the concentration typically used in a single anti-infective systemic dose application. In certain aspects, the anti- infective agent is released from the composition in effective concentrations in a time period that may be measured from the time of infiltration into tissue adjacent to the device, which ranges from about less than 1 day to about 180 days. Generally, the release time may also be from about less than 1 day to about 180 days; from about 7 days to about 14 days; from about 14 days to about 28 days; from about 28 days to about 56 days; from about 56 days to about 90 days; from about 90 days to about 180 days.
The exemplary anti-infective agents, used alone or in combination, should be administered under the following dosing guidelines. The total amount (dose) of anti-infective agent in the composition can be in the range of about 0.01 μg-1 μg, or about 1 μg-10 μg, or about 10 μg-1 mg, or about 1 mg to 10 mg, or about 10 mg-100 mg, or about 100 mg to 250 mg, or about 250 mg-1000 mg. The dose (amount) of anti-infective agent per unit area of device or tissue surface to which the agent is applied may be in the range of about 0.01 μg/mm2 - 1 μg/mm2, or about 1 μg/mm2 - 10 μg/mm2, or about 10 μg/mm2 - 100 μg/mm2, or about 100 μg/mm2 to 250 μg/mm2, or about 250 μg/mm2 - 1000 μg/mm2. As different compositions will release the anti-infective agent at differing rates, the above dosing parameters should be utilized in combination with the release rate of the drug from the composition such that a minimum concentration of about 10~8 to 10"7, or about 10"7 to 106 about 10"6 to 10-5 or about lO'5 to 10"4Of the agent is maintained on the tissue surface.
It should be readily evident based upon the discussions provided herein that combinations of anthracyclines (e.g., doxorubicin or mitoxantrone), fluoropyrimidines (e.g., 5-fluorouracil), folic acid antagonists (e.g., methotrexate), quinolones, and/or podophylotoxins (e.g., etoposide) may be utilized to enhance the antibacterial activity of the composition. Films and Meshes
In one aspect, the subject compositions may be infiltrated into tissue adjacent to a film or mesh. The subject compositions may contain a therapeutic agent (e.g., an anti-scarring and/or anti-infective agent). Infiltration of the subject composition comprising a fibrosis-inhibiting agent and/or anti-infective agent into tissue adjacent to the film or mesh can minimize fibrosis (or scarring) in the vicinity of the implant and may reduce or prevent the formation of adhesions between the implant and the surrounding tissue and/or may inhibit or prevent infection in the vicinity of the implant site. In certain aspects, the film or mesh may be used as a drug-delivery vehicle (e.g., as a perivascular delivery device for the prevention of neointimal hyperplasia at an anastomotic site).
Representative examples of fims and meshes that may benefit from having the subject compositions infiltrated into adjacent tissue are provided above in conjunction with the coating of medical devices. Numerous agents or compositions for use with films and meshes have been described above which may be infiltrated into the tissue adjacent to the device (preferably near the device-tissue interface).
A variety of polymeric compositions have been described that may be used in conjunction with the films and meshes of the invention. Such compositions may be in the form of, for example, gels, sprays, liquids, and pastes, or may be polymerized from monomeric or prepolymeric constituents in situ. For example, the composition may be a polymeric tissue coating which is formed by applying a polymerization initiator to the tissue and then covering it with a water-soluble macromer that is polymerizable using free radical initiators under the influence of UV light. See, e.g., U.S. Patent Nos. 6,177,095 and 6,083,524. The composition may be an aqueous composition including a surfactant, pentoxifylline and a polyoxyalkylene polyether. See, e.g., U.S. Patent No. 6,399,624. The composition may be a hydrogel-forming, self-solvating, absorbable polyester copolymers capable of selective, segmental association into compliant hydrogels mass upon contact with an aqueous environment. See, e.g., U.S. Patent No. 5,612,052. The composition may be composed of fluent pre- polymeric material that is emitted to the tissue surface and then exposed to activating energy in situ to initiate conversion of the applied material to non- fluent polymeric form. See, e.g., U.S. Patent Nos. 6,004,547 and 5,612,050. The composition may be composed of a gas mixture of oxygen present in a volume ratio of 1 to 20%. See, e.g., U.S. Patent No. 6,428,500. The composition may be composed of an anionic polymer having an acid sulfate and sulfur content greater than 5% which acts to inhibit monocyte or macrophage invasion. See, e.g., U.S. Patent No. 6,417,173. The composition may be composed of a non-gelling polyoxyalkylene composition with or without a therapeutic agent. See, e.g., U.S. Patent No. 6,436,425. The composition may be coated onto tissue surfaces and may be composed of an aqueous solution of a hydrophilic, polymeric material (e.g., polypeptides or polysaccharide) having greater than 50,000 molecular weight and a concentration range of 0.01% to 15% by weight. See, e.g., U.S. Patent No. 6,464,970.
Other representative examples of polymeric compositions which may be infiltrated into tissue adjacent to the film or mesh include poly(ethylene glycol)-based systems, hyaluronic acid and crosslinked hyaluronic acid compositions. These compositions can be applied as the final composition or they can be applied as materials that form crosslinked gel in situ.
Other compositions that can be used in conjunction with films and meshes, include, but are not limited to: (a) sprayable PEG-containing formulations such as COSEAL, SPRAYGEL, FOCALSEAL or DURASEAL; (b) hyaluronic acid-containing formulations such as RESTYLANE, HYLAFORM, PERLANE, SYNVISC, SEPRAFILM, SEPRACOAT, INTERGEL, (c) polymeric gels such as REPEL or FLOWGEL, (d) dextran sulfate gels such as the ADCON range of products, (e) lipid based compositions such as ADSURF (Brittania Pharmaceuticals). The film or mesh (or device comprising the film or mesh) may be made sterile either by preparing them under aseptic environment and/or they may be terminally sterilized using methods known in the art, such as gamma radiation or electron beam sterilization methods or a combination of both of these methods.
Films and meshes may be applied to any bodily conduit or any tissue that may be prone to the development of fibrosis or intimal hyperplasia. Prior to implantation, the film or mesh may be trimmed or cut from a sheet of bulk material to match the configuration of the widened foramen, canal, or dissection region, or at a minimum, to overlay the exposed tissue area. The film or mesh may be bent or shaped to match the particular configuration of the placement region. The film or mesh may also be rolled in a cuff shape or cylindrical shape and placed around the exterior periphery of the desired tissue. The film or mesh may be provided in a relatively large bulk sheet and then cut into shapes to mold the particular structure and surface topography of the tissue or device to be wrapped. Alternatively, the film or mesh may be pre-shaped into one or more patterns for subsequent use. The films and meshes may be typically rectangular in shape and be placed at the desired location within the surgical site by direct surgical placement or by endoscopic techniques. The film or mesh may be secured into place by wrapping it onto itself {i.e., self-adhesive), or by securing it with sutures, staples, sealant, and the like. Alternatively, the film or mesh may adhere readily to tissue and therefore, additional securing mechanisms may not be required.
The films or meshes of the invention may be used for a variety of indications, including, without limitation: (a) prevention of surgical adhesions between tissues following surgery (e.g., gynecologic surgery, vasovasostomy, hernia repair, nerve root decompression surgery and laminectomy); (b) prevention of hypertrophic scars or keloids {e.g., resulting from tissue burns or other wounds); (c) prevention of intimal hyperplasia and/or restenosis (e.g., resulting from insertion of vascular grafts or hemodialysis access devices); (d) may be used in affiliation with devices and implants that lead to scarring as described herein (e.g., as a sleeve or mesh around a breast implant to reduce or inhibit scarring); (e) prevention of infection (e.g., resulting from tissue burns, surgery or other wounds); or (f) may be used in affiliate with devices and implants that lead to infection as described herein.
In one embodiment, films or meshes may be used to prevent adhesions that occur between tissues following surgery, injury or disease. Adhesion formation, a complex process in which bodily tissues that are normally separate grow together, occurs most commonly as a result of surgical intervention and/or trauma. Generally, adhesion formation is an inflammatory reaction in which factors are released, increasing vascular permeability and resulting in fibrinogen influx and fibrin deposition. This deposition forms a matrix that bridges the abutting tissues. Fibroblasts accumulate, attach to the matrix, deposit collagen and induce angiogenesis. If this cascade of events can be prevented within 4 to 5 days following surgery, then adhesion formation can be inhibited. Adhesion formation or unwanted scar tissue accumulation and encapsulation complicates a variety of surgical procedures and virtually any open or endoscopic surgical procedure in the abdominal or pelvic cavity. Encapsulation of surgical implants also complicates breast reconstruction surgery, joint replacement surgery, hernia repair surgery, artificial vascular graft surgery, and neurosurgery. In each case, the implant becomes encapsulated by a fibrous connective tissue capsule which compromises or impairs the function of the surgical implant (e.g., breast implant, artificial joint, surgical mesh, vascular graft, dural patch). Chronic inflammation and scarring also occurs during surgery to correct chronic sinusitis or removal of other regions of chronic inflammation (e.g., foreign bodies, infections (fungal, mycobacterium). Surgical procedures that may lead to surgical adhesions may include cardiac, spinal, neurologic, pleural, thoracic and gynecologic surgeries. However, adhesions may also develop as a result of other processes, including, but not limited to, non-surgical mechanical injury, ischemia, hemorrhage, radiation treatment, infection-related inflammation, pelvic inflammatory disease and/or foreign body reaction. This abnormal scarring interferes with normal physiological functioning and, in come cases, can force and/or interfere with follow-up, corrective or other surgical operations. For example, these post-operative surgical adhesions occur in 60 to 90% of patients undergoing major gynecologic surgery and represent one of the most common causes of intestinal obstruction in the industrialized world. These adhesions are a major cause of failed surgical therapy and are the leading cause of bowel obstruction and infertility. Other adhesion- treated complications include chronic pelvic pain, urethral obstruction and voiding dysfunction.
Currently, preventative therapies, administered 4 to 5 days following surgery, are used to inhibit adhesion formation. Various modes of adhesion prevention have been examined, including (1) prevention of fibrin deposition, (2) reduction of local-tissue inflammation, and (3) removal of fibrin deposits. Fibrin deposition is prevented through the use of physical adhesion barriers that are either mechanical or comprised of viscous solutions. Although many investigators are utilizing adhesion prevention barriers, a number of technical difficulties exist.
In one aspect, the present invention provides films and meshes having the subject composition comprising an anti-scarring agent infiltrated into adjacent tissue for use as surgical adhesion barriers.
In one aspect, films and meshes having the subject composition comprising an anti-scarring agent infiltrated into adjacent tissue may be used to prevent surgical adhesions in the epidural and dural tissue which is a factor contributing to failed back surgeries and complications associated with spinal injuries (e.g., compression and crush injuries). Scar formation within dura and around nerve roots has been implicated in rendering subsequent spine operations technically more difficult. To gain access to the spinal foramen during back surgeries, vertebral bone tissue is often disrupted. Back surgeries, such as laminectomies and diskectomies, often leave the spinal dura exposed and unprotected. As a result, scar tissue frequently forms between the dura and the surrounding tissue. This scar is formed from the damaged erector spinae muscles that overlay the laminectomy site. This results in adhesion development between the muscle tissue and the fragile dura, thereby, reducing mobility of the spine and nerve roots which leads to pain and slow post-operative recovery. To circumvent adhesion development, a scar-reducing barrier may be inserted between the dural sleeve and the paravertebral musculature post- laminotomy. This reduces cellular and vascular invasion into the epidural space from the overlying muscle and exposed cancellous bone and thus, reduces the complications associated with the canal housing the spinal chord and/or nerve roots.
In another aspect, films and meshes having the subject composition comprising an anti-scarring agent infiltrated into adjacent tissue may be used to prevent the fibrosis from occurring between a hernia repair mesh and the surrounding tissue. Hernias are abnormal protrusions (outpouchings) of an organ or other body structure through a defect or natural opening in a covering membrane, muscle or bone. Hernias themselves are not dangerous, but can become extremely problematic if they become incarcerated. Surgical prostheses used in hernia repair (referred to herein as "hernia meshes") include prosthetic mesh-or gauze- like materials, which support the repaired hernia or other body structures during the healing process. Hernias are often repaired surgically to prevent complications. Conditions in which a hernia mesh may need to be used include, without limitation, the repair of inguinal (i.e., groin), umbilical, ventral, femoral, abdominal, diaphragmatic, epigastric, gastroesophageal, hiatal, intermuscular, mesenteric, paraperitoneal, rectovaginal, rectocecal, uterine, and vesical hernias. Hernia repair typically involves returning the viscera to its normal location and the defect in the wall is primarily closed with sutures, but for bigger gaps, a mesh is placed over the defect to close the hernia opening. Infiltration of the subject composition comprising an anti-scarring agent into tissue adjacent to a hernia repair mesh may reduce or prevent fibrosis proximate to the implanted hernia mesh, thereby minimizing the possibility of adhesions between the abdominal wall or other tissues and the mesh itself, and reducing further complications and abdominal pain.
In yet another aspect, films or meshes having the subject composition comprising an anti-scarring agent infiltrated into adjacent tissue may be used to prevent hypertrophic scars or keloids (e.g., resulting from tissue bums or other wounds). Hypertrophic scars and keloids are the result of an excessive fibroproliferative wound healing response. Briefly, healing of wounds and scar formation occurs in three phases: inflammation, proliferation, and maturation. The first phase, inflammation, occurs in response to an injury which is severe enough to break the skin. During this phase, which lasts 3 to 4 days, blood and tissue fluid form an adhesive coagulum and fibrinous network which serves to bind the wound surfaces together. This is then followed by a proliferative phase in which there is ingrowth of capillaries and connective tissue from the wound edges, and closure of the skin defect. Finally, once capillary and fibroblastic proliferation has ceased, the maturation process begins wherein the scar contracts and becomes less cellular, less vascular, and appears flat and white. This final phase may take between 6 and 12 months. If too much connective tissue is produced and the wound remains persistently cellular, the scar may become red and raised. If the scar remains within the boundaries of the original wound it is referred to as a hypertrophic scar, but if it extends beyond the original scar and into the surrounding tissue, the lesion is referred to as a keloid. Hypertrophic scars and keloids are produced during the second and third phases of scar formation. Several wounds are particularly prone to excessive endothelial and fibroblastic proliferation, including burns, open wounds, and infected wounds. With hypertrophic scars, some degree of maturation occurs and gradual improvement occurs. In the case of keloids however, an actual tumor is produced which can become quite large. Spontaneous improvement in such cases rarely occurs. A film or mesh having the subject composition comprising an anti-scarring agent infiltrated into adjacent tissue may be placed in contact with a wound or burn site in order to prevent formation of hypertrophic scar or keloids.
In yet another aspect, films and meshes having the subject composition comprising an anti-scarring agent infiltrated into adjacent tissue are provided that may be used for delivering an anti-scarring agent to an external portion (surface) of a body passageway or cavity. Examples of body passageways include arteries, veins, the heart, the esophagus, the stomach, the duodenum, the small intestine, the large intestine, biliary tracts, the ureter, the bladder, the urethra, lacrimal ducts, the trachea, bronchi, bronchiole, nasal airways, Eustachian tubes, the external auditory mayal, vas deferens and fallopian tubes. Examples of cavities include the abdominal cavity, the buccal cavity, the peritoneal cavity, the pericardial cavity, the pelvic cavity, perivisceral cavity, pleural cavity and uterine cavity.
Examples of conditions that may be treated or prevented with films and meshes having the subject composition comprising an anti- scarring agent infiltrated into adjacent tissue include iatrogenic complications of arterial and venous catheterization, complications of vascular dissection, complications of gastrointestinal passageway rupture and dissection, restonotic complications associated with vascular surgery (e.g., bypass surgery), and intimal hyperplasia.
In one aspect, an anti-scarring agent may be delivered from the subject composition infiltrated into tissue adjacent to a film or mesh to the external walls of body passageways or cavities for the purpose of preventing and/or reducing a proliferative biological response that may obstruct or hinder the optimal functioning of the passageway or cavity, including, for example, iatrogenic complications of arterial and venous catheterization, aortic dissection, cardiac rupture, aneurysm, cardiac valve dehiscence, graft placement (e.g., A-V-bypass, peripheral bypass, CABG), fistula formation, passageway rupture and surgical wound repair.
The films or meshes may be used in the form of a perivascular wrap to prevent restenosis at anastomotic sites resulting from insertion of vascular grafts or hemodialysis access devices. In this case, perivascular wraps having the subject composition containing an anti-scarring agent infiltrated into adjacent tissue may be used in conjunction with a vascular graft to inhibit scarring at an anastomotic site. These films or meshes may be placed or wrapped in a perivascular (periadventitial) manner around the outside of the anastomosis at the time of surgery. Film and mesh implants having the subject composition containing an anti-scarring agent infiltrated into adjacent tissue may be used with synthetic bypass grafts (femoral- popliteal, femoral-femoral, axillary-femoral etc.), vein grafts (peripheral and coronary), internal mammary (coronary) grafts or hemodialysis grafts (AV fistulas, AV access grafts).
In order to further the understanding of such conditions, representative complications leading to compromised body passageway or cavity integrity are discussed in more detail below.
In one aspect, the subject compositions may be infiltrated into tissue adjacent to a coronary artery bypass graft ("CABG"). The subject compositions may contain a therapeutic agent (e.g., an anti-scarring and/or anti-infective agent).
In one aspect, the subject compositions may be infiltrated into tissue adjacent to a peripheral bypass surgery site. The subject compositions may contain a therapeutic agent (e.g., an anti-scarring and/or anti-infective agent).
In one aspect, the subject compositions may be infiltrated into tissue adjacent to an arterio-venous fistula. The subject compositions may contain a therapeutic agent (e.g., an anti-scarring and/or anti-infective agent). In one aspect, the subject compositions may be infiltrated into tissue adjacent to a peripheral bypass surgery site. The subject compositions may contain a therapeutic agent (e.g., an anti-scarring and/or anti-infective agent).
In one aspect, the subject compositions may be infiltrated into tissue adjacent to an anastomotic closure device. The subject compositions may contain a therapeutic agent (e.g., an anti-scarring and/or anti-infective agent).
In one aspect, the subject compositions may be infiltrated into tissue adjacent to a transplant surgery site. The subject compositions may contain a therapeutic agent (e.g., an anti-scarring and/or anti-infective agent).
According to the one aspect, any anti-scarring agent described above may be utilized in the practice of the present invention. In one aspect of the invention, the subject compositions infiltrated into tissue adjacent to films and meshes may be adapted to contain and/or release an agent that inhibits one or more of the four general components of the process of fibrosis (or scarring), including: formation of new blood vessels (angiogenesis), migration and proliferation of connective tissue cells (such as fibroblasts or smooth muscle cells), deposition of extracellular matrix (ECM), and remodeling (maturation and organization of the fibrous tissue).
Examples of fibrosis-inhibiting agents for use in the present invention include the following: ZD-6474, AP-23573, synthadotin, S-0885, aplidine, ixabepilone, IDN-5390, SB-2723005, ABT-518, combretastatin, anecortave acetate, SB-715992, temsirolimus, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, as well as analogues and derivatives of the aforementioned.
Additional examples of anti-scarring agents which can be used include those having a high potency in the assays described herein (1- 10OnM IC50 range) such as isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, dolastatin 15, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17- DMAG, and tacrolimus; those having a mid-potency in the assays described herein (100-500 nM IC50 range) such as loteprednoi etabonate, juglone, prednisolone, puromycin, 3-BAABE, cladribine, and mannose-6-phosphate; and those having a low potency in the assays described herein (500- 1000nm range IC50 range) such as 5-azacytidine, Ly333531(ruboxistaurin), and simvastatin.
The drug dose administered from the present compositions for prevention or inhibition of fibrosis in accordance with the present invention will depend on a variety of factors, including the type of formulation, the location of the treatment site, and the type of condition being treated. As films and meshes are made in a variety of configurations and sizes, the exact dose administered will also vary with device size, surface area and design. However, certain principles can be applied in the application of this art. Drug dose can be calculated as a function of dose per unit area (of the treatment site), total drug dose administered can be measured and appropriate surface concentrations of active drug can be determined. Drugs are to be used at concentrations that range from several times more than to 50%, 20%, 10%, 5%, or even less than 1 % of the concentration typically used in a single chemotherapeutic systemic dose application. In certain aspects, the anti-scarring agent is released from the composition in effective concentrations in a time period that may be measured from the time of infiltration into tissue adjacent to the device, which ranges from about less than 1 day to about 180 days. Generally, the release time may also be from about less than 1 day to about 180 days; from about 7 days to about 14 days; from about 14 days to about 28 days; from about 28 days to about 56 days; from about 56 days to about 90 days; from about 90 days to about 180 days.
The exemplary anti-fibrosing agents, used alone or in combination, should be administered under the following dosing guidelines. The total amount (dose) of. anti-scarring agent in the composition can be in the range of about 0.01 μg-10 μg, or about 10 μg-10 mg, or about 10 mg- 250 mg, or about 250 mg-1000 mg, or about 1000 mg-2500 mg. The dose (amount) of anti-scarring agent per unit area of device or tissue surface to which the agent is applied may be in the range of about 0.01 μg/mm2 - 1 μg/mm2, or about 1 μg/mm2 - 10 μg/mm2, or about 10 μg/mm2 - 250 μg/mm2, or about 250 μg/mm2 - 1000 μg/mm2, or about 1000 μg/mm2 - 2500 μg/mm2.
According to another aspect, any anti-infective agent described above may be used in the practice of the present invention. Exemplary anti-infective agents include (A) anthracyclines (e.g., doxorubicin and mitoxantrone), (B) fluoropyrimidines (e.g., 5-FU), (C) folic acid antagonists (e.g., methotrexate), (D) podophylotoxins (e.g., etoposide), (E) camptothecins, (F) hydroxyureas, and (G) platinum complexes (e.g., cisplatin), as well as analogues and derivatives of the aforementioned.
The drug dose administered from the present compositions for prevention or inhibition of infection in accordance with the present invention will depend on a variety of factors, including the type of formulation, the location of the treatment site, and the type of condition being treated. However, certain principles can be applied in the application of this art. Drug dose can be calculated as a function of dose per unit area (of the treatment site), total drug dose administered can be measured and appropriate surface concentrations of active drug can be determined. Drugs are to be used at concentrations that range from several times more than to 50%, 20%, 10%, 5%, or even less than 1% of the concentration typically used in a single anti-infective systemic dose application. In certain aspects, the anti- infective agent is released from the composition in effective concentrations in a time period that may be measured from the time of infiltration into tissue adjacent to the device, which ranges from about less than 1 day to about 180 days. Generally, the release time may also be from about less than 1 day to about 180 days; from about 7 days to about 14 days; from about 14 days to about 28 days; from about 28 days to about 56 days; from about 56 days to about 90 days; from about 90 days to about 180 days.
The exemplary anti-infective agents, used alone or in combination, should be administered under the following dosing guidelines. The total amount (dose) of anti-infective agent in the composition can be in the range of about 0.01 μg-1 μg, or about 1 μg-10 μg, or about 10 μg-1 mg, or about 1 mg to 10 mg, or about 10 mg-100 mg, or about 100 mg to 250 mg, or about 250 mg-1000 mg. The dose (amount) of anti-infective agent per unit area of device or tissue surface to which the agent is applied may be in the range of about 0.01 μg/mm2 - 1 μg/mm2, or about 1 μg/mm2 - 10 μg/mm2, or about 10 μg/mm2 - 100 μg/mm2, or about 100 μg/mm2 to 250 μg/mm2, or about 250 μg/mm2 - 1000 μg/mm2. As different compositions will release the anti-infective agent at differing rates, the above dosing parameters should be utilized in combination with the release rate of the drug from the composition such that a minimum concentration of about 10"8 to 10"7, or about 10r7 to iθ"6 about 10"6 to 10"5or about lO'5 to 10'4 of the agent is maintained on the tissue surface.
It should be readily evident based upon the discussions provided herein that combinations of anthracyclines (e.g., doxorubicin or mitoxantrone), fluoropyrimidines (e.g., 5-fluorouracil), folic acid antagonists (e.g., methotrexate), quinolones, and/or podophylotoxins (e.g., etoposide) may be utilized to enhance the antibacterial activity of the composition.
Glaucoma Drainage Devices
In one aspect, the subject compositions may be infiltrated into tissue adjacent to a glaucoma drainage device. The subject compositions may contain a therapeutic agent (e.g., an anti-scarring and/or anti-infective agent).
Representative examples glaucoma drainage devides that may benefit from having the subject compositions infiltrated into adjacent tissue are provided above in conjunction with the coating of medical devices. Numerous agents or compositions for use with glaucoma drainage devices have been described above which may be infiltrated into the tissue adjacent to the device (preferably near the device-tissue interface).
Therapeutic agents or pharmaceutical compositionsmay be infiltrated around implanted glaucoma drainage devices by applying the composition directly and/or indirectly into and/or onto (a) tissue adjacent to the glaucoma drainage device; (b) the vicinity of the glaucoma drainage device-tissue interface; (c) the region around the glaucoma drainage device; and (d) tissue surrounding the glaucoma drainage device. Methods for infiltrating the subject compositions into tissue adjacent to a glaucoma drainage device include delivering the composition: (a) to the glaucoma drainage device surface (e.g., as an injectable, paste, gel or mesh) during the implantation procedure; (b) to the surface of the tissue (e.g., as an injectable, paste, gel, in situ forming gel or mesh) immediately prior to, or during, implantation of the glaucoma drainage device; (c) to the surface of the glaucoma drainage device and/or the tissue surrounding the implanted glaucoma drainage device (e.g., as an injectable, paste, gel, in situ forming gel or mesh) immediately after the implantation of the glaucoma drainage device; (d) by topical application of the composition into the anatomical space where the glaucoma drainage device may be placed (particularly useful for this embodiment is the use of polymeric carriers which release the therapeutic agent over a period ranging from several hours to several weeks - fluids, suspensions, emulsions, microemulsions, microspheres, pastes, gels, microparticulates, sprays, aerosols, solid implants and other formulations which release the agent may be delivered into the region where the device may be inserted); (e) via percutaneous injection into the tissue surrounding the glaucoma drainage device as a solution as an infusate or as a sustained release preparation; (f) by any combination of the aforementioned methods. Combination therapies (i.e., combinations of therapeutic agents and combinations with antithrombotic and/or antiplatelet agents) may also be used. In all cases it is understood that the subject compositions may be infiltrated into tissue adjacent to all or a portion of the device.
In one aspect, the methods above can be used to infiltrate the subject composition into tissue adjacent to all or portions of the plate of the device.
In another aspect, the methods above can be used to infiltrate the subject composition into tissue adjacent to all or portions of the tube of the device.
In yet another aspect, the methods above can be used to infiltrate the subject composition into tissue adjacent to all or potions of both the plate and the tube of the device.
According to the present invention, any fibrosis-inhibiting and/or anti-infective agent described above can be utilized in the practice of the present invention. In one aspect of the invention, the subject compositions infiltrated into tissue adjacent to glaucoma drainage devices may be adapted to release an agent that inhibits one or more of the four general components of the process of fibrosis (or scarring), including: formation of new blood vessels (angiogenesis), migration and proliferation of connective tissue cells (such as fibroblasts or smooth muscle cells), deposition of extracellular matrix (ECM), and remodeling (maturation and organization of the fibrous tissue). By inhibiting one or more of the components of fibrosis (or scarring), the overgrowth of granulation tissue may be inhibited or reduced. Examples of fibrosis-inhibiting agents for use in the present invention include the following: ZD-6474, AP-23573, synthadotin, S-0885, aplidine, ixabepilone, IDN-5390, SB-2723005, ABT- 518, combretastatin, anecortave acetate, SB-715992, temsirolimus, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, as well as analogues and derivatives of the aforementioned.
Additional examples of anti-scarring agents which can be used include those having a high potency in the assays described herein (1- 10OnM IC50 range) such as isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, dolastatin 15, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17- DMAG, and tacrolimus; those having a mid-potency in the assays described herein (100-500 nM IC50 range) such as loteprednol etabonate, juglone, prednisolone, puromycin, 3-BAABE, cladribine, and mannose-6-phosphate; and those having a low potency in the assays described herein (500- 1000nm range IC50 range) such as 5-azacytidine, Ly333531(ruboxistaurin), and simvastatin.
The drug dose administered from the present compositions for prevention or inhibition of fibrosis in accordance with the present invention will depend on a variety of factors, including the type of formulation, the location of the treatment site, and the type of condition being treated. As glaucoma drainage devices are made in a variety of configurations and sizes, the exact dose administered will also vary with device size, surface area and design. However, certain principles can be applied in the application of this art. Drug dose can be calculated as a function of dose per unit area (of the treatment site), total drug dose administered can be measured and appropriate surface concentrations of active drug can be determined. Drugs are to be used at concentrations that range from several times more than to 50%, 20%, 10%, 5%, or even less than 1% of the concentration typically used in a single chemotherapeutic systemic dose application. In certain aspects, the anti-scarring agent is released from the composition in effective concentrations in a time period that may be measured from the time of infiltration into tissue adjacent to the device, which ranges from about less than 1 day to about 180 days. Generally, the release time may also be from about less than 1 day to about 180 days; from about 7 days to about 14 days; from about 14 days to about 28 days; from about 28 days to about 56 days; from about 56 days to about 90 days; from about 90 days to about 180 days.
The exemplary anti-fibrosing agents, used alone or in combination, should be administered under the following dosing guidelines. The total amount (dose) of anti-scarring agent in the composition can be in the range of about 0.01 μg-10 μg, or about 10 μg-10 mg, or about 10 mg- 250 mg, or about 250 mg-1000 mg, or about 1000 mg-2500 mg. The dose (amount) of anti-scarring agent per unit area of device or tissue surface to which the agent is applied may be in the range of about 0.01 μg/mm2 - 1 μg/mm2, or about 1 μg/mm2 - 10 μg/mm2, or about 10 μg/mm2 - 250 μg/mm2, or about 250 μg/mm2 - 1000 μg/mm2, or about 1000 μg/mm2 - 2500 μg/mm2.
According to another aspect, any anti-infective agent described above may be used in the practice of the present invention. Exemplary anti-infective agents include (A) anthracyclines (e.g., doxorubicin and mitoxantrone), (B) fluoropyrimidines (e.g., 5-FU), (C) folic acid antagonists (e.g., methotrexate), (D) podophylotoxins (e.g., etoposide), (E) camptothecins, (F) hydroxyureas, and (G) platinum complexes (e.g., cisplatin), as well as analogues and derivatives of the aforementioned.
The drug dose administered from the present compositions for prevention or inhibition of infection in accordance with the present invention will depend on a variety of factors, including the type of formulation, the location of the treatment site, and the type of condition being treated. However, certain principles can be applied in the application of this art. Drug dose can be calculated as a function of dose per unit area (of the treatment site), total drug dose administered can be measured and appropriate surface concentrations of active drug can be determined. Drugs are to be used at concentrations that range from several times more than to 50%, 20%, 10%, 5%, or even less than 1% of the concentration typically used in a single anti-infective systemic dose application. In certain aspects, the anti- infective agent is released from the composition in effective concentrations in a time period that may be measured from the time of infiltration into tissue adjacent to the device, which ranges from about less than 1 day to about 180 days. Generally, the release time may also be from about less than 1 day to about 180 days; from about 7 days to about 14 days; from about 14 days to about 28 days; from about 28 days to about 56 days; from about 56 days to about 90 days; from about 90 days to about 180 days.
The exemplary anti-infective agents, used alone or in combination, should be administered under the following dosing guidelines. The total amount (dose) of anti-infective agent in the composition can be in the range of about 0.01 μg-1 μg, or about 1 μg-10 μg, or about 10 μg-1 mg, or about 1 mg to 10 mg, or about 10 mg-100 mg, or about 100 mg to 250 mg, or about 250 mg-1000 mg. The dose (amount) of anti-infective agent per unit area of device or tissue surface to which the agent is applied may be in the range of about 0.01 μg/mm2 - 1 μg/mm2, or about 1 μg/mm2 - 10 μg/mm2, or about 10 μg/mm2 - 100 μg/mm2, or about 100 μg/mm2 to 250 μg/mm2, or about 250 μg/mm2 - 1000 μg/mm2. As different compositions will release the anti-infective agent at differing rates, the above dosing parameters should be utilized in combination with the release rate of the drug from the composition such that a minimum concentration of about 10"8 to 10'7, or about 1O-7 to 106 about 10"6 to 10"5or about 10"5 to 104 of the agent is maintained on the tissue surface.
It should be readily evident based upon the discussions provided herein that combinations of anthracyclines (e.g., doxorubicin or mitoxantrone), fluoropyrimidines (e.g., 5-fluorouracil), folic acid antagonists (e.g., methotrexate), quinolones, and/or podophylotoxins (e.g., etoposide) may be utilized to enhance the antibacterial activity of the composition.
Prosthetic Heart Valves
In one aspect, the subject compositions may be infiltrated into tissue adjacent to a prosthetic heart valve. The subject compositions may contain a therapeutic agent (e.g., an anti-scarring and/or anti-infective agent).
Representative examples of prosthetic heart valves that may benefit from having the subject compositions infiltrated into adjacent tissue are provided above in conjunction with the coating of medical devices. Numerous agents or compositions for use with prosthetic heart valves have been described above which may be infiltrated into the tissue adjacent to the device (preferably near the device-tissue interface).
Therapeutic agents or pharmaceutical compositionsmay be infiltrated around implanted prosthetic heart valves by applying the composition directly and/or indirectly into and/or onto (a) tissue adjacent to the prosthetic heart valve; (b) the vicinity of the prosthetic heart valve-tissue interface; (c) the region around the prosthetic heart valve; and (d) tissue surrounding the prosthetic heart valve. Methods for infiltrating the subject compositions into tissue adjacent to a prosthetic heart valve include delivering the composition: (a) to the prosthetic heart valve surface (e.g., as an injectable, paste, gel or mesh) during the implantation procedure; (b) to the surface of the tissue (e.g., as an injectable, paste, gel, in situ forming gel or mesh) immediately prior to, or during, implantation of the prosthetic heart valve; (c) to the surface of the prosthetic heart valve and/or the tissue surrounding the implanted prosthetic heart valve (e.g., as an injectable, paste, gel, in situ forming gel or mesh) immediately after the implantation of the prosthetic heart valve; (d) by topical application of the composition into the anatomical space where the prosthetic heart valve may be placed (particularly useful for this embodiment is the use of polymeric carriers which release the therapeutic agent over a period ranging from several hours to several weeks - fluids, suspensions, emulsions, mic'roemulsions, microspheres, pastes, gels, microparticulates, sprays, aerosols, solid implants and other formulations which release the agent may be delivered into the region where the device may be inserted); (e) via percutaneous injection into the tissue surrounding the prosthetic heart valve as a solution as an infusate or as a sustained release preparation; (f) by any combination of the aforementioned methods. Combination therapies (i.e., combinations of therapeutic agents and combinations with antithrombotic and/or antiplatelet agents) may also be used. In all cases it is understood that the subject compositions may be infiltrated into tissue adjacent to all or a portion of the device.
In some aspects, the subject compositions may be infiltrated into tissue adjacent to: (a) the surface of the annular ring (particularly mechanical valves); (b) the surface of the valve leaflets (particularly bioprosthetic valves); and/or (c) any combination of the aforementioned.
According to one aspect, any fibrosis-inhibiting and/or anti- infective agent described above may be utilized in the practice of the present invention. In one aspect of the invention, the subject compositions infiltrated into tissue adjacent to prosthetic heart valves may be adapted to release an agent that inhibits one or more of the four general components of the process of fibrosis (or scarring), including: formation of new blood vessels (angiogenesis), migration and proliferation of connective tissue cells (such as fibroblasts or smooth muscle cells), deposition of extracellular matrix (ECM), and remodeling (maturation and organization of the fibrous tissue). By inhibiting one or more of the components of fibrosis (or scarring), the overgrowth of granulation tissue may be inhibited or reduced.
Examples of fibrosis-inhibiting agents for use in the present invention include the following: ZD-6474, AP-23573, synthadotin, S-0885, aplidine, ixabepilone, IDN-5390, SB-2723005, ABT-518, combretastatin, anecortave acetate, SB-715992, temsirolimus, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, as well as analogues and derivatives of the aforementioned.
Additional examples of anti-scarring agents which can be used include those having a high potency in the assays described herein (1- 10OnM IC50 range) such as isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, dolastatin 15, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17- DMAG, and tacrolimus; those having a mid-potency in the assays described herein (100-500 nM IC50 range) such as loteprednol etabonate, juglone, prednisolone, puromycin, 3-BAABE, cladribine, and mannose-6-phosphate; and those having a low potency in the assays described herein (500- IOOOnm range IC50 range) such as 5-azacytidine, Ly333531(ruboxistaurin), and simvastatin.
The drug dose administered from the present compositions for prevention or inhibition of fibrosis in accordance with the present invention will depend on a variety of factors, including the type of formulation, the location of the treatment site, and the type of condition being treated. As prosthetic heart valves are made in a variety of configurations and sizes, the exact dose administered will also vary with device size, surface area and design. However, certain principles can be applied in the application of this art. Drug dose can be calculated as a function of dose per unit area (of the treatment site), total drug dose administered can be measured and appropriate surface concentrations of active drug can be determined. Drugs are to be used at concentrations that range from several times more than to 50%, 20%, 10%, 5%, or even less than 1% of the concentration typically used in a single chemotherapeutic systemic dose application. In certain aspects, the anti-scarring agent is released from the composition in effective concentrations in a time period that may be measured from the time of infiltration into tissue adjacent to the device, which ranges from about less than 1 day to about 180 days. Generally, the release time may also be from about less than 1 day to about 180 days; from about 7 days to about 14 days; from about 14 days to about 28 days; from about 28 days to about 56 days; from about 56 days to about 90 days; from about 90 days to about 180 days.
The exemplary anti-fibrosing agents, used alone or in combination, should be administered under the following dosing guidelines. The total amount (dose) of anti-scarring agent in the composition can be in the range of about 0.01 μg-10 μg, or about 10 μg-10 mg, or about 10 mg- 250 mg, or about 250 mg-1000 mg, or about 1000 mg-2500 mg. The dose (amount) of anti-scarring agent per unit area of device or tissue surface to which the agent is applied may be in the range of about 0.01 μg/mm2 - 1 μg/mm2, or about 1 μg/mm2 - 10 μg/mm2, or about 10 μg/mm2 - 250 μg/mm2, or about 250 μg/mm2 - 1000 μg/mm2, or about 1000 μg/mm2 - 2500 μg/mm2.
According to another aspect, any anti-infective agent described above may be used in the practice of the present invention. Exemplary anti-infective agents include (A) anthracyclines (e.g., doxorubicin and mitoxantrone), (B) fluoropyrimidines (e.g., 5-FU), (C) folic acid antagonists (e.g., methotrexate), (D) podophylotoxins (e.g., etoposide), (E) camptothecins, (F) hydroxyureas, and (G) platinum complexes (e.g., cisplatin), as well as analogues and derivatives of the aforementioned.
The drug dose administered from the present compositions for prevention or inhibition of infection in accordance with the present invention will depend on a variety of factors, including the type of formulation, the location of the treatment site, and the type of condition being treated. However, certain principles can be applied in the application of this art. Drug dose can be calculated as a function of dose per unit area (of the treatment site), total drug dose administered can be measured and appropriate surface concentrations of active drug can be determined. Drugs are to be used at concentrations that range from several times more than to 50%, 20%, 10%, 5%, or even less than 1% of the concentration typically used in a single anti-infective systemic dose application. In certain aspects, the anti- infective agent is released from the composition in effective concentrations in a time period that may be measured from the time of infiltration into tissue adjacent to the device, which ranges from about less than 1 day to about 180 days. Generally, the release time may also be from about less than 1 day to about 180 days; from about 7 days to about 14 days; from about 14 days to about 28 days; from about 28 days to about 56 days; from about 56 days to about 90 days; from about 90 days to about 180 days.
The exemplary anti-infective agents, used alone or in combination, should be administered under the following dosing guidelines. The total amount (dose) of anti-infective agent in the composition can be in the range of about 0.01 μg-1 μg, or about 1 μg-10 μg, or about 10 μg-1 mg, or about 1 mg to 10 mg, or about 10 mg-100 mg, or about 100 mg to 250 mg, or about 250 mg-1000 mg. The dose (amount) of anti-infective agent per unit area of device or tissue surface to which the agent is applied may be in the range of about 0.01 μg/mm2 - 1 μg/mm2, or about 1 μg/mm2 - 10 μg/mm2, or about 10 μg/mm2 - 100 μg/mm2, or about 100 μg/mm2 to 250 μg/mm2, or about 250 μg/mm2 - 1000 μg/mm2. As different compositions will release the anti-infective agent at differing rates, the above dosing parameters should be utilized in combination with the release rate of the drug from the composition such that a minimum concentration of about 10"8 to 10"7, or about 10"7 to 10"6 about 10"6 to 10"5or about 10"5 to 10"4 of the agent is maintained on the tissue surface.
It should be readily evident based upon the discussions provided herein that combinations of anthracyclines (e.g., doxorubicin or mitoxantrone), fluoropyrimidines (e.g., 5-fluorouracil), folic acid antagonists (e.g., methotrexate), quinolones, and/or podophylotoxins (e.g., etoposide) may be utilized to enhance the antibacterial activity of the composition..
Penile Implants
In one aspect, the subject compositions may be infiltrated into tissue adjacent to a penile implant device. The subject compositions may contain a therapeutic agent {e.g., an anti-scarring and/or anti-infective agent). In one aspect, the subject compositions infiltrated into tissue adjacent to penile implants are loaded with an anti-scarring drug to prevent fibrous encapsulation. In another aspect, the subject compositions infiltrated into tissue adjacent to penile implants are loaded with an anti- infective agent (either alone or in conjunction with an anti-scarring drug) to prevent fibrous infection and/or encapsulation
Representative examples of penile implants that may benefit from having the subject compositions infiltrated into adjacent tissue are provided above in conjunction with the coating of medical devices. Numerous agents or compositions for use with penile implants have been described above which may be infiltrated into the tissue adjacent to the device (preferably near the device-tissue interface).
Therapeutic agents or pharmaceutical compositionsmay be infiltrated around implanted penile implants by applying the composition directly and/or indirectly into and/or onto (a) tissue adjacent to the penile implant; (b) the vicinity of the penile implant-tissue interface; (c) the region around the penile implant; and (d) tissue surrounding the penile implant. Methods for infiltrating the subject compositions into tissue adjacent to a penile implant include delivering the composition: (a) to the penile implant surface {e.g., as an injectable, paste, gel or mesh) during the implantation procedure; (b) to the surface of the tissue (e.g., as an injectable, paste, gel, in situ forming gel or mesh) immediately prior to, or during, implantation of the penile implant; (c) to the surface of the penile implant and/or the tissue surrounding the implanted penile implant (e.g., as an injectable, paste, gel, in situ forming gel or mesh) immediately after the implantation of the penile implant; (d) by topical application of the composition into the anatomical space where the penile implant may be placed (particularly useful for this embodiment is the use of polymeric carriers which release the therapeutic agent over a period ranging from several hours to several weeks - fluids, suspensions, emulsions, microemulsions, microspheres, pastes, gels, microparticulates, sprays, aerosols, solid implants and other formulations which release the agent may be delivered into the region where the device may be inserted); (e) via percutaneous injection into the tissue surrounding the penile implant as a solution as an infusate or as a sustained release preparation; (f) by any combination of the aforementioned methods. Combination therapies (i.e., combinations of therapeutic agents and combinations with antithrombotic and/or antiplatelet agents) may also be used. In all cases it is understood that the subject compositions may be infiltrated into tissue adjacent to all or a portion of the device. The placement of penile implants can be complicated by infection (usually in the first 6 months after surgery) with Coagulase Negative Staphylococci (including Staphylococcus epidermidis), Staphylococcus aureus, Pseudomonas aeruginosa, Enterococci, Serratia and Candida. Infection is characterized by fever, erythema, induration and purulent drainage from the operative site. The usual route of infection is through the incision at the time of surgery and up to 3% of penile implants become infected despite the best sterile surgical technique. To help combat this, intraoperative irrigation with antibiotic solutions is often employed.
Infiltrating into the tissue adjacent to the penile implant a composition containing an anti-infective agent can allow bacteriocidal drug levels to be achieved locally, thus reducing the incidence of bacterial colonization (and subsequent development of local infection and device failure), while producing negligible systemic exposure to the drugs.
According to the one aspect, any fibrosis-inhibiting and/or anti- infective agent described above may be utilized in the practice of the present invention. In one aspect of the invention, the subject compositions infiltrated into tissue adjacent to penile implants may be adapted to release an agent that inhibits one or more of the four general components of the process of fibrosis (or scarring), including: formation of new blood vessels (angiogenesis), migration and proliferation of connective tissue cells (such as fibroblasts or smooth muscle cells), deposition of extracellular matrix (ECM), and remodeling (maturation and organization of the fibrous tissue). By inhibiting one or more of the components of fibrosis (or scarring), the overgrowth of granulation tissue may be inhibited or reduced.
Examples of fibrosis-inhibiting agents for use in the present invention include the following: ZD-6474, AP-23573, synthadotin, S-0885, aplidine, ixabepilone, IDN-5390, SB-2723005, ABT-518, combretastatin, anecortave acetate, SB-715992, temsirolimus, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, as well as analogues and derivatives of the aforementioned. Additional examples of anti-scarring agents which can be used include those having a high potency in the assays described herein (1- 10OnM IC50 range) such as isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, dolastatin 15, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17- DMAG, and tacrolimus; those having a mid-potency in the assays described herein (100-500 nM IC50 range) such as loteprednol etabonate, juglone, prednisolone, puromycin, 3-BAABE, cladribine, and mannose-6-phosphate; and those having a low potency in the assays described herein (500- 1000nm range IC5O range) such as 5-azacytidine, Ly333531(ruboxistaurin), and simvastatin.
The drug dose administered from the present compositions for prevention or inhibition of fibrosis in accordance with the present invention will depend on a variety of factors, including the type of formulation, the location of the treatment site, and the type of condition being treated. As penile implants are made in a variety of configurations and sizes, the exact dose administered will also vary with device size, surface area and design. However, certain principles can be applied in the application of this art. Drug dose can be calculated as a function of dose per unit area (of the treatment site), total drug dose administered can be measured and appropriate surface concentrations of active drug can be determined. Drugs are to be used at concentrations that range from several times more than to 50%, 20%, 10%, 5%, or even less than 1% of the concentration typically used in a single chemotherapeutic systemic dose application. In certain aspects, the anti-scarring agent is released from the composition in effective concentrations in a time period that may be measured from the time of infiltration into tissue adjacent to the device, which ranges from about less than 1 day to about 180 days. Generally, the release time may also be from about less than 1 day to about 180 days; from about 7 days to about 14 days; from about 14 days to about 28 days; from about 28 days to about 56 days; from about 56 days to about 90 days; from about 90 days to about 180 days.
The exemplary anti-fibrosing agents, used alone or in combination, should be administered under the following dosing guidelines. The total amount (dose) of anti-scarring agent in the composition can be in the range of about 0.01 μg-10 μg, or about 10 μg-10 mg, or about 10 mg- 250 mg, or about 250 mg-1000 mg, or about 1000 mg-2500 mg. The dose (amount) of anti-scarring agent per unit area of device or tissue surface to which the agent is applied may be in the range of about 0.01 μg/mm2 - 1 μg/mm2, or about 1 μg/mm2 - 10 μg/mm2, or about 10 μg/mm2 - 250 μg/mm2, or about 250 μg/mm2 - 1000 μg/mm2, or about 1000 μg/mm2 - 2500 μg/mm2.
According to another aspect, any anti-infective agent described above may be used in the practice of the present invention. Exemplary anti-infective agents include (A) anthracyclines (e.g., doxorubicin and mitoxantrone), (B) fluoropyrimidines (e.g., 5-FU), (C) folic acid antagonists (e.g., methotrexate), (D) podophylotoxins (e.g., etoposide), (E) camptothecins, (F) hydroxyureas, and (G) platinum complexes (e.g., cisplatin), as well as analogues and derivatives of the aforementioned.
The drug dose administered from the present compositions for prevention or inhibition of infection in accordance with the present invention will depend on a variety of factors, including the type of formulation, the location of the treatment site, and the type of condition being treated. However, certain principles can be applied in the application of this art. Drug dose can be calculated as a function of dose per unit area (of the treatment site), total drug dose administered can be measured and appropriate surface concentrations of active drug can be determined. Drugs are to be used at concentrations that range from several times more than to 50%, 20%, 10%, 5%, or even less than 1% of the concentration typically used in a single anti-infective systemic dose application. In certain aspects, the anti-infective agent is released from the composition in effective concentrations in a time period that may be measured from the time of infiltration into tissue adjacent to the device, which ranges from about less than 1 day to about 180 days. Generally, the release time may also be from about less than 1 day to about 180 days; from about 7 days to about 14 days; from about 14 days to about 28 days; from about 28 days to about 56 days; from about 56 days to about 90 days; from about 90 days to about 180 days.
The exemplary anti-infective agents, used alone or in combination, should be administered under the following dosing guidelines. The total amount (dose) of anti-infective agent in the composition can be in the range of about 0.01 μg-1 μg, or about 1 μg-10 μg, or about 10 μg-1 mg, or about 1 mg to 10 mg, or about 10 mg-100 mg, or about 100 mg to 250 mg, or about 250 mg-1000 mg. The dose (amount) of anti-infective agent per unit area of device or tissue surface to which the agent is applied may be in the range of about 0.01 μg/mm2 - 1 μg/mm2, or about 1 μg/mm2 - 10 μg/mm2, or about 10 μg/mm2 - 100 μg/mm2, or about 100 μg/mm2 to 250 μg/mm2, or about 250 μg/mm2 - 1000 μg/mm2. As different compositions will release the anti-infective agent at differing rates, the above dosing parameters should be utilized in combination with the release rate of the drug from the composition such that a minimum concentration of about 10~8 to 10"7, or about 10"7 to 10"6 about 10"6 to 10'5 or about 10"5 to 10"4 of the agent is maintained on the tissue surface.
It should be readily evident based upon the discussions provided herein that combinations of anthracyclines (e.g., doxorubicin or mitoxantrone), fluoropyrimidines (e.g., 5-fluorouracil), folic acid antagonists (e.g., methotrexate), quinolones, and/or podophylotoxins (e.g., etoposide) may be utilized to enhance the antibacterial activity of the composition.
Endotracheal and Tracheostomy Tubes In one aspect, the subject compositions may be infiltrated into tissue adjacent to endotracheal and tracheostomy tube devices. The subject compositions may contain a therapeutic agent (e.g., an anti-scarring and/or anti-infective agent). Association of an anti-scarring agent with an endotracheal or a tracheostomy tube (e.g., chest tube), or adjacent tissue, may be used to prevent stenosis and/or infection of the artificial airway.
Representative examples of endotracheal and tracheostomy tubes that may benefit from having the subject compositions infiltrated into adjacent tissue are provided above in conjunction with the coating of medical devices. Numerous agents or compositions for use with endotracheal and tracheostomy tubes have been described above which may be infiltrated into the tissue adjacent to the device (preferably near the device-tissue interface).
Therapeutic agents or pharmaceutical compositionsmay be infiltrated around implanted endotracheal and tracheostomy tube devices by applying the composition directly and/or indirectly into and/or onto (a) tissue adjacent to the endotracheal or tracheostomy tube device; (b) the vicinity of the endotracheal or tracheostomy tube device-tissue interface; (c) the region around the endotracheal or tracheostomy tube device; and (d) tissue surrounding the endotracheal or tracheostomy tube device. Methods for infiltrating the subject compositions into tissue adjacent to endotracheal or tracheostomy tube devices include delivering the composition: (a) to the endotracheal or tracheostomy tube device surface (e.g., as an injectable, paste, gel or mesh) during the implantation procedure; (b) to the surface of the tissue (e.g., as an injectable, paste, gel, in situ forming gel or mesh) immediately prior to, or during, implantation of the endotracheal or tracheostomy tube device; (c) to the surface of the endotracheal or tracheostomy tube device and/or the tissue surrounding the implanted endotracheal or tracheostomy tube device (e.g., as an injectable, paste, gel, in situ forming gel or mesh) immediately after the implantation of the endotracheal or tracheostomy tube device; (d) by topical application of the composition into the anatomical space where the endotracheal or tracheostomy tube device may be placed (particularly useful for this embodiment is the use of polymeric carriers which release the therapeutic agent over a period ranging from several hours to several weeks - fluids, suspensions, emulsions, microemulsions, microspheres, pastes, gels, microparticulates, sprays, aerosols, solid implants and other formulations which release the agent may be delivered into the region where the device may be inserted); (e) via percutaneous injection into the tissue surrounding the endotracheal or tracheostomy tube device as a solution as an infusate or as a sustained release preparation; (f) by any combination of the aforementioned methods. Combination therapies (i.e., combinations of therapeutic agents and combinations with antithrombotic and/or antiplatelet agents) may also be used. In all cases it is understood that the subject compositions may be infiltrated into tissue adjacent to all or a portion of the device.
According to one aspect, any fibrosis-inhibiting and/or anti- infective agent described above may be utilized in the practice of the present invention. In one aspect of the invention, the subject compositions infiltrated into tissue adjacent to endotracheal and tracheostomy tube devices may be adapted to release an agent that inhibits one or more of the four general components of the process of fibrosis (or scarring), including: formation of new blood vessels (angiogenesis), migration and proliferation of connective tissue cells (such as fibroblasts or smooth muscle cells), deposition of extracellular matrix (ECM), and remodeling (maturation and organization of the fibrous tissue). By inhibiting one or more of the components of fibrosis (or scarring), the overgrowth of granulation tissue may be inhibited or reduced.
Examples of fibrosis-inhibiting agents for use in the present invention include the following: ZD-6474, AP-23573, synthadotin, S-0885, aplidine, ixabepilone, IDN-5390, SB-2723005, ABT-518, combretastatin, anecortave acetate, SB-715992, temsirolimus, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, as well as analogues and derivatives of the aforementioned. Additional examples of anti-scarring agents which can be used include those having a high potency in the assays described herein (1- 10OnM IC50 range) such as isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, dolastatin 15, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17- DMAG, and tacrolimus; those having a mid-potency in the assays described herein (100-500 nM IC50 range) such as loteprednol etabonate, juglone, prednisolone, puromycin, 3-BAABE, cladribine, and mannose-6-phosphate; and those having a low potency in the assays described herein (500- 1000nm range IC50 range) such as 5-azacytidine, Ly333531(ruboxistaurin), and simvastatin.
The drug dose administered from the present compositions for prevention or inhibition of fibrosis in accordance with the present invention will depend on a variety of factors, including the type of formulation, the location of the treatment site, and the type of condition being treated. As endotracheal and tracheostomy tube devices are made in a variety of configurations and sizes, the exact dose administered will also vary with device size, surface area and design. However, certain principles can be applied in the application of this art. Drug dose can be calculated as a function of dose per unit area (of the treatment site), total drug dose administered can be measured and appropriate surface concentrations of active drug can be determined. Drugs are to be used at concentrations that range from several times more than to 50%, 20%, 10%, 5%, or even less than 1% of the concentration typically used in a single chemotherapeutic systemic dose application. In certain aspects, the anti-scarring agent is released from the composition in effective concentrations in a time period that may be measured from the time of infiltration into tissue adjacent to the device, which ranges from about less than 1 day to about 180 days. Generally, the release time may also be from about less than 1 day to about 180 days; from about 7 days to about 14 days; from about 14 days to about 28 days; from about 28 days to about 56 days; from about 56 days to about 90 days; from about 90 days to about 180 days.
The exemplary anti-fibrosing agents, used alone or in combination, should be administered under the following dosing guidelines. The total amount (dose) of anti-scarring agent in the composition can be in the range of about 0.01 μg-10 μg, or about 10 μg-10 mg, or about 10 mg- 250 mg, or about 250 mg-1000 mg, or about 1000 mg-2500 mg. The dose (amount) of anti-scarring agent per unit area of device or tissue surface to which the agent is applied may be in the range of about 0.01 μg/mm2 - 1 μg/mm2, or about 1 μg/mm2 - 10 μg/mm2, or about 10 μg/mm2 - 250 μg/mm2, or about 250 μg/mm2 - 1000 μg/mm2, or about 1000 μg/mm2 - 2500 μg/mm2.
According to another aspect, any anti-infective agent described above may be used in the practice of the present invention. Exemplary anti-infective agents include (A) anthracyclines (e.g., doxorubicin and mitoxantrone), (B) fluoropyrimidines (e.g., 5-FU), (C) folic acid antagonists (e.g., methotrexate), (D) podophylotoxins (e.g., etoposide), (E) camptothecins, (F) hydroxyureas, and (G) platinum complexes (e.g., cisplatin), as well as analogues and derivatives of the aforementioned.
The drug dose administered from the present compositions for prevention or inhibition of infection in accordance with the present invention will depend on a variety of factors, including the type of formulation, the location of the treatment site, and the type of condition being treated. However, certain principles can be applied in the application of this art. Drug dose can be calculated as a function of dose per unit area (of the treatment site), total drug dose administered can be measured and appropriate surface concentrations of active drug can be determined. ' Drugs are to be used at concentrations that range from several times more than to 50%, 20%, 10%, 5%, or even less than 1% of the concentration typically used in a single anti-infective systemic dose application. In certain aspects, the anti- infective agent is released from the composition in effective concentrations in a time period that may be measured from the time of infiltration into tissue adjacent to the device, which ranges from about less than 1 day to about 180 days. Generally, the release time may also be from about less than 1 day to about 180 days; from about 7 days to about 14 days; from about 14 days to about 28 days; from about 28 days to about 56 days; from about 56 days to about 90 days; from about 90 days to about 180 days.
The exemplary anti-infective agents, used alone or in combination, should be administered under the following dosing guidelines. The total amount (dose) of anti-infective agent in the composition can be in the range of about 0.01 μg-1 μg, or about 1 μg-10 μg, or about 10 μg-1 mg, or about 1 mg to 10 mg, or about 10 mg-100 mg, or about 100 mg to 250 mg, or about 250 mg-1000 mg. The dose (amount) of anti-infective agent per unit area of device or tissue surface to which the agent is applied may be in the range of about 0.01 μg/mm2 - 1 μg/mm2, or about 1 μg/mm2 - 10 μg/mm2, or about 10 μg/mm2 - 100 μg/mm2, or about 100 μg/mm2 to 250 μg/mm2, or about 250 μg/mm2 - 1000 μg/mm2. As different compositions will release the anti-infective agent at differing rates, the above dosing parameters should be utilized in combination with the release rate of the drug from the composition such that a minimum concentration of about 10"8 to 10"7, or about 10'7 to 10"6 about 106 to 10'5Or about 10"5 to 104 of the agent is maintained on the tissue surface.
It should be readily evident based upon the discussions provided herein that combinations of anthracyclines (e.g., doxorubicin or mitoxantrone), fluoropyrimidines (e.g., 5-fluorouracil), folic acid antagonists (e.g., methotrexate), quinolones, and/or podophylotoxins (e.g., etoposide) may be utilized to enhance the antibacterial activity of the composition.
Peritoneal Dialysis Catheters
In one aspect, the subject compositions may be infiltrated into tissue adjacent to a peritoneal dialysis catheter or a peritoneal implant for drug delivery. The subject compositions may contain a therapeutic agent (e.g., an anti-scarring and/or anti-infective agent).
Representative examples of peritoneal dialysis catheters that may benefit from having the subject compositions infiltrated into adjacent tissue are provided above in conjunction with the coating of medical devices. Numerous agents or compositions for use with peritoneal dialysis catheters have been described above which may be infiltrated into the tissue adjacent to the device (preferably near the device-tissue interface).
Therapeutic agents or pharmaceutical compositionsmay be infiltrated around implanted peritoneal access catheters and implants by applying the composition directly and/or indirectly into and/or onto (a) tissue adjacent to the peritoneal access catheter or implant; (b) the vicinity of the peritoneal access catheter or implant-tissue interface; (c) the region around the peritoneal access catheter or implant; and (d) tissue surrounding the peritoneal access catheter or implant. Methods for infiltrating the subject compositions into tissue adjacent to a peritoneal access catheter or implant include delivering the composition: (a) to the peritoneal access catheter or implant surface (e.g., as an injectable, paste, gel or mesh) during the implantation procedure; (b) to the surface of the tissue (e.g., as an injectable, paste, gel, in situ forming gel or mesh) immediately prior to, or during, implantation of the peritoneal access catheter or implant; (c) to the surface of the peritoneal access catheter or implant and/or the tissue surrounding the implanted peritoneal access catheter or implant (e.g., as an injectable, paste, gel, in situ forming gel or mesh) immediately after the implantation of the peritoneal access catheter or implant; (d) by topical application of the composition into the anatomical space where the peritoneal access catheter or implant may be placed (particularly useful for this embodiment is the use of polymeric carriers which release the therapeutic agent over a period ranging from several hours to several weeks - fluids, suspensions, emulsions, microemulsions, microspheres, pastes, gels, microparticulates, sprays, aerosols, solid implants and other formulations which release the agent may be delivered into the region where the device may be inserted); (e) via percutaneous injection into the tissue surrounding the peritoneal access catheter or implant as a solution as an infusate or as a sustained release preparation; (f) by any combination of the aforementioned methods. Combination therapies (i.e., combinations of therapeutic agents and combinations with antithrombotic and/or antiplatelet agents) may also be used. In all cases it is understood that the subject compositions may be infiltrated into tissue adjacent to all or a portion of the device.
According to one aspect, any fibrosis-inhibiting and/or anti- infective agent described above may be utilized in the practice of the present invention. In one aspect of the invention, the subject compositions infiltrated into tissue adjacent to peritoneal dialysis implants and catheters may be adapted to release an agent that inhibits one or more of the four general components of the process of fibrosis (or scarring), including: formation of new blood vessels (angiogenesis), migration and proliferation of connective tissue cells (such as fibroblasts or smooth muscle cells), deposition of extracellular matrix (ECM), and remodeling (maturation and organization of the fibrous tissue). By inhibiting one or more of the components of fibrosis (or scarring), the overgrowth of granulation tissue may be inhibited or reduced.
Examples of fibrosis-inhibiting agents for use in the present invention include the following: ZD-6474, AP-23573, synthadotin, S-0885, aplidine, ixabepilone, IDN-5390, SB-2723005, ABT-518, combretastatin, anecortave acetate, SB-715992, temsirolimus, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, as well as analogues and derivatives of the aforementioned.
Additional examples of anti-scarring agents which can be used include those having a high potency in the assays described herein (1- 10OnM IC50 range) such as isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, dolastatin 15, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17- DMAG, and tacrolimus; those having a mid-potency in the assays described herein (100-500 nM IC50 range) such as loteprednol etabonate, juglone, prednisolone, puromycin, 3-BAABE, cladribine, and mannose-6-phosphate; and those having a low potency in the assays described herein (500- 1000nm range IC50 range) such as 5-azacytidine, Ly333531(ruboxistaurin), and simvastatin.
The drug dose administered from the present compositions for prevention or inhibition of fibrosis in accordance with the present invention will depend on a variety of factors, including the type of formulation, the location of the treatment site, and the type of condition being treated. As peritoneal dialysis implants and catheters are made in a variety of configurations and sizes, the exact dose administered will also vary with device size, surface area and design. However, certain principles can be applied in the application of this art. Drug dose can be calculated as a function of dose per unit area (of the treatment site), total drug dose administered can be measured and appropriate surface concentrations of active drug can be determined. Drugs are to be used at concentrations that range from several times more than to 50%, 20%, 10%, 5%, or even less than 1% of the concentration typically used in a single chemotherapeutic systemic dose application. In certain aspects, the anti-scarring agent is released from the composition in effective concentrations in a time period that may be measured from the time of infiltration into tissue adjacent to the device, which ranges from about less than 1 day to about 180 days. Generally, the release time may also be from about less than 1 day to about 180 days; from about 7 days to about 14 days; from about 14 days to about 28 days; from about 28 days to about 56 days; from about 56 days to about 90 days; from about 90 days to about 180 days.
The exemplary anti-fibrosing agents, used alone or in combination, should be administered under the following dosing guidelines. The total amount (dose) of anti-scarring agent in the composition can be in the range of about 0.01 μg-10 μg, or about 10 μg-10 mg, or about 10 mg- 250 mg, or about 250 mg-1000 mg, or about 1000 mg-2500 mg. The dose (amount) of anti-scarring agent per unit area of device or tissue surface to which the agent is applied may be in the range of about 0.01 μg/mm2 - 1 μg/mm2, or about 1 μg/mm2 - 10 μg/mm2, or about 10 μg/mm2 - 250 μg/mm2, or about 250 μg/mm2 - 1000 μg/mm2, or about 1000 μg/mm2 - 2500 μg/mm2.
According to another aspect, any anti-infective agent described above may be used in the practice of the present invention. Exemplary anti-infective agents include (A) anthracyclines (e.g., doxorubicin and mitoxantrone), (B) fluoropyrimidines (e.g., 5-FU), (C) folic acid antagonists (e.g., methotrexate), (D) podophylotoxins (e.g., etoposide), (E) camptothecins, (F) hydroxyureas, and (G) platinum complexes (e.g., cisplatin), as well as analogues and derivatives of the aforementioned.
The drug dose administered from the present compositions for prevention or inhibition of infection in accordance with the present invention will depend on a variety of factors, including the type of formulation, the location of the treatment site, and the type of condition being treated. However, certain principles can be applied in the application of this art. Drug dose can be calculated as a function of dose per unit area (of the treatment site), total drug dose administered can be measured and appropriate surface concentrations of active drug can be determined. Drugs are to be used at concentrations that range from several times more than to 50%, 20%, 10%, 5%, or even less than 1% of the concentration typically used in a single anti-infective systemic dose application. In certain aspects, the anti- infective agent is released from the composition in effective concentrations in a time period that may be measured from the time of infiltration into tissue adjacent to the device, which ranges from about less than 1 day to about 180 days. Generally, the release time may also be from about less than 1 day to about 180 days; from about 7 days to about 14 days; from about 14 days to about 28 days; from about 28 days to about 56 days; from about 56 days to about 90 days; from about 90 days to about 180 days.
The exemplary anti-infective agents, used alone or in combination, should be administered under the following dosing guidelines. The total amount (dose) of anti-infective agent in the composition can be in the range of about 0.01 μg-1 μg, or about 1 μg-10 μg, or about 10 μg-1 mg, or about 1 mg to 10 mg, or about 10 mg-100 mg, or about 100 mg to 250 mg, or about 250 mg-1000 mg. The dose (amount) of anti-infective agent per unit area of device or tissue surface to which the agent is applied may be in the range of about 0.01 μg/mm2 - 1 μg/mm2, or about 1 μg/mm2 - 10 μg/mm2, or about 10 μg/mm2 - 100 μg/mm2, or about 100 μg/mm2 to 250 μg/mm2, or about 250 μg/mm2 - 1000 μg/mm2. As different compositions will release the anti-infective agent at differing rates, the above dosing parameters should be utilized in combination with the release rate of the drug from the composition such that a minimum concentration of about 10"8 to 10"7, or about 10'7 to 10"6 about 10"6 to 10~5or about 10"5 to 10"4 Of the agent is maintained on the tissue surface.
It should be readily evident based upon the discussions provided herein that combinations of anthracyclines (e.g., doxorubicin or mitoxantrone), fluoropyrimidines (e.g., 5-fluorouracil), folic acid antagonists (e.g., methotrexate), quinolones, and/or podophylotoxins (e.g., etoposide) may be utilized to enhance the antibacterial activity of the composition.
Central Nervous System Shunts and Pressure Monitoring Devices
In one aspect, the subject compositions may be infiltrated into tissue adjacent to a central nervous system (CNS) device, such as a CNS shunt or a pressure monitoring device. The subject compositions may contain a therapeutic agent (e.g., an anti-scarring and/or anti-infective agent). CNS devices having the subject composition comprising an anti- scarring agent infiltrated into adjacent tissue are capable of preventing stenosis and obstruction of the device leading to hydrocephalus and increased intercranial pressure. CNS devices having the subject composition comprising an anti-infective agent infiltrated into adjacent tissue are capable of preventing or inhibiting infection in the tissue surrounding the device.
Representative examples of CNS and pressure monitoring devices that may benefit from having the subject compositions infiltrated into adjacent tissue are provided above in conjunction with the coating of medical devices. Numerous agents or compositions for use with CNS and pressure monitoring devices have been described above which may be infiltrated into the tissue adjacent to the device (preferably near the device- tissue interface).
Therapeutic agents or pharmaceutical compositionsmay be infiltrated around implanted CNS devices by applying the composition directly and/or indirectly into and/or onto (a) tissue adjacent to the CNS device; (b) the vicinity of the CNS device-tissue interface; (c) the region around the CNS device; and (d) tissue surrounding the CNS device. Methods for infiltrating the subject compositions into tissue adjacent to a CNS device include delivering the composition: (a) to the CNS device surface (e.g., as an injectable, paste, gel or mesh) during the implantation procedure; (b) to the surface of the tissue (e.g., as an injectable, paste, gel, in situ forming gel or mesh) immediately prior to, or during, implantation of the CNS device; (c) to the surface of the CNS device and/or the tissue surrounding the implanted CNS device (e.g., as an injectable, paste, gel, in situ forming gel or mesh) immediately after the implantation of the CNS device; (d) by topical application of the composition into the anatomical space where the CNS device may be placed (particularly useful for this embodiment is the use of polymeric carriers which release the therapeutic agent over a period ranging from several hours to several weeks - fluids, suspensions, emulsions, microemulsions, microspheres, pastes, gels, microparticulates, sprays, aerosols, solid implants and other formulations which release the agent may be delivered into the region where the device may be inserted); (e) via percutaneous injection into the tissue surrounding the CNS device as a solution as an infusate or as a sustained release preparation; (f) by any combination of the aforementioned methods. Combination therapies (i.e., combinations of therapeutic agents and combinations with antithrombotic and/or antiplatelet agents) may also be used. In all cases it is understood that the subject compositions may be infiltrated into tissue adjacent to all or a portion of the device.
According to one aspect, any fibrosis-inhibiting and/or anti- infective agent described above may be utilized in the practice of the present invention. In one aspect of the invention, the subject compositions infiltrated into tissue adjacent to CNS devices may be adapted to release an agent that inhibits one or more of the four general components of the process of fibrosis (or scarring), including: formation of new blood vessels (angiogenesis), migration and proliferation of connective tissue cells (such as fibroblasts or smooth muscle cells), deposition of extracellular matrix (ECM), and remodeling (maturation and organization of the fibrous tissue). By inhibiting one or more of the components of fibrosis (or scarring), the overgrowth of granulation tissue may be inhibited or reduced.
Examples of fibrosis-inhibiting agents for use in the present invention include the following: ZD-6474, AP-23573, synthadotin, S-0885, aplidine, ixabepilone, IDN-5390, SB-2723005, ABT-518, combretastatin, anecortave acetate, SB-715992, temsirolimus, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, as well as analogues and derivatives of the aforementioned.
Additional examples of anti-scarring agents which can be used include those having a high potency in the assays described herein (1- 10OnM IC50 range) such as isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, dolastatin 15, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17- DMAG, and tacrolimus; those having a mid-potency in the assays described herein (100-500 nM IC50 range) such as loteprednol etabonate, juglone, prednisolone, puromycin, 3-BAABE, cladribine, and mannose-6-phosphate; and those having a low potency in the assays described herein (500- 1000nm range IC50 range) such as 5-azacytidine, Ly333531(ruboxistaurin), and simvastatin.
The drug dose administered from the present compositions for prevention or inhibition of fibrosis in accordance with the present invention will depend on a variety of factors, including the type of formulation, the location of the treatment site, and the type of condition being treated. As CNS devices are made in a variety of configurations and sizes, the exact dose administered will also vary with device size, surface area and design. However, certain principles can be applied in the application of this art. Drug dose can be calculated as a function of dose per unit area (of the treatment site), total drug dose administered can be measured and appropriate surface concentrations of active drug can be determined. Drugs are to be used at concentrations that range from several times more than to 50%, 20%, 10%, 5%, or even less than 1% of the concentration typically used in a single chemotherapeutic systemic dose application. In certain aspects, the anti-scarring agent is released from the composition in effective concentrations in a time period that may be measured from the time of infiltration into tissue adjacent to the device, which ranges from about less than 1 day to about 180 days. Generally, the release time may also be from about less than 1 day to about 180 days; from about 7 days to about 14 days; from about 14 days to about 28 days; from about 28 days to about 56 days; from about 56 days to about 90 days; from about 90 days to about 180 days.
The exemplary anti-fibrosing agents, used alone or in combination, should be administered under the following dosing guidelines. The total amount (dose) of anti-scarring agent in the composition can be in the range of about 0.01 μg-10 μg, or about 10 μg-10 mg, or about 10 mg- 250 mg, or about 250 mg-1000 mg, or about 1000 mg-2500 mg. The dose (amount) of anti-scarring agent per unit area of device or tissue surface to which the agent is applied may be in the range of about 0.01 μg/mm2 - 1 μg/mm2, or about 1 μg/mm2 - 10 μg/mm2, or about 10 μg/mm2 - 250 μg/mm2, or about 250 μg/mm2 - 1000 μg/mm2, or about 1000 μg/mm2 - 2500 μg/mm2.
According to another aspect, any anti-infective agent described above may be used in the practice of the present invention. Exemplary anti-infective agents include (A) anthracyclines (e.g., doxorubicin and mitoxantrone), (B) fluoropyrimidines (e.g., 5-FU), (C) folic acid antagonists (e.g., methotrexate), (D) podophylotoxins (e.g., etoposide), (E) camptothecins, (F) hydroxyureas, and (G) platinum complexes (e.g., cisplatin), as well as analogues and derivatives of the aforementioned.
The drug dose administered from the present compositions for prevention or inhibition of infection in accordance with the present invention will depend on a variety of factors, including the type of formulation, the location of the treatment site, and the type of condition being treated. However, certain principles can be applied in the application of this art. Drug dose can be calculated as a function of dose per unit area (of the treatment site), total drug dose administered can be measured and appropriate surface concentrations of active drug can be determined. Drugs are to be used at concentrations that range from several times more than to 50%, 20%, 10%, 5%, or even less than 1% of the concentration typically used in a single anti-infective systemic dose application. In certain aspects, the anti- infective agent is released from the composition in effective concentrations in a time period that may be measured from the time of infiltration into tissue adjacent to the device, which ranges from about less than 1 day to about 180 days. Generally, the release time may also be from about less than 1 day to about 180 days; from about 7 days to about 14 days; from about 14 days to about 28 days; from about 28 days to about 56 days; from about 56 days to about 90 days; from about 90 days to about 180 days. The exemplary anti-infective agents, used alone or in combination, should be administered under the following dosing guidelines. The total amount (dose) of anti-infective agent in the composition can be in the range of about 0.01 μg-1 μg, or about 1 μg-10 μg, or about 10 μg-1 mg, or about 1 mg to 10 mg, or about 10 mg-100 mg, or about 100 mg to 250 mg, or about 250 mg-1000 mg. The dose (amount) of anti-infective agent per unit area of device or tissue surface to which the agent is applied may be in the range of about 0.01 μg/mm2 - 1 μg/mm2, or about 1 μg/mm2 - 10 μg/mm2, or about 10 μg/mm2 - 100 μg/mm2, or about 100 μg/mm2 to 250 μg/mm2, or about 250 μg/mm2 - 1000 μg/mm2. As different compositions will release the anti-infective agent at differing rates, the above dosing parameters should be utilized in combination with the release rate of the drug from the composition such that a minimum concentration of about 10"8 to 10"7, or about 10~7 to 106 about 10'6 to 10"5or about 10'5 to 10"4of the agent is maintained on the tissue surface.
It should be readily evident based upon the discussions provided herein that combinations of anthracyclines (e.g., doxorubicin or mitoxantrone), fluoropyrimidines (e.g., 5-fluorouracil), folic acid antagonists (e.g., methotrexate), quinolones, and/or podophylotoxins (e.g., etoposide) may be utilized to enhance the antibacterial activity of the composition.
Inferior Vena Cava Filters
In one aspect, the subject compositions may be infiltrated into tissue adjacent to an inferior vena cava filter device. The subject compositions may contain a therapeutic agent (e.g., an anti-scarring and/or anti-infective agent).
Representative examples of inferior vena cava filters that may benefit from having the subject compositions infiltrated into adjacent tissue are provided above in conjunction with the coating of medical devices. Numerous agents or compositions for use with inferior vena cava filters have been described above which may be infiltrated into the tissue adjacent to the device (preferably near the device-tissue interface).
Therapeutic agents or pharmaceutical compositionsmay be infiltrated around implanted inferior vena cava filter devices by applying the composition directly and/or indirectly into and/or onto (a) tissue adjacent to the inferior vena cava filter device; (b) the vicinity of the inferior vena cava filter device-tissue interface; (c) the region around the inferior vena cava filter device; and (d) tissue surrounding the inferior vena cava filter device. Methods for infiltrating the subject compositions into tissue adjacent to an inferior vena cava filter device include delivering the composition: (a) to the inferior vena cava filter device surface (e.g., as an injectable, paste, gel or mesh) during the implantation procedure; (b) to the surface of the tissue (e.g., as an injectable, paste, gel, in situ forming gel or mesh) immediately prior to, or during, implantation of the inferior vena cava filter device; (c) to the surface of the inferior vena cava filter device and/or the tissue surrounding the implanted inferior vena cava filter device (e.g., as an injectable, paste, gel, in situ forming gel or mesh) immediately after the implantation of the inferior vena cava filter device; (d) by topical application of the composition into the anatomical space where the inferior vena cava filter device may be placed (particularly useful for this embodiment is the use of polymeric carriers which release the therapeutic agent over a period ranging from several hours to several weeks - fluids, suspensions, emulsions, microemulsions, microspheres, pastes, gels, microparticulates, sprays, aerosols, solid implants and other formulations which release the agent may be delivered into the region where the device may be inserted); (e) via percutaneous injection into the tissue surrounding the inferior vena cava filter device as a solution as an infusate or as a sustained release preparation; (f) by any combination of the aforementioned methods. Combination therapies (i.e., combinations of therapeutic agents and combinations with antithrombotic and/or antiplatelet agents) may also be used. In all cases it is understood that the subject compositions may be infiltrated into tissue adjacent to all or a portion of the device.
According to one aspect, any fibrosis-inhibiting and/or anti- infective agent described above may be utilized in the practice of the present invention. In one aspect of the invention, the subject compositions infiltrated into tissue adjacent to vena cava filters (e.g., inferior vena cava filters) may be adapted to release an agent that inhibits one or more of the four general components of the process of fibrosis (or scarring), including: formation of new blood vessels (angiogenesis), migration and proliferation of connective tissue cells (such as fibroblasts or smooth muscle cells), deposition of extracellular matrix (ECM), and remodeling (maturation and organization of the fibrous tissue). By inhibiting one or more of the components of fibrosis (or scarring), the overgrowth of granulation tissue may be inhibited or reduced.
Examples of fibrosis-inhibiting agents for use in the present invention include the following: ZD-6474, AP-23573, synthadotin, S-0885, aplidine, ixabepilone, IDN-5390, SB-2723005, ABT-518, combretastatin, anecortave acetate, SB-715992, temsirolimus, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, as well as analogues and derivatives of the aforementioned.
Additional examples of anti-scarring agents which can be used include those having a high potency in the assays described herein (1- 10OnM IC50 range) such as isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, dolastatin 15, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17- DMAG, and tacrolimus; those having a mid-potency in the assays described herein (100-500 nM IC50 range) such as loteprednol etabonate, juglone, prednisolone, puromycin, 3-BAABE, cladribine, and mannose-6-phosphate; and those having a low potency in the assays described herein (500- 1000nm range IC50 range) such as 5-azacytidine, Ly333531(ruboxistaurin), and simvastatin. The drug dose administered from the present compositions for prevention or inhibition of fibrosis in accordance with the present invention will depend on a variety of factors, including the type of formulation, the location of the treatment site, and the type of condition being treated. As inferior vena cava filter devices are made in a variety of configurations and sizes, the exact dose administered will also vary with device size, surface area and design. However, certain principles can be applied in the application of this art. Drug dose can be calculated as a function of dose per unit area (of the treatment site), total drug dose administered can be measured and appropriate surface concentrations of active drug can be determined. Drugs are to be used at concentrations that range from several times more than to 50%, 20%, 10%, 5%, or even less than 1% of the concentration typically used in a single chemotherapeutic systemic dose application. In certain aspects, the anti-scarring agent is released from the composition in effective concentrations in a time period that may be measured from the time of infiltration into tissue adjacent to the device, which ranges from about less than 1 day to about 180 days. Generally, the release time may also be from about less than 1 day to about 180 days; from about 7 days to about 14 days; from about 14 days to about 28 days; from about 28 days to about 56 days; from about 56 days to about 90 days; from about 90 days to about 180 days.
The exemplary anti-fibrosing agents, used alone or in combination, should be administered under the following dosing guidelines. The total amount (dose) of anti-scarring agent in the composition can be in the range of about 0.01 μg-10 μg, or about 10 μg-10 mg, or about 10 mg- 250 mg, or about 250 mg-1000 mg, or about 1000 mg-2500 mg. The dose (amount) of anti-scarring agent per unit area of device or tissue surface to which the agent is applied may be in the range of about 0.01 μg/mm2 - 1 μg/mm2, or about 1 μg/mm2 - 10 μg/mm2, or about 10 μg/mm2 - 250 μg/mm2, or about 250 μg/mm2 - 1000 μg/mm2, or about 1000 μg/mm2 - 2500 μg/mm2. According to another aspect, any anti-infective agent described above may be used in the practice of the present invention. Exemplary anti-infective agents include (A) anthracyclines (e.g., doxorubicin and mitoxantrone), (B) fluoropyrimidines (e.g., 5-FU), (C) folic acid antagonists (e.g., methotrexate), (D) podophylotoxins (e.g., etoposide), (E) camptothecins, (F) hydroxyureas, and (G) platinum complexes (e.g., cisplatin), as well as analogues and derivatives of the aforementioned.
The drug dose administered from the present compositions for prevention or inhibition of infection in accordance with the present invention will depend on a variety of factors, including the type of formulation, the location of the treatment site, and the type of condition being treated. However, certain principles can be applied in the application of this art. Drug dose can be calculated as a function of dose per unit area (of the treatment site), total drug dose administered can be measured and appropriate surface concentrations of active drug can be determined. Drugs are to be used at concentrations that range from several times more than to 50%, 20%, 10%, 5%, or even less than 1% of the concentration typically used in a single anti-infective systemic dose application. In certain aspects, the anti- infective agent is released from the composition in effective concentrations in a time period that may be measured from the time of infiltration into tissue adjacent to the device, which ranges from about less than 1 day to about 180 days. Generally, the release time may also be from about less than 1 day to about 180 days; from about 7 days to about 14 days; from about 14 days to about 28 days; from about 28 days to about 56 days; from about 56 days to about 90 days; from about 90 days to about 180 days.
The exemplary anti-infective agents, used alone or in combination, should be administered under the following dosing guidelines. The total amount (dose) of anti-infective agent in the composition can be in the range of about 0.01 μg-1 μg, or about 1 μg-10 μg, or about 10 μg-1 mg, or about 1 mg to 10 mg, or about 10 mg-100 mg, or about 100 mg to 250 mg, or about 250 mg-1000 mg. The dose (amount) of anti-infective agent per unit area of device or tissue surface to which the agent is applied may be in the range of about 0.01 μg/mm2 - 1 μg/mm2, or about 1 μg/mm2 - 10 μg/mm2, or about 10 μg/mm2 - 100 μg/mm2, or about 100 μg/mm2 to 250 μg/mm2, or about 250 μg/mm2 - 1000 μg/mm2. As different compositions will release the anti-infective agent at differing rates, the above dosing parameters should be utilized in combination with the release rate of the drug from the composition such that a minimum concentration of about 10"8 to 10'7, or about 10'7 to 10-6 about 10"6 to 10'5or about 10"5 to 10"4 of the agent is maintained on the tissue surface.
It should be readily evident based upon the discussions provided herein that combinations of anthracyclines (e.g., doxorubicin or mitoxantrone), fluoropyrimidines (e.g., 5-fluorouracil), folic acid antagonists (e.g., methotrexate), quinolones, and/or podophylotoxins (e.g., etoposide) may be utilized to enhance the antibacterial activity of the composition.
Gastrointestinal Devices
In one aspect, the subject compositions may be infiltrated into tissue adjacent to a gastrointestinal (Gl) device. The subject compositions may contain a therapeutic agent (e.g., an anti-scarring and/or anti-infective agent).
Representative examples of Gl devices that may benefit from having the subject compositions infiltrated into adjacent tissue are provided above in conjunction with the coating of medical devices. Numerous agents or compositions for use with Gl devices have been described above which may be infiltrated into the tissue adjacent to the device (preferably near the device-tissue interface).
Therapeutic agents or pharmaceutical compositionsmay be infiltrated around implanted Gl devices by applying the composition directly and/or indirectly into and/or onto (a) tissue adjacent to the Gl device; (b) the vicinity of the Gl device-tissue interface; (c) the region around the Gl device; and (d) tissue surrounding the Gl device. Methods for infiltrating the subject compositions into tissue adjacent to a Gl device include delivering the composition: (a) to the Gl device surface (e.g., as an injectable, paste, gel or mesh) during the implantation procedure; (b) to the surface of the tissue (e.g., as an injectable, paste, gel, in situ forming gel or mesh) immediately prior to, or during, implantation of the Gl device; (c) to the surface of the Gl device and/or the tissue surrounding the implanted Gl device (e.g., as an injectable, paste, gel, in situ forming gel or mesh) immediately after the implantation of the Gl device; (d) by topical application of the composition into the anatomical space where the Gl device may be placed (particularly useful for this embodiment is the use of polymeric carriers which release the therapeutic agent over a period ranging from several hours to several weeks - fluids, suspensions, emulsions, microemulsions, microspheres, pastes, gels, microparticulates, sprays, aerosols, solid implants and other formulations which release the agent may be delivered into the region where the device may be inserted); (e) via percutaneous injection into the tissue surrounding the Gl device as a solution as an infusate or as a sustained release preparation; (f) by any combination of the aforementioned methods. Combination therapies (i.e., combinations of therapeutic agents and combinations with antithrombotic and/or antiplatelet agents) may also be used. In all cases it is understood that the subject compositions may be infiltrated into tissue adjacent to all or a portion of the device.
According to one aspect, any anti-scarring and/or anti-infective agent described above may be utilized in the practice of the present invention. In one aspect of the invention, the subject compositions infiltrated into tissue adjacent to Gl devices may be adapted to release an agent that inhibits one or more of the four general components of the process of fibrosis (or scarring), including: formation of new blood vessels (angiogenesis), migration and proliferation of connective tissue cells (such as fibroblasts or smooth muscle cells), deposition of extracellular matrix (ECM), and remodeling (maturation and organization of the fibrous tissue). By inhibiting one or more of the components of fibrosis (or scarring), the overgrowth of granulation tissue may be inhibited or reduced.
Examples of fibrosis-inhibiting agents for use in the present invention include the following: ZD-6474, AP-23573, synthadotin, S-0885, aplidine, ixabepilone, IDN-5390, SB-2723005, ABT-518, combretastatin, anecortave acetate, SB-715992, temsirolimus, adalimumab, erucyiphosphocholine, alphastatin, etanercept, humicade, gefitinib, as well as analogues and derivatives of the aforementioned.
Additional examples of anti-scarring agents which can be used include those having a high potency in the assays described herein (1- 10OnM IC50 range) such as isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, dolastatin 15, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17- DMAG, and tacrolimus; those having a mid-potency in the assays described herein (100-500 nM IC5O range) such as loteprednol etabonate, juglone, prednisolone, puromycin, 3-BAABE, cladribine, and mannose-6-phosphate; and those having a low potency in the assays described herein (500- 1000nm range IC50 range) such as 5-azacytidine, Ly333531(ruboxistaurin), and simvastatin.
The drug dose administered from the present compositions for prevention or inhibition of fibrosis in accordance with the present invention will depend on a variety of factors, including the type of formulation, the location of the treatment site, and the type of condition being treated. As Gl devices are made in a variety of configurations and sizes, the exact dose administered will also vary with device size, surface area and design. However, certain principles can be applied in the application of this art. Drug dose can be calculated as a function of dose per unit area (of the treatment site), total drug dose administered can be measured and appropriate surface concentrations of active drug can be determined. Drugs are to be used at concentrations that range from several times more than to 50%, 20%, 10%, 5%, or even less than 1% of the concentration typically used in a single chemotherapeutic systemic dose application. In certain aspects, the anti-scarring agent is released from the composition in effective concentrations in a time period that may be measured from the time of infiltration into tissue adjacent to the device, which ranges from about less than 1 day to about 180 days. Generally, the release time may also be from about less than 1 day to about 180 days; from about 7 days to about 14 days; from about 14 days to about 28 days; from about 28 days to about 56 days; from about 56 days to about 90 days; from about 90 days to about 180 days.
The exemplary anti-fibrosing agents, used alone or in combination, should be administered under the following dosing guidelines. The total amount (dose) of anti-scarring agent in the composition can be in the range of about 0.01 μg-10 μg, or about 10 μg-10 mg, or about 10 mg- 250 mg, or about 250 mg-1000 mg, or about 1000 mg-2500 mg. The dose (amount) of anti-scarring agent per unit area of device or tissue surface to which the agent is applied may be in the range of about 0.01 μg/mm2 - 1 μg/mm2, or about 1 μg/mm2 - 10 μg/mm2, or about 10 μg/mm2 - 250 μg/mm2, or about 250 μg/mm2 - 1000 μg/mm2, or about 1000 μg/mm2 - 2500 μg/mm2.
According to another aspect, any anti-infective agent described above may be used in the practice of the present invention. Exemplary anti-infective agents include (A) anthracyclines (e.g., doxorubicin and mitoxantrone), (B) fluoropyrimidines (e.g., 5-FU), (C) folic acid antagonists (e.g., methotrexate), (D) podophylotoxins (e.g., etoposide), (E) camptothecins, (F) hydroxyureas, and (G) platinum complexes (e.g., cisplatin), as well as analogues and derivatives of the aforementioned.
The drug dose administered from the present compositions for prevention or inhibition of infection in accordance with the present invention will depend on a variety of factors, including the type of formulation, the location of the treatment site, and the type of condition being treated. However, certain principles can be applied in the application of this art. Drug dose can be calculated as a function of dose per unit area (of the treatment site), total drug dose administered can be measured and appropriate surface concentrations of active drug can be determined. Drugs are to be used at concentrations that range from several times more than to 50%, 20%, 10%, 5%, or even less than 1% of the concentration typically used in a single anti-infective systemic dose application. In certain aspects, the anti- infective agent is released from the composition in effective concentrations in a time period that may be measured from the time of infiltration into tissue adjacent to the device, which ranges from about less than 1 day to about 180 days. Generally, the release time may also be from about less than 1 day to about 180 days; from about 7 days to about 14 days; from about 14 days to about 28 days; from about 28 days to about 56 days; from about 56 days to about 90 days; from about 90 days to about 180 days.
The exemplary anti-infective agents, used alone or in combination, should be administered under the following dosing guidelines. The total amount (dose) of anti-infective agent in the composition can be in the range of about 0.01 μg-1 μg, or about 1 μg-10 μg, or about 10 μg-1 mg, or about 1 mg to 10 mg, or about 10 mg-100 mg, or about 100 mg to 250 mg, or about 250 mg-1000 mg. The dose (amount) of anti-infective agent per unit area of device or tissue surface to which the agent is applied may be in the range of about 0.01 μg/mm2 - 1 μg/mm2, or about 1 μg/mm2 - 10 μg/mm2, or about 10 μg/mm2 - 100 μg/mm2, or about 100 μg/mm2 to 250 μg/mm2, or about 250 μg/mm2 - 1000 μg/mm2. As different compositions will release the anti-infective agent at differing rates, the above dosing parameters should be utilized in combination with the release rate of the drug from the composition such that a minimum concentration of about 10~8 to 10~7, or about 10"7 to 10"6 about 10'6 to 10"5or about 10"5 to 10"4 Of the agent is maintained on the tissue surface.
It should be readily evident based upon the discussions provided herein that combinations of anthracyclines (e.g., doxorubicin or mitoxantrone), fluoropyrimidines (e.g., 5-fluorouracil), folic acid antagonists (e.g., methotrexate), quinolones, and/or podophylotoxins (e.g., etoposide) may be utilized to enhance the antibacterial activity of the composition.
Central Venous Catheters
In one aspect, the subject compositions may be infiltrated into tissue adjacent to a central venous catheter (CVC) device. The subject compositions may contain a therapeutic agent (e.g., an anti-scarring and/or anti-infective agent),such that the overgrowth of granulation tissue is inhibited or reduced and/or infection at the site of the CVC device is inhibited or prevented.
Representative examples genital-urinary (GU) stents that may benefit from having the subject compositions infiltrated into adjacent tissue are provided above in conjunction with the coating of medical devices. Numerous agents or compositions for use with tracheal and bronchial have been described above which may be infiltrated into the tissue adjacent to the device (preferably near the device-tissue interface). Polymeric compositions may be infiltrated around implanted CVC devices by applying the composition directly and/or indirectly into and/or onto (a) tissue adjacent to the CVC device; (b) the vicinity of the CVC device-tissue interface; (c) the region around the CVC device; and (d) tissue surrounding the CVC device. Methods for infiltrating the subject compositions into tissue adjacent to a CVC device include delivering the composition: (a) to the CVC device surface (e.g., as an injectable, paste, gel or mesh) during the implantation procedure; (b) to the surface of the tissue (e.g., as an injectable, paste, gel, in situ forming gel or mesh) immediately prior to, or during, implantation of the CVC device; (c) to the surface of the CVC device and/or the tissue surrounding the implanted CVC device (e.g., as an injectable, paste, gel, in situ forming gel or mesh) immediately after the implantation of the CVC device; (d) by topical application of the composition into the anatomical space where the CVC device may be placed (particularly useful for this embodiment is the use of polymeric carriers which release the therapeutic agent over a period ranging from several hours to several weeks - fluids, suspensions, emulsions, microemulsions, microspheres, pastes, gels, microparticulates, sprays, aerosols, solid implants and other formulations which release the agent may be delivered into the region where the device may be inserted); (e) via percutaneous injection into the tissue surrounding the CVC device as a solution as an infusate or as a sustained release preparation; (f) by any combination of the aforementioned methods. Combination therapies (i.e., combinations of therapeutic agents and combinations with antithrombotic and/or antiplatelet agents) may also be used. In all cases it is understood that the subject compositions may be infiltrated into tissue adjacent to all or a portion of the device.
In some aspects, the subject compositions may infiltrated into tissue adjacent to: (a) the exterior surface of the intravascular portion of the CVC device and/or the segment of the CVC device that traverses the skin; (b) exterior surface of the intravascular portion of the CVC device and/or the segment of the CVC device that traverses the skin, where the interior and/or exterior of the CVC device is coated with a composition comprising a therapeutic agent (e.g., an anti-infective agent); (c) the surface of, a subcutaneous "cuff" around the CVC device; (d) other surfaces of the CVC device; and (e) any combination of the aforementioned.
According to one aspect, any anti-scarring and/or anti-infective agent described above may be utilized in the practice of the present invention. In one aspect of the invention, the subject compositions infiltrated into tissue adjacent to CVC devices may be adapted to release an agent that inhibits one or more of the four general components of the process of fibrosis (or scarring), including: formation of new blood vessels (angiogenesis), migration and proliferation of connective tissue cells (such as fibroblasts or smooth muscle cells), deposition of extracellular matrix (ECM), and remodeling (maturation and organization of the fibrous tissue). By inhibiting one or more of the components of fibrosis (or scarring), the overgrowth of granulation tissue may be inhibited or reduced.
Examples of fibrosis-inhibiting agents for use in the present invention include the following: ZD-6474, AP-23573, synthadotin, S-0885, aplidine, ixabepilone, IDN-5390, SB-2723005, ABT-518, combretastatin, anecortave acetate, SB-715992, temsirolimus, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, as well as analogues and derivatives of the aforementioned.
Additional examples of anti-scarring agents which can be used include those having a high potency in the assays described herein (1- 10OnM IC50 range) such as isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, dolastatin 15, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17- DMAG, and tacrolimus; those having a mid-potency in the assays described herein (100-500 nM IC50 range) such as loteprednol etabonate, juglone, prednisolone, puromycin, 3-BAABE, cladribine, and mannose-6-phosphate; and those having a low potency in the assays described herein (500- 1000nm range IC50 range) such as 5-azacytidine, Ly333531(ruboxistaurin), and simvastatin.
The drug dose administered from the present compositions for prevention or inhibition of fibrosis in accordance with the present invention will depend on a variety of factors, including the type of formulation, the location of the treatment site, and the type of condition being treated. As CVC devices are made in a variety of configurations and sizes, the exact dose administered will also vary with device size, surface area and design. However, certain principles can be applied in the application of this art. Drug dose can be calculated as a function of dose per unit area (of the treatment site), total drug dose administered can be measured and appropriate surface concentrations of active drug can be determined. Drugs are to be used at concentrations that range from several times more than to 50%, 20%, 10%, 5%, or even less than 1% of the concentration typically Drug dose can be calculated as a function of dose per unit area (of the treatment site), total drug dose administered can be measured and appropriate surface concentrations of active drug can be determined. Drugs are to be used at concentrations that range from several times more than to 50%, 20%, 10%, 5%, or even less than 1% of the concentration typically used in a single anti-infective systemic dose application. In certain aspects, the anti- infective agent is released from the composition in effective concentrations in a time period that may be measured from the time of infiltration into tissue adjacent to the device, which ranges from about less than 1 day to about 180 days. Generally, the release time may also be from about less than 1 day to about 180 days; from about 7 days to about 14 days; from about 14 days to about 28 days; from about 28 days to about 56 days; from about 56 days to about 90 days; from about 90 days to about 180 days.
The exemplary anti-infective agents, used alone or in combination, should.be administered under the following dosing guidelines. The total amount (dose) of anti-infective agent in the composition can be in the range of about 0.01 μg-1 μg, or about 1 μg-10 μg, or about 10 μg-1 mg, or about 1 mg to 10 mg, or about 10 mg-100 mg, or about 100 mg to 250 mg, or about 250 mg-1000 mg. The dose (amount) of anti-infective agent per unit area of device or tissue surface to which the agent is applied may be in the range of about 0.01 μg/mm2 - 1 μg/mm2, or about 1 μg/mm2 - 10 μg/mm2, or about 10 μg/mm2 - 100 μg/mm2, or about 100 μg/mm2 to 250 μg/mm2, or about 250 μg/mm2 - 1000 μg/mm2. As different compositions will release the anti-infective agent at differing rates, the above dosing parameters should be utilized in combination with the release rate of the drug from the composition such that a minimum concentration of about 10'8 to 10"7, or about 10"7 to 106 about 10 to 10"5 or about 10"5 to 104 of the agent is maintained on the tissue surface.
It should be readily evident based upon the discussions provided herein that combinations of anthracyclines (e.g., doxorubicin or
768 used in a single chemotherapeutic systemic dose application. In certain aspects, the anti-scarring agent is released from the composition in effective concentrations in a time period that may be measured from the time of infiltration into tissue adjacent to the device, which ranges from about less than 1 day to about 180 days. Generally, the release time may also be from about less than 1 day to about 180 days; from about 7 days to about 14 days; from about 14 days to about 28 days; from about 28 days to about 56 days; from about 56 days to about 90 days; from about 90 days to about 180 days.
The exemplary anti-fibrosing agents, used alone or in combination, should be administered under the following dosing guidelines. The total amount (dose) of anti-scarring agent in the composition can be in the range of about 0.01 μg-10 μg, or about 10 μg-10 mg, or about 10 mg- 250 mg, or about 250 mg-1000 mg, or about 1000 mg-2500 mg. The dose (amount) of anti-scarring agent per unit area of device or tissue surface to which the agent is applied may be in the range of about 0.01 μg/mm2 - 1 μg/mm2, or about 1 μg/mm2 - 10 μg/mm2, or about 10 μg/mm2 - 250 μg/mm2, or about 250 μg/mm2 - 1000 μg/mm2, or about 1000 μg/mm2 - 2500 μg/mm2.
According to another aspect, any anti-infective agent described above may be used in the practice of the present invention. Exemplary anti-infective agents include (A) anthracyclines (e.g., doxorubicin and mitoxantrone), (B) fluoropyrimidines (e.g., 5-FU), (C) folic acid antagonists (e.g., methotrexate), (D) podophylotoxins (e.g., etoposide), (E) camptothecins, (F) hydroxyureas, and (G) platinum complexes (e.g., cisplatin), as well as analogues and derivatives of the aforementioned.
The drug dose administered from the present compositions for prevention or inhibition of infection in accordance with the present invention will depend on a variety of factors, including the type of formulation, the location of the treatment site, and the type of condition being treated. However, certain principles can be applied in the application of this art.
767 mitoxantrone), fluoropyrimidines (e.g., 5-fluorouracil), folic acid antagonists (e.g., methotrexate), quinolones, and/or podophylotoxins (e.g., etoposide) may be utilized to enhance the antibacterial activity of the composition.
Ventricular Assist Devices
In one aspect, the subject compositions may be infiltrated into tissue adjacent to a ventricular assist device (VAD). The subject compositions may contain a therapeutic agent (e.g., an anti-scarring and/or anti-infective agent).
Representative examples of VAD's that may benefit from having the subject compositions infiltrated into adjacent tissue are provided above in conjunction with the coating of medical devices. Numerous agents or compositions for use with VAD's have been described above which may be infiltrated into the tissue adjacent to the device (preferably near the device-tissue interface).
Therapeutic agents or pharmaceutical compositionsmay be infiltrated around implanted VADs by applying the composition directly and/or indirectly into and/or onto (a) tissue adjacent to the VAD; (b) the vicinity of the VAD -tissue interface; (c) the region around the VAD; and (d) tissue surrounding the VAD. Methods for infiltrating the subject compositions into tissue adjacent to a VAD include delivering the composition: (a) to the VAD surface (e.g., as an injectable, paste, gel or mesh) during the implantation procedure; (b) to the surface of the tissue (e.g., as an injectable, paste, gel, in situ forming gel or mesh) immediately prior to, or during, implantation of the VAD; (c) to the surface of the VAD and/or the tissue surrounding the implanted VAD (e.g., as an injectable, paste, gel, in situ forming gel or mesh) immediately after the implantation of the VAD; (d) by topical application of the composition into the anatomical space where the VAD may be placed (particularly useful for this embodiment is the use of polymeric carriers which release the therapeutic agent over a period ranging from several hours to several weeks - fluids, suspensions, emulsions, microemulsions, microspheres, pastes, gels, microparticulates, sprays, aerosols, solid implants and other formulations which release the agent may be delivered into the region where the device may be inserted); (e) via percutaneous injection into the tissue surrounding the VAD as a solution as an infusate or as a sustained release preparation; (f) by any combination of the aforementioned methods. Combination therapies (i.e., combinations of therapeutic agents and combinations with antithrombotic and/or antiplatelet agents) may also be used. In all cases it is understood that the subject compositions may be infiltrated into tissue adjacent to all or a portion of the device.
According to the one aspect, any anti-scarring and/or anti- infective agent described above may be utilized in the practice of the present invention. In one aspect of the invention, the subject compositions infiltrated into tissue adjacent to VADs (e.g., LVAD's) may be adapted to release an agent that inhibits one or more of the four general components of the process of fibrosis (or scarring), including: formation of new blood vessels (angiogenesis), migration and proliferation of connective tissue cells (such as fibroblasts or smooth muscle cells), deposition of extracellular matrix (ECM), and remodeling (maturation and organization of the fibrous tissue). By inhibiting one or more of the components of fibrosis (or scarring), the overgrowth of granulation tissue may be inhibited or reduced.
Examples of fibrosis-inhibiting agents for use in the present invention include the following: ZD-6474, AP-23573, synthadotin, S-0885, aplidine, ixabepilone, IDN-5390, SB-2723005, ABT-518, combretastatin, anecortave acetate, SB-715992, temsirolimus, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, as well as analogues and derivatives of the aforementioned.
Additional examples of anti-scarring agents which can be used include those having a high potency in the assays described herein (1- 10OnM IC50 range) such as isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, dolastatin 15, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17- DMAG, and tacrolimus; those having a mid-potency in the assays described herein (100-500 nM IC50 range) such as loteprednol etabonate, juglone, prednisolone, puromycin, 3-BAABE, cladribine, and mannose-6-phosphate; and those having a low potency in the assays described herein (500- 1000nm range IC50 range) such as 5-azacytidine, Ly333531(ruboxistaurin), and simvastatin.
The drug dose administered from the present compositions for prevention or inhibition of fibrosis in accordance with the present invention will depend on a variety of factors, including the type of formulation, the location of the treatment site, and the type of condition being treated. As VADs are made in a variety of configurations and sizes, the exact dose administered will also vary with device size, surface area and design. However, certain principles can be applied in the application of this art. Drug dose can be calculated as a function of dose per unit area (of the treatment site), total drug dose administered can be measured and appropriate surface concentrations of active drug can be determined. Drugs are to be used at concentrations that range from several times more than to 50%, 20%, 10%, 5%, or even less than 1% of the concentration typically used in a single chemotherapeutic systemic dose application. In certain aspects, the anti-scarring agent is released from the composition in effective concentrations in a time period that may be measured from the time of infiltration into tissue adjacent to the device, which ranges from about less than 1 day to about 180 days. Generally, the release time may also be from about less than 1 day to about 180 days; from about 7 days to about 14 days; from about 14 days to about 28 days; from about 28 days to about 56 days; from about 56 days to about 90 days; from about 90 days to about 180 days.
The exemplary anti-fibrosing agents, used alone or in combination, should be administered under the following dosing guidelines. The total amount (dose) of anti-scarring agent in the composition can be in the range of about 0.01 μg-10 μg, or about 10 μg-10 mg, or about 10 mg- 250 mg, or about 250 mg-1000 mg, or about 1000 mg-2500 mg. The dose (amount) of anti-scarring agent per unit area of device or tissue surface to which the agent is applied may be in the range of about 0.01 μg/mm2 - 1 μg/mm2, or about 1 μg/mm2 - 10 μg/mm2, or about 10 μg/mm2 - 250 μg/mm2, or about 250 μg/mm2 - 1000 μg/mm2, or about 1000 μg/mm2 - 2500 μg/mm2.
According to another aspect, any anti-infective agent described above may be used in the practice of the present invention. Exemplary anti-infective agents include (A) anthracyclines (e.g., doxorubicin and mitoxantrone), (B) fluoropyrimidines (e.g., 5-FU), (C) folic acid antagonists (e.g., methotrexate), (D) podophylotoxins (e.g., etoposide), (E) camptothecins, (F) hydroxyureas, and (G) platinum complexes (e.g., cisplatin), as well as analogues and derivatives of the aforementioned.
The drug dose administered from the present compositions for prevention or inhibition of infection in accordance with the present invention will depend on a variety of factors, including the type of formulation, the location of the treatment site, and the type of condition being treated. However, certain principles can be applied in the application of this art. Drug dose can be calculated as a function of dose per unit area (of the treatment site), total drug dose administered can be measured and appropriate surface concentrations of active drug can be determined. Drugs are to be used at concentrations that range from several times more than to 50%, 20%, 10%, 5%, or even less than 1% of the concentration typically used in a single anti-infective systemic dose application. In certain aspects, the anti- infective agent is released from the composition in effective concentrations in a time period that may be measured from the time of infiltration into tissue adjacent to the device, which ranges from about less than 1 day to about 180 days. Generally, the release time may also be from about less than 1 day to about 180 days; from about 7 days to about 14 days; from about 14 days to about 28 days; from about 28 days to about 56 days; from about 56 days to about 90 days; from about 90 days to about 180 days.
The exemplary anti-infective agents, used alone or in combination, should be administered under the following dosing guidelines. The total amount (dose) of anti-infective agent in the composition can be in the range of about 0.01 μg-1 μg, or about 1 μg-10 μg, or about 10 μg-1 mg, or about 1 mg to 10 mg, or about 10 mg-100 mg, or about 100 mg to 250 mg, or about 250 mg-1000 mg. The dose (amount) of anti-infective agent per unit area of device or tissue surface to which the agent is applied may be in the range of about 0.01 μg/mm2 - 1 μg/mm2, or about 1 μg/mm2 - 10 μg/mm2, or about 10 μg/mm2 - 100 μg/mm2, or about 100 μg/mm2 to 250 μg/mm2, or about 250 μg/mm2 - 1000 μg/mm2. As different compositions will release the anti-infective agent at differing rates, the above dosing parameters should be utilized in combination with the release rate of the drug from the composition such that a minimum concentration of about 10"8 to 10"7, or about lO'7 to 106 about 10"6 to 10'5 or about 10'5 to 10'4 of the agent is maintained on the tissue surface.
It should be readily evident based upon the discussions provided herein that combinations of anthracyclines (e.g., doxorubicin or mitoxantrone), fluoropyrimidines (e.g., 5-fluorouracil), folic acid antagonists (e.g., methotrexate), quinolones, and/or podophylotoxins (e.g., etoposide) may be utilized to enhance the antibacterial activity of the composition.
Spinal Implants
In one aspect, the subject compositions may be infiltrated into tissue adjacent to a spinal implant (e.g., a spinal prosthesis). The subject compositions may contain a therapeutic agent (e.g., an anti-scarring and/or anti-infective agent).
Representative examples of spinal implants that may benefit from having the subject compositions infiltrated into adjacent tissue are provided above in conjunction with the coating of medical devices. Numerous agents or compositions for use with spinal implants have been described above which may be infiltrated into the tissue adjacent to the device (preferably near the device-tissue interface). Infiltration of the subject compositions comprising a fibrosis-inhibiting agent and/or anti- infective agent into tissue adjacent to a spinal implant can minimize fibrosis (or scarring) in the vicinity of the implant and/or may reduce or prevent the formation of adhesions between the implant and the surrounding tissue and/or may inhibit or prevent infection in the vicinity of the implant.
Therapeutic agents or pharmaceutical compositionsmay be infiltrated around implanted spinal implants by applying the composition directly and/or indirectly into and/or onto (a) tissue adjacent to the spinal implant; (b) the vicinity of the spinal implant-tissue interface; (c) the region around the spinal implant; and (d) tissue surrounding the spinal implant. Methods for infiltrating the subject compositions into tissue adjacent to a spinal implant include delivering the composition: (a) to the spinal implant surface (e.g., as an injectable, paste, gel or mesh) during the implantation procedure; (b) to the surface of the tissue (e.g., as an injectable, paste, gel, in situ forming gel or mesh) immediately prior to, or during, implantation of the spinal implant; (c) to the surface of the spinal implant and/or the tissue surrounding the implanted spinal implant (e.g., as an injectable, paste, gel, in situ forming gel or mesh) immediately after the implantation of the spinal implant; (d) by topical application of the composition into the anatomical space where the spinal implant may be placed (particularly useful for this embodiment is the use of polymeric carriers which release the therapeutic agent over a period ranging from several hours to several weeks - fluids, suspensions, emulsions, microemulsions, microspheres, pastes, gels, microparticulates, sprays, aerosols, solid implants and other formulations which release the agent may be delivered into the region where the device may be inserted); (e) via percutaneous injection into the tissue surrounding the spinal implant as a solution as an infusate or as a sustained release preparation; (f) by any combination of the aforementioned methods. Combination therapies (i.e., combinations of therapeutic agents and combinations with antithrombotic and/or antiplatelet agents) may also be used. In all cases it is understood that the subject compositions may be infiltrated into tissue adjacent to all or a portion of the device.
In one aspect, the subject composition comprising an anti- scarring and/or anti-infective agent is infiltrated into the tissue adjacent to a spinal implant (e.g., an implantable cages or disc). In certain aspects, the spinal implant may be coated with (or adapted to contain) a fibrosis-inducing agent (e.g., silk or talc) on one part of the device and the subject composition comprising an anti-scarring may be infiltrated into tissue adjacent to another part of the device. For example, the outer surface of the implant (e.g., a vertebral implant) may be coated with a fibrosis-inducing agent to improve adhesion between the device and the surrounding tissue, while the subject composition comprising an anti-scarring may be infiltrated into tissue adjacent to the interior of the device to minimize adhesion of tissue to the interior of the implant. Examples of fibrosis-inducing agents and methods of using fibrosis-inducing agents in combination with spinal implants are described in co-pending application entitled, "Medical Implants and Fibrosis-inducing Agents," filed November 20, 2003 (U.S. Ser. No. 60/524,023) and June 9, 2004 (U.S. Ser. No. 60/578,471). Additional examples of fibrosis-inducing agents include angiolytic agents (i.e., agents which cause leakage of vessels) such as EXHERIN from Adherex Technologies Inc. (Canada). .
According to one aspect, any adhesion or fibrosis-inhibiting agent and/or anti-infective agent described above can be utilized in the practice of the present invention. In one aspect of the invention, the subject compositions infiltrated into tissue adjacent to spinal implants may be adapted to release an agent that inhibits one or more of the four general components of the process of fibrosis (or scarring), including: formation of new blood vessels (angiogenesis), migration and proliferation of connective tissue cells (such as fibroblasts or smooth muscle cells), deposition of extracellular matrix (ECM), and remodeling (maturation and organization of the fibrous tissue). By inhibiting one or more of the components of fibrosis (or scarring), the overgrowth of granulation tissue may be inhibited or reduced.
Examples of fibrosis-inhibiting agents for use in the present invention include the following: ZD-6474, AP-23573, synthadotin, S-0885, aplidine, ixabepilone, IDN-5390, SB-2723005, ABT-518, combretastatin, anecortave acetate, SB-715992, temsirolimus, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, as well as analogues and derivatives of the aforementioned.
Additional examples of anti-scarring agents which can be used include those having a high potency in the assays described herein (1- 10OnM IC50 range) such as isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, dolastatin 15, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17- DMAG, and tacrolimusrthose having a mid-potency in the. assays described herein (100-500 nM IC50 range) such as loteprednol etabonate, juglone, prednisolone, puromycin, 3-BAABE, cladribine, and mannose-6-phosphate; and those having a low potency in the assays described herein (500- 1000nm range IC50 range) such as 5-azacytidine, Ly333531(ruboxistaurin), and simvastatin.
The drug dose administered from the present compositions for prevention or inhibition of fibrosis in accordance with the present invention will depend on a variety of factors, including the type of formulation, the location of the treatment site, and the type of condition being treated. As spinal implants are made in a variety of configurations and sizes, the exact dose administered will also vary with device size, surface area and design. However, certain principles can be applied in the application of this art. Drug dose can be calculated as a function of dose per unit area (of the treatment site), total drug dose administered can be measured and appropriate surface concentrations of active drug can be determined. Drugs are to be used at concentrations that range from several times more than to 50%, 20%, 10%, 5%, or even less than 1% of the concentration typically used in a single chemotherapeutic systemic dose application. In certain aspects, the anti-scarring agent is released from the composition in effective concentrations in a time period that may be measured from the time of infiltration into tissue adjacent to the device, which ranges from about less than 1 day to about 180 days. Generally, the release time may also be from about less than 1 day to about 180 days; from about 7 days to about 14 days; from about 14 days to about 28 days; from about 28 days to about 56 days; from about 56 days to about 90 days; from about 90 days to about 180 days.
The exemplary anti-fibrosing agents, used alone or in combination, should be administered under the following dosing guidelines. The total amount (dose) of anti-scarring agent in the composition can be in the range of about 0.01 μg-10 μg, or about 10 μg-10 mg, or about 10 mg- 250 mg, or about 250 mg-1000 mg, or about 1000 mg-2500 mg. The dose (amount) of anti-scarring agent per unit area of device or tissue surface to which the agent is applied may be in the range of about 0.01 μg/mm2 - 1 μg/mm2, or about 1 μg/mm2 - 10 μg/mm2, or about 10 μg/mm2 - 250 μg/mm2, or about 250 μg/mm2 - 1000 μg/mm2, or about 1000 μg/mm2 - 2500 μg/mm2.
According to another aspect, any anti-infective agent described above may be used in the practice of the present invention. Exemplary anti-infective agents include (A) anthracyclines (e.g., doxorubicin and mitoxantrone), (B) fluoropyrimidines (e.g., 5-FU), (C) folic acid antagonists (e.g., methotrexate), (D) podophylotoxins (e.g., etoposide), (E) camptothecins, (F) hydroxyureas, and (G) platinum complexes (e.g., cisplatin), (H) quinolones, as well as analogues and derivatives of the aforementioned.
The drug dose administered from the present compositions for prevention or inhibition of infection in accordance with the present invention will depend on a variety of factors, including the type of formulation, the location of the treatment site, and the type of condition being treated. However, certain principles can be applied in the application of this art. Drug dose can be calculated as a function of dose per unit area (of the treatment site), total drug dose administered can be measured and appropriate surface concentrations of active drug can be determined. Drugs are to be used at concentrations that range from several times more than to 50%, 20%, 10%, 5%, or even less than 1% of the concentration typically used in a single anti-infective systemic dose application. In certain aspects, the anti- infective agent is released from the composition in effective concentrations in a time period that may be measured from the time of infiltration into tissue adjacent to the device, which ranges from about less than 1 day to about 180 days. Generally, the release time may also be from about less than 1 day to about 180 days; from about 7 days to about 14 days; from about 14 days to about 28 days; from about 28 days to about 56 days; from about 56 days to about 90 days; from about 90 days to about 180 days.
The exemplary anti-infective agents, used alone or in combination, should be administered under the following dosing guidelines. The total amount (dose) of anti-infective agent in the composition can be in the range of about 0.01 μg-1 μg, or about 1 μg-10 μg, or about 10 μg-1 mg, or about 1 mg to 10 mg, or about 10 mg-100 mg, or about 100 mg to 250 mg, or about 250 mg-1000 mg. The dose (amount) of anti-infective agent per unit area of device or tissue surface to which the agent is applied may be in the range of about 0.01 μg/mm2 - 1 μg/mm2, or about 1 μg/mm2 - 10 μg/mm2, or about 10 μg/mm2 - 100 μg/mm2, or about 100 μg/mm2 to 250 μg/mm2, or about 250 μg/mm2 - 1000 μg/mm2. As different compositions will release the anti-infective agent at differing rates, the above dosing parameters should be utilized in combination with the release rate of the drug from the composition such that a minimum concentration of about 10~8 to 10"7, or about 10"7 to 10"6 about 10"6 to 10"5 or about 10'5 to 10"4 of the agent is maintained on the tissue surface.
It should be readily evident based upon the discussions provided herein that combinations of anthracyclines (e.g., doxorubicin or mitoxantrone), fluoropyrimidines (e.g., 5-fluorouracil), folic acid antagonists (e.g., methotrexate), quinolones, (e.g., methotrexate), quinolones, and/or podophylotoxins (e.g., etoposide) may be utilized to enhance the antibacterial activity of the composition.
The present invention, in various aspects, provides the following itemized embodiments:
1. A device, comprising a medical device (e.g., an implant) and an anti-scarring agent or a composition comprising an anti-scarring agent, wherein the agent inhibits scarring between the device and a host into which the device is implanted.
2. The device of item 1 wherein the agent is an adensosine A2A receptor antagonist.
3. The device of item 1 wherein the agent is an AKT inhibitor.
4. The device of item 1 wherein the agent is an alpha 2 integrin antagonist, wherein the alpha 2 integrin antagonist is Pharmaprojects No. 5754 (Merck KgaA).
5. The device of item 1 wherein the agent an alpha 4 integrin antagonist. 6. The device of item 1 wherein the agent is an alpha 7 nicotinic receptor agonist.
7. The device of item 1 wherein the agent is an angiogenesis inhibitor selected from the group consisting of AG-12,958 (Pfizer), ATN-161 (Attention LLC), neovastat, an angiogenesis inhibitor from Jerina AG (Germany), NM-3 (Mercian), VGA-1155 (Taisho), FCE-26644 (Pfizer), FCE-26950 (Pfizer), FPMA (Meiji Daries), FR-111142 (Fujisawa), GGTI-298, GM-1306 (Ligand), GPA-1734 (Novartis), NNC-47-0011 (Novo Nordisk), herbamycin (Nippon Kayaku), lenalidomide (Celegene), IP-10 (NIH), ABT-828 (Abbott), KIN-841 (Tokushima University, Japan), SF-1126 (Semafore Pharmaceuticals), laminin technology (NIH), CHIR-258 (Chiron), NVP-AEW541 (Novartis), NVP-AEW541 (Novartis), Vt16907 (Alchemia), OXI-8007 (Oxigene), EG-3306 (Ark Therapeutics), Maspin (Arriva), ABT- 567 (Abbott), PPI-2458 (Praecis Pharmaceuticals), CC-5079, CC-4089 (Celgene), HIF-1 alpha inhibitors (Xenova), S-247 (Pfizer), AP-23573 (Ariad), AZD-9935 (Astra Zeneca), mebendazole (Introgen Therapeutics), MetAP-2 inhibitors (GlaxoSmithKline), AG-615 (Angiogene Pharmaceuticals), Tie-2 antagonists (Hybrigenics), NC-381 , CYC-381, NC-169, NC-219, NC-383, NC-384, NC-407 (Lorus Therapeutics), ATN-224 (Attenuon), ON-01370 (Onconova), Vitronectin antagonists (Amgen), SDX-103 (Salmedix), Vitronectin antagonists (Shire), CHP (Riemser), TEK (Amgen), Anecortave acetate (Alcon), T46.2 (Matrix Therapeutics), HG-2 (Heptagen), TEM antagonists (Genzyme), Oxi-4500 (Oxigene), ATN-161 (Attenuon), WX-293 (Wilex), M-2025 (Metris Therapeutics), Alphastatin (BioActa), YH-16 (Yantai Rongchang), BIBF-1120 (Boehringer Ingelheim), BAY-57-9352 (Bayer), AS- 1404 (Cancer Research Technology), SC-77964 (Pfizer), glycomimetics (BioTie Therapies), TIE-2 Inhibitors (Ontogen), DIMI, Octamer (Octamer), ABR-215050 (Active Biotech), ABT-518 (Abbott), KDR inhibitors (Abbott), BSF-466895 (Abbott), SCH-221153 (Schering-Plough), DAC:antiangiogenic (ConjuChem), TFPI (EntreMed), AZD-2171 (Astra-Zenaca), CDC-394 (Celgene), LY290293 (EIi Lilly), IDN-5390 (Indena), Kdr Kinase Inhibitors (Merck), CT-113020, CT-116433, CT-116563, CT-31890, CT-32228) (Cell Therapeutics), A-299620 (Abbott), TWEAK Inhibitor (Amgen), VEGF modulators (Johnson and Johnson), Tum-N53, tumstatin (Genzyme), Thios- 1 , Thios-2 (Thios Pharmaceuticals), MV-6401 (Miravant Medical Technologies), Spisulosine (PharmaMar), CEP-7055 (Cephalon), AUV-201 (Auvation), LM-609 (EIi Lilly), SKF-106615 (AnorMED), Oglufanide disodium (Cytran), BW-114 (Pharminox), Calreticulin (NIH), WX-678 (Wilex), SD-7784 (Pfizer), WX-UK1 (Wilex), SH-268 (Schering AG), 2-Me-PGA (Celgene), S- 137 (Pfizer), ZD-6126 (Angiogene Pharmaceuticals), SG-292 (SignalGen), Benefin (Lane Labs), A6, A36 (Angstrom), SB-2723005 (GlaxoSmithKline), SC-7 (Cell Therapeutics), ZEN-014 (AEterna Zentaris), 2-methoxyestradiol (EntreMed), NK-130119 (Nippon Kayaku), CC-10004 (Celgene), AVE- 8062A (Ajinomoto), Tacedinaline (Pfizer), Actinonin (Tokyo Metropolitan Institute of Medical Science), Lenalidomide (Celgene), VGA-1155, BTO-956 (SRI International), ER-68203-00 (Eisai), CT-6685 (U6B), JKC-362 (Phoenix Pharmaceuticals), DMI-3798 (DMI Biosciences, Angiomate (Ipsen), ZD-6474 (AstraZeneca), CEP-5214 (Cephalon), Canstatin (Genzyme), NM-3 (Mercian), Oridigm (MediQuest Therapeutics), Exherin (Adherex), BLS-0597 (Boston Life Sciences), PTC-299 (PTC Therapeutics), NPI-2358 (Nereus Pharmaceuticals), CGP-79787 (Novartis), AEE-788 (Novartis), CKD-732 (Chong Kun Dang), CP-564959 (OSI Pharmaceuticals), CM-101 (CarboMed), CT-2584, CT3501 (Cell Therapeutics), combretastatin and analogues and derivatives thereof (Oxigene), Rebimastat (Bristol-Meyers Squibb), Dextrin 2-sulfate (ML Laboratories), Cilengitide (Merk KGaA), NSC-706704 (Pharminox), KRN- 951 (Kirin Brewery), Ukrain, NSC-631570 (Nowicky Pharma), Tecogalan sodium (Daiichi Pharmaceutical), Tz-93 (Tsumura), TBC-1635 (Encysive Pharmaceuticals), TAN-1120 (Takeda), Semaxanib (Pfizer), BDI-7800 (Biopharmacopae), SD-186, SD-983 (Bristol-Meyers Squibb), SB-223245 (GlaxoSmithKline), SC-236 (Pfizer), RWJ-590973 (Johnson and Johnson), ILX-1850 (Genzyme), SC-68488, S-836 (Pfizer), CG-55069-11 (CuraGen), Ki-23057 (Kirin Brewery), CCX-700 (Chemoentryx), Pegaptanib octasodium (Giled Sciences), ANGIOCOL (available from Biostratum Inc.), or an analogue or derivative thereof.
8. The device of item 1 wherein the agent is an apoptosis antagonist.
9. The device of item 1 wherein the agent is an apoptosis activator.
10. The device of item 1 wherein the agent is a beta 1 integrin antagonist.
11. The device of item 1 wherein the agent is a beta tubulin inhibitor.
12. The device of item 1 wherein the agent is a blocker of enzyme production in Hepatitis C.
13. The device of item 1 wherein the agent is a Bruton's tyrosine kinase inhibitor.
14. The device of item 1 wherein the agent is a calcineurin inhibitor.
15. The device of item 1 wherein the agent is a caspase 3 inhibitor.
16. The device of item 1 wherein the agent is a CC chemokine receptor antagonist. 17. The device of item 1 wherein the agent is a cell cycle inhibitor selected from the group consisting of SNS-595 (Sunesis), synthadotin, KRX-0403, and an analogue or derivative thereof.
18. The device of item 1 wherein the agent is a cathepsin B inhibitor.
19. The device of item 1 wherein the agent is a cathepsin K inhibitor, wherein the cathepsin K inhibitor is 462795 (GlaxoSmithKline), INPL-022-D6 (Amura Therapeutics), an analogue or derivative thereof).
20. The device of item 1 wherein the agent is a cathepsin L inhibitor.
21. The device of item 1 wherein the agent is a CD40 antagonist. . . -
22. The device of item 1 wherein the agent is a chemokine receptor agonist.
23. The device of item 1 wherein the agent is a chymase inhibitor.
24. The device of item 1 wherein the agent is a collagenase antagonist.
25. The device of item 1 wherein the agent is a CXCR antagonist.
26. The device of item 1 wherein the agent is a cyclin dependent kinase inhibitor selected from the group consisting of a CDK-1 inhibitor, a CDK-2 inhibitor, a CDK- 4 inhibitor, a CDK-6 inhibitor, a CAK1 inhibitor from GPC Biotech and Bristol-Myers Squibb, RGB-286199 (GPC Biotech), an anticancer agent from Astex Technology, a CAK1 inhibitor from GPC Biotech, a CDK inhibitor from Sanofi-Aventis, a CDK1/CDK2 inhibitor from Amgen, a CDK2 inhibitor from SUGEN-2 (Pfizer), a hearing loss therapy agent (Sound Pharmaceuticals), PD-0332991 (Pfizer), RGB-286199 (GPC Biotech), Ro-0505124. (Hoffmann-La Roche), a Ser/Thr kinase inhibitor from Lilly (EIi Lilly), CVT-2584 (CAS No. 199986-75-9) (CV Therapeutics), and an analogue or derivative thereof.
27. The device of item 1 wherein the agent is a cyclooxygenase 1 inhibitor.
28. The device of item 1 wherein the agent is a DHFR inhibitor.
29. The device of item 1 wherein the agent is a dual integrin inhibitor.
30. The device of item 1 wherein the agent is an elastase inhibitor.
31. The device of item 1 wherein the agent is an elongation factor-1 alpha inhibitor.
32. The device of item 1 wherein the agent is an endothelial growth factor antagonist.
33. The device of item 1 wherein the agent is an endothelial growth factor receptor kinase inhibitor selected from the group consisting of sorafenib tosylate (Bayer), AAL-993 (Novartis), ABP-309 (Novartis), BAY- 57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), E-7080 (Eisai), EG- 3306 (Ark Therapeutics), EXEL-2880 (Exelixis), GW-654652 (GlaxoSmithKline), a KDR inhibitor from LG Life Sciences, CT-6685 and CT- 6729 (UCB), KRN-633 and KRN-951 (Kirin Brewery), OSI-930 (OSI Pharmaceuticals), SP-5.2 (Supratek Pharma), SU-11657 (Pfizer), a Tie-2 antagonist (Hybrigenics), a VEGFR-2 kinase inhibitor (Bristol-Myers Squibb), XL-647 (Exelixis), a KDR inhibitor from Abbott Laboratories, sorafenib tosylate, and an analogue or derivative thereof.
34. The device of item 1 wherein the agent is an endotoxin antagonist.
35. The device of item 1 wherein the agent is an epothilone and tubulin binder.
36. The device of item 1 wherein the agent is an estrogen receptor antagonist.
37. The device of item 1 wherein the agent is an FGF inhibitor.
38. The device of item 1 wherein the agent is a farnexyl transferase inhibitor.
39. The device of item 1 wherein the agent is famesyltransferase inhibitor selected from the group of A-197574 (Abbott), a farnesyltransferase inhibitor from Servier, FPTIII (Strathclyde Institute for Drug R), LB-42908 (LG Life Sciences), Pharmaprojects No. 5063 (Genzyme), Pharmaprojects No. 5597 (Ipsen), Yissum Project No. B-1055 (Yissum), and an analogue or derivative thereof. 40. The device of item 1 wherein the agent is an FLT-3 kinase inhibitor.
41. The device of item 1 wherein the agent is an FGF receptor kinase inhibitor.
42. The device of item 1 wherein the agent is a fibrinogen antagonist selected from the group consisting of AUV-201 (Auvation), MG- 13926 (Sanofi-Aventis), plasminogen activator (CAS No. 105913-11-9) (from Sanofi-Aventis or UCB), plasminogen activator-2 (tPA-2) (Sanofi- Aventis), pro-urokinase (CAS No. 82657-92-9) (Sanofi-Aventis), and an analogue or derivative thereof.
43. The device of item 1 wherein the agent is a heat shock protein 90 antagonist selected from the group consisting of SRN-005 (Sirenade), geldanamycin, NSC-33050 (17-allylaminogeldanamycin; 17- AAG), 17-dimethylaminoethylamino-17-demethoxy-geldanamycin (17- DMAG), rifabutin (rifamycin XIV, 1\4-didehydro-1-deoxy-1,4-dihydro-5'-(2- methylpropyl)-1-oxo-), and an analogue or derivative thereof.
44. The device of item 1 wherein the agent is a histone deacetylase inhibitor.
45. The device of item 1 wherein the agent is an HMGCoA reductase inhibitor selected from the group consisting of an atherosclerosis therapeutic from Lipid Sciences, ATI-16000 (ARYx Therapeutics), KS-01- 019 (Kos Pharmaceuticals), Pharmaprojects No. 2197 (Sanofi-Aventi), RP 61969 (Sanofi-Aventis), and an analogue or derivative thereof.
46. The device of item 1 wherein the agent is an ICAM inhibitor. 47. The device of item 1 wherein the agent is an IL, ICE and IRAK antagonist, wherein the antagonist is a CJ-14877, CP-424174 (Pfizer), NF-61 (Negma-Lerads), and an analogue or derivative thereof.
48. The device of item 1 wherein the agent is an IL-2 inhibitor.
49. The device of item 1 wherein the agent is an immunosuppressant selected from the group consisting of teriflunomide (Sanofi Aventis), chlorsulfaquinoxalone (NSC-339004), chlorsulfaquinoxalone sulfate, CS-712 (Sankyo), ismomultin alfa (CAS No. 457913-93-8) (Akzo Nobel), antiallergics from GenPat77, antiinflammatories or AT-005 (Androclus Therapeutics), autoimmune disease therapeutics from EpiVax, BN-007 (Bone), budesonide (CAS No. 51333-22- 3) (MAP Pharmaceuticals), CO-14 (Genzyme), edratide (CAS No. 433922- 67-9) (Teva), EP-314 (Enanta), eprovafen.(CAS No. 101335-99-3) (Sanofi- Aventis), HWA-131 (CAS No. 118788-41-3) (Sanofi-Aventis), immunomodulators from MerLion Pharmaceuticals, immunosuppressives from Alchemia, IPL-12 (Inflazyme), MDL-9563 (CAS No. 27086-86-8) (Sanofi-Aventis), Pharmaprojects No. 2330 (Sanofi-Aventis), Pharmaprojects No. 6426 (Abgenix), PXS-25 (Pharmaxis), rosmarinic acid (CAS No. 20283-92-5) (Sanofi-Aventis), RP 42927 or RP 54745 (CAS No. 135330-08-4) (Sanofi-Aventis), SGN-35 (Seattle Genetics), ST-1959 (Sigma-Tau), type I diabetes therapy from SYNX Pharma, UNIL-88 (Debiopharm), VP-025 (Vasogen), VR-694 (Vectura), PRTX-001 (Protalex), and an analogue or derivative thereof.
50. The device of item 1 wherein the agent is an IMPDH (inosine monophosphate). 51. The device of item 1 wherein the agent is an integrin antagonist.
52. The device of item 1 wherein the agent is an interleukin antagonist.
53. The device of item 1 wherein the agent is an inhibitor of type III receptor tyrosine kinase.
54. The device of item 1 wherein the agent is an irreversible inhibitor of enzyme methionine aminopeptidase type 2.
55. The device of item 1 wherein the agent is an isozyme selective delta protein kinase C inhibitor.
56. The device of item 1 wherein the agent a JAK3 enzyme inhibitor.
57. The device of item 1 wherein the agent is a JNK inhibitor.
58. The device of item 1 wherein the agent is a kinase inhibitor.
59. The device of item 1 wherein the agent is kinesin antagonist.
60. The device of item 1 wherein the agent is a kinesin antagonist. 61. The device of item 1 wherein the agent is a leukotriene inhibitor and antagonist selected from the group consisting of ambicromil (CAS No. 58805-38-2) (Sanofi-Aventis), amelubant (CAS No. 346735-24-8) (Boehringer Ingelheim), DW-1141 (Dong Wha), ebselen (Daiichi Pharmaceutical), ibudilast (Kyorin), leucotriene inhibitors from Sanofi- Aventis, lymphotoxin-beta receptor (LT-D) from Biogen Idee, Pharmaprojects No. 1535 and 2728 (CAS No. 119340-33-9) (Sanofi- Aventis), R-112 (Rigel), Rev-5367 (CAS No. 92532-05-3) (Sanofi-Aventis), RG-14893 (CAS No. 141835-49-6) (Sanofi-Aventis), RG-5901-A (CAS No. 101910-24-1), 92532-23-5, RP 66153 (CAS No. 142422-79-5), RP 66364 (CAS No.186916-92-5), RP 69698 (CAS No. 141748-00-7) (Sanofi-Aventis), SC-411930 (Pfizer), SC-41930 (CAS No. 120072-59-5) (Pfizer), SC-50605 (CAS No. 138828-39-4) (Pfizer), SC-51146 (CAS No. 141059-52-1), SC- 53228 (CAS No. 153633-01-3) (Pfizer), spaglumic acid (ZY-15106) (CAS No.3106-85-2), 80619-64-3 (Novartis), tipredane (CAS No. 85197-77-9) (Bristol-Myers Squibb), U-75302 (CAS No. 119477-85-9) (Pfizer), and analogue or derivative thereof.
62. The device of item 1 wherein the agent is an MAP kinase inhibitor.
63. The device of item 1 wherein the agent is a matrix metalloproteinase inhibitor.
64. The device of item 1 wherein the agent is an MCP- CCR2 inhibitor.
65. The device of item 1 wherein the agent is an mTOR inhibitor. 66. The device of item 1 wherein the agent is an mTOR kinase inhibitor.
67. The device of item Iwherein the agent is a microtubule inhibitor selected from the group consisting of antibody-maytansinoid conjugates from Biogen Idee, colchicines (MantiCore Pharmaceuticals), anticancer immunoconjugates from Johnson & Johnson, DIME from Octamer, gni-1f (GNI), huC242-DM4, huMy9-6-DM1 (ImmunoGen), IDN- 5404 (Indena), IMO-098, IMOderm (Imotep), mebendazole (Introgen Therapeutics), microtubule poisons from Cambridge Enterprise, paclitaxel such as LOTAX from Aphios (CAS No. 33069-62-4), Genexol-PM from Samyang, Pharmaprojects No. 6383 (Tapestry Pharmaceuticals), RPR-
112378 (Sanofi-Aventis), SGN-75 (Seattle Genetics), SPL-7435 (Starpharma), SSR-250411 (Sanofi-Aventis), trastuzumab-DM1 (Genentech), vinorelbine, and an analogue or derivative thereof.
68. The device of item 1 wherein the agent is an MIF inhibitor.
69. The device of item 1 wherein the agent is an MMP inhibitor.
70. The device of item 1 wherein the agent is a neurokinin (NK) antagonist selected from the group consisting of anthrotainin (CAS No. 148084-40-6) (Sanofi-Aventis), an IBS thereapeutic from ArQuIe, MDL- 105212A (CAS No. 167261-60-1) (Ssanofi-Aventis), Pharmaprojects No. 2744, 3258 (CAS No. 139167-47-8) 4006, 4201, or 5986 (Sanofi-Aventis), RP 67580 (CAS No. 135911-02-3), SR-144190 (CAS No. 201152-86-5), SSR-240600, SSR-241586 (Sanofi-Aventis), TKA-457 (Novartis), vestipitant mesylate (CAS No. 334476-64-1) (GlaxoSmithKline), Win-64821 (Sanofi- Aventis), PRX-96026 (Predix Pharmaceuticals), and an analogue or derivative thereof.
71. The device of item 1 wherein the agent is an NF kappa B inhibitor selected from the group consisting of AVE-0545 or AVE-0547 (Sanofi-Aventis), bortezomib (CAS No. 179324-69-7) (Millennium Pharmaceuticals), dexanabinol (CAS No. 112924-45-5) (Pharmos), dexlipotam (Viatris), Pharmaprojects No. 6283 (INDRA) (OXiGENE), IPL- 576092 (CAS No. 137571-30-3) (Inflazyme), NFKB decoy (Corgentech), NFKB decoy oligo (AnGes MG), NFKB's from Ariad, osteoporosis treatments or S5 (F005) from Fulcrum Pharmaceuticals, P61 (Phytopharm), R-flurbiprofen (CAS No. 5104-49-4) (Encore Pharmaceuticals), and an analogue or derivative thereof.
72. The device of item 1 wherein the agent is a nitric oxide agonist.
73. The device of item 1 wherein the agent is an ornithine decarboxylase inhibitor.
74. The device of item 1 wherein the agent is a p38 MAP kinase inhibitor selected from the group consisting of AZD-6703 (AstraZeneca), JX-401 (Jexys Pharmaceuticals), BMS-2 (Bristol-Myers Squibb), a p38 MAP kinase inhibitor from Novartis, a p38-alpha MAP kinase inhibitor from Amphora, Pharmaprojects No. 5704 (Pharmacopeia), RPR- 200765A (Sanofi-Aventis), SD-282 (Johnson & Johnson), TAK-715 (Takeda), and an analogue or derivative thereof.
75. The device of item 1 wherein the agent is a palmitoyl- protein thioesterase inhibitor. 76. The device of item 1 wherein the agent is a PDGF receptor kinase inhibitor selected from the group consisting of AAL-993, AMN-107, or ABP-309 (Novartis), AMG-706 (Amgen), BAY-57-9352 (Bayer), CDP-860 (UCB), E-7080 (Eisai), imatinib (CAS No. 152459-95-5) (Novartis), OSI-930 (OSI Pharmaceuticals), RPR-127963E (Sanofi-Aventis), RWJ-540973 (Johnson & Johnson), sorafenib tosylate (Bayer), SU-11657 (Pfizer), tandutinib (CAS No. 387867-13-2) (Millennium Pharmaceuticals), vatalanib (Novartis), ZK-CDK (Schering AG), and an analogue or derivative thereof.
77. The device of item 1 wherein the agent is (-)-halofenate (Metabolex), AMG-131 (Amgen), antidiabetics from Japan Tobacco, AZD- 4619, AZD-8450, AZD-8677 (AstraZeneca), DRF-10945, balaglitazone (Dr Reddy's), CS-00088, CS-00098 (Chipscreen Biosciences), E-3030 (Eisai), etalocib (CAS No. 161172-51-6) (EIi Lilly), GSK-641597 (Ligand), GSK- 677954 (GlaxoSmithKline), GW-409544 (Ligand), GW-590735 (GlaxoSmithKline), K-111 (Hoffmann-La Roche), LY-518674 (EIi Lilly), LY- 674 (Ligand), LY-929 (Ligand), MC-3001, MC-3002 (MaxoCore Pharmaceuticals), metformin HCI + pioglitazone (CAS No. 1115-70-4 and 112529-15-4), ACTOPLUS MET from Andrx), muraglitazar (CAS No. 331741-94-7) (Bristol-Myers Squibb), naveglitazar (Ligand), oleoylethanolamide (Kadmus Pharmaceuticals), ONO-5129, pioglitazone hydrochloride (CAS No. 111025-46-8 and 112529-15-4) (Takeda), PLX-204 (Plexxikon), PPAR agonists from Genfit, PPAR delta agonists from EIi Lilly, PPAR-alpha agonists from CrystalGenomics, PPAR-gamma modulators and PPAR-β modulators from CareX, rosiglitazone maleate (CAS No. 122324- 73-4 or 155141-29-0) (GlaxoSmithKline), rosiglitazone maleate/glimepir (CAS No. 155141-29-0 and 93479-97-1), AVANDARYL, rosiglitazone maleate/metformin extend (CAS No. 155141-29-0 and 657-24-9), AVANDAMET, rosiglitazone maieate+metformin, AVANDAMET (GlaxoSmithKline), tesaglitazar (AstraZeneca), and an analogue or derivative thereof.
78. The device of item 1 wherein the agent is a phosphatase inhibitor.
79. The device of item 1 wherein the agent is a phosphodiesterase (PDE) inhibitor selected from the group consisting of avanafil (Tanabe Seiyaku), dasantafil (CAS No. 569351-91-3) (Schering- Plough), A-906119 (CAS No. 134072-58-5), DL-850 (Sanofi-Aventis), GRC- 3015, GRC-3566, GRC-3886 (Glenmark), HWA-153 (CAS No. 56395-66-5) (Sanofi-Aventis), hydroxypumafentrine (Altana), IBFB-130011 , IBFB-14-016, IBFB-140301, IBFB-150007, IBFB-211913 (IBFB Pharma), L-826141 (Merck & Co), medorinone (CAS No. 88296-61-1) (Sanofi-Aventis), MEM- 1917 (Memory Pharmaceuticals), ND-1251 (Neuro3d), PDE inhibitors from ICOS, PDE IV inhibitors from Memory Pharmaceuticals and CrystalGenomics, Pharmaprojects No. 2742 and 6141 (Sanofi-Aventis), QAD-171 (Novartis), RHC-2963 (CAS No. 76993-12-9 and 76993-14-1), RPR-117658, RPR-122818 derivatives, SR-24870 , and RPR-132294 (Sanofi-Aventis), SK-350 (ln2Gen), stroke therapy agents from deCODE Genetics, TAS-203 (Taiho), tofimilast (CAS No. 185954-27-2) (Pfizer), UK- 371800 (Pfizer), WIN-65579 (CAS No. 158020-82-7) (Sanofi-Aventis), IBFB- 130020 (IBFB Pharma), OPC-6535 (CAS No. 145739-56-6) (Otsuka), a phosphodiesterase III inhibitor, enoximone, a phosphodiesterase IV inhibitor, fosfosal, Atopik (Barrier Therapeutics), triflusal, a phosphodiesterase V inhibitor, and an analogue or derivative thereof.
80. The device of item 1 wherein the agent is a PKC inhibitor. 81. The device of item 1 wherein the agent is a platelet activating factor antagonist.
82. The device of item 1 wherein the agent is a platelet- derived growth factor receptor kinase inhibitor.
83. The device of item 1 wherein the agent is a prolyl hydroxylase inhibitor.
84. The device of item 1 wherein the agent is a polymorphonuclear neutrophil inhibitor.
85. The device of item 1 wherein the agent is a protein kinase B inhibitor.
86. The device of item 1 wherein the agent is a protein kinase C stimulant.
87. The device of item 1 wherein the agent is a purine nucleoside analogue.
88. The device of item 1 wherein the agent is a purinoreceptor P2X antagonist.
89. The device of item 1 wherein the agent is a Raf kinase inhibitor.
90. The device of item 1 wherein the agent is a reversible inhibitor of ErbB1 and ErbB2. 91. The device of item 1 wherein the agent is a ribonucleoside triphosphate reductase inhibitor.
92. The device of item 1 wherein the agent is an SDF-1 antagonist.
93. The device of item 1 wherein the agent is a sheddase inhibitor.
94. The device of item 1 wherein the agent is an SRC inhibitor.
95. The device of item 1 wherein the agent is a stromelysin inhibitor.
96. The device of item 1 wherein the agent is an Syk kinase inhibitor.
97. The device of item 1 wherein the agent is a telomerase inhibitor.
98. The device of item 1 wherein the agent is a TGF beta inhibitor selected from the group consisting of pirfenidone (CAS No. 53179- 13-8) (MARNAC), tranϋast (CAS No. 53902-12-8) (Kissei), IN-1130 (ln2Gen), mannose-6-phosphate (BTG), TGF-β antagonists from Inflazyme (Pharmaprojects No. 6075), TGF-β antagonists from Sydney, non-industrial source), TGF-β I receptor kinase inhibitors from EIi Lilly, TGF-β receptor inhibitors from Johnson & Johnson, and an analogue or derivative thereof.
99. The device of item 1 wherein the agent is a TNFα antagonist or TACE inhibitor selected from the group consisting of adalimumab (CAS No. 331731-18-1) (Cambridge Antibody Technology), AGIX-4207 (AtheroGenics), AGT-1 (Advanced Biotherapy), an antiinflammatory from Borean Pharma, Cellzome, or Paradigm Therapeutics, anti-inflammatory vaccine (TNF-alpha kinoid) from Neovacs, humanized anti-TNF antibody or an anti-TNF MAb (CB0006) Celltech (UCB), apratastat (CAS No. 287445-51-0) (Wyeth), BMS-561392 (Bristol-Myers Squibb), BN- 006 (Bone), certolizumab pegol (CAS No. 428863-50-7 or CH-138 (UCB), cilomilast (CAS No. 153259-65-5) (GlaxoSmithKline), CR-1 (Nuada Pharmaceuticals), CRx-119 (CombinatoRx), D-5410 (UCB), dacopafant (CAS No. 125372-33-0) (Sanofi-Aventis), dersalazine (CAS No. 188913-57- 7/188913-58-8) (Uriach), etanercept (CAS No. 185243-69-0) (Amgen), ethyl pyruvate (Critical (Critical Therapeutics), golimumab (CAS No. 476181-74-5) (Johnson & Johnson), hormono-immunotherapy from Ipsen, CDP571 (e.g., Humicade from UCB), IC-485 (ICOS), infliximab (CAS No. 170277-31-3) (Johnson & Johnson), IP-751 (Manhattan Pharmaceuticals), ISIS-104838 (CAS No. 250755-32-9) (ISIS Pharmaceuticals), lenalidomide. (CAS No. 191732-72-6) (Celgene), lentinan (CAS No. 37339-90-5) (Ajinomoto), MDL- 201112 (CAS No. 142130-73-2) (Sanofi-Aventis), medroxyprogesterone (CAS No. 520-85-4) (InKine Pharmaceutical), N-acetylcysteine (CAS No. 616-91-1) (Zambon), NBE-P2 (DIREVO Biotech), nerelimomab (CAS No. 162774-06-3) (Chiron), OM-294DP (OM PHARMA), onercept (CAS No. 199685-57-9) (Yeda), PASSTNF-alpha (Verigen), pentoxifylline or oxypentifylline (Sanofi-Aventis), Pharmaprojects No. 4091, 4241, 4295, or 4488 (Sanofi-Aventis), Pharmaprojects No. 5480 (Amgen), Pharmaprojects No. 6749 (Cengent), pirfenidone (CAS No. 53179-13-8) (MARNAC), RPR- 132294 (Sanofi-Aventis), S5 (F002) (Fulcrum Pharmaceuticals), simvastatin (CAS No. 79902-63-9) (Merck & Co), STA-6292 (Synta Pharmaceuticals), tacrolimus (CAS No. 104987-11-3) (Fujisawa LifeCycle Pharma), talactoferrin alfa (CAS No. 308240-58-6) (Agennix), thalidomide (CAS No. 50-35-1) (Celgene), TNF antagonists form ProStrakan, and Synergen, TNF inhibitors (Amgen), TNF-alpha antagonists from Dynavax Technologies and Jerina AG (Germany), TNF-alpha inhibitors from IBFB Pharma and Xencor (Xencor), torbafylline (CAS No. 105102-21-4) (Sanofi-Aventis), UR-1505 (Uriach), VT-346 (Viron Therapeutics), YSIL6 (Y's Therapeutics), YSTH2 (Y's Therapeutics), NPI-1302a-3 (Nereus Pharmaceuticals, a TNF antagonist from Jerina AG (Germany), dersalazine, and an analogue or derivative thereof.
100. The device of item 1 wherein the agent is a Toll receptor inhibitor.
101. The device of item 1 wherein the agent is a tubulin antagonist.
102. The device of item 1 wherein the agent is a tyrosine kinase inhibitor selected from the group consisting of SU-011248, SUTENT from Pfizer Inc. (New York, NY)1 BMS-354825, PN-355 (Paracelsian Pharmaceuticals), AGN-199659 (Allergan), AAL-993 or ABP-309 (Novartis), adaphostin (NIH), AEE-788 (Novartis), AG-013736 (OSI Pharmaceuticals), AG-13736 (Pfizer), ALT-110 (Alteris Therapeutics), AMG-706 (Amgen), anticancer MAbs from Xencor, anti-EGFrvlll MAbs from Abgenix, anti-HER2 MAb from Abiogen, AZD-2171 or AZD-9935 (AstraZeneca), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), CEP-5214 (Cephalon), CEP- 7055 (Cephalon), cetuximab (ImClone Systems), CHIR-200131 and CHIR- 258 (Chiron), CP-547632 (OSI Pharmaceuticals), CP-724714 (Pfizer), CT- 301 (Creabilis Therapeutics), D-69491 (Baxter International), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EGFR/ErbB2 inhibitors from Array BioPharma, erlotinib (CAS No. 183319-69-9) (OSI Pharmaceuticals), EXEL-2880 (Exelixis), FK-778 (Sanofi-Aventis) , gefitinib (CAS No. 184475-35-2) (AstraZeneca), GW-2286 or GW-654652 (GlaxoSmithKline), her2/neu antigen from AlphaVax, HER-2/neu inhibitor from Generex, Herzyme (Medipad) (Sirna Therapeutics), HKI-272 (Wyeth), HuMax-EGFr (Genmab), 105. The device of item 1 wherein the agent is ZD-6474 (an angiogenesis inhibitor).
106. The device of item 1 wherein the agent is AP-23573 (an mTOR inhibitor).
107. The device of item 1 wherein the agent is synthadotin (a tubulin antagonist).
108. The device of item 1 wherein the agent is S-0885 (a collagenase inhibitor).
109. The device of item 1 wherein the agent is aplidine (an elongation factor-1 alpha inhibitor).
110. The device of item 1 wherein the agent is ixabepilone (an epithilone).
111. The device of item 1 wherein the agent is IDN-5390 (an angiogenesis inhibitor).
112. The device of item 1 wherein the agent is SB-2723005 (an angiogenesis inhibitor).
113. The device of item 1 wherein the agent is ABT-518 (an angiogenesis inhibitor).
114. The device of item 1 wherein the agent is combretastatin (an angiogenesis inhibitor).
799 idronoxil (CAS No. 81267-65-4) (Novogen), IGF-1 inhibitors from Ontogen, IMC-11 F8 (ImCIone Systems), kahalalide F (CAS No. 149204-42-2) (PharmaMar), KDR inhibitor from LG Life Sciences, KDR inhibitors from Abbott Laboratories, KDR kinase inhibitors (UCB), Kdr kinase inhibitors from Merck & Co, KRN-633 and KRN-951 (Kirin Brewery), KSB-102 (Xenova), lapatinib ditosylate (CAS No. 388082-78-8) (GlaxoSmithKline), matuzumab (Merck KGaA), MDX-214 (Medarex), ME-103 (Pharmexa), MED-A300 (Gerolymatos), MNAC-13 (Lay Line Genomics), nimotuzumab (Center of Molecular Immunology), NSC-330507 or NSC-707545 (NIH), NV-50 (Novogen), OSI-930 (OSI Pharmaceuticals), panitumumab (Abgenix), pelitinib (CAS No. 287933-82-7) (Wyeth), pertuzumab (CAS No. 380610-27- 5) (Genentech), Pharmaprojects No. 3985 (Sanofi-Aventis), prostate cancer therapeutics from Sequenom (SQPC35, SQPC36, SQPC90), removab and remoxab (Trion Pharma), RG-13022 (CAS No. 136831-48-6), RG-13291 (CAS No. 138989-50-1), or RG-14620 (CAS No. 136831-49-7) (Sanofi- Aventis), RM-6427 (Romark), RNAi breast cancer therapy from Benitec, RP 53801 (CAS No. 125882-88-4) (Sanofi-Aventis), sorafenib tosylate (Bayer), SU-11657 (Pfizer), Tie-2 antagonists from Semaia (Hybrigenics), Tie-2 inhibitors from Ontogen, trastuzumab (CAS No. 180288-69-1) (Genentech), tyrosine kinase inhibitors from Sanofi-Aventis, U3-1287, U3-1565, U3-1784, U3-1800 (U3 Pharma), vatalanib (Novartis), VEGFR-2 kinase inhibitor from Bristol-Myers Squibb, XL-647 (Exelixis), ZD-6474 (AstraZeneca), ZK-CDK (Schering AG), an EGFR tyrosine kinase inhibitor, EKB-569 (Wyeth), and an analogue or derivative thereof.
103. The device of item 1 wherein the agent is a VEGF inhibitor.
104. The device of item 1 wherein the agent is a vitamin D receptor agonist.
798 115. The device of item 1 wherein the agent is anecortave acetate (an angiogenesis inhibitor).
116. The device of item 1 wherein the agent is SB-715992 (a kinesin antagonist).
117. The device of item 1 wherein the agent is temsirolimus (an mTOR inhibitor).
118. The device of item 1 wherein the agent is adalimumab (a TNFα antagonist).
119. The device of item 1 , further comprising a polymer.
120. The device of item 1 wherein the anti-scarring agent inhibits adhesion between the device and a host into which the device is implanted.
121. The device of item 1 wherein the device delivers the anti-scarring agent locally to tissue proximate to the device.
122. The device of item 1, further comprising a coating, wherein the coating comprises the anti-scarring agent.
123. The device of item 1 , further comprising a coating, wherein the coating is disposed on a surface of the device.
124. The device of item 1 , further comprising a coating, wherein the coating directly contacts the device. 125. The device of item 1 , further comprising a coating, wherein the coating indirectly contacts the device.
126. The device of item 1 , further comprising a coating, wherein the coating partially covers the device.
127. The device of item 1 , further comprising a coating, wherein the coating completely covers the device.
128. The device of item 1, further comprising a coating, wherein the coating is a uniform coating.
129. The device of item 1 , further comprising a coating, wherein the coating is a non-uniform coating.
130. The device of item 1 , further comprising a coating, wherein the coating is a discontinuous coating.
131. The device of item 1 , further comprising a coating, wherein the coating is a patterned coating.
132. The device of item 1, further comprising a coating, wherein the coating has a thickness of 100 Dm or less.
133. The device of item 1, further comprising a coating, wherein the coating has a thickness of 10 Dm or less.
134. The device of item 1, further comprising a coating, wherein the coating adheres to the surface of the device upon deployment of the device. 135. The device of item 1 , further comprising a coating, wherein the coating is stable at room temperature for a period of 1 year.
136. The device of item 1 , further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 0.0001% to about 1% by weight.
137. The device of item 1 , further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 1% to about 10% by weight.
138. The device of item 1 , further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 10% to about 25% by weight.
139. The device of item 1 , further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 25% to about 70% by weight.
140. The device of item 1 , further comprising a coating, wherein the coating further comprises a polymer.
141. The device of item 1 , further comprising a first coating having a first composition and the second coating having a second composition.
142. The device of item 1 , further comprising a first coating having a first composition and the second coating having a second composition, wherein the first composition and the second composition are different. 143. The device of item 1, further comprising a polymeric carrier, wherein the polymeric carrier comprises a copolymer.
144. The device of item 1 , further comprising a polymeric carrier, wherein the polymeric carrier comprises a block copolymer.
145. The device of item 1 , further comprising a polymeric carrier, wherein the polymeric carrier comprises a random copolymer.
146. The device of item 1, further comprising a polymeric carrier, wherein the polymeric carrier comprises a biodegradable polymer.
147. The device of item 1, further comprising a polymeric carrier, wherein the polymeric carrier comprises a non-biodegradable polymer.
148. The device of item 1, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrophilic polymer.
149. The device of item 1 , further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrophobic polymer.
150. The device of item 1 , further comprising a polymeric carrier, wherein the polymeric carrier comprises a polymer having hydrophilic domains.
151. The device of item 1 , further comprising a polymeric carrier, wherein the polymeric carrier comprises a polymer having hydrophobic domains. 152. The device of item 1, further comprising a polymeric carrier, wherein the polymeric carrier comprises a non-conductive polymer.
153. The device of item 1, further comprising a polymeric carrier, wherein the polymeric carrier comprises an elastomer.
154. The device of item 1, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrogel.
155. The device of item 1 , further comprising a polymeric carrier, wherein the polymeric carrier comprises a silicone polymer.
156. The device of item 1 , further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrocarbon polymer.
157. The device of item 1 , further comprising a polymeric carrier, wherein the polymeric carrier comprises a styrene-derived polymer.
158. The device of item 1 , further comprising a polymeric carrier, wherein the polymeric carrier comprises a butadiene polymer.
159. The device of item 1, further comprising a polymeric carrier, wherein the polymeric carrier comprises a macromer.
160. The device of item 1 , further comprising a polymeric carrier, wherein the polymeric carrier comprises a poly(ethylene glycol) polymer.
161. The device of item 1 , further comprising a polymeric carrier, wherein the polymeric carrier comprises an amorphous polymer. 162. The device of item 1, further comprising a lubricious coating.
163. The device of item 1 wherein the anti-scarring agent is located within pores or holes of the device.
164. The device of item 1 wherein the anti-scarring agent is located within a channel, lumen, or divet of the device.
165. The device of item 1, further comprising a second pharmaceutically active agent.
166. The device of item 1 , further comprising an antiinflammatory agent.
-167. The device of item 1 , further comprising an agent that inhibits infection.
168. The device of item 1 , further comprising an agent that inhibits infection, wherein the agent is an anthracycline.
169. The device of item 1 , further comprising an agent that inhibits infection, wherein the agent is doxorubicin.
170. The device of item 1 , further comprising an agent that inhibits infection, wherein the agent is mitoxantrone.
171. The device of item 1 , further comprising an agent that inhibits infection, wherein the agent is a fluoropyrimidine. 172. The device of item 1, further comprising an agent that inhibits infection, wherein the agent is 5-fluorouracil (5-FU).
173. The device of item 1 , further comprising an agent that inhibits infection, wherein the agent is a folic acid antagonist.
174. The device of item 1, further comprising an agent that inhibits infection, wherein the agent is methotrexate.
175. The device of item 1 , further comprising an agent that inhibits infection, wherein the agent is a podophylotoxin.
176. The device of item 1 , further comprising an agent that inhibits infection, wherein the agent is etoposide.
177. The device of item 1 , further comprising an agent that inhibits infection, wherein the agent is a camptothecin.
178. The device of item 1, further comprising an agent that inhibits infection, wherein the agent is a hydroxyurea.
179. The device of item 1 , further comprising an agent that inhibits infection, wherein the agent is a platinum complex.
180. The device of item 1 , further comprising an agent that inhibits infection, wherein the agent is cisplatin.
181. The device of item 1 , further comprising an antithrombotic agent. 182. The device of item 1 , further comprising a visualization agent.
183. The device of item 1 , further comprising a visualization agent, wherein the visualization agent is a radiopaque material, wherein the radiopaque material comprises a metal, a halogenated compound, or a barium containing compound.
184. The device of item 1 , further comprising a visualization agent, wherein the visualization agent is a radiopaque material, wherein the radiopaque material comprises barium, tantalum, or technetium.
185. The device of item 1 , further comprising a visualization agent, wherein the visualization agent is a MRI responsive material.
186. The device of item 1 , further comprising a visualization agent, wherein the visualization agent comprises a gadolinium chelate.
187. The device of item 1 , further comprising a visualization agent, wherein the visualization agent comprises iron, magnesium, manganese, copper, or chromium.
188. The device of item 1, further comprising a visualization agent, wherein the visualization agent comprises an iron oxide compound.
189. The device of item 1 , further comprising a visualization agent, wherein the visualization agent comprises a dye, pigment, or colorant.
190. The device of item 1 , further comprising an echogenic material. 191. The device of item 1 , further comprising an echogenic material, wherein the echogenic material is in the form of a coating.
192. The device of item 1 wherein the device is sterile.
193. The device of item 1 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device.
194. The device of item 1 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is connective tissue.
195. The device of item 1 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is muscle tissue.
196. The device of item 1 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is nerve tissue.
197. The device of item 1 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is epithelium tissue.
198. The device of item 1 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from the time of deployment of the device to about 1 year. 199. The device of item 1 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from about 1 month to 6 months.
200. The device of item 1 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from about 1 - 90 days.
201. The device of item 1 wherein the anti-scarring agent is released in effective concentrations from the device at a constant rate.
202. The device of item 1 wherein the anti-scarring agent is released in effective concentrations from the device at an increasing rate.
203. The device of item 1 wherein the anti-scarring agent is released ineffective concentrations from the device at a decreasing rate.
204. The device of item 1 wherein the anti-scarring agent is released in effective concentrations from the composition comprising the anti-scarring agent by diffusion over a period ranging from the time of deployment of the device to about 90 days.
205. The device of item 1 wherein the anti-scarring agent is released in effective concentrations from the composition comprising the anti-scarring agent by erosion of the composition over a period ranging from the time of deployment of the device to about 90 days.
206. The device of item 1 wherein the device comprises about 0.01 Dg to about 10 Dg of the anti-scarring agent. 207. The device of item 1 wherein the device comprises about 10 Dg to about 10 mg of the anti-scarring agent.
208. The device of item 1 wherein the device comprises about 10 mg to about 250 mg of the anti-scarring agent.
209. The device of item 1 wherein the device comprises about 250 mg to about 1000 mg of the anti-scarring agent.
210. The device of item 1 wherein the device comprises about 1000 mg to about 2500 mg of the anti-scarring agent.
211. The device of item 1 wherein a surface of the device comprises less than 0.01 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
212. The device of item 1 wherein a surface of the device comprises about 0.01 Dg to about 1 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
213. The device of item 1 wherein a surface of the device comprises about 1 Dg to about 10 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
214. The device of item 1 wherein a surface of the device comprises about 10 Dg to about 250 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
215. The device of item 1 wherein a surface of the device comprises about 250 Dg to about 1000 Dg of the anti-scarring agent of anti- scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
216. The device of item 1 wherein a surface of the device comprises about 1000 Dg to about 2500 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
217. The device of any one of items 1-216, wherein the device is an intravascular device.
218. The device of any one of items 1-216, wherein the device is a gastrointestinal stent.
219. The device of any one of items 1-216, wherein the device is a tracheal and bronchial stent.
220. The device of any one of items 1-216, wherein the device is a genital urinary stent.
221. The device of any one of items 1-216, wherein the device is an ear and nose stent.
222. The device of any one of items 1-216, wherein the device is an ear ventilation device.
223. The device of any one of items 1-216, wherein the device is an intraocular implant.
224. The device of any one of items 1-216, wherein the device is a vascular graft. 225. The device of any one of items 1-216, wherein the device comprises a film or a mesh.
226. The device of any one of items 1-216, wherein the device is a glaucoma drainage device.
227. The device of any one of items 1-216, wherein the device is a prosthetic heart valve or a component thereof.
228. The device of any one of items 1-216, wherein the device is a penile implant.
229. The device of any one of items 1-216, wherein the device is an endotracheal or tracheostomy tube.
230. The device of any one of items 1-216fwherein the device is a peritoneal dialysis catheter.
231. The device of any one of items 1-216, wherein the device is a central nervous system shunt or a pressure monitoring device.
232. The device of any one of items 1-216, wherein the device is an inferior vena cava filter.
233. The device of any one of items 1-216, wherein the device is a gastrointestinal device.
234. The device of any one of items 1-216, wherein the device is a central venous catheter. 235. The device of any one of items 1-216, wherein the device is a ventricular assist device.
236. The device of any one of items 1-216, wherein the device is a spinal implant.
237. The device of any one of items 1-216, wherein the device is an implantable electrical device.
238. The device of any one of items 1-216, wherein the device is an implantable sensor.
239. The device of any one of items 1-216, wherein the device is a soft tissue implant.
240. The device of any-one of items 1-216, wherein the device is a soft tissue implant.
241. A device, comprising an intravascular device (e.g., an implant) and an anti-scarring agent or a composition comprising an anti- scarring agent, wherein the agent inhibits scarring between the device and a host into which the device is implanted.
242. The device of item 241 wherein the agent is an adensosine A2A receptor antagonist.
243. The device of item 241 wherein the agent is an AKT inhibitor. 244. The device of item 241 wherein the agent is an alpha 2 integrin antagonist, wherein the alpha 2 integrin antagonist is Pharmaprojects No. 5754 (Merck KgaA).
245. The device of item 241 wherein the agent an alpha 4 integrin antagonist.
246. The device of item 241 wherein the agent is an alpha 7 nicotinic receptor agonist.
247. The device of item 241 wherein the agent is an angiogenesis inhibitor selected from the group consisting of AG-12,958 (Pfizer), ATN-161 (Attention LLC), neovastat, an angiogenesis inhibitor from Jerina AG (Germany), NM-3 (Mercian), VGA-1155 (Taisho), FCE-26644 (Pfizer), FCE-26950 (Pfizer), FPMA (Meiji Daries), FR-111142 (Fujisawa), GGTI-298, GM-1306 (Ligand), GPA-1734 (Novartis), NNC-47-0011 (Novo Nordisk), herbamycin (Nippon Kayaku), lenalidomide (Celegene), IP-10 (NIH), ABT-828 (Abbott), KIN-841 (Tokushima University, Japan), SF-1126 (Semafore Pharmaceuticals), laminin technology (NIH), CHIR-258 (Chiron), NVP-AEW541 (Novartis), NVP-AEW541 (Novartis), Vt16907 (Alchemia), OXI-8007 (Oxigene), EG-3306 (Ark Therapeutics), Maspin (Arriva), ABT- 567 (Abbott), PPI-2458 (Praecis Pharmaceuticals), CC-5079, CC-4089 (Celgene), HIF-1 alpha inhibitors (Xenova), S-247 (Pfizer), AP-23573 (Ariad), AZD-9935 (Astra Zeneca), mebendazole (Introgen Therapeutics), MetAP-2 inhibitors (GlaxoSmithKline), AG-615 (Angiogene Pharmaceuticals), Tie-2 antagonists (Hybrigenics), NC-381, CYC-381, NC-169, NC-219, NC-383, NC-384, NC-407 (Lorus Therapeutics), ATN-224 (Attenuon), ON-01370 (Onconova), Vitronectin antagonists (Amgen), SDX-103 (Salmedix), Vitronectin antagonists (Shire), CHP (Riemser), TEK (Amgen), Anecortave acetate (Alcon), T46.2 (Matrix Therapeutics), HG-2 (Heptagen), TEM antagonists (Genzyme), Oxi-4500 (Oxigene), ATN-161 (Attenuon), WX-293 (Wilex), M-2025 (Metris Therapeutics), Alphastatin (BioActa), YH-16 (Yantai Rongchang), BIBF-1120 (Boehringer Ingelheim), BAY-57-9352 (Bayer), AS- 1404 (Cancer Research Technology), SC-77964 (Pfizer), glycomimetics (BioTie Therapies), TIE-2 Inhibitors (Ontogen), DIMI, Octamer (Octamer), ABR-215050 (Active Biotech), ABT-518 (Abbott), KDR inhibitors (Abbott), BSF-466895 (Abbott), SCH-221153 (Schering-Plough), DAC:antiangiogenic (ConjuChem), TFPI (EntreMed), AZD-2171 (Astra-Zenaca), CDC-394 (Celgene), LY290293 (EIi Lilly), IDN-5390 (Indena), Kdr Kinase Inhibitors (Merck), CT-113020, CT-116433, CT-116563, CT-31890, CT-32228) (Cell Therapeutics), A-299620 (Abbott), TWEAK Inhibitor (Amgen), VEGF modulators (Johnson and Johnson), Tum-N53, tumstatin (Genzyme), Thios- 1 , Thios-2 (Thios Pharmaceuticals), MV-6401 (Miravant Medical Technologies), Spisulosine (PharmaMar), CEP-7055 (Cephalon), AUV-201 (Auvation), LM-609 (EIi Lilly), SKF-106615 (AnorMED), Oglufanide disodium (Cytran), BW-114 (Pharminox), Calreticulin (NIH)1 WX-678 (Wilex), SD-7784 (Pfizer), WX-UK1 (Wilex), SH-268 (Schering.AG), 2-Me-PGA (Celgene), S- 137 (Pfizer), ZD-6126 (Angiogene Pharmaceuticals), SG-292 (SignalGen), Benefin (Lane Labs), A6, A36 (Angstrom), SB-2723005 (GlaxoSmithKline), SC-7 (Cell Therapeutics), ZEN-014 (AEterna Zentaris), 2-methoxyestradiol (EntreMed), NK-130119 (Nippon Kayaku), CC-10004 (Celgene), AVE- 8062A (Ajinomoto), Tacedinaline (Pfizer), Actinonin (Tokyo Metropolitan Institute of Medical Science), Lenalidomide (Celgene), VGA-1155, BTO-956 (SRI International), ER-68203-00 (Eisai), CT-6685 (UCB), JKC-362 (Phoenix Pharmaceuticals), DMI-3798 (DMI Biosciences, Angiomate (Ipsen), ZD-6474 (AstraZeneca), CEP-5214 (Cephalon), Canstatin (Genzyme), NM-3 (Mercian), Oridigm (MediQuest Therapeutics), Exherin (Adherex), BLS-0597 (Boston Life Sciences), PTC-299 (PTC Therapeutics), NPI-2358 (Nereus Pharmaceuticals), CGP-79787 (Novartis), AEE-788 (Novartis), CKD-732 (Chong Kun Dang), CP-564959 (OSI Pharmaceuticals), CM-101 (CarboMed), CT-2584, CT3501 (Cell Therapeutics), combretastatin and analogues and derivatives thereof (Oxigene), Rebimastat (Bristol-Meyers Squibb), Dextrin 2-sulfate (ML Laboratories), Cilengitide (Merk KGaA), NSC-706704 (Pharminox), KRN- 951 (Kirin Brewery), Ukrain, NSC-631570 (Nowicky Pharma), Tecogalan sodium (Daiichi Pharmaceutical), Tz-93 (Tsumura), TBC-1635 (Encysive Pharmaceuticals), TAN-1120 (Takeda), Semaxanib (Pfizer), BDI-7800 (Biopharmacopae), SD-186, SD-983 (Bristol-Meyers Squibb), SB-223245 (GlaxoSmithKline), SC-236 (Pfizer), RWJ-590973 (Johnson and Johnson), ILX-1850 (Genzyme), SC-68488, S-836 (Pfizer), CG-55069-11 (CuraGen), Ki-23057 (Kirin Brewery), CCX-700 (Chemoentryx), Pegaptanib octasodium (Giled Sciences), ANGIOCOL (available from Biostratum Inc.), or an analogue or derivative thereof.
248. The device of item 241 wherein the agent is an apoptosis antagonist.
249. The device of item 241 wherein the agent is an apoptosis activator.
250. The device of item 241 wherein the agent is a beta 1 integrin antagonist.
251. The device of item 241 wherein the agent is a beta tubulin inhibitor.
252. The device of item 241 wherein the agent is a blocker of enzyme production in Hepatitis C.
253. The device of item 241 wherein the agent is a Bruton's tyrosine kinase inhibitor. 254. The device of item 241 wherein the agent is a calcineurin inhibitor.
255. The device of item 241 wherein the agent is a caspase 3 inhibitor.
256. The device of item 241 wherein the agent is a CC chemokine receptor antagonist.
257. The device of item 241 wherein the agent is a cell cycle inhibitor selected from the group consisting of SNS-595 (Sunesis), synthadotin, KRX-0403, and an analogue or derivative thereof.
258. The device of item 241 wherein the agent is a cathepsin B inhibitor.
259. The device of item 241 wherein the agent is a cathepsin K inhibitor, wherein the cathepsin K inhibitor is 462795 (GlaxoSmithKline), INPL-022-D6 (Amura Therapeutics), an analogue or derivative thereof).
260. The device of item 241 wherein the agent is a cathepsin L inhibitor.
261. The device of item 241 wherein the agent is a CD40 antagonist.
262. The device of item 241 wherein the agent is a chemokine receptor agonist.
263. The device of item 241 wherein the agent is a chymase inhibitor. 264. The device of item 241 wherein the agent is a collagenase antagonist.
265. The device of item 241 wherein the agent is a CXCR antagonist.
266. The device of item 241 wherein the agent is a cyclin dependent kinase inhibitor selected from the group consisting of a CDK-1 inhibitor, a CDK-2 inhibitor, a CDK- 4 inhibitor, a CDK-6 inhibitor, a CAK1 inhibitor from GPC Biotech and Bristol-Myers Squibb, RGB-286199 (GPC Biotech), an anticancer agent from Astex Technology, a CAK1 inhibitor from GPC Biotech, a CDK inhibitor from Sanofi-Aventis, a CDK1/CDK2 inhibitor from Amgen, a CDK2 inhibitor from SUGEN-2 (Pfizer), a hearing loss therapy agent (Sound Pharmaceuticals), PD-0332991 (Pfizer), RGB-286199 (GPC Biotech), Ro-0505124 (Hoffmann-La Roche), a Ser/Thr kinase inhibitor from Lilly (EIi Lilly), CVT-2584 (CAS No. 199986-75-9) (CV Therapeutics), and an analogue or derivative thereof.
267. The device of item 241 wherein the agent is a cyclooxygenase 1 inhibitor.
268. The device of item 241 wherein the agent is a DHFR inhibitor.
269. The device of item 241 wherein the agent is a dual integrin inhibitor.
270. The device of item 241 wherein the agent is an elastase inhibitor. 271. The device of item 241 wherein the agent is an elongation factor-1 alpha inhibitor.
272. The device of item 241 wherein the agent is an endothelial growth factor antagonist.
273. The device of item 241 wherein the agent is an endothelial growth factor receptor kinase inhibitor selected from the group consisting of sorafenib tosylate (Bayer), AAL-993 (Novartis), ABP-309 (Novartis), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), E- 7080 (Eisai), EG-3306 (Ark Therapeutics), EXEL-2880 (Exelixis), GW- 654652 (GlaxoSmithKline), a KDR inhibitor from LG Life Sciences, CT-6685 and CT-6729 (UCB), KRN-633 and KRN-951 (Kirin Brewery), OSI-930 (OSI Pharmaceuticals), SP-5.2 (Supratek Pharma), SU-11657 (Pfizer), a Tie-2 antagonist (Hybrigenics), a VEGFR-2 kinase inhibitor (Bristol-Myers Squibb), XL^647 (Exelixis), a KDR inhibitor from Abbott Laboratories, sorafenib tosylate, and an analogue or derivative thereof.
274. The device of item 241 wherein the agent is an endotoxin antagonist.
275. The device of item 241 wherein the agent is an epothilone and tubulin binder.
276. The device of item 241 wherein the agent is an estrogen receptor antagonist.
277. The device of item 241 wherein the agent is an FGF inhibitor. 278. The device of item 241 wherein the agent is a farnexyl transferase inhibitor.
279. The device of item 241 wherein the agent is famesyltransferase inhibitor selected from the group of A-197574 (Abbott), a farnesyltransferase inhibitor from Servier, FPTIII (Strathclyde Institute for Drug R), LB-42908 (LG Life Sciences), Pharmaprojects No. 5063 (Genzyme), Pharmaprojects No. 5597 (Ipsen), Yissum Project No. B-1055 (Yissum), and an analogue or derivative thereof.
280. The device of item 241 wherein the agent is an FLT-3 kinase inhibitor.
281. The device of item 241 wherein the agent is an FGF receptor kinase inhibitor.
282. The device of item 241 wherein the agent is a fibrinogen antagonist selected from the group consisting of AUV-201 (Auvation), MG-13926 (Sanofi-Aventis), plasminogen activator (CAS No. 105913-11-9) (from Sanofi-Aventis or UCB), plasminogen activator-2 (tPA- 2) (Sanofi-Aventis), pro-urokinase (CAS No. 82657-92-9) (Sanofi-Aventis), and an analogue or derivative thereof.
283. The device of item 241 wherein the agent is a heat shock protein 90 antagonist selected from the group consisting of SRN-005 (Sirenade), geldanamycin, NSC-33050 (17-allylaminogeldanamycin; 17- AAG), 17-dimethylaminoethylamino-17-demethoxy-geldanamycin (17- DMAG), rifabutin (rifamycin XIV, r,4-didehydro-1-deoxy-1 ,4-dihydro-5'-(2- methylpropyl)-1-oxo-), and an analogue or derivative thereof. 284. The device of item 241 wherein the agent is a histone deacetylase inhibitor.
285. The device of item 241 wherein the agent is an HMGCoA reductase inhibitor selected from the group consisting of an atherosclerosis therapeutic from Lipid Sciences, ATI- 16000 (ARYx Therapeutics), KS-01-019 (Kos Pharmaceuticals), Pharmaprojects No. 2197 (Sanofi-Aventi), RP 61969 (Sanofi-Aventis), and an analogue or derivative thereof.
286. The device of item 241 wherein the agent is an ICAM inhibitor.
287. The device of item 241 wherein the agent is an IL, ICE and IRAK antagonist, wherein the antagonist is a CJ-14877, CP-424174 (Pfizer), NF-61 (Negma-Lerads), and an analogue or derivative thereof.
288. The device of item 241 wherein the agent is an IL-2 inhibitor.
289. The device of item 241 wherein the agent is an immunosuppressant selected from the group consisting of teriflunomide (Sanofi Aventis), chlorsulfaquinoxalone (NSC-339004), chlorsulfaquinoxalone sulfate, CS-712 (Sankyo), ismomultin alfa (CAS No. 457913-93-8) (Akzo Nobel), antiallergics from GenPat77, antiinflammatories or AT-005 (Androclus Therapeutics), autoimmune disease therapeutics from EpiVax, BN-007 (Bone), budesonide (CAS No. 51333-22- 3) (MAP Pharmaceuticals), CO-14 (Genzyme), edratide (CAS No. 433922- 67-9) (Teva), EP-314 (Enanta), eprovafen (CAS No. 101335-99-3) (Sanofi- Aventis), HWA-131 (CAS No. 118788-41-3) (Sanofi-Aventis), immunomodulators from MerLion Pharmaceuticals, immunosuppressives from Alchemia, IPL-12 (Inflazyme), MDL-9563 (CAS No. 27086-86-8) (Sanofi-Aventis), Pharmaprojects No. 2330 (Sanofi-Aventis), Pharmaprojects No. 6426 (Abgenix), PXS-25 (Pharmaxis), rosmarinic acid (CAS No. 20283-92-5) (Sanofi-Aventis), RP 42927 or RP 54745 (CAS No. 135330-08-4) (Sanofi-Aventis), SGN-35 (Seattle Genetics), ST-1959 (Sigma-Tau), type I diabetes therapy from SYNX Pharma, UNIL-88 (Debiopharm), VP-025 (Vasogen), VR-694 (Vectura), PRTX-001 (Protalex), and an analogue or derivative thereof.
290. The device of item 241 wherein the agent is an IMPDH (inosine monophosphate).
291. The device of item 241 wherein the agent is an integrin antagonist.
292. The device of item 241 wherein the agent is an interleukin antagonist.
293. The device of item 241 wherein the agent is an inhibitor of type III receptor tyrosine kinase.
294. The device of item 241 wherein the agent is an irreversible inhibitor of enzyme methionine aminopeptidase type 2.
295. The device of item 241 wherein the agent is an isozyme selective delta protein kinase C inhibitor.
296. The device of item 241 wherein the agent a JAK3 enzyme inhibitor. 297. The device of item 241 wherein the agent is a JNK inhibitor.
298. The device of item 241 wherein the agent is a kinase inhibitor.
299. The device of item 241 wherein the agent is kinesin antagonist.
300. The device of item 241 wherein the agent is a kinesin antagonist.
301. The device of item 241 wherein the agent is a leukotriene inhibitor and antagonist selected from the group consisting of ambicromil (CAS No. 58805-38-2) (Sanofi-Aventis), amelubant (CAS No. 346735-24-8) (Boehringer Ingelheim), DV\M 141 (Dong Wha), ebselen (Daiichi Pharmaceutical), ibudilast (Kyorin), leucotriene inhibitors from Sanofi-Aventis, lymphotoxin-beta receptor (LT-D) from Biogen Idee, Pharmaprojects No. 1535 and 2728 (CAS No. 119340-33-9) (Sanofi- Aventis), R-112 (Rigel), Rev-5367 (CAS No. 92532-05-3) (Sanofi-Aventis), RG-14893 (CAS No. 141835-49-6) (Sanofi-Aventis), RG-5901-A (CAS No. 101910-24-1), 92532-23-5, RP 66153 (CAS No. 142422-79-5), RP 66364 (CAS No.186916-92-5), RP 69698 (CAS No. 141748-00-7) (Sanofi-Aventis), SC-411930 (Pfizer), SC-41930 (CAS No. 120072-59-5) (Pfizer), SC-50605 (CAS No. 138828-39-4) (Pfizer), SC-51146 (CAS No. 141059-52-1), SC- 53228 (CAS No. 153633-01-3) (Pfizer), spaglumic acid (ZY-15106) (CAS No.3106-85-2), 80619-64-3 (Novartis), tipredane (CAS No. 85197-77-9) (Bristol-Myers Squibb), U-75302 (CAS No. 119477-85-9) (Pfizer), and analogue or derivative thereof. 302. The device of item 241 wherein the agent is an MAP kinase inhibitor.
303. The device of item 241 wherein the agent is a matrix metalloproteinase inhibitor.
304. The device of item 241 wherein the agent is an MCP- CCR2 inhibitor.
305. The device of item 241 wherein the agent is an mTOR inhibitor.
306. The device of item 241 wherein the agent is an mTOR kinase inhibitor.
307. The device of item 241 wherein the agent is a microtubule inhibitor selected from the group consisting of antibody- maytansinoid conjugates from Biogen Idee, colchicines (MantiCore Pharmaceuticals), anticancer immunoconjugates from Johnson & Johnson, DIME from Octamer, gni-1f (GNI), huC242-DM4, huMy9-6-DM1 (ImmunoGen), IDN-5404 (Indena), IMO-098, IMOderm (Imotep), mebendazole (Introgen Therapeutics), microtubule poisons from Cambridge Enterprise, paclitaxel such as LOTAX from Aphios (CAS No. 33069-62-4), Genexol-PM from Samyang, Pharmaprojects No. 6383 (Tapestry Pharmaceuticals), RPR-112378 (Sanofi-Aventis), SGN-75 (Seattle Genetics), SPL-7435 (Starpharma), SSR-250411 (Sanofi-Aventis), trastuzumab-DM1 (Genentech), vinorelbine, and an analogue or derivative thereof.
308. The device of item 241 wherein the agent is an MIF inhibitor. 309. The device of item 241 wherein the agent is an MMP inhibitor.
310. The device of item 241 wherein the agent is a neurokinin (NK) antagonist selected from the group consisting of anthrotainin (CAS No. 148084-40-6) (Sanofi-Aventis), an IBS thereapeutic from ArQuIe, MDL-105212A (CAS No. 167261-60-1) (Ssanofi-Aventis), Pharmaprojects No. 2744, 3258 (CAS No. 139167-47-8) 4006, 4201, or 5986 (Sanofi-Aventis), RP 67580 (CAS No. 135911-02-3), SR-144190 (CAS No. 201152-86-5), SSR-240600, SSR-241586 (Sanofi-Aventis), TKA-457 (Novartis), vestipitant mesylate (CAS No. 334476-64-1) (GlaxoSmithKline), Win-64821 (Sanofi-Aventis), PRX-96026 (Predix Pharmaceuticals), and an analogue or derivative thereof.
311. The device of item 241 wherein the agent is an NF kappa B inhibitor selected from the group consisting of AVE-0545 or AVE- 0547 (Sanofi-Aventis), bortezomib (CAS No. 179324-69-7) (Millennium Pharmaceuticals), dexanabinol (CAS No. 112924-45-5) (Pharmos), dexlipotam (Viatris), Pharmaprojects No. 6283 (INDRA) (OXiGENE), IPL- 576092 (CAS No. 137571-30-3) (Inflazyme), NFKB decoy (Corgentech), NFKB decoy oligo (AnGes MG), NFKB's from Ariad, osteoporosis treatments or S5 (F005) from Fulcrum Pharmaceuticals, P61 (Phytopharm), R-flurbiprofen (CAS No. 5104-49-4) (Encore Pharmaceuticals), and an analogue or derivative thereof.
312. The device of item 241 wherein the agent is a nitric oxide agonist.
313. The device of item 241 wherein the agent is an ornithine decarboxylase inhibitor. 314. The device of item 241 wherein the agent is a p38 MAP kinase inhibitor selected from the group consisting of AZD-6703 (AstraZeneca), JX-401 (Jexys Pharmaceuticals), BMS-2 (Bristol-Myers Squibb), a p38 MAP kinase inhibitor from Novartis, a p38-alpha MAP kinase inhibitor from Amphora, Pharmaprojects No. 5704 (Pharmacopeia), RPR- 200765A (Sanofi-Aventis), SD-282 (Johnson & Johnson), TAK-715 (Takeda), and an analogue or derivative thereof.
315. The device of item 241 wherein the agent is a palmitoyl- protein thioesterase inhibitor.
316. The device of item 241 wherein the agent is a PDGF receptor kinase inhibitor selected from the group consisting of AAL-993, AMN-107, or ABP-309 (Novartis), AMG-706 (Amgen), BAY-57-9352 (Bayer), CDP-860 (UCB), E-7080 (Eisai), imatinib (CAS No. 152459-95-5) (Novartis), OSI-930 (OSI Pharmaceuticals), RPR-127963E (Sanofi-Aventis), RWJ-540973 (Johnson & Johnson), sorafenib tosylate (Bayer), SU-11657 (Pfizer), tandutinib (CAS No. 387867-13-2) (Millennium Pharmaceuticals), vatalanib (Novartis), ZK-CDK (Schering AG), and an analogue or derivative thereof.
317. The device of item 241 wherein the agent is (-)- halofenate (Metabolex), AMG-131 (Amgen), antidiabetics from Japan Tobacco, AZD-4619, AZD-8450, AZD-8677 (AstraZeneca), DRF-10945, balaglitazone (Dr Reddy's), CS-00088, CS-00098 (Chipscreen Biosciences), E-3030 (Eisai), etalocib (CAS No. 161172-51-6) (EIi Lilly), GSK-641597 (Ligand), GSK-677954 (GlaxoSmithKline), GW-409544 (Ligand), GW- 590735 (GlaxoSmithKline), K-111 (Hoffmann-La Roche), LY-518674 (EIi Lilly), LY-674 (Ligand), LY-929 (Ligand), MC-3001, MC-3002 (MaxoCore Pharmaceuticals), metformin HCI + pioglitazone (CAS No. 1115-70-4 and 112529-15-4), ACTOPLUS MET from Andrx), muraglitazar (CAS No. 331741-94-7) (Bristol-Myers Squibb), naveglitazar (Ligand), oleoylethanolamide (Kadmus Pharmaceuticals), ONO-5129, pioglitazone hydrochloride (CAS No. 111025-46-8 and 112529-15-4) (Takeda), PLX-204 (Plexxikon), PPAR agonists from Genfit, PPAR delta agonists from EIi Lilly, PPAR-alpha agonists from CrystalGenomics, PPAR-gamma modulators and PPAR-β modulators from CareX, rosiglitazone maleate (CAS No. 122324- 73-4 or 155141-29-0) (GlaxoSmithKline), rosiglitazone maleate/glimepir (CAS No. 155141-29-0 and 93479-97-1), AVANDARYL, rosiglitazone maleate/metformin extend (CAS No. 155141-29-0 and 657-24-9), AVANDAMET, rosiglitazone maleate+metformin, AVANDAMET (GlaxoSmithKline), tesaglitazar (AstraZeneca), and an analogue or derivative thereof.
318. The device of item 241 wherein the agent is a phosphatase inhibitor.
319. The device of item 241 wherein the agent is a phosphodiesterase (PDE) inhibitor selected from the group consisting of avanafil (Tanabe Seiyaku), dasantafil (CAS No. 569351-91-3) (Schering- Plough), A-906119 (CAS No. 134072-58-5), DL-850 (Sanofi-Aventis), GRC- 3015, GRC-3566, GRC-3886 (Glenmark), HWA-153 (CAS No. 56395-66-5) (Sanofi-Aventis), hydroxypumafentrine (Altana), IBFB-130011 , IBFB-14-016, IBFB-140301 , IBFB-150007, IBFB-211913 (IBFB Pharma), L-826141 (Merck & Co), medorinone (CAS No. 88296-61-1) (Sanofi-Aventis), MEM- 1917 (Memory Pharmaceuticals), ND-1251 (Neuro3d), PDE inhibitors from ICOS, PDE IV inhibitors from Memory Pharmaceuticals and CrystalGenomics, Pharmaprojects No. 2742 and 6141 (Sanofi-Aventis), QAD-171 (Novartis), RHC-2963 (CAS No. 76993-12-9 and 76993-14-1), RPR-117658, RPR-122818 derivatives, SR-24870 , and RPR-132294 (Sanofi-Aventis), SK-350 (ln2Gen), stroke therapy agents from deCODE Genetics, TAS-203 (Taiho), tofimilast (CAS No. 185954-27-2) (Pfizer), UK- 371800 (Pfizer), WIN-65579 (CAS No. 158020-82-7) (Sanofi-Aventis), IBFB- 130020 (IBFB Pharma), OPC-6535 (CAS No. 145739-56-6) (Otsuka), a phosphodiesterase III inhibitor, enoximone, a phosphodiesterase IV inhibitor, fosfosal, Atopik (Barrier Therapeutics), triflusal, a phosphodiesterase V inhibitor, and an analogue or derivative thereof.
320. The device of item 241 wherein the agent is a PKC inhibitor.
321. The device of item 241 wherein the agent is a platelet activating factor antagonist.
322. The device of item 241 wherein the agent is a platelet- derived growth factor receptor kinase inhibitor.
323. The device of item 241 wherein the agent is a prolyl hydroxylase inhibitor.
324. The device of item 241 wherein the agent is a polymorphonuclear neutrophil inhibitor.
325. The device of item 241 wherein the agent is a protein kinase B inhibitor.
326. The device of item 241 wherein the agent is a protein kinase C stimulant.
327. The device of item 241 wherein the agent is a purine nucleoside analogue. 328. The device of item 241 wherein the agent is a purinoreceptor P2X antagonist.
329. The device of item 241 wherein the agent is a Raf kinase inhibitor.
330. The device of item 241 wherein the agent is a reversible inhibitor of ErbB1 and ErbB2.
331. The device of item 241 wherein the agent is a ribonucleoside triphosphate reductase inhibitor.
332. The device of item 241 wherein the agent is an SDF-1 antagonist.
333. The device of item 241 wherein the agent is a sheddase inhibitor.
334. The device of item 241 wherein the agent is an SRC inhibitor.
335. The device of item 241 wherein the agent is a stromelysin inhibitor.
336. The device of item 241 wherein the agent is an Syk kinase inhibitor.
337. The device of item 241 wherein the agent is a telomerase inhibitor. 338. The device of item 241 wherein the agent is a TGF beta inhibitor selected from the group consisting of pirfenidone (CAS No. 53179- 13-8) (MARNAC), tranilast (CAS No. 53902-12-8) (Kissei), IN-1130 (ln2Gen), mannose-6-phosphate (BTG), TGF-β antagonists from Inflazyme (Pharmaprojects No. 6075), TGF-β antagonists from Sydney, non-industrial source), TGF-β I receptor kinase inhibitors from EIi Lilly, TGF-β receptor inhibitors from Johnson & Johnson, and an analogue or derivative thereof.
339. The device of item 241 wherein the agent is a TNFα antagonist or TACE inhibitor selected from the group consisting of adalimumab (CAS No. 331731-18-1) (Cambridge Antibody Technology), AGlX-4207 (AtheroGenics), AGT-1 (Advanced Biotherapy), an antiinflammatory from Borean Pharma, Cellzome, or Paradigm Therapeutics, anti-inflammatory vaccine (TNF-alpha kinoid) from Neovacs, humanized anti-TNF antibody or an anti-TNF MAb (CB0006) Celltech (UCB), apratastat (CAS No..287445-51-0) (Wyeth), BMSr561392 (Bristol-Myers Squibb), BN- 006 (Bone), certolizumab pegol (CAS No. 428863-50-7 or CH-138 (UCB), cilomilast (CAS No. 153259-65-5) (GlaxoSmithKline), CR-1 (Nuada Pharmaceuticals), CRx-119 (CombinatoRx), D-5410 (UCB), dacopafant (CAS No. 125372-33-0) (Sanofi-Aventis), dersalazine (CAS No. 188913-57- 7/188913-58-8) (Uriach), etanercept (CAS No. 185243-69-0) (Amgen), ethyl pyruvate (Critical (Critical Therapeutics), golimumab (CAS No. 476181-74-5) (Johnson & Johnson), hormono-immunotherapy from Ipsen, CDP571 (e.g., Humicade from UCB), IC-485 (ICOS), infliximab (CAS No. 170277-31-3) (Johnson & Johnson), IP-751 (Manhattan Pharmaceuticals), ISIS-104838 (CAS No. 250755-32-9) (ISIS Pharmaceuticals), lenalidomide (CAS No. 191732-72-6) (Celgene), lentinan (CAS No. 37339-90-5) (Ajinomoto), MDL- 201112 (CAS No. 142130-73-2) (Sanofi-Aventis), medroxyprogesterone (CAS No. 520-85-4) (InKine Pharmaceutical), N-acetylcysteine (CAS No. 616-91-1) (Zambon), NBE-P2 (DIREVO Biotech), nerelimomab (CAS No. 162774-06-3) (Chiron), OM-294DP (OM PHARMA), onercept (CAS No. (Bayer), BIBF-1120 (Boehringer Ingelheim), CEP-5214 (Cephalon), CEP- 7055 (Cephalon), cetuximab (ImClone Systems), CHIR-200131 and CHIR- 258 (Chiron), CP-547632 (OSI Pharmaceuticals), CP-724714 (Pfizer), CT- 301 (Creabilis Therapeutics), D-69491 (Baxter International), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EGFR/ErbB2 inhibitors from Array BioPharma, erlotinib (CAS No. 183319-69-9) (OSI Pharmaceuticals), EXEL-2880 (Exelixis), FK-778 (Sanofi-Aventis) , gefitinib (CAS No. 184475-35-2) (AstraZeneca), GW-2286 or GW-654652 (GlaxoSmithKline), her2/neu antigen from AlphaVax, HER-2/neu inhibitor from Generex, Herzyme (Medipad) (Sirna Therapeutics), HKI-272 (Wyeth), HuMax-EGFr (Genmab), idronoxil (CAS No. 81267-65-4) (Novogen), IGF-1 inhibitors from Ontogen, IMC-11 F8 (ImClone Systems), kahalalide F (CAS No. 149204-42-2) (PharmaMar), KDR inhibitor from LG Life Sciences, KDR inhibitors from Abbott Laboratories, KDR kinase inhibitors (UCB), Kdr kinase inhibitors from Merck & Co, KRN-633 and KRN-951 (Kirin Brewery), KSB-102 (Xenova), lapatinib ditosylate (CAS No. 388082-78-8) (GlaxoSmithKline), matuzumab (Merck KGaA), MDX-214 (Medarex), ME-103 (Pharmexa), MED-A300 (Gerolymatos), MNAC-13 (Lay Line Genomics), nimotuzumab (Center of Molecular Immunology), NSC-330507 or NSC-707545 (NIH), NV-50 (Novogen), OSI-930 (OSI Pharmaceuticals), panitumumab (Abgenix), pelitinib (CAS No. 287933-82-7) (Wyeth), pertuzumab (CAS No. 380610-27- 5) (Genentech), Pharmaprojects No. 3985 (Sanofi-Aventis), prostate cancer therapeutics from Sequenom (SQPC35, SQPC36, SQPC90), removab and remoxab (Trion Pharma), RG-13022 (CAS No. 136831-48-6), RG-13291 (CAS No. 138989-50-1), or RG-14620 (CAS No. 136831-49-7) (Sanofi- Aventis), RM-6427 (Romark), RNAi breast cancer therapy from Benitec, RP 53801 (CAS No. 125882-88-4) (Sanofi-Aventis), sorafenib tosylate (Bayer), SU-11657 (Pfizer), Tie-2 antagonists from Semaia (Hybrigenics), Tie-2 inhibitors from Ontogen, trastuzumab (CAS No. 180288-69-1) (Genentech), tyrosine kinase inhibitors from Sanofi-Aventis, U3-1287, U3-1565, U3-1784, U3-1800 (U3 Pharma), vatalanib (Novartis), VEGFR-2 kinase inhibitor from
832 199685-57-9) (Yeda), PASSTNF-alpha (Verigen), pentoxifylline or oxypentifylline (Sanofi-Aventis), Pharmaprojects No. 4091 , 4241, 4295, or 4488 (Sanofi-Aventis), Pharmaprojects No. 5480 (Amgen), Pharmaprojects No. 6749 (Cengent), pirfenidone (CAS No. 53179-13-8) (MARNAC), RPR- 132294 (Sanofi-Aventis), S5 (F002) (Fulcrum Pharmaceuticals), simvastatin (CAS No. 79902-63-9) (Merck & Co), STA-6292 (Synta Pharmaceuticals), tacrolimus (CAS No. 104987-11-3) (Fujisawa LifeCycle Pharma), talactoferrin alfa (CAS No. 308240-58-6) (Agennix), thalidomide (CAS No. 50-35-1) (Celgene), TNF antagonists form ProStrakan, and Synergen, TNF inhibitors (Amgen), TNF-alpha antagonists from Dynavax Technologies and Jerina AG (Germany), TNF-alpha inhibitors from IBFB Pharma and Xencor (Xencor), torbafylline (CAS No. 105102-21-4) (Sanofi-Aventis), UR-1505 (Uriach), VT-346 (Viron Therapeutics), YSIL6 (Y's Therapeutics), YSTH2 (Y's Therapeutics), NPI-1302a-3 (Nereus Pharmaceuticals, a TNF antagonist from Jerina AG (Germany), dersalazine, and an analogue or derivative thereof.
340. The device of item 241 wherein the agent is a Toll receptor inhibitor.
341. The device of item 241 wherein the agent is a tubulin antagonist.
342. The device of item 241 wherein the agent is a tyrosine kinase inhibitor selected from the group consisting of SU-011248, SUTENT from Pfizer Inc. (New York, NY), BMS-354825, PN-355 (Paracelsian Pharmaceuticals), AGN-199659 (Allergan), AAL-993 or ABP-309 (Novartis), adaphostin (NIH), AEE-788 (Novartis), AG-013736 (OSI Pharmaceuticals), AG-13736 (Pfizer), ALT-110 (Alteris Therapeutics), AMG-706 (Amgen), anticancer MAbs from Xencor, anti-EGFrvlll MAbs from Abgenix, anti-HER2 MAb from Abiogen, AZD-2171 or AZD-9935 (AstraZeneca), BAY-57-9352
831 Bristol-Myers Squibb, XL-647 (Exelixis), ZD-6474 (AstraZeneca), ZK-CDK (Schering AG), an EGFR tyrosine kinase inhibitor, EKB-569 (Wyeth), and an analogue or derivative thereof.
343. The device of item 241 wherein the agent is a VEGF inhibitor.
344. The device of item 241 wherein the agent is a vitamin D receptor agonist.
345. The device of item 241 wherein the agent is ZD-6474 (an angiogenesis inhibitor).
346. The device of item 241 wherein the agent is AP-23573 (an mTOR inhibitor).
347. The device of item 241 wherein the agent is synthadotin (a tubulin antagonist).
348. The device of item 241 wherein the agent is S-0885 (a collagenase inhibitor).
349. The device of item 241 wherein the agent is aplidine (an elongation factor-1 alpha inhibitor).
350. The device of item 241 wherein the agent is ixabepilone (an epithilone).
351. The device of item 241 wherein the agent is IDN-5390 (an angiogenesis inhibitor). 352. The device of item 241 wherein the agent is SB- 2723005 (an angiogenesis inhibitor).
353. The device of item 241 wherein the agent is ABT-518 (an angiogenesis inhibitor).
354. The device of item 241 wherein the agent is combretastatin (an angiogenesis inhibitor).
355. The device of item 241 wherein the agent is anecortave acetate (an angiogenesis inhibitor).
356. The device of item 241 wherein the agent is SB-715992 (a kinesin antagonist).
357. The device of item 241 wherein the agent is temsirolimus (an mTOR inhibitor).
358. The device of item 241 wherein the agent is adalimumab (a TNFα antagonist).
359. The device of item 241, further comprising a polymer.
360. The device of item 241 wherein the anti-scarring agent inhibits adhesion between the device and a host into which the device is implanted.
361. The device of item 241 wherein the device delivers the anti-scarring agent locally to tissue proximate to the device. 362. The device of item 241 , further comprising a coating, wherein the coating comprises the anti-scarring agent.
363. The device of item 241 , further comprising a coating, wherein the coating is disposed on a surface of the device.
364. The device of item 241, further comprising a coating, wherein the coating directly contacts the device.
365. The device of item 241 , further comprising a coating, wherein the coating indirectly contacts the device.
366. The device of item 241 , further comprising a coating, wherein the coating partially covers the device.
367. The device of item 241 , further comprising a coating, wherein the coating completely covers the device.
368. The device of item 241 , further comprising a coating, wherein the coating is a uniform coating.
369. The device of item 241 , further comprising a coating, wherein the coating is a non-uniform coating.
370. The device of item 241 , further comprising a coating, wherein the coating is a discontinuous coating.
371. The device of item 241 , further comprising a coating, wherein the coating is a patterned coating. 372. The device of item 241 , further comprising a coating, wherein the coating has a thickness of 100 Dm or less.
373. The device of item 241, further comprising a coating, wherein the coating has a thickness of 10 Dm or less.
374. The device of item 241 , further comprising a coating, wherein the coating adheres to the surface of the device upon deployment of the device.
375. The device of item 241 , further comprising a coating, wherein the coating is stable at room temperature for a period of 1 year.
376. The device of item 241 , further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 0.0001 % to about 1 % by weight.
377. The device of item 241 , further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 1% to about 10% by weight.
378. The device of item 241 , further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 10% to about 25% by weight.
379. The device of item 241 , further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 25% to about 70% by weight.
380. The device of item 241 , further comprising a coating, wherein the coating further comprises a polymer. 381. The device of item 241 , further comprising a first coating having a first composition and the second coating having a second composition.
382. The device of item 241 , further comprising a first coating having a first composition and the second coating having a second composition, wherein the first composition and the second composition are different.
383. The device of item 241 , further comprising a polymeric carrier, wherein the polymeric carrier comprises a copolymer.
384. The device of item 241 , further comprising a polymeric carrier, wherein the polymeric carrier comprises a block copolymer.
385. The device of item 241 , further comprising a polymeric carrier, wherein the polymeric carrier comprises a random copolymer.
386. The device of item 241 , further comprising a polymeric carrier, wherein the polymeric carrier comprises a biodegradable polymer.
387. The device of item 241 , further comprising a polymeric carrier, wherein the polymeric carrier comprises a non-biodegradable polymer.
388. The device of item 241 , further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrophilic polymer.
389. The device of item 241 , further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrophobic polymer. 390. The device of item 241, further comprising a polymeric carrier, wherein the polymeric carrier comprises a polymer having hydrophilic domains.
391. The device of item 241 , further comprising a polymeric carrier, wherein the polymeric carrier comprises a polymer having hydrophobic domains.
392. The device of item 241 , further comprising a polymeric carrier, wherein the polymeric carrier comprises a non-conductive polymer.
393. The device of item 241 , further comprising a polymeric carrier, wherein the polymeric carrier comprises an elastomer.
394. The device of item 241 , further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrogel.
395. The device of item 241 , further comprising a polymeric carrier, wherein the polymeric carrier comprises a silicone polymer.
396. The device of item 241 , further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrocarbon polymer.
397. The device of item 241 , further comprising a polymeric carrier, wherein the polymeric carrier comprises a styrene-derived polymer.
398. The device of item 241, further comprising a polymeric carrier, wherein the polymeric carrier comprises a butadiene polymer.
399. The device of item 241 , further comprising a polymeric carrier, wherein the polymeric carrier comprises a macromer. 400. The device of item 241, further comprising a polymeric carrier, wherein the polymeric carrier comprises a poly(ethylene glycol) polymer.
401. The device of item 241 , further comprising a polymeric carrier, wherein the polymeric carrier comprises an amorphous polymer.
402. The device of item 241 , further comprising a lubricious coating.
403. The device of item 241 wherein the anti-scarring agent is located within pores or holes of the device.
404. The device of item 241 wherein the anti-scarring agent is located within a channel, lumen, or divet of the device.
405. The device of item 241 , further comprising a second pharmaceutically active agent.
406. The device of item 241 , further comprising an antiinflammatory agent.
407. The device of item 241 , further comprising an agent that inhibits infection.
408. The device of item 241 , further comprising an agent that inhibits infection, wherein the agent is an anthracycline.
409. The device of item 241 , further comprising an agent that inhibits infection, wherein the agent is doxorubicin. 410. The device of item 241 , further comprising an agent that inhibits infection, wherein the agent is mitoxantrone.
411. The device of item 241 , further comprising an agent that inhibits infection, wherein the agent is a fluoropyrimidine.
412. The device of item 241, further comprising an agent that inhibits infection, wherein the agent is 5-fluorouracil (5-FU).
413. The device of item 241 , further comprising an agent that inhibits infection, wherein the agent is a folic acid antagonist.
414. The device of item 241 , further comprising an agent that inhibits infection, wherein the agent is methotrexate.
415. The device of item 241 , further comprising an agent that inhibits infection, wherein the agent is a podophylotoxin.
416. The device of item 241, further comprising an agent that inhibits infection, wherein the agent is etoposide.
417. The device of item 241 , further comprising an agent that inhibits infection, wherein the agent is a camptothecin.
418. The device of item 241 , further comprising an agent that inhibits infection, wherein the agent is a hydroxyurea.
419. The device of item 241 , further comprising an agent that inhibits infection, wherein the agent is a platinum complex. 420. The device of item 241 , further comprising an agent that inhibits infection, wherein the agent is cisplatin.
421. The device of item 241 , further comprising an antithrombotic agent.
422. The device of item 241 further comprising a visualization agent.
423. The device of item 241 , further comprising a visualization agent, wherein the visualization agent is a radiopaque material, wherein the radiopaque material comprises a metal, a halogenated compound, or a barium containing compound.
424. The device of item 241 , further comprising a visualization agent, wherein the visualization agent is a radiopaque material, wherein the radiopaque material comprises barium, tantalum, or technetium.
425. The device of item 241 , further comprising a visualization agent, wherein the visualization agent is a MRI responsive material.
426. The device of item 241, further comprising a visualization agent, wherein the visualization agent comprises a gadolinium chelate.
427. The device of item 241, further comprising a visualization agent, wherein the visualization agent comprises iron, magnesium, manganese, copper, or chromium. 428. The device of item 241 , further comprising a visualization agent, wherein the visualization agent comprises an iron oxide compound.
429. The device of item 241 , further comprising a visualization agent, wherein the visualization agent comprises a dye, pigment, or colorant.
430. The device of item 241 , further comprising an echogenic material.
431. The device of item 241, further comprising an echogenic material, wherein the echogenic material is in the form of a coating.
432. The device of item 241 wherein the device is sterile.
433. The device of item 241 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device.
434. The device of item 241 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is connective tissue.
435. The device of item 241 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is muscle tissue. 436. The device of item 241 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is nerve tissue.
437. The device of item 241 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is epithelium tissue.
438. The device of item 241 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from the time of deployment of the device to about 1 year.
439. The device of item 241 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from about 1 month to 6 months.
440. The device of item 241 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from about 1 - 90 days.
441. The device of item 241 wherein the anti-scarring agent is released in effective concentrations from the device at a constant rate.
442. The device of item 241 wherein the anti-scarring agent is released in effective concentrations from the device at an increasing rate.
443. The device of item 241 wherein the anti-scarring agent is released in effective concentrations from the device at a decreasing rate.
444. The device of item 241 wherein the anti-scarring agent is released in effective concentrations from the composition comprising the anti-scarring agent by diffusion over a period ranging from the time of deployment of the device to about 90 days.
445. The device of item 241 wherein the anti-scarring agent is released in effective concentrations from the composition comprising the anti-scarring agent by erosion of the composition over a period ranging from the time of deployment of the device to about 90 days.
446. The device of item 241 wherein the device comprises about 0.01 Dg to about 10 Dg of the anti-scarring agent.
447. The device of item 241 wherein the device comprises about 10 Dg to about 10 mg of the anti-scarring agent.
448. The device of item 241 wherein the device comprises about 10 mg to about 250 mg of the anti-scarring agent.
449. The device of item 241 wherein the device comprises about 250 mg to about 1000 mg of the anti-scarring agent.
450. The device of item 241 wherein the device comprises about 1000 mg to about 2500 mg of the anti-scarring agent.
451. The device of item 241 wherein a surface of the device comprises less than 0.01 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
452. The device of item 241 wherein a surface of the device comprises about 0.01 Dg to about 1 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied. 453. The device of item 241 wherein a surface of the device comprises about 1 Dg to about 10 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
454. The device of item 241 wherein a surface of the device comprises about 10 Og to about 250 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
455. The device of item 241 wherein a surface of the device comprises about 250 Dg to about 1000 Dg of the anti-scarring agent of anti- scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
456. The device of item 241 wherein a surface of the device comprises about 1000 Dg to about 2500 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
457. The device of any one of items 241-456, wherein the device is a catheter.
458. The device of any one of items 241-456, wherein the device is a balloon catheter.
459. The device of any one of items 241-456, wherein the device is a balloon.
460. The device of any one of items 241-456, wherein the device is a stent graft.
461. The device of any one of items 241-456, wherein the device is a guidewire. 462. The device of any one of items 241-456, wherein the device is a stent.
463. The device of any one of items 241-456, wherein the device is an intravascular stent.
464. The device of any one of items 241-456, wherein the device is a metallic stent.
465. The device of any one of items 241-456, wherein the device is a polymeric stent.
466. The device of any one of items 241-456, wherein the device is a biodegradable stent.
467. The device of any one of items 241-456, wherein the device is a non-biodegradable stent.
468. The device of any one of items 241-456, wherein the device is a self-expandable stent.
469. The device of any one of items 241-456, wherein the device is a balloon expandable stent.
470. The device of any one of items 241-456, wherein the device is a covered stent.
471. The device of any one of items 241-456, wherein the device is a drug eluting stent. 472. The device of any one of items 241-456, wherein the device is a stent that comprises a radio-opaque material.
473. The device of any one of items 241-456, wherein the device is a stent that comprises an echogenic material.
474. The device of any one of items 241-456, wherein the device is a stent that comprises an MRI responsive material.
475. The device of any one of items 241-456, wherein the device is an anastomotic connector device.
476. The device of any one of items 241-456, wherein the device is an artery to artery anastomotic connector device.
477. The device of any one of items 241-456, wherein the device is a vein to artery anastomotic connector device.
478. The device of any one of items 241-456, wherein the device is an artery to vein anastomotic connector device.
479. The device of any one of items 241-456, wherein the device is an artery to synthetic graft anastomotic connector device.
480. The device of any one of items 241-456, wherein the device is a synthetic graft to artery anastomotic connector device.
481. The device of any one of items 241-456, wherein the device is a vein to synthetic graft anastomotic connector device. 482. The device of any one of items 241-456, wherein the device is a synthetic graft to vein anastomotic connector device.
483. The device of any one of items 241-456, wherein the device is a vascular clip.
484. The device of any one of items 241-456, wherein the device is a vascular clamp.
485. The device of any one of items 241-456, wherein the device is a suturing device.
486. The device of any one of items 241-456, wherein the device is an anastomotic coupler.
487. . The device of any one of items 241-456, wherein the device is an automated or modified suture device.
488. The device of any one of items 241-456, wherein the device is a micromechanical anastomotic connector device.
489. The device of any one of items 241-456, wherein the device is an anastomotic coupling device that facilitates automated attachment of a graft or vessel to an aperature or orifice in a target vessel without the sue of sutures or staples.
490. The device of any one of items 241-456, wherein the device is an anastomotic coupling device that comprises a tubular graft conduit and may be placed in a side wall of a target vessel so that the tubular graft conduit may be extended from the target vessel. 491. The device of any one of items 241-456, wherein the device is an anastomotic coupler in the form of a frame.
492. The device of any one of items 241-456, wherein the device is an anastomotic coupler in a ring-like form.
493. The device of any one of items 241-456, wherein the device is a resorbable anastomotic coupler.
494. The device of any one of items 241-456, wherein the device is an anastomotic coupler that comprises a bioabsorbable and elastomeric material.
495. The device of any one of items 241-456, wherein the device is an anastomotic coupler adapted to connect a first blood vessel with a second blood vessel with a graft vessel.
496. The device of any one of items 241-456, wherein the device is an anastomotic coupler adapted to connect a first blood vessel with a second blood vessel without a graft vessel.
497. The device of any one of items 241-456, wherein the device is an anastomotic coupler that is incorporated in the design of a vascular graft.
498. The device of any one of items 241-456, wherein the device is an anastomotic coupler that comprises a graft that incorporates a fixation mechanism. 499. The device of any one of items 241-456, wherein the device is an anastomotic coupler that comprises a compressible, expandable fitting for securing the ends of a bypass graft to two vessels.
500. The device of any one of items 241-456, wherein the device is an anastomotic coupler that comprises a pair of coupling disc members for joining two vessels in an end to end or end to side fashion.
501. The device of any one of items 241-456, wherein the device is a proximal aortic connector.
502. The device of any one of items 241-456, wherein the device is a distal coronary connector.
503. The device of any one of items 241-456, wherein the device is a bypass device made of a biocompatible material.
504. The device of any one of items 241-456, wherein the device is a bypass device made of at least partially a metal or metal alloy.
505. The device of any one of items 241-456, wherein the device is a bypass device made of at least partially a synthetic polymer.
506. The device of any one of items 241-456, wherein the device is a bypass device made of at least partially naturally derived polymer.
507. The device of any one of items 241-456, wherein the device is a tubular anastomotic coupler that comprises a tubular structure that may be attached directly to a proximal blood vessel. 508. The device of any one of items 241-456, wherein the device is a tubular anastomotic coupler that comprises a tubular structure that may be attached directly to a distal blood vessel.
509. The device of any one of items 241-456, wherein the device is a tubular anastomotic coupler that comprises a tubular structure that has a proximal end attachable to a proximal vessel and a distal end attachable to a bypass graft.
510. The device of any one of items 241-456, wherein the device is a tubular anastomotic coupler that has a proximal end attachable to a graft Bessel that is secured to a proximal blood vessel and a distal end attachable to a distal blood vessel.
511. The device of any one of items 241-456, wherein the device is an anastomotic connector device adapted for end to end anastomosis procedures.
512. The device of any one of items 241-456, wherein the device is an anastomotic stent.
513. The device of any one of items 241-456, wherein the device is an anastomotic sleeve.
514. The device of any one of items 241-456, wherein the device is an anastomotic connector device adapted for end to side anastomosis procedures.
515. The device of any one of items 241-456, wherein the device is a single lumen bypass device. 516. The device of any one of items 241-456, wherein the device is a multi-lumen bypass device.
517. The device of any one of items 241-456, wherein the device is an anastomotic coupling device that comprises a single tubular portion that may be used as a shunt to divert blood from a source vessel to a graft vessel.
518. The device of any one of items 241-456, wherein the device is an anastomotic coupling device that comprises more than one tubular portion, and wherein at least one tubular portion may be used as a shunt for diverting blood between a source vessel and a target vessel.
519. The device of any one of items 241-456, wherein the device is an anastomotic connector device that comprises a tubular portion, and wherein one or more ends of the tubular portion may be inserted into the end or into the side of one or more blood vessels.
520. The device of any one of items 241-456, wherein the device is a multi-lumen anastomotic connector device that at least one arm of the device may be attached to a graft vessel.
521. The device of any one of items 241-456, wherein the device is an anastomotic connector device that includes three or more tubular arms that extend from a junction site.
522. The device of any one of items 241-456, wherein the device is a multi-lumen anastomotic connector device that is generally T- shaped. 523. The device of any one of items 241-456, wherein the device is a multi-lumen anastomotic connector device that is generally Y- shaped.
524. The device of any one of items 241-456, wherein the device is an anastomotic connector device that comprises a tube for bypassing blood flow directly from a portion of the heart to a coronary artery.
525. The device of any one of items 241-456, wherein the device is an anastomotic connector device that comprises a network of interconnected tubular conduits.
526. The device of any one of items 241-456, wherein the device is an anastomotic connector device that is configured with two or more termini that provide a vessel interface without the need for sutures and a fluid communication through an intersecting lumen.
527. A device, comprising a gastrointestinal stent (e.g., an implant) and an anti-scarring agent or a composition comprising an anti- scarring agent, wherein the agent inhibits scarring between the device and a host into which the device is implanted.
528. The device of item 527 wherein the agent is an adensosine A2A receptor antagonist.
529. The device of item 527 wherein the agent is an AKT inhibitor.
530. The device of item 527 wherein the agent is an alpha 2 integrin antagonist, wherein the alpha 2 integrin antagonist is Pharmaprojects No. 5754 (Merck KgaA). 531. The device of item 527 wherein the agent an alpha 4 integrin antagonist.
532. The device of item 527 wherein the agent is an alpha 7 nicotinic receptor agonist.
533. The device of item 527 wherein the agent is an angiogenesis inhibitor selected from the group consisting of AG-12,958 (Pfizer), ATN-161 (Attention LLC), neovastat, an angiogenesis inhibitor from Jerina AG (Germany), NM-3 (Mercian), VGA-1155 (Taisho), FCE-26644 (Pfizer), FCE-26950 (Pfizer), FPMA (Meiji Daries), FR-111142 (Fujisawa), GGTI-298, GM-1306 (Ligand), GPA-1734 (Novartis), NNC-47-0011 (Novo Nordisk), herbamycin (Nippon Kayakυ), lenalidomide (Celegene), IP-10 (NIH), ABT-828 (Abbott), KIN-841 (Tokushima University, Japan), SF-1126 (Semafore Pharmaceuticals), laminin technology (NIH), CHIR-258 (Chiron), NVP-AEW541 (Novartis), NVP-AEW541 (Novartis), VU6907 (Alchemia), OXI-8007 (Oxigene), EG-3306 (Ark Therapeutics), Maspin (Arriva), ABT- 567 (Abbott), PPI-2458 (Praecis Pharmaceuticals), CC-5079, CC-4089 (Celgene), HIF-1 alpha inhibitors (Xenova), S-247 (Pfizer), AP-23573 (Ariad), AZD-9935 (Astra Zeneca), mebendazole (Introgen Therapeutics), MetAP-2 inhibitors (GlaxoSmithKline), AG-615 (Angiogene Pharmaceuticals), Tie-2 antagonists (Hybrigenics), NC-381 , CYC-381, NC-169, NC-219, NC-383, NC-384, NC-407 (Lorus Therapeutics), ATN-224 (Attenuon), ON-01370 (Onconova), Vitronectin antagonists (Amgen), SDX-103 (Salmedix), Vitronectin antagonists (Shire), CHP (Riemser), TEK (Amgen), Anecortave acetate (Alcon), T46.2 (Matrix Therapeutics), HG-2 (Heptagen), TEM antagonists (Genzyme), Oxi-4500 (Oxigene), ATN-161 (Attenuon), WX-293 (Wilex), M-2025 (Metris Therapeutics), Alphastatin (BioActa), YH-16 (Yantai Rongchang), BIBF-1120 (Boehringer Ingelheim), BAY-57-9352 (Bayer), AS- 1404 (Cancer Research Technology), SC-77964 (Pfizer), glycomimetics (BioTie Therapies), TIE-2 Inhibitors (Ontogen), DIMI, Octamer (Octamer), ABR-215050 (Active Biotech), ABT-518 (Abbott), KDR inhibitors (Abbott), BSF-466895 (Abbott), SCH-221153 (Schering-Plough), DAC:antiangiogenic (ConjuChem), TFPI (EntreMed), AZD-2171 (Astra-Zenaca), CDC-394 (Celgene), LY290293 (EIi Lilly), IDN-5390 (Indena), Kdr Kinase Inhibitors (Merck), CT-113020, CT-116433, CT-116563, CT-31890, CT-32228) (Cell Therapeutics), A-299620 (Abbott), TWEAK Inhibitor (Amgen), VEGF modulators (Johnson and Johnson), Tum-N53, tumstatin (Genzyme), Thios- 1 , Thios-2 (Thios Pharmaceuticals), MV-6401 (Miravant Medical Technologies), Spisulosine (PharmaMar), CEP-7055 (Cephalon), AUV-201 (Auvation), LM-609 (EIi Lilly), SKF-106615 (AnorMED), Oglufanide disodium (Cytran), BW-114 (Pharminox), Calreticulin (NIH), WX-678 (Wilex), SD-7784 (Pfizer), WX-UK1 (Wilex), SH-268 (Schering AG), 2-Me-PGA (Celgene), S- 137 (Pfizer), ZD-6126 (Angiogene Pharmaceuticals), SG-292 (SignalGen), Benefin (Lane Labs), A6, A36 (Angstrom), SB-2723005 (GlaxoSmithKline), SC-7 (Cell Therapeutics), ZEN-014 (AEtema Zentaris), 2-methoxyestradiol (EntreMed), NK-130119 (Nippon Kayaku), CC-10004 (Celgene), AVE- 8062A (Ajinomoto), Tacedinaline (Pfizer), Actinonin (Tokyo Metropolitan Institute of Medical Science), Lenalidomide (Celgene), VGA-1155, BTO-956 (SRI International), ER-68203-00 (Eisai), CT-6685 (UCB), JKC-362 (Phoenix Pharmaceuticals), DMI-3798 (DMI Biosciences, Angiomate (Ipsen), ZD-6474 (AstraZeneca), CEP-5214 (Cephalon), Canstatin (Genzyme), NM-3 (Mercian), Oridigm (MediQuest Therapeutics), Exherin (Adherex), BLS-0597 (Boston Life Sciences), PTC-299 (PTC Therapeutics), NPI-2358 (Nereus Pharmaceuticals), CGP-79787 (Novartis), AEE-788 (Novartis), CKD-732 (Chong Kun Dang), CP-564959 (OSI Pharmaceuticals), CM-101 (CarboMed), CT-2584, CT3501 (Cell Therapeutics), combretastatin and analogues and derivatives thereof (Oxigene), Rebimastat (Bristol-Meyers Squibb), Dextrin 2-sulfate (ML Laboratories), Cilengitide (Merk KGaA), NSC-706704 (Pharminox), KRN- 951 (Kirin Brewery), Ukrain, NSC-631570 (Nowicky Pharma), Tecogalan sodium (Daiichi Pharmaceutical), Tz-93 (Tsumura), TBC-1635 (Encysive Pharmaceuticals), TAN-1120 (Takeda), Semaxanib (Pfizer), BDI-7800 (Biopharmacopae), SD-186, SD-983 (Bristol-Meyers Squibb), SB-223245 (GlaxoSmithKline), SC-236 (Pfizer), RWJ-590973 (Johnson and Johnson), ILX-1850 (Genzyme), SC-68488, S-836 (Pfizer), CG-55069-11 (CuraGen), Ki-23057 (Kirin Brewery), CCX-700 (Chemoentryx), Pegaptanib octasodium (Giled Sciences), ANGIOCOL (available from Biostratum Inc.), or an analogue or derivative thereof.
534. The device of item 527 wherein the agent is an apoptosis antagonist.
535. The device of item 527 wherein the agent is an apoptosis activator.
536. The device of item 527 wherein the agent is a beta 1 integrin antagonist.
537. The device of item 527 wherein the agent is a beta tubulin inhibitor.
538. The device of item 527 wherein the agent is a blocker of enzyme production in Hepatitis C.
539. The device of item 527 wherein the agent is a Bruton's tyrosine kinase inhibitor.
540. The device of item 527 wherein the agent is a calcineurin inhibitor.
541. The device of item 527 wherein the agent is a caspase 3 inhibitor. 542. The device of item 527 wherein the agent is a CC chemokine receptor antagonist.
543. The device of item 527 wherein the agent is a cell cycle inhibitor selected from the group consisting of SNS-595 (Sunesis), synthadotin, KRX-0403, and an analogue or derivative thereof.
544. The device of item 527 wherein the agent is a cathepsin B inhibitor.
545. The device of item 527 wherein the agent is a cathepsin K inhibitor, wherein the cathepsin K inhibitor is 462795 (GlaxoSmithKline), INPL-022-D6 (Amura Therapeutics), an analogue or derivative thereof).
546. The device of item 527 wherein the agent is a cathepsin L inhibitor.
547. The device of item 527 wherein the agent is a CD40 antagonist.
548. The device of item 527 wherein the agent is a chemokine receptor agonist.
549. The device of item 527 wherein the agent is a chymase inhibitor.
550. The device of item 527 wherein the agent is a collagenase antagonist.
551. The device of item 527 wherein the agent is a CXCR antagonist. 552. The device of item 527 wherein the agent is a cyclin dependent kinase inhibitor selected from the group consisting of a CDK-1 inhibitor, a CDK-2 inhibitor, a CDK- 4 inhibitor, a CDK-6 inhibitor, a CAK1 inhibitor from GPC Biotech and Bristol-Myers Squibb, RGB-286199 (GPC Biotech), an anticancer agent from Astex Technology, a CAK1 inhibitor from GPC Biotech, a CDK inhibitor from Sanofi-Aventis, a CDK1/CDK2 inhibitor from Amgen, a CDK2 inhibitor from SUGEN-2 (Pfizer), a hearing loss therapy agent (Sound Pharmaceuticals), PD-0332991 (Pfizer), RGB-286199 (GPC Biotech), Ro-0505124 (Hoffmann-La Roche), a Ser/Thr kinase inhibitor from Lilly (EIi Lilly), CVT-2584 (CAS No. 199986-75-9) (CV Therapeutics), and an analogue or derivative thereof.
553. The device of item 527 wherein the agent is a cyclooxygenase 1 inhibitor.
554. The device of item 527 wherein the agent is a DHFR inhibitor.
555. The device of item 527 wherein the agent is a dual integrin inhibitor.
556. The device of item 527 wherein the agent is an elastase inhibitor.
557. The device of item 527 wherein the agent is an elongation factor-1 alpha inhibitor.
558. The device of item 527 wherein the agent is an endothelial growth factor antagonist. 559. The device of item 527 wherein the agent is an endothelial growth factor receptor kinase inhibitor selected from the group consisting of sorafenib tosylate (Bayer), AAL-993 (Novartis), ABP-309 (Novartis), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), E- 7080 (Eisai), EG-3306 (Ark Therapeutics), EXEL-2880 (Exelixis), GW- 654652 (GlaxoSmithKline), a KDR inhibitor from LG Life Sciences, CT-6685 and CT-6729 (UCB), KRN-633 and KRN-951 (Kirin Brewery), OSI-930 (OSI Pharmaceuticals), SP-5.2 (Supratek Pharma), SlM 1657 (Pfizer), a Tie-2 antagonist (Hybrigenics), a VEGFR-2 kinase inhibitor (Bristol-Myers Squibb), XL-647 (Exelixis), a KDR inhibitor from Abbott Laboratories, sorafenib tosylate, and an analogue or derivative thereof.
560. The device of item 527 wherein the agent is an endotoxin antagonist.
561. The device of-item 527 wherein the agent is an epothilone and tubulin binder.
562. The device of item 527 wherein the agent is an estrogen receptor antagonist.
563. The device of item 527 wherein the agent is an FGF inhibitor.
564. The device of item 527 wherein the agent is a famexyl transferase inhibitor.
565. The device of item 527 wherein the agent is farnesyltransferase inhibitor selected from the group of A-197574 (Abbott), a famesyltransferase inhibitor from Servier, FPTIII (Strathclyde Institute for Drug R), LB-42908 (LG Life Sciences), Pharmaprojects No. 5063 (Genzyme), Pharmaprojects No. 5597 (Ipsen), Yissum Project No. B-1055 (Yissum), and an analogue or derivative thereof.
566. The device of item 527 wherein the agent is an FLT-3 kinase inhibitor.
567. The device of item 527 wherein the agent is an FGF receptor kinase inhibitor.
568. The device of item 527 wherein the agent is a fibrinogen antagonist selected from the group consisting of AUV-201 (Auvation), MG-13926 (Sanofi-Aventis), plasminogen activator (CAS No. 105913-11-9) (from Sanofi-Aventis or UCB), plasminogen activator-2 (tPA- 2) (Sanofi-Aventis), pro-urokinase (CAS No. 82657-92-9) (Sanofi-Aventis), and an analogue or derivative thereof.
569. The device of item 527 wherein the agent is a heat shock protein 90 antagonist selected from the group consisting of SRN-005 (Sirenade), geldanamycin, NSC-33050 (17-allylaminogeldanamycin; 17- AAG), 17-dimethylaminoethylamino-17-demethoxy-geldanamycin (17- DMAG), rifabutin (rifamycin XIV, 1\4-didehydro-1-deoxy-1 ,4-dihydro-5'-(2- methylpropyl)-1-oxo-), and an analogue or derivative thereof.
570. The device of item 527 wherein the agent is a histone deacetylase inhibitor.
571. The device of item 527 wherein the agent is an HMGCoA reductase inhibitor selected from the group consisting of an atherosclerosis therapeutic from Lipid Sciences, ATI-16000 (ARYx Therapeutics), KS-01-019 (Kos Pharmaceuticals), Pharmaprojects No. 2197 (Sanofi-Aventi), RP 61969 (Sanofi-Aventis), and an analogue or derivative thereof.
572. The device of item 527 wherein the agent is an ICAM inhibitor.
573. The device of item 527 wherein the agent is an IL, ICE and IRAK antagonist, wherein the antagonist is a CJ-14877, CP-424174 (Pfizer), NF-61 (Negma-Lerads), and an analogue or derivative thereof.
574. The device of item 527 wherein the agent is an IL-2 inhibitor.
575. The device of item 527 wherein the agent is an immunosuppressant selected from the group consisting of teriflunomide (Sanofi Aventis), chlorsulfaquinoxalone (NSC-339004), chlorsulfaquinoxalone sulfate, CS-712 (Sankyo), ismomultin alfa (CAS No. 457913-93-8) (Akzo Nobel), antiallergics from GenPat77, antiinflammatories or AT-005 (Androclus Therapeutics), autoimmune disease therapeutics from EpiVax, BN-007 (Bone), budesonide (CAS No. 51333-22- 3) (MAP Pharmaceuticals), CO-14 (Genzyme), edratide (CAS No. 433922- 67-9) (Teva), EP-314 (Enanta), eprovafen (CAS No. 101335-99-3) (Sanofi- Aventis), HWA-131 (CAS No. 118788-41-3) (Sanofi-Aventis), immunomodulators from MerLion Pharmaceuticals, immunosuppressives from Alchemia, IPL-12 (Inflazyme), MDL-9563 (CAS No. 27086-86-8) (Sanofi-Aventis), Pharmaprojects No. 2330 (Sanofi-Aventis), Phaπmaprojects No. 6426 (Abgenix), PXS-25 (Pharmaxis), rosmarinic acid (CAS No. 20283-92-5) (Sanofi-Aventis), RP 42927 or RP 54745 (CAS No. 135330-08-4) (Sanofi-Aventis), SGN-35 (Seattle Genetics), ST-1959 (Sigma-Tau), type I diabetes therapy from SYNX Pharma, UNIL-88 (Debiopharm), VP-025 (Vasogen), VR-694 (Vectura), PRTX-001 (Protalex), and an analogue or derivative thereof.
576. The device of item 527 wherein the agent is an IMPDH (inosine monophosphate).
577. The device of item 527, wherein the agent is an integrin antagonist.
578. The device of item 527 wherein the agent is an interleukin antagonist.
579. The device of item 527 wherein the agent is an inhibitor of type III receptor tyrosine kinase.
580. The device of item 527 wherein the agent is an irreversible inhibitor of enzyme methionine aminopeptidase type 2.
581. The device of item 527 wherein the agent is an isozyme selective delta protein kinase C inhibitor.
582. The device of item 527 wherein the agent a JAK3 enzyme inhibitor.
583. The device of item 527 wherein the agent is a JNK inhibitor.
584. The device of item 527 wherein the agent is a kinase inhibitor. 585. The device of item 527 wherein the agent is kinesin antagonist.
586. The device of item 527 wherein the agent is a kinesin antagonist.
587. The device of item 527 wherein the agent is a leukotriene inhibitor and antagonist selected from the group consisting of ambicromil (CAS No. 58805-38-2) (Sanofi-Aventis), amelubant (CAS No. 346735-24-8) (Boehringer Ingelheim), DW-1141 (Dong Wha), ebselen (Daiichi Pharmaceutical), ibudilast (Kyorin), leucotriene inhibitors from Sanofi-Aventis, lymphotoxin-beta receptor (LT-D) from Biogen Idee, Pharmaprojects No. 1535 and 2728 (CAS No. 119340-33-9) (Sanofi- Aventis), R-112 (Rigel), Rev-5367 (CAS No. 92532-05-3) (Sanofi-Aventis), RG-14893 (CAS No. 141835-49-6) (Sanofi-Aventis), RG-5901-A (CAS No. 101910-24-1), 92532-23-5, RP 66153 (CAS No. 142422-79-5), RP 66364 (CAS No.186916-92-5), RP 69698 (CAS No. 141748-00-7) (Sanofi-Aventis), SC-411930 (Pfizer), SC-41930 (CAS No. 120072-59-5) (Pfizer), SC-50605 (CAS No. 138828-39-4) (Pfizer), SC-51146 (CAS No. 141059-52-1), SC- 53228 (CAS No. 153633-01-3) (Pfizer), spaglumic acid (ZY-15106) (CAS No.3106-85-2), 80619-64-3 (Novartis), tipredane (CAS No. 85197-77-9) (Bristol-Myers Squibb), U-75302 (CAS No. 119477-85-9) (Pfizer), and analogue or derivative thereof.
588. The device of item 527 wherein the agent is an MAP kinase inhibitor.
589. The device of item 527 wherein the agent is a matrix metalloproteinase inhibitor. 590. The device of item 527 wherein the agent is an MCP- CCR2 inhibitor.
591. The device of item 527 wherein the agent is an mTOR inhibitor.
592. The device of item 527 wherein the agent is an mTOR kinase inhibitor.
593. The device of item 527 wherein the agent is a microtubule inhibitor selected from the group consisting of antibody- maytansinoid conjugates from Biogen Idee, colchicines (MantiCore Pharmaceuticals), anticancer immunoconjugates from Johnson & Johnson, DIME from Octamer, gni-1f (GNI), huC242-DM4, huMy9-6-DM1 (ImmunoGen), IDN-5404 (Indena), IMO-098, IMOderm (Imotep), mebendazole (Introgen Therapeutics), microtubule poisons from Cambridge Enterprise, paclitaxel such as LOTAX from Aphios (CAS No. 33069-62-4), Genexol-PM from Samyang, Pharmaprojects No. 6383 (Tapestry Pharmaceuticals), RPR-112378 (Sanofi-Aventis), SGN-75 (Seattle Genetics), SPL-7435 (Starpharma), SSR-250411 (Sanofi-Aventis), trastuzumab-DM1 (Genentech), vinorelbine, and an analogue or derivative thereof.
594. The device of item 527 wherein the agent is an MIF inhibitor.
595. The device of item 527 wherein the agent is an MMP inhibitor.
596. The device of item 527 wherein the agent is a neurokinin (NK) antagonist selected from the group consisting of anthrotainin (CAS No. 148084-40-6) (Sanofi-Aventis), an IBS thereapeutic from ArQuIe, MDL-105212A (CAS No. 167261-60-1) (Ssanofi-Aventis), Pharmaprojects No. 2744, 3258 (CAS No. 139167-47-8) 4006, 4201 , or 5986 (Sanofi-Aventis), RP 67580 (CAS No. 135911-02-3), SR-144190 (CAS No. 201152-86-5), SSR-240600, SSR-241586 (Sanofi-Aventis), TKA-457 (Novartis), vestipitant mesylate (CAS No. 334476-64-1) (GlaxoSmithKline), Win-64821 (Sanofi-Aventis), PRX-96026 (Predix Pharmaceuticals), and an analogue or derivative thereof.
597. The device of item 527 wherein the agent is an NF kappa B inhibitor selected from the group consisting of AVE-0545 or AVE- 0547 (Sanofi-Aventis), bortezomib (CAS No. 179324-69-7) (Millennium Pharmaceuticals), dexanabinol (CAS No. 112924-45-5) (Pharmos), dexlipotam (Viatris), Pharmaprojects No. 6283 (INDRA) (OXiGENE), IPL- 576092 (CAS No. 137571-30-3) (Inflazyme), NFKB decoy (Corgentech), NFKB decoy oligo (AnGes MG), NFKB's from Ariad, osteoporosis treatments or S5 (F005) from Fulcrum Pharmaceuticals, P61 (Phytopharm), R-flurbiprofen (CAS No. 5104-49-4) (Encore Pharmaceuticals), and an analogue or derivative thereof.
598. The device of item 527 wherein the agent is a nitric oxide agonist.
599. The device of item 527 wherein the agent is an ornithine decarboxylase inhibitor.
600. The device of item 527 wherein the agent is a p38 MAP kinase inhibitor selected from the group consisting of AZD-6703 (AstraZeneca), JX-401 (Jexys Pharmaceuticals), BMS-2 (Bristol-Myers Squibb), a p38 MAP kinase inhibitor from Novartis, a p38-alpha MAP kinase inhibitor from Amphora, Pharmaprojects No. 5704 (Pharmacopeia), RPR- 200765A (Sanofi-Aventis), SD-282 (Johnson & Johnson), TAK-715 (Takeda), and an analogue or derivative thereof.
601. The device of item 527 wherein the agent is a palmitoyl- protein thioesterase inhibitor.
602. The device of item 527 wherein the agent is a PDGF receptor kinase inhibitor selected from the group consisting of AAL-993, AMN-107, or ABP-309 (Novartis), AMG-706 (Amgen), BAY-57-9352 (Bayer), CDP-860 (UCB), E-7080 (Eisai), imatinib (CAS No. 152459-95-5) (Novartis), OSI-930 (OSI Pharmaceuticals), RPR-127963E (Sanofi-Aventis), RWJ-540973 (Johnson & Johnson), sorafenib tosylate (Bayer), SU-11657 (Pfizer), tandutinib (CAS No. 387867-13-2) (Millennium Pharmaceuticals), vatalanib (Novartis), ZK-CDK (Schering AG), and an analogue or derivative thereof.
603. The device of item 527 wherein the agent is (-)- halofenate (Metabolex), AMG-131 (Amgen), antidiabetics from Japan Tobacco, AZD-4619, AZD-8450, AZD-8677 (AstraZeneca), DRF-10945, balaglitazone (Dr Reddy's), CS-00088, CS-00098 (Chipscreen Biosciences), E-3030 (Eisai), etalocib (CAS No. 161172-51-6) (EIi Lilly), GSK-641597 (Ligand), GSK-677954 (GlaxoSmithKline), GW-409544 (Ligand), GW- 590735 (GlaxoSmithKline), K-111 (Hoffmann-La Roche), LY-518674 (EIi Lilly), LY-674 (Ligand), LY-929 (Ligand), MC-3001 , MC-3002 (MaxoCore Pharmaceuticals), metformin HCI + pioglitazone (CAS No. 1115-70-4 and
112529-15-4), ACTOPLUS MET from Andrx), muraglitazar (CAS No. 331741-94-7) (Bristol-Myers Squibb), naveglitazar (Ligand), oleoylethanolamide (Kadmus Pharmaceuticals), ONO-5129, pioglitazone hydrochloride (CAS No. 111025-46-8 and 112529-15-4) (Takeda), PLX-204 (Plexxikon), PPAR agonists from Genfit, PPAR delta agonists from EIi Lilly, PPAR-alpha agonists from CrystalGenomics, PPAR-gamma modulators and PPAR-β modulators from CareX, rosiglitazone maleate (CAS No. 122324- 73-4 or 155141-29-0) (GlaxoSmithKline), rosiglitazone maleate/glimepir (CAS No. 155141-29-0 and 93479-97-1), AVANDARYL, rosiglitazone maleate/metformin extend (CAS No. 155141-29-0 and 657-24-9), AVANDAMET, rosiglitazone maleate+metformin, AVANDAMET (GlaxoSmithKline), tesaglitazar (AstraZeneca), and an analogue or derivative thereof.
604. The device of item 527 wherein the agent is a phosphatase inhibitor.
605. The device of item 527 wherein the agent is a phosphodiesterase (PDE) inhibitor selected from the group consisting of avanafil (Tanabe Seiyaku), dasantafil (CAS No. 569351-91-3) (Schering- Plough), A-906119 (CAS No. 134072-58-5), DL-850 (Sanofi-Aventis), GRC- 3015, GRC-3566, GRC-3886 (Glenmark), HWA-153 (CAS No. 56395-66-5) (Sanofi-Aventis), hydroxypumafentrine (Altana), IBFB-130011 , IBFB-14-016, IBFB-140301 , IBFB-150007, IBFB-211913 (IBFB Pharma), L-826141 (Merck & Co), medorinone (CAS No. 88296-61-1) (Sanofi-Aventis), MEM- 1917 (Memory Pharmaceuticals), ND-1251 (Neuro3d), PDE inhibitors from ICOS, PDE IV inhibitors from Memory Pharmaceuticals and CrystalGenomics, Pharmaprojects No. 2742 and 6141 (Sanofi-Aventis), QAD-171 (Novartis), RHC-2963 (CAS No. 76993-12-9 and 76993-14-1), RPR-117658, RPR-122818 derivatives, SR-24870 , and RPR-132294 (Sanofi-Aventis), SK-350 (ln2Gen), stroke therapy agents from deCODE Genetics, TAS-203 (Taiho), tofimilast (CAS No. 185954-27-2) (Pfizer), UK- 371800 (Pfizer), WIN-65579 (CAS No. 158020-82-7) (Sanofi-Aventis), IBFB- 130020 (IBFB Pharma), OPC-6535 (CAS No. 145739-56-6) (Otsuka), a phosphodiesterase III inhibitor, enoximone, a phosphodiesterase IV inhibitor, fosfosal, Atopik (Barrier Therapeutics), triflusal, a phosphodiesterase V inhibitor, and an analogue or derivative thereof. 606. The device of item 527 wherein the agent is a PKC inhibitor.
607. The device of item 527 wherein the agent is a platelet activating factor antagonist.
608. The device of item 527 wherein the agent is a platelet- derived growth factor receptor kinase inhibitor.
609. The device of item 527 wherein the agent is a prolyl hydroxylase inhibitor.
610. The device of item 527 wherein the agent is a polymorphonuclear neutrophil inhibitor.
6-11. The device of item 527 wherein the agent is a protein kinase B inhibitor.
612. The device of item 527 wherein the agent is a protein kinase C stimulant.
613. The device of item 527 wherein the agent is a purine nucleoside analogue.
614. The device of item 527 wherein the agent is a purinoreceptor P2X antagonist.
615. The device of item 527 wherein the agent is a Raf kinase inhibitor. 616. The device of item 527 wherein the agent is a reversible inhibitor of ErbB1 and ErbB2.
617. The device of item 527 wherein the agent is a ribonucleoside triphosphate reductase inhibitor.
618. The device of item 527 wherein the agent is an SDF-1 antagonist.
619. The device of item 527 wherein the agent is a sheddase inhibitor.
620. The device of item 527 wherein the agent is an SRC inhibitor.
621. - The device of item 527 wherein the agent is a stromelysin inhibitor.
622. The device of item 527 wherein the agent is an Syk kinase inhibitor.
623. The device of item 527 wherein the agent is a telomerase inhibitor.
624. The device of item 527 wherein the agent is a TGF beta inhibitor selected from the group consisting of pirfenidone (CAS No. 53179- 13-8) (MARNAC), tranilast (CAS No. 53902-12-8) (Kissei), IN-1130 (ln2Gen), mannose-6-phosphate (BTG), TGF-β antagonists from Inflazyme (Pharmaprojects No. 6075), TGF-β antagonists from Sydney, non-industrial source), TGF-β I receptor kinase inhibitors from EIi Lilly, TGF-β receptor inhibitors from Johnson & Johnson, and an analogue or derivative thereof. 625. The device of item 527 wherein the agent is a TNFα antagonist or TACE inhibitor selected from the group consisting of adalimumab (CAS No. 331731-18-1) (Cambridge Antibody Technology), AGIX-4207 (AtheroGenics), AGT-1 (Advanced Biotherapy), an antiinflammatory from Borean Pharma, Cellzome, or Paradigm Therapeutics, anti-inflammatory vaccine (TNF-alpha kinoid) from Neovacs, humanized anti-TNF antibody or an anti-TNF MAb (CB0006) Celltech (UCB), apratastat (CAS No. 287445-51-0) (Wyeth), BMS-561392 (Bristol-Myers Squibb), BN- 006 (Bone), certolizumab pegol (CAS No. 428863-50-7 or CH-138 (UCB), cilomilast (CAS No. 153259-65-5) (GlaxoSmithKline), CR-1 (Nuada Pharmaceuticals), CRx-119 (CombinatoRx), D-5410 (UCB), dacopafant (CAS No. 125372-33-0) (Sanofi-Aventis), dersalazine (CAS No. 188913-57- 7/188913-58-8) (Uriach), etanercept (CAS No. 185243-69-0) (Amgen), ethyl pyruvate (Critical (Critical Therapeutics), golimumab (CAS No. 476181-74-5) (Johnson & Johnson), hormono-immunotherapy from Ipsen, CDP571 (e.g., Humicade from UCB), IC-485 (ICOS), infliximab (CAS No. 170277-31-3) (Johnson & Johnson), IP-751 (Manhattan Pharmaceuticals), ISIS-104838 (CAS No. 250755-32-9) (ISIS Pharmaceuticals), lenalidomide (CAS No. 191732-72-6) (Celgene), lentinan (CAS No. 37339-90-5) (Ajinomoto), MDL- 201112 (CAS No. 142130-73-2) (Sanofi-Aventis), medroxyprogesterone (CAS No. 520-85-4) (InKine Pharmaceutical), N-acetylcysteine (CAS No. 616-91-1) (Zambon), NBE-P2 (DIREVO Biotech), nerelimomab (CAS No. 162774-06-3) (Chiron), OM-294DP (OM PHARMA), onercept (CAS No. 199685-57-9) (Yeda), PASSTNF-alpha (Verigen), pentoxifylline or oxypentifylline (Sanofi-Aventis), Pharmaprojects No. 4091, 4241, 4295, or 4488 (Sanofi-Aventis), Pharmaprojects No. 5480 (Amgen), Pharmaprojects No. 6749 (Cengent), pirfenidone (CAS No. 53179-13-8) (MARNAC), RPR- 132294 (Sanofi-Aventis), S5 (F002) (Fulcrum Pharmaceuticals), simvastatin (CAS No. 79902-63-9) (Merck & Co), STA-6292 (Synta Pharmaceuticals), tacrolimus (CAS No. 104987-11-3) (Fujisawa LifeCycle Pharma), talactoferrin alfa (CAS No. 308240-58-6) (Agennix), thalidomide (CAS No. 50-35-1) (Celgene), TNF antagonists form ProStrakan, and Synergen, TNF inhibitors (Amgen), TNF-alpha antagonists from Dynavax Technologies and Jerina AG (Germany), TNF-alpha inhibitors from IBFB Pharma and Xencor (Xencor), torbafylline (CAS No. 105102-21-4) (Sanofi-Aventis), UR-1505 (Uriach), VT-346 (Viron Therapeutics), YSIL6 (Y1S Therapeutics), YSTH2 (Y's Therapeutics), NPI-1302a-3 (Nereus Pharmaceuticals, a TNF antagonist from Jerina AG (Germany), dersalazine, and an analogue or derivative thereof.
626. The device of item 527 wherein the agent is a Toll receptor inhibitor.
627. The device of item 527 wherein the agent is a tubulin antagonist.
628. The device of item 527 wherein the agent is a tyrosine kinase inhibitor selected from the group consisting of SU-011248, SUTENT from Pfizer Inc. (New York, NY), BMS-354825, PN-355 (Paracelsian Pharmaceuticals), AGN-199659 (Allergan), AAL-993 or ABP-309 (Novartis), adaphostin (NIH), AEE-788 (Novartis), AG-013736 (OSI Pharmaceuticals), AG-13736 (Pfizer), ALT-110 (Alteris Therapeutics), AMG-706 (Amgen), anticancer MAbs from Xencor, anti-EGFrvlll MAbs from Abgenix, anti-HER2 MAb from Abiogen, AZD-2171 or AZD-9935 (AstraZeneca), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), CEP-5214 (Cephalon), CEP- 7055 (Cephalon), cetuximab (ImClone Systems), CHIR-200131 and CHIR- 258 (Chiron), CP-547632 (OSI Pharmaceuticals), CP-724714 (Pfizer), CT- 301 (Creabilis Therapeutics), D-69491 (Baxter International), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EGFR/ErbB2 inhibitors from Array BioPharma, erlotinib (CAS No. 183319-69-9) (OSI Pharmaceuticals), EXEL-2880 (Exelixis), FK-778 (Sanofi-Aventis) , gefitinib (CAS No. 184475-35-2) (AstraZeneca), GW-2286 or GW-654652 (GlaxoSmithKline), her2/neu antigen from AlphaVax, HER-2/neu inhibitor from Generex, Herzyme (Medipad) (Sirna Therapeutics), HKI-272 (Wyeth), HuMax-EGFr (Genmab), idronoxil (CAS No. 81267-65-4) (Novogen), IGF-1 inhibitors from Ontogen, IMC-11F8 (ImClone Systems), kahalalide F (CAS No. 149204-42-2) (PharmaMar), KDR inhibitor from LG Life Sciences, KDR inhibitors from Abbott Laboratories, KDR kinase inhibitors (UCB), Kdr kinase inhibitors from Merck & Co, KRN-633 and KRN-951 (Kirin Brewery), KSB-102 (Xenova), lapatinib ditosylate (CAS No. 388082-78-8) (GlaxoSmithKline), matuzumab (Merck KGaA), MDX-214 (Medarex), ME-103 (Pharmexa), MED-A300 (Gerolymatos), MNAC-13 (Lay Line Genomics), nimotuzumab (Center of Molecular Immunology), NSC-330507 or NSC-707545 (NIH), NV-50 (Novogen), OSI-930 (OSI Pharmaceuticals), panitumumab (Abgenix), pelitinib (CAS No. 287933-82-7) (Wyeth), pertuzumab (CAS No. 380610-27- 5) (Genentech), Pharmaprojects No. 3985 (Sanofi-Aventis), prostate cancer therapeutics from Sequenom (SQPC35, SQPC36, SQPC90), removab and remόxab (Tfion Pharma), RG-13022 (CAS No. 136831-48-6); RG-13291 (CAS No. 138989-50-1), or RG-14620 (CAS No. 136831-49-7) (Sanofi- Aventis), RM-6427 (Romark), RNAi breast cancer therapy from Benitec, RP 53801 (CAS No. 125882-88-4) (Sanofi-Aventis), sorafenib tosylate (Bayer), SLM 1657 (Pfizer), Tie-2 antagonists from Semaia (Hybrigenics), Tie-2 inhibitors from Ontogen, trastuzumab (CAS No. 180288-69-1) (Genentech), tyrosine kinase inhibitors from Sanofi-Aventis, U3-1287, U3-1565, U3-1784, U3-1800 (U3 Pharma), vatalanib (Novartis), VEGFR-2 kinase inhibitor from Bristol-Myers Squibb, XL-647 (Exelixis), ZD-6474 (AstraZeneca), ZK-CDK (Schering AG), an EGFR tyrosine kinase inhibitor, EKB-569 (Wyeth), and an analogue or derivative thereof.
629. The device of item 527 wherein the agent is a VEGF inhibitor. 630. The device of item 527 wherein the agent is a vitamin D receptor agonist.
631. The device of item 527 wherein the agent is ZD-6474 (an angiogenesis inhibitor).
632. The device of item 527 wherein the agent is AP-23573 (an mTOR inhibitor).
633. The device of item 527 wherein the agent is synthadotin (a tubulin antagonist).
634. The device of item 527 wherein the agent is S-0885 (a collagenase inhibitor).
635. The device of item 527 wherein the agent is aplidine (an elongation factor-1 alpha inhibitor).
636. The device of item 527 wherein the agent is ixabepilone (an epithilone).
637. The device of item 527 wherein the agent is IDN-5390 (an angiogenesis inhibitor).
638. The device of item 527 wherein the agent is SB- 2723005 (an angiogenesis inhibitor).
639. The device of item 527 wherein the agent is ABT-518 (an angiogenesis inhibitor). 640. The device of item 527 wherein the agent is combretastatin (an angiogenesis inhibitor).
641. The device of item 527 wherein the agent is anecortave acetate (an angiogenesis inhibitor).
642. The device of item 527 wherein the agent is SB-715992 (a kinesin antagonist).
643. The device of item 527 wherein the agent is temsirolimus (an mTOR inhibitor).
644. The device of item 527 wherein the agent is adalimumab (a TNFα antagonist).
645. The device of item 527, further comprising a polymer.
646. The device of item 527 wherein the anti-scarring agent inhibits adhesion between the device and a host into which the device is implanted.
647. The device of item 527 wherein the device delivers the anti-scarring agent locally to tissue proximate to the device.
648. The device of item 527, further comprising a coating, wherein the coating comprises the anti-scarring agent.
649. The device of item 527, further comprising a coating, wherein the coating is disposed on a surface of the device. 650. The device of item 527, further comprising a coating, wherein the coating directly contacts the device.
651. The device of item 527, further comprising a coating, wherein the coating indirectly contacts the device.
652. The device of item 527, further comprising a coating, wherein the coating partially covers the device.
653. The device of item 527, further comprising a coating, wherein the coating completely covers the device.
654. The device of item 527, further comprising a coating, wherein the coating is a uniform coating.
655. The device of item 527, further comprising a coating, wherein the coating is a non-uniform coating.
656. The device of item 527, further comprising a coating, wherein the coating is a discontinuous coating.
657. The device of item 527, further comprising a coating, wherein the coating is a patterned coating.
658. The device of item 527, further comprising a coating, wherein the coating has a thickness of 100 Dm or less.
659. The device of item 527, further comprising a coating, wherein the coating has a thickness of 10 Dm or less. 660. The device of item 527, further comprising a coating, wherein the coating adheres to the surface of the device upon deployment of the device.
661. The device of item 527, further comprising a coating, wherein the coating is stable at room temperature for a period of 1 year.
662. The device of item 527, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 0.0001 % to about 1% by weight.
663. The device of item 527, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 1% to about 10% by weight.
664. The device of item 527, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 10% to about 25% by weight.
665. The device of item 527, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 25% to about 70% by weight.
666. The device of item 527, further comprising a coating, wherein the coating further comprises a polymer.
667. The device of item 527, further comprising a first coating having a first composition and the second coating having a second composition. 668. The device of item 527, further comprising a first coating having a first composition and the second coating having a second composition, wherein the first composition and the second composition are different.
669. The device of item 527, further comprising a polymeric carrier, wherein the polymeric carrier comprises a copolymer.
670. The device of item 527, further comprising a polymeric carrier, wherein the polymeric carrier comprises a block copolymer.
671. The device of item 527, further comprising a polymeric carrier, wherein the polymeric carrier comprises a random copolymer.
672. The device of item 527, further comprising a polymeric carrier, wherein the polymeric carrier comprises a biodegradable polymer.
673. The device of item 527, further comprising a polymeric carrier, wherein the polymeric carrier comprises a non-biodegradable polymer.
674. The device of item 527, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrophilic polymer.
675. The device of item 527, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrophobic polymer.
676. The device of item 527, further comprising a polymeric carrier, wherein the polymeric carrier comprises a polymer having hydrophilic domains. 677. The device of item 527, further comprising a polymeric carrier, wherein the polymeric carrier comprises a polymer having hydrophobic domains.
678. The device of item 527, further comprising a polymeric carrier, wherein the polymeric carrier comprises a non-conductive polymer.
679. The device of item 527, further comprising a polymeric carrier, wherein the polymeric carrier comprises an elastomer.
680. The device of item 527, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrogel.
681. The device of item 527, further comprising a polymeric carrier, wherein the polymeric carrier comprises a silicone polymer.
682. The device of item 527, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrocarbon polymer.
683. The device of item 527, further comprising a polymeric carrier, wherein the polymeric carrier comprises a styrene-derived polymer.
684. The device of item 527, further comprising a polymeric carrier, wherein the polymeric carrier comprises a butadiene polymer.
685. The device of item 527, further comprising a polymeric carrier, wherein the polymeric carrier comprises a macromer.
686. The device of item 527, further comprising a polymeric carrier, wherein the polymeric carrier comprises a poly(ethylene glycol) polymer. 687. The device of item 527, further comprising a polymeric carrier, wherein the polymeric carrier comprises an amorphous polymer.
688. The device of item 527, further comprising a lubricious coating.
689. The device of item 527 wherein the anti-scarring agent is located within pores or holes of the device.
690. The device of item 527 wherein the anti-scarring agent is located within a channel, lumen, or divet of the device.
691. The device of item 527, further comprising a second pharmaceutically active agent.
692. The device of item 527, further comprising an antiinflammatory agent.
693. The device of item 527, further comprising an agent that inhibits infection.
694. The device of item 527, further comprising an agent that inhibits infection, wherein the agent is an anthracycline.
695. The device of item 527, further comprising an agent that inhibits infection, wherein the agent is doxorubicin.
696. The device of item 527, further comprising an agent that inhibits infection, wherein the agent is mitoxantrone. 697. The device of item 527, further comprising an agent that inhibits infection, wherein the agent is a fluoropyrimidine.
698. The device of item 527, further comprising an agent that inhibits infection, wherein the agent is 5-fluorouracil (5-FU).
699. The device of item 527, further comprising an agent that inhibits infection, wherein the agent is a folic acid antagonist.
700. The device of item 527, further comprising an agent that inhibits infection, wherein the agent is methotrexate.
701. The device of item 527, further comprising an agent that inhibits infection, wherein the agent is a podophylotoxin.
702. The device of item 527, further comprising an agent that inhibits infection, wherein the agent is etoposide.
703. The device of item 527, further comprising an agent that inhibits infection, wherein the agent is a camptothecin.
704. The device of item 527, further comprising an agent that inhibits infection, wherein the agent is a hydroxyurea.
705. The device of item 527, further comprising an agent that inhibits infection, wherein the agent is a platinum complex.
706. The device of item 527, further comprising an agent that inhibits infection, wherein the agent is cisplatin. 707. The device of item 527, further comprising an antithrombotic agent.
708. The device of item 527, further comprising a visualization agent.
709. The device of item 527, further comprising a visualization agent, wherein the visualization agent is a radiopaque material, wherein the radiopaque material comprises a metal, a halogenated compound, or a barium containing compound.
710. The device of item 527, further comprising a visualization agent, wherein the visualization agent is a radiopaque material, wherein the radiopaque material comprises barium, tantalum, or technetium.
711. The device of item 527, further comprising a visualization agent, wherein the visualization agent is a MRI responsive material.
712. The device of item 527, further comprising a visualization agent, wherein the visualization agent comprises a gadolinium chelate.
713. The device of item 527, further comprising a visualization agent, wherein the visualization agent comprises iron, magnesium, manganese, copper, or chromium.
714. The device of item 527, further comprising a visualization agent, wherein the visualization agent comprises an iron oxide compound. 715. The device of item 527, further comprising a visualization agent, wherein the visualization agent comprises a dye, pigment, or colorant.
716. The device of item 527, further comprising an echogenic material.
717. The device of item 527, further comprising an echogenic material, wherein the echogenic material is in the form of a coating.
718. The device of item 527 wherein the device is sterile.
719. The device of item 527 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device.
720. The device of item 527 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is connective tissue.
721. The device of item 527 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is muscle tissue.
722. The device of item 527 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is nerve tissue. 723. The device of item 527 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is epithelium tissue.
724. The device of item 527 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from the time of deployment of the device to about 1 year.
725. The device of item 527 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from about 1 month to 6 months.
726. The device of item 527 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from about 1 - 90 days.
727. The device of item 527 wherein the anti-scarring agent is released in effective concentrations from the device at a constant rate.
728. The device of item 527 wherein the anti-scarring agent is released in effective concentrations from the device at an increasing rate.
729. The device of item 527 wherein the anti-scarring agent is released in effective concentrations from the device at a decreasing rate.
730. The device of item 527 wherein the anti-scarring agent is released in effective concentrations from the composition comprising the anti-scarring agent by diffusion over a period ranging from the time of deployment of the device to about 90 days. 731. The device of item 527 wherein the anti-scarring agent is released in effective concentrations from the composition comprising the anti-scarring agent by erosion of the composition over a period ranging from the time of deployment of the device to about 90 days.
732. The device of item 527 wherein the device comprises about 0.01 Dg to about 10 Dg of the anti-scarring agent.
733. The device of item 527 wherein the device comprises about 10 Dg to about 10 mg of the anti-scarring agent.
734. The device of item 527 wherein the device comprises about 10 mg to about 250 mg of the anti-scarring agent.
735. The device of item 527 wherein the device comprises about 250 mg to about 1000 mg of the anti-scarring agent.
736. The device of item 527 wherein the device comprises about 1000 mg to about 2500 mg of the anti-scarring agent.
737. The device of item 527 wherein a surface of the device comprises less than 0.01 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
738. The device of item 527 wherein a surface of the device comprises about 0.01 Dg to about 1 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.]
739. The device of item 527 wherein a surface of the device comprises about 1 Dg to about 10 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied. 740. The device of item 527 wherein a surface of the device comprises about 10 Dg to about 250 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
741. The device of item 527 wherein a surface of the device comprises about 250 Dg to about 1000 Dg of the anti-scarring agent of anti- scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
742. The device of item 527 wherein a surface of the device comprises about 1000 Dg to about 2500 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
743. A method as in any one of items 527-742, wherein the device is an esophageal stent.
744. A method as in any one of items 527-742, wherein the device is a biliary stent.
745. A method as in any one of items 527-742, wherein the device is a colonic stent.
746. A method as in any one of items 527-742, wherein the device is a pancreatic stent.
747. A device, comprising a tracheal and bronchial stent (e.g., an implant) and an anti-scarring agent or a composition comprising an anti-scarring agent, wherein the agent inhibits scarring between the device and a host into which device is implanted. 748. The device of item 747 wherein the agent is an adensosine A2A receptor antagonist.
749. The device of item 747 wherein the agent is an AKT inhibitor.
750. The device of item 747 wherein the agent is an alpha 2 integrin antagonist, wherein the alpha 2 integrin antagonist is Pharmaprojects No. 5754 (Merck KgaA).
751. The device of item 747 wherein the agent an alpha 4 integrin antagonist.
752. The device of item 747 wherein the agent is an alpha 7 nicotinic receptor agonist.
753. The device of item 747 wherein the agent is an angiogenesis inhibitor selected from the group consisting of AG-12,958 (Pfizer), ATN-161 (Attention LLC), neovastat, an angiogenesis inhibitor from Jerina AG (Germany), NM-3 (Mercian), VGA-1155 (Taisho), FCE-26644 (Pfizer), FCE-26950 (Pfizer), FPMA (Meiji Daries), FR-111142 (Fujisawa), GGTI-298, GM-1306 (Ligand), GPA-1734 (Novartis), NNC-47-0011 (Novo Nordisk), herbamycin (Nippon Kayaku), lenalidomide (Celegene), IP-10 (NIH), ABT-828 (Abbott), KIN-841 (Tokushima University, Japan), SF-1126 (Semafore Pharmaceuticals), laminin technology (NIH), CHIR-258 (Chiron), NVP-AEW541 (Novartis), NVP-AEW541 (Novartis), Vt16907 (Alchemia), OXI-8007 (Oxigene), EG-3306 (Ark Therapeutics), Maspin (Arriva), ABT- 567 (Abbott), PPI-2458 (Praecis Pharmaceuticals), CC-5079, CC-4089 (Celgene), HIF-1 alpha inhibitors (Xenova), S-247 (Pfizer), AP-23573 (Ariad), AZD-9935 (Astra Zeneca), mebendazole (Introgen Therapeutics), MetAP-2 inhibitors (GlaxoSmithKline), AG-615 (Angiogene Pharmaceuticals), Tie-2 antagonists (Hybrigenics), NC-381 , CYC-381, NC-169, NC-219, NC-383, NC-384, NC-407 (Lorus Therapeutics), ATN-224 (Attenuon), ON-01370 (Onconova), Vitronectin antagonists (Amgen), SDX-103 (Salmedix), Vitronectin antagonists (Shire), CHP (Riemser), TEK (Amgen), Anecortave acetate (Alcon), T46.2 (Matrix Therapeutics), HG-2 (Heptagen), TEM antagonists (Genzyme), Oxi-4500 (Oxigene), ATN-161 (Attenuon), WX-293 (Wilex), M-2025 (Metris Therapeutics), Alphastatin (BioActa), YH-16 (Yantai Rongchang), BIBF-1120 (Boehringer Ingelheim), BAY-57-9352 (Bayer), AS- 1404 (Cancer Research Technology), SC-77964 (Pfizer), glycomimetics (BioTie Therapies), TIE-2 Inhibitors (Ontogen), DIMI, Octamer (Octamer), ABR-215050 (Active Biotech), ABT-518 (Abbott), KDR inhibitors (Abbott), BSF-466895 (Abbott), SCH-221153 (Schering-Plough), DAC:antiangiogenic (ConjuChem), TFPI (EntreMed), AZD-2171 (Astra-Zenaca), CDC-394 (Celgene), LY290293 (EIi Lilly), IDN-5390 (Indena), Kdr Kinase Inhibitors (Merck), CT-113020, CT-116433, CT-116563, CT-31890, CT-32228) (Cell Therapeutics), A-299620 (Abbott), TWEAK Inhibitor (Amgen), VEGF modulators (Johnson and Johnson), Tum-N53, tumstatin (Genzyme), Thios- 1, Thios-2 (Thios Pharmaceuticals), MV-6401 (Miravant Medical Technologies), Spisulosine (PharmaMar), CEP-7055 (Cephalon), AUV-201 (Auvation), LM-609 (EIi Lilly), SKF-106615 (AnorMED), Oglufanide disodium (Cytran), BW-114 (Pharminox), Calreticulin (NIH), WX-678 (Wilex), SD-7784 (Pfizer), WX-UK1 (Wilex), SH-268 (Schering AG), 2-Me-PGA (Celgene), S- 137 (Pfizer), ZD-6126 (Angiogene Pharmaceuticals), SG-292 (SignalGen), Benefin (Lane Labs), A6, A36 (Angstrom), SB-2723005 (GlaxoSmithKline), SC-7 (Cell Therapeutics), ZEN-014 (AEterna Zentaris), 2-methoxyestradiol (EntreMed), NK-130119 (Nippon Kayaku), CC-10004 (Celgene), AVE- 8062A (Ajinomoto), Tacedinaline (Pfizer), Actinonin (Tokyo Metropolitan Institute of Medical Science), Lenalidomide (Celgene), VGA-1155, BTO-956 (SRI International), ER-68203-00 (Eisai), CT-6685 (UCB), JKC-362 (Phoenix Pharmaceuticals), DMI-3798 (DMI Biosciences, Angiomate (Ipsen), ZD-6474 (AstraZeneca), CEP-5214 (Cephalon), Canstatin (Genzyme), NM-3 (Mercian), Oridigm (MediQuest Therapeutics), Exherin (Adherex), BLS-0597 (Boston Life Sciences), PTC-299 (PTC Therapeutics), NPI-2358 (Nereus Pharmaceuticals), CGP-79787 (Novartis), AEE-788 (Novartis), CKD-732 (Chong Kun Dang), CP-564959 (OSI Pharmaceuticals), CM-101 (CarboMed), CT-2584, CT3501 (Cell Therapeutics), combretastatin and analogues and derivatives thereof (Oxigene), Rebimastat (Bristol-Meyers Squibb), Dextrin 2-sulfate (ML Laboratories), Cilengitide (Merk KGaA), NSC-706704 (Pharminox), KRN- 951 (Kirin Brewery), Ukrain, NSC-631570 (Nowicky Pharma), Tecogalan sodium (Daiichi Pharmaceutical), Tz-93 (Tsumura), TBC-1635 (Encysive Pharmaceuticals), TAN-1120 (Takeda), Semaxanib (Pfizer), BDI-7800 (Biopharmacopae), SD-186, SD-983 (Bristol-Meyers Squibb), SB-223245 (GlaxoSmithKline), SC-236 (Pfizer), RWJ-590973 (Johnson and Johnson), ILX-1850 (Genzyme), SC-68488, S-836 (Pfizer), CG-55069-11 (CuraGen), Ki-23057 (Kirin Brewery), CCX-700 (Chemoentryx), Pegaptanib octasodium (Giled Sciences), ANGIOCOL (available from Biostratum Inc.), or an analogue or derivative thereof.
754. The device of item 747 wherein the agent is an apoptosis antagonist.
755. The device of item 747 wherein the agent is an apoptosis activator.
756. The device of item 747 wherein the agent is a beta 1 integrin antagonist.
757. The device of item 747 wherein the agent is a beta tubulin inhibitor. 758. The device of item 747 wherein the agent is a blocker of enzyme production in Hepatitis C.
759. The device of item 747 wherein the agent is a Bruton's tyrosine kinase inhibitor.
760. The device of item 747 wherein the agent is a calcineurin inhibitor.
761. The device of item 747 wherein the agent is a caspase 3 inhibitor.
762. The device of item 747 wherein the agent is a CC chemokine receptor antagonist.
763. The device of item 747 wherein the agent is a cell cycle inhibitor selected from the group consisting of SNS-595 (Sunesis), synthadotin, KRX-0403, and an analogue or derivative thereof.
764. The device of item 747 wherein the agent is a cathepsin B inhibitor.
765. The device of item 747 wherein the agent is a cathepsin K inhibitor, wherein the cathepsin K inhibitor is 462795 (GlaxoSmithKline), INPL-022-D6 (Amura Therapeutics), an analogue or derivative thereof).
766. The device of item 747 wherein the agent is a cathepsin L inhibitor.
767. The device of item 747 wherein the agent is a CD40 antagonist. 768. The device of item 747 wherein the agent is a chemokine receptor agonist.
769. The device of item 747 wherein the agent is a chymase inhibitor.
770. The device of item 747 wherein the agent is a collagenase antagonist.
771. The device of item 747 wherein the agent is a CXCR antagonist.
772. The device of item 747 wherein the agent is a cyclin dependent kinase inhibitor selected from the group consisting of a CDK-1 inhibitor, a CDK-2 inhibitor, a CDK- 4 inhibitor, a CDK-6 inhibitor, a CAK1 inhibitor from GPC Biotech and Bristol-Myers Squibb, RGB-2861-99 (GPC Biotech), an anticancer agent from Astex Technology, a CAK1 inhibitor from GPC Biotech, a CDK inhibitor from Sanofi-Aventis, a CDK1/CDK2 inhibitor from Amgen, a CDK2 inhibitor from SUGEN-2 (Pfizer), a hearing loss therapy agent (Sound Pharmaceuticals), PD-0332991 (Pfizer), RGB-286199 (GPC Biotech), Ro-0505124 (Hoffmann-La Roche), a Ser/Thr kinase inhibitor from Lilly (EIi Lilly), CVT-2584 (CAS No. 199986-75-9) (CV Therapeutics), and an analogue or derivative thereof.
773. The device of item 747 wherein the agent is a cyclooxygenase 1 inhibitor.
774. The device of item 747 wherein the agent is a DHFR inhibitor. 775. The device of item 747 wherein the agent is a dual integrin inhibitor.
776. The device of item 747 wherein the agent is an elastase inhibitor.
777. The device of item 747 wherein the agent is an elongation factor-1 alpha inhibitor.
778. The device of item 747 wherein the agent is an endothelial growth factor antagonist.
779. The device of item 747 wherein the agent is an endothelial growth factor receptor kinase inhibitor selected from the group consisting of sorafenib tosylate (Bayer), AAL-993 (Novartis), ABP-309 (Novartis), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), E- 7080 (Eisai), EG-3306 (Ark Therapeutics), EXEL-2880 (Exelixis), GW- 654652 (GlaxoSmithKline), a KDR inhibitor from LG Life Sciences, CT-6685 and CT-6729 (UCB)1 KRN-633 and KRN-951 (Kirin Brewery), OSI-930 (OSI Pharmaceuticals), SP-5.2 (Supratek Pharma), SU-11657 (Pfizer), a Tie-2 antagonist (Hybrigenics), a VEGFR-2 kinase inhibitor (Bristol-Myers Squibb), XL-647 (Exelixis), a KDR inhibitor from Abbott Laboratories, sorafenib tosylate, and an analogue or derivative thereof.
780. The device of item 747 wherein the agent is an endotoxin antagonist.
781. The device of item 747 wherein the agent is an epothilone and tubulin binder. 782. The device of item 747 wherein the agent is an estrogen receptor antagonist.
783. The device of item 747 wherein the agent is an FGF inhibitor.
784. The device of item 747 wherein the agent is a farnexyl transferase inhibitor.
785. The device of item 747 wherein the agent is farnesyltransferase inhibitor selected from the group of A-197574 (Abbott), a famesyltransferase inhibitor from Servier, FPTIII (Strathclyde Institute for Drug R), LB-42908 (LG Life Sciences), Pharmaprojects No. 5063 (Genzyme), Pharmaprojects No. 5597 (Ipsen), Yissum Project No. B-1055 (Yissum), and an analogue or derivative thereof.
786. The device of item 747 wherein the agent is an FLT-3 kinase inhibitor.
787. The device of item 747 wherein the agent is an FGF receptor kinase inhibitor.
788. The device of item 747 wherein the agent is a fibrinogen antagonist selected from the group consisting of AUV-201 (Auvation), MG-13926 (Sanofi-Aventis), plasminogen activator (CAS No. 105913-11-9) (from Sanofi-Aventis or UCB), plasminogen activator-2 (tPA- 2) (Sanofi-Aventis), pro-urokinase (CAS No. 82657-92-9) (Sanofi-Aventis), and an analogue or derivative thereof.
789. The device of item 747 wherein the agent is a heat shock protein 90 antagonist selected from the group consisting of SRN-005 (Sirenade), geldanamycin, NSC-33050 (17-allylaminogeldanamycin; 17- AAG), ^-dimethylaminoethylamino-^-demethoxy-geldanamycin (17- DMAG), rifabutin (rifamycin XIV, r,4-didehydro-1-deoxy-1 ,4-dihydro-5'-(2- methylpropyl)-1-oxo-), and an analogue or derivative thereof.
790. The device of item 747 wherein the agent is a histone deacetylase inhibitor.
791. The device of item 747 wherein the agent is an HMGCoA reductase inhibitor selected from the group consisting of an atherosclerosis therapeutic from Lipid Sciences, ATI-16000 (ARYx Therapeutics), KS-01-019 (Kos Pharmaceuticals), Pharmaprojects No. 2197 (Sanofi-Aventi), RP 61969 (Sanofi-Aventis), and an analogue or derivative thereof.
792. The device of item 747 wherein the agent is an ICAM inhibitor.
793. The device of item 747 wherein the agent is an IL, ICE and IRAK antagonist, wherein the antagonist is a CJ-14877, CP-424174 (Pfizer), NF-61 (Negma-Lerads), and an analogue or derivative thereof.
794. The device of item 747 wherein the agent is an IL-2 inhibitor.
795. The device of item 747 wherein the agent is an immunosuppressant selected from the group consisting of teriflunomide (Sanofi Aventis), chlorsulfaquinoxalone (NSC-339004), chlorsulfaquinoxalone sulfate, CS-712 (Sankyo), ismomultin alfa (CAS No. 457913-93-8) (Akzo Nobel), antiallergics from GenPat.77, antiinflammatories or AT-005 (Androclus Therapeutics), autoimmune disease therapeutics from EpiVax, BN-007 (Bone), budesonide (CAS No. 51333-22- 3) (MAP Pharmaceuticals), CO-14 (Genzyme), edratide (CAS No. 433922- 67-9) (Teva), EP-314 (Enanta), eprovafen (CAS No. 101335-99-3) (Sanofi- Aventis), HWA-131 (CAS No. 118788-41-3) (Sanofi-Aventis), immunomodulators from MerLion Pharmaceuticals, immunosuppressives from Alchemia, IPL-12 (Inflazyme), MDL-9563 (CAS No. 27086-86-8) (Sanofi-Aventis), Pharmaprojects No. 2330 (Sanofi-Aventis), Pharmaprojects No. 6426 (Abgenix), PXS-25 (Pharmaxis), rosmarinic acid (CAS No. 20283-92-5) (Sanofi-Aventis), RP 42927 or RP 54745 (CAS No. 135330-08-4) (Sanofi-Aventis), SGN-35 (Seattle Genetics), ST-1959 (Sigma-Tau), type I diabetes therapy from SYNX Pharma, UNIL-88 (Debiopharm), VP-025 (Vasogen), VR-694 (Vectura), PRTX-001 (Protalex), and an analogue or derivative thereof.
796. The device of item 747 wherein the agent is an IMPDH (inosine monophosphate).
797. The device of item 747 wherein the agent is an integrin antagonist.
798. The device of item 747 wherein the agent is an interleukin antagonist.
799. The device of item 747 wherein the agent is an inhibitor of type III receptor tyrosine kinase.
800. The device of item 747 wherein the agent is an irreversible inhibitor of enzyme methionine aminopeptidase type 2.
801. The device of item 747 wherein the agent is an isozyme selective delta protein kinase C inhibitor. 802. The device of item 747 wherein the agent a JAK3 enzyme inhibitor.
803. The device of item 747 wherein the agent is a JNK inhibitor.
804. The device of item 747 wherein the agent is a kinase inhibitor.
805. The device of item 747 wherein the agent is kinesin antagonist.
806. The device of item 747 wherein the agent is a kinesin antagonist.
807. The device of item 747 wherein the agent is a leukotriene inhibitor and antagonist selected from the group consisting of ambicromil (CAS No. 58805-38-2) (Sanofi-Aventis), amelubant (CAS No. 346735-24-8) (Boehringer Ingelheim), DW-1141 (Dong Wha), ebselen (Daiichi Pharmaceutical), ibudilast (Kyorin), leucotriene inhibitors from Sanofi-Aventis, lymphotoxin-beta receptor (LT-D) from Biogen Idee, Pharmaprojects No. 1535 and 2728 (CAS No. 119340-33-9) (Sanofi- Aventis), R-112 (Rigel), Rev-5367 (CAS No. 92532-05-3) (Sanofi-Aventis), RG-14893 (CAS No. 141835-49-6) (Sanofi-Aventis), RG-5901-A (CAS No. 101910-24-1), 92532-23-5, RP 66153 (CAS No. 142422-79-5), RP 66364 (CAS No.186916-92-5), RP 69698 (CAS No. 141748-00-7) (Sanofi-Aventis), SC-411930 (Pfizer), SC-41930 (CAS No. 120072-59-5) (Pfizer), SC-50605 (CAS No. 138828-39-4) (Pfizer), SC-51146 (CAS No. 141059-52-1), SC- 53228 (CAS No. 153633-01-3) (Pfizer), spaglumic acid (ZY-15106) (CAS No.3106-85-2), 80619-64-3 (Novartis), tipredane (CAS No. 85197-77-9) (Bristol-Myers Squibb), U-75302 (CAS No. 119477-85-9) (Pfizer), and analogue or derivative thereof.
808. The device of item 747 wherein the agent is an MAP kinase inhibitor.
809. The device of item 747 wherein the agent is a matrix metalloproteinase inhibitor.
810. The device of item 747 wherein the agent is an MCP- CCR2 inhibitor.
811. The device of item 747 wherein the agent is an mTOR inhibitor.
812. The device of item 747 wherein the agent is an mTOR kinase inhibitor.
813. The device of item 747 wherein the agent is a microtubule inhibitor selected from the group consisting of antibody- maytansinoid conjugates from Biogen Idee, colchicines (MantiCore Pharmaceuticals), anticancer immunoconjugates from Johnson & Johnson, DIME from Octamer, gni-1f (GNI), huC242-DM4, huMy9-6-DM1 (ImmunoGen), IDN-5404 (Indena), IMO-098, IMOderm (Imotep), mebendazole (Introgen Therapeutics), microtubule poisons from Cambridge Enterprise, paclitaxel such as LOTAX from Aphios (CAS No. 33069-62-4), Genexol-PM from Samyang, Pharmaprojects No. 6383 (Tapestry Pharmaceuticals), RPR-112378 (Sanofi-Aventis), SGN-75 (Seattle Genetics), SPL-7435 (Starpharma), SSR-250411 (Sanofi-Aventis), trastuzumab-DM1 (Genentech), vinorelbine, and an analogue or derivative thereof. 814. The device of item 747 wherein the agent is an MIF inhibitor.
815. The device of item 747 wherein the agent is an MMP inhibitor.
816. The device of item 747 wherein the agent is a neurokinin (NK) antagonist selected from the group consisting of anthrotainin (CAS No. 148084-40-6) (Sanofi-Aventis), an IBS thereapeutic from ArQuIe, MDL-105212A (CAS No. 167261-60-1) (Ssanofi-Aventis), Pharmaprojects No. 2744, 3258 (CAS No. 139167-47-8) 4006, 4201, or 5986 (Sanofi-Aventis), RP 67580 (CAS No. 135911-02-3), SR-144190 (CAS No. 201152-86-5), SSR-240600, SSR-241586 (Sanofi-Aventis), TKA-457 (Novartis), vestipitant mesylate (CAS No. 334476-64-1) (GlaxoSmithKline), Win-64821 (Sanofi-Aventis), PRX-96026 (Predix Pharmaceuticals), and an analogue or derivative thereof.
817. The device of item 747 wherein the agent is an NF kappa B inhibitor selected from the group consisting of AVE-0545 or AVE- 0547 (Sanofi-Aventis), bortezomib (CAS No. 179324-69-7) (Millennium Pharmaceuticals), dexanabinol (CAS No. 112924-45-5) (Pharmos), dexlipotam (Viatris), Pharmaprojects No. 6283 (INDRA) (OXiGENE), IPL- 576092 (CAS No. 137571-30-3) (lnflazyme), NFKB decoy (Corgentech), NFKB decoy oligo (AnGes MG), NFKB's from Ariad, osteoporosis treatments or S5 (F005) from Fulcrum Pharmaceuticals, P61 (Phytopharm), R-flurbiprofen (CAS No. 5104-49-4) (Encore Pharmaceuticals), and an analogue or derivative thereof.
818. The device of item 747 wherein the agent is a nitric oxide agonist. 819. The device of item 747 wherein the agent is an ornithine decarboxylase inhibitor.
820. The device of item 747 wherein the agent is a p38 MAP kinase inhibitor selected from the group consisting of AZD-6703 (AstraZeneca), JX-401 (Jexys Pharmaceuticals), BMS-2 (Bristol-Myers Squibb), a p38 MAP kinase inhibitor from Novartis, a p38-alpha MAP kinase inhibitor from Amphora, Pharmaprojects No. 5704 (Pharmacopeia), RPR- 200765A (Sanofi-Aventis), SD-282 (Johnson & Johnson), TAK-715 (Takeda), and an analogue or derivative thereof.
821. The device of item 747 wherein the agent is a palmitoyl- protein thioesterase inhibitor.
822. The device of item 747 wherein the agent is a PDGF receptor kinase inhibitor selected from the group consisting of AAL-993, AMN-107, or ABP-309 (Novartis), AMG-706 (Amgen), BAY-57-9352 (Bayer), CDP-860 (UCB), E-7080 (Eisai), imatinib (CAS No. 152459-95-5) (Novartis), OSI-930 (OSI Pharmaceuticals), RPR-127963E (Sanofi-Aventis), RWJ-540973 (Johnson & Johnson), sorafenib tosylate (Bayer), SU-11657 (Pfizer), tandutinib (CAS No. 387867-13-2) (Millennium Pharmaceuticals), vatalanib (Novartis), ZK-CDK (Schering AG), and an analogue or derivative thereof.
823. The device of item 747 wherein the agent is (-)- halofenate (Metabolex), AMG-131 (Amgen), antidiabetics from Japan Tobacco, AZD-4619, AZD-8450, AZD-8677 (AstraZeneca), DRF-10945, balaglitazone (Dr Reddy's), CS-00088, CS-00098 (Chipscreen Biosciences), E-3030 (Eisai), etalocib (CAS No. 161172-51-6) (EIi Lilly), GSK-641597 (Ligand), GSK-677954 (GlaxoSmithKline), GW-409544 (Ligand), GW- 590735 (GlaxoSmithKline), K-111 (Hoffmann-La Roche), LY-518674 (EIi Lilly), LY-674 (Ligand), LY-929 (Ligand), MC-3001 , MC-3002 (MaxoCore Pharmaceuticals), metformin HCI + pioglitazone (CAS No. 1115-70-4 and 112529-15-4), ACTOPLUS MET from Andrx), muraglitazar (CAS No. 331741-94-7) (Bristol-Myers Squibb), naveglitazar (Ligand), oleoylethanolamide (Kadmus Pharmaceuticals), ONO-5129, pioglitazone hydrochloride (CAS No. 111025-46-8 and 112529-15-4) (Takeda), PLX-204 (Plexxikon), PPAR agonists from Genfit, PPAR delta agonists from EIi Lilly, PPAR-alpha agonists from CrystalGenomics, PPAR-gamma modulators and PPAR-β modulators from CareX, rosiglitazone maleate (CAS No. 122324- 73-4 or 155141-29-0) (GlaxoSmithKline), rosiglitazone maleate/glimepir (CAS No. 155141-29-0 and 93479-97-1), AVANDARYL, rosiglitazone maleate/metformin extend (CAS No. 155141-29-0 and 657-24-9), AVANDAMET, rosiglitazone maleate+metformin, AVANDAMET (GlaxoSmithKline), tesaglitazar (AstraZeneca), and an analogue or derivative thereof.
824. The device of item 747 wherein the agent is a phosphatase inhibitor.
825. The device of item 747 wherein the agent is a phosphodiesterase (PDE) inhibitor selected from the group consisting of avanafil (Tanabe Seiyaku), dasantafil (CAS No. 569351-91-3) (Schering- Plough), A-906119 (CAS No. 134072-58-5), DL-850 (Sanofi-Aventis), GRC- 3015, GRC-3566, GRC-3886 (Glenmark), HWA-153 (CAS No. 56395-66-5) (Sanofi-Aventis), hydroxypumafentrine (Altana), IBFB-130011, IBFB-14-016, IBFB-140301, IBFB-150007, IBFB-211913 (IBFB Pharma), L-826141 (Merck & Co), medorinone (CAS No. 88296-61-1) (Sanofi-Aventis), MEM- 1917 (Memory Pharmaceuticals), ND-1251 (Neuro3d), PDE inhibitors from ICOS, PDE IV inhibitors from Memory Pharmaceuticals and CrystalGenomics, Pharmaprojects No. 2742 and 6141 (Sanofi-Aventis), QAD-171 (Novartis), RHC-2963 (CAS No. 76993-12-9 and 76993-14-1), RPR-117658, RPR-122818 derivatives, SR-24870 , and RPR-132294 (Sanofi-Aventis), SK-350 (ln2Gen), stroke therapy agents from deCODE Genetics, TAS-203 (Taiho), tofimilast (CAS No. 185954-27-2) (Pfizer), UK- 371800 (Pfizer), WIN-65579 (CAS No. 158020-82-7) (Sanofi-Aventis), IBFB- 130020 (IBFB Pharma), OPC-6535 (CAS No. 145739-56-6) (Otsuka), a phosphodiesterase III inhibitor, enoximone, a phosphodiesterase IV inhibitor, fosfosal, Atopik (Barrier Therapeutics), triflusal, a phosphodiesterase V inhibitor, and an analogue or derivative thereof.
826. The device of item 747 wherein the agent is a PKC inhibitor.
827. The device of item 747 wherein the agent is a platelet activating factor antagonist.
828. The device of item 747 wherein the agent is a platelet- derived growth factor receptor kinase inhibitor.
829. The device of item 747 wherein the agent is a prolyl hydroxylase inhibitor.
830. The device of item 747 wherein the agent is a polymorphonuclear neutrophil inhibitor.
831. The device of item 747 wherein the agent is a protein kinase B inhibitor.
832. The device of item 747 wherein the agent is a protein kinase C stimulant. 833. The device of item 747 wherein the agent is a purine nucleoside analogue.
834. The device of item 747 wherein the agent is a purinoreceptor P2X antagonist.
835. The device of item 747 wherein the agent is a Raf kinase inhibitor.
836. The device of item 747 wherein the agent is a reversible inhibitor of ErbB1 and ErbB2.
837. The device of item 747 wherein the agent is a ribonucleoside triphosphate reductase inhibitor.
838. The device of item 747 wherein the agent is an SDF-1 antagonist.
839. The device of item 747 wherein the agent is a sheddase inhibitor.
840. The device of item 747 wherein the agent is an SRC inhibitor.
841. The device of item 747 wherein the agent is a stromelysin inhibitor.
842. The device of item 747 wherein the agent is an Syk kinase inhibitor. 843. The device of item 747 wherein the agent is a telomerase inhibitor.
844. The device of item 747 wherein the agent is a TGF beta inhibitor selected from the group consisting of pirfenidone (CAS No. 53179- 13-8) (MARNAC)1 tranilast (CAS No. 53902-12-8) (Kissei), IN-1130 (ln2Gen), mannose-6-phosphate (BTG), TGF-β antagonists from Inflazyme (Pharmaprojects No. 6075), TGF-β antagonists from Sydney, non-industrial source), TGF-β I receptor kinase inhibitors from EIi Lilly, TGF-β receptor inhibitors from Johnson & Johnson, and an analogue or derivative thereof.
845. The device of item 747 wherein the agent is a TNFα antagonist or TACE inhibitor selected from the group consisting of adalimumab (CAS No. 331731-18-1) (Cambridge Antibody Technology), AGIX-4207 (AtheroGenics), AGT-1 (Advanced Biotherapy), an antiinflammatory from Borean Pharma, Cellzome, or Paradigm Therapeutics, anti-inflammatory vaccine (TNF-alpha kinoid) from Neovacs, humanized anti-TNF antibody or an anti-TNF MAb (CB0006) Celltech (UCB), apratastat (CAS No. 287445-51-0) (Wyeth), BMS-561392 (Bristol-Myers Squibb), BN- 006 (Bone), certolizumab pegol (CAS No. 428863-50-7 or CH-138 (UCB), cilomilast (CAS No. 153259-65-5) (GlaxoSmithKline), CR-1 (Nuada Pharmaceuticals), CRx-119 (CombinatoRx), D-5410 (UCB), dacopafant (CAS No. 125372-33-0) (Sanofi-Aventis), dersalazine (CAS No. 188913-57- 7/188913-58-8) (Uriach), etanercept (CAS No. 185243-69-0) (Amgen), ethyl pyruvate (Critical (Critical Therapeutics), golimumab (CAS No. 476181-74-5) (Johnson & Johnson), hormono-immunotherapy from Ipsen, CDP571 (e.g., Humicade from UCB), IC-485 (ICOS), infliximab (CAS No. 170277-31-3) (Johnson & Johnson), IP-751 (Manhattan Pharmaceuticals), ISIS-104838 (CAS No. 250755-32-9) (ISIS Pharmaceuticals), lenalidomide (CAS No. 191732-72-6) (Celgene), lentinan (CAS No. 37339-90-5) (Ajinomoto), MDL- 201112 (CAS No. 142130-73-2) (Sanofi-Aventis), medroxyprogesterone (CAS No. 520-85-4) (InKine Pharmaceutical), N-acetylcysteine (CAS No. 616-91-1) (Zambon), NBE-P2 (DIREVO Biotech), nerelimomab (CAS No. 162774-06-3) (Chiron), OM-294DP (OM PHARMA), onercept (CAS No. 199685-57-9) (Yeda), PASSTNF-alpha (Verigen), pentoxifylline or oxypentifylline (Sanofi-Aventis), Pharmaprojects No. 4091, 4241, 4295, or 4488 (Sanofi-Aventis), Pharmaprojects No. 5480 (Amgen), Pharmaprojects No. 6749 (Cengent), pirfenidone (CAS No. 53179-13-8) (MARNAC), RPR- 132294 (Sanofi-Aventis), S5 (F002) (Fulcrum Pharmaceuticals), simvastatin (CAS No. 79902-63-9) (Merck & Co), STA-6292 (Synta Pharmaceuticals), tacrolimus (CAS No. 104987-11-3) (Fujisawa LifeCycle Pharma), talactoferrin alfa (CAS No. 308240-58-6) (Agennix), thalidomide (CAS No. 50-35-1) (Celgene), TNF antagonists form ProStrakan, and Synergen, TNF inhibitors (Amgen), TNF-alpha antagonists from Dynavax Technologies and Jerina AG (Germany), TNF-alpha inhibitors from IBFB Pharma and Xencor (Xencor), torbafylline (CAS No. 105102-21-4) (Sanofi-Aventis), UR-1505 (Uriach), VT-346 (Viron Therapeutics), YSIL6 (Y's Therapeutics), YSTH2 (Y's Therapeutics), NPI-1302a-3 (Nereus Pharmaceuticals, a TNF antagonist from Jerina AG (Germany), dersalazine, and an analogue or derivative thereof.
846. The device of item 747 wherein the agent is a Toll receptor inhibitor.
847. The device of item 747 wherein the agent is a tubulin antagonist.
848. The device of item 747 wherein the agent is a tyrosine kinase inhibitor selected from the group consisting of SU-011248, SUTENT from Pfizer Inc. (New York, NY), BMS-354825, PN-355 (Paracelsian Pharmaceuticals), AGN-199659 (Allergan), AAL-993 or ABP-309 (Novartis), adaphostin (NIH), AEE-788 (Novartis), AG-013736 (OSI Pharmaceuticals), AG-13736 (Pfizer), ALT-110 (Alteris Therapeutics), AMG-706 (Amgen), anticancer MAbs from Xencor, anti-EGFrvlll MAbs from Abgenix, anti-HER2 MAb from Abiogen, AZD-2171 or AZD-9935 (AstraZeneca), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), CEP-5214 (Cephalon), CEP- 7055 (Cephalon), cetuximab (ImCIone Systems), CHIR-200131 and CHIR- 258 (Chiron), CP-547632 (OSI Pharmaceuticals), CP-724714 (Pfizer), CT- 301 (Creabilis Therapeutics), D-69491 (Baxter International), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EGFR/ErbB2 inhibitors from Array BioPharma, erlotinib (CAS No. 183319-69-9) (OSI Pharmaceuticals), EXEL-2880 (Exelixis), FK-778 (Sanofi-Aventis) , gefitinib (CAS No. 184475-35-2) (AstraZeneca), GW-2286 or GW-654652 (GlaxoSmithKline), her2/neu antigen from AlphaVax, HER-2/neu inhibitor from Generex, Herzyme (Medipad) (Sima Therapeutics), HKI-272 (Wyeth), HuMax-EGFr (Genmab), idronoxil (CAS No. 81267-65-4) (Novogen), IGF-1 inhibitors from Ontogen, IMC-11F8 (ImCIone Systems), kahalalide F (CAS No. 149204-42-2) (PharmaMar), KDR inhibitor from LG Life Sciences, KDR inhibitors from Abbott Laboratories, KDR kinase inhibitors (UCB), Kdr kinase inhibitors from Merck & Co, KRN-633 and KRN-951 (Kirin Brewery), KSB-102 (Xenova), lapatinib ditosylate (CAS No. 388082-78-8) (GlaxoSmithKline), matuzumab (Merck KGaA), MDX-214 (Medarex), ME-103 (Pharmexa), MED-A300 (Gerolymatos), MNAC-13 (Lay Line Genomics), nimotuzumab (Center of Molecular Immunology), NSC-330507 or NSC-707545 (NIH), NV-50 (Novogen), OSI-930 (OSI Pharmaceuticals), panitumumab (Abgenix), pelitinib (CAS No. 287933-82-7) (Wyeth), pertuzumab (CAS No. 380610-27- 5) (Genentech), Pharmaprojects No. 3985 (Sanofi-Aventis), prostate cancer therapeutics from Sequenom (SQPC35, SQPC36, SQPC90), removab and remoxab (Trion Pharma), RG-13022 (CAS No. 136831-48-6), RG-13291 (CAS No. 138989-50-1), or RG-14620 (CAS No. 136831-49-7) (Sanofi- Aventis), RM-6427 (Romark), RNAi breast cancer therapy from Benitec, RP 53801 (CAS No. 125882-88-4) (Sanofi-Aventis), sorafenib tosylate (Bayer), SU-11657 (Pfizer), Tie-2 antagonists from Semaia (Hybrigenics), Tie-2 inhibitors from Ontogen, trastuzumab (CAS No. 180288-69-1) (Genentech), tyrosine kinase inhibitors from Sanofi-Aventis, U3-1287, U3-1565, U3-1784, U3-1800 (U3 Pharma), vatalanib (Novartis), VEGFR-2 kinase inhibitor from Bristol-Myers Squibb, XL-647 (Exelixis), ZD-6474 (AstraZeneca), ZK-CDK (Schering AG), an EGFR tyrosine kinase inhibitor, EKB-569 (Wyeth), and an analogue or derivative thereof.
849. The device of item 747 wherein the agent is a VEGF inhibitor.
850. The device of item 747 wherein the agent is a vitamin D receptor agonist.
851. The device of item 747 wherein the agent is ZD-6474 (an angiogenesis inhibitor).
852. The device of item 747 wherein the agent is AP-23573 (an mTOR inhibitor).
853. The device of item 747 wherein the agent is synthadotin (a tubulin antagonist).
854. The device of item 747 wherein the agent is S-0885 (a collagenase inhibitor).
855. The device of item 747 wherein the agent is aplidine (an elongation factor-1 alpha inhibitor).
856. The device of item 747 wherein the agent is ixabepilone (an epithilone). 857. The device of item 747 wherein the agent is IDN-5390 (an angiogenesis inhibitor).
858. The device of item 747 wherein the agent is SB- 2723005 (an angiogenesis inhibitor).
859. The device of item 747 wherein the agent is ABT-518 (an angiogenesis inhibitor).
860. The device of item 747 wherein the agent is combretastatin (an angiogenesis inhibitor).
861. The device of item 747 wherein the agent is anecortave acetate (an angiogenesis inhibitor).
862. The device of item 747 wherein the agent is SB-715992 (a kinesin antagonist).
863. The device of item 747 wherein the agent is temsirolimus (an mTOR inhibitor).
864. The device of item 747 wherein the agent is adalimumab (a TNFα antagonist).
865. The device of item 747, further comprising a polymer.
866. The device of item 747 wherein the anti-scarring agent inhibits adhesion between the device and a host into which the device is implanted. 867. The device of item 747 wherein the device delivers the anti-scarring agent locally to tissue proximate to the device.
868. The device of item 747, further comprising a coating, wherein the coating comprises the anti-scarring agent.
869. The device of item 747, further comprising a coating, wherein the coating is disposed on a surface of the device.
870. The device of item 747, further comprising a coating, wherein the coating directly contacts the device.
871. The device of item 747, further comprising a coating, wherein the coating indirectly contacts the device.
872. The device of item 747, further comprising a coating, wherein the coating partially covers the device.
873. The device of item 747, further comprising a coating, wherein the coating completely covers the device.
874. The device of item 747, further comprising a coating, wherein the coating is a uniform coating.
875. The device of item 747, further comprising a coating, wherein the coating is a non-uniform coating.
876. The device of item 747, further comprising a coating, wherein the coating is a discontinuous coating. 877. The device of item 747, further comprising a coating, wherein the coating is a patterned coating.
878. The device of item 747, further comprising a coating, wherein the coating has a thickness of 100 Dm or less.
879. The device of item 747, further comprising a coating, wherein the coating has a thickness of 10 Dm or less.
880. The device of item 747, further comprising a coating, wherein the coating adheres to the surface of the device upon deployment of the device.
881. The device of item 747, further comprising a coating, wherein the coating is stable at room temperature for a period of 1 year.
882. The device of item 747, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 0.0001% to about 1% by weight.
883. The device of item 747, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 1 % to about 10% by weight.
884. The device of item 747, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 10% to about 25% by weight.
885. The device of item 747, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 25% to about 70% by weight. 886. The device of item 747, further comprising a coating, wherein the coating further comprises a polymer.
887. The device of item 747, further comprising a first coating having a first composition and the second coating having a second composition.
888. The device of item 747, further comprising a first coating having a first composition and the second coating having a second composition, wherein the first composition and the second composition are different.
889. The device of item 747, further comprising a polymeric carrier, wherein the polymeric carrier comprises a copolymer.
890. The device of item 747, further comprising a polymeric carrier, wherein the polymeric carrier comprises a block copolymer.
891. The device of item 747, further comprising a polymeric carrier, wherein the polymeric carrier comprises a random copolymer.
892. The device of item 747, further comprising a polymeric carrier, wherein the polymeric carrier comprises a biodegradable polymer.
893. The device of item 747, further comprising a polymeric carrier, wherein the polymeric carrier comprises a non-biodegradable polymer.
894. The device of item 747, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrophilic polymer. 895. The device of item 747, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrophobic polymer.
896. The device of item 747, further comprising a polymeric carrier, wherein the polymeric carrier comprises a polymer having hydrophilic domains.
897. The device of item 747, further comprising a polymeric carrier, wherein the polymeric carrier comprises a polymer having hydrophobic domains.
898. The device of item 747, further comprising a polymeric carrier, wherein the polymeric carrier comprises a non-conductive polymer.
899. The device of item 747, further comprising a polymeric carrier, wherein the polymeric carrier comprises an elastomer.
900. The device of item 747, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrogel.
901. The device of item 747, further comprising a polymeric carrier, wherein the polymeric carrier comprises a silicone polymer.
902. The device of item 747, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrocarbon polymer.
903. The device of item 747, further comprising a polymeric carrier, wherein the polymeric carrier comprises a styrene-derived polymer.
904. The device of item 747, further comprising a polymeric carrier, wherein the polymeric carrier comprises a butadiene polymer. 905. The device of item 747, further comprising a polymeric carrier, wherein the polymeric carrier comprises a macromer.
906. The device of item 747, further comprising a polymeric carrier, wherein the polymeric carrier comprises a poly(ethylene glycol) polymer.
907. The device of item 747, further comprising a polymeric carrier, wherein the polymeric carrier comprises an amorphous polymer.
908. The device of item 747, further comprising a lubricious coating.
909. The device of item 747 wherein the anti-scarring agent is located within pores or holes of the device.
910. The device of item 747 wherein the anti-scarring agent is located within a channel, lumen, or divet of the device.]
911. The device of item 747, further comprising a second pharmaceutically active agent.
912. The device of item 747, further comprising an antiinflammatory agent.
913. The device of item 747, further comprising an agent that inhibits infection.
914. The device of item 747, further comprising an agent that inhibits infection, wherein the agent is an anthracycline. 915. The device of item 747, further comprising an agent that inhibits infection, wherein the agent is doxorubicin.
916. The device of item 747, further comprising an agent that inhibits infection, wherein the agent is mitoxantrone.
917. The device of item 747, further comprising an agent that inhibits infection, wherein the agent is a fluoropyrimidine.
918. The device of item 747, further comprising an agent that inhibits infection, wherein the agent is 5-fluorouracil (5-FU).
919. The device of item 747, further comprising an agent that inhibits infection, wherein the agent is a folic acid antagonist.
920. The device of item 747, further comprising an agent that inhibits infection, wherein the agent is methotrexate.
921. The device of item 747, further comprising an agent that inhibits infection, wherein the agent is a podophylotoxin.
922. The device of item 747, further comprising an agent that inhibits infection, wherein the agent is etoposide.
923. The device of item 747, further comprising an agent that inhibits infection, wherein the agent is a camptothecin.
924. The device of item 747, further comprising an agent that inhibits infection, wherein the agent is a hydroxyurea. 925. The device of item 747, further comprising an agent that inhibits infection, wherein the agent is a platinum complex.
926. The device of item 747, further comprising an agent that inhibits infection, wherein the agent is cisplatin.
927. The device of item 747, further comprising an antithrombotic agent.
928. The device of item 747, further comprising a visualization agent.
929. The device of item 747, further comprising a visualization agent, wherein the visualization agent is a radiopaque material, wherein the radiopaque material comprises a metal, a halogenated compound, or a barium containing compound. .
930. The device of item 747, further comprising a visualization agent, wherein the visualization agent is a radiopaque material, wherein the radiopaque material comprises barium, tantalum, or technetium.
931. The device of item 747, further comprising a visualization agent, wherein the visualization agent is a MRI responsive material.
932. The device of item 747, further comprising a visualization agent, wherein the visualization agent comprises a gadolinium chelate. 933. The device of item 747, further comprising a visualization agent, wherein the visualization agent comprises iron, magnesium, manganese, copper, or chromium.
934. The device of item 747, further comprising a visualization agent, wherein the visualization agent comprises an iron oxide compound.
935. The device of item 747, further comprising a visualization agent, wherein the visualization agent comprises a dye, pigment, or colorant.
936. The device of item 747, further comprising an echogenic material.
937. The device of item 747, further comprising an echogenic material, wherein the echogenic material is in the form of a coating.
938. The device of item 747 wherein the device is sterile.
939. The device of item 747 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device.
940. The device of item 747 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is connective tissue. 941. The device of item 747 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is muscle tissue.
942. The device of item 747 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is nerve tissue.
943. The device of item 747 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is epithelium tissue.
944. The device of item 747 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from the time of deployment of the device to about 1 year.
945. The device of item 747 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from about 1 month to 6 months.
946. The device of item 747 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from about 1 - 90 days.
947. The device of item 747 wherein the anti-scarring agent is released in effective concentrations from the device at a constant rate.
948. The device of item 747 wherein the anti-scarring agent is released in effective concentrations from the device at an increasing rate. 949. The device of item 747 wherein the anti-scarring agent is released in effective concentrations from the device at a decreasing rate.
950. The device of item 747 wherein the anti-scarring agent is released in effective concentrations from the composition comprising the anti-scarring agent by diffusion over a period ranging from the time of deployment of the device to about 90 days.
951. The device of item 747 wherein the anti-scarring agent is released in effective concentrations from the composition comprising the anti-scarring agent by erosion of the composition over a period ranging from the time of deployment of the device to about 90 days.
952. The device of item 747 wherein the device comprises about 0.01 Dg to about 10 Dg of the anti-scarring agent.
953. The device of item 747 wherein the device comprises about 10 Dg to about 10 mg of the anti-scarring agent.
954. The device of item 747 wherein the device comprises about 10 mg to about 250 mg of the anti-scarring agent.
955. The device of item 747 wherein the device comprises about 250 mg to about 1000 mg of the anti-scarring agent.
956. The device of item 747 wherein the device comprises about 1000 mg to about 2500 mg of the anti-scarring agent.
957. The device of item 747 wherein a surface of the device comprises less than 0.01 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied. 958. The device of item 747 wherein a surface of the device comprises about 0.01 Dg to about 1 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
959. The device of item 747 wherein a surface of the device comprises about 1 Dg to about 10 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
960. The device of item 747 wherein a surface of the device comprises about 10 ϋg to about 250 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
961. The device of item 747 wherein a surface of the device comprises about 250 Dg to about 1000 Dg of the anti-scarring agent of anti- scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
962. The device of item 747 wherein a surface of the device comprises about 1000 Dg to about 2500 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
963. A device of any one of items 747-962, wherein the device is a tracheal stent.
964. A device of any one of items 747-962, wherein the device is a bronchial stent.
965. A device of any one of items 747-962, wherein the device is a metallic tracheal stent. 966. A device of any one of items 747-962, wherein the device is a metallic broachial stent.
967. A device of any one of items 747-962, wherein the device is a polymeric tracheal stent.
968. A device of any one of items 747-962, wherein the device is a polymeric bronchial stent.
969. A device, comprising a genital urinary stent (e.g., an implant) and an anti-scarring agent or a composition comrpsing an anti- scarring agent, wherein the agent inhibits scarring between the device and a host into which the device is implanted.
970. The device of item 969 wherein the agent is an adensosine A2A receptor antagonist.
971. The device of item 969 wherein the agent is an AKT inhibitor.
972. The device of item 969 wherein the agent is an alpha 2 integrin antagonist, wherein the alpha 2 integrin antagonist is Pharmaprojects No. 5754 (Merck KgaA).
973. The device of item 969 wherein the agent an alpha 4 integrin antagonist.
974. The device of item 969 wherein the agent is an alpha 7 nicotinic receptor agonist. 975. The device of item 969 wherein the agent is an angiogenesis inhibitor selected from the group consisting of AG-12,958 (Pfizer), ATN-161 (Attention LLC), neovastat, an angiogenesis inhibitor from Jerina AG (Germany), NM-3 (Mercian), VGA-1155 (Taisho), FCE-26644 (Pfizer), FCE-26950 (Pfizer), FPMA (Meiji Daries), FR-111142 (Fujisawa), GGTI-298, GM-1306 (Ligand), GPA-1734 (Novartis), NNC-47-0011 (Novo Nordisk), herbamycin (Nippon Kayaku), Ienalidomide (Celegene), IP-10 (NIH), ABT-828 (Abbott), KIN-841 (Tokushima University, Japan), SF-1126 (Semafore Pharmaceuticals), laminin technology (NIH), CHIR-258 (Chiron), NVP-AEW541 (Novartis), NVP-AEW541 (Novartis), Vt16907 (Alchemia), OXI-8007 (Oxigene), EG-3306 (Ark Therapeutics), Maspin (Arriva), ABT- 567 (Abbott), PPI-2458 (Praecis Pharmaceuticals), CC-5079, CC-4089 (Celgene), HIF-1 alpha inhibitors (Xenova), S-247 (Pfizer), AP-23573 (Ariad), AZD-9935 (Astra Zeneca), mebendazole (Introgen Therapeutics), MetAP-2 inhibitors (GlaxoSmithKline), AG-615 (Angiogene Pharmaceuticals), Tie-2 antagonists (Hybrigenics), NC-381 , CYC-381, NC-169, NC-219, NC-383, NC-384, NC-407 (Lorus Therapeutics), ATN-224 (Attenuon), ON-01370 (Onconova), Vitronectin antagonists (Amgen), SDX-103 (Salmedix), Vitronectin antagonists (Shire), CHP (Riemser), TEK (Amgen), Anecortave acetate (Alcon), T46.2 (Matrix Therapeutics), HG-2 (Heptagen), TEM antagonists (Genzyme), Oxi-4500 (Oxigene), ATN-161 (Attenuon), WX-293 (Wilex), M-2025 (Metris Therapeutics), Alphastatin (BioActa), YH-16 (Yantai Rongchang), BIBF-1120 (Boehringer Ingelheim), BAY-57-9352 (Bayer), AS- 1404 (Cancer Research Technology), SC-77964 (Pfizer), glycomimetics (BioTie Therapies), TIE-2 Inhibitors (Ontogen), DIMI, Octamer (Octamer), ABR-215050 (Active Biotech), ABT-518 (Abbott), KDR inhibitors (Abbott), BSF-466895 (Abbott), SCH-221153 (Schering-Plough), DAC:antiangiogenic (ConjuChem), TFPI (EntreMed), AZD-2171 (Astra-Zenaca), CDC-394 (Celgene), LY290293 (EIi Lilly), IDN-5390 (Indena), Kdr Kinase Inhibitors (Merck), CT-113020, CT-116433, CT-116563, CT-31890, CT-32228) (Cell Therapeutics), A-299620 (Abbott), TWEAK Inhibitor (Amgen), VEGF modulators (Johnson and Johnson), Tυm-N53, tumstatin (Genzyme), Thios- 1 , Thios-2 (Thios Pharmaceuticals), MV-6401 (Miravant Medical Technologies), Spisulosine (PharmaMar), CEP-7055 (Cephalon), AUV-201 (Auvation), LM-609 (EIi Lilly), SKF-106615 (AnorMED), Oglufanide disodium (Cytran), BW-114 (Pharminox), Calreticulin (NIH), WX-678 (Wilex), SD-7784 (Pfizer), WX-UK1 (Wilex), SH-268 (Schering AG), 2-Me-PGA (Celgene), S- 137 (Pfizer), ZD-6126 (Angiogene Pharmaceuticals), SG-292 (SignalGen), Benefin (Lane Labs), A6, A36 (Angstrom), SB-2723005 (GlaxoSmithKline), SC-7 (Cell Therapeutics), ZEN-014 (AEterna Zentaris), 2-methoxyestradiol (EntreMed), NK-130119 (Nippon Kayaku), CC-10004 (Celgene), AVE- 8062A (Ajinomoto), Tacedinaline (Pfizer), Actinonin (Tokyo Metropolitan Institute of Medical Science), Lenalidomide (Celgene), VGA-1155, BTO-956 (SRI International), ER-68203-00 (Eisai), CT-6685 (UCB), JKC-362 (Phoenix Pharmaceuticals), DMI-3798 (DMI Biosciences, Angiomate (Ipsen), ZD-6474 (AstraZeneca), CEP-5214 (Cephalon), Canstatin (Genzyme), NM-3 (Mercian), Oridigm (MediQuest Therapeutics), Exherin. (Adherex), BLS-0597 (Boston Life Sciences), PTC-299 (PTC Therapeutics), NPI-2358 (Nereus Pharmaceuticals), CGP-79787 (Novartis), AEE-788 (Novartis), CKD-732 (Chong Kun Dang), CP-564959 (OSI Pharmaceuticals), CM-101 (CarboMed), CT-2584, CT3501 (Cell Therapeutics), combretastatin and analogues and derivatives thereof (Oxigene), Rebimastat (Bristol-Meyers Squibb), Dextrin 2-sulfate (ML Laboratories), Cilengitide (Merk KGaA), NSC-706704 (Pharminox), KRN- 951 (Kirin Brewery), Ukrain, NSC-631570 (Nowicky Pharma), Tecogalan sodium (Daiichi Pharmaceutical), Tz-93 (Tsumura), TBC-1635 (Encysive Pharmaceuticals), TAN-1120 (Takeda), Semaxanib (Pfizer), BDI-7800 (Biopharmacopae), SD-186, SD-983 (Bristol-Meyers Squibb), SB-223245 (GlaxoSmithKline), SC-236 (Pfizer), RWJ-590973 (Johnson and Johnson), ILX-1850 (Genzyme), SC-68488, S-836 (Pfizer), CG-55069-11 (CuraGen), Ki-23057 (Kirin Brewery), CCX-700 (Chemoentryx), Pegaptanib octasodium (Giled Sciences), ANGIOCOL (available from Biostratum Inc.), or an analogue or derivative thereof.
976. The device of item 969 wherein the agent is an apoptosis antagonist.
977. The device of item 969 wherein the agent is an apoptosis activator.
978. The device of item 969 wherein the agent is a beta 1 integrin antagonist.
979. The device of item 969 wherein the agent is a beta tubulin inhibitor.
980. The device of item 969 wherein the agent is a blocker of enzyme production in Hepatitis C.
981. The device of item 969 wherein the agent is a Bruton's tyrosine kinase inhibitor.
982. The device of item 969 wherein the agent is a calcineurin inhibitor.
983. The device of item 969 wherein the agent is a caspase 3 inhibitor.
984. The device of item 969 wherein the agent is a CC chemokine receptor antagonist. 985. The device of item 969 wherein the agent is a cell cycle inhibitor selected from the group consisting of SNS-595 (Sunesis), synthadotin, KRX-0403, and an analogue or derivative thereof.
986. The device of item 969 wherein the agent is a cathepsin B inhibitor.
987. The device of item 969 wherein the agent is a cathepsin K inhibitor, wherein the cathepsin K inhibitor is 462795 (GlaxoSmithKline), INPL-022-D6 (Amura Therapeutics), an analogue or derivative thereof).
988. The device of item 969 wherein the agent is a cathepsin L inhibitor.
989. The device of item 969 wherein the agent is a CD40 antagonist.
990. The device of item 969 wherein the agent is a chemokine receptor agonist.
991. The device of item 969 wherein the agent is a chymase inhibitor.
992. The device of item 969 wherein the agent is a collagenase antagonist.
993. The device of item 969 wherein the agent is a CXCR antagonist.
994. The device of item 969 wherein the agent is a cyclin dependent kinase inhibitor selected from the group consisting of a CDK-1 inhibitor, a CDK-2 inhibitor, a CDK- 4 inhibitor, a CDK-6 inhibitor, a CAK1 inhibitor from GPC Biotech and Bristol-Myers Squibb, RGB-286199 (GPC Biotech), an anticancer agent from Astex Technology, a CAK1 inhibitor from GPC Biotech, a CDK inhibitor from Sanofi-Aventis, a CDK1/CDK2 inhibitor from Amgen, a CDK2 inhibitor from SUGEN-2 (Pfizer), a hearing loss therapy agent (Sound Pharmaceuticals), PD-0332991 (Pfizer), RGB-286199 (GPC Biotech), Ro-0505124 (Hoffmann-La Roche), a Ser/Thr kinase inhibitor from Lilly (EIi Lilly), CVT-2584 (CAS No. 199986-75-9) (CV Therapeutics), and an analogue or derivative thereof.
995. The device of item 969 wherein the agent is a cyclooxygenase 1 inhibitor.
996. The device of item 969 wherein the agent is a DHFR inhibitor.
997. The device of item 969 wherein the agent is a dual integrin inhibitor.
998. The device of item 969 wherein the agent is an elastase inhibitor.
999. The device of item 969 wherein the agent is an elongation factor-1 alpha inhibitor.
1000. The device of item 969 wherein the agent is an endothelial growth factor antagonist.
1001. The device of item 969 wherein the agent is an endothelial growth factor receptor kinase inhibitor selected from the group consisting of sorafenib tosylate (Bayer), AAL-993 (Novartis), ABP-309 (Novartis), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), E- 7080 (Eisai), EG-3306 (Ark Therapeutics), EXEL-2880 (Exelixis), GW- 654652 (GlaxoSmithKline), a KDR inhibitor from LG Life Sciences, CT-6685 and CT-6729 (UCB), KRN-633 and KRN-951 (Kirin Brewery), OSI-930 (OSI Pharmaceuticals), SP-5.2 (Supratek Pharma), SU-11657 (Pfizer), a Tie-2 antagonist (Hybrigenics), a VEGFR-2 kinase inhibitor (Bristol-Myers Squibb), XL-647 (Exelixis), a KDR inhibitor from Abbott Laboratories, sorafenib tosylate, and an analogue or derivative thereof.
1002. The device of item 969 wherein the agent is an endotoxin antagonist.
1003. The device of item 969 wherein the agent is an epothilone and tubulin binder.
1004. The device of item 969 wherein the agent is an estrogen receptor antagonist.
1005. The device of item 969 wherein the agent is an FGF inhibitor.
1006. The device of item 969 wherein the agent is a farnexyl transferase inhibitor.
1007. The device of item 969 wherein the agent is farnesyltransferase inhibitor selected from the group of A-197574 (Abbott), a famesyltransferase inhibitor from Servier, FPTIII (Strathclyde Institute for Drug R), LB-42908 (LG Life Sciences), Pharmaprojects No. 5063 (Genzyme), Pharmaprojects No. 5597 (Ipsen), Yissum Project No. B-1055 (Yissum), and an analogue or derivative thereof. 1008. The device of item 969 wherein the agent is an FLT-3 kinase inhibitor.
1009. The device of item 969 wherein the agent is an FGF receptor kinase inhibitor.
1010. The device of item 969 wherein the agent is a fibrinogen antagonist selected from the group consisting of AUV-201 (Auvation), MG-13926 (Sanofi-Aventis), plasminogen activator (CAS No. 105913-11-9) (from Sanofi-Aventis or UCB), plasminogen activator-2 (tPA- 2) (Sanofi-Aventis), pro-urokinase (CAS No. 82657-92-9) (Sanofi-Aventis), and an analogue or derivative thereof.
1011. The device of item 969 wherein the agent is a heat shock protein 90 antagonist selected from the group consisting of SRN-005 (Sirenade), geldanamycin, NSC-33050 (17-allylaminogeldanamycin; 17- AAG), 17-dimethylaminoethylamino-17-demethoxy-geldanamycin (17- DMAG), rifabutin (rifamycin XIV, 1',4-didehydro-1-deoxy-1 ,4-dihydro-5'-(2- methyipropyl)-1-oxo-), and an analogue or derivative thereof.
1012. The device of item 969 wherein the agent is a histone deacetylase inhibitor.
1013. The device of item 969 wherein the agent is an HMGCoA reductase inhibitor selected from the group consisting of an atherosclerosis therapeutic from Lipid Sciences, ATI-16000 (ARYx Therapeutics), KS-01-019 (Kos Pharmaceuticals), Pharmaprojects No. 2197 (Sanofi-Aventi), RP 61969 (Sanofi-Aventis), and an analogue or derivative thereof. 1014. The device of item 969 wherein the agent is an ICAM inhibitor.
1015. The device of item 969 wherein the agent is an IL, ICE and IRAK antagonist, wherein the antagonist is a CJ-14877, CP-424174 (Pfizer), NF-61 (Negma-Lerads), and an analogue or derivative thereof.
1016. The device of item 969 wherein the agent is an IL-2 inhibitor.
1017. The device of item 969 wherein the agent is an immunosuppressant selected from the group consisting of teriflunomide (Sanofi Aventis), chlorsulfaquinoxalone (NSC-339004), chlorsulfaquinoxalone sulfate, CS-712 (Sankyo), ismomultin alfa (CAS No. 457913-93-8) (Akzo Nobel), antiallergics from GenPat77, antiinflammatories or AT-005 (Androclus Therapeutics), autoimmune disease therapeutics from EpiVax, BN-007 (Bone), budesonide (CAS No. 51333-22- 3) (MAP Pharmaceuticals), CO-14 (Genzyme), edratide (CAS No. 433922- 67-9) (Teva), EP-314 (Enanta), eprovafen (CAS No. 101335-99-3) (Sanofi- Aventis), HWA-131 (CAS No. 118788-41-3) (Sanofi-Aventis), immunomodulators from MerLion Pharmaceuticals, immunosuppressives from Alchemia, IPL-12 (Inflazyme), MDL-9563 (CAS No. 27086-86-8) (Sanofi-Aventis), Pharmaprojects No. 2330 (Sanofi-Aventis), Pharmaprojects No. 6426 (Abgenix), PXS-25 (Pharmaxis), rosmarinic acid (CAS No. 20283-92-5) (Sanofi-Aventis), RP 42927 or RP 54745 (CAS No. 135330-08-4) (Sanofi-Aventis), SGN-35 (Seattle Genetics), ST-1959 (Sigma-Tau), type I diabetes therapy from SYNX Pharma, UNIL-88 (Debiopharm), VP-025 (Vasogen), VR-694 (Vectura), PRTX-001 (Protalex), and an analogue or derivative thereof. 1018. The device of item 969 wherein the agent is an IMPDH (inosine monophosphate).
1019. The device of item 969 wherein the agent is an integrin antagonist.
1020. The device of item 969 wherein the agent is an interleukin antagonist.
1021. The device of item 969 wherein the agent is an inhibitor of type III receptor tyrosine kinase.
1022. The device of item 969 wherein the agent is an irreversible inhibitor of enzyme methionine aminopeptidase type 2.
1023. The device of item 969 wherein the agent is an isozyme selective delta protein kinase C inhibitor.
1024. The device of item 969 wherein the agent a JAK3 enzyme inhibitor.
1025. The device of item 969 wherein the agent is a JNK inhibitor.
1026. The device of item 969 wherein the agent is a kinase inhibitor.
1027. The device of item 969 wherein the agent is kinesin antagonist. 1028. The device of item 969 wherein the agent is a kinesin antagonist.
1029. The device of item 969 wherein the agent is a leukotriene inhibitor and antagonist selected from the group consisting of ambicromil (CAS No. 58805-38-2) (Sanofi-Aventis), amelubant (CAS No. 346735-24-8) (Boehringer Ingelheim), DW-1141 (Dong Wha), ebselen (Daiichi Pharmaceutical), ibudilast (Kyorin), leucotriene inhibitors from Sanofi-Aventis, lymphotoxin-beta receptor (LT-D) from Biogen Idee, Pharmaprojects No. 1535 and 2728 (CAS No. 119340-33-9) (Sanofi- Aventis), R-112 (Rigel), Rev-5367 (CAS No. 92532-05-3) (Sanofi-Aventis), RG-14893 (CAS No. 141835-49-6) (Sanofi-Aventis), RG-5901-A (CAS No. 101910-24-1), 92532-23-5, RP 66153 (CAS No. 142422-79-5), RP 66364 (CAS No.186916-92-5), RP 69698 (CAS No. 141748-00-7) (Sanofi-Aventis), SC-411930 (Pfizer), SC-41930 (CAS No. 120072-59-5) (Pfizer), SC-50605 (CAS No. 138828-39-4) (Pfizer), SC-51146 (CAS No. 141059-52-1), SC- 53228 (CAS No. 153633-01-3) (Pfizer), spaglumic acid (ZY-15106) (CAS No.3106-85-2), 80619-64-3 (Novartis), tipredane (CAS No. 85197-77-9) (Bristol-Myers Squibb), U-75302 (CAS No. 119477-85-9) (Pfizer), and analogue or derivative thereof.
1030. The device of item 969 wherein the agent is an MAP kinase inhibitor.
1031. The device of item 969 wherein the agent is a matrix metalloproteinase inhibitor.
1032. The device of item 969 wherein the agent is an MCP- CCR2 inhibitor. 1033. The device of item 969 wherein the agent is an mTOR inhibitor.
1034. The device of item 969 wherein the agent is an mTOR kinase inhibitor.
1035. The device of item 969 wherein the agent is a microtubule inhibitor selected from the group consisting of antibody- maytansinoid conjugates from Biogen Idee, colchicines (MantiCore Pharmaceuticals), anticancer immunoconjugates from Johnson & Johnson, DIME from Octamer, gni-1f (GNI), huC242-DM4, huMy9-6-DM1 (ImmunoGen), IDN-5404 (Indena), IMO-098, IMOderm (Imotep), mebendazole (Introgen Therapeutics), microtubule poisons from Cambridge Enterprise, paclitaxel such as LOTAX from Aphios (CAS No. 33069-62-4), Genexol-PM from Samyang, Pharmaprojects No. 6383 (Tapestry Pharmaceuticals), RPR-112378 (Sanofi-Aventis), SGN-75 (Seattle Genetics), SPL-7435 (Starpharma), SSR-250411 (Sanofi-Aventis), trastuzumab-DM1 (Genentech), vinorelbine, and an analogue or derivative thereof.
1036. The device of item 969 wherein the agent is an MIF inhibitor.
1037. The device of item 969 wherein the agent is an MMP inhibitor.
1038. The device of item 969 wherein the agent is a neurokinin (NK) antagonist selected from the group consisting of anthrotainin (CAS No. 148084-40-6) (Sanofi-Aventis), an IBS thereapeutic from ArQuIe, MDL-105212A (CAS No. 167261-60-1) (Ssanofi-Aventis), Pharmaprojects No. 2744, 3258 (CAS No. 139167-47-8) 4006, 4201, or 5986 (Sanofi-Aventis), RP 67580 (CAS No. 135911-02-3), SR-144190 (CAS No. 201152-86-5), SSR-240600, SSR-241586 (Sanofi-Aventis), TKA-457 (Novartis), vestipitant mesylate (CAS No. 334476-64-1) (GlaxoSmithKline), Win-64821 (Sanofi-Aventis), PRX-96026 (Predix Pharmaceuticals), and an analogue or derivative thereof.
1039. The device of item 969 wherein the agent is an NF kappa B inhibitor selected from the group consisting of AVE-0545 or AVE- 0547 (Sanofi-Aventis), bortezomib (CAS No. 179324-69-7) (Millennium Pharmaceuticals), dexanabinol (CAS No. 112924-45-5) (Pharmos), dexlipotam (Viatris), Pharmaprojects No. 6283 (INDRA) (OXiGENE), IPL- 576092 (CAS No. 137571-30-3) (Inflazyme), NFKB decoy (Corgentech), NFKB decoy oligo (AnGes MG), NFKB's from Ariad, osteoporosis treatments or S5 (F005) from Fulcrum Pharmaceuticals, P61 (Phytopharm), R-flurbiprofen (CAS No. 5104-49-4) (Encore Pharmaceuticals), and an analogue or derivative thereof.
1040. The device of item 969 wherein the agent is a nitric oxide agonist.
1041. The device of item 969 wherein the agent is an ornithine decarboxylase inhibitor.
1042. The device of item 969 wherein the agent is a p38 MAP kinase inhibitor selected from the group consisting of AZD-6703 (AstraZeneca), JX-401 (Jexys Pharmaceuticals), BMS-2 (Bristol-Myers Squibb), a p38 MAP kinase inhibitor from Novartis, a p38-alpha MAP kinase inhibitor from Amphora, Pharmaprojects No. 5704 (Pharmacopeia), RPR- 200765A (Sanofi-Aventis), SD-282 (Johnson & Johnson), TAK-715 (Takeda), and an analogue or derivative thereof. 1043. The device of item 969 wherein the agent is a palmitoyl- protein thioesterase inhibitor.
1044. The device of item 969 wherein the agent is a PDGF receptor kinase inhibitor selected from the group consisting of AAL-993, AMN-107, or ABP-309 (Novartis), AMG-706 (Amgen), BAY-57-9352 (Bayer), CDP-860 (UCB), E-7080 (Eisai), imatinib (CAS No. 152459-95-5) (Novartis), OSI-930 (OSI Pharmaceuticals), RPR-127963E (Sanofi-Aventis), RWJ-540973 (Johnson & Johnson), sorafenib tosylate (Bayer), SU-11657 (Pfizer), tandutinib (CAS No. 387867-13-2) (Millennium Pharmaceuticals), vatalanib (Novartis), ZK-CDK (Schering AG), and an analogue or derivative thereof.
1045. The device of item 969 wherein the agent is (-)- halofenate (Metabolex), AMG-131 (Amgen), antidiabetics from Japan Tobacco, AZD-4619, AZD-8450, AZD-8677 (AstraZeneca), DRF-10945, balaglitazone (Dr Reddy's), CS-00088, CS-00098 (Chipscreen Biosciences), E-3030 (Eisai), etalocib (CAS No. 161172-51-6) (EIi Lilly), GSK-641597 (Ligand), GSK-677954 (GlaxoSmithKline), GW-409544 (Ligand), GW- 590735 (GlaxoSmithKline), K-111 (Hoffmann-La Roche), LY-518674 (EIi Lilly), LY-674 (Ligand), LY-929 (Ligand), MC-3001 , MC-3002 (MaxoCore Pharmaceuticals), metformin HCI + pioglitazone (CAS No. 1115-70-4 and
112529-15-4), ACTOPLUS MET from Andrx), muraglitazar (CAS No. 331741-94-7) (Bristol-Myers Squibb), naveglitazar (Ligand), oleoylethanolamide (Kadmus Pharmaceuticals), ONO-5129, pioglitazone hydrochloride (CAS No. 111025-46-8 and 112529-15-4) (Takeda), PLX-204 (Plexxikon), PPAR agonists from Genfit, PPAR delta agonists from EIi Lilly, PPAR-alpha agonists from CrystalGenomics, PPAR-gamma modulators and PPAR-β modulators from CareX, rosiglitazone maleate (CAS No. 122324- 73-4 or 155141-29-0) (GlaxoSmithKline), rosiglitazone maleate/glimepir (CAS No. 155141-29-0 and 93479-97-1), AVANDARYL, rosiglitazone maleate/metformin extend (CAS No. 155141-29-0 and 657-24-9), AVANDAMET, rosiglitazone maleate+metformin, AVANDAMET (GlaxoSmithKline), tesaglitazar (AstraZeneca), and an analogue or derivative thereof.
1046. The device of item 969 wherein the agent is a phosphatase inhibitor.
1047. The device of item 969 wherein the agent is a phosphodiesterase (PDE) inhibitor selected from the group consisting of avanafil (Tanabe Seiyaku), dasantafil (CAS No. 569351-91-3) (Schering- Plough), A-906119 (CAS No. 134072-58-5), DL-850 (Sanofi-Aventis), GRC- 3015, GRC-3566, GRC-3886 (Glenmark), HWA-153 (CAS No. 56395-66-5) (Sanofi-Aventis), hydroxypumafentrine (Altana), IBFB-130011 , IBFB-14-016, IBFB-140301 , IBFB-150007, IBFB-211913 (IBFB Pharma), L-826141 (Merck & Co), medorinone (CAS No. 88296-61-1) (Sanofi-Aventis), MEM- 1917 (Memory Pharmaceuticals), ND-1251 (Neuro3d), PDE inhibitors from ICOS, PDE IV inhibitors from Memory Pharmaceuticals and CrystalGenomics, Pharmaprojects No. 2742 and 6141 (Sanofi-Aventis), QAD-171 (Novartis), RHC-2963 (CAS No. 76993-12-9 and 76993-14-1), RPR-117658, RPR-122818 derivatives, SR-24870 , and RPR-132294 (Sanofi-Aventis), SK-350 (ln2Gen), stroke therapy agents from deCODE Genetics, TAS-203 (Taiho), tofimilast (CAS No. 185954-27-2) (Pfizer), UK- 371800 (Pfizer), WIN-65579 (CAS No. 158020-82-7) (Sanofi-Aventis), IBFB- 130020 (IBFB Pharma), OPC-6535 (CAS No. 145739-56-6) (Otsuka), a phosphodiesterase III inhibitor, enoximone, a phosphodiesterase IV inhibitor, fosfosal, Atopik (Barrier Therapeutics), triflusal, a phosphodiesterase V inhibitor, and an analogue or derivative thereof.
1048. The device of item 969 wherein the agent is a PKC inhibitor. 1049. The device of item 969 wherein the agent is a platelet activating factor antagonist.
1050. The device of item 969 wherein the agent is a platelet- derived growth factor receptor kinase inhibitor.
1051. The device of item 969 wherein the agent is a prolyl hydroxylase inhibitor.
1052. The device of item 969 wherein the agent is a polymorphonuclear neutrophil inhibitor.
1053. The device of item 969 wherein the agent is a protein kinase B inhibitor.
1054. The device of item 969 wherein the agent is a protein kinase C stimulant.
1055. The device of item 969 wherein the agent is a purine nucleoside analogue.
1056. The device of item 969 wherein the agent is a purinoreceptor P2X antagonist.
1057. The device of item 969 wherein the agent is a Raf kinase inhibitor.
1058. The device of item 969 wherein the agent is a reversible inhibitor of ErbB1 and ErbB2. 1059. The device of item 969 wherein the agent is a ribonucleoside triphosphate reductase inhibitor.
1060. The device of item 969 wherein the agent is an SDF-1 antagonist.
1061. The device of item 969 wherein the agent is a sheddase inhibitor.
1062. The device of item 969 wherein the agent is an SRC inhibitor.
1063. The device of item 969 wherein the agent is a stromelysin inhibitor.
1064. The device of item 969 wherein the agent is an Syk kinase inhibitor.
1065. The device of item 969 wherein the agent is a telomerase inhibitor.
1066. The device of item 969 wherein the agent is a TGF beta inhibitor selected from the group consisting of pirfenidone (CAS No. 53179- 13-8) (MARNAC), tranilast (CAS No. 53902-12-8) (Kissei), IN-1130 (ln2Gen), mannose-6-phosphate (BTG), TGF-β antagonists from Inflazyme (Pharmaprojects No. 6075), TGF-β antagonists from Sydney, non-industrial source), TGF-β I receptor kinase inhibitors from EIi Lilly, TGF-β receptor inhibitors from Johnson & Johnson, and an analogue or derivative thereof.
1067. The device of item 969 wherein the agent is a TNFα antagonist or TACE inhibitor selected from the group consisting of adalimumab (CAS No. 331731-18-1) (Cambridge Antibody Technology), AGIX-4207 (AtheroGenics), AGT-1 (Advanced Biotherapy), an antiinflammatory from Borean Pharma, Cellzome, or Paradigm Therapeutics, anti-inflammatory vaccine (TNF-alpha kinoid) from Neovacs, humanized anti-TNF antibody or an anti-TNF MAb (CB0006) Celltech (UCB), apratastat (CAS No. 287445-51-0) (Wyeth), BMS-561392 (Bristol-Myers Squibb), BN- 006 (Bone), certolizumab pegol (CAS No. 428863-50-7 or CH-138 (UCB), cilomilast (CAS No. 153259-65-5) (GlaxoSmithKline), CR-1 (Nuada Pharmaceuticals), CRx-119 (CombinatoRx), D-5410 (UCB), dacopafant (CAS No. 125372-33-0) (Sanofi-Aventis), dersalazine (CAS No. 188913-57- 7/188913-58-8) (Uriach), etanercept (CAS No. 185243-69-0) (Amgen), ethyl pyruvate (Critical (Critical Therapeutics), golimumab (CAS No. 476181-74-5) (Johnson & Johnson), hormono-immunotherapy from Ipsen, CDP571 (e.g., Humicade from UCB), IC-485 (ICOS), infliximab (CAS No. 170277-31-3) (Johnson & Johnson), IP-751 (Manhattan Pharmaceuticals), ISIS-104838 (CAS No. 250755-32-9) (ISIS Pharmaceuticals), lenalidomide (CAS No, 191732-72-6) (Celgene), lentinan (CAS No. 37339-90-5) (Ajinomoto), MDL- 201112 (CAS No. 142130-73-2) (Sanofi-Aventis), medroxyprogesterone (CAS No. 520-85-4) (InKine Pharmaceutical), N-acetylcysteine (CAS No. 616-91-1) (Zambon), NBE-P2 (DIREVO Biotech), nerelimomab (CAS No. 162774-06-3) (Chiron), OM-294DP (OM PHARMA), onercept (CAS No. 199685-57-9) (Yeda), PASSTNF-alpha (Verigen), pentoxifylline or oxypentifylline (Sanofi-Aventis), Pharmaprojects No. 4091 , 4241, 4295, or 4488 (Sanofi-Aventis), Pharmaprojects No. 5480 (Amgen), Pharmaprojects No. 6749 (Cengent), pirfenidone (CAS No. 53179-13-8) (MARNAC), RPR- 132294 (Sanofi-Aventis), S5 (F002) (Fulcrum Pharmaceuticals), simvastatin (CAS No. 79902-63-9) (Merck & Co), STA-6292 (Synta Pharmaceuticals), tacrolimus (CAS No. 104987-11-3) (Fujisawa LifeCycle Pharma), talactoferrin alfa (CAS No. 308240-58-6) (Agennix), thalidomide (CAS No. 50-35-1) (Celgene), TNF antagonists form ProStrakan, and Synergen, TNF inhibitors (Amgen), TNF-alpha antagonists from Dynavax Technologies and Jerina AG (Germany), TNF-alpha inhibitors from IBFB Pharma and Xencor (Xencor), torbafylline (CAS No. 105102-21-4) (Sanofi-Aventis), UR-1505 (Uriach), VT-346 (Viron Therapeutics), YS1L6 (Y's Therapeutics), YSTH2 (Y's Therapeutics), NPI-1302a-3 (Nereus Pharmaceuticals, a TNF antagonist from Jerina AG (Germany), dersalazine, and an analogue or derivative thereof.
1068. The device of item 969 wherein the agent is a Toll receptor inhibitor.
1069. The device of item 969 wherein the agent is a tubulin antagonist.
1070. The device of item 969 wherein the agent is a tyrosine kinase inhibitor selected from the group consisting of SU-011248, SUTENT from Pfizer Inc. (New York, NY), BMS-354825, PN-355.(Paracelsian Pharmaceuticals), AGN-199659 (Allergan), AAL-993 or ABP-309 (Novartis), adaphostin (NIH), AEE-788 (Novartis), AG-013736 (OSI Pharmaceuticals), AG-13736 (Pfizer), ALT-110 (Alteris Therapeutics), AMG-706 (Amgen), anticancer MAbs from Xencor, anti-EGFrvlll MAbs from Abgenix, anti-HER2 MAb from Abiogen, AZD-2171 or AZD-9935 (AstraZeneca), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), CEP-5214 (Cephalon), CEP- 7055 (Cephalon), cetuximab (ImClone Systems), CHIR-200131 and CHlR- 258 (Chiron), CP-547632 (OSI Pharmaceuticals), CP-724714 (Pfizer), CT- 301 (Creabilis Therapeutics), D-69491 (Baxter International), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EGFR/ErbB2 inhibitors from Array BioPharma, erlotinib (CAS No. 183319-69-9) (OSI Pharmaceuticals), EXEL-2880 (Exelixis), FK-778 (Sanofi-Aventis) , gefitinib (CAS No. 184475-35-2) (AstraZeneca), GW-2286 or GW-654652 (GlaxoSmithKline), her2/neu antigen from AlphaVax, HER-2/neu inhibitor from Generex, Herzyme (Medipad) (Sirna Therapeutics), HKI-272 (Wyeth), HuMax-EGFr (Genmab), idronoxil (CAS No. 81267-65-4) (Novogen), IGF-1 inhibitors from Ontogen, IMC-11F8 (ImCIone Systems), kahalalide F (CAS No. 149204-42-2) (PharmaMar), KDR inhibitor from LG Life Sciences, KDR inhibitors from Abbott Laboratories, KDR kinase inhibitors (UCB), Kdr kinase inhibitors from Merck & Co, KRN-633 and KRN-951 (Kirin Brewery), KSB-102 (Xenova), lapatinib ditosylate (CAS No. 388082-78-8) (GlaxoSmithKline), matuzumab (Merck KGaA), MDX-214 (Medarex), ME-103 (Pharmexa), MED-A300 (Gerolymatos), MNAC-13 (Lay Line Genomics), nimotuzumab (Center of Molecular Immunology), NSC-330507 or NSC-707545 (NIH), NV-50 (Novogen), OSI-930 (OSI Pharmaceuticals), panitumumab (Abgenix), pelitinib (CAS No. 287933-82-7) (Wyeth), pertuzumab (CAS No. 380610-27- 5) (Genentech), Pharmaprojects No. 3985 (Sanofi-Aventis), prostate cancer therapeutics from Sequenom (SQPC35, SQPC36, SQPC90), removab and remoxab (Trion Pharma), RG-13022 (CAS No. 136831-48-6), RG-13291 (CAS No. 138989-50-1), or RG-14620 (CAS No. 136831-49-7) (Sanofi- Aventis), RM-6427 (Romark), RNAi breast cancer therapy from Benite.c, RP 53801 (CAS No. 125882-88-4) (Sanofi-Aventis), sorafenib tosylate (Bayer), SU-11657 (Pfizer), Tie-2 antagonists from Semaia (Hybrigenics), Tie-2 inhibitors from Ontogen, trastuzumab (CAS No. 180288-69-1) (Genentech), tyrosine kinase inhibitors from Sanofi-Aventis, U3-1287, U3-1565, U3-1784, U3-1800 (U3 Pharma), vatalanib (Novartis), VEGFR-2 kinase inhibitor from Bristol-Myers Squibb, XL-647 (Exelixis), ZD-6474 (AstraZeneca), ZK-CDK (Schering AG), an EGFR tyrosine kinase inhibitor, EKB-569 (Wyeth), and an analogue or derivative thereof.
1071. The device of item 969 wherein the agent is a VEGF inhibitor.
1072. The device of item 969 wherein the agent is a vitamin D receptor agonist. 1073. The device of item 969 wherein the agent is ZD-6474 (an angiogenesis inhibitor).
1074. The device of item 969 wherein the agent is AP-23573 (an mTOR inhibitor).
1075. The device of item 969 wherein the agent is synthadotin (a tubulin antagonist).
1076. The device of item 969 wherein the agent is S-0885 (a collagenase inhibitor).
1077. The device of item 969 wherein the agent is aplidine (an elongation factor-1 alpha inhibitor).
1078. The device of item 969 wherein the agent is ixabepilone (an epithilone).
1079. The device of item 969 wherein the agent is IDN-5390 (an angiogenesis inhibitor).
1080. The device of item 969 wherein the agent is SB- 2723005 (an angiogenesis inhibitor).
1081. The device of item 969 wherein the agent is ABT-518 (an angiogenesis inhibitor).
1082. The device of item 969 wherein the agent is combretastatin (an angiogenesis inhibitor). 1083. The device of item 969 wherein the agent is anecortave acetate (an angiogenesis inhibitor).
1084. The device of item 969 wherein the agent is SB-715992 (a kinesin antagonist).
1085. The device of item 969 wherein the agent is temsirolimus (an mTOR inhibitor).
1086. The device of item 969 wherein the agent is adalimumab (a TNFα antagonist).
1087. The device of item 969, further comprising a polymer.
1088. The device of item 969 wherein the anti-scarring agent inhibits adhesion between the device and a host into which the device is implanted.
1089. The device of item 969 wherein the device delivers the anti-scarring agent locally to tissue proximate to the device.
1090. The device of item 969, further comprising a coating, wherein the coating comprises the anti-scarring agent.
1091. The device of item 969, further comprising a coating, wherein the coating is disposed on a surface of the device.
1092. The device of item 969, further comprising a coating, wherein the coating directly contacts the device. 1093. The device of item 969, further comprising a coating, wherein the coating indirectly contacts the device.
1094. The device of item 969, further comprising a coating, wherein the coating partially covers the device.
1095. The device of item 969, further comprising a coating, wherein the coating completely covers the device.
1096. The device of item 969, further comprising a coating, wherein the coating is a uniform coating.
1097. The device of item 969, further comprising a coating, wherein the coating is a non-uniform coating.
1098. The device of item 969, further comprising a coating, wherein the coating is a discontinuous coating.
1099. The device of item 969, further comprising a coating, wherein the coating is a patterned coating.
1100. The device of item 969, further comprising a coating, wherein the coating has a thickness of 100 Dm or less.
1101. The device of item 969, further comprising a coating, wherein the coating has a thickness of 10 Dm or less.
1102. The device of item 969, further comprising a coating, wherein the coating adheres to the surface of the device upon deployment of the device. 1103. The device of item 969, further comprising a coating, wherein the coating is stable at room temperature for a period of 1 year.
1104. The device of item 969, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 0.0001% to about 1% by weight.
1105. The device of item 969, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 1% to about 10% by weight.
1106. The device of item 969, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 10% to about 25% by weight.
1107. The device of item 969, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 25% to about 70% by weight.
1108. The device of item 969, further comprising a coating, wherein the coating further comprises a polymer.
1109. The device of item 969, further comprising a first coating having a first composition and the second coating having a second composition.
1110. The device of item 969, further comprising a first coating having a first composition and the second coating having a second composition, wherein the first composition and the second composition are different. 1111. The device of item 969, further comprising a polymeric carrier, wherein the polymeric carrier comprises a copolymer.
1112. The device of item 969, further comprising a polymeric carrier, wherein the polymeric carrier comprises a block copolymer.
1113. The device of item 969, further comprising a polymeric carrier, wherein the polymeric carrier comprises a random copolymer.
1114. The device of item 969, further comprising a polymeric carrier, wherein the polymeric carrier comprises a biodegradable polymer.
1115. The device of item 969, further comprising a polymeric carrier, wherein the polymeric carrier comprises a non-biodegradable polymer.
1116. The device of item 969, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrophilic polymer.
1117. The device of item 969, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrophobic polymer.
1118. The device of item 969, further comprising a polymeric carrier, wherein the polymeric carrier comprises a polymer having hydrophilic domains.
1119. The device of item 969, further comprising a polymeric carrier, wherein the polymeric carrier comprises a polymer having hydrophobic domains. 1120. The device of item 969, further comprising a polymeric carrier, wherein the polymeric carrier comprises a non-conductive polymer.
1121. The device of item 969, further comprising a polymeric carrier, wherein the polymeric carrier comprises an elastomer.
1122. The device of item 969, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrogel.
1123. The device of item 969, further comprising a polymeric carrier, wherein the polymeric carrier comprises a silicone polymer.
1124. The device of item 969, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrocarbon polymer.
1125. The device of item 969, further comprising a polymeric carrier, wherein the polymeric carrier comprises a styrene-derived polymer.
1126. The device of item 969, further comprising a polymeric carrier, wherein the polymeric carrier comprises a butadiene polymer.
1127. The device of item 969, further comprising a polymeric carrier, wherein the polymeric carrier comprises a macromer.
1128. The device of item 969, further comprising a polymeric carrier, wherein the polymeric carrier comprises a poly(ethylene glycol) polymer.
1129. The device of item 969, further comprising a polymeric carrier, wherein the polymeric carrier comprises an amorphous polymer. 1130. The device of item 969, further comprising a Iubricious coating.
1131. The device of item 969 wherein the anti-scarring agent is located within pores or holes of the device.
1132. The device of item 969 wherein the anti-scarring agent is located within a channel, lumen, or divet of the device.
1133. The device of item 969, further comprising a second pharmaceutically active agent.
1134. The device of item 969, further comprising an antiinflammatory agent.
1135. The device of item 969, further comprising an agent that inhibits infection.
1136. The device of item 969, further comprising an agent that inhibits infection, wherein the agent is an anthracycline.
1137. The device of item 969, further comprising an agent that inhibits infection, wherein the agent is doxorubicin.
1138. The device of item 969, further comprising an agent that inhibits infection, wherein the agent is mitoxantrone.
1139. The device of item 969, further comprising an agent that inhibits infection, wherein the agent is a fluoropyrimidine. 1140. The device of item 969, further comprising an agent that inhibits infection, wherein the agent is 5-fluorouracil (5-FU).
1141. The device of item 969, further comprising an agent that inhibits infection, wherein the agent is a folic acid antagonist.
1142. The device of item 969, further comprising an agent that inhibits infection, wherein the agent is methotrexate.
1143. The device of item 969, further comprising an agent that inhibits infection, wherein the agent is a podophylotoxin.
1144. The device of item 969, further comprising an agent that inhibits infection, wherein the agent is etoposide.
1145. The device of item 969, further comprising an agent that inhibits infection, wherein the agent is a camptothecin.
1146. The device of item 969, further comprising an agent that inhibits infection, wherein the agent is a hydroxyurea.
1147. The device of item 969, further comprising an agent that inhibits infection, wherein the agent is a platinum complex.
1148. The device of item 969, further comprising an agent that inhibits infection, wherein the agent is cisplatin.
1149. The device of item 969, further comprising an antithrombotic agent. 1150. The device of item 969, further comprising a visualization agent.
1151. The device of item 969, further comprising a visualization agent, wherein the visualization agent is a radiopaque material, wherein the radiopaque material comprises a metal, a halogenated compound, or a barium containing compound.
1152. The device of item 969, further comprising a visualization agent, wherein the visualization agent is a radiopaque material, wherein the radiopaque material comprises barium, tantalum, or technetium.
1153. The device of item 969, further comprising a visualization agent, wherein the visualization agent is a MRI responsive material.
1154. The device of item 969, further comprising a visualization agent, wherein the visualization agent comprises a gadolinium chelate.
1155. The device of item 969, further comprising a visualization agent, wherein the visualization agent comprises iron, magnesium, manganese, copper, or chromium.
1156. The device of item 969, further comprising a visualization agent, wherein the visualization agent comprises an iron oxide compound.
1157. The device of item 969, further comprising a visualization agent, wherein the visualization agent comprises a dye, pigment, or colorant. 1158. The device of item 969, further comprising an echogenic material.
1159. The device of item 969, further comprising an echogenic material, wherein the echogenic material is in the form of a coating.
1160. The device of item 969 wherein the device is sterile.
1161. The device of item 969 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device.
1162. The device of item 969 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the
- device, wherein the tissue is connective tissue. .
1163. The device of item 969 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is muscle tissue.
1164. The device of item 969 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is nerve tissue.
1165. The device of item 969 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is epithelium tissue. 1166. The device of item 969 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from the time of deployment of the device to about 1 year.
1167. The device of item 969 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from about 1 month to 6 months.
1168. The device of item 969 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from about 1 - 90 days.
1169. The device of item 969 wherein the anti-scarring agent is released in effective concentrations from the device at a constant rate.
1170. The device of item 969 wherein the anti-scarring~agent is released in effective concentrations from the device at an increasing rate.
1171. The device of item 969 wherein the anti-scarring agent is released in effective concentrations from the device at a decreasing rate.
1172. The device of item 969 wherein the anti-scarring agent is released in effective concentrations from the composition comprising the anti-scarring agent by diffusion over a period ranging from the time of deployment of the device to about 90 days.
1173. The device of item 969 wherein the anti-scarring agent is released in effective concentrations from the composition comprising the anti-scarring agent by erosion of the composition over a period ranging from the time of deployment of the device to about 90 days. 1174. The device of item 969 wherein the device comprises about 0.01 Dg to about 10 Dg of the anti-scarring agent.
1175. The device of item 969 wherein the device comprises about 10 Dg to about 10 mg of the anti-scarring agent.
1176. The device of item 969 wherein the device comprises about 10 mg to about 250 mg of the anti-scarring agent.
1177. The device of item 969 wherein the device comprises about 250 mg to about 1000 mg of the anti-scarring agent.
1178. The device of item 969 wherein the device comprises about 1000 mg to about 2500 mg of the anti-scarring agent.
1179. The device of item 969 wherein a surface of the device comprises less than 0.01 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
1180. The device of item 969 wherein a surface of the device comprises about 0.01 Dg to about 1 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
1181. The device of item 969 wherein a surface of the device comprises about 1 Dg to about 10 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
1182. The device of item 969 wherein a surface of the device comprises about 10 Dg to about 250 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied. 1183. The device of item 969 wherein a surface of the device comprises about 250 Dg to about 1000 Dg of the anti-scarring agent of anti- scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
1184. The device of item 969 wherein a surface of the device comprises about 1000 Dg to about 2500 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
1185. A device of any one of items 969-1184, wherein the device is a ureteric stent.
1186. A device of any one of items 969-1184, wherein the device is a aurethral stent.
1187. A device of any one of items 969-1184, wherein the device is a fallopian tube stent.
1188. A device of any one of items 969-1184, wherein the device is a prostate stent.
1189. A device of any one of items 969-1184, wherein the device is a metallic genital urinary stent.
1190. A device of any one of items 969-1184, wherein the device is a polymeric genital urinary stent.
1191. A device, comprising an ear and nose stent (e.g., an implant) and an anti-scarring agent or a composition comprising an anti- scarring agent, wherein the agent inhibits scarring between the device and a host into which the device is implanted. 1192. The device of item 1191 wherein the agent is an adensosine A2A receptor antagonist.
1193. The device of item 1191 wherein the agent is an AKT inhibitor.
1194. The device of item 1191 wherein the agent is an alpha 2 integrin antagonist, wherein the alpha 2 integrin antagonist is Pharmaprojects No. 5754 (Merck KgaA).
1195. The device of item 1191 wherein the agent an alpha 4 integrin antagonist.
1196. The device of item 1191 wherein the agent is an alpha 7 nicotinic receptor agonist.
1197. The device of item 1191 wherein the agent is an angiogenesis inhibitor selected from the group consisting of AG-12,958 (Pfizer), ATN-161 (Attention LLC), neovastat, an angiogenesis inhibitor from Jerina AG (Germany), NM-3 (Mercian), VGA-1155 (Taisho), FCE-26644 (Pfizer), FCE-26950 (Pfizer), FPMA (Meiji Daries), FR-111142 (Fujisawa), GGTI-298, GM-1306 (Ligand), GPA-1734 (Novartis), NNC-47-0011 (Novo Nordisk), herbamycin (Nippon Kayaku), lenalidomide (Celegene), IP-10 (NIH), ABT-828 (Abbott), KIN-841 (Tokushima University, Japan), SF-1126 (Semafore Pharmaceuticals), laminin technology (NIH), CHIR-258 (Chiron), NVP-AEW541 (Novartis), NVP-AEW541 (Novartis), Vt16907 (Alchemia), OXI-8007 (Oxigene), EG-3306 (Ark Therapeutics), Maspin (Arriva), ABT- 567 (Abbott), PPI-2458 (Praecis Pharmaceuticals), CC-5079, CC-4089 (Celgene), HIF-1 alpha inhibitors (Xenova), S-247 (Pfizer), AP-23573 (Ariad), AZD-9935 (Astra Zeneca), mebendazole (Introgen Therapeutics), MetAP-2 inhibitors (GlaxoSmithKline), AG-615 (Angiogene Pharmaceuticals), Tie-2 antagonists (Hybrigenics), NC-381 , CYC-381, NC-169, NC-219, NC-383, NC-384, NC-407 (Lorus Therapeutics), ATN-224 (Attention), ON-01370 (Onconova), Vitronectin antagonists (Amgen), SDX-103 (Salmedix), Vitronectin antagonists (Shire), CHP (Riemser), TEK (Amgen), Anecortave acetate (Alcon), T46.2 (Matrix Therapeutics), HG-2 (Heptagen), TEM antagonists (Genzyme), Oxi-4500 (Oxigene), ATN-161 (Attenuon), WX-293 (Wϋex), M-2025 (Metris Therapeutics), Alphastatin (BioActa), YH-16 (Yantai Rongchang), BIBF-1120 (Boehringer Ingelheim), BAY-57-9352 (Bayer), AS- 1404 (Cancer Research Technology), SC-77964 (Pfizer), glycomimetics (BioTie Therapies), TIE-2 Inhibitors (Ontogen), DIMI, Octamer (Octamer), ABR-215050 (Active Biotech), ABT-518 (Abbott), KDR inhibitors (Abbott), BSF-466895 (Abbott), SCH-221153 (Schering-Plough), DAC:antiangiogenic (ConjuChem), TFPI (EntreMed), AZD-2171 (Astra-Zenaca), CDC-394 (Celgene), LY290293 (EIi Lilly), IDN-5390 (Indena), Kdr Kinase Inhibitors (Merck), CT-113020, CT-116433, CT-116563, CT-31890, CT-32228) (Cell Therapeutics), A-299620 (Abbott), TWEAK Inhibitor (Amgen), VEGF modulators (Johnson and Johnson), Tum-N53, tumstatin (Genzyme), Thios- 1 , Thios-2 (Thios Pharmaceuticals), MV-6401 (Miravant Medical Technologies), Spisulosine (PharmaMar), CEP-7055 (Cephalon), AUV-201 (Auvation), LM-609 (EIi Lilly), SKF-106615 (AnorMED), Oglufanide disodium (Cytran), BW-114 (Pharminox), Calreticulin (NIH), WX-678 (Wilex), SD-7784 (Pfizer), WX-UK1 (Wilex), SH-268 (Schering AG), 2-Me-PGA (Celgene), S- 137 (Pfizer), ZD-6126 (Angiogene Pharmaceuticals), SG-292 (SignalGen), Benefin (Lane Labs), A6, A36 (Angstrom), SB-2723005 (GlaxoSmithKline), SC-7 (Cell Therapeutics), ZEN-014 (AEterna Zentaris), 2-methoxyestradiol (EntreMed), NK-130119 (Nippon Kayaku), CC-10004 (Celgene), AVE- 8062A (Ajinomoto), Tacedinaline (Pfizer), Actinonin (Tokyo Metropolitan Institute of Medical Science), Lenalidomide (Celgene), VGA-1155, BTO-956 (SRI International), ER-68203-00 (Eisai), CT-6685 (UCB), JKC-362 (Phoenix Pharmaceuticals), DMI-3798 (DMI Biosciences, Angiomate (Ipsen), ZD-6474 (AstraZeneca), CEP-5214 (Cephalon), Canstatin (Genzyme), NM-3 (Mercian), Oridigm (MediQuest Therapeutics), Exherin (Adherex), BLS-0597 (Boston Life Sciences), PTC-299 (PTC Therapeutics), NPI-2358 (Nereus Pharmaceuticals), CGP-79787 (Novartis), AEE-788 (Novartis), CKD-732 (Chong Kun Dang), CP-564959 (OSI Pharmaceuticals), CM-101 (CarboMed), CT-2584, CT3501 (Cell Therapeutics), combretastatin and analogues and derivatives thereof (Oxigene), Rebimastat (Bristol-Meyers Squibb), Dextrin 2-sulfate (ML Laboratories), Cilengitide (Merk KGaA), NSC-706704 (Pharminox), KRN- 951 (Kirin Brewery), Ukrain, NSC-631570 (Nowicky Pharma), Tecogalan sodium (Daiichi Pharmaceutical), Tz-93 (Tsumura), TBC-1635 (Encysive Pharmaceuticals), TAN-1120 (Takeda), Semaxanib (Pfizer), BDI-7800 (Biopharmacopae), SD-186, SD-983 (Bristol-Meyers Squibb), SB-223245 (GlaxoSmithKline), SC-236 (Pfizer), RWJ-590973 (Johnson and Johnson), ILX-1850 (Genzyme), SC-68488, S-836 (Pfizer), CG-55069-11 (CuraGen), Ki-23057 (Kirin Brewery), CCX-700 (Chemoentryx), Pegaptanib octasodium ^(Giled Sciences), ANGIOCOL (available from Biostratum Inc.), or an analogue or derivative thereof.
1198. The device of item 1191 wherein the agent is an apoptosis antagonist.
1199. The device of item 1191 wherein the agent is an apoptosis activator.
1200. The device of item 1191 wherein the agent is a beta 1 integrin antagonist.
1201. The device of item 1191 wherein the agent is a beta tubulin inhibitor. 1202. The device of item 1191 wherein the agent is a blocker of enzyme production in Hepatitis C.
1203. The device of item 1191 wherein the agent is a Bruton's tyrosine kinase inhibitor.
1204. The device of item 1191 wherein the agent is a calcineurin inhibitor.
1205. The device of item 1191 wherein the agent is a caspase 3 inhibitor.
1206. The device of item 1191 wherein the agent is a CC chemokine receptor antagonist.
1207. The device of item 1191 wherein, the agentjs ;.a cell cycle inhibitor selected from the group consisting of SNS-595 (Sunesis), synthadotin, KRX-0403, and an analogue or derivative thereof.
1208. The device of item 1191 wherein the agent is a cathepsin B inhibitor.
1209. The device of item 1191 wherein the agent is a cathepsin K inhibitor, wherein the cathepsin K inhibitor is 462795 (GlaxoSmithKline), INPL-022-D6 (Amura Therapeutics), an analogue or derivative thereof).
1210. The device of item 1191 wherein the agent is a cathepsin L inhibitor. 1211. The device of item 1191 wherein the agent is a CD40 antagonist.
\ 1212. The device of item 1191 wherein the agent is a chemokine receptor agonist.
1213. The device of item 1191 wherein the agent is a chymase inhibitor.
1214. The device of item 1191 wherein the agent is a collagenase antagonist.
1215. The device of item 1191 wherein the agent is a CXCR antagonist.
1216. The device of item 1191 wherein the agent is a cyclin_ . . dependent kinase inhibitor selected from the group consisting of a CDK-1 inhibitor, a CDK-2 inhibitor, a CDK- 4 inhibitor, a CDK-6 inhibitor, a CAK1 inhibitor from GPC Biotech and Bristol-Myers Squibb, RGB-286199 (GPC Biotech), an anticancer agent from Astex Technology, a CAK1 inhibitor from GPC Biotech, a CDK inhibitor from Sanofi-Aventis, a CDK1/CDK2 inhibitor from Amgen, a CDK2 inhibitor from SUGEN-2 (Pfizer), a hearing loss therapy agent (Sound Pharmaceuticals), PD-0332991 (Pfizer), RGB-286199 (GPC Biotech), Ro-0505124 (Hoffmann-La Roche), a Ser/Thr kinase inhibitor from Lilly (EIi Lilly), CVT-2584 (CAS No. 199986-75-9) (CV Therapeutics), and an analogue or derivative thereof.
1217. The device of item 1191 wherein the agent is a cyclooxygenase 1 inhibitor. 1218. The device of item 1191 wherein the agent is a DHFR inhibitor.
1219. The device of item 1191 wherein the agent is a dual integrin inhibitor.
1220. The device of item 1191 wherein the agent is an elastase inhibitor.
1221. The device of item 1191 wherein the agent is an elongation factor-1 alpha inhibitor.
1222. The device of item 1191 wherein the agent is an endothelial growth factor antagonist.
- - 1223. The device of item 1191 wherein the agent is an . . endothelial growth factor receptor kinase inhibitor selected from the group consisting of sorafenib tosylate (Bayer), AAL-993 (Novartis), ABP-309 (Novartis), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), E- 7080 (Eisai), EG-3306 (Ark Therapeutics), EXEL-2880 (Exelixis), GW- 654652 (GlaxoSmithKline), a KDR inhibitor from LG Life Sciences, CT-6685 and CT-6729 (UCB), KRN-633 and KRN-951 (Kirin Brewery), OSI-930 (OSI Pharmaceuticals), SP-5.2 (Supratek Pharma), SU-11657 (Pfizer), a Tie-2 antagonist (Hybrigenics), a VEGFR-2 kinase inhibitor (Bristol-Myers Squibb), XL-647 (Exelixis), a KDR inhibitor from Abbott Laboratories, sorafenib tosylate, and an analogue or derivative thereof.
1224. The device of item 1191 wherein the agent is an endotoxin antagonist. 1225. The device of item 1191 wherein the agent is an epothilone and tubulin binder.
1226. The device of item 1191 wherein the agent is an estrogen receptor antagonist.
1227. The device of item 1191 wherein the agent is an FGF inhibitor.
1228. The device of item 1191 wherein the agent is a farnexyl transferase inhibitor.
1229. The device of item 1191 wherein the agent is farnesyltransferase inhibitor selected from the group of A-197574 (Abbott), a famesyltransferase inhibitor from Servier, FPTIII (Strathclyde Institute for Drug R), LB-42908 (LG Life Sciences), Pharmaprojects No. 5063 (Genzyme), Pharmaprojects No. 5597 (Ipsen), Yissum Project No. B-1055 (Yissum), and an analogue or derivative thereof.
1230. The device of item 1191 wherein the agent is an FLT-3 kinase inhibitor.
1231. The device of item 1191 wherein the agent is an FGF receptor kinase inhibitor.
1232. The device of item 1191 wherein the agent is a fibrinogen antagonist selected from the group consisting of AUV-201 (Auvation), MG-13926 (Sanofi-Aventis), plasminogen activator (CAS No. 105913-11-9) (from Sanofi-Aventis or UCB), plasminogen activator-2 (tPA- 2) (Sanofi-Aventis), pro-urokinase (CAS No. 82657-92-9) (Sanofi-Aventis), and an analogue or derivative thereof. 1233. The device of item 1191 wherein the agent is a heat shock protein 90 antagonist selected from the group consisting of SRN-005 (Sirenade), geldanamycin, NSC-33050 (17-allylaminogeldanamycin; 17- AAG), ^-dimethylaminoethylamino-^-demethoxy-geldanamycin (17- DMAG), rifabutin (rifamycin XIV, 1',4-didehydro-1-deoxy-1 ,4-dihydro-5'-(2- methylpropyl)-1-oxo-), and an analogue or derivative thereof.
1234. The device of item 1191 wherein the agent is a histone deacetylase inhibitor.
1235. The device of item 1191 wherein the agent is an HMGCoA reductase inhibitor selected from the group consisting of an atherosclerosis therapeutic from Lipid Sciences, ATI-16000 (ARYx Therapeutics), KS-01-019 (Kos Pharmaceuticals), Pharmaprojects No. 2197 (Sanofi-Aventi), RP 61969 (Sanofi-Aventis), and an analogue or derivative thereof. - . , . ..„
1236. The device of item 1191 wherein the agent is an ICAM inhibitor.
1237. The device of item 1191 wherein the agent is an IL, ICE and IRAK antagonist, wherein the antagonist is a CJ-14877, CP-424174 (Pfizer), NF-61 (Negma-Lerads), and an analogue or derivative thereof.
1238. The device of item 1191 wherein the agent is an IL-2 inhibitor.
1239. The device of item 1191 wherein the agent is an immunosuppressant selected from the group consisting of teriflunomide (Sanofi Aventis), chlorsulfaquinoxalone (NSC-339004), chlorsulfaquinoxalone sulfate, CS-712 (Sankyo), ismomultin alfa (CAS No. 457913-93-8) (Akzo Nobel), antiallergics from GenPat77, antiinflammatories or AT-005 (Androclus Therapeutics), autoimmune disease therapeutics from EpiVax, BN-007 (Bone), budesonide (CAS No. 51333-22- 3) (MAP Pharmaceuticals), CO-14 (Genzyme), edratide (CAS No. 433922- 67-9) (Teva), EP-314 (Enanta), eprovafen (CAS No. 101335-99-3) (Sanofi- Aventis), HWA- 131 (CAS No. 118788-41-3) (Sanofi-Aventis), immunomodulators from MerLion Pharmaceuticals, immunosuppressives from Alchemia, IPL-12 (Inflazyme), MDL-9563 (CAS No. 27086-86-8) (Sanofi-Aventis), Pharmaprojects No. 2330 (Sanofi-Aventis), Pharmaprojects No. 6426 (Abgenix), PXS-25 (Pharmaxis), rosmarinic acid (CAS No. 20283-92-5) (Sanofi-Aventis), RP 42927 or RP 54745 (CAS No. 135330-08-4) (Sanofi-Aventis), SGN-35 (Seattle Genetics), ST-1959 (Sigma-Tau), type I diabetes therapy from SYNX Pharma, UNIL-88 (Debiopharm), VP-025 (Vasogen), VR-694 (Vectura), PRTX-001 (Protalex), and an analogue or derivative thereof.
1240. The device of item 1191 wherein the agent is an IMPDH (inosine monophosphate).
1241. The device of item 1191 wherein the agent is an integrin antagonist.
1242. The device of item 1191 wherein the agent is an interleukin antagonist.
1243. The device of item 1191 wherein the agent is an inhibitor of type III receptor tyrosine kinase.
1244. The device of item 1191 wherein the agent is an irreversible inhibitor of enzyme methionine aminopeptidase type 2. 1245. The device of item 1191 wherein the agent is an isozyme selective delta protein kinase C inhibitor.
1246. The device of item 1191 wherein the agent a JAK3 enzyme inhibitor.
1247. The device of item 1191 wherein the agent is a JNK inhibitor.
1248. The device of item 1191 wherein the agent is a kinase inhibitor.
1249. The device of item 1191 wherein the agent is kinesin antagonist.
1250. The device of item 1191 wherein the agent is a kinesin antagonist.
1251. The device of item 1191 wherein the agent is a leukotriene inhibitor and antagonist selected from the group consisting of ambicromil (CAS No. 58805-38-2) (Sanofi-Aventis), amelubant (CAS No. 346735-24-8) (Boehringer Ingelheim), DW-1141 (Dong Wha), ebselen (Daiichi Pharmaceutical), ibudilast (Kyorin), leucotriene inhibitors from Sanofi-Aventis, lymphotoxin-beta receptor (LT-D) from Biogen Idee, Pharmaprojects No. 1535 and 2728 (CAS No. 119340-33-9) (Sanofi- Aventis), R-112 (Rigel), Rev-5367 (CAS No. 92532-05-3) (Sanofi-Aventis), RG-14893 (CAS No. 141835-49-6) (Sanofi-Aventis), RG-5901-A (CAS No. 101910-24-1), 92532-23-5, RP 66153 (CAS No. 142422-79-5), RP 66364 (CAS No.186916-92-5), RP 69698 (CAS No. 141748-00-7) (Sanofi-Aventis), SC-411930 (Pfizer), SC-41930 (CAS No. 120072-59-5) (Pfizer), SC-50605 (CAS No. 138828-39-4) (Pfizer), SC-51146 (CAS No. 141059-52-1), SC- 53228 (CAS No. 153633-01-3) (Pfizer), spaglumic acid (ZY-15106) (CAS No.3106-85-2), 80619-64-3 (Novartis), tipredane (CAS No. 85197-77-9) (Bristol-Myers Squibb), U-75302 (CAS No. 119477-85-9) (Pfizer), and analogue or derivative thereof.
1252. The device of item 1191 wherein the agent is an MAP kinase inhibitor.
1253. The device of item 1191 wherein the agent is a matrix metalloproteinase inhibitor.
1254. The device of item 1191 wherein the agent is an MCP- CCR2 inhibitor.
1255. The device of item 1191 wherein the agent is an mTOR inhibitor.
1256. The device of item 1191 wherein the agent is an mTOR kinase inhibitor.
1257. The device of item 1191 wherein the agent is a microtubule inhibitor selected from the group consisting of antibody- maytansinoid conjugates from Biogen Idee, colchicines (MantiCore Pharmaceuticals), anticancer immunoconjugates from Johnson & Johnson, DIME from Octamer, gni-1f (GNI), huC242-DM4, huMy9-6-DM1 (ImmunoGen), IDN-5404 (Indena), IMO-098, IMOderm (Imotep), mebendazole (Introgen Therapeutics), microtubule poisons from Cambridge Enterprise, paclitaxel such as LOTAX from Aphios (CAS No. 33069-62-4), Genexol-PM from Samyang, Pharmaprojects No. 6383 (Tapestry Pharmaceuticals), RPR-112378 (Sanofi-Aventis), SGN-75 (Seattle Genetics), SPL-7435 (Starpharma), SSR-250411 (Sanofi-Aventis), trastuzumab-DM1 (Genentech), vinorelbine, and an analogue or derivative thereof.
1258. The device of item 1191 wherein the agent is an MIF inhibitor.
1259. The device of item 1191 wherein the agent is an MMP inhibitor.
1260. The device of item 1191 wherein the agent is a neurokinin (NK) antagonist selected from the group consisting of anthrotainin (CAS No. 148084-40-6) (Sanofi-Aventis), an IBS thereapeutic from ArQuIe, MDL-105212A (CAS No. 167261-60-1) (Ssanofi-Aventis), Pharmaprojects No. 2744, 3258 (CAS No. 139167-47-8) 4006, 4201, or 5986 (Sanofi-Aventis), RP 67580 (CAS No. 135911-02-3), SR-144190 (CAS No. 201152-86-5), SSR-240600, SSR-241586 (Sanofi-Aventis), TKA-457 (Novartis), vestipitant mesylate (CAS No. 334476-64-1) (GlaxoSmithKline), Win-64821 (Sanofi-Aventis), PRX-96026 (Predix Pharmaceuticals), and an analogue or derivative thereof.
1261. The device of item 1191 wherein the agent is an NF kappa B inhibitor selected from the group consisting of AVE-0545 or AVE- 0547 (Sanofi-Aventis), bortezomib (CAS No. 179324-69-7) (Millennium Pharmaceuticals), dexanabinol (CAS No. 112924-45-5) (Pharmos), dexlipotam (Viatris), Pharmaprojects No. 6283 (INDRA) (OXiGENE), IPL- 576092 (CAS No. 137571-30-3) (Inflazyme), NFKB decoy (Corgentech), NFKB decoy oligo (AnGes MG), NFKB's from Ariad, osteoporosis treatments or S5 (F005) from Fulcrum Pharmaceuticals, P61 (Phytopharm), R-flurbiprofen (CAS No. 5104-49-4) (Encore Pharmaceuticals), and an analogue or derivative thereof. 1262. The device of item 1191 wherein the agent is a nitric oxide agonist.
1263. The device of item 1191 wherein the agent is an ornithine decarboxylase inhibitor.
1264. The device of item 1191 wherein the agent is a p38 MAP kinase inhibitor selected from the group consisting of AZD-6703 (AstraZeneca), JX-401 (Jexys Pharmaceuticals), BMS-2 (Bristol-Myers Squibb), a p38 MAP kinase inhibitor from Novartis, a p38-alpha MAP kinase inhibitor from Amphora, Pharmaprojects No. 5704 (Pharmacopeia), RPR- 200765A (Sanofi-Aventis), SD-282 (Johnson & Johnson), TAK-715 (Takeda), and an analogue or derivative thereof.
1265. The device of item 1191 wherein the agent is a palmitoyl-protein thioesterase inhibitor.
1266. The device of item 1191 wherein the agent is a PDGF receptor kinase inhibitor selected from the group consisting of AAL-993, AMN-107, or ABP-309 (Novartis), AMG-706 (Amgen), BAY-57-9352 (Bayer), CDP-860 (UCB), E-7080 (Eisai), imatinib (CAS No. 152459-95-5) (Novartis), OSI-930 (OSI Pharmaceuticals), RPR-127963E (Sanofi-Aventis), RWJ-540973 (Johnson & Johnson), sorafenib tosylate (Bayer), SU-11657 (Pfizer), tandutinib (CAS No. 387867-13-2) (Millennium Pharmaceuticals), vatalanib (Novartis), ZK-CDK (Schering AG), and an analogue or derivative thereof.
1267. The device of item 1191 wherein the agent is (-)- halofenate (Metabolex), AMG-131 (Amgen), antidiabetics from Japan Tobacco, AZD-4619, AZD-8450, AZD-8677 (AstraZeneca), DRF-10945, balaglitazone (Dr Reddy's), CS-00088, CS-00098 (Chipscreen Biosciences), E-3030 (Eisai), etalocib (CAS No. 161172-51-6) (EIi Lilly), GSK-641597 (Ligand), GSK-677954 (GlaxoSmithKline), GW-409544 (Ligand), GW- 590735 (GlaxoSmithKline), K-111 (Hoffmann-La Roche), LY-518674 (EIi Lilly), LY-674 (Ligand), LY-929 (Ligand), MC-3001, MC-3002 (MaxoCore Pharmaceuticals), metformin HCI + pioglitazone (CAS No. 1115-70-4 and 112529-15-4), ACTOPLUS MET from Andrx), muraglitazar (CAS No. 331741-94-7) (Bristol-Myers Squibb), naveglitazar (Ligand), oleoylethanolamide (Kadmus Pharmaceuticals), ONO-5129, pioglitazone hydrochloride (CAS No. 111025-46-8 and 112529-15-4) (Takeda), PLX-204 (Plexxikon), PPAR agonists from Genfit, PPAR delta agonists from EIi Lilly, PPAR-alpha agonists from CrystalGenomics, PPAR-gamma modulators and PPAR-β modulators from CareX, rosiglitazone maleate (CAS No. 122324- 73-4 or 155141-29-0) (GlaxoSmithKline), rosiglitazone maleate/glimepir (CAS No. 155141-29-0 and 93479-97-1), AVANDARYL, rosiglitazone maleate/metformin extend (CAS No. 155141-29-0 and 657-24-9), AVANDAMET, rosiglitazone maleate+metformin, AVANDAMET - (GlaxoSmithKline), tesaglitazar (AstraZeneca), and an analogue or derivative thereof.
1268. The device of item 1191 wherein the agent is a phosphatase inhibitor.
1269. The device of item 1191 wherein the agent is a phosphodiesterase (PDE) inhibitor selected from the group consisting of avanafil (Tanabe Seiyaku), dasantafil (CAS No. 569351-91-3) (Schering- Plough), A-906119 (CAS No. 134072-58-5), DL-850 (Sanofi-Aventis), GRC- 3015, GRC-3566, GRC-3886 (Glenmark), HWA-153 (CAS No. 56395-66-5) (Sanofi-Aventis), hydroxypumafentrine (Altana), IBFB-130011 , IBFB-14-016, IBFB-140301 , IBFB-150007, IBFB-211913 (IBFB Pharma), L-826141 (Merck & Co), medorinone (CAS No. 88296-61-1) (Sanofi-Aventis), MEM- 1917 (Memory Pharmaceuticals), ND-1251 (Neuro3d), PDE inhibitors from ICOS, PDE IV inhibitors from Memory Pharmaceuticals and CrystalGenomics, Pharmaprojects No. 2742 and 6141 (Sanofi-Aventis), QAD-171 (Novartis), RHC-2963 (CAS No. 76993-12-9 and 76993-14-1), RPR-117658, RPR-122818 derivatives, SR-24870 , and RPR-132294 (Sanofi-Aventis), SK-350 (ln2Gen), stroke therapy agents from deCODE Genetics, TAS-203 (Taiho), tofimilast (CAS No. 185954-27-2) (Pfizer), UK- 371800 (Pfizer), WIN-65579 (CAS No. 158020-82-7) (Sanofi-Aventis), IBFB- 130020 (IBFB Pharma), OPC-6535 (CAS No. 145739-56-6) (Otsuka), a phosphodiesterase III inhibitor, enoximone, a phosphodiesterase IV inhibitor, fosfosal, Atopik (Barrier Therapeutics), triflusal, a phosphodiesterase V inhibitor, and an analogue or derivative thereof.
1270. The device of item 1191 wherein the agent is a PKC inhibitor.
1271. The device of item 1191 wherein the agent is a platelet activating factor antagonist.
1272. The device of item 1191 wherein the agent is a platelet- derived growth factor receptor kinase inhibitor.
1273. The device of item 1191 wherein the agent is a prolyl hydroxylase inhibitor.
1274. The device of item 1191 wherein the agent is a polymorphonuclear neutrophil inhibitor.
1275. The device of item 1191 wherein the agent is a protein kinase B inhibitor. 1276. The device of item 1191 wherein the agent is a protein kinase C stimulant.
1277. The device of item 1191 wherein the agent is a purine nucleoside analogue.
1278. The device of item 1191 wherein the agent is a purinoreceptor P2X antagonist.
1279. The device of item 1191 wherein the agent is a Raf kinase inhibitor.
1280. The device of item 1191 wherein the agent is a reversible inhibitor of ErbB1 and ErbB2.
- - - - 1281. The device of item 1191 wherein the agent is a ribonucleoside triphosphate reductase inhibitor.
1282. The device of item 1191 wherein the agent is an SDF-1 antagonist.
1283. The device of item 1191 wherein the agent is a sheddase inhibitor.
1284. The device of item 1191 wherein the agent is an SRC inhibitor.
1285. The device of item 1191 wherein the agent is a stromelysin inhibitor. 1286. The device of item 1191 wherein the agent is an Syk kinase inhibitor.
1287. The device of item 1191 wherein the agent is a telomerase inhibitor.
1288. The device of item 1191 wherein the agent is a TGF beta inhibitor selected from the group consisting of pirfenidone (CAS No. 53179-13-8) (MARNAC), tranilast (CAS No. 53902-12-8) (Kissei), IN-1130 (ln2Gen), mannose-6-phosphate (BTG), TGF-β antagonists from Inflazyme (Pharmaprojects No. 6075), TGF-β antagonists from Sydney, non-industrial source), TGF-β I receptor kinase inhibitors from EIi Lilly, TGF-β receptor inhibitors from Johnson & Johnson, and an analogue or derivative thereof.
1289. The device of item 1191 wherein the agent is a TNFα antagonist or TACE inhibitor selected from the group consisting of - - adalimumab (CAS No. 331731-18-1) (Cambridge Antibody Technology), AGIX-4207 (AtheroGenics), AGT-1 (Advanced Biotherapy), an antiinflammatory from Borean Pharma, Cellzome, or Paradigm Therapeutics, anti-inflammatory vaccine (TNF-alpha kinoid) from Neovacs, humanized anti-TNF antibody or an anti-TNF MAb (CB0006) Celltech (UCB), apratastat (CAS No. 287445-51-0) (Wyeth), BMS-561392 (Bristol-Myers Squibb), BN- 006 (Bone), certolizumab pegol (CAS No. 428863-50-7 or CH-138 (UCB), cilomilast (CAS No. 153259-65-5) (GlaxoSmithKline), CR-1 (Nuada Pharmaceuticals), CRx-119 (CombinatoRx), D-5410 (UCB), dacopafant (CAS No. 125372-33-0) (Sanofi-Aventis), dersalazine (CAS No. 188913-57- 7/188913-58-8) (Uriach), etanercept (CAS No. 185243-69-0) (Amgen), ethyl pyruvate (Critical (Critical Therapeutics), golimumab (CAS No. 476181-74-5) (Johnson & Johnson), hormono-immunotherapy from Ipsen, CDP571 (e.g., Humicade from UCB), IC-485 (ICOS), infliximab (CAS No. 170277-31-3) (Johnson & Johnson), IP-751 (Manhattan Pharmaceuticals), ISIS-104838 (CAS No. 250755-32-9) (ISIS Pharmaceuticals), lenalidomide (CAS No. 191732-72-6) (Celgene), lentinan (CAS No. 37339-90-5) (Ajinomoto), MDL- 201112 (CAS No. 142130-73-2) (Sanofi-Aventis), medroxyprogesterone (CAS No. 520-85-4) (InKine Pharmaceutical), N-acetylcysteine (CAS No. 616-91-1) (Zambon), NBE-P2 (DIREVO Biotech), nerelimomab (CAS No. 162774-06-3) (Chiron), OM-294DP (OM PHARMA), onercept (CAS No. 199685-57-9) (Yeda), PASSTNF-alpha (Verigen), pentoxifylline or oxypentifylline (Sanofi-Aventis), Pharmaprojects No. 4091 , 4241 , 4295, or 4488 (Sanofi-Aventis), Pharmaprojects No. 5480 (Amgen), Pharmaprojects No. 6749 (Cengent), pirfenidone (CAS No. 53179-13-8) (MARNAC), RPR- 132294 (Sanofi-Aventis), S5 (F002) (Fulcrum Pharmaceuticals), simvastatin (CAS No. 79902-63-9) (Merck & Co), STA-6292 (Synta Pharmaceuticals), tacrolimus (CAS No. 104987-11-3) (Fujisawa LifeCycle Pharma), talactoferrin alfa (CAS No. 308240-58-6) (Agennix), thalidomide (CAS No. 50-35-1) (Celgene), TNF antagonists form ProStrakan, and Synergen, TNF inhibitors (Amgen), TNF-alpha antagonists from Dynavax Technologies and Jerina AG (Germany), TNF-alpha inhibitors from IBFB Pharma and Xencor (Xencor), torbafylline (CAS No. 105102-21-4) (Sanofi-Aventis), UR-1505 (Uriach), VT-346 (Viron Therapeutics), YSIL6 (Y's Therapeutics), YSTH2 (Y's Therapeutics), NPI-1302a-3 (Nereus Pharmaceuticals, a TNF antagonist from Jerina AG (Germany), dersalazine, and an analogue or derivative thereof.
1290. The device of item 1191 wherein the agent is a Toll receptor inhibitor.
1291. The device of item 1191 wherein the agent is a tubulin antagonist.
1292. The device of item 1191 wherein the agent is a tyrosine kinase inhibitor selected from the group consisting of SU-011248, SUTENT from Pfizer Inc. (New York, NY), BMS-354825, PN-355 (Paracelsian Pharmaceuticals), AGN-199659 (Allergan), AAL-993 or ABP-309 (Novartis), adaphostin (NIH), AEE-788 (Novartis), AG-013736 (OS) Pharmaceuticals), AG-13736 (Pfizer), ALT-110 (Alteris Therapeutics), AMG-706 (Amgen), anticancer MAbs from Xencor, anti-EGFrvlll MAbs from Abgenix, anti-HER2 MAb from Abiogen, AZD-2171 or AZD-9935 (AstraZeneca), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), CEP-5214 (Cephalon), CEP- 7055 (Cephalon), cetuximab (ImClone Systems), CHIR-200131 and CHIR- 258 (Chiron), CP-547632 (OSI Pharmaceuticals), CP-724714 (Pfizer), CT- 301 (Creabilis Therapeutics), D-69491 (Baxter International), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EGFR/ErbB2 inhibitors from Array BioPharma, erlotinib (CAS No. 183319-69-9) (OSI Pharmaceuticals), EXEL-2880 (Exelixis), FK-778 (Sanofi-Aventis) , gefitinib (CAS No. 184475-35-2) (AstraZeneca), GW-2286 or GW-654652 (GlaxoSmithKline), her2/neu antigen from AlphaVax, HER-2/neu inhibitor from Generex, Herzyme (Medipad) (Sirna Therapeutics), HKI-272 (Wyeth), HuMax-EGFr (Genmab), idronoxil (CAS No. 81267-65-4) (Novogen), IGF-1 inhibitors from Ontogen, IMC-11F8 (ImClone Systems), kahalalide F (CAS No. 149204-42-2) (PharmaMar), KDR inhibitor from LG Life Sciences, KDR inhibitors from Abbott Laboratories, KDR kinase inhibitors (UCB), Kdr kinase inhibitors from Merck & Co, KRN-633 and KRN-951 (Kirin Brewery), KSB-102 (Xenova), lapatinib ditosylate (CAS No. 388082-78-8) (GlaxoSmithKline), matuzumab (Merck KGaA), MDX-214 (Medarex), ME-103 (Pharmexa), MED-A300 (Gerolymatos), MNAC-13 (Lay Line Genomics), nimotuzumab (Center of Molecular Immunology), NSC-330507 or NSC-707545 (NIH)1 NV-50 (Novogen), OSI-930 (OSl Pharmaceuticals), panitumumab (Abgenix), pelitinib (CAS No. 287933-82-7) (Wyeth), pertuzumab (CAS No. 380610-27- 5) (Genentech), Pharmaprojects No. 3985 (Sanofi-Aventis), prostate cancer therapeutics from Sequenom (SQPC35, SQPC36, SQPC90), removab and remoxab (Trion Pharma), RG-13022 (CAS No. 136831-48-6), RG-13291 (CAS No. 138989-50-1), or RG-14620 (CAS No. 136831-49-7) (Sanofi- Aventis), RM-6427 (Romark), RNAi breast cancer therapy from Benitec, RP 53801 (CAS No. 125882-88-4) (Sanofi-Aventis), sorafenib tosylate (Bayer), SU-11657 (Pfizer), Tie-2 antagonists from Semaia (Hybrigenics), Tie-2 inhibitors from Ontogen, trastuzumab (CAS No. 180288-69-1) (Genentech), tyrosine kinase inhibitors from Sanofi-Aventis, U3-1287, U3-1565, U3-1784, U3-1800 (U3 Pharma), vatalanib (Novartis), VEGFR-2 kinase inhibitor from Bristol-Myers Squibb, XL-647 (Exelixis), ZD-6474 (AstraZeneca), ZK-CDK (Schering AG), an EGFR tyrosine kinase inhibitor, EKB-569 (Wyeth), and an analogue or derivative thereof.
1293. The device of item 1191 wherein the agent is a VEGF inhibitor.
1294. The device of item 1191 wherein the agent is a vitamin D receptor agonist.
1295. The device of item 1191 wherein the agent is ZD-6474 (an angiogenesis inhibitor).
1296. The device of item 1191 wherein the agent is AP-23573 (an mTOR inhibitor).
1297. The device of item 1191 wherein the agent is synthadotin (a tubulin antagonist).
1298. The device of item 1191 wherein the agent is S-0885 (a collagenase inhibitor).
1299. The device of item 1191 wherein the agent is aplidine (an elongation factor-1 alpha inhibitor). 1300. The device of item 1191 wherein the agent is ixabepilone (an epithilone).
1301. The device of item 1191 wherein the agent is IDN-5390 (an angiogenesis inhibitor).
1302. The device of item 1191 wherein the agent is SB- 2723005 (an angiogenesis inhibitor).
1303. The device of item 1191 wherein the agent is ABT-518 (an angiogenesis inhibitor).
1304. The device of item 1191 wherein the agent is combretastatin (an angiogenesis inhibitor).
1305. The device of item 1191 wherein the agent is . anecortave acetate (an angiogenesis inhibitor).
1306. The device of item 1191 wherein the agent is SB- 715992 (a kinesin antagonist).
1307. The device of item 1191 wherein the agent is temsirolimus (an mTOR inhibitor).
1308. The device of item 1191 wherein the agent is adalimumab (a TNFα antagonist).
1309. The device of item 1191, further comprising a polymer. 1310. The device of item 1191 wherein the anti-scarring agent inhibits adhesion between the device and a host into which the device is implanted.
1311. The device of item 1191 wherein the device delivers the anti-scarring agent locally to tissue proximate to the device.
1312. The device of item 1191 , further comprising a coating, wherein the coating comprises the anti-scarring agent.
1313. The device of item 1191 , further comprising a coating, wherein the coating is disposed on a surface of the device.
1314. The device of item 1191 , further comprising a coating, wherein the coating directly contacts the device.
1315. The device of item 1191 , further comprising a coating, wherein the coating indirectly contacts the device.
1316. The device of item 1191 , further comprising a coating, wherein the coating partially covers the device.
1317. The device of item 1191 , further comprising a coating, wherein the coating completely covers the device.
1318. The device of item 1191 , further comprising a coating, wherein the coating is a uniform coating.
1319. The device of item 1191 , further comprising a coating, wherein the coating is a non-uniform coating. 1320. The device of item 1191 , further comprising a coating, wherein the coating is a discontinuous coating.
1321. The device of item 1191 , further comprising a coating, wherein the coating is a patterned coating.
1322. The device of item 1191, further comprising a coating, wherein the coating has a thickness of 100 Dm or less.
1323. The device of item 1191 , further comprising a coating, wherein the coating has a thickness of 10 Dm or less.
1324. The device of item 1191 , further comprising a coating, wherein the coating adheres to the surface of the device upon deployment of the device.
1325. The device of item 1191 , further comprising a coating, wherein the coating is stable at room temperature for a period of 1 year.
1326. The device of item 1191 , further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 0.0001% to about 1% by weight.
1327. The device of item 1191, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 1% to about 10% by weight.
1328. The device of item 1191, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 10% to about 25% by weight. 1329. The device of item 1191 , further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 25% to about 70% by weight.
1330. The device of item 1191 , further comprising a coating, wherein the coating further comprises a polymer.
1331. The device of item 1191 , further comprising a first coating having a first composition and the second coating having a second composition.
1332. The device of item 1191 , further comprising a first coating having a first composition and the second coating having a second composition, wherein the first composition and the second composition are different.
1333. The device of item 1191 , further comprising a polymeric carrier, wherein the polymeric carrier comprises a copolymer.
1334. The device of item 1191 , further comprising a polymeric carrier, wherein the polymeric carrier comprises a block copolymer.
1335. The device of item 1191 , further comprising a polymeric carrier, wherein the polymeric carrier comprises a random copolymer.
1336. The device of item 1191, further comprising a polymeric carrier, wherein the polymeric carrier comprises a biodegradable polymer.
1337. The device of item 1191 , further comprising a polymeric carrier, wherein the polymeric carrier comprises a non-biodegradable polymer. 1338. The device of item 1191 , further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrophilic polymer.
1339. The device of item 1191, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrophobic polymer.
1340. The device of item 1191 , further comprising a polymeric carrier, wherein the polymeric carrier comprises a polymer having hydrophilic domains.
1341. The device of item 1191, further comprising a polymeric carrier, wherein the polymeric carrier comprises a polymer having hydrophobic domains.
1342. The device of item 1191 , further comprising a polymeric carrier, wherein the polymeric carrier comprises a non-conductive polymer.
1343. The device of item 1191, further comprising a polymeric carrier, wherein the polymeric carrier comprises an elastomer.
1344. The device of item 1191, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrogel.
1345. The device of item 1191 , further comprising a polymeric carrier, wherein the polymeric carrier comprises a silicone polymer.
1346. The device of item 1191, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrocarbon polymer.
1347. The device of item 1191 , further comprising a polymeric carrier, wherein the polymeric carrier comprises a styrene-derived polymer. 1348. The device of item 1191 , further comprising a polymeric carrier, wherein the polymeric carrier comprises a butadiene polymer.
1349. The device of item 1191, further comprising a polymeric carrier, wherein the polymeric carrier comprises a macromer.
1350. The device of item 1191 , further comprising a polymeric carrier, wherein the polymeric carrier comprises a poly(ethylene glycol) polymer.
1351. The device of item 1191 , further comprising a polymeric carrier, wherein the polymeric carrier comprises an amorphous polymer.
1352. The device of item 1191 , further comprising a lubricious coating.
1353. The device of item 1191 wherein the anti-scarring agent is located within pores or holes of the device.
1354. The device of item 1191 wherein the anti-scarring agent is located within a channel, lumen, or divet of the device.
1355. The device of item 1191, further comprising a second pharmaceutically active agent.
1356. The device of item 1191 , further comprising an antiinflammatory agent.
1357. The device of item 1191 , further comprising an agent that inhibits infection. 1358. The device of item 1191 , further comprising an agent that inhibits infection, wherein the agent is an anthracycline.
1359. The device of item 1191 , further comprising an agent that inhibits infection, wherein the agent is doxorubicin.
1360. The device of item 1191, further comprising an agent that inhibits infection, wherein the agent is mitoxantrone.
1361. The device of item 1191 , further comprising an agent that inhibits infection, wherein the agent is a fluoropyrimidine.
1362. The device of item 1191 , further comprising an agent that inhibits infection, wherein the agent is 5-fluorouracil (5-FU).
1363. The device of item 1191 , further, comprising an agent that inhibits infection, wherein the agent is a folic acid antagonist.
1364. The device of item 1191, further comprising an agent that inhibits infection, wherein the agent is methotrexate.
1365. The device of item 1191 , further comprising an agent that inhibits infection, wherein the agent is a podophylotoxin.
1366. The device of item 1191 , further comprising an agent that inhibits infection, wherein the agent is etoposide.
1367. The device of item 1191, further comprising an agent that inhibits infection, wherein the agent is a camptothecin. 1368. The device of item 1191 , further comprising an agent that inhibits infection, wherein the agent is a hydroxyurea.
1369. The device of item 1191 , further comprising an agent that inhibits infection, wherein the agent is a platinum complex.
1370. The device of item 1191 , further comprising an agent that inhibits infection, wherein the agent is cisplatin.
1371. The device of item 1191, further comprising an antithrombotic agent.
1372. The device of item 1191, further comprising a visualization agent.
1373. The device of item 1191 , further comprising a visualization agent, wherein the visualization agent is a radiopaque material, wherein the radiopaque material comprises a metal, a halogenated compound, or a barium containing compound.
1374. The device of item 1191 , further comprising a visualization agent, wherein the visualization agent is a radiopaque material, wherein the radiopaque material comprises barium, tantalum, or technetium.
1375. The device of item 1191 , further comprising a visualization agent, wherein the visualization agent is a MRI responsive material.
1376. The device of item 1191, further comprising a visualization agent, wherein the visualization agent comprises a gadolinium chelate. 1377. The device of item 1191 , further comprising a visualization agent, wherein the visualization agent comprises iron, magnesium, manganese, copper, or chromium.
1378. The device of item 1191 , further comprising a visualization agent, wherein the visualization agent comprises an iron oxide compound.
1379. The device of item 1191 , further comprising a visualization agent, wherein the visualization agent comprises a dye, pigment, or colorant.
1380. The device of item 1191, further comprising an echogenic material.
1381. The device of item 1191 , further comprising an echogenic material, wherein the echogenic material is in the form of a coating.
1382. The device of item 1191 wherein the device is sterile.
1383. The device of item 1191 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device.
1384. The device of item 1191 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is connective tissue. 1385. The device of item 1191 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is muscle tissue.
1386. The device of item 1191 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is nerve tissue.
1387. The device of item 1191 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is epithelium tissue.
1388. The device of item 1191 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from the time of deployment of the device to about 1 year.
1389. The device of item 1191 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from about 1 month to 6 months.
1390. The device of item 1191 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from about 1 - 90 days.
1391. The device of item 1191 wherein the anti-scarring agent is released in effective concentrations from the device at a constant rate.
1392. The device of item 1191 wherein the anti-scarring agent is released in effective concentrations from the device at an increasing rate. 1393. The device of item 1191 wherein the anti-scarring agent is released in effective concentrations from the device at a decreasing rate.
1394. The device of item 1191 wherein the anti-scarring agent is released in effective concentrations from the composition comprising the anti-scarring agent by diffusion over a period ranging from the time of deployment of the device to about 90 days.
1395. The device of item 1191 wherein the anti-scarring agent is released in effective concentrations from the composition comprising the anti-scarring agent by erosion of the composition over a period ranging from the time of deployment of the device to about 90 days.
1396. The device of item 1191 wherein the device comprises about 0.01 Dg to about 10 Dg of the anti-scarring agent.
1397. The device of item 1191 wherein the device comprises about 10 Dg to about 10 mg of the anti-scarring agent.
1398. The device of item 1191 wherein the device comprises about 10 mg to about 250 mg of the anti-scarring agent.
1399. The device of item 1191 wherein the device comprises about 250 mg to about 1000 mg of the anti-scarring agent.
1400. The device of item 1191 wherein the device comprises about 1000 mg to about 2500 mg of the anti-scarring agent.
1401. The device of item 1191 wherein a surface of the device comprises less than 0.01 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied. 1402. The device of item 1191 wherein a surface of the device comprises about 0.01 Dg to about 1 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
1403. The device of item 1191 wherein a surface of the device comprises about 1 Dg to about 10 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
1404. The device of item 1191 wherein a surface of the device comprises about 10 Dg to about 250 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
1405. The device of item 1191 wherein a surface of the device comprises about 250 Dg to about 1000 Dg of the anti-scarring agent of anti- scarring agent per mm2 of device surface to which the anti-scarring agent is applied. -
1406. The device of item 1191 wherein a surface of the device comprises about 1000 Dg to about 2500 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
1407. The device of any one of items 1191-1406, wherein the device is a lacrimal duct stent.
1408. The device of any one of items 1191-1406, wherein the device is an Eustachian tube stent.
1409. The device of any one of items 1191-1406, wherein the device is a nasal stent. 1410. The device of any one of items 1191-1406, wherein the device is a sinus stent.
1411. A device, comprising an ear ventilation tube (e.g., an implant) and an anti-scarring agent or a composition comprising an anti- scarring agent, wherein the agent inhibits scarring between the device and a host into which the device is implanted.
1412. The device of item 1411 wherein the agent is an adensosine A2A receptor antagonist.
1413. The device of item 1411 wherein the agent is an AKT inhibitor.
1414. The device of item 1411 wherein the agent is an alpha 2 integrin antagonist, wherein the alpha 2 integrin antagonist is - Pharmaprojects No. 5754 (Merck KgaA).
1415. The device of item 1411 wherein the agent an alpha 4 integrin antagonist.
1416. The device of item 1411 wherein the agent is an alpha 7 nicotinic receptor agonist.
1417. The device of item 1411 wherein the agent is an angiogenesis inhibitor selected from the group consisting of AG-12,958 (Pfizer), ATN-161 (Attenuon LLC), neovastat, an angiogenesis inhibitor from Jerina AG (Germany), NM-3 (Mercian), VGA-1155 (Taisho), FCE-26644 (Pfizer), FCE-26950 (Pfizer), FPMA (Meiji Daries), FR-111142 (Fujisawa), GGTI-298, GM-1306 (Ligand), GPA-1734 (Novartis), NNC-47-0011 (Novo Nordisk), herbamycin (Nippon Kayaku), lenalidomide (Celegene), IP-10 (NIH), ABT-828 (Abbott), KIN-841 (Tokushima University, Japan), SF-1126 (Semafore Pharmaceuticals), laminin technology (NIH), CHIR-258 (Chiron), NVP-AEW541 (Novartis), NVP-AEW541 (Novartis), Vt16907 (Alchemia), OXI-8007 (Oxigene), EG-3306 (Ark Therapeutics), Maspin (Arriva), ABT- 567 (Abbott), PPI-2458 (Praecis Pharmaceuticals), CC-5079, CC-4089 (Celgene), HIF-1 alpha inhibitors (Xenova), S-247 (Pfizer), AP-23573 (Ariad), AZD-9935 (Astra Zeneca), mebendazole (Introgen Therapeutics), MetAP-2 inhibitors (GlaxoSmithKline), AG-615 (Angiogene Pharmaceuticals), Tie-2 antagonists (Hybrigenics), NC-381 , CYC-381, NC-169, NC-219, NC-383, NC-384, NC-407 (Lorus Therapeutics), ATN-224 (Attenuon), ON-01370 (Onconova), Vitronectin antagonists (Amgen), SDX-103 (Salmedix), Vitronectin antagonists (Shire), CHP (Riemser), TEK (Amgen), Anecortave acetate (Alcon), T46.2 (Matrix Therapeutics), HG-2 (Heptagen), TEM antagonists (Genzyme), Oxi-4500 (Oxigene), ATN-161 (Attenuon), WX-293 (Wilex), M-2025 (Metris Therapeutics), Alphastatin (BioActa), YH-16 (Yantai Rongchang), BIBF-1120 (Boehringer Ingelheim), BAY-57-9352 (Bayer), ASr 1404 (Cancer Research Technology), SC-77964 (Pfizer), glycomimetics (BioTie Therapies), TIE-2 Inhibitors (Ontogen), DIMI, Octamer (Octamer), ABR-215050 (Active Biotech), ABT-518 (Abbott), KDR inhibitors (Abbott), BSF-466895 (Abbott), SCH-221153 (Schering-Plough), DAC:antiangiogenic (ConjuChem), TFPI (EntreMed), AZD-2171 (Astra-Zenaca), CDC-394 (Celgene), LY290293 (EIi Lilly), IDN-5390 (Indena), Kdr Kinase Inhibitors (Merck), CT-113020, CT-116433, CT-116563, CT-31890, CT-32228) (Cell Therapeutics), A-299620 (Abbott), TWEAK Inhibitor (Amgen), VEGF modulators (Johnson and Johnson), Tum-N53, tumstatin (Genzyme), Thios- 1, Thios-2 (Thios Pharmaceuticals), MV-6401 (Miravant Medical Technologies), Spisulosine (PharmaMar), CEP-7055 (Cephalon), AUV-201 (Auvation), LM-609 (EIi Lilly), SKF-106615 (AnorMED), Oglufanide disodium (Cytran), BW-114 (Pharminox), Calreticulin (NIH), WX-678 (Wilex), SD-7784 (Pfizer), WX-UK1 (Wilex), SH-268 (Schering AG), 2-Me-PGA (Celgene), S- 137 (Pfizer), ZD-6126 (Angiogene Pharmaceuticals), SG-292 (SignalGen), Benefin (Lane Labs), A6, A36 (Angstrom), SB-2723005 (GlaxoSmithKline), SC-7 (Cell Therapeutics), ZEN-014 (AEtema Zentaris), 2-methoxyestradiol (EntreMed), NK-130119 (Nippon Kayaku), CC-10004 (Celgene), AVE- 8062A (Ajinomoto), Tacedinaline (Pfizer), Actinonin (Tokyo Metropolitan Institute of Medical Science), Lenalidomide (Celgene), VGA-1155, BTO-956 (SRI International), ER-68203-00 (Eisai), CT-6685 (UCB), JKC-362 (Phoenix Pharmaceuticals), DMI-3798 (DMI Biosciences, Angiomate (Ipsen), ZD-6474 (AstraZeneca), CEP-5214 (Cephalon), Canstatin (Genzyme), NM-3 (Mercian), Oridigm (MediQuest Therapeutics), Exherin (Adherex), BLS-0597 (Boston Life Sciences), PTC-299 (PTC Therapeutics), NPI-2358 (Nereus Pharmaceuticals), CGP-79787 (Novartis), AEE-788 (Novartis), CKD-732 (Chong Kun Dang), CP-564959 (OSI Pharmaceuticals), CM-101 (CarboMed), CT-2584, CT3501 (Cell Therapeutics), combretastatin and analogues and derivatives thereof (Oxigene), Rebimastat (Bristol-Meyers Squibb), Dextrin 2-sulfate (ML Laboratories), Cilengitide (Merk KGaA), NSC-706704 (Pharminox), KRN- 951 (Kirin Brewery), Ukrain, NSC-631570 (Nowicky Pharma), Tecogalan sodium (Daiichi Pharmaceutical), Tz-93 (Tsumura), TBC-1635 (Encysive Pharmaceuticals), TAN-1120 (Takeda), Semaxanib (Pfizer), BDI-7800 (Biopharmacopae), SD-186, SD-983 (Bristol-Meyers Squibb), SB-223245 (GlaxoSmithKline), SC-236 (Pfizer), RWJ-590973 (Johnson and Johnson), ILX-1850 (Genzyme), SC-68488, S-836 (Pfizer), CG-55069-11 (CuraGen), Ki-23057 (Kirin Brewery), CCX-700 (Chemoentryx), Pegaptanib octasodium (Giled Sciences), ANGIOCOL (available from Biostratum Inc.), or an analogue or derivative thereof.
1418. The device of item 1411 wherein the agent is an apoptosis antagonist.
1419. The device of item 1411 wherein the agent is an apoptosis activator. 1420. The device of item 1411 wherein the agent is a beta 1 integrin antagonist.
1421. The device of item 1411 wherein the agent is a beta tubulin inhibitor.
1422. The device of item 1411 wherein the agent is a blocker of enzyme production in Hepatitis C.
1423. The device of item 1411 wherein the agent is a Bruton's tyrosine kinase inhibitor.
1424. The device of item 1411 wherein the agent is a calcineurin inhibitor.
1425. The device of item 1411 wherein the agent is a caspase 3 inhibitor.
1426. The device of item 1411 wherein the agent is a CC chemokine receptor antagonist.
1427. The device of item 1411 wherein the agent is a cell cycle inhibitor selected from the group consisting of SNS-595 (Sunesis), synthadotin, KRX-0403, and an analogue or derivative thereof.
1428. The device of item 1411 wherein the agent is a cathepsin B inhibitor.
1429. The device of item 1411 wherein the agent is a cathepsin K inhibitor, wherein the cathepsin K inhibitor is 462795 (GlaxoSmithKIine), INPL-022-D6 (Amura Therapeutics), an analogue or derivative thereof).
1430. The device of item 1411 wherein the agent is a cathepsin L inhibitor.
1431. The device of item 1411 wherein the agent is a CD40 antagonist.
1432. The device of item 1411 wherein the agent is a chemokine receptor agonist.
1433. The device of item 1411 wherein the agent is a chymase inhibitor.
1434. The device of item 1411 wherein the agent is a collagenase antagonist.
1435. The device of item 1411 wherein the agent is a CXCR antagonist.
1436. The device of item 1411 wherein the agent is a cyclin dependent kinase inhibitor selected from the group consisting of a CDK-1 inhibitor, a CDK-2 inhibitor, a CDK- 4 inhibitor, a CDK-6 inhibitor, a CAK1 inhibitor from GPC Biotech and Bristol-Myers Squibb, RGB-286199 (GPC Biotech), an anticancer agent from Astex Technology, a CAK1 inhibitor from GPC Biotech, a CDK inhibitor from Sanofi-Aventis, a CDK1/CDK2 inhibitor from Amgen, a CDK2 inhibitor from SUGEN-2 (Pfizer), a hearing loss therapy agent (Sound Pharmaceuticals), PD-0332991 (Pfizer), RGB-286199 (GPC Biotech), Ro-0505124 (Hoffmann-La Roche), a Ser/Thr kinase inhibitor from Lilly (EIi Lilly), CVT-2584 (CAS No. 199986-75-9) (CV Therapeutics), and an analogue or derivative thereof.
1437. The device of item 1411 wherein the agent is a cyclooxygenase 1 inhibitor.
1438. The device of item 1411 wherein the agent is a DHFR inhibitor.
1439. The device of item 1411 wherein the agent is a dual integrin inhibitor.
1440. The device of item 1411 wherein the agent is an elastase inhibitor.
1441. The device of item 1411 wherein the agent is an elongation factor-1 alpha inhibitor.
1442. The device of item 1411 wherein the agent is an endothelial growth factor antagonist.
1443. The device of item 1411 wherein the agent is an endothelial growth factor receptor kinase inhibitor selected from the group consisting of sorafenib tosylate (Bayer), AAL-993 (Novartis), ABP-309 (Novartis), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), E- 7080 (Eisai), EG-3306 (Ark Therapeutics), EXEL-2880 (Exelixis), GW- 654652 (GlaxoSmithKline), a KDR inhibitor from LG Life Sciences, CT-6685 and CT-6729 (UCB), KRN-633 and KRN-951 (Kirin Brewery), OSI-930 (OSI Pharmaceuticals), SP-5.2 (Supratek Pharma), SU-11657 (Pfizer), a Tie-2 antagonist (Hybrigenics), a VEGFR-2 kinase inhibitor (Bristol-Myers Squibb), XL-647 (Exelixis), a KDR inhibitor from Abbott Laboratories, sorafenib tosylate, and an analogue or derivative thereof.
1444. The device of item 1411 wherein the agent is an endotoxin antagonist.
1445. The device of item 1411 wherein the agent is an epothilone and tubulin binder.
1446. The device of item 1411 wherein the agent is an estrogen receptor antagonist.
1447. The device of item 1411 wherein the agent is an FGF inhibitor.
1448. The device of item 1411 wherein the agent is a farnexyl transferase inhibitor.
1449. The device of item 1411 wherein the agent is famesyltransferase inhibitor selected from the group of A-197574 (Abbott), a famesyltransferase inhibitor from Servier, FPTIII (Strathclyde Institute for Drug R), LB-42908 (LG Life Sciences), Pharmaprojects No. 5063 (Genzyme), Pharmaprojects No. 5597 (Ipsen), Yissum Project No. B-1055 (Yissum), and an analogue or derivative thereof.
1450. The device of item 1411 wherein the agent is an FLT-3 kinase inhibitor.
1451. The device of item 1411 wherein the agent is an FGF receptor kinase inhibitor. 1452. The device of item 1411 wherein the agent is a fibrinogen antagonist selected from the group consisting of AUV-201 (Auvation), MG-13926 (Sanofi-Aventis), plasminogen activator (CAS No. 105913-11-9) (from Sanofi-Aventis or UCB), plasminogen activator-2 (tPA- 2) (Sanofi-Aventis), pro-urokinase (CAS No. 82657-92-9) (Sanofi-Aventis), and an analogue or derivative thereof.
1453. The device of item 1411 wherein the agent is a heat shock protein 90 antagonist selected from the group consisting of SRN-005 (Sirenade), geldanamycin, NSC-33050 (17-allylaminogeldanamycin; 17- AAG), 17-dimethylaminoethylamino-17-demethoxy-geldanamycin (17- DMAG), rifabutin (rifamycin XIV, 1l,4-didehydro-1-deoxy-1 ,4-dihydro-5'-(2- methylpropyl)-1-oxo-), and an analogue or derivative thereof.
1454. The device of item 1411 wherein the agent is a histone deacetylase inhibitor.
1455. The device of item 1411 wherein the agent is an HMGCoA reductase inhibitor selected from the group consisting of an atherosclerosis therapeutic from Lipid Sciences, ATI-16000 (ARYx Therapeutics), KS-01-019 (Kos Pharmaceuticals), Pharmaprojects No. 2197 (Sanofi-Aventi), RP 61969 (Sanofi-Aventis), and an analogue or derivative thereof.
1456. The device of item 1411 wherein the agent is an ICAM inhibitor.
1457. The device of item 1411 wherein the agent is an IL, ICE and IRAK antagonist, wherein the antagonist is a CJ-14877, CP-424174 (Pfizer), NF-61 (Negma-Lerads), and an analogue or derivative thereof. 1458. The device of item 1411 wherein the agent is an IL-2 inhibitor.
1459. The device of item 1411 wherein the agent is an immunosuppressant selected from the group consisting of teriflunomide (Sanofi Aventis), chlorsulfaquinoxalone (NSC-339004), chlorsulfaquinoxalone sulfate, CS-712 (Sankyo), ismomultin alfa (CAS No. 457913-93-8) (Akzo Nobel), antiallergics from GenPat77, antiinflammatories or AT-005 (Androclus Therapeutics), autoimmune disease therapeutics from EpiVax, BN-007 (Bone), budesonide (CAS No. 51333-22- 3) (MAP Pharmaceuticals), CO-14 (Genzyme), edratide (CAS No. 433922- 67-9) (Teva), EP-314 (Enanta), eprovafen (CAS No. 101335-99-3) (Sanofi- Aventis), HWA-131 (CAS No. 118788-41-3) (Sanofi-Aventis), immunomodulators from MerLion Pharmaceuticals, immunosuppressives from Alchemia, IPL-12 (Inflazyme), MDL-9563 (CAS No. 27086-86-8) (Sanofi-Aventis), Pharmaprojects No. 2330 (Sanofi-Aventis),- Pharmaprojects No. 6426 (Abgenix), PXS-25 (Pharmaxis), rosmarinic acid (CAS No. 20283-92-5) (Sanofi-Aventis), RP 42927 or RP 54745 (CAS No. 135330-08-4) (Sanofi-Aventis), SGN-35 (Seattle Genetics), ST-1959 (Sigma-Tau), type I diabetes therapy from SYNX Pharma, UNIL-88 (Debiopharm), VP-025 (Vasogen), VR-694 (Vectura), PRTX-001 (Protalex), and an analogue or derivative thereof.
1460. The device of item 1411 wherein the agent is an IMPDH (inosine monophosphate).
1461. The device of item 1411 wherein the agent is an integrin antagonist.
1462. The device of item 1411 wherein the agent is an interleukin antagonist. 1463. The device of item 1411 wherein the agent is an inhibitor of type III receptor tyrosine kinase.
1464. The device of item 1411 wherein the agent is an irreversible inhibitor of enzyme methionine aminopeptidase type 2.
1465. The device of item 1411 wherein the agent is an isozyme selective delta protein kinase C inhibitor.
1466. The device of item 1411 wherein the agent a JAK3 enzyme inhibitor.
1467. The device of item 1411 wherein the agent is a JNK inhibitor.
1468. The device of item 1411 wherein the agent is a kinase inhibitor.
1469. The device of item 1411 wherein the agent is kinesin antagonist.
1470. The device of item 1411 wherein the agent is a kinesin antagonist.
1471. The device of item 1411 wherein the agent is a leukotriene inhibitor and antagonist selected from the group consisting of ambicromil (CAS No. 58805-38-2) (Sanofi-Aventis), amelubant (CAS No. 346735-24-8) (Boehringer Ingelheim), DW-1141 (Dong Wha), ebselen (Daiichi Pharmaceutical), ibudilast (Kyorin), leucotriene inhibitors from Sanofi-Aventis, lymphotoxin-beta receptor (LT-ϋ) from Biogen Idee, Pharmaprojects No. 1535 and 2728 (CAS No. 119340-33-9) (Sanofi- Aventis), R-112 (Rigel), Rev-5367 (CAS No. 92532-05-3) (Sanofi-Aventis), RG-14893 (CAS No. 141835-49-6) (Sanofi-Aventis), RG-5901-A (CAS No. 101910-24-1), 92532-23-5, RP 66153 (CAS No. 142422-79-5), RP 66364 (CAS No.186916-92-5), RP 69698 (CAS No. 141748-00-7) (Sanofi-Aventis), SC-411930 (Pfizer), SC-41930 (CAS No. 120072-59-5) (Pfizer), SC-50605 (CAS No. 138828-39-4) (Pfizer), SC-51146 (CAS No. 141059-52-1), SC- 53228 (CAS No. 153633-01-3) (Pfizer), spaglumic acid (ZY-15106) (CAS No.3106-85-2), 80619-64-3 (Novartis), tipredane (CAS No. 85197-77-9) (Bristol-Myers Squibb), U-75302 (CAS No. 119477-85-9) (Pfizer), and analogue or derivative thereof.
1472. The device of item 1411 wherein the agent is an MAP kinase inhibitor.
1473. The device of item 1411 wherein the agent is a matrix metalloproteinase inhibitor. -
1474. The device of item 1411 wherein the agent is an MCP- CCR2 inhibitor.
1475. The device of item 1411 wherein the agent is an mTOR inhibitor.
1476. The device of item 1411 wherein the agent is an mTOR kinase inhibitor.
1477. The device of item 1411 wherein the agent is a microtubule inhibitor selected from the group consisting of antibody- maytansinoid conjugates from Biogen Idee, colchicines (MantiCore Pharmaceuticals), anticancer immunoconjugates from Johnson & Johnson, DIME from Octamer, gni-1f (GNI), huC242-DM4, huMy9-6-DM1 (ImmunoGen), IDN-5404 (Indena), IMO-098, IMOderm (Imotep), mebendazole (Introgen Therapeutics), microtubule poisons from Cambridge Enterprise, paclitaxel such as LOTAX from Aphios (CAS No. 33069-62-4), Genexol-PM from Samyang, Pharmaprojects No. 6383 (Tapestry Pharmaceuticals), RPR-112378 (Sanofi-Aventis), SGN-75 (Seattle Genetics), SPL-7435 (Starpharma), SSR-250411 (Sanofi-Aventis), trastuzumab-DM1 (Genentech), vinorelbine, and an analogue or derivative thereof.
1478. The device of item 1411 wherein the agent is an MIF inhibitor.
1479. The device of item 1411 wherein the agent is an MMP inhibitor.
1480. The device of item 1411 wherein the agent-is a neurokinin (NK) antagonist selected from the group consisting of anthrotainin (CAS No. 148084-40-6) (Sanofi-Aventis), an IBS thereapeutic from ArQuIe, MDL-105212A (CAS No. 167261-60-1) (Ssanofi-Aventis), Pharmaprojects No. 2744, 3258 (CAS No. 139167-47-8) 4006, 4201 , or 5986 (Sanofi-Aventis), RP 67580 (CAS No. 135911-02-3), SR-144190 (CAS No. 201152-86-5), SSR-240600, SSR-241586 (Sanofi-Aventis), TKA-457 (Novartis), vestipitant mesylate (CAS No. 334476-64-1) (GlaxoSmithKline), Win-64821 (Sanofi-Aventis), PRX-96026 (Predix Pharmaceuticals), and an analogue or derivative thereof.
1481. The device of item 1411 wherein the agent is an NF kappa B inhibitor selected from the group consisting of AVE-0545 or AVE- 0547 (Sanofi-Aventis), bortezomib (CAS No. 179324-69-7) (Millennium Pharmaceuticals), dexanabinol (CAS No. 112924-45-5) (Pharmos), dexlipotam (Viatris), Pharmaprojects No. 6283 (INDRA) (OXiGENE), IPL- 576092 (CAS No. 137571-30-3) (Inflazyme), NFKB decoy (Corgentech), NFKB decoy oligo (AnGes MG), NFKB's from Ariad, osteoporosis treatments or S5 (F005) from Fulcrum Pharmaceuticals, P61 (Phytopharm), R-flurbiprofen (CAS No. 5104-49-4) (Encore Pharmaceuticals), and an analogue or derivative thereof.
1482. The device of item 1411 wherein the agent is a nitric oxide agonist.
1483. The device of item 1411 wherein the agent is an ornithine decarboxylase inhibitor.
1484. The device of item 1411 wherein the agent is a p38 MAP kinase inhibitor selected from the group consisting of AZD-6703 (AstraZeneca), JX-401 (Jexys Pharmaceuticals), BMS-2 (Bristol-Myers Squibb), a p38 MAP kinase inhibitor frora Novartis, a p38-alpha MAP kinase inhibitor from Amphora, Pharmaprojects No. 5704 (Pharmacopeia), RPR- 200765A (Sanofi-Aventis), SD-282 (Johnson & Johnson), TAK-715 (Takeda), and an analogue or derivative thereof.
1485. The device of item 1411 wherein the agent is a palmitoyl-protein thioesterase inhibitor.
1486. The device of item 1411 wherein the agent is a PDGF receptor kinase inhibitor selected from the group consisting of AAL-993, AMN-107, or ABP-309 (Novartis), AMG-706 (Amgen), BAY-57-9352 (Bayer), CDP-860 (UCB), E-7080 (Eisai), imatinib (CAS No. 152459-95-5) (Novartis), OSI-930 (OSI Pharmaceuticals), RPR-127963E (Sanofi-Aventis), RWJ-540973 (Johnson & Johnson), sorafenib tosylate (Bayer), SU-11657 (Pfizer), tandutinib (CAS No. 387867-13-2) (Millennium Pharmaceuticals), vatalanib (Novartis), ZK-CDK (Schering AG), and an analogue or derivative thereof.
1487. The device of item 1411 wherein the agent is (-)- haiofenate (Metabolex), AMG-131 (Amgen), antidiabetics from Japan Tobacco, AZD-4619, AZD-8450, AZD-8677 (AstraZeneca), DRF-10945, balaglitazone (Dr Reddy's), CS-00088, CS-00098 (Chipscreen Biosciences), E-3030 (Eisai), etalocib (CAS No. 161172-51-6) (EIi Lilly), GSK-641597 (Ligand), GSK-677954 (GlaxoSmithKline), GW-409544 (Ligand), GW- 590735 (GlaxoSmithKline), K-111 (Hoffmann-La Roche), LY-518674 (EIi Lilly), LY-674 (Ligand), LY-929 (Ligand), MC-3001 , MC-3002 (MaxoCore Pharmaceuticals), metformin HCI + pioglitazone (CAS No. 1115-70-4 and 112529-15-4), ACTOPLUS MET from Andrx), muraglitazar (CAS No. 331741-94-7) (Bristol-Myers Squibb), naveglitazar (Ligand), oleoylethanolamide (Kadmus Pharmaceuticals), ONO-5129, pioglitazone hydrochloride (CAS No. 111025-46-8 and 112529-15-4) (Takeda),.PLX-204 (Plexxikon), PPAR agonists from Genfit, PPAR delta agonists from EIi Lilly, PPAR-alpha agonists from CrystalGenomics, PPAR-gamma modulators and PPAR-β modulators from CareX, rosiglitazone maleate (CAS No. 122324- 73-4 or 155141-29-0) (GlaxoSmithKline), rosiglitazone maleate/glimepir (CAS No. 155141-29-0 and 93479-97-1), AVANDARYL, rosiglitazone maleate/metformin extend (CAS No. 155141-29-0 and 657-24-9), AVANDAMET, rosiglitazone maleate+metformin, AVANDAMET (GlaxoSmithKline), tesaglitazar (AstraZeneca), and an analogue or derivative thereof.
1488. The device of item 1411 wherein the agent is a phosphatase inhibitor.
1489. The device of item 1411 wherein the agent is a phosphodiesterase (PDE) inhibitor selected from the group consisting of avanafil (Tanabe Seiyaku), dasantafil (CAS No. 569351-91-3) (Schering- Plough), A-906119 (CAS No. 134072-58-5), DL-850 (Sanofi-Aventis), GRC- 3015, GRC-3566, GRC-3886 (Glenmark), HWA-153 (CAS No. 56395-66-5) (Sanofi-Aventis), hydroxypumafentrine (Altana), IBFB-130011, IBFB-14-016, IBFB-140301, IBFB-150007, IBFB-211913 (IBFB Pharma), L-826141 (Merck & Co), medorinone (CAS No. 88296-61-1) (Sanofi-Aventis), MEM- 1917 (Memory Pharmaceuticals), ND-1251 (Neuro3d), PDE inhibitors from ICOS, PDE IV inhibitors from Memory Pharmaceuticals and CrystalGenomics, Pharmaprojects No. 2742 and 6141 (Sanofi-Aventis), QAD-171 (Novartis), RHC-2963 (CAS No. 76993-12-9 and 76993-14-1), RPR-117658, RPR-122818 derivatives, SR-24870 , and RPR-132294 (Sanofi-Aventis), SK-350 (ln2Gen), stroke therapy agents from deCODE Genetics, TAS-203 (Taiho), tofimilast (CAS No. 185954-27-2) (Pfizer), UK- 371800 (Pfizer), WIN-65579 (CAS No. 158020-82-7) (Sanofi-Aventis), IBFB- 130020 (IBFB Pharma), OPC-6535 (CAS No. 145739-56-6) (Otsuka), a phosphodiesterase III inhibitor, enoximone, a phosphodiesterase IV inhibitor, fosfosal, Atopik (Barrier Therapeutics), triflusal, a phosphodiesterase V inhibitor, and an analogue or derivative thereof.
1490. The device of item 1411 wherein the agent is a PKC inhibitor.
1491. The device of item 1411 wherein the agent is a platelet activating factor antagonist.
1492. The device of item 1411 wherein the agent is a platelet- derived growth factor receptor kinase inhibitor.
1493. The device of item 1411 wherein the agent is a prolyl hydroxylase inhibitor. 1494. The device of item 1411 wherein the agent is a polymorphonuclear neutrophil inhibitor.
1495. The device of item 1411 wherein the agent is a protein kinase B inhibitor.
1496. The device of item 1411 wherein the agent is a protein kinase C stimulant.
1497. The device of item 1411 wherein the agent is a purine nucleoside analogue.
1498. The device of item 1411 wherein the agent is a purinoreceptor P2X antagonist.
1499. The device of item 14.11 wherein the agent is a Raf kinase inhibitor.
1500. The device of item 1411 wherein the agent is a reversible inhibitor of ErbB1 and ErbB2.
1501. The device of item 1411 wherein the agent is a ribonucleoside triphosphate reductase inhibitor.
1502. The device of item 1411 wherein the agent is an SDF-1 antagonist.
1503. The device of item 1411 wherein the agent is a sheddase inhibitor. 1504. The device of item 1411 wherein the agent is an SRC inhibitor.
1505. The device of item 1411 wherein the agent is a stromelysin inhibitor.
1506. The device of item 1411 wherein the agent is an Syk kinase inhibitor.
1507. The device of item 1411 wherein the agent is a telomerase inhibitor.
1508. The device of item 1411 wherein the agent is a TGF beta inhibitor selected from the group consisting of pirfenidone (CAS No. 53179-13-8) (MARNAC), tranilast (CAS No. 53902-12-8) (Kissei), IN-1130 (ln2Gen), mannose-6-phosphate (BTG), TGF-β antagonists from Inflazyme (Pharmaprojects No. 6075), TGF-β antagonists from Sydney, non-industrial source), TGF-β I receptor kinase inhibitors from EIi Lilly, TGF-β receptor inhibitors from Johnson & Johnson, and an analogue or derivative thereof.
1509. The device of item 1411 wherein the agent is a TNFα antagonist or TACE inhibitor selected from the group consisting of adalimumab (CAS No. 331731-18-1) (Cambridge Antibody Technology), AGIX-4207 (AtheroGenics), AGT-1 (Advanced Biotherapy), an antiinflammatory from Borean Pharma, Cellzome, or Paradigm Therapeutics, anti-inflammatory vaccine (TNF-alpha kinoid) from Neovacs, humanized anti-TNF antibody or an anti-TNF MAb (CB0006) Celltech (UCB), apratastat (CAS No. 287445-51-0) (Wyeth), BMS-561392 (Bristol-Myers Squibb), BN- 006 (Bone), certolizumab pegol (CAS No. 428863-50-7 or CH-138 (UCB), cilomilast (CAS No. 153259-65-5) (GlaxoSmithKline), CR-1 (Nuada Pharmaceuticals), CRx-119 (CombinatoRx), D-5410 (UCB), dacopafant (CAS No. 125372-33-0) (Sanofi-Aventis), dersalazine (CAS No. 188913-57- 7/188913-58-8) (Uriach), etanercept (CAS No. 185243-69-0) (Amgen), ethyl pyruvate (Critical (Critical Therapeutics), golimumab (CAS No. 476181-74-5) (Johnson & Johnson), hormono-immunotherapy from Ipsen, CDP571 (e.g., Humicade from UCB), IC-485 (ICOS), infliximab (CAS No. 170277-31-3) (Johnson & Johnson), IP-751 (Manhattan Pharmaceuticals), ISIS-104838 (CAS No. 250755-32-9) (ISIS Pharmaceuticals), lenalidomide (CAS No. 191732-72-6) (Celgene), lentinan (CAS No. 37339-90-5) (Ajinomoto), MDL- 201112 (CAS No. 142130-73-2) (Sanofi-Aventis), medroxyprogesterone (CAS No. 520-85-4) (InKine Pharmaceutical), N-acetylcysteine (CAS No. 616-91-1) (Zambon), NBE-P2 (DIREVO Biotech), nerelimomab (CAS No. 162774-06-3) (Chiron), OM-294DP (OM PHARMA), onercept (CAS No. 199685-57-9) (Yeda), PASSTNF-alpha (Verigen), pentoxifylline or oxypentifylline (Sanofi-Aventis), Pharmaprojects No. 4091 , 4241 , 4295, or 4488 (Sanofi-Aventis), Pharmaprojects No. 5480 (Amgen), Pharmaprojects No. 6749 (Cengent), pirfenidone (CAS No. 53179-13-8) (MARNAC), RPR- 132294 (Sanofi-Aventis), S5 (F002) (Fulcrum Pharmaceuticals), simvastatin (CAS No. 79902-63-9) (Merck & Co), STA-6292 (Synta Pharmaceuticals), tacrolimus (CAS No. 104987-11-3) (Fujisawa LifeCycle Pharma), talactoferrin alfa (CAS No. 308240-58-6) (Agennix), thalidomide (CAS No. 50-35-1) (Celgene), TNF antagonists form ProStrakan, and Synergen, TNF inhibitors (Amgen), TNF-alpha antagonists from Dynavax Technologies and Jerina AG (Germany), TNF-alpha inhibitors from IBFB Pharma and Xencor (Xencor), torbafylline (CAS No. 105102-21-4) (Sanofi-Aventis), UR-1505 (Uriach), VT-346 (Viron Therapeutics), YSIL6 (Y1S Therapeutics), YSTH2 (Y's Therapeutics), NPI-1302a-3 (Nereus Pharmaceuticals, a TNF antagonist from Jerina AG (Germany), dersalazine, and an analogue or derivative thereof.
1510. The device of item 1411 wherein the agent is a Toll receptor inhibitor. 1511. The device of item 1411 wherein the agent is a tubulin antagonist.
1512. The device of item 1411 wherein the agent is a tyrosine kinase inhibitor selected from the group consisting of SU-011248, SUTENT from Pfizer Inc. (New York, NY)1 BMS-354825, PN-355 (Paracetsian Pharmaceuticals), AGN-199659 (Allergan), AAL-993 or ABP-309 (Novartis), adaphostin (NIH), AEE-788 (Novartis), AG-013736 (OSI Pharmaceuticals), AG-13736 (Pfizer), ALT-110 (Alteris Therapeutics), AMG-706 (Amgen), anticancer MAbs from Xencor, anti-EGFrvlll MAbs from Abgenix, anti-HER2 MAb from Abiogen, AZD-2171 or AZD-9935 (AstraZeneca), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), CEP-5214 (Cephalon), CEP- 7055 (Cephalon), cetuximab (ImClone Systems), CHIR-200131 and CHIR- 258 (Chiron), CP-547632 (OSI Pharmaceuticals), CP-724714 (Pfizer), CT- 301 (Creabilis Therapeutics), D-69491 (Baxter International), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EGFR/ErbB2 inhibitors from Array BioPharma, erlotinib (CAS No. 183319-69-9) (OSI Pharmaceuticals), EXEL-2880 (Exelixis), FK-778 (Sanofi-Aventis) , gefitinib (CAS No. 184475-35-2) (AstraZeneca), GW-2286 or GW-654652 (GlaxoSmithKline), her2/neu antigen from AlphaVax, HER-2/neu inhibitor from Generex, Herzyme (Medipad) (Sirna Therapeutics), HKI-272 (Wyeth), HuMax-EGFr (Genmab), idronoxil (CAS No. 81267-65-4) (Novogen), IGF-1 inhibitors from Ontogen, IMC-11F8 (ImClone Systems), kahalalide F (CAS No. 149204-42-2) (PharmaMar), KDR inhibitor from LG Life Sciences, KDR inhibitors from Abbott Laboratories, KDR kinase inhibitors (UCB), Kdr kinase inhibitors from Merck & Co, KRN-633 and KRN-951 (Kirin Brewery), KSB-102 (Xenova), lapatinib ditosylate (CAS No. 388082-78-8) (GlaxoSmithKline), matuzumab (Merck KGaA), MDX-214 (Medarex), ME-103 (Pharmexa), MED-A300 (Gerolymatos), MNAC-13 (Lay Line Genomics), nimotuzumab (Center of Molecular Immunology), NSC-330507 or NSC-707545 (NIH), NV-50 (Novogen), OSI-930 (OSI Pharmaceuticals), panitumumab (Abgenix), pelitinib (CAS No. 287933-82-7) (Wyeth), pertuzumab (CAS No. 380610-27- 5) (Genentech), Pharmaprojects No. 3985 (Sanofi-Aventis), prostate cancer therapeutics from Sequenom (SQPC35, SQPC36, SQPC90), removab and remoxab (Trion Pharma), RG-13022 (CAS No. 136831-48-6), RG-13291 (CAS No. 138989-50-1), or RG- 14620 (CAS No. 136831-49-7) (Sanofi- Aventis), RM-6427 (Romark), RNAi breast cancer therapy from Benitec, RP 53801 (CAS No. 125882-88-4) (Sanofi-Aventis), sorafenib tosylate (Bayer), SU-11657 (Pfizer), Tie-2 antagonists from Semaia (Hybrigenics), Tie-2 inhibitors from Ontogen, trastuzumab (CAS No. 180288-69-1) (Genentech), tyrosine kinase inhibitors from Sanofi-Aventis, U3-1287, U3-1565, U3-1784, U3-1800 (U3 Pharma), vatalanib (Novartis), VEGFR-2 kinase inhibitor from Bristol-Myers Squibb, XL-647 (Exelixis), ZD-6474 (AstraZeneca), ZK-CDK (Schering AG), an EGFR tyrosine kinase inhibitor, EKB-569 (Wyeth), and an analogue or derivative thereof.
1513. The device of item 1411 wherein, the agent is a VEGF inhibitor.
1514. The device of item 1411 wherein the agent is a vitamin D receptor agonist.
1515. The device of item 1411 wherein the agent is ZD-6474 (an angiogenesis inhibitor).
1516. The device of item 1411 wherein the agent is AP-23573 (an mTOR inhibitor).
1517. The device of item 1411 wherein the agent is synthadotin (a tubulin antagonist). 1518. The device of item 1411 wherein the agent is S-0885 (a collagenase inhibitor).
1519. The device of item 1411 wherein the agent is aplidine (an elongation factor-1 alpha inhibitor).
1520. The device of item 1411 wherein the agent is ixabepilone (an epithilone).
1521. The device of item 1411 wherein the agent is IDN-5390 (an angiogenesis inhibitor).
1522. The device of item 1411 wherein the agent is SB- 2723005 (an angiogenesis inhibitor).
1523. The device of item 1411 wherein the agent is ABT-518 (an angiogenesis inhibitor).
1524. The device of item 1411 wherein the agent is combretastatin (an angiogenesis inhibitor).
1525. The device of item 1411 wherein the agent is anecortave acetate (an angiogenesis inhibitor).
1526. The device of item 1411 wherein the agent is SB- 715992 (a kinesin antagonist).
1527. The device of item 1411 wherein the agent is temsirolimus (an mTOR inhibitor). 1528. The device of item 1411 wherein the agent is adalimumab (a TNFα antagonist).
1529. The device of item 1411, further comprising a polymer.
1530. The device of item 1411 wherein the anti-scarring agent inhibits adhesion between the device and a host into which the device is implanted.
1531. The device of item 1411 wherein the device delivers the anti-scarring agent locally to tissue proximate to the device.
1532. The device of item 1411 , further comprising a coating, wherein the coating comprises the anti-scarring agent.
1533. The device of item 1411, further comprising a coating; wherein the coating is disposed on a surface of the device.
1534. The device of item 1411, further comprising a coating, wherein the coating directly contacts the device.
1535. The device of item 1411 , further comprising a coating, wherein the coating indirectly contacts the device.
1536. The device of item 1411, further comprising a coating, wherein the coating partially covers the device.
1537. The device of item 1411 , further comprising a coating, wherein the coating completely covers the device. 1538. The device of item 1411 , further comprising a coating, wherein the coating is a uniform coating.
1539. The device of item 1411 , further comprising a coating, wherein the coating is a non-uniform coating.
1540. The device of item 1411 , further comprising a coating, wherein the coating is a discontinuous coating.
1541. The device of item 1411, further comprising a coating, wherein the coating is a patterned coating.
1542. The device of item 1411, further comprising a coating, wherein the coating has a thickness of 100 Dm or less.
1543. The device of item 1411, further comprising a coating, wherein the coating has a thickness of 10 Dm or less.
1544. The device of item 1411, further comprising a coating, wherein the coating adheres to the surface of the device upon deployment of the device.
1545. The device of item 1411, further comprising a coating, wherein the coating is stable at room temperature for a period of 1 year.
1546. The device of item 1411, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 0.0001% to about 1% by weight. 1547. The device of item 1411 , further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 1% to about 10% by weight.
1548. The device of item 1411 , further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 10% to about 25% by weight.
1549. The device of item 1411 , further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 25% to about 70% by weight.
1550. The device of item 1411 , further comprising a coating, wherein the coating further comprises a polymer.
1551. The device of item 1411, further comprising a first coating having a first composition and the second coating having a second composition.
1552. The device of item 1411 , further comprising a first coating having a first composition and the second coating having a second composition, wherein the first composition and the second composition are different.
1553. The device of item 1411, further comprising a polymeric carrier, wherein the polymeric carrier comprises a copolymer.
1554. The device of item 1411 , further comprising a polymeric carrier, wherein the polymeric carrier comprises a block copolymer. 1555. The device of item 1411 , further comprising a polymeric carrier, wherein the polymeric carrier comprises a random copolymer.
1556. The device of item 1411 , further comprising a polymeric carrier, wherein the polymeric carrier comprises a biodegradable polymer.
1557. The device of item 1411 , further comprising a polymeric carrier, wherein the polymeric carrier comprises a non-biodegradable polymer.
1558. The device of item 1411, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrophilic polymer.
1559. The device of item 1411, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrophobic polymer.
1560. The device of item 1411 , further comprising a polymeric carrier, wherein the polymeric carrier comprises a polymer having hydrophilic domains.
1561. The device of item 1411 , further comprising a polymeric carrier, wherein the polymeric carrier comprises a polymer having hydrophobic domains.
1562. The device of item 1411, further comprising a polymeric carrier, wherein the polymeric carrier comprises a non-conductive polymer.
1563. The device of item 1411, further comprising a polymeric carrier, wherein the polymeric carrier comprises an elastomer. 1564. The device of item 1411, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrogel.
1565. The device of item 1411, further comprising a polymeric carrier, wherein the polymeric carrier comprises a silicone polymer.
1566. The device of item 1411, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrocarbon polymer.
1567. The device of item 1411, further comprising a polymeric carrier, wherein the polymeric carrier comprises a styrene-derived polymer.
1568. The device of item 1411, further comprising a polymeric carrier, wherein the polymeric carrier comprises a butadiene polymer.
1569. The device of item 1411, further comprising a polymeric carrier, wherein the polymeric carrier comprises a macromer.
1570. The device of item 1411 , further comprising a polymeric carrier, wherein the polymeric carrier comprises a poly(ethylene glycol) polymer.
1571. The device of item 1411, further comprising a polymeric carrier, wherein the polymeric carrier comprises an amorphous polymer.
1572. The device of item 1411, further comprising a lubricious coating.
1573. The device of item 1411 wherein the anti-scarring agent is located within pores or holes of the device. 1574. The device of item 1411 wherein the anti-scarring agent is located within a channel, lumen, or divet of the device.
1575. The device of item 1411 , further comprising a second pharmaceutically active agent.
1576. The device of item 1411 , further comprising an antiinflammatory agent.
1577. The device of item 1411 , further comprising an agent that inhibits infection.
1578. The device of item 1411 , further comprising an agent that inhibits infection, wherein the agent is an anthracycline.
1579. The device of item 1411 , further comprising an agent that inhibits infection, wherein the agent is doxorubicin.
1580. The device of item 1411 , further comprising an agent that inhibits infection, wherein the agent is mitoxantrone.
1581. The device of item 1411 , further comprising an agent that inhibits infection, wherein the agent is a fluoropyrimidine.
1582. The device of item 1411 , further comprising an agent that inhibits infection, wherein the agent is 5-fluorouracil (5-FU).
1583. The device of item 1411 , further comprising an agent that inhibits infection, wherein the agent is a folic acid antagonist. 1584. The device of item 1411 , further comprising an agent that inhibits infection, wherein the agent is methotrexate.
1585. The device of item 1411 , further comprising an agent that inhibits infection, wherein the agent is a podophylotoxin.
1586. The device of item 1411 , further comprising an agent that inhibits infection, wherein the agent is etoposide.
1587. The device of item 1411 , further comprising an agent that inhibits infection, wherein the agent is a camptothecin.
1588. The device of item 1411 , further comprising an agent that inhibits infection, wherein the agent is a hydroxyurea.
1589. The device of item 1411, further comprising, an agent that inhibits infection, wherein the agent is a platinum complex.
1590. The device of item 1411 , further comprising an agent that inhibits infection, wherein the agent is cisplatin.
1591. The device of item 1411 , further comprising an antithrombotic agent.
1592. The device of item 1411 , further comprising a visualization agent.
1593. The device of item 1411 , further comprising a visualization agent, wherein the visualization agent is a radiopaque material, wherein the radiopaque material comprises a metal, a halogenated compound, or a barium containing compound. 1594. The device of item 1411 , further comprising a visualization agent, wherein the visualization agent is a radiopaque material, wherein the radiopaque material comprises barium, tantalum, or technetium.
1595. The device of item 1411, further comprising a visualization agent, wherein the visualization agent is a MRI responsive material.
1596. The device of item 1411 , further comprising a visualization agent, wherein the visualization agent comprises a gadolinium chelate.
1597. The device of item 1411, further comprising a visualization agent, wherein the visualization agent comprises iron, magnesium, manganese, copper, or chromium.
1598. The device of item 1411 , further comprising a visualization agent, wherein the visualization agent comprises an iron oxide compound.
1599. The device of item 1411, further comprising a visualization agent, wherein the visualization agent comprises a dye, pigment, or colorant.
1600. The device of item 1411 , further comprising an echogenic material.
1601. The device of item 1411, further comprising an echogenic material, wherein the echogenic material is in the form of a coating. 1602. The device of item 1411 wherein the device is sterile.
1603. The device of item 1411 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device.
1604. The device of item 1411 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is connective tissue.
1605. The device of item 1411 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is muscle tissue.
1606. The device of item 1411 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is nerve tissue.
1607. The device of item 1411 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is epithelium tissue.
1608. The device of item 1411 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from the time of deployment of the device to about 1 year.
1609. The device of item 1411 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from about 1 month to 6 months. 1610. The device of item 1411 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from about 1 - 90 days.
1611. The device of item 1411 wherein the anti-scarring agent is released in effective concentrations from the device at a constant rate.
1612. The device of item 1411 wherein the anti-scarring agent is released in effective concentrations from the device at an increasing rate.
1613. The device of item 1411 wherein the anti-scarring agent is released in effective concentrations from the device at a decreasing rate.
1614. The device of item 1411 wherein the anti-scarring agent is released in effective concentrations from the composition comprising the anti-scarring agent by diffusion oveca period- ranging from the time of deployment of the device to about 90 days.
1615. The device of item 1411 wherein the anti-scarring agent is released in effective concentrations from the composition comprising the anti-scarring agent by erosion of the composition over a period ranging from the time of deployment of the device to about 90 days.
1616. The device of item 1411 wherein the device comprises about 0.01 Dg to about 10 Dg of the anti-scarring agent.
1617. The device of item 1411 wherein the device comprises about 10 Dg to about 10 mg of the anti-scarring agent.
1618. The device of item 1411 wherein the device comprises about 10 mg to about 250 mg of the anti-scarring agent. 1619. The device of item 1411 wherein the device comprises about 250 mg to about 1000 mg of the anti-scarring agent.
1620. The device of item 1411 wherein the device comprises about 1000 mg to about 2500 mg of the anti-scarring agent.
1621. The device of item 1411 wherein a surface of the device comprises less than 0.01 Gg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
1622. The device of item 1411 wherein a surface of the device comprises about 0.01 Dg to about 1 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
1623. The device of item 1411 wherein a surface of the device comprises about 1 Dg to about 10 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
1624. The device of item 1411 wherein a surface of the device comprises about 10 Dg to about 250 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
1625. The device of item 1411 wherein a surface of the device comprises about 250 Dg to about 1000 Dg of the anti-scarring agent of anti- scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
1626. The device of item 1411 wherein a surface of the device comprises about 1000 Dg to about 2500 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied. 1627. A method as in any one of items 1411-1626, wherein the device is a grommet shaped tube.
1628. A method as in any one of items 1411-1626, wherein the device is a T-tube.
1629. A method as in any one of items 1411-1626, wherein the device is a tympanostomy tube
1630. A method as in any one of items 1411-1626, wherein the device is a drain tube.
1631. A method as in any one of items 1411-1626, wherein the device is a tympanic tube.
1632. A method as in any one of items 1411-1626, wherein the device is a otological tube.
1633. A method as in any one of items 1411-1626, wherein the device is a myringotomy tube.
1634. A method as in any one of items 1411-1626, wherein the device is an artificial Eustachian tube.
1635. A method as in any one of items 1411-1626, wherein the device is an Eustachian tube prosthesis.
1636. A method as in any one of items 1411-1626, wherein the device is an Eustachian stent. 1637. A device, comprising an intraocular implant (e.g., an implant) and an anti-scarring agent or a composition comprising an anti- scarring agent, wherein the agent inhibits scarring between the device and a host into which the device is implanted.
1638. The device of item 1637 wherein the agent is an adensosine A2A receptor antagonist.
1639. The device of item 1637 wherein the agent is an AKT inhibitor.
1640. The device of item 1637 wherein the agent is an alpha 2 integrin antagonist, wherein the alpha 2 integrin antagonist is Pharmaprojects No. 5754 (Merck KgaA).
1641. The device of item 1637 wherein the agent an alpha 4 integrin antagonist.
1642. The device of item 1637 wherein the agent is an alpha 7 nicotinic receptor agonist.
1643. The device of item 1637 wherein the agent is an angiogenesis inhibitor selected from the group consisting of AG-12,958 (Pfizer), ATN-161 (Attenuon LLC), neovastat, an angiogenesis inhibitor from Jerina AG (Germany), NM-3 (Mercian), VGA-1155 (Taisho), FCE-26644 (Pfizer), FCE-26950 (Pfizer), FPMA (Meiji Daries), FR-111142 (Fujisawa), GGTI-298, GM-1306 (Ligand), GPA-1734 (Novartis), NNC-47-0011 (Novo Nordisk), herbamycin (Nippon Kayaku), lenalidomide (Celegene), IP-10 (NIH), ABT-828 (Abbott), KIN-841 (Tokushima University, Japan), SF-1126 (Semafore Pharmaceuticals), laminin technology (NIH), CHIR-258 (Chiron), NVP-AEW541 (Novartis), NVP-AEW541 (Novartis), Vt16907 (Alchemia), OXI-8007 (Oxigene), EG-3306 (Ark Therapeutics), Maspin (Arriva), ABT- 567 (Abbott), PPI-2458 (Praecis Pharmaceuticals), CC-5079, CC-4089 (Celgene), HIF-1 alpha inhibitors (Xenova), S-247 (Pfizer), AP-23573 (Ariad), AZD-9935 (Astra Zeneca), mebendazole (Introgen Therapeutics), MetAP-2 inhibitors (GlaxoSmithKline), AG-615 (Angiogene Pharmaceuticals), Tie-2 antagonists (Hybrigenics), NC-381 , CYC-381 , NC-169, NC-219, NC-383, NC-384, NC-407 (Lorus Therapeutics), ATN-224 (Attenuon), ON-01370 (Onconova), Vitronectin antagonists (Amgen), SDX-103 (Salmedix), Vitronectin antagonists (Shire), CHP (Riemser), TEK (Amgen), Anecortave acetate (Alcon), T46.2 (Matrix Therapeutics), HG-2 (Heptagen), TEM antagonists (Genzyme), Oxi-4500 (Oxigene), ATN-161 (Attenuon), WX-293 (Wilex), M-2025 (Metris Therapeutics), Alphastatin (BioActa), YH-16 (Yantai Rongchang), BIBF-1120 (Boehringer Ingelheim), BAY-57-9352 (Bayer), AS- 1404 (Cancer Research Technology), SC-77964 (Pfizer), glycomimetics (BioTie Therapies), TIE-2 Inhibitors (Ontogen), DIMI, Octamer (Octamer), ABR-215050 (Active Biotech), ABT-518 (Abbott), KDR inhibitors (Abbott), BSF-466895 (Abbott), SCH-221153 (Schering-Plough), DAC:antiangiogenic (ConjuChem), TFPI (EntreMed), AZD-2171 (Astra-Zenaca), CDC-394 (Celgene), LY290293 (EIi Lilly), IDN-5390 (Indena), Kdr Kinase Inhibitors (Merck), CT-113020, CT-116433, CT-116563, CT-31890, CT-32228) (Cell Therapeutics), A-299620 (Abbott), TWEAK Inhibitor (Amgen), VEGF modulators (Johnson and Johnson), Tum-N53, tumstatin (Genzyme), Thios- 1, Thios-2 (Thios Pharmaceuticals), MV-6401 (Miravant Medical Technologies), Spisulosine (PharmaMar), CEP-7055 (Cephalon), AUV-201 (Auvation), LM-609 (EIi Lilly), SKF-106615 (AnorMED), Oglufanide disodium (Cytran), BW-114 (Pharminox), Calreticulin (NIH), WX-678 (Wilex), SD-7784 (Pfizer), WX-UK1 (Wilex), SH-268 (Schering AG), 2-Me-PGA (Celgene), S- 137 (Pfizer), ZD-6126 (Angiogene Pharmaceuticals), SG-292 (SignalGen), Benefin (Lane Labs), A6, A36 (Angstrom), SB-2723005 (GlaxoSmithKline), SC-7 (Cell Therapeutics), ZEN-014 (AEtema Zentaris), 2-methoxyestradiol (EntreMed), NK-130119 (Nippon Kayaku), CC-10004 (Celgene), AVE- 8062A (Ajinomoto), Tacedinaline (Pfizer), Actinonin (Tokyo Metropolitan Institute of Medical Science), Lenalidomide (Celgene), VGA-1155, BTO-956 (SRI International), ER-68203-00 (Eisai), CT-6685 (UCB), JKC-362 (Phoenix Pharmaceuticals), DMI-3798 (DMI Biosciences, Angiomate (Ipsen), ZD-6474 (AstraZeneca), CEP-5214 (Cephalon), Canstatin (Genzyme), NM-3 (Mercian), Oridigm (MediQuest Therapeutics), Exherin (Adherex), BLS-0597 (Boston Life Sciences), PTC-299 (PTC Therapeutics), NPI-2358 (Nereus Pharmaceuticals), CGP-79787 (Novartis), AEE-788 (Novartis), CKD-732 (Chong Kun Dang), CP-564959 (OSI Pharmaceuticals), CM-101 (CarboMed), CT-2584, CT3501 (Cell Therapeutics), combretastatin and analogues and derivatives thereof (Oxigene), Rebimastat (Bristol-Meyers Squibb), Dextrin 2-sulfate (ML Laboratories), Cilengitide (Merk KGaA), NSC-706704 (Pharminox), KRN- 951 (Kirin Brewery), Ukrain, NSC-631570 (Nowicky Pharma), Tecogalan sodium (Daiichi Pharmaceutical), Tz-93 (Tsumura), TBC-1635 (Encysive Pharmaceuticals), TAN-1120 (Takeda), Semaxanib (Pfizer), BDI-7800 (Biopharmacopae), SD-186, SD-983 (Bristol-Meyers Squibb), SB-223245 (GlaxoSmithKline), SC-236 (Pfizer), RWJ-590973 (Johnson and Johnson), ILX-1850 (Genzyme), SC-68488, S-836 (Pfizer), CG-55069-11 (CuraGen), Ki-23057 (Kirin Brewery), CCX-700 (Chemoentryx), Pegaptanib octasodium (Giled Sciences), ANGIOCOL (available from Biostratum Inc.), or an analogue or derivative thereof.
1644. The device of item 1637 wherein the agent is an apoptosis antagonist.
1645. The device of item 1637 wherein the agent is an apoptosis activator.
1646. The device of item 1637 wherein the agent is a beta 1 integrin antagonist. 1647. The device of item 1637 wherein the agent is a beta tubulin inhibitor.
1648. The device of item 1637 wherein the agent is a blocker of enzyme production in Hepatitis C.
1649. The device of item 1637 wherein the agent is a Bruton's tyrosine kinase inhibitor.
1650. The device of item 1637 wherein the agent is a calcineurin inhibitor.
1651. The device of item 1637 wherein the agent is a caspase 3 inhibitor.
1652. The device of item 1637 wherein the agent is a CC chemokine receptor antagonist.
1653. The device of item 1637 wherein the agent is a cell cycle inhibitor selected from the group consisting of SNS-595 (Sunesis), synthadotin, KRX-0403, and an analogue or derivative thereof.
1654. The device of item 1637 wherein the agent is a cathepsin B inhibitor.
1655. The device of item 1637 wherein the agent is a cathepsin K inhibitor, wherein the cathepsin K inhibitor is 462795 (GlaxoSmithKline), INPL-022-D6 (Amura Therapeutics), an analogue or derivative thereof). 1656. The device of item 1637 wherein the agent is a cathepsin L inhibitor.
1657. The device of item 1637 wherein the agent is a CD40 antagonist.
1658. The device of item 1637 wherein the agent is a chemokine receptor agonist.
1659. The device of item 1637 wherein the agent is a chymase inhibitor.
1660. The device of item 1637 wherein the agent is a collagenase antagonist.
1661. The device of item 1637 wherein the agent is a CXCR antagonist.
1662. The device of item 1637 wherein the agent is a cyclin dependent kinase inhibitor selected from the group consisting of a CDK-1 inhibitor, a CDK-2 inhibitor, a CDK- 4 inhibitor, a CDK-6 inhibitor, a CAK1 inhibitor from GPC Biotech and Bristol-Myers Squibb, RGB-286199 (GPC Biotech), an anticancer agent from Astex Technology, a CAK1 inhibitor from GPC Biotech, a CDK inhibitor from Sanofi-Aventis, a CDK1/CDK2 inhibitor from Amgen, a CDK2 inhibitor from SUGEN-2 (Pfizer), a hearing loss therapy agent (Sound Pharmaceuticals), PD-0332991 (Pfizer), RGB-286199 (GPC Biotech), Ro-0505124 (Hoffmann-La Roche), a Ser/Thr kinase inhibitor from Lilly (EIi Lilly), CVT-2584 (CAS No. 199986-75-9) (CV Therapeutics), and an analogue or derivative thereof. 1663. The device of item 1637 wherein the agent is a cyclooxygenase 1 inhibitor.
1664. The device of item 1637 wherein the agent is a DHFR inhibitor.
1665. The device of item 1637 wherein the agent is a dual integrin inhibitor.
1666. The device of item 1637 wherein the agent is an elastase inhibitor.
1667. The device of item 1637 wherein the agent is an elongation factor-1 alpha inhibitor.
1668. The device of item 1637 wherein the agent is an endothelial growth factor antagonist.
1669. The device of item 1637 wherein the agent is an endothelial growth factor receptor kinase inhibitor selected from the group consisting of sorafenib tosylate (Bayer), AAL-993 (Novartis), ABP-309 (Novartis), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), E- 7080 (Eisai), EG-3306 (Ark Therapeutics), EXEL-2880 (Exelixis), GW- 654652 (GlaxoSmithKline), a KDR inhibitor from LG Life Sciences, CT-6685 and CT-6729 (UCB), KRN-633 and KRN-951 (Kirin Brewery), OSI-930 (OSI Pharmaceuticals), SP-5.2 (Supratek Pharma), SU-11657 (Pfizer), a Tie-2 antagonist (Hybrigenics), a VEGFR-2 kinase inhibitor (Bristol-Myers Squibb), XL-647 (Exelixis), a KDR inhibitor from Abbott Laboratories, sorafenib tosylate, and an analogue or derivative thereof. 1670. The device of item 1637 wherein the agent is an endotoxin antagonist.
1671. The device of item 1637 wherein the agent is an epothilone and tubulin binder.
1672. The device of item 1637 wherein the agent is an estrogen receptor antagonist.
1673. The device of item 1637 wherein the agent is an FGF inhibitor.
1674. The device of item 1637 wherein the agent is a farnexyl transferase inhibitor.
1675. The device of item 1637 wherejn the agent is famesyltransferase inhibitor selected from the group of A-197574 (Abbott), a farnesyltransferase inhibitor from Servier, FPTIII (Strathclyde Institute for Drug R), LB-42908 (LG Life Sciences), Pharmaprojects No. 5063 (Genzyme), Pharmaprojects No. 5597 (Ipsen), Yissum Project No. B-1055 (Yissum), and an analogue or derivative thereof.
1676. The device of item 1637 wherein the agent is an FLT-3 kinase inhibitor.
1677. The device of item 1637 wherein the agent is an FGF receptor kinase inhibitor.
1678. The device of item 1637 wherein the agent is a fibrinogen antagonist selected from the group consisting of AUV-201 (Auvation), MG-13926 (Sanofi-Aventis), plasminogen activator (CAS No. 105913-11-9) (from Sanofi-Aventis or UCB)1 plasminogen activator-2 (tPA- 2) (Sanofi-Aventis), pro-urokinase (CAS No. 82657-92-9) (Sanofi-Aventis), and an analogue or derivative thereof.
1679. The device of item 1637 wherein the agent is a heat shock protein 90 antagonist selected from the group consisting of SRN-005 (Sirenade), geldanamycin, NSC-33050 (17-allylaminogeldanamycin; 17- AAG), 17-dimethylaminoethylamino-17-demethoxy-geldanamycin (17- DMAG), rifabutin (rifamycin XIV, 1',4-didehydro-1-deoxy-1 ,4-dihydro-5'-(2- methy!propyl)-1-oxo-), and an analogue or derivative thereof.
1680. The device of item 1637 wherein the agent is a histone deacetylase inhibitor.
1681. The device of item 1637 wherein the agent is an HMGCoA reductase inhibitor selected from the group consisting of an atherosclerosis therapeutic from Lipid Sciences, ATI-16000 (ARYx Therapeutics), KS-01-019 (Kos Pharmaceuticals), Pharmaprojects No. 2197 (Sanofi-Aventi), RP 61969 (Sanofi-Aventis), and an analogue or derivative thereof.
1682. The device of item 1637 wherein the agent is an ICAM inhibitor.
1683. The device of item 1637 wherein the agent is an IL, ICE and IRAK antagonist, wherein the antagonist is a CJ-14877, CP-424174 (Pfizer), NF-61 (Negma-Lerads), and an analogue or derivative thereof.
1684. The device of item 1637 wherein the agent is an IL-2 inhibitor. 1685. The device of item 1637 wherein the agent is an immunosuppressant selected from the group consisting of teriflunomide (Sanofi Aventis), chlorsulfaquinoxalone (NSC-339004), chlorsulfaquinoxaione sulfate, CS-712 (Sankyo), ismomultin alfa (CAS No. 457913-93-8) (Akzo Nobel), antiallergics from GenPat77, antiinflammatories or AT-005 (Androclus Therapeutics), autoimmune disease therapeutics from EpiVax, BN-007 (Bone), budesonide (CAS No. 51333-22- 3) (MAP Pharmaceuticals), CO-14 (Genzyme), edratide (CAS No. 433922- 67-9) (Teva), EP-314 (Enanta), eprovafen (CAS No. 101335-99-3) (Sanofi- Aventis), HWA-131 (CAS No. 118788-41-3) (Sanofi-Aventis), immunomodulators from MerLion Pharmaceuticals, immunosuppressives from Alchemia, IPL-12 (Inflazyme), MDL-9563 (CAS No. 27086-86-8) (Sanofi-Aventis), Pharmaprojects No. 2330 (Sanofi-Aventis), Pharmaprojects No. 6426 (Abgenix), PXS-25 (Pharmaxis), rosmarinic acid (CAS No. 20283-92-5) (Sanofi-Aventis), RP 42927 or RP 54745 (CAS No. 135330-08-4) (Sanofi-Aventis), SGN-35 (Seattle Genetics), ST-1959 (Sigma-Tau), type I diabetes therapy from SYNX Pharma, UNIL-88 (Debiopharm), VP-025 (Vasogen), VR-694 (Vectura), PRTX-O01 (Protalex), and an analogue or derivative thereof.
1686. The device of item 1637 wherein the agent is an IMPDH (inosine monophosphate).
1687. The device of item 1637 wherein the agent is an integrin antagonist.
1688. The device of item 1637 wherein the agent is an interleukin antagonist.
1689. The device of item 1637 wherein the agent is an inhibitor of type III receptor tyrosine kinase. 1690. The device of item 1637 wherein the agent is an irreversible inhibitor of enzyme methionine aminopeptidase type 2.
1691. The device of item 1637 wherein the agent is an isozyme selective delta protein kinase C inhibitor.
1692. The device of item 1637 wherein the agent a JAK3 enzyme inhibitor.
1693. The device of item 1637 wherein the agent is a JNK inhibitor.
1694. The device of item 1637 wherein the agent is a kinase inhibitor.
1695. The device of item 1637 wherein the agent is kinesin antagonist.
1696. The device of item 1637 wherein the agent is a kinesin antagonist.
1697. The device of item 1637 wherein the agent is a leukotriene inhibitor and antagonist selected from the group consisting of ambicromil (CAS No. 58805-38-2) (Sanofi-Aventis), amelubant (CAS No. 346735-24-8) (Boehringer Ingelheim), DW-1141 (Dong Wha), ebselen (Daiichi Pharmaceutical), ibudilast (Kyorin), leucotriene inhibitors from Sanofi-Aventis, lymphotoxin-beta receptor (LT-D) from Biogen Idee, Pharmaprojects No. 1535 and 2728 (CAS No. 119340-33-9) (Sanofi- Aventis), R-112 (Rigel), Rev-5367 (CAS No. 92532-05-3) (Sanofi-Aventis), RG-14893 (CAS No. 141835-49-6) (Sanofi-Aventis), RG-5901-A (CAS No. 101910-24-1), 92532-23-5, RP 66153 (CAS No. 142422-79-5), RP 66364 (CAS No.186916-92-5), RP 69698 (CAS No. 141748-00-7) (Sanofi-Aventis), SC-411930 (Pfizer), SC-41930 (CAS No. 120072-59-5) (Pfizer), SC-50605 (CAS No. 138828-39-4) (Pfizer), SC-51146 (CAS No. 141059-52-1), SC- 53228 (CAS No. 153633-01-3) (Pfizer), spaglumic acid (ZY-15106) (CAS No.3106-85-2), 80619-64-3 (Novartis), tipredane (CAS No. 85197-77-9) (Bristol-Myers Squibb), U-75302 (CAS No. 119477-85-9) (Pfizer), and analogue or derivative thereof.
1698. The device of item 1637 wherein the agent is an MAP kinase inhibitor.
1699. The device of item 1637 wherein the agent is a matrix metailoproteinase inhibitor.
1700. The device of item 1637 wherein the agent is an MCP- CCR2 inhibitor.
1701. The device of item 1637 wherein the agent is an mTOR inhibitor.
1702. The device of item 1637 wherein the agent is an mTOR kinase inhibitor.
1703. The device of item 1637 wherein the agent is a microtubule inhibitor selected from the group consisting of antibody- maytansinoid conjugates from Biogen Idee, colchicines (MantiCore Pharmaceuticals), anticancer immunoconjugates from Johnson & Johnson, DIME from Octamer, gni-1f (GNI), huC242-DM4, huMy9-6-DM1 (ImmunoGen), IDN-5404 (Indena), IMO-098, IMOderm (Imotep), mebendazole (Introgen Therapeutics), microtubule poisons from Cambridge Enterprise, paclitaxel such as LOTAX from Aphios (CAS No. 33069-62-4), Genexol-PM from Samyang, Pharmaprojects No. 6383 (Tapestry Pharmaceuticals), RPR-112378 (Sanofi-Aventis), SGN-75 (Seattle Genetics), SPL-7435 (Starpharma), SSR-250411 (Sanofi-Aventis), trastuzumab-DM1 (Genentech), vinorelbine, and an analogue or derivative thereof.
1704. The device of item 1637 wherein the agent is an MIF inhibitor.
1705. The device of item 1637 wherein the agent is an MMP inhibitor.
1706. The device of item 1637 wherein the agent is a neurokinin (NK) antagonist selected from the group consisting of anthrotainin (CAS No. 148084-40-6) (Sanofi-Aventis), an IBS thereapeutic from ArQuIe, MDL-105212A (CAS No. 167261-60-1) (Ssanofi-Aventis), Pharmaprojects No. 2744, 3258 (CAS No. 139167-47-8) 4006, 4201 , or 5986 (Sanofi-Aventis), RP 67580 (CAS No. 135911-02-3), SR-144190 (CAS No. 201152-86-5), SSR-240600, SSR-241586 (Sanofi-Aventis), TKA-457 (Novartis), vestipitant mesylate (CAS No. 334476-64-1) (GlaxoSmithKline), Win-64821 (Sanofi-Aventis), PRX-96026 (Predix Pharmaceuticals), and an analogue or derivative thereof.
1707. The device of item 1637 wherein the agent is an NF kappa B inhibitor selected from the group consisting of AVE-0545 or AVE- 0547 (Sanofi-Aventis), bortezomib (CAS No. 179324-69-7) (Millennium Pharmaceuticals), dexanabinol (CAS No. 112924-45-5) (Pharmos), dexiipotam (Viatris), Pharmaprojects No. 6283 (INDRA) (OXiGENE), IPL- 576092 (CAS No. 137571-30-3) (Inflazyme), NFKB decoy (Corgentech), NFKB decoy oligo (AnGes MG), NFKB's from Ariad, osteoporosis treatments or S5 (F005) from Fulcrum Pharmaceuticals, P61 (Phytopharm), R-flurbiprofen (CAS No. 5104-49-4) (Encore Pharmaceuticals), and an analogue or derivative thereof.
1708. The device of item 1637 wherein the agent is a nitric oxide agonist.
1709. The device of item 1637 wherein the agent is an ornithine decarboxylase inhibitor.
1710. The device of item 1637 wherein the agent is a p38 MAP kinase inhibitor selected from the group consisting of AZD-6703 (AstraZeneca), JX-401 (Jexys Pharmaceuticals), BMS-2 (Bristol-Myers Squibb), a p38 MAP kinase inhibitor from Novartis, a p38-alpha MAP kinase inhibitor from Amphora, Pharmaprojects No. 5704 (Pharmacopeia), RPR- 200765A (Sanofi-Aventis), SD-282 (Johnson & Johnson), TAK-715 (Takeda), and an analogue or derivative thereof.
1711. The device of item 1637 wherein the agent is a palmitoyl-protein thioesterase inhibitor.
1712. The device of item 1637 wherein the agent is a PDGF receptor kinase inhibitor selected from the group consisting of AAL-993, AMN-107, or ABP-309 (Novartis), AMG-706 (Amgen), BAY-57-9352 (Bayer), CDP-860 (UCB), E-7080 (Eisai), imatinib (CAS No. 152459-95-5) (Novartis), OSI-930 (OSI Pharmaceuticals), RPR-127963E (Sanofi-Aventis), RWJ-540973 (Johnson & Johnson), sorafenib tosylate (Bayer), SU-11657 (Pfizer), tandutinib (CAS No. 387867-13-2) (Millennium Pharmaceuticals), vatalanib (Novartis), ZK-CDK (Schering AG), and an analogue or derivative thereof. 1713. The device of item 1637 wherein the agent is (-)- halofenate (Metabolex), AMG-131 (Amgen), antidiabetics from Japan Tobacco, AZD-4619, AZD-8450, AZD-8677 (AstraZeneca), DRF-10945, balaglitazone (Dr Reddy's), CS-00088, CS-00098 (Chipscreen Biosciences), E-3030 (Eisai), etalocib (CAS No. 161172-51-6) (EIi Lilly), GSK-641597 (Ligand), GSK-677954 (GlaxoSmithKIine), GW-409544 (Ligand), GW- 590735 (GlaxoSmithKIine), K-111 (Hoffmann-La Roche), LY-518674 (EIi Lilly), LY-674 (Ligand), LY-929 (Ligand), MC-3001, MC-3002 (MaxoCore Pharmaceuticals), metformin HCI + pioglitazone (CAS No. 1115-70-4 and 112529-15-4), ACTOPLUS MET from Andrx), muraglitazar (CAS No. 331741-94-7) (Bristol-Myers Squibb), naveglitazar (Ligand), oleoylethanolamide (Kadmus Pharmaceuticals), ONO-5129, pioglitazone hydrochloride (CAS No. 111025-46-8 and 112529-15-4) (Takeda), PLX-204 (Plexxikon), PPAR agonists from Genfit, PPAR delta agonists from EIi Lilly, PPAR-alpha agonists from CrystalGenomics, PPAR-gamma modulators and PPAR-β modulators from CareX, rosiglitazone maleate (CAS No. 122324- 73-4 or 155141-29-0) (GlaxoSmithKIine), rosiglitazone maleate/glimepir (CAS No. 155141-29-0 and 93479-97-1), AVANDARYL, rosiglitazone maleate/metformin extend (CAS No. 155141-29-0 and 657-24-9), AVANDAMET, rosiglitazone maleate+metformin, AVANDAMET (GlaxoSmithKIine), tesaglitazar (AstraZeneca), and an analogue or derivative thereof.
1714. The device of item 1637 wherein the agent is a phosphatase inhibitor.
1715. The device of item 1637 wherein the agent is a phosphodiesterase (PDE) inhibitor selected from the group consisting of avanafil (Tanabe Seiyaku), dasantafil (CAS No. 569351-91-3) (Schering- Plough), A-906119 (CAS No. 134072-58-5), DL-850 (Sanofi-Aventis), GRC- 3015, GRC-3566, GRC-3886 (Glenmark), HWA-153 (CAS No. 56395-66-5) (Sanofi-Aventis), hydroxypumafentrine (Altana), IBFB-130011, IBFB-14-016, 1BFB-140301, IBFB-150007, IBFB-211913 (IBFB Pharma), L-826141 (Merck & Co), medorinone (CAS No. 88296-61-1) (Sanofi-Aventis), MEM- 1917 (Memory Pharmaceuticals), ND-1251 (Neuro3d), PDE inhibitors from ICOS, PDE IV inhibitors from Memory Pharmaceuticals and CrystalGenomics, Pharmaprojects No. 2742 and 6141 (Sanofi-Aventis), QAD-171 (Novartis), RHC-2963 (CAS No. 76993-12-9 and 76993-14-1), RPFM 17658, RPR-122818 derivatives, SR-24870 , and RPR-132294 (Sanofi-Aventis), SK-350 (ln2Gen), stroke therapy agents from deCODE Genetics, TAS-203 (Taiho), tofimilast (CAS No. 185954-27-2) (Pfizer), UK- 371800 (Pfizer), WIN-65579 (CAS No. 158020-82-7) (Sanofi-Aventis), IBFB- 130020 (IBFB Pharma), OPC-6535 (CAS No. 145739-56-6) (Otsuka), a phosphodiesterase 111 inhibitor, enoximone, a phosphodiesterase IV inhibitor, fosfosal, Atopik (Barrier Therapeutics), triflusal, a phosphodiesterase V inhibitor, and an analogue or derivative thereof.
1716. The device of item 1637 wherein the agent is a PKC inhibitor.
1717. The device of item 1637 wherein the agent is a platelet activating factor antagonist.
1718. The device of item 1637 wherein the agent is a platelet- derived growth factor receptor kinase inhibitor.
1719. The device of item 1637 wherein the agent is a prolyl hydroxylase inhibitor.
1720. The device of item 1637 wherein the agent is a polymorphonuclear neutrophil inhibitor. 1721. The device of item 1637 wherein the agent is a protein kinase B inhibitor.
1722. The device of item 1637 wherein the agent is a protein kinase C stimulant.
1723. The device of item 1637 wherein the agent is a purine nucleoside analogue.
1724. The device of item 1637 wherein the agent is a purinoreceptor P2X antagonist.
1725. The device of item 1637 wherein the agent is a Raf kinase inhibitor.
1726. The device of item 1637 wherein the agent is a reversible inhibitor of ErbB1 and ErbB2.
1727. The device of item 1637 wherein the agent is a ribonucleoside triphosphate reductase inhibitor.
1728. The device of item 1637 wherein the agent is an SDF-1 antagonist.
1729. The device of item 1637 wherein the agent is a sheddase inhibitor.
1730. The device of item 1637 wherein the agent is an SRC inhibitor. 1731. The device of item 1637 wherein the agent is a stromelysin inhibitor.
1732. The device of item 1637 wherein the agent is an Syk kinase inhibitor.
1733. The device of item 1637 wherein the agent is a telomerase inhibitor.
1734. The device of item 1637 wherein the agent is a TGF beta inhibitor selected from the group consisting of pirfenidone (CAS No. 53179-13-8) (MARNAC), tranilast (CAS No. 53902-12-8) (Kissei), IN-1130 (ln2Gen), mannose-6-phosphate (BTG), TGF-β antagonists from Inflazyme (Pharmaprojects No. 6075), TGF-β antagonists from Sydney, non-industrial source), TGF-β I receptor kinase inhibitors from EIi Lilly, TGF-β receptor inhibitors from Johnson & Johnson, and an analogue or derivative thereof.
1735. The device of item 1637 wherein the agent is a TNFα antagonist or TACE inhibitor selected from the group consisting of adalimumab (CAS No. 331731-18-1) (Cambridge Antibody Technology), AGIX-4207 (AtheroGenics), AGT-1 (Advanced Biotherapy), an antiinflammatory from Borean Pharma, Cellzome, or Paradigm Therapeutics, anti-inflammatory vaccine (TNF-alpha kinoid) from Neovacs, humanized anti-TNF antibody or an anti-TNF MAb (CB0006) Celltech (UCB), apratastat (CAS No. 287445-51-0) (Wyeth), BMS-561392 (Bristol-Myers Squibb), BN- 006 (Bone), certolizumab pegol (CAS No. 428863-50-7 or CH-138 (UCB), cilomilast (CAS No. 153259-65-5) (GlaxoSmithKline), CR-1 (Nuada Pharmaceuticals), CRx-119 (CombinatoRx), D-5410 (UCB), dacopafant (CAS No. 125372-33-0) (Sanofi-Aventis), dersalazine (CAS No. 188913-57- 7/188913-58-8) (Uriach), etanercept (CAS No. 185243-69-0) (Amgen), ethyl pyruvate (Critical (Critical Therapeutics), golimumab (CAS No. 476181-74-5) (Johnson & Johnson), hormono-immunotherapy from Ipsen, CDP571 (e.g., Humicade from UCB), IC-485 (ICOS), infliximab (CAS No. 170277-31-3) (Johnson & Johnson), IP-751 (Manhattan Pharmaceuticals), ISIS-104838 (CAS No. 250755-32-9) (ISIS Pharmaceuticals), lenalidomide (CAS No. 191732-72-6) (Celgene), lentinan (CAS No. 37339-90-5) (Ajinomoto), MDL- 201112 (CAS No. 142130-73-2) (Sanofi-Aventis), medroxyprogesterone (CAS No. 520-85-4) (InKine Pharmaceutical), N-acetylcysteine (CAS No. 616-91-1) (Zambon), NBE-P2 (DIREVO Biotech), nerelimomab (CAS No. 162774-06-3) (Chiron), OM-294DP (OM PHARMA)1 onercept (CAS No. 199685-57-9) (Yeda), PASSTNF-alpha (Verigen), pentoxifylline or oxypentifylline (Sanofi-Aventis), Pharmaprojects No. 4091 , 4241 , 4295, or 4488 (Sanofi-Aventis), Pharmaprojects No. 5480 (Amgen), Pharmaprojects No. 6749 (Cengent), pirfenidone (CAS No. 53179-13-8) (MARNAC), RPR- 132294 (Sanofi-Aventis), S5 (F002) (Fulcrum Pharmaceuticals), simvastatin (CAS No. 79902-63-9) (Merck & Co), STA-6292 (Synta Pharmaceuticals), tacrolimus (CAS No. 104987-11-3) (Fujisawa LifeCycle Pharma), talactoferrin alfa (CAS No. 308240-58-6) (Agennix), thalidomide (CAS No. 50-35-1) (Celgene), TNF antagonists form ProStrakan, and Synergen, TNF inhibitors (Amgen), TNF-alpha antagonists from Dynavax Technologies and Jerina AG (Germany), TNF-alpha inhibitors from IBFB Pharma and Xencor (Xencor), torbafylline (CAS No. 105102-21-4) (Sanofi-Aventis), UR-1505 (Uriach), VT-346 (Viron Therapeutics), YSIL6 (Y's Therapeutics), YSTH2 (Y's Therapeutics), NPI-1302a-3 (Nereus Pharmaceuticals, a TNF antagonist from Jerina AG (Germany), dersalazine, and an analogue or derivative thereof.
1736. The device of item 1637 wherein the agent is a Toll receptor inhibitor.
1737. The device of item 1637 wherein the agent is a tubulin antagonist 1738. The device of item 1637 wherein the agent is a tyrosine kinase inhibitor selected from the group consisting of SU-011248, SUTENT from Pfizer Inc. (New York, NY), BMS-354825, PN-355 (Paracelsian Pharmaceuticals), AGN-199659 (Allergan), AAL-993 or ABP-309 (Novartis), adaphostin (NIH), AEE-788 (Novartis), AG-013736 (OSI Pharmaceuticals), AG-13736 (Pfizer), ALT-110 (Alteris Therapeutics), AMG-706 (Amgen), anticancer MAbs from Xencor, anti-EGFrvlll MAbs from Abgenix, anti-HER2 MAb from Abiogen, AZD-2171 or AZD-9935 (AstraZeneca), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), CEP-5214 (Cephalon), CEP- 7055 (Cephalon), cetuximab (ImClone Systems), CHIR-200131 and CHIR- 258 (Chiron), CP-547632 (OSI Pharmaceuticals), CP-724714 (Pfizer), CT- 301 (Creabilis Therapeutics), D-69491 (Baxter International), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EGFR/ErbB2 inhibitors from Array BioPharma, erlotinib (CAS No. 183319-69-9) (OSI Pharmaceuticals), EXEL-2880 (Exelixis), FK-778 (Sanofi-Aventis) , gefitinib (CAS No. 184475-35-2) (AstraZeneca), GW-2286 or GW-654652 (GlaxoSmithKline), her2/neu antigen from AlphaVax, HER-2/neu inhibitor from Generex, Herzyme (Medipad) (Sirna Therapeutics), HKI-272 (Wyeth), HuMax-EGFr (Genmab), idronoxil (CAS No. 81267-65-4) (Novogen), IGF-1 inhibitors from Ontogen, IMC-11F8 (ImClone Systems), kahalalide F (CAS No. 149204-42-2) (PharmaMar), KDR inhibitor from LG Life Sciences, KDR inhibitors from Abbott Laboratories, KDR kinase inhibitors (UCB), Kdr kinase inhibitors from Merck & Co, KRN-633 and KRN-951 (Kirin Brewery), KSB-102 (Xenova), lapatinib ditosylate (CAS No. 388082-78-8) (GlaxoSmithKline), matuzumab (Merck KGaA), MDX-214 (Medarex), ME-103 (Pharmexa), MED-A300 (Gerolymatos), MNAC-13 (Lay Line Genomics), nimotuzumab (Center of Molecular Immunology), NSC-330507 or NSC-707545 (NIH), NV-50 (Novogen), OSI-930 (OSI Pharmaceuticals), panitumumab (Abgenix), pelitinib (CAS No. 287933-82-7) (Wyeth), pertuzumab (CAS No. 380610-27- 5) (Genentech), Pharmaprojects No. 3985 (Sanofi-Aventis), prostate cancer therapeutics from Sequenom (SQPC35, SQPC36, SQPC90), removab and remoxab (Trion Pharma), RG-13022 (CAS No. 136831-48-6), RG-13291 (CAS No. 138989-50-1), or RG-14620 (CAS No. 136831-49-7) (Sanofi- Aventis), RM-6427 (Romark), RNAi breast cancer therapy from Benitec, RP 53801 (CAS No. 125882-88-4) (Sanofi-Aventis), sorafenib tosylate (Bayer), SU-11657 (Pfizer), Tie-2 antagonists from Semaia (Hybrigenics), Tie-2 inhibitors from Ontogen, trastuzumab (CAS No. 180288-69-1) (Genentech), tyrosine kinase inhibitors from Sanofi-Aventis, U3-1287, U3-1565, U3-1784, U3-1800 (U3 Pharma), vatalanib (Novartis), VEGFR-2 kinase inhibitor from Bristol-Myers Squibb, XL-647 (Exelixis), ZD-6474 (AstraZeneca), ZK-CDK (Schering AG), an EGFR tyrosine kinase inhibitor, EKB-569 (Wyeth), and an analogue or derivative thereof.
1739. The device of item 1637 wherein the agent is a VEGF inhibitor.
1740. The device of item 1637-wherein the agent is a vitamin D receptor agonist.
1741. The device of item 1637 wherein the agent is ZD-6474 (an angiogenesis inhibitor).
1742. The device of item 1637 wherein the agent is AP-23573 (an mTOR inhibitor).
1743. The device of item 1637 wherein the agent is synthadotin (a tubulin antagonist).
1744. The device of item 1637 wherein the agent is S-0885 (a collagenase inhibitor). 1745. The device of item 1637 wherein the agent is aplidine (an elongation factor-1 alpha inhibitor).
1746. The device of item 1637 wherein the agent is ixabepilone (an epithilone).
1747. The device of item 1637 wherein the agent is IDN-5390 (an angiogenesis inhibitor).
1748. The device of item 1637 wherein the agent is SB- 2723005 (an angiogenesis inhibitor).
1749. The device of item 1637 wherein the agent is ABT-518 (an angiogenesis inhibitor).
1750. The device of Jtenr 1637 wherein the agent is combretastatin (an angiogenesis inhibitor).
1751. The device of item 1637 wherein the agent is anecortave acetate (an angiogenesis inhibitor).
1752. The device of item 1637 wherein the agent is SB- 715992 (a kinesin antagonist).
1753. The device of item 1637 wherein the agent is temsirolimus (an rnTOR inhibitor).
1754. The device of item 1637 wherein the agent is adalimumab (a TNFα antagonist).
1755. The device of item 1637, further comprising a polymer. 1756. The device of item 1637 wherein the anti-scarring agent inhibits adhesion between the device and a host into which the device is implanted.
1757. The device of item 1637 wherein the device delivers the anti-scarring agent locally to tissue proximate to the device.
1758. The device of item 1637, further comprising a coating, wherein the coating comprises the anti-scarring agent.
1759. The device of item 1637, further comprising a coating, wherein the coating is disposed on a surface of the device.
1760. The device of item 1637, further comprising a coating, wherein the coating directly contacts the device.
1761. The device of item 1637, further comprising a coating, wherein the coating indirectly contacts the device.
1762. The device of item 1637, further comprising a coating, wherein the coating partially covers the device.
1763. The device of item 1637, further comprising a coating, wherein the coating completely covers the device.
1764. The device of item 1637, further comprising a coating, wherein the coating is a uniform coating.
1765. The device of item 1637, further comprising a coating, wherein the coating is a non-uniform coating. 1766. The device of item 1637, further comprising a coating, wherein the coating is a discontinuous coating.
1767. The device of item 1637, further comprising a coating, wherein the coating is a patterned coating.
1768. The device of item 1637, further comprising a coating, wherein the coating has a thickness of 100 Dm or less.
1769. The device of item 1637, further comprising a coating, wherein the coating has a thickness of 10 Om or less.
1770. The device of item 1637, further comprising a coating, wherein the coating adheres to the surface of the device upon deployment of the device.
1771. The device of item 1637, further comprising a coating, wherein the coating is stable at room temperature for a period of 1 year.
1772. The device of item 1637, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 0.0001% to about 1% by weight.
1773. The device of item 1637, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 1% to about 10% by weight.
1774. The device of item 1637, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 10% to about 25% by weight. 1775. The device of item 1637, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 25% to about 70% by weight.
1776. The device of item 1637, further comprising a coating, wherein the coating further comprises a polymer.
1777. The device of item 1637, further comprising a first coating having a first composition and the second coating having a second composition.
1778. The device of item 1637, further comprising a first coating having a first composition and the second coating having a second composition, wherein the first composition and the second composition are different.
1779. The device of item 1637, further comprising a polymeric carrier, wherein the polymeric carrier comprises a copolymer.
1780. The device of item 1637, further comprising a polymeric carrier, wherein the polymeric carrier comprises a block copolymer.
1781. The device of item 1637, further comprising a polymeric carrier, wherein the polymeric carrier comprises a random copolymer.
1782. The device of item 1637, further comprising a polymeric carrier, wherein the polymeric carrier comprises a biodegradable polymer.
1783. The device of item 1637, further comprising a polymeric carrier, wherein the polymeric carrier comprises a non-biodegradable polymer. 1784. The device of item 1637, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrophilic polymer.
1785. The device of item 1637, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrophobic polymer.
1786. The device of item 1637, further comprising a polymeric carrier, wherein the polymeric carrier comprises a polymer having hydrophilic domains.
1787. The device of item 1637, further comprising a polymeric carrier, wherein the polymeric carrier comprises a polymer having hydrophobic domains.
1788. The device of item 1637, further comprising a polymeric carrier, wherein the polymeric carrier comprises a non-conductive polymer.
1789. The device of item 1637, further comprising a polymeric carrier, wherein the polymeric carrier comprises an elastomer.
1790. The device of item 1637, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrogel.
1791. The device of item 1637, further comprising a polymeric carrier, wherein the polymeric carrier comprises a silicone polymer.
1792. The device of item 1637, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrocarbon polymer.
1793. The device of item 1637, further comprising a polymeric carrier, wherein the polymeric carrier comprises a styrene-derived polymer. 1794. The device of item 1637, further comprising a polymeric carrier, wherein the polymeric carrier comprises a butadiene polymer.
1795. The device of item 1637, further comprising a polymeric carrier, wherein the polymeric carrier comprises a macromer.
1796. The device of item 1637, further comprising a polymeric carrier, wherein the polymeric carrier comprises a poly(ethylene glycol) polymer.
1797. The device of item 1637, further comprising a polymeric carrier, wherein the polymeric carrier comprises an amorphous polymer.
1798. The device of item 1637, further comprising a lubricious coating.
1799. The device of item 1637 wherein the anti-scarring agent is located within pores or holes of the device.
1800. The device of item 1637 wherein the anti-scarring agent is located within a channel, lumen, or divet of the device.
1801. The device of item 1637, further comprising a second pharmaceutically active agent.
1802. The device of item 1637, further comprising an antiinflammatory agent.
1803. The device of item 1637, further comprising an agent that inhibits infection. 1804. The device of item 1637, further comprising an agent that inhibits infection, wherein the agent is an anthracycline.
1805. The device of item 1637, further comprising an agent that inhibits infection, wherein the agent is doxorubicin.
1806. The device of item 1637, further comprising an agent that inhibits infection, wherein the agent is mitoxantrone.
1807. The device of item 1637, further comprising an agent that inhibits infection, wherein the agent is a fluoropyrimidine.
1808. The device of item 1637, further comprising an agent that inhibits infection, wherein the agent is 5-fluorouracil (5-FU).
1809. The device of item 1637, further comprising an agent that inhibits infection, wherein the agent is a folic acid antagonist.
1810. The device of item 1637, further comprising an agent that inhibits infection, wherein the agent is methotrexate.
1811. The device of item 1637, further comprising an agent that inhibits infection, wherein the agent is a podophylotoxin.
1812. The device of item 1637, further comprising an agent that inhibits infection, wherein the agent is etoposide.
1813. The device of item 1637, further comprising an agent that inhibits infection, wherein the agent is a camptothecin. 1814. The device of item 1637, further comprising an agent that inhibits infection, wherein the agent is a hydroxyurea.
1815. The device of item 1637, further comprising an agent that inhibits infection, wherein the agent is a platinum complex.
1816. The device of item 1637, further comprising an agent that inhibits infection, wherein the agent is cisplatin.
1817. The device of item 1637, further comprising an antithrombotic agent.
1818. The device of item 1637, further comprising a visualization agent.
1819. The device of item 1637, further comprising a visualization agent, wherein the visualization agent is a radiopaque material, wherein the radiopaque material comprises a metal, a halogenated compound, or a barium containing compound.
1820. The device of item 1637, further comprising a visualization agent, wherein the visualization agent is a radiopaque material, wherein the radiopaque material comprises barium, tantalum, or technetium.
1821. The device of item 1637, further comprising a visualization agent, wherein the visualization agent is a MRI responsive material.
1822. The device of item 1637, further comprising a visualization agent, wherein the visualization agent comprises a gadolinium chelate. 1823. The device of item 1637, further comprising a visualization agent, wherein the visualization agent comprises iron, magnesium, manganese, copper, or chromium.
1824. The device of item 1637, further comprising a visualization agent, wherein the visualization agent comprises an iron oxide compound.
1825. The device of item 1637, further comprising a visualization agent, wherein the visualization agent comprises a dye, pigment, or colorant.
1826. The device of item 1637, further comprising an echogenic material.
1827. The device of item 1637, further comprising an echogenic material, wherein the echogenic material is in the form of a coating.
1828. The device of item 1637 wherein the device is sterile.
1829. The device of item 1637 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device.
1830. The device of item 1637 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is connective tissue. 1831. The device of item 1637 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is muscle tissue.
1832. The device of item 1637 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is nerve tissue.
1833. The device of item 1637 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is epithelium tissue.
1834. The device of item 1637 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from the time of deployment of the device to about 1 year.
1835. The device of item 1637 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from about 1 month to 6 months.
1836. The device of item 1637 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from about 1 - 90 days.
1837. The device of item 1637 wherein the anti-scarring agent is released in effective concentrations from the device at a constant rate.
1838. The device of item 1637 wherein the anti-scarring agent is released in effective concentrations from the device at an increasing rate. 1839. The device of item 1637 wherein the anti-scarring agent is released in effective concentrations from the device at a decreasing rate.
1840. The device of item 1637 wherein the anti-scarring agent is released in effective concentrations from the composition comprising the anti-scarring agent by diffusion over a period ranging from the time of deployment of the device to about 90 days.
1841. The device of item 1637 wherein the anti-scarring agent is released in effective concentrations from the composition comprising the anti-scarring agent by erosion of the composition over a period ranging from the time of deployment of the device to about 90 days.
1842. The device of item 1637 wherein the device comprises about 0.01 Dg to about 10 Dg of the anti-scarring agent.
1843. The device of item 1637 wherein the device comprises about 10 Dg to about 10 mg of the anti-scarring agent.
1844. The device of item 1637 wherein the device comprises about 10 mg to about 250 mg of the anti-scarring agent.
1845. The device of item 1637 wherein the device comprises about 250 mg to about 1000 mg of the anti-scarring agent.
1846. The device of item 1637 wherein the device comprises about 1000 mg to about 2500 mg of the anti-scarring agent.
1847. The device of item 1637 wherein a surface of the device comprises less than 0.01 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied. 1848. The device of item 1637 wherein a surface of the device comprises about 0.01 Dg to about 1 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
1849. The device of item 1637 wherein a surface of the device comprises about 1 Dg to about 10 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
1850. The device of item 1637 wherein a surface of the device comprises about 10 Dg to about 250 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
1851. The device of item 1637 wherein a surface of the device comprises about 250 Dg to about 1000 Dg of the anti-scarring agent of anti- scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
1852. The device of item 1637 wherein a surface of the device comprises about 1000 Dg to about 2500 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
1853. A method as in any one of items 1637-1852, wherein the device is an intraocular lens device for preventing lens opacification.
1854. A method as in any one of items 1637-1852, wherein the device is a polymethylmethacrylate intraocular lens.
1855. A method as in any one of items 1637-1852, wherein the device is a silicone intraocular lens. 1856. A method as in any one of items 1637-1852, wherein the device is an achromatic lens.
1857. A method as in any one of items 1637-1852, wherein the device is a pseudophako.
1858. A method as in any one of items 1637-1852, wherein the device is a phakic lens.
1859. A method as in any one of items 1637-1852, wherein the device is an aphakic lens.
1860. A method as in any one of items 1637-1852, wherein the device is a multi-focal intraocular lens.
1861. A method as in any one of items 1637-1852, wherein the device is a hydrophilic and hydrophobic acrylic intraocular lens.
1862. A method as in any one of items 1637-1852, wherein the device is an intraocular implant.
1863. A method as in any one of items 1637-1852, wherein the device is an optic lens.
1864. A method as in any one of items 1637-1852, wherein the device is a rigid gas permeable lens.
1865. A method as in any one of items 1637-1852, wherein the device is a foldable intraocular lens. 1866. A method as in any one of items 1637-1852, wherein the device is a rigid intraocular lens.
1867. A method as in any one of items 1637-1852, wherein the device is a corrective implant for vision impairment.
1868. A method as in any one of items 1637-1852, wherein the device is an intraocular implant adapted for being used in conjunction with a transplant for the cornea.
1869. A method as in any one of items 1637-1852, wherein the device is an intraocular implant adapted for being used in conjunction with treatment of secondary cataract after extracapsular cataract extraction.
1870. A device, comprising a medical device for treating hypertropic scar or keloid (e.g., an implant) and an anti-scarring agent or a composition comprising an anti-scarring agent, wherein the agent inhibits scarring between the device and a host into which the device is implanted.
1871. The device of item 1870 wherein the agent is an adensosine A2A receptor antagonist.
1872. The device of item 1870 wherein the agent is an AKT inhibitor.
1873. The device of item 1870 wherein the agent is an alpha 2 integrin antagonist, wherein the alpha 2 integrin antagonist is Pharmaprojects No. 5754 (Merck KgaA).
1874. The device of item 1870 wherein the agent an alpha 4 integrin antagonist. 1875. The device of item 1870 wherein the agent is an alpha 7 nicotinic receptor agonist.
1876. The device of item 1870 wherein the agent is an angiogenesis inhibitor selected from the group consisting of AG-12,958 (Pfizer), ATN-161 (Attenuon LLC), neovastat, an angiogenesis inhibitor from Jerina AG (Germany), NM-3 (Mercian), VGA-1155 (Taisho), FCE-26644 (Pfizer), FCE-26950 (Pfizer), FPMA (Meiji Daries), FR-111142 (Fujisawa), GGTI-298, GM-1306 (Ligand), GPA-1734 (Novartis), NNC-47-0011 (Novo Nordisk), herbamycin (Nippon Kayaku), lenalidomide (Celegene), IP-10 (NIH), ABT-828 (Abbott), KIN-841 (Tokushima University, Japan), SF-1126 (Semafore Pharmaceuticals), laminin technology (NIH), CHIR-258 (Chiron), NVP-AEW541 (Novartis), NVP-AEW541 (Novartis), Vt16907 (Alchemia), OXI-8007 (Oxigene), EG-3306 (Ark Therapeutics), Maspin (Arriva), ABT- 567 (Abbott), PPI-2458 (Praecis Pharmaceuticals), CC-5079, CC-4089 (Celgene), HIF-1 alpha inhibitors (Xenova), S-247 (Pfizer), AP-23573 (Ariad), AZD-9935 (Astra Zeneca), mebendazole (Introgen Therapeutics), MetAP-2 inhibitors (GlaxoSmithKline), AG-615 (Angiogene Pharmaceuticals), Tie-2 antagonists (Hybrigenics), NC-381 , CYC-381, NC-169, NC-219, NC-383, NC-384, NC-407 (Lorus Therapeutics), ATN-224 (Attenuon), ON-01370 (Onconova), Vitronectin antagonists (Amgen), SDX-103 (Salmedix), Vitronectin antagonists (Shire), CHP (Riemser), TEK (Amgen), Anecortave acetate (Alcon), T46.2 (Matrix Therapeutics), HG-2 (Heptagen), TEM antagonists (Genzyme), Oxi-4500 (Oxigene), ATN-161 (Attenuon), WX-293 (Wilex), M-2025 (Metris Therapeutics), Alphastatin (BioActa), YH-16 (Yantai Rongchang), BIBF-1120 (Boehringer Ingelheim), BAY-57-9352 (Bayer), AS- 1404 (Cancer Research Technology), SC-77964 (Pfizer), glycomimetics (BioTie Therapies), TIE-2 Inhibitors (Ontogen), DlMI, Octamer (Octamer), ABR-215050 (Active Biotech), ABT-518 (Abbott), KDR inhibitors (Abbott), BSF-466895 (Abbott), SCH-221153 (Schering-Plough), DAC:antiangiogenic (ConjuChem), TFPI (EntreMed), AZD-2171 (Astra-Zenaca), CDC-394 (Celgene), LY290293 (EIi Lilly), IDN-5390 (Indena), Kdr Kinase Inhibitors (Merck), CT-113020, CT-116433, CT-116563, CT-31890, CT-32228) (Cell Therapeutics), A-299620 (Abbott), TWEAK Inhibitor (Amgen), VEGF modulators (Johnson and Johnson), Tum-N53, tumstatin (Genzyme), Thios- 1, Thios-2 (Thios Pharmaceuticals), MV-6401 (Miravant Medical Technologies), Spisulosine (PharmaMar), CEP-7055 (Cephalon), AUV-201 (Auvation), LM-609 (EIi Lilly), SKF-106615 (AnorMED), Oglufanide disodium (Cytran), BW-114 (Pharminox), Calreticulin (NIH), WX-678 (Wilex), SD-7784 (Pfizer), WX-UK1 (Wilex), SH-268 (Schering AG), 2-Me-PGA (Celgene), S- 137 (Pfizer), ZD-6126 (Angiogene Pharmaceuticals), SG-292 (SignalGen), Benefin (Lane Labs), A6, A36 (Angstrom), SB-2723005 (GlaxoSmithKline), SC-7 (Cell Therapeutics), ZEN-014 (AEterna Zentaris), 2-methoxyestradiol (EntreMed), NK-130119 (Nippon Kayaku), CC-10004 (Celgene), AVE- 8062A (Ajinomoto), Tacedinaline (Pfizer), Actinonin (Tokyo Metropolitan Institute of Medical Science), Lenalidomide (Celgene), VGA-1155, BTO-956 (SRI International), ER-68203-00 (Eisai), CT-6685 (UCB), JKC-362 (Phoenix Pharmaceuticals), DMI-3798 (DMI Biosciences, Angiomate (Ipsen), ZD-6474 (AstraZeneca), CEP-5214 (Cephalon), Canstatin (Genzyme), NM-3 (Mercian), Oridigm (MediQuest Therapeutics), Exherin (Adherex), BLS-0597 (Boston Life Sciences), PTC-299 (PTC Therapeutics), NPI-2358 (Nereus Pharmaceuticals), CGP-79787 (Novartis), AEE-788 (Novartis), CKD-732 (Chong Kun Dang), CP-564959 (OSI Pharmaceuticals), CM-101 (CarboMed), CT-2584, CT3501 (Cell Therapeutics), combretastatin and analogues and derivatives thereof (Oxigene), Rebimastat (Bristol-Meyers Squibb), Dextrin 2-sulfate (ML Laboratories), Cilengitide (Merk KGaA), NSC-706704 (Pharminox), KRN- 951 (Kirin Brewery), Ukrain, NSC-631570 (Nowicky Pharma), Tecogalan sodium (Daiichi Pharmaceutical), Tz-93 (Tsumura), TBC-1635 (Encysive Pharmaceuticals), TAN-1120 (Takeda), Semaxanib (Pfizer), BDI-7800 (Biopharmacopae), SD-186, SD-983 (Bristol-Meyers Squibb), SB-223245 (GlaxoSmithKline), SC-236 (Pfizer), RWJ-590973 (Johnson and Johnson), ILX-1850 (Genzyme), SC-68488, S-836 (Pfizer), CG-55069-11 (CuraGen), Ki-23057 (Kirin Brewery), CCX-700 (Chemoentryx), Pegaptanib octasodium (Giled Sciences), ANGIOCOL (available from Biostratum Inc.), or an analogue or derivative thereof.
1877. The device of item 1870 wherein the agent is an apoptosis antagonist.
1878. The device of item 1870 wherein the agent is an apoptosis activator.
1879. The device of item 1870 wherein the agent is a beta 1 integrin antagonist.
1880. The device of item 1870 wherein the agent is a beta tubulin inhibitor.
1881. The device of item 1870 wherein the agent is a blocker of enzyme production in Hepatitis C.
1882. The device of item 1870 wherein the agent is a Bruton's tyrosine kinase inhibitor.
1883. The device of item 1870 wherein the agent is a calcineurin inhibitor.
1884. The device of item 1870 wherein the agent is a caspase 3 inhibitor.
1885. The device of item 1870 wherein the agent is a CC chemokine receptor antagonist. 1886. The device of item 1870 wherein the agent is a cell cycle inhibitor selected from the group consisting of SNS-595 (Sunesis), synthadotin, KRX-0403, and an analogue or derivative thereof.
1887. The device of item 1870 wherein the agent is a cathepsin B inhibitor.
1888. The device of item 1870 wherein the agent is a cathepsin K inhibitor, wherein the cathepsin K inhibitor is 462795 (GlaxoSmithKline), INPL-022-D6 (Amura Therapeutics), an analogue or derivative thereof).
1889. The device of item 1870 wherein the agent is a cathepsin L inhibitor.
1890. The device of item 1870 wherein the agent is a CD40 antagonist.
1891. The device of item 1870 wherein the agent is a chemokine receptor agonist.
1892. The device of item 1870 wherein the agent is a chymase inhibitor.
1893. The device of item 1870 wherein the agent is a collagenase antagonist.
1894. The device of item 1870 wherein the agent is a CXCR antagonist. 1895. The device of item 1870 wherein the agent is a cyclin dependent kinase inhibitor selected from the group consisting of a CDK-1 inhibitor, a CDK-2 inhibitor, a CDK- 4 inhibitor, a CDK-6 inhibitor, a CAK1 inhibitor from GPC Biotech and Bristol-Myers Squibb, RGB-286199 (GPC Biotech), an anticancer agent from Astex Technology, a CAK1 inhibitor from GPC Biotech, a CDK inhibitor from Sanofi-Aventis, a CDK1/CDK2 inhibitor from Amgen, a CDK2 inhibitor from SUGEN-2 (Pfizer), a hearing loss therapy agent (Sound Pharmaceuticals), PD-0332991 (Pfizer), RGB-286199 (GPC Biotech), Ro-0505124 (Hoffmann-La Roche), a Ser/Thr kinase inhibitor from Lilly (EIi Lilly), CVT-2584 (CAS No. 199986-75-9) (CV Therapeutics), and an analogue or derivative thereof.
1896. The device of item 1870 wherein the agent is a cyclooxygenase 1 inhibitor.
1897. The device of item 1870 wherein the agent is. a DHFR inhibitor.
1898. The device of item 1870 wherein the agent is a dual integrin inhibitor.
1899. The device of item 1870 wherein the agent is an elastase inhibitor.
1900. The device of item 1870 wherein the agent is an elongation factor-1 alpha inhibitor.
1901. The device of item 1870 wherein the agent is an endothelial growth factor antagonist. 1902. The device of item 1870 wherein the agent is an endothelial growth factor receptor kinase inhibitor selected from the group consisting of sorafenib tosylate (Bayer), AAL-993 (Novartis), ABP-309 (Novartis), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), E- 7080 (Eisai), EG-3306 (Ark Therapeutics), EXEL-2880 (Exelixis), GW- 654652 (GlaxoSmithKline), a KDR inhibitor from LG Life Sciences, CT-6685 and CT-6729 (UCB), KRN-633 and KRN-951 (Kirin Brewery), OSI-930 (OSI Pharmaceuticals), SP-5.2 (Supratek Pharma), SU-11657 (Pfizer), a Tie-2 antagonist (Hybrigenics), a VEGFR-2 kinase inhibitor (Bristol-Myers Squibb), XL-647 (Exelixis), a KDR inhibitor from Abbott Laboratories, sorafenib tosylate, and an analogue or derivative thereof.
1903. The device of item 1870 wherein the agent is an endotoxin antagonist.
1904. The device of item 1870 wherein the agent is an epothilone and tubulin binder.
1905. The device of item 1870 wherein the agent is an estrogen receptor antagonist.
1906. The device of item 1870 wherein the agent is an FGF inhibitor.
1907. The device of item 1870 wherein the agent is a farnexyl transferase inhibitor.
1908. The device of item 1870 wherein the agent is farnesyltransferase inhibitor selected from the group of A-197574 (Abbott), a famesyltransferase inhibitor from Servier, FPTIII (Strathclyde Institute for Drug R), LB-42908 (LG Life Sciences), Pharmaprojects No. 5063 (Genzyme), Pharmaprojects No. 5597 (Ipsen), Yissum Project No. B-1055 (Yissum), and an analogue or derivative thereof.
1909. The device of item 1870 wherein the agent is an FLT-3 kinase inhibitor.
1910. The device of item 1870 wherein the agent is an FGF receptor kinase inhibitor.
1911. The device of item 1870 wherein the agent is a fibrinogen antagonist selected from the group consisting of AUV-201 (Auvation), MG-13926 (Sanofi-Aventis), plasminogen activator (CAS No. 105913-11-9) (from Sanofi-Aventis or UCB), plasminogen activator-2 (tPA- 2) (Sanofi-Aventis), pro-urokinase (CAS No. 82657-92-9) (Sanofi-Aventis), and an analogue or derivative thereof.
1912. The device of item 1870 wherein the agent is a heat shock protein 90 antagonist selected from the group consisting of SRN-005 (Sirenade), geldanamycin, NSC-33050 (17-allylaminogeldanamycin; 17- AAG), 17-dimethylaminoethylamino-17-demethoxy-geldanamycin (17- DMAG), rifabutin (rifamycin XIV, 1\4-didehydro-1-deoxy-1 ,4-dihydro-5'-(2- methylpropyl)-1-oxo-), and an analogue or derivative thereof.
1913. The device of item 1870 wherein the agent is a histone deacetylase inhibitor.
1914. The device of item 1870 wherein the agent is an HMGCoA reductase inhibitor selected from the group consisting of an atherosclerosis therapeutic from Lipid Sciences, ATI-16000 (ARYx Therapeutics), KS-01-019 (Kos Pharmaceuticals), Pharmaprojects No. 2197 (Sanofi-Aventi), RP 61969 (Sanofi-Aventis), and an analogue or derivative thereof.
1915. The device of item 1870 wherein the agent is an ICAM inhibitor.
1916. The device of item 1870 wherein the agent is an IL, ICE and IRAK antagonist, wherein the antagonist is a CJ-14877, CP-424174 (Pfizer), NF-61 (Negma-Lerads), and an analogue or derivative thereof.
1917. The device of item 1870 wherein the agent is an IL-2 inhibitor.
1918. The device of item 1870 wherein the agent is an immunosuppressant selected from the group consisting of teriflunomide (Sanofi Aventis), chlorsulfaquinoxalone (NSC-339004), chlorsulfaquinoxalone sulfate, CS-712 (Sankyo), ismomultin alfa (CAS No. 457913-93-8) (Akzo Nobel), antiallergics from GenPat77, antiinflammatories or AT-005 (Androclus Therapeutics), autoimmune disease therapeutics from EpiVax, BN-007 (Bone), budesonide (CAS No. 51333-22- 3) (MAP Pharmaceuticals), CO-14 (Genzyme), edratide (CAS No. 433922- 67-9) (Teva), EP-314 (Enanta), eprovafen (CAS No. 101335-99-3) (Sanofi- Aventis), HWA-131 (CAS No. 118788-41-3) (Sanofi-Aventis), immunomodulators from MerLion Pharmaceuticals, immunosuppressives from Alchemia, IPL-12 (Inflazyme), MDL-9563 (CAS No. 27086-86-8) (Sanofi-Aventis), Pharmaprojects No. 2330 (Sanofi-Aventis), Pharmaprojects No. 6426 (Abgenix), PXS-25 (Pharmaxis), rosmarinic acid (CAS No. 20283-92-5) (Sanofi-Aventis), RP 42927 or RP 54745 (CAS No. 135330-08-4) (Sanofi-Aventis), SGN-35 (Seattle Genetics), ST-1959 (Sigma-Tau), type I diabetes therapy from SYNX Pharma, UNIL-88 (Debiopharm), VP-025 (Vasogen), VR-694 (Vectura), PRTX-001 (Protalex), and an analogue or derivative thereof.
1919. The device of item 1870 wherein the agent is an IMPDH (inosine monophosphate).
1920. The device of item 1870 wherein the agent is an integrin antagonist.
1921. The device of item 1870 wherein the agent is an interleukin antagonist.
1922. The device of item 1870 wherein the agent is an inhibitor of type III receptor tyrosine kinase.
1923. The device of item 187Q. wherein the agent is an irreversible inhibitor of enzyme methionine aminopeptidase type 2.
1924. The device of item 1870 wherein the agent is an isozyme selective delta protein kinase C inhibitor.
1925. The device of item 1870 wherein the agent a JAK3 enzyme inhibitor.
1926. The device of item 1870 wherein the agent is a JNK inhibitor.
1927. The device of item 1870 wherein the agent is a kinase inhibitor. 1928. The device of item 1870 wherein the agent is kinesin antagonist.
1929. The device of item 1870 wherein the agent is a kinesin antagonist.
1930. The device of item 1870 wherein the agent is a leukotriene inhibitor and antagonist selected from the group consisting of ambicromil (CAS No. 58805-38-2) (Sanofi-Aventis), amelubant (CAS No. 346735-24-8) (Boehringer Ingelheim), DW-1141 (Dong Wha), ebselen (Daiichi Pharmaceutical), ibudilast (Kyorin), leucotriene inhibitors from Sanofi-Aventis, lymphotoxin-beta receptor (LT-D) from Biogen Idee, Pharmaprojects No. 1535 and 2728 (CAS No. 119340-33-9) (Sanofi- Aventis), R-112 (Rigel), Rev-5367 (CAS No. 92532-05-3) (Sanofi-Aventis), RG-14893 (CAS No. 141835-49-6) (Sanofi-Aventis), RG-5901-A (CAS No. 101910-24-1), 92532-23-5, RP 66153 (CAS No. 142422-79-5), RP 66364 (CAS No.186916-92-5), RP 69698 (CAS No. 141748-00-7) (Sanofi-Aventis), SC-411930 (Pfizer), SC-41930 (CAS No. 120072-59-5) (Pfizer), SC-50605 (CAS No. 138828-39-4) (Pfizer), SC-51146 (CAS No. 141059-52-1), SC- 53228 (CAS No. 153633-01-3) (Pfizer), spaglumic acid (ZY-15106) (CAS No.3106-85-2), 80619-64-3 (Novartis), tipredane (CAS No. 85197-77-9) (Bristol-Myers Squibb), U-75302 (CAS No. 119477-85-9) (Pfizer), and analogue or derivative thereof.
1931. The device of item 1870 wherein the agent is an MAP kinase inhibitor.
1932. The device of item 1870 wherein the agent is a matrix metalloproteinase inhibitor. 1933. The device of item 1870 wherein the agent is an MCP- CCR2 inhibitor.
1934. The device of item 1870 wherein the agent is an mTOR inhibitor.
1935. The device of item 1870 wherein the agent is an mTOR kinase inhibitor.
1936. The device of item 1870 wherein the agent is a microtubule inhibitor selected from the group consisting of antibody- maytansinoid conjugates from Biogen Idee, colchicines (MantiCore Pharmaceuticals), anticancer immunoconjugates from Johnson & Johnson, DIME from Octamer, gni-1f (GNI), huC242-DM4, huMy9-6-DM1 (ImmunoGen), IDN-5404 (Indena), IMO-098, IMOderm (Imotep), mebendazole (Introgen Therapeutics), microtubule poisons from Cambridge Enterprise, paclitaxel such as LOTAX from Aphios (CAS No. 33069-62-4), Genexol-PM from Samyang, Pharmaprojects No. 6383 (Tapestry Pharmaceuticals), RPR-112378 (Sanofi-Aventis), SGN-75 (Seattle Genetics), SPL-7435 (Starpharma), SSR-250411 (Sanofi-Aventis), trastuzumab-DM1 (Genentech), vinorelbine, and an analogue or derivative thereof.
1937. The device of item 1870 wherein the agent is an MIF inhibitor.
1938. The device of item 1870 wherein the agent is an MMP inhibitor.
1939. The device of item 1870 wherein the agent is a neurokinin (NK) antagonist selected from the group consisting of anthrotainin (CAS No. 148084-40-6) (Sanofi-Aventis), an IBS thereapeutic from ArQuIe, MDL-105212A (CAS No. 167261-60-1) (Ssanofi-Aventis), Pharmaprojects No. 2744, 3258 (CAS No. 139167-47-8) 4006, 4201 , or 5986 (Sanofi-Aventis), RP 67580 (CAS No. 135911-02-3), SR-144190 (CAS No. 201152-86-5), SSR-240600, SSR-241586 (Sanofi-Aventis), TKA-457 (Novartis), vestipitant mesylate (CAS No. 334476-64-1) (GlaxoSmithKline), Win-64821 (Sanofi-Aventis), PRX-96026 (Predix Pharmaceuticals), and an analogue or derivative thereof.
1940. The device of item 1870 wherein the agent is an NF kappa B inhibitor selected from the group consisting of AVE-0545 or AVE- 0547 (Sanofi-Aventis), bortezomib (CAS No. 179324-69-7) (Millennium Pharmaceuticals), dexanabinol (CAS No. 112924-45-5) (Pharmos), dexlipotam (Viatris), Pharmaprojects No. 6283 (INDRA) (OXiGENE), IPL- 576092 (CAS No. 137571-30-3) (Inflazyme), NFKB decoy (Corgentech), NFKB decoy oligo (AnGes MG), NFKB's from Ariad, osteoporosis treatments or S5 (F005) from Fulcrum Pharmaceuticals, P61 (Phytopharm), R-flurbiprofen (CAS No. 5104-49-4) (Encore Pharmaceuticals), and an analogue or derivative thereof.
1941. The device of item 1870 wherein the agent is a nitric oxide agonist.
1942. The device of item 1870 wherein the agent is an ornithine decarboxylase inhibitor.
1943. The device of item 1870 wherein the agent is a p38 MAP kinase inhibitor selected from the group consisting of AZD-6703 (AstraZeneca), JX-401 (Jexys Pharmaceuticals), BMS-2 (Bristol-Myers Squibb), a p38 MAP kinase inhibitor from Novartis, a p38-alpha MAP kinase inhibitor from Amphora, Pharmaprojects No. 5704 (Pharmacopeia), RPR- 200765A (Sanofi-Aventis), SD-282 (Johnson & Johnson), TAK-715 (Takeda), and an analogue or derivative thereof.
1944. The device of item 1870 wherein the agent is a palmitoyl-protein thioesterase inhibitor.
1945. The device of item 1870 wherein the agent is a PDGF receptor kinase inhibitor selected from the group consisting of AAL-993, AMN-107, or ABP-309 (Novartis), AMG-706 (Amgen), BAY-57-9352 (Bayer), CDP-860 (UCB), E-7080 (Eisai), imatinib (CAS No. 152459-95-5) (Novartis), OSI-930 (OSI Pharmaceuticals), RPR-127963E (Sanofi-Aventis), RWJ-540973 (Johnson & Johnson), sorafenib tosylate (Bayer), SU-11657 (Pfizer), tandutinib (CAS No. 387867-13-2) (Millennium Pharmaceuticals), vatalanib (Novartis), ZK-CDK (Schering AG), and an analogue or derivative thereof.
1946. The device of item 1870 wherein the agent is (-)- halofenate (Metabolex), AMG-131 (Amgen), antidiabetics from Japan Tobacco, AZD-4619, AZD-8450, AZD-8677 (AstraZeneca), DRF-10945, balaglitazone (Dr Reddy's), CS-00088, CS-00098 (Chipscreen Biosciences), E-3030 (Eisai), etalocib (CAS No. 161172-51-6) (EIi Lilly), GSK-641597 (Ligand), GSK-677954 (GlaxoSmithKline), GW-409544 (Ligand), GW- 590735 (GlaxoSmithKline), K-111 (Hoffmann-La Roche), LY-518674 (EIi Lilly), LY-674 (Ligand), LY-929 (Ligand), MC-3001 , MC-3002 (MaxoCore Pharmaceuticals), metformin HCI + pioglitazone (CAS No. 1115-70-4 and
112529-15-4), ACTOPLUS MET from Andrx), muraglitazar (CAS No. 331741-94-7) (Bristol-Myers Squibb), naveglitazar (Ligand), oleoylethanolamide (Kadmus Pharmaceuticals), ONO-5129, pioglitazone hydrochloride (CAS No. 111025-46-8 and 112529-15-4) (Takeda), PLX-204 (Plexxikon), PPAR agonists from Genfit, PPAR delta agonists from EIi Lilly, PPAR-alpha agonists from CrystalGenomics, PPAR-gamma modulators and PPAR-β modulators from CareX, rosiglitazone maleate (CAS No. 122324- 73-4 or 155141-29-0) (GlaxoSmithKline), rosiglitazone maleate/glimepir (CAS No. 155141-29-0 and 93479-97-1), AVANDARYL, rosiglitazone maleate/metformin extend (CAS No. 155141-29-0 and 657-24-9), AVANDAMET, rosiglitazone maleate+metformin, AVANDAMET (GlaxoSmithKline), tesaglitazar (AstraZeneca), and an analogue or derivative thereof.
1947. The device of item 1870 wherein the agent is a phosphatase inhibitor.
1948. The device of item 1870 wherein the agent is a phosphodiesterase (PDE) inhibitor selected from the group consisting of avanafil (Tanabe Seiyaku), dasantafil (CAS No. 569351-91-3) (Schering- Plough), A-906119 (CAS No. 134072-58-5), DL-850 (Sanofi-Aventis), GRC- 3015, GRC-3566, GRC-3886 (Glenmark), HWA-153 (CAS No. 56395-66-5) (Sanofi-Aventis), hydroxypumafentrine (Altana), IBFB-130011 , IBFB-14-016, IBFB-140301, IBFB-150007, IBFB-211913 (IBFB Pharma), L-826141 (Merck & Co), medorinone (CAS No. 88296-61-1) (Sanofi-Aventis), MEM- 1917 (Memory Pharmaceuticals), ND-1251 (Neuro3d), PDE inhibitors from ICOS, PDE IV inhibitors from Memory Pharmaceuticals and CrystalGenomics, Pharmaprojects No. 2742 and 6141 (Sanofi-Aventis), QAD-171 (Novartis), RHC-2963 (CAS No. 76993-12-9 and 76993-14-1), RPR-117658, RPR-122818 derivatives, SR-24870 , and RPR-132294 (Sanofi-Aventis), SK-350 (ln2Gen), stroke therapy agents from deCODE Genetics, TAS-203 (Taiho), tofimilast (CAS No. 185954-27-2) (Pfizer), UK- 371800 (Pfizer), WIN-65579 (CAS No. 158020-82-7) (Sanofi-Aventis), IBFB- 130020 (IBFB Pharma), OPC-6535 (CAS No. 145739-56-6) (Otsuka), a phosphodiesterase III inhibitor, enoximone, a phosphodiesterase IV inhibitor, fosfosal, Atopik (Barrier Therapeutics), triflusal, a phosphodiesterase V inhibitor, and an analogue or derivative thereof. 1949. The device of item 1870 wherein the agent is a PKC inhibitor.
1950. The device of item 1870 wherein the agent is a platelet activating factor antagonist.
1951. The device of item 1870 wherein the agent is a platelet- derived growth factor receptor kinase inhibitor.
1952. The device of item 1870 wherein the agent is a prolyl hydroxylase inhibitor.
1953. The device of item 1870 wherein the agent is a polymorphonuclear neutrophil inhibitor.
1954. The device of item 1870 wherein the agent is a protein kinase B inhibitor.
1955. The device of item 1870 wherein the agent is a protein kinase C stimulant.
1956. The device of item 1870 wherein the agent is a purine nucleoside analogue.
1957. The device of item 1870 wherein the agent is a purinoreceptor P2X antagonist.
1958. The device of item 1870 wherein the agent is a Raf kinase inhibitor. 1959. The device of item 1870 wherein the agent is a reversible inhibitor of ErbB1 and ErbB2.
1960. The device of item 1870 wherein the agent is a ribonucleoside triphosphate reductase inhibitor.
1961. The device of item 1870 wherein the agent is an SDF-1 antagonist.
1962. The device of item 1870 wherein the agent is a sheddase inhibitor.
1963. The device of item 1870 wherein the agent is an SRC inhibitor.
1964. The device of item 1870 whereinJhe agent is a stromelysin inhibitor.
1965. The device of item 1870 wherein the agent is an Syk kinase inhibitor.
1966. The device of item 1870 wherein the agent is a telomerase inhibitor.
1967. The device of item 1870 wherein the agent is a TGF beta inhibitor selected from the group consisting of pirfenidone (CAS No. 53179-13-8) (MARNAC), tranilast (CAS No. 53902-12-8) (Kissei), IN-1130 (ln2Gen), mannose-6-phosphate (BTG), TGF-β antagonists from Inflazyme (Pharmaprojects No. 6075), TGF-β antagonists from Sydney, non-industrial source), TGF-β I receptor kinase inhibitors from EIi Lilly, TGF-β receptor inhibitors from Johnson & Johnson, and an analogue or derivative thereof. 1968. The device of item 1870 wherein the agent is a TNFα antagonist or TACE inhibitor selected from the group consisting of adalimumab (CAS No. 331731-18-1) (Cambridge Antibody Technology), AGIX-4207 (AtheroGenics), AGT-1 (Advanced Biotherapy), an antiinflammatory from Borean Pharma, Cellzome, or Paradigm Therapeutics, anti-inflammatory vaccine (TNF-alpha kinoid) from Neovacs, humanized anti-TNF antibody or an anti-TNF MAb (CB0006) Celltech (UCB), apratastat (CAS No. 287445-51-0) (Wyeth), BMS-561392 (Bristol-Myers Squibb), BN- 006 (Bone), certolizumab pegol (CAS No. 428863-50-7 or CH-138 (UCB), cilomilast (CAS No. 153259-65-5) (GlaxoSmithKline), CR-1 (Nuada Pharmaceuticals), CRx-119 (CombinatoRx), D-5410 (UCB)1 dacopafant (CAS No. 125372-33-0) (Sanofi-Aventis), dersalazine (CAS No. 188913-57- 7/188913-58-8) (Uriach), etanercept (CAS No. 185243-69-0) (Amgen), ethyl pyruvate (Critical (Critical Therapeutics), golimumab (CAS No. 476181-74-5) (Johnson & Johnson), hormono-immunotherapy from Ipsen, CDP571 (e.g., Humicade from UCB), IC-485 (ICOS), infliximab (CAS No. 170277-31-3) (Johnson & Johnson), IP-751 (Manhattan Pharmaceuticals), ISIS-104838 (CAS No. 250755-32-9) (ISIS Pharmaceuticals), lenalidomide (CAS No. 191732-72-6) (Celgene), lentinan (CAS No. 37339-90-5) (Ajinomoto), MDL- 201112 (CAS No. 142130-73-2) (Sanofi-Aventis), medroxyprogesterone (CAS No. 520-85-4) (InKine Pharmaceutical), N-acetylcysteine (CAS No. 616-91-1) (Zambon), NBE-P2 (DIREVO Biotech), nerelimomab (CAS No. 162774-06-3) (Chiron), OM-294DP (OM PHARMA), onercept (CAS No. 199685-57-9) (Yeda), PASSTNF-alpha (Verigen), pentoxifylline or oxypentifylline (Sanofi-Aventis), Pharmaprojects No. 4091 , 4241 , 4295, or 4488 (Sanofi-Aventis), Pharmaprojects No. 5480 (Amgen), Pharmaprojects No. 6749 (Cengent), pirfenidone (CAS No. 53179-13-8) (MARNAC), RPR- 132294 (Sanofi-Aventis), S5 (F002) (Fulcrum Pharmaceuticals), simvastatin (CAS No. 79902-63-9) (Merck & Co), STA-6292 (Synta Pharmaceuticals), tacrolimus (CAS No. 104987-11-3) (Fujisawa LifeCycle Pharma), talactoferrin alfa (CAS No. 308240-58-6) (Agennix), thalidomide (CAS No. 50-35-1) (Celgene), TNF antagonists form ProStrakan, and Synergen, TNF inhibitors (Amgen), TNF-alpha antagonists from Dynavax Technologies and Jerina AG (Germany), TNF-alpha inhibitors from IBFB Pharma and Xencor (Xencor), torbafylline (CAS No. 105102-21-4) (Sanofi-Aventis), UR-1505 (Uriach), VT-346 (Viron Therapeutics), YSIL6 (Y's Therapeutics), YSTH2 (Y's Therapeutics), NPI-1302a-3 (Nereus Pharmaceuticals, a TNF antagonist from Jerina AG (Germany), dersalazine, and an analogue or derivative thereof.
1969. The device of item 1870 wherein the agent is a Toll receptor inhibitor.
1970. The device of item 1870 wherein the agent is a tubulin antagonist.
1971. The device of item 1870 wherein the agent is a tyrosine kinase inhibitor selected from the group consisting of SU-011248, SUTENT from Pfizer Inc. (New York, NY), BMS-354825, PN-355 (Paracelsian Pharmaceuticals), AGN-199659 (Allergan), AAL-993 or ABP-309 (Novartis), adaphostin (NIH), AEE-788 (Novartis), AG-013736 (OSI Pharmaceuticals), AG-13736 (Pfizer), ALT-110 (Alteris Therapeutics), AMG-706 (Amgen), anticancer MAbs from Xencor, anti-EGFrvlll MAbs from Abgenix, anti-HER2 MAb from Abiogen, AZD-2171 or AZD-9935 (AstraZeneca), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), CEP-5214 (Cephalon), CEP- 7055 (Cephalon), cetuximab (ImClone Systems), CHIR-200131 and CHIR- 258 (Chiron), CP-547632 (OSI Pharmaceuticals), CP-724714 (Pfizer), CT- 301 (Creabilis Therapeutics), D-69491 (Baxter International), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EGFR/ErbB2 inhibitors from Array BioPharma, erlotinib (CAS No. 183319-69-9) (OSI Pharmaceuticals), EXEL-2880 (Exelixis), FK-778 (Sanofi-Aventis) , gefitinib (CAS No. 184475-35-2) (AstraZeneca), GW-2286 or GW-654652 (GlaxoSmithKline), her2/neu antigen from AlphaVax, HER-2/neu inhibitor from Generex, Herzyme (Medipad) (Sirna Therapeutics), HKI-272 (Wyeth), HuMax-EGFr (Genmab), idronoxil (CAS No. 81267-65-4) (Novogen), IGF-1 inhibitors from Ontogen, IMC-11F8 (ImCione Systems), kahalalide F (CAS No. 149204-42-2) (PharmaMar), KDR inhibitor from LG Life Sciences, KDR inhibitors from Abbott Laboratories, KDR kinase inhibitors (UCB), Kdr kinase inhibitors from Merck & Co, KRN-633 and KRN-951 (Kirin Brewery), KSB-102 (Xenova), lapatinib ditosylate (CAS No. 388082-78-8) (GlaxoSmithKline), matuzumab (Merck KGaA), MDX-214 (Medarex), ME-103 (Pharmexa), MED-A300 (Gerolymatos), MNAC-13 (Lay Line Genomics), nimotuzumab (Center of Molecular Immunology), NSC-330507 or NSC-707545 (NIH)1 NV-50 (Novogen), OSI-930 (OSI Pharmaceuticals), panitumumab (Abgenix), pelitinib (CAS No. 287933-82-7) (Wyeth), pertuzumab (CAS No. 380610-27- 5) (Genentech), Pharmaprojects No. 3985 (Sanofi-Aventis), prostate cancer therapeutics from Sequenom (SQPC35, SQPC36, SQPC90), removab and remoxab (Trion Pharma), RG-13022 (CAS No. 136831-48-6), RG-13291 - (CAS No. 138989-50-1), or RG-14620 (CAS No. 136831-49-7) (Sanofi- Aventis), RM-6427 (Romark), RNAi breast cancer therapy from Benitec, RP 53801 (CAS No. 125882-88-4) (Sanofi-Aventis), sorafenib tosylate (Bayer), SU-11657 (Pfizer), Tie-2 antagonists from Semaia (Hybrigenics), Tie-2 inhibitors from Ontogen, trastuzumab (CAS No. 180288-69-1) (Genentech), tyrosine kinase inhibitors from Sanofi-Aventis, U3-1287, U3-1565, U3-1784, U3-1800 (U3 Pharma), vatalanib (Novartis), VEGFR-2 kinase inhibitor from Bristol-Myers Squibb, XL-647 (Exelixis), ZD-6474 (AstraZeneca), ZK-CDK (Schering AG), an EGFR tyrosine kinase inhibitor, EKB-569 (Wyeth), and an analogue or derivative thereof.
1972. The device of item 1870 wherein the agent is a VEGF inhibitor. 1973. The device of item 1870 wherein the agent is a vitamin D receptor agonist.
1974. The device of item 1870 wherein the agent is ZD-6474 (an angiogenesis inhibitor).
1975. The device of item 1870 wherein the agent is AP-23573 (an mTOR inhibitor).
1976. The device of item 1870 wherein the agent is synthadotin (a tubulin antagonist).
1977. The device of item 1870 wherein the agent is S-0885 (a collagenase inhibitor).
1978. The device of item 1870 wherein the agent is aplidine (an elongation factor-1 alpha inhibitor).
1979. The device of item 1870 wherein the agent is ixabepilone (an epithilone).
1980. The device of item 1870 wherein the agent is IDN-5390 (an angiogenesis inhibitor).
1981. The device of item 1870 wherein the agent is SB- 2723005 (an angiogenesis inhibitor).
1982. The device of item 1870 wherein the agent is ABT-518 (an angiogenesis inhibitor). 1983. The device of item 1870 wherein the agent is combretastatin (an angiogenesis inhibitor).
1984. The device of item 1870 wherein the agent is anecortave acetate (an angiogenesis inhibitor).
1985. The device of item 1870 wherein the agent is SB- 715992 (a kinesin antagonist).
1986. The device of item 1870 wherein the agent is temsirolimus (an mTOR inhibitor).
1987. The device of item 1870 wherein the agent is adalimumab (a TNFα antagonist).
1988. The device of item 1870, further comprising a polymer.
1989. The device of item 1870 wherein the anti-scarring agent inhibits adhesion between the device and a host into which the device is implanted.
1990. The device of item 1870 wherein the device delivers the anti-scarring agent locally to tissue proximate to the device.
1991. The device of item 1870, further comprising a coating, wherein the coating comprises the anti-scarring agent.
1992. The device of item 1870, further comprising a coating, wherein the coating is disposed on a surface of the device. 1993. The device of item 1870, further comprising a coating, wherein the coating directly contacts the device.
1994. The device of item 1870, further comprising a coating, wherein the coating indirectly contacts the device.
1995. The device of item 1870, further comprising a coating, wherein the coating partially covers the device.
1996. The device of item 1870, further comprising a coating, . wherein the coating completely covers the device.
1997. The device of item 1870, further comprising a coating, wherein the coating is a uniform coating.
1998. The device of item 1870, further comprising a coating, wherein the coating is a non-uniform coating.
1999. The device of item 1870, further comprising a coating, wherein the coating is a discontinuous coating.
2000. The device of item 1870, further comprising a coating, wherein the coating is a patterned coating.
2001. The device of item 1870, further comprising a coating, wherein the coating has a thickness of 100 Dm or less.
2002. The device of item 1870, further comprising a coating, wherein the coating has a thickness of 10 Dm or less. 2003. The device of item 1870, further comprising a coating, wherein the coating adheres to the surface of the device upon deployment of the device.
2004. The device of item 1870, further comprising a coating, wherein the coating is stable at room temperature for a period of 1 year.
2005. The device of item 1870, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 0.0001% to about 1% by weight.
2006. The device of item 1870, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 1 % to about 10% by weight.
- 2007. The device of item 187.0, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 10% to about 25% by weight.
2008. The device of item 1870, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 25% to about 70% by weight.
2009. The device of item 1870, further comprising a coating, wherein the coating further comprises a polymer.
2010. The device of item 1870, further comprising a first coating having a first composition and the second coating having a second composition. 2011. The device of item 1870, further comprising a first coating having a first composition and the second coating having a second composition, wherein the first composition and the second composition are different.
2012. The device of item 1870, further comprising a polymeric carrier, wherein the polymeric carrier comprises a copolymer.
2013. The device of item 1870, further comprising a polymeric carrier, wherein the polymeric carrier comprises a block copolymer.
2014. The device of item 1870, further comprising a polymeric carrier, wherein the polymeric carrier comprises a random copolymer.
2015. The device of item 1870, further comprising a polymeric carrier, wherein the polymeric carrier comprises a biodegradable polymer.
2016. The device of item 1870, further comprising a polymeric carrier, wherein the polymeric carrier comprises a non-biodegradable polymer.
2017. The device of item 1870, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrophilic polymer.
2018. The device of item 1870, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrophobic polymer.
2019. The device of item 1870, further comprising a polymeric carrier, wherein the polymeric carrier comprises a polymer having hydrophilic domains. 2020. The device of item 1870, further comprising a polymeric carrier, wherein the polymeric carrier comprises a polymer having hydrophobic domains.
2021. The device of item 1870, further comprising a polymeric carrier, wherein the polymeric carrier comprises a non-conductive polymer.
2022. The device of item 1870, further comprising a polymeric carrier, wherein the polymeric carrier comprises an elastomer.
2023. The device of item 1870, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrogel.
2024. The device of item 1870, further comprising a polymeric carrier, wherein the polymeric carrier comprises a silicone polymer.
2025. The device of item 1870, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrocarbon polymer.
2026. The device of item 1870, further comprising a polymeric carrier, wherein the polymeric carrier comprises a styrene-derived polymer.
2027. The device of item 1870, further comprising a polymeric carrier, wherein the polymeric carrier comprises a butadiene polymer.
2028. The device of item 1870, further comprising a polymeric carrier, wherein the polymeric carrier comprises a macromer.
2029. The device of item 1870, further comprising a polymeric carrier, wherein the polymeric carrier comprises a poly(ethyiene glycol) polymer. 2030. The device of item 1870, further comprising a polymeric carrier, wherein the polymeric carrier comprises an amorphous polymer.
2031. The device of item 1870, further comprising a lubricious coating.
2032. The device of item 1870 wherein the anti-scarring agent is located within pores or holes of the device.
2033. The device of item 1870 wherein the anti-scarring agent is located within a channel, lumen, or divet of the device.
2034. The device of item 1870, further comprising a second pharmaceutically active agent.
2035. The device of item 1870, further comprising an antiinflammatory agent.
2036. The device of item 1870, further comprising an agent that inhibits infection.
2037. The device of item 1870, further comprising an agent that inhibits infection, wherein the agent is an anthracycline.
2038. The device of item 1870, further comprising an agent that inhibits infection, wherein the agent is doxorubicin.
2039. The device of item 1870, further comprising an agent that inhibits infection, wherein the agent is mitoxantrone. 2040. The device of item 1870, further comprising an agent that inhibits infection, wherein the agent is a fluoropyrimidine.
2041. The device of item 1870, further comprising an agent that inhibits infection, wherein the agent is 5-fluorouracil (5-FU).
2042. The device of item 1870, further comprising an agent that inhibits infection, wherein the agent is a folic acid antagonist.
2043. The device of item 1870, further comprising an agent that inhibits infection, wherein the agent is methotrexate.
2044. The device of item 1870, further comprising an agent that inhibits infection, wherein the agent is a podophylotoxin.
2045. The device of item 1870, further comprising an agent that inhibits infection, wherein the agent is etoposide.
2046. The device of item 1870, further comprising an agent that inhibits infection, wherein the agent is a camptothecin.
2047. The device of item 1870, further comprising an agent that inhibits infection, wherein the agent is a hydroxyurea.
2048. The device of item 1870, further comprising an agent that inhibits infection, wherein the agent is a platinum complex.
2049. The device of item 1870, further comprising an agent that inhibits infection, wherein the agent is cisplatin. 2050. The device of item 1870, further comprising an antithrombotic agent.
2051. The device of item 1870, further comprising a visualization agent.
2052. The device of item 1870, further comprising a visualization agent, wherein the visualization agent is a radiopaque material, wherein the radiopaque material comprises a metal, a halogenated compound, or a barium containing compound.
2053. The device of item 1870, further comprising a visualization agent, wherein the visualization agent is a radiopaque material, wherein the radiopaque material comprises barium, tantalum, or technetium.
2054. The device of item 1870, further comprising a visualization agent, wherein the visualization agent is a MRI responsive material.
2055. The device of item 1870, further comprising a visualization agent, wherein the visualization agent comprises a gadolinium chelate.
2056. The device of item 1870, further comprising a visualization agent, wherein the visualization agent comprises iron, magnesium, manganese, copper, or chromium.
2057. The device of item 1870, further comprising a visualization agent, wherein the visualization agent comprises an iron oxide compound. 2058. The device of item 1870, further comprising a visualization agent, wherein the visualization agent comprises a dye, pigment, or colorant.
2059. The device of item 1870, further comprising an echogenic material.
2060. The device of item 1870, further comprising an echogenic material, wherein the echogenic material is in the form of a coating.
2061. The device of item 1870 wherein the device is sterile.
2062. The device of item 1870 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device. . .
2063. The device of item 1870 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is connective tissue.
2064. The device of item 1870 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is muscle tissue.
2065. The device of item 1870 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is nerve tissue. 2066. The device of item 1870 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is epithelium tissue.
2067. The device of item 1870 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from the time of deployment of the device to about 1 year.
2068. The device of item 1870 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from about 1 month to 6 months.
2069. The device of item 1870 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from about 1 - 90 days.
2070. The device of item 1870 wherein the anti-scarring agent is released in effective concentrations from the device at a constant rate.
2071. The device of item 1870 wherein the anti-scarring agent is released in effective concentrations from the device at an increasing rate.
2072. The device of item 1870 wherein the anti-scarring agent is released in effective concentrations from the device at a decreasing rate.
2073. The device of item 1870 wherein the anti-scarring agent is released in effective concentrations from the composition comprising the anti-scarring agent by diffusion over a period ranging from the time of deployment of the device to about 90 days. 2074. The device of item 1870 wherein the anti-scarring agent is released in effective concentrations from the composition comprising the anti-scarring agent by erosion of the composition over a period ranging from the time of deployment of the device to about 90 days.
2075. The device of item 1870 wherein the device comprises about 0.01 Dg to about 10 Dg of the anti-scarring agent.
2076. The device of item 1870 wherein the device comprises about 10 Dg to about 10 mg of the anti-scarring agent.
2077. The device of item 1870 wherein the device comprises about 10 mg to about 250 mg of the anti-scarring agent.
2078. The device of item 1870 wherein the device comprises about 250 mg to about 1000 mg of the anti-scarring agent.
2079. The device of item 1870 wherein the device comprises about 1000 mg to about 2500 mg of the anti-scarring agent.
2080. The device of item 1870 wherein a surface of the device comprises less than 0.01 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
2081. The device of item 1870 wherein a surface of the device comprises about 0.01 Dg to about 1 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
2082. The device of item 1870 wherein a surface of the device comprises about 1 Dg to about 10 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied. 2083. The device of item 1870 wherein a surface of the device comprises ab oouutt 1100 DDgg ttoo aabboouutt 225500 DDgg ooff tthhee aannttii--ssccaarrrriinngg ; agent per mm2 of device surface to which the anti-scarring agent is applied.
2084. The device of item 1870 wherein a surface of the device comprises about 250 Dg to about 1000 Dg of the anti-scarring agent of anti- scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
2085. The device of item 1870 wherein a surface of the device comprises about 1000 Dg to about 2500 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
2086. A method as in any one of items 1870-2085, wherein the device is a device for treating hypertropic scar or keloid that comprises an external tissue expansion device.
2087. A method as in any one of items 1870-2085, wherein the device is a device for treating hypertropic scar or keloid that comprises a masking element, and wherein the masking element may be pressed onto the scar tissue.
2088. A method as in any one of items 1870-2085, wherein the device is a device for treating hypertropic scar or keloid that comprises a locking element and a grasping structure so that the dermal and epidermal layers of a skin wound can be pushed together.
2089. A device, comprising a vascular graft (e.g., an implant) and an anti-scarring agent or a composition comprising an anti-scarring agent, wherein the agent inhibits scarring between the device and a host into which the device is implanted. 2090. The device of item 2089 wherein the agent is an adensosine A2A receptor antagonist.
2091. The device of item 2089 wherein the agent is an AKT inhibitor.
2092. The device of item 2089 wherein the agent is an alpha 2 integrin antagonist, wherein the alpha 2 integrin antagonist is Pharmaprojects No. 5754 (Merck KgaA).
2093. The device of item 2089 wherein the agent an alpha 4 integrin antagonist.
2094. The device of item 2089 wherein the agent is an alpha 7 nicotinic receptor agonist.
2095. The device of item 2089 wherein the agent is an angiogenesis inhibitor selected from the group consisting of AG-12,958 (Pfizer), ATN-161 (Attenuon LLC), neovastat, an angiogenesis inhibitor from Jerina AG (Germany), NM-3 (Mercian), VGA-1155 (Taisho), FCE-26644 (Pfizer), FCE-26950 (Pfizer), FPMA (Meiji Daries), FR-111142 (Fujisawa), GGTI-298, GM-1306 (Ligand), GPA-1734 (Novartis), NNC-47-0011 (Novo Nordisk), herbamycin (Nippon Kayaku), lenalidomide (Celegene), IP-10 (NIH), ABT-828 (Abbott), KIN-841 (Tokushima University, Japan), SF-1126 (Semafore Pharmaceuticals), laminin technology (NIH), CHIR-258 (Chiron), NVP-AEW541 (Novartis), NVP-AEW541 (Novartis), Vt16907 (Alchemia), OXI-8007 (Oxigene), EG-3306 (Ark Therapeutics), Maspin (Arriva), ABT- 567 (Abbott), PPI-2458 (Praecis Pharmaceuticals), CC-5079, CC-4089 (Celgene), HIF-1 alpha inhibitors (Xenova), S-247 (Pfizer), AP-23573 (Ariad), AZD-9935 (Astra Zeneca), mebendazole (Introgen Therapeutics), MetAP-2 inhibitors (GlaxoSmithKline), AG-615 (Angiogene Pharmaceuticals), Tie-2 antagonists (Hybrigenics), NC-381, CYC-381, NC-169, NC-219, NC-383, NC-384, NC-407 (Lorus Therapeutics), ATN-224 (Attenuon), ON-01370 (Onconova), Vitronectin antagonists (Amgen), SDX-103 (Salmedix), Vitronectin antagonists (Shire), CHP (Riemser), TEK (Amgen), Anecortave acetate (Alcon), T46.2 (Matrix Therapeutics), HG-2 (Heptagen), TEM antagonists (Genzyme), Oxi-4500 (Oxigene), ATN-161 (Attenuon), WX-293 (Wilex), M-2025 (Metris Therapeutics), Alphastatin (BioActa), YH-16 (Yantai Rongchang), BIBF-1120 (Boehringer Ingelheim), BAY-57-9352 (Bayer), AS- 1404 (Cancer Research Technology), SC-77964 (Pfizer), glycomimetics (BioTie Therapies), TIE-2 Inhibitors (Ontogen), DIMI, Octamer (Octamer), ABR-215050 (Active Biotech), ABT-518 (Abbott), KDR inhibitors (Abbott), BSF-466895 (Abbott), SCH-221153 (Schering-Plough), DAC:antiangiogenic (ConjuChem), TFPI (EntreMed), AZD-2171 (Astra-Zenaca), CDC-394 (Celgene), LY290293 (EIi Lilly), IDN-5390 (Indena), Kdr Kinase Inhibitors (Merck), CT-113020, CT-116433, CT-116563, CT-31890, CT-32228) (Cell Therapeutics), A-299620 (Abbott), TWEAK Inhibitor (Amgen), VEGF modulators (Johnson and Johnson), Tum-N53, tumstatin (Genzyme), Thios- 1 , Thios-2 (Thios Pharmaceuticals), MV-6401 (Miravant Medical Technologies), Spisulosine (PharmaMar), CEP-7055 (Cephalon), AUV-201 (Auvation), LM-609 (EIi Lilly), SKF-106615 (AnorMED), Oglufanide disodium (Cytran), BW-114 (Pharminox), Calreticulin (NIH), WX-678 (Wilex), SD-7784 (Pfizer), WX-UK1 (Wilex), SH-268 (Schering AG), 2-Me-PGA (Celgene), S- 137 (Pfizer), ZD-6126 (Angiogene Pharmaceuticals), SG-292 (SignalGen), Benefin (Lane Labs), A6, A36 (Angstrom), SB-2723005 (GlaxoSmithKline), SC-7 (Cell Therapeutics), ZEN-014 (AEterna Zentaris), 2-methoxyestradiol (EntreMed), NK-130119 (Nippon Kayaku), CC-10004 (Celgene), AVE- 8062A (Ajinomoto), Tacedinaline (Pfizer), Actinonin (Tokyo Metropolitan Institute of Medical Science), Lenalidomide (Celgene), VGA-1155, BTO-956 (SRI International), ER-68203-00 (Eisai), CT-6685 (UCB), JKC-362 (Phoenix Pharmaceuticals), DMI-3798 (DMI Biosciences, Angiomate (Ipsen), ZD-6474 (AstraZeneca), CEP-5214 (Cephalon), Canstatin (Genzyme), NM-3 (Mercian), Oridigm (MediQuest Therapeutics), Exherin (Adherex), BLS-0597 (Boston Life Sciences), PTC-299 (PTC Therapeutics), NPI-2358 (Nereus Pharmaceuticals), CGP-79787 (Novartis), AEE-788 (Novartis), CKD-732 (Chong Kun Dang), CP-564959 (OSI Pharmaceuticals), CM-101 (CarboMed), CT-2584, CT3501 (Cell Therapeutics), combretastatin and analogues and derivatives thereof (Oxigene), Rebimastat (Bristol-Meyers Squibb), Dextrin 2-sulfate (ML Laboratories), Cilengitide (Merk KGaA), NSC-706704 (Pharminox), KRN- 951 (Kirin Brewery), Ukrain, NSC-631570 (Nowicky Pharma), Tecogalan sodium (Daiichi Pharmaceutical), Tz-93 (Tsumura), TBC-1635 (Encysive Pharmaceuticals), TAN-1120 (Takeda), Semaxanib (Pfizer), BDI-7800 (Biopharmacopae), SD-186, SD-983 (Bristol-Meyers Squibb), SB-223245 (GlaxoSmithKline), SC-236 (Pfizer), RWJ-590973 (Johnson and Johnson), ILX-1850 (Genzyme), SC-68488, S-836 (Pfizer), CG-55069-11 (CuraGen), Ki-23057 (Kirin Brewery), CCX-700 (Chemoentryx), Pegaptanib octasodium (Giled Sciences), ANGIOCOL (available from Biostratum Inc.), or an analogue or derivative thereof.
2096. The device of item 2089 wherein the agent is an apoptosis antagonist.
2097. The device of item 2089 wherein the agent is an apoptosis activator.
2098. The device of item 2089 wherein the agent is a beta 1 integrin antagonist.
2099. The device of item 2089 wherein the agent is a beta tubulin inhibitor. 2100. The device of item 2089 wherein the agent is a blocker of enzyme production in Hepatitis C.
2101. The device of item 2089 wherein the agent is a Bruton's tyrosine kinase inhibitor.
2102. The device of item 2089 wherein the agent is a calcineurin inhibitor.
2103. The device of item 2089 wherein the agent is a caspase 3 inhibitor.
2104. The device of item 2089 wherein the agent is a CC chemokine receptor antagonist.
2105. The device of item 2089 wherein the agent is a cell cycle inhibitor selected from the group consisting of SNS-595 (Sunesis), synthadotin, KRX-0403, and an analogue or derivative thereof.
2106. The device of item 2089 wherein the agent is a cathepsin B inhibitor.
2107. The device of item 2089 wherein the agent is a cathepsin K inhibitor, wherein the cathepsin K inhibitor is 462795 (GlaxoSmithKline), INPL-022-D6 (Amura Therapeutics), an analogue or derivative thereof).
2108. The device of item 2089 wherein the agent is a cathepsin L inhibitor. 2109. The device of item 2089 wherein the agent is a CD40 antagonist.
2110. The device of item 2089 wherein the agent is a chemokine receptor agonist.
2111. The device of item 2089 wherein the agent is a chymase inhibitor.
2112. The device of item 2089 wherein the agent is a collagenase antagonist.
2113. The device of item 2089 wherein the agent is a CXCR antagonist.
2114. The device of item 2089 wherein the agent is a cyclin dependent kinase inhibitor selected from the group consisting of a CDK-1 inhibitor, a CDK-2 inhibitor, a CDK- 4 inhibitor, a CDK-6 inhibitor, a CAK1 inhibitor from GPC Biotech and Bristol-Myers Squibb, RGB-286199 (GPC Biotech), an anticancer agent from Astex Technology, a CAK1 inhibitor from GPC Biotech, a CDK inhibitor from Sanofi-Aventis, a CDK1/CDK2 inhibitor from Amgen, a CDK2 inhibitor from SUGEN-2 (Pfizer), a hearing loss therapy agent (Sound Pharmaceuticals), PD-0332991 (Pfizer), RGB-286199 (GPC Biotech), Ro-0505124 (Hoffmann-La Roche), a Ser/Thr kinase inhibitor from Lilly (EIi Lilly), CVT-2584 (CAS No. 199986-75-9) (CV Therapeutics), and an analogue or derivative thereof.
2115. The device of item 2089 wherein the agent is a cyclooxygenase 1 inhibitor. 2116. The device of item 2089 wherein the agent is a DHFR inhibitor.
2117. The device of item 2089 wherein the agent is a dual integrin inhibitor.
2118. The device of item 2089 wherein the agent is an elastase inhibitor.
2119. The device of item 2089 wherein the agent is an elongation factor-1 alpha inhibitor.
2120. The device of item 2089 wherein the agent is an endothelial growth factor antagonist.
2121. The device of item 2089 wherein the agent is an endothelial growth factor receptor kinase inhibitor selected from the group consisting of sorafenib tosylate (Bayer), AAL-993 (Novartis), ABP-309 (Novartis), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), E- 7080 (Eisai), EG-3306 (Ark Therapeutics), EXEL-2880 (Exelixis), GW- 654652 (GlaxoSmithKline), a KDR inhibitor from LG Life Sciences, CT-6685 and CT-6729 (UCB), KRN-633 and KRN-951 (Kirin Brewery), OSI-930 (OSI Pharmaceuticals), SP-5.2 (Supratek Pharma), SLM 1657 (Pfizer), a Tie-2 antagonist (Hybrigenics), a VEGFR-2 kinase inhibitor (Bristol-Myers Squibb), XL-647 (Exelixis), a KDR inhibitor from Abbott Laboratories, sorafenib tosylate, and an analogue or derivative thereof.
2122. The device of item 2089 wherein the agent is an endotoxin antagonist. 2123. The device of item 2089 wherein the agent is an epothilone and tubulin binder.
2124. The device of item 2089 wherein the agent is an estrogen receptor antagonist.
2125. The device of item 2089 wherein the agent is an FGF inhibitor.
2126. The device of item 2089 wherein the agent is a famexyl transferase inhibitor.
2127. The device of item 2089 wherein the agent is fa mesyltransf erase inhibitor selected from the group of A-197574 (Abbott), a farnesyltransferase inhibitor from Servier, FPTIII (Strathclyde Institute for Drug. R)1. L-B-429Q8 (LG Life Sciences), Pharmaprojects No. 5063 (Genzyme), Pharmaprojects No. 5597 (Ipsen), Yissum Project No. B-1055 (Yissum), and an analogue or derivative thereof.
2128. The device of item 2089 wherein the agent is an FLT-3 kinase inhibitor.
2129. The device of item 2089 wherein the agent is an FGF receptor kinase inhibitor.
2130. The device of item 2089 wherein the agent is a fibrinogen antagonist selected from the group consisting of AUV-201 (Auvation), MG-13926 (Sanofi-Aventis), plasminogen activator (CAS No. 105913-11-9) (from Sanofi-Aventis or UCB), plasminogen activator-2 (tPA- 2) (Sanofi-Aventis), pro-urokinase (CAS No. 82657-92-9) (Sanofi-Aventis), and an analogue or derivative thereof. 2131. The device of item 2089 wherein the agent is a heat shock protein 90 antagonist selected from the group consisting of SRN-005 (Sirenade), geldanamycin, NSC-33050 (17-allylaminogeldanamycin; 17- AAG), ^-dimethylaminoethylamino-^-demethoxy-geldanamycin (17- DMAG), rifabutin (rifamycin XIV, 1\4-didehydro-1-deoxy-1,4-dihydro-5'-(2- methylpropyl)-1-oxo-), and an analogue or derivative thereof.
2132. The device of item 2089 wherein the agent is a histone deacetylase inhibitor.
2133. The device of item 2089 wherein the agent is an HMGCoA reductase inhibitor selected from the group consisting of an atherosclerosis therapeutic from Lipid Sciences, ATI-16000 (ARYx Therapeutics), KS-01-019 (Kos Pharmaceuticals), Pharmaprojects No. 2197 (Sanofi-Aventi), RP 61969 (Sanofi-Aventis), and an analogue or derivative thereof. . _
2134. The device of item 2089 wherein the agent is an ICAM inhibitor.
2135. The device of item 2089 wherein the agent is an IL, ICE and IRAK antagonist, wherein the antagonist is a CJ-14877, CP-424174 (Pfizer), NF-61 (Negma-Lerads), and an analogue or derivative thereof.
2136. The device of item 2089 wherein the agent is an IL-2 inhibitor.
2137. The device of item 2089 wherein the agent is an immunosuppressant selected from the group consisting of teriflunomide (Sanofi Aventis), chlorsulfaquinoxalone (NSC-339004), chlorsulfaquinoxalone sulfate, CS-712 (Sankyo), ismomultin alfa (CAS No. 457913-93-8) (Akzo Nobel), antiallergics from GenPat77, antiinflammatories or AT-005 (Androclus Therapeutics), autoimmune disease therapeutics from EpiVax, BN-007 (Bone), budesonide (CAS No. 51333-22- 3) (MAP Pharmaceuticals), CO-14 (Genzyme), edratide (CAS No. 433922- 67-9) (Teva), EP-314 (Enanta), eprovafen (CAS No. 101335-99-3) (Sanofi- Aventis), HWA-131 (CAS No. 118788-41-3) (Sanofi-Aventis), immunomodulators from MerLion Pharmaceuticals, immunosuppressives from Alchemia, IPL-12 (Inflazyme), MDL-9563 (CAS No. 27086-86-8) (Sanofi-Aventis), Pharmaprojects No. 2330 (Sanofi-Aventis), Pharmaprojects No. 6426 (Abgenix), PXS-25 (Pharmaxis), rosmarinic acid (CAS No. 20283-92-5) (Sanofi-Aventis), RP 42927 or RP 54745 (CAS No. 135330-08-4) (Sanofi-Aventis), SGN-35 (Seattle Genetics), ST-1959 (Sigma-Tau), type I diabetes therapy from SYNX Pharma, UNIL-88 (Debiopharm), VP-025 (Vasogen), VR-694 (Vectura), PRTX-001 (Protalex), and an analogue or derivative thereof.
2138. The device of item 2089 wherein the agent is an IMPDH (inosine monophosphate).
2139. The device of item 2089 wherein the agent is an integrin antagonist.
2140. The device of item 2089 wherein the agent is an interleukin antagonist.
2141. The device of item 2089 wherein the agent is an inhibitor of type III receptor tyrosine kinase.
2142. The device of item 2089 wherein the agent is an irreversible inhibitor of enzyme methionine aminopeptidase type 2. 2143. The device of item 2089 wherein the agent is an isozyme selective delta protein kinase C inhibitor.
2144. The device of item 2089 wherein the agent a JAK3 enzyme inhibitor.
2145. The device of item 2089 wherein the agent is a JNK inhibitor.
2146. The device of item 2089 wherein the agent is a kinase inhibitor.
2147. The device of item 2089 wherein the agent is kinesin antagonist.
2148. The device of item .2089 wherein the agent is a kinesin antagonist.
2149. The device of item 2089 wherein the agent is a leukotriene inhibitor and antagonist selected from the group consisting of ambicromil (CAS No. 58805-38-2) (Sanofi-Aventis), amelubant (CAS No. 346735-24-8) (Boehringer Ingelheim), DW-1141 (Dong Wha), ebselen (Daiichi Pharmaceutical), ibudilast (Kyorin), leucotriene inhibitors from Sanofi-Aventis, lymphotoxin-beta receptor (LT-D) from Biogen Idee, Pharmaprojects No. 1535 and 2728 (CAS No. 119340-33-9) (Sanofi- Aventis), R-112 (Rigel), Rev-5367 (CAS No. 92532-05-3) (Sanofi-Aventis), RG-14893 (CAS No. 141835-49-6) (Sanofi-Aventis), RG-5901-A (CAS No. 101910-24-1), 92532-23-5, RP 66153 (CAS No. 142422-79-5), RP 66364 (CAS No.186916-92-5), RP 69698 (CAS No. 141748-00-7) (Sanofi-Aventis), SC-411930 (Pfizer), SC-41930 (CAS No. 120072-59-5) (Pfizer), SC-50605 (CAS No. 138828-39-4) (Pfizer), SC-51146 (CAS No. 141059-52-1), SC- 53228 (CAS No. 153633-01-3) (Pfizer), spaglumic acid (ZY-15106) (CAS No.3106-85-2), 80619-64-3 (Novartis), tipredane (CAS No. 85197-77-9) (Bristol-Myers Squibb), U-75302 (CAS No. 119477-85-9) (Pfizer), and analogue or derivative thereof.
2150. The device of item 2089 wherein the agent is an MAP kinase inhibitor.
2151. The device of item 2089 wherein the agent is a matrix metalloproteinase inhibitor.
2152. The device of item 2089 wherein the agent is an MCP- CCR2 inhibitor.
2153. The device of item 2089 wherein the agent is an mTOR inhibitor.
2154. The device of item 2089 wherein the agent is an mTOR kinase inhibitor.
2155. The device of item 2089 wherein the agent is a microtubule inhibitor selected from the group consisting of antibody- maytansinoid conjugates from Biogen Idee, colchicines (MantiCore Pharmaceuticals), anticancer immunoconjugates from Johnson & Johnson, DIME from Octamer, gni-1f (GNI), huC242-DM4, huMy9-6-DM1 (ImmunoGen), IDN-5404 (Indena), IMO-098, IMOderm (Imotep), mebendazole (Introgen Therapeutics), microtubule poisons from Cambridge Enterprise, paclitaxel such as LOTAX from Aphios (CAS No. 33069-62-4), Genexol-PM from Samyang, Pharmaprojects No. 6383 (Tapestry Pharmaceuticals), RPR-112378 (Sanofi-Aventis), SGN-75 (Seattle Genetics), SPL-7435 (Starpharma), SSR-250411 (Sanofi-Aventis), trastuzumab-DM1 (Genentech), vinorelbine, and an analogue or derivative thereof.
2156. The device of item 2089 wherein the agent is an MIF inhibitor.
2157. The device of item 2089 wherein the agent is an MMP inhibitor.
2158. The device of item 2089 wherein the agent is a neurokinin (NK) antagonist selected from the group consisting of anthrotainin (CAS No. 148084-40-6) (Sanofi-Aventis), an IBS thereapeutic from ArQuIe, MDL-105212A (CAS No. 167261-60-1) (Ssanofi-Aventis), Pharmaprojects No. 2744, 3258 (CAS No. 139167-47-8) 4006, 4201 , or 5986 (Sanofi-Aventis), RP 67580 (CAS No. 135911-02-3), SR-144190 (CAS No. 201152-86-5), SSR:240600, SSR-241586 (Sanofi-Aventis), TKA-457 . (Novartis), vestipitant mesylate (CAS No. 334476-64-1) (GlaxoSmithKline), Win-64821 (Sanofi-Aventis), PRX-96026 (Predix Pharmaceuticals), and an analogue or derivative thereof.
2159. The device of item 2089 wherein the agent is an NF kappa B inhibitor selected from the group consisting of AVE-0545 or AVE- 0547 (Sanofi-Aventis), bortezomib (CAS No. 179324-69-7) (Millennium Pharmaceuticals), dexanabinol (CAS No. 112924-45-5) (Pharmos), dexlipotam (Viatris), Pharmaprojects No. 6283 (INDRA) (OXiGENE), IPL- 576092 (CAS No. 137571-30-3) (Inflazyme), NFKB decoy (Corgentech), NFKB decoy oligo (AnGes MG), NFKB's from Ariad, osteoporosis treatments or S5 (F005) from Fulcrum Pharmaceuticals, P61 (Phytopharm), R-flurbiprofen (CAS No. 5104-49-4) (Encore Pharmaceuticals), and an analogue or derivative thereof. 2160. The device of item 2089 wherein the agent is a nitric oxide agonist.
2161. The device of item 2089 wherein the agent is an ornithine decarboxylase inhibitor.
2162. The device of item 2089 wherein the agent is a p38 MAP kinase inhibitor selected from the group consisting of AZD-6703 (AstraZeneca), JX-401 (Jexys Pharmaceuticals), BMS-2 (Bristol-Myers Squibb), a p38 MAP kinase inhibitor from Novartis, a p38-alpha MAP kinase inhibitor from Amphora, Pharmaprojects No. 5704 (Pharmacopeia), RPR- 200765A (Sanofi-Aventis), SD-282 (Johnson & Johnson), TAK-715 (Takeda), and an analogue or derivative thereof.
2163. The device of item 2089 wherein the agent is a palmitoyl-protein thioesterase inhibitor. .._ . -
2164. The device of item 2089 wherein the agent is a PDGF receptor kinase inhibitor selected from the group consisting of AAL-993, AMN-107, or ABP-309 (Novartis), AMG-706 (Amgen), BAY-57-9352 (Bayer), CDP-860 (UCB), E-7080 (Eisai), imatinib (CAS No. 152459-95-5) (Novartis), OSI-930 (OSI Pharmaceuticals), RPR-127963E (Sanofi-Aventis), RWJ-540973 (Johnson & Johnson), sorafenib tosylate (Bayer), SU-11657 (Pfizer), tandutinib (CAS No. 387867-13-2) (Millennium Pharmaceuticals), vatalanib (Novartis), ZK-CDK (Schering AG), and an analogue or derivative thereof.
2165. The device of item 2089 wherein the agent is (-)- halofenate (Metabolex), AMG-131 (Amgen), antidiabetics from Japan Tobacco, AZD-4619, AZD-8450, AZD-8677 (AstraZeneca), DRF-10945, balaglitazone (Dr Reddy's), CS-00088, CS-00098 (Chipscreen Biosciences), E-3030 (Eisai), etalocib (CAS No. 161172-51-6) (EIi Lilly), GSK-641597 (Ligand), GSK-677954 (GlaxoSmithKline), GW-409544 (Ligand), GW- 590735 (GlaxoSmithKline), K-111 (Hoffmann-La Roche), LY-518674 (EIi Lilly), LY-674 (Ligand), LY-929 (Ligand), MC-3001 , MC-3002 (MaxoCore Pharmaceuticals), metformin HCI + pioglitazone (CAS No. 1115-70-4 and 112529-15-4), ACTOPLUS MET from Andrx), muraglitazar (CAS No. 331741-94-7) (Bristol-Myers Squibb), naveglitazar (Ligand), oleoylethanolamide (Kadmus Pharmaceuticals), ONO-5129, pioglitazone hydrochloride (CAS No. 111025-46-8 and 112529-15-4) (Takeda), PLX-204 (Plexxikon), PPAR agonists from Genfit, PPAR delta agonists from EIi Lilly, PPAR-alpha agonists from CrystalGenomics, PPAR-gamma modulators and PPAR-β modulators from CareX, rosiglitazone maleate (CAS No. 122324- 73-4 or 155141-29-0) (GlaxoSmithKline), rosiglitazone maleate/glimepir (CAS No. 155141-29-0 and 93479-97-1), AVANDARYL, rosiglitazone maleate/metformin extend (CAS No. 155141-29-0 and 657-24-9), AVANDAMET, rosiglitazone maleate+metformin, AVANDAMET (GlaxoSmithKline), tesaglitazar (AstraZeneca), and an analogue or derivative thereof.
2166. The device of item 2089 wherein the agent is a phosphatase inhibitor.
2167. The device of item 2089 wherein the agent is a phosphodiesterase (PDE) inhibitor selected from the group consisting of avanafil (Tanabe Seiyaku), dasantafil (CAS No. 569351-91-3) (Schering- Plough), A-906119 (CAS No. 134072-58-5), DL-850 (Sanofi-Aventis), GRC- 3015, GRC-3566, GRC-3886 (Glenmark), HWA-153 (CAS No. 56395-66-5) (Sanofi-Aventis), hydroxypumafentrine (Altana), IBFB-130011 , IBFB-14-016, IBFB-140301 , IBFB-150007, IBFB-211913 (IBFB Pharma), L-826141 (Merck & Co), medorinone (CAS No. 88296-61-1) (Sanofi-Aventis), MEM- 1917 (Memory Pharmaceuticals), ND-1251 (Neuro3d), PDE inhibitors from ICOS, PDE IV inhibitors from Memory Pharmaceuticals and CrystalGenomics, Pharmaprojects No. 2742 and 6141 (Sanofi-Aventis), QAD-171 (Novartis), RHC-2963 (CAS No. 76993-12-9 and 76993-14-1), RPR-117658, RPR-122818 derivatives, SR-24870 , and RPR-132294 (Sanofi-Aventis), SK-350 (ln2Gen), stroke therapy agents from deCODE Genetics, TAS-203 (Taiho), tofimilast (CAS No. 185954-27-2) (Pfizer), UK- 371800 (Pfizer), WIN-65579 (CAS No. 158020-82-7) (Sanofi-Aventis), IBFB- 130020 (IBFB Pharma), OPC-6535 (CAS No. 145739-56-6) (Otsuka), a phosphodiesterase III inhibitor, enoximone, a phosphodiesterase IV inhibitor, fosfosal, Atopik (Barrier Therapeutics), triflusal, a phosphodiesterase V inhibitor, and an analogue or derivative thereof.
2168. The device of item 2089 wherein the agent is a PKC inhibitor.
2169. The device of item 2089 wherein the agent is a platelet activating factor antagonist.
2170. The device of item 2089 wherein the agent is a platelet- derived growth factor receptor kinase inhibitor.
2171. The device of item 2089 wherein the agent is a prolyl hydroxylase inhibitor.
2172. The device of item 2089 wherein the agent is a polymorphonuclear neutrophil inhibitor.
2173. The device of item 2089 wherein the agent is a protein kinase B inhibitor. 2174. The device of item 2089 wherein the agent is a protein kinase C stimulant.
2175. The device of item 2089 wherein the agent is a purine nucleoside analogue.
2176. The device of item 2089 wherein the agent is a purinoreceptor P2X antagonist.
2177. The device of item 2089 wherein the agent is a Raf kinase inhibitor.
2178. The device of item 2089 wherein the agent is a reversible inhibitor of ErbB1 and ErbB2.
2179. The device of item 2089 wherein the agent is a - . ribonucleoside triphosphate reductase inhibitor.
2180. The device of item 2089 wherein the agent is an SDF-1 antagonist.
2181. The device of item 2089 wherein the agent is a sheddase inhibitor.
2182. The device of item 2089 wherein the agent is an SRC inhibitor.
2183. The device of item 2089 wherein the agent is a stromelysin inhibitor. 2184. The device of item 2089 wherein the agent is an Syk kinase inhibitor.
2185. The device of item 2089 wherein the agent is a telomerase inhibitor.
2186. The device of item 2089 wherein the agent is a TGF beta inhibitor selected from the group consisting of pirfenidone (CAS No. 53179-13-8) (MARNAC), tranilast (CAS No. 53902-12-8) (Kissei), IN-1130 (ln2Gen), mannose-6-phosphate (BTG), TGF-β antagonists from Inflazyme (Pharmaprojects No. 6075), TGF-β antagonists from Sydney, non-industrial source), TGF-β I receptor kinase inhibitors from EIi Lilly, TGF-β receptor inhibitors from Johnson & Johnson, and an analogue or derivative thereof.
2187. The device of item 2089 wherein the agent is a TNFα antagonist or TACE inhibitor selected from the group consisting of adalimumab (CAS No. 331731-18-1) (Cambridge Antibody Technology), AGIX-4207 (AtheroGenics), AGT-1 (Advanced Biotherapy), an antiinflammatory from Borean Pharma, Cellzome, or Paradigm Therapeutics, anti-inflammatory vaccine (TNF-alpha kinoid) from Neovacs, humanized anti-TNF antibody or an anti-TNF MAb (CB0006) Celltech (UCB), apratastat (CAS No. 287445-51-0) (Wyeth), BMS-561392 (Bristol-Myers Squibb), BN- 006 (Bone), certolizumab pegol (CAS No. 428863-50-7 or CH-138 (UCB), cilomilast (CAS No. 153259-65-5) (GlaxoSmithKline), CR-1 (Nuada Pharmaceuticals), CRx-119 (CombinatoRx), D-5410 (UCB), dacopafant (CAS No. 125372-33-0) (Sanofi-Aventis), dersalazine (CAS No. 188913-57- 7/188913-58-8) (Uriach), etanercept (CAS No. 185243-69-0) (Amgen), ethyl pyruvate (Critical (Critical Therapeutics), golimumab (CAS No. 476181-74-5) (Johnson & Johnson), hormono-immunotherapy from Ipsen, CDP571 (e.g., Humicade from UCB), IC-485 (ICOS), infliximab (CAS No. 170277-31-3) (Johnson & Johnson), IP-751 (Manhattan Pharmaceuticals), ISIS-104838 (CAS No. 250755-32-9) (ISIS Pharmaceuticals), lenalidomide (CAS No. 191732-72-6) (Celgene), lentinan (CAS No. 37339-90-5) (Ajinomoto), MDL- 201112 (CAS No. 142130-73-2) (Sanofi-Aventis), medroxyprogesterone (CAS No. 520-85-4) (InKine Pharmaceutical), N-acetylcysteine (CAS No. 616-91-1) (Zambon), NBE-P2 (DIREVO Biotech), nerelimomab (CAS No. 162774-06-3) (Chiron), OM-294DP (OM PHARMA), onercept (CAS No. 199685-57-9) (Yeda), PASSTNF-alpha (Verigen), pentoxifylline or oxypentifylline (Sanofi-Aventis), Pharmaprojects No. 4091 , 4241, 4295, or 4488 (Sanofi-Aventis), Pharmaprojects No. 5480 (Amgen), Pharmaprojects No. 6749 (Cengent), pirfenidone (CAS No. 53179-13-8) (MARNAC), RPR- 132294 (Sanofi-Aventis), S5 (F002) (Fulcrum Pharmaceuticals), simvastatin (CAS No. 79902-63-9) (Merck & Co), STA-6292 (Synta Pharmaceuticals), tacrolimus (CAS No. 104987-11-3) (Fujisawa LifeCycle Pharma), talactoferrin alfa (CAS No. 308240-58-6) (Agennix), thalidomide (CAS No. 50-35-1) (Celgene), TNF antagonists form ProStrakan, and Synergen, TNF inhibitors (Amgen), TNF-alpha antagonists from Dynavax Technologies and Jerina AG (Germany), TNF-alpha inhibitors from IBFB Pharma and Xencor (Xencor), torbafylline (CAS No. 105102-21-4) (Sanofi-Aventis), UR-1505 (Uriach), VT-346 (Viron Therapeutics), YSIL6 (Y's Therapeutics), YSTH2 (Y's Therapeutics), NPI-1302a-3 (Nereus Pharmaceuticals, a TNF antagonist from Jerina AG (Germany), dersalazine, and an analogue or derivative thereof.
2188. The device of item 2089 wherein the agent is a Toll receptor inhibitor.
2189. The device of item 2089 wherein the agent is a tubulin antagonist.
2190. The device of item 2089 wherein the agent is a tyrosine kinase inhibitor selected from the group consisting of SU-011248, SUTENT from Pfizer Inc. (New York, NY), BMS-354825, PN-355 (Paracelsian Pharmaceuticals), AGN-199659 (Allergan), AAL-993 or ABP-309 (Novartis), adaphostin (NIH), AEE-788 (Novartis), AG-013736 (OSI Pharmaceuticals), AG-13736 (Pfizer), ALT-110 (Alteris Therapeutics), AMG-706 (Amgen), anticancer MAbs from Xencor, anti-EGFrvlll MAbs from Abgenix, anti-HER2 MAb from Abiogen, AZD-2171 or AZD-9935 (AstraZeneca), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), CEP-5214 (Cephalon), CEP- 7055 (Cephalon), cetuximab (ImClone Systems), CHIR-200131 and CHIR- 258 (Chiron), CP-547632 (OSI Pharmaceuticals), CP-724714 (Pfizer), CT- 301 (Creabilis Therapeutics), D-69491 (Baxter International), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EGFR/ErbB2 inhibitors from Array BioPharma, erlotinib (CAS No. 183319-69-9) (OSI Pharmaceuticals), EXEL-2880 (Exelixis), FK-778 (Sanofi-Aventis) , gefitinib (CAS No. 184475-35-2) (AstraZeneca), GW-2286 or GW-654652 (GlaxoSmithKline), her2/neu antigen from AlphaVax, HER-2/neu inhibitor from Generex, Herzyme (Medipad) (Sirna Therapeutics), HKI-272 (Wyeth), HuMax-EGFr (Genmab), idronoxil (CAS No. 81267-65-4) (Novogen), IGF-1 inhibitors from Ontogen, IMC-11 F8 (ImClone Systems), kahalalide F (CAS No. 149204-42-2) (PharmaMar), KDR inhibitor from LG Life Sciences, KDR inhibitors from Abbott Laboratories, KDR kinase inhibitors (UCB), Kdr kinase inhibitors from Merck & Co, KRN-633 and KRN-951 (Kirin Brewery), KSB-102 (Xenova), lapatinib ditosylate (CAS No. 388082-78-8) (GlaxoSmithKline), matuzumab (Merck KGaA), MDX-214 (Medarex), ME-103 (Pharmexa), MED-A300 (Gerolymatos), MNAC-13 (Lay Line Genomics), nimotuzumab (Center of Molecular Immunology), NSC-330507 or NSC-707545 (NIH), NV-50 (Novogen), OSI-930 (OSI Pharmaceuticals), panitumumab (Abgenix), pelitinib (CAS No. 287933-82-7) (Wyeth), pertuzumab (CAS No. 380610-27- 5) (Genentech), Pharmaprojects No. 3985 (Sanofi-Aventis), prostate cancer therapeutics from Sequenom (SQPC35, SQPC36, SQPC90), removab and remoxab (Trion Pharma), RG-13022 (CAS No. 136831-48-6), RG-13291 (CAS No. 138989-50-1), or RG-14620 (CAS No. 136831-49-7) (Sanofi- Aventis), RM-6427 (Romark), RNAi breast cancer therapy from Benitec, RP 53801 (CAS No. 125882-88-4) (Sanofi-Aventis), sorafenib tosylate (Bayer), SU-11657 (Pfizer), Tie-2 antagonists from Semaia (Hybrigenics), Tie-2 inhibitors from Ontogen, trastuzumab (CAS No. 180288-69-1) (Genentech), tyrosine kinase inhibitors from Sanofi-Aventis, U3-1287, U3-1565, U3-1784, U3-1800 (U3 Pharma), vatalanib (Novartis), VEGFR-2 kinase inhibitor from Bristol-Myers Squibb, XL-647 (Exelixis), ZD-6474 (AstraZeneca), ZK-CDK (Schering AG), an EGFR tyrosine kinase inhibitor, EKB-569 (Wyeth), and an analogue or derivative thereof.
2191. The device of item 2089 wherein the agent is a VEGF inhibitor.
2192. The device of item 2089 wherein the agent is a vitamin D receptor agonist.
2193. The device of item 2089 wherein the agent is ZD-6474 (an angiogenesis inhibitor).
2194. The device of item 2089 wherein the agent is AP-23573 (an mTOR inhibitor).
2195. The device of item 2089 wherein the agent is synthadotin (a tubulin antagonist).
2196. The device of item 2089 wherein the agent is S-0885 (a collagenase inhibitor).
2197. The device of item 2089 wherein the agent is aplidϊne (an elongation factor-1 alpha inhibitor). 2198. The device of item 2089 wherein the agent is ixabepilone (an epithilone).
2199. The device of item 2089 wherein the agent is IDN-5390 (an angiogenesis inhibitor).
2200. The device of item 2089 wherein the agent is SB- 2723005 (an angiogenesis inhibitor).
2201. The device of item 2089 wherein the agent is ABT-518 (an angiogenesis inhibitor).
2202. The device of item 2089 wherein the agent is combretastatin (an angiogenesis inhibitor).
2203. The device of item 2089 wherein the agent is anecortave acetate (an angiogenesis inhibitor).
2204. The device of item 2089 wherein the agent is SB- 715992 (a kinesin antagonist).
2205. The device of item 2089 wherein the agent is temsirolimus (an mTOR inhibitor).
2206. The device of item 2089 wherein the agent is adalimumab (a TNFα antagonist).
2207. The device of item 2089, further comprising a polymer. 2208. The device of item 2089 wherein the anti-scarring agent inhibits adhesion between the device and a host into which the device is implanted.
2209. The device of item 2089 wherein the device delivers the anti-scarring agent locally to tissue proximate to the device.
2210. The device of item 2089, further comprising a coating, wherein the coating comprises the anti-scarring agent.
2211. The device of item 2089, further comprising a coating, wherein the coating is disposed on a surface of the device.
2212. The device of item 2089, further comprising a coating, wherein the coating directly contacts the device.
2213. The device of item 2089, further comprising a coating, wherein the coating indirectly contacts the device.
2214. The device of item 2089, further comprising a coating, wherein the coating partially covers the device.
2215. The device of item 2089, further comprising a coating, wherein the coating completely covers the device.
2216. The device of item 2089, further comprising a coating, wherein the coating is a uniform coating.
2217. The device of item 2089, further comprising a coating, wherein the coating is a non-uniform coating. 2218. The device of item 2089, further comprising a coating, wherein the coating is a discontinuous coating.
2219. The device of item 2089, further comprising a coating, wherein the coating is a patterned coating.
2220. The device of item 2089, further comprising a coating, wherein the coating has a thickness of 100 Dm or less.
2221. The device of item 2089, further comprising a coating, wherein the coating has a thickness of 10 Dm or less.
2222. The device of item 2089, further comprising a coating, wherein the coating adheres to the surface of the device upon deployment of the device.
2223. The device of item 2089, further comprising a coating, wherein the coating is stable at room temperature for a period of 1 year.
2224. The device of item 2089, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 0.0001% to about 1% by weight.
2225. The device of item 2089, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 1% to about 10% by weight.
2226. The device of item 2089, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 10% to about 25% by weight. 2227. The device of item 2089, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 25% to about 70% by weight.
2228. The device of item 2089, further comprising a coating, wherein the coating further comprises a polymer.
2229. The device of item 2089, further comprising a first coating having a first composition and the second coating having a second composition.
2230. The device of item 2089, further comprising a first coating having a first composition and the second coating having a second composition, wherein the first composition and the second composition are different.
2231. The device of item 2089, further comprising a polymeric carrier, wherein the polymeric carrier comprises a copolymer.
2232. The device of item 2089, further comprising a polymeric carrier, wherein the polymeric carrier comprises a block copolymer.
2233. The device of item 2089, further comprising a polymeric carrier, wherein the polymeric carrier comprises a random copolymer.
2234. The device of item 2089, further comprising a polymeric carrier, wherein the polymeric carrier comprises a biodegradable polymer.
2235. The device of item 2089, further comprising a polymeric carrier, wherein the polymeric carrier comprises a non-biodegradable polymer. 2246. The device of item 2089, further comprising a polymeric carrier, wherein the polymeric carrier comprises a butadiene polymer.
2247. The device of item 2089, further comprising a polymeric carrier, wherein the polymeric carrier comprises a macromer.
2248. The device of item 2089, further comprising a polymeric carrier, wherein the polymeric carrier comprises a poly(ethylene glycol) polymer.
2249. The device of item 2089, further comprising a polymeric carrier, wherein the polymeric carrier comprises an amorphous polymer.
2250. The device of item 2089, further comprising a lubricious coating.
2251. The device of item 2089 wherein the anti-scarring agent is located within pores or holes of the device.
2252. The device of item 2089 wherein the anti-scarring agent is located within a channel, lumen, or divet of the device.
2253. The device of item 2089, further comprising a second pharmaceutically active agent.
2254. The device of item 2089, further comprising an antiinflammatory agent.
2255. The device of item 2089, further comprising an agent that inhibits infection.
1107 2236. The device of item 2089, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrophilic polymer.
2237. The device of item 2089, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrophobic polymer.
2238. The device of item 2089, further comprising a polymeric carrier, wherein the polymeric carrier comprises a polymer having hydrophilic domains.
2239. The device of item 2089, further comprising a polymeric carrier, wherein the polymeric carrier comprises a polymer having hydrophobic domains.
2240. The device of item 2089, further comprising a polymeric carrier, wherein the polymeric carrier comprises a non-conductive polymer.
2241. The device of item 2089, further comprising a polymeric carrier, wherein the polymeric carrier comprises an elastomer.
2242. The device of item 2089, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrogel.
2243. The device of item 2089, further comprising a polymeric carrier, wherein the polymeric carrier comprises a silicone polymer.
2244. The device of item 2089, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrocarbon polymer.
2245. The device of item 2089, further comprising a polymeric carrier, wherein the polymeric carrier comprises a styrene-derived polymer.
1106 2256. The device of item 2089, further comprising an agent that inhibits infection, wherein the agent is an anthracyciine.
2257. The device of item 2089, further comprising an agent that inhibits infection, wherein the agent is doxorubicin.
2258. The device of item 2089, further comprising an agent that inhibits infection, wherein the agent is mitoxantrone.
2259. The device of item 2089, further comprising an agent that inhibits infection, wherein the agent is a fluoropyrimidine.
2260. The device of item 2089, further comprising an agent that inhibits infection, wherein the agent is 5-fluorouracil (5-FU).
2261. The device of item 2089, further comprising an agent that inhibits infection, wherein the agent is a folic acid antagonist.
2262. The device of item 2089, further comprising an agent that inhibits infection, wherein the agent is methotrexate.
2263. The device of item 2089, further comprising an agent that inhibits infection, wherein the agent is a podophylotoxin.
2264. The device of item 2089, further comprising an agent that inhibits infection, wherein the agent is etoposide.
2265. The device of item 2089, further comprising an agent that inhibits infection, wherein the agent is a camptothecin. 2266. The device of item 2089, further comprising an agent that inhibits infection, wherein the agent is a hydroxyurea.
2267. The device of item 2089, further comprising an agent that inhibits infection, wherein the agent is a platinum complex.
2268. The device of item 2089, further comprising an agent that inhibits infection, wherein the agent is cisplatin.
2269. The device of item 2089, further comprising an antithrombotic agent.
2270. The device of item 2089, further comprising a visualization agent.
2271. The device of item 2089, further comprising a visualization agent, wherein the visualization agent is a radiopaque material, wherein the radiopaque material comprises a metal, a halogenated compound, or a barium containing compound.
2272. The device of item 2089, further comprising a visualization agent, wherein the visualization agent is a radiopaque material, wherein the radiopaque material comprises barium, tantalum, or technetium.
2273. The device of item 2089, further comprising a visualization agent, wherein the visualization agent is a MRI responsive material.
2274. The device of item 2089, further comprising a visualization agent, wherein the visualization agent comprises a gadolinium chelate. 2275. The device of item 2089, further comprising a visualization agent, wherein the visualization agent comprises iron, magnesium, manganese, copper, or chromium.
2276. The device of item 2089, further comprising a visualization agent, wherein the visualization agent comprises an iron oxide compound.
2277. The device of item 2089, further comprising a visualization agent, wherein the visualization agent comprises a dye, pigment, or colorant.
2278. The device of item 2089, further comprising an echogenic material.
2279. The device of item 2089, further comprising an echogenic material, wherein the echogenic material is in the form of a coating.
2280. The device of item 2089 wherein the device is sterile.
2281. The device of item 2089 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device.
2282. The device of item 2089 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is connective tissue. 2283. The device of item 2089 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is muscle tissue.
2284. The device of item 2089 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is nerve tissue.
2285. The device of item 2089 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is epithelium tissue.
2286. The device of item 2089 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from the time of deployment of the device to about 1 year.
2287. The device of item 2089 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from about 1 month to 6 months.
2288. The device of item 2089 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from about 1 - 90 days.
2289. The device of item 2089 wherein the anti-scarring agent is released in effective concentrations from the device at a constant rate.
2290. The device of item 2089 wherein the anti-scarring agent is released in effective concentrations from the device at an increasing rate. 2291. The device of item 2089 wherein the anti-scarring agent is released in effective concentrations from the device at a decreasing rate.
2292. The device of item 2089 wherein the anti-scarring agent is released in effective concentrations from the composition comprising the anti-scarring agent by diffusion over a period ranging from the time of deployment of the device to about 90 days.
2293. The device of item 2089 wherein the anti-scarring agent is released in effective concentrations from the composition comprising the anti-scarring agent by erosion of the composition over a period ranging from the time of deployment of the device to about 90 days.
2294. The device of item 2089 wherein the device comprises about 0.01 Dg to about 10 Dg of the anti-scarring agent.
2295. The device of item 2089 wherein the device comprises about 10 Dg to about 10 mg of the anti-scarring agent.
2296. The device of item 2089 wherein the device comprises about 10 mg to about 250 mg of the anti-scarring agent.
2297. The device of item 2089 wherein the device comprises about 250 mg to about 1000 mg of the anti-scarring agent.
2298. The device of item 2089 wherein the device comprises about 1000 mg to about 2500 mg of the anti-scarring agent.
2299. The device of item 2089 wherein a surface of the device comprises less than 0.01 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied. 2300. The device of item 2089 wherein a surface of the device comprises about 0.01 Dg to about 1 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
2301. The device of item 2089 wherein a surface of the device comprises about 1 Dg to about 10 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
2302. The device of item 2089 wherein a surface of the device comprises about 10 Dg to about 250 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
2303. The device of item 2089 wherein a surface of the device comprises about 250 Dg to about 1000 Dg of the anti-scarring agent of anti- scarring agent per mm2 of device surface to which the anti-scarring agent is applied. -
2304. The device of item 2089 wherein a surface of the device comprises about 1000 Dg to about 2500 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
2305. A method as in any one of items 2089-2304, wherein the device is an extravascular graft.
2306. A method as in any one of items 2089-2304, wherein the device is an intravascular graft.
2307. A method as in any one of items 2089-2304, wherein the device is a vascular graft adapted for replacing a blood vessel damaged by aneurysm. 2308. A method as in any one of items 2089-2304, wherein the device is a vascular graft adapted for replacing a blood vessel damaged by intimal hyperplasia.
2309. A method as in any one of items 2089-2304, wherein the device is a vascular graft adapted for replacing a blood vessel damaged by thrombosis.
2310. A method as in any one of items 2089-2304, wherein the device is a vascular graft adapted for providing access to blood vessel.
2311. A method as in any one of items 2089-2304, wherein the device is a vascular graft adapted for providing an alternative conduit for blood flow through a damaged or diseased area in a vein.
2312. A method as in any one of items 2089-2304, wherein the device is a vascular graft adapted for providing an alternative conduit for blood flow through a damaged or diseased area in an artery.
2313. A method as in any one of items 2089-2304, wherein the device is a synthetic bypass graft.
2314. A method as in any one of items 2089-2304, wherein the device is a femoral-popliteal bypass graft.
2315. A method as in any one of items 2089-2304, wherein the device is a femoral-femoral bypass graft.
2316. A method as in any one of items 2089-2304, wherein the device is an axillary-femoral bypass graft. 2317. A method as in any one of items 2089-2304, wherein the device is a vein graft.
2318. A method as in any one of items 2089-2304, wherein the device is a peripheral vein graft.
2319. A method as in any one of items 2089-2304, wherein the device is a coronary vein graft.
2320. A method as in any one of items 2089-2304, wherein the device is an internal mammary graft.
2321. A method as in any one of items 2089-2304, wherein the device is a bifurcated vascular graft.
2322. A method as in any one of items 2089-2304, wherein the device is an intraluminal graft.
2323. A method as in any one of items 2089-2304, wherein the device is a prosthetic vascular graft.
2324. A device, comprising a hemodialysis access device (e.g., an implant) and an anti-scarring agent or a composition comprising an anti-scarring agent, wherein the agent inhibits scarring between the device and a host into which the device is implanted.
2325. The device of item 2324 wherein the agent is an adensosine A2A receptor antagonist.
2326. The device of item 2324 wherein the agent is an AKT inhibitor. 2327. The device of item 2324 wherein the agent is an alpha 2 integrin antagonist, wherein the alpha 2 integrin antagonist is Pharmaprojects No. 5754 (Merck KgaA).
2328. The device of item 2324 wherein the agent an alpha 4 integrin antagonist.
2329. The device of item 2324 wherein the agent is an alpha 7 nicotinic receptor agonist.
2330. The device of item 2324 wherein the agent is an angiogenesis inhibitor selected from the group consisting of AG-12,958 (Pfizer), ATN-161 (Attention LLC), neovastat, an angiogenesis inhibitor from Jerina AG (Germany), NM-3 (Mercian), VGA-1155 (Taisho), FCE-26644 (Pfizer), FCE-26950 (Pfizer), FPMA (Meiji Daries), FR-111142 (Fujisawa), GGTI-298, GM-1306 (Ligand), GPA-1734 (Novartis), NNC-47-0011 (Novo Nordisk), herbamycin (Nippon Kayaku), lenalidomide (Celegene), IP-10 (NIH), ABT-828 (Abbott), KIN-841 (Tokushima University, Japan), SF-1126 (Semafore Pharmaceuticals), laminin technology (NIH), CHIR-258 (Chiron), NVP-AEW541 (Novartis), NVP-AEW541 (Novartis), Vt16907 (Alchemia), OXI-8007 (Oxigene), EG-3306 (Ark Therapeutics), Maspin (Arriva), ABT- 567 (Abbott), PPI-2458 (Praecis Pharmaceuticals), CC-5079, CC-4089 (Celgene), HIF-1 alpha inhibitors (Xenova), S-247 (Pfizer), AP-23573 (Ariad), AZD-9935 (Astra Zeneca), mebendazole (Introgen Therapeutics), MetAP-2 inhibitors (GlaxoSmithKline), AG-615 (Angiogene Pharmaceuticals), Tie-2 antagonists (Hybrigenics), NC-381 , CYC-381 , NC-169, NC-219, NC-383, NC-384, NC-407 (Lorus Therapeutics), ATN-224 (Attenuon), ON-01370 (Onconova), Vitronectin antagonists (Amgen), SDX-103 (Salmedix), Vitronectin antagonists (Shire), CHP (Riemser), TEK (Amgen), Anecortave acetate (Alcon), T46.2 (Matrix Therapeutics), HG-2 (Heptagen), TEM antagonists (Genzyme), Oxi-4500 (Oxigene), ATN-161 (Attenuon), WX-293 (Wilex), M-2025 (Metris Therapeutics), Alphastatin (BioActa), YH-16 (Yantai Rongchang), BIBF-1120 (Boehringer Ingelheim), BAY-57-9352 (Bayer), AS- 1404 (Cancer Research Technology), SC-77964 (Pfizer), glycomimetics (BioTie Therapies), TIE-2 Inhibitors (Ontogen), DIMI, Octamer (Octamer), ABR-215050 (Active Biotech), ABT-518 (Abbott), KDR inhibitors (Abbott), BSF-466895 (Abbott), SCH-221153 (Schering-Plough), DAC:antiangiogenic (ConjuChem), TFPI (EntreMed), AZD-2171 (Astra-Zenaca), CDC-394 (Celgene), LY290293 (EIi Lilly), IDN-5390 (Indena), Kdr Kinase Inhibitors (Merck), CT-113020, CT-116433, CT-116563, CT-31890, CT-32228) (Cell Therapeutics), A-299620 (Abbott), TWEAK Inhibitor (Amgen), VEGF modulators (Johnson and Johnson), Tum-N53, tumstatin (Genzyme), Thios- 1 , Thios-2 (Thios Pharmaceuticals), MV-6401 (Miravant Medical Technologies), Spisulosine (PharmaMar), CEP-7055 (Cephalon), AUV-201 (Auvation), LM-609 (EIi Lilly), SKF-106615 (AnorMED), Oglufanide disodium (Cytran), BW-114 (Pharminox), Calreticulin (NIH), WX-678 (Wilex), SD-7784 (Pfizer), WX-UK1 (Wilex), SH-268 (Schering AG), 2-Me-PGA (Celgene), S- 137 (Pfizer), ZD-6126 (Angiogene Pharmaceuticals), SG-292 (SignalGen), Benefin (Lane Labs), A6, A36 (Angstrom), SB-2723005 (GlaxoSmithKline), SC-7 (Cell Therapeutics), ZEN-014 (AEterna Zentaris), 2-methoxyestradiol (EntreMed), NK-130119 (Nippon Kayaku), CC-10004 (Celgene), AVE- 8062A (Ajinomoto), Tacedinaline (Pfizer), Actinonin (Tokyo Metropolitan Institute of Medical Science), Lenalidomide (Celgene), VGA-1155, BTO-956 (SRI International), ER-68203-00 (Eisai), CT-6685 (UCB), JKC-362 (Phoenix Pharmaceuticals), DMI-3798 (DMI Biosciences, Angiomate (Ipsen), ZD-6474 (AstraZeneca), CEP-5214 (Cephalon), Canstatin (Genzyme), NM-3 (Mercian), Oridigm (MediQuest Therapeutics), Exherin (Adherex), BLS-0597 (Boston Life Sciences), PTC-299 (PTC Therapeutics), NPI-2358 (Nereus Pharmaceuticals), CGP-79787 (Novartis), AEE-788 (Novartis), CKD-732 (Chong Kun Dang), CP-564959 (OSI Pharmaceuticals), CM-101 (CarboMed), CT-2584, CT3501 (Cell Therapeutics), combretastatin and analogues and derivatives thereof (Oxigene), Rebimastat (Bristol-Meyers Squibb), Dextrin 2-sulfate (ML Laboratories), Cilengitide (Merk KGaA), NSC-706704 (Pharminox), KRN- 951 (Kirin Brewery), Ukrain, NSC-631570 (Nowicky Pharma), Tecogalan sodium (Daiichi Pharmaceutical), Tz-93 (Tsumura), TBC-1635 (Encysive Pharmaceuticals), TAN-1120 (Takeda), Semaxanib (Pfizer), BDI-7800 (Biopharmacopae), SD-186, SD-983 (Bristol-Meyers Squibb), SB-223245 (GlaxoSmithKline), SC-236 (Pfizer), RWJ-590973 (Johnson and Johnson), ILX-1850 (Genzyme), SC-68488, S-836 (Pfizer), CG-55069-11 (CuraGen), Ki-23057 (Kirin Brewery), CCX-700 (Chemoentryx), Pegaptanib octasodium (Giled Sciences), ANGIOCOL (available from Biostratum Inc.), or an analogue or derivative thereof.
2331. The device of item 2324 wherein the agent is an apoptosis antagonist.
2332. The device of item 2324 wherein the agent is an apoptosis activator.
2333. The device of item 2324 wherein the agent is a beta 1 integrin antagonist.
2334. The device of item 2324 wherein the agent is a beta tubulin inhibitor.
2335. The device of item 2324 wherein the agent is a blocker of enzyme production in Hepatitis C.
2336. The device of item 2324 wherein the agent is a Bruton's tyrosine kinase inhibitor. 2337. The device of item 2324 wherein the agent is a calcineurin inhibitor.
2338. The device of item 2324 wherein the agent is a caspase 3 inhibitor.
2339. The device of item 2324 wherein the agent is a CC chemokine receptor antagonist.
2340. The device of item 2324 wherein the agent is a cell cycle inhibitor selected from the group consisting of SNS-595 (Sunesis), synthadotin, KRX-0403, and an analogue or derivative thereof.
2341. The device of item 2324 wherein the agent is a cathepsin B inhibitor.
2342. The device of item 2324 wherein the agent is a cathepsin K inhibitor, wherein the cathepsin K inhibitor is 462795 (GlaxoSmithKline), INPL-022-D6 (Amura Therapeutics), an analogue or derivative thereof).
2343. The device of item 2324 wherein the agent is a cathepsin L inhibitor.
2344. The device of item 2324 wherein the agent is a CD40 antagonist.
2345. The device of item 2324 wherein the agent is a chemokine receptor agonist. 2346. The device of item 2324 wherein the agent is a chymase inhibitor.
2347. The device of item 2324 wherein the agent is a collagenase antagonist.
2348. The device of item 2324 wherein the agent is a CXCR antagonist.
2349. The device of item 2324 wherein the agent is a cyclin dependent kinase inhibitor selected from the group consisting of a CDK-1 inhibitor, a CDK-2 inhibitor, a CDK- 4 inhibitor, a CDK-6 inhibitor, a CAK1 inhibitor from GPC Biotech and Bristol-Myers Squibb, RGB-286199 (GPC Biotech), an anticancer agent from Astex Technology, a CAK1 inhibitor from GPC Biotech, a CDK inhibitor from Sanofi-Aventis, a CDK1/CDK2 inhibitor from Amgen, a CDK2 inhibitor from SUGEN-2 (Pfizer), a hearing loss therapy agent (Sound Pharmaceuticals), PD-0332991 (Pfizer), RGB-286199 (GPC Biotech), Ro-0505124 (Hoffmann-La Roche), a Ser/Thr kinase inhibitor from Lilly (EIi Lilly), CVT-2584 (CAS No. 199986-75-9) (CV Therapeutics), and an analogue or derivative thereof.
2350. The device of item 2324 wherein the agent is a cyclooxygenase 1 inhibitor.
2351. The device of item 2324 wherein the agent is a DHFR inhibitor.
2352. The device of item 2324 wherein the agent is a dual integrin inhibitor. 2353. The device of item 2324 wherein the agent is an elastase inhibitor.
2354. The device of item 2324 wherein the agent is an elongation factor-1 alpha inhibitor.
2355. The device of item 2324 wherein the agent is an endothelial growth factor antagonist.
2356. The device of item 2324 wherein the agent is an endothelial growth factor receptor kinase inhibitor selected from the group consisting of sorafenib tosylate (Bayer), AAL-993 (Novartis), ABP-309 (Novartis), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), E- 7080 (Eisai), EG-3306 (Ark Therapeutics), EXEL-2880 (Exelixis), GW- 654652 (GlaxoSmithKline), a KDR inhibitor from LG Life Sciences, CT-6685 and CT-6729 (UCB), KRN-633 and KRN-951 (Kirin Brewery), OSI-930 (OSI Pharmaceuticals), SP-5.2 (Supratek Pharma), SU-11657 (Pfizer), a Tie-2 antagonist (Hybrigenics), a VEGFR-2 kinase inhibitor (Bristol-Myers Squibb), XL-647 (Exelixis), a KDR inhibitor from Abbott Laboratories, sorafenib tosylate, and an analogue or derivative thereof.
2357. The device of item 2324 wherein the agent is an endotoxin antagonist.
2358. The device of item 2324 wherein the agent is an epothilone and tubulin binder.
2359. The device of item 2324 wherein the agent is an estrogen receptor antagonist. 2360. The device of item 2324 wherein the agent is an FGF inhibitor.
2361. The device of item 2324 wherein the agent is a famexyl transferase inhibitor.
2362. The device of item 2324 wherein the agent is farnesyltransferase inhibitor selected from the group of A-197574 (Abbott), a farnesyltransferase inhibitor from Servier, FPTIII (Strathclyde Institute for Drug R), LB-42908 (LG Life Sciences), Pharmaprojects No. 5063 (Genzyme), Pharmaprojects No. 5597 (Ipsen), Yissum Project No. B-1055 (Yissum), and an analogue or derivative thereof.
2363. The device of item 2324 wherein the agent is an FLT-3 kinase inhibitor.
2364. The device of item 2324 wherein the agent is an FGF receptor kinase inhibitor.
2365. The device of item 2324 wherein the agent is a fibrinogen antagonist selected from the group consisting of AUV-201 (Auvation), MG-13926 (Sanofi-Aventis), plasminogen activator (CAS No. 105913-11-9) (from Sanofi-Aventis or UCB), plasminogen activator-2 (tPA- 2) (Sanofi-Aventis), pro-urokinase (CAS No. 82657-92-9) (Sanofi-Aventis), and an analogue or derivative thereof.
2366. The device of item 2324 wherein the agent is a heat shock protein 90 antagonist selected from the group consisting of SRN-005 (Sirenade), geldanamycin, NSC-33050 (17-allylaminogeldanamycin; 17- AAG), 17-dimethylaminoethylamino-17-demethoxy-geldanamycin (17- DMAG), rifabutin (rifamycin XIV, r,4-didehydro-1-deoxy-1 ,4-dihydro-5'-(2- methylpropyl)-1-oxo-), and an analogue or derivative thereof.
2367. The device of item 2324 wherein the agent is a histone deacetylase inhibitor.
2368. The device of item 2324 wherein the agent is an HMGCoA reductase inhibitor selected from the group consisting of an atherosclerosis therapeutic from Lipid Sciences, ATM 6000 (ARYx Therapeutics), KS-01-019 (Kos Pharmaceuticals), Pharmaprojects No. 2197 (Sanofi-Aventi), RP 61969 (Sanofi-Aventis), and an analogue or derivative thereof.
2369. The device of item 2324 wherein the agent is an ICAM inhibitor.
2370. The device of item 2324 wherein the agent is an IL, ICE and IRAK antagonist, wherein the antagonist is a CJ-14877, CP-424174 (Pfizer), NF-61 (Negma-Lerads), and an analogue or derivative thereof.
2371. The device of item 2324 wherein the agent is an IL-2 inhibitor.
2372. The device of item 2324 wherein the agent is an immunosuppressant selected from the group consisting of teriflunomide (Sanofi Aventis), chlorsulfaquinoxalone (NSC-339004), chlorsulfaquinoxalone sulfate, CS-712 (Sankyo), ismomultin alfa (CAS No. 457913-93-8) (Akzo Nobel), antiallergics from GenPat77, antiinflammatories or AT-005 (Androclus Therapeutics), autoimmune disease therapeutics from EpiVax, BN-007 (Bone), budesonide (CAS No. 51333-22- 3) (MAP Pharmaceuticals), CO-14 (Genzyme), edratide (CAS No. 433922- 67-9) (Teva), EP-314 (Enanta), eprovafen (CAS No. 101335-99-3) (Sanofi- Aventis), HWA-131 (CAS No. 118788-41-3) (Sanofi-Aventis), immunomodulators from MerLion Pharmaceuticals, immunosuppressives from Alchemia, IPL-12 (Inflazyme), MDL-9563 (CAS No. 27086-86-8) (Sanofi-Aventis), Pharmaprojects No. 2330 (Sanofi-Aventis), Pharmaprojects No. 6426 (Abgenix), PXS-25 (Pharmaxis), rosmarinic acid (CAS No. 20283-92-5) (Sanofi-Aventis), RP 42927 or RP 54745 (CAS No. 135330-08-4) (Sanofi-Aventis), SGN-35 (Seattle Genetics), ST-1959 (Sigma-Tau), type I diabetes therapy from SYNX Pharma, UNIL-88 (Debiopharm), VP-025 (Vasogen), VR-694 (Vectura), PRTX-001 (Protalex), and an analogue or derivative thereof.
2373. The device of item 2324 wherein the agent is an IMPDH (inosine monophosphate).
- - - 2374. The device of item 2324-wherein the agent is an integrin antagonist.
2375. The device of item 2324 wherein the agent is an interleukin antagonist.
2376. The device of item 2324 wherein the agent is an inhibitor of type III receptor tyrosine kinase.
2377. The device of item 2324 wherein the agent is an irreversible inhibitor of enzyme methionine aminopeptidase type 2.
2378. The device of item 2324 wherein the agent is an isozyme selective delta protein kinase C inhibitor. 2379. The device of item 2324 wherein the agent a JAK3 enzyme inhibitor.
2380. The device of item 2324 wherein the agent is a JNK inhibitor.
2381. The device of item 2324 wherein the agent is a kinase inhibitor.
2382. The device of item 2324 wherein the agent is kinesin antagonist.
2383. The device of item 2324 wherein the agent is a kinesin antagonist.
2384. The device of item 2324 wherein the agent is a leukotriene inhibitor and antagonist selected from the group consisting of ambicromil (CAS No. 58805-38-2) (Sanofi-Aventis), amelubant (CAS No. 346735-24-8) (Boehringer Ingelheim), DW-1141 (Dong Wha), ebselen (Daiichi Pharmaceutical), ibudilast (Kyorin), leucotriene inhibitors from Sanofi-Aventis, lymphotoxin-beta receptor (LT-D) from Biogen Idee, Pharmaprojects No. 1535 and 2728 (CAS No. 119340-33-9) (Sanofi- Aventis), R-112 (Rigel), Rev-5367 (CAS No. 92532-05-3) (Sanofi-Aventis), RG-14893 (CAS No. 141835-49-6) (Sanofi-Aventis), RG-5901-A (CAS No. 101910-24-1), 92532-23-5, RP 66153 (CAS No. 142422-79-5), RP 66364 (CAS No.186916-92-5), RP 69698 (CAS No. 141748-00-7) (Sanofi-Aventis), SC-411930 (Pfizer), SC-41930 (CAS No. 120072-59-5) (Pfizer), SC-50605 (CAS No. 138828-39-4) (Pfizer), SC-51146 (CAS No. 141059-52-1), SC- 53228 (CAS No. 153633-01-3) (Pfizer), spaglumic acid (ZY-15106) (CAS No.3106-85-2), 80619-64-3 (Novartis), tipredane (CAS No. 85197-77-9) (Bristol-Myers Squibb), U-75302 (CAS No. 119477-85-9) (Pfizer), and analogue or derivative thereof.
2385. The device of item 2324 wherein the agent is an MAP kinase inhibitor.
2386. The device of item 2324 wherein the agent is a matrix metalloproteinase inhibitor.
2387. The device of item 2324 wherein the agent is an MCP- CCR2 inhibitor.
2388. The device of item 2324 wherein the agent is an mTOR inhibitor.
2389: The device of item 2324 wherein the agent is an mTOR kinase inhibitor.
2390. The device of item 2324 wherein the agent is a microtubule inhibitor selected from the group consisting of antibody- maytansinoid conjugates from Biogen Idee, colchicines (MantiCore Pharmaceuticals), anticancer immunoconjugates from Johnson & Johnson, DIME from Octamer, gni-1f (GNI), huC242-DM4, huMy9-6-DM1 (ImmunoGen), IDN-5404 (Indena), lMO-098, IMOderm (Imotep), mebendazole (Introgen Therapeutics), microtubule poisons from Cambridge Enterprise, paclitaxel such as LOTAX from Aphios (CAS No. 33069-62-4), Genexol-PM from Samyang, Pharmaprojects No. 6383 (Tapestry Pharmaceuticals), RPR-112378 (Sanofi-Aventis), SGN-75 (Seattle Genetics), SPL-7435 (Starpharma), SSR-250411 (Sanofi-Aventis), trastuzumab-DM1 (Genentech), vinorelbine, and an analogue or derivative thereof. 2391. The device of item 2324 wherein the agent is an MIF inhibitor.
2392. The device of item 2324 wherein the agent is an MMP inhibitor.
2393. The device of item 2324 wherein the agent is a neurokinin (NK) antagonist selected from the group consisting of anthrotainin (CAS No. 148084-40-6) (Sanofi-Aventis), an IBS thereapeutic from ArQuIe1 MDL-105212A (CAS No. 167261-60-1) (Ssanofi-Aventis), Pharmaprojects No. 2744, 3258 (CAS No. 139167-47-8) 4006, 4201 , or 5986 (Sanofi-Aventis), RP 67580 (CAS No. 135911-02-3), SR-144190 (CAS No. 201152-86-5), SSR-240600, SSR-241586 (Sanofi-Aventis), TKA-457 (Novartis), vestipitant mesylate (CAS No. 334476-64-1) (GlaxoSmithKline), Win-64821 (Sanofi-Aventis), PRX-96026 (Predix Pharmaceuticals), and an analogue or derivative thereof.
2394. The device of item 2324 wherein the agent is an NF kappa B inhibitor selected from the group consisting of AVE-0545 or AVE- 0547 (Sanofi-Aventis), bortezomib (CAS No. 179324-69-7) (Millennium Pharmaceuticals), dexanabinol (CAS No. 112924-45-5) (Pharmos), dexlipotam (Viatris), Pharmaprojects No. 6283 (INDRA) (OXiGENE), IPL- 576092 (CAS No. 137571-30-3) (Inflazyme), NFKB decoy (Corgentech), NFKB decoy oligo (AnGes MG), NFKB's from Ariad, osteoporosis treatments or S5 (F005) from Fulcrum Pharmaceuticals, P61 (Phytopharm), R-flurbiprofen (CAS No. 5104-49-4) (Encore Pharmaceuticals), and an analogue or derivative thereof.
2395. The device of item 2324 wherein the agent is a nitric oxide agonist. 2396. The device of item 2324 wherein the agent is an ornithine decarboxylase inhibitor.
2397. The device of item 2324 wherein the agent is a p38 MAP kinase inhibitor selected from the group consisting of AZD-6703 (AstraZeneca), JX-401 (Jexys Pharmaceuticals), BMS-2 (Bristol-Myers Squibb), a p38 MAP kinase inhibitor from Novartis, a p38-alpha MAP kinase inhibitor from Amphora, Pharmaprojects No. 5704 (Pharmacopeia), RPR- 200765A (Sanofi-Aventis), SD-282 (Johnson & Johnson), TAK-715 (Takeda), and an analogue or derivative thereof.
2398. The device of item 2324 wherein the agent is a palmitoyl-protein thioesterase inhibitor.
2399. The device of item 2324 wherein the agent is a PDGF receptor kinase inhibitor selected from the group consisting of AAL-993, AMN-107, or ABP-309 (Novartis), AMG-706 (Amgen), BAY-57-9352 (Bayer), CDP-860 (UCB), E-7080 (Eisai), imatinib (CAS No. 152459-95-5) (Novartis), OSI-930 (OSI Pharmaceuticals), RPR-127963E (Sanofi-Aventis), RWJ-540973 (Johnson & Johnson), sorafenib tosylate (Bayer), SU-11657 (Pfizer), tandutinib (CAS No. 387867-13-2) (Millennium Pharmaceuticals), vatalanib (Novartis), ZK-CDK (Schering AG), and an analogue or derivative thereof.
2400. The device of item 2324 wherein the agent is (-)- halofenate (Metabolex), AMG-131 (Amgen), antidiabetics from Japan Tobacco, AZD-4619, AZD-8450, AZD-8677 (AstraZeneca), DRF-10945, balaglitazone (Dr Reddy's), CS-00088, CS-00098 (Chipscreen Biosciences), E-3030 (Eisai), etalocib (CAS No. 161172-51-6) (EIi Lilly), GSK-641597 (Ligand), GSK-677954 (GlaxoSmithKline), GW-409544 (Ligand), GW- 590735 (GlaxoSmithKline), K-111 (Hoffmann-La Roche), LY-518674 (EIi Lilly), LY-674 (Ligand), LY-929 (Ligand), MC-3001 , MC-3002 (MaxoCore Pharmaceuticals), metformin HCI + pioglitazone (CAS No. 1115-70-4 and 112529-15-4), ACTOPLUS MET from Andrx), muraglitazar (CAS No. 331741-94-7) (Bristol-Myers Squibb), naveglitazar (Ligand), oleoylethanolamide (Kadmus Pharmaceuticals), ONO-5129, pioglitazone hydrochloride (CAS No. 111025-46-8 and 112529-15-4) (Takeda), PLX-204 (Plexxikon), PPAR agonists from Genfit, PPAR delta agonists from EIi Lilly, PPAR-alpha agonists from CrystalGenomics, PPAR-gamma modulators and PPAR-β modulators from CareX, rosiglitazone maleate (CAS No. 122324- 73-4 or 155141-29-0) (GlaxoSmithKline), rosiglitazone maleate/glimepir (CAS No. 155141-29-0 and 93479-97-1), AVANDARYL, rosiglitazone maleate/metformin extend (CAS No. 155141-29-0 and 657-24-9), AVANDAMET, rosiglitazone maleate+metformin, AVANDAMET (GlaxoSmithKline), tesaglitazar (AstraZeneca), and an analogue or derivative thereof.
2401. The device of item 2324 wherein the agent is a phosphatase inhibitor.
2402. The device of item 2324 wherein the agent is a phosphodiesterase (PDE) inhibitor selected from the group consisting of avanafil (Tanabe Seiyaku), dasantafil (CAS No. 569351-91-3) (Schering- Plough), A-906119 (CAS No. 134072-58-5), DL-850 (Sanofi-Aventis), GRC- 3015, GRC-3566, GRC-3886 (Glenmark), HWA-153 (CAS No. 56395-66-5) (Sanofi-Aventis), hydroxypumafentrine (Altana), IBFB-130011 , IBFB-14-016, IBFB-140301 , IBFB-150007, IBFB-211913 (IBFB Pharma), L-826141 (Merck & Co), medorinone (CAS No. 88296-61-1) (Sanofi-Aventis), MEM- 1917 (Memory Pharmaceuticals), ND-1251 (Neuro3d), PDE inhibitors from ICOS, PDE IV inhibitors from Memory Pharmaceuticals and CrystalGenomics, Pharmaprojects No. 2742 and 6141 (Sanofi-Aventis), QAD-171 (Novartis), RHC-2963 (CAS No. 76993-12-9 and 76993-14-1), RPR-117658, RPR-122818 derivatives, SR-24870 , and RPR-132294 (Sanofi-Aventis), SK-350 (ln2Gen), stroke therapy agents from deCODE Genetics, TAS-203 (Taiho), tofimilast (CAS No. 185954-27-2) (Pfizer), UK- 371800 (Pfizer), WIN-65579 (CAS No. 158020-82-7) (Sanofi-Aventis), IBFB- 130020 (IBFB Pharma), OPC-6535 (CAS No. 145739-56-6) (Otsuka), a phosphodiesterase III inhibitor, enoximone, a phosphodiesterase IV inhibitor, fosfosal, Atopik (Barrier Therapeutics), triflusal, a phosphodiesterase V inhibitor, and an analogue or derivative thereof.
2403. The device of item 2324 wherein the agent is a PKC inhibitor.
2404. The device of item 2324 wherein the agent is a platelet activating factor antagonist.
- 2405. The device of item 2324 wherein the agent is a platelet- derived growth factor receptor kinase inhibitor.
2406. The device of item 2324 wherein the agent is a prolyl hydroxylase inhibitor.
2407. The device of item 2324 wherein the agent is a polymorphonuclear neutrophil inhibitor.
2408. The device of item 2324 wherein the agent is a protein kinase B inhibitor.
2409. The device of item 2324 wherein the agent is a protein kinase C stimulant. 2410. The device of item 2324 wherein the agent is a purine nucleoside analogue.
2411. The device of item 2324 wherein the agent is a p u ri no receptor P2X antagonist.
2412. The device of item 2324 wherein the agent is a Raf kinase inhibitor.
2413. The device of item 2324 wherein the agent is a reversible inhibitor of ErbB1 and ErbB2.
2414. The device of item 2324 wherein the agent is a ribonucleoside triphosphate reductase inhibitor.
2415. The device of item 2324 wherein the agent is an SDF-1 antagonist.
2416. The device of item 2324 wherein the agent is a sheddase inhibitor.
2417. The device of item 2324 wherein the agent is an SRC inhibitor.
2418. The device of item 2324 wherein the agent is a stromelysin inhibitor.
2419. The device of item 2324 wherein the agent is an Syk kinase inhibitor. 2420. The device of item 2324 wherein the agent is a telomerase inhibitor.
2421. The device of item 2324 wherein the agent is a TGF beta inhibitor selected from the group consisting of pirfenidone (CAS No. 53179-13-8) (MARNAC), tranilast (CAS No. 53902-12-8) (Kissei), IN-1130 (ln2Gen), mannose-6-phosphate (BTG), TGF-β antagonists from Inflazyme (Pharmaprojects No. 6075), TGF-β antagonists from Sydney, non-industrial source), TGF-β I receptor kinase inhibitors from EIi Lilly, TGF-β receptor inhibitors from Johnson & Johnson, and an analogue or derivative thereof.
2422. The device of item 2324 wherein the agent is a TNFα antagonist or TACE inhibitor selected from the group consisting of adalimumab (CAS No. 331731-18-1) (Cambridge Antibody Technology), AGIX-4207 (AtheroGenics), AGT-1 (Advanced Biotherapy), an antiinflammatory from Borean Pharma, Cellzome, or Paradigm Therapeutics, -- anti-inflammatory vaccine (TNF-alpha kinoid) from Neovacs, humanized anti-TNF antibody or an anti-TNF MAb (CB0006) Celltech (UCB), apratastat (CAS No. 287445-51-0) (Wyeth), BMS-561392 (Bristol-Myers Squibb), BN- 006 (Bone), certolizumab pegol (CAS No. 428863-50-7 or CH-138 (UCB), cilomilast (CAS No. 153259-65-5) (GlaxoSmithKline), CR-1 (Nuada Pharmaceuticals), CRx-119 (CombinatoRx), D-5410 (UCB), dacopafant (CAS No. 125372-33-0) (Sanofi-Aventis), dersalazine (CAS No. 188913-57- 7/188913-58-8) (Uriach), etanercept (CAS No. 185243-69-0) (Amgen), ethyl pyruvate (Critical (Critical Therapeutics), golimumab (CAS No. 476181-74-5) (Johnson & Johnson), hormono-immunotherapy from Ipsen, CDP571 (e.g., Humicade from UCB), IC-485 (ICOS), infliximab (CAS No. 170277-31-3) (Johnson & Johnson), IP-751 (Manhattan Pharmaceuticals), ISIS-104838 (CAS No. 250755-32-9) (ISIS Pharmaceuticals), lenalidomide (CAS No. 191732-72-6) (Celgene), lentinan (CAS No. 37339-90-5) (Ajinomoto), MDL- 201112 (CAS No. 142130-73-2) (Sanofi-Aventis), medroxyprogesterone (CAS No. 520-85-4) (InKine Pharmaceutical), N-acetylcysteine (CAS No. 616-91-1) (Zambon), NBE-P2 (DIREVO Biotech), nerelimomab (CAS No. 162774-06-3) (Chiron), OM-294DP (OM PHARMA), onercept (CAS No. 199685-57-9) (Yeda), PASSTNF-alpha (Verigen), pentoxifylline or oxypentifylline (Sanofi-Aventis), Pharmaprojects No. 4091 , 4241 , 4295, or 4488 (Sanofi-Aventis), Pharmaprojects No. 5480 (Amgen), Pharmaprojects No. 6749 (Cengent), pirfenidone (CAS No. 53179-13-8) (MARNAC), RPR- 132294 (Sanofi-Aventis), S5 (F002) (Fulcrum Pharmaceuticals), simvastatin (CAS No. 79902-63-9) (Merck & Co), STA-6292 (Synta Pharmaceuticals), tacrolimus (CAS No. 104987-11-3) (Fujisawa LifeCycle Pharma), talactoferrin alfa (CAS No. 308240-58-6) (Agennix), thalidomide (CAS No. 50-35-1) (Celgene), TNF antagonists form ProStrakan, and Synergen, TNF inhibitors (Amgen), TNF-alpha antagonists from Dynavax Technologies and Jerina AG (Germany), TNF-alpha inhibitors from IBFB Pharma and Xencor (Xencor), torbafylline (CAS No. 105102-21-4) (Sanofi-Aventis), UR-1505 (Uriach), VT-346 (Viron Therapeutics), YSIL6 (Y's Therapeutics), YSTH2 (Y's Therapeutics), NPI-1302a-3 (Nereus Pharmaceuticals, a TNF antagonist from Jerina AG (Germany), dersalazine, and an analogue or derivative thereof.
2423. The device of item 2324 wherein the agent is a Toll receptor inhibitor.
2424. The device of item 2324 wherein the agent is a tubulin antagonist.
2425. The device of item 2324 wherein the agent is a tyrosine kinase inhibitor selected from the group consisting of SU-011248, SUTENT from Pfizer Inc. (New York, NY), BMS-354825, PN-355 (Paracelsian Pharmaceuticals), AGN-199659 (Allergan), AAL-993 or ABP-309 (Novartis), adaphostin (NIH), AEE-788 (Novartis), AG-013736 (OSI Pharmaceuticals), AG-13736 (Pfizer), ALT-110 (Alteris Therapeutics), AMG-706 (Amgen), anticancer MAbs from Xencor, anti-EGFrvlll MAbs from Abgenix, anti-HER2 MAb from Abiogen, AZD-2171 or AZD-9935 (AstraZeneca), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), CEP-5214 (Cephalon), CEP- 7055 (Cephalon), cetuximab (ImCIone Systems), CHIR-200131 and CHIR- 258 (Chiron), CP-547632 (OSI Pharmaceuticals), CP-724714 (Pfizer), CT- 301 (Creabilis Therapeutics), D-69491 (Baxter International), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EGFR/ErbB2 inhibitors from Array BioPharma, erlotinib (CAS No. 183319-69-9) (OSI Pharmaceuticals), EXEL-2880 (Exelixis), FK-778 (Sanofi-Aventis) , gefitinib (CAS No. 184475-35-2) (AstraZeneca), GW-2286 or GW-654652 (GlaxoSmithKline), her2/neu antigen from AlphaVax, HER-2/neu inhibitor from Generex, Herzyme (Medipad) (Sirna Therapeutics), HKI-272 (Wyeth), HuMax-EGFr (Genmab), idronoxil (CAS No. 81267-65-4) (Novogen), IGF-1 inhibitors from Ontogen, IMC-11F8 (ImCIone Systems), kahalalide F (CAS No. 149204-42-2) (PharmaMar), KDR inhibitor from LG Life Sciences, KDR inhibitors from Abbott Laboratories, KDR kinase inhibitors (UCB), Kdr kinase inhibitors from Merck & Co, KRN-633 and KRN-951 (Kirin Brewery), KSB-102 (Xenova), lapatinib ditosylate (CAS No. 388082-78-8) (GlaxoSmithKline), matuzumab (Merck KGaA), MDX-214 (Medarex), ME-103 (Pharmexa), MED-A300 (Gerolymatos), MNAC-13 (Lay Line Genomics), nimotuzumab (Center of Molecular Immunology), NSC-330507 or NSC-707545 (NIH), NV-50 (Novogen), OSI-930 (OSI Pharmaceuticals), panitumumab (Abgenix), pelitinib (CAS No. 287933-82-7) (Wyeth), pertuzumab (CAS No. 380610-27- 5) (Genentech), Pharmaprojects No. 3985 (Sanofi-Aventis), prostate cancer therapeutics from Sequenom (SQPC35, SQPC36, SQPC90), removab and remoxab (Trion Pharma), RG-13022 (CAS No. 136831-48-6), RG-13291 (CAS No. 138989-50-1), or RG-14620 (CAS No. 136831-49-7) (Sanofi- Aventis), RM-6427 (Romark), RNAi breast cancer therapy from Benitec, RP 53801 (CAS No. 125882-88-4) (Sanofi-Aventis), sorafenib tosylate (Bayer), SU-11657 (Pfizer), Tie-2 antagonists from Semaia (Hybrigenics), Tie-2 inhibitors from Ontogen, trastuzumab (CAS No. 180288-69-1) (Genentech), tyrosine kinase inhibitors from Sanofi-Aventis, U3-1287, U3-1565, U3-1784, U3-1800 (U3 Pharma), vatalanib (Novartis), VEGFR-2 kinase inhibitor from Bristol-Myers Squibb, XL-647 (Exelixis), ZD-6474 (AstraZeneca), ZK-CDK (Schering AG), an EGFR tyrosine kinase inhibitor, EKB-569 (Wyeth), and an analogue or derivative thereof.
2426. The device of item 2324 wherein the agent is a VEGF inhibitor.
2427. The device of item 2324 wherein the agent is a vitamin D receptor agonist.
2428. The device of item 2324 wherein the agent is ZD-6474 (an angiogenesis inhibitor).
2429. The device of item 2324 wherein the agent is AP-23573 (an mTOR inhibitor).
2430. The device of item 2324 wherein the agent is synthadotin (a tubulin antagonist).
2431. The device of item 2324 wherein the agent is S-0885 (a collagenase inhibitor).
2432. The device of item 2324 wherein the agent is aplidine (an elongation factor-1 alpha inhibitor).
2433. The device of item 2324 wherein the agent is ixabepilone (an epithilone). 2434. The device of item 2324 wherein the agent is IDN-5390 (an angiogenesis inhibitor).
2435. The device of item 2324 wherein the agent is SB- 2723005 (an angiogenesis inhibitor).
2436. The device of item 2324 wherein the agent is ABT-518 (an angiogenesis inhibitor).
2437. The device of item 2324 wherein the agent is combretastatin (an angiogenesis inhibitor).
2438. The device of item 2324 wherein the agent is anecortave acetate (an angiogenesis inhibitor).
2439. The device of item 2324 wherein the agent is SB- 715992 (a kinesin antagonist).
2440. The device of item 2324 wherein the agent is temsirolimus (an mTOR inhibitor).
2441. The device of item 2324 wherein the agent is adalimumab (a TNFα antagonist).
2442. The device of item 2324, further comprising a polymer.
2443. The device of item 2324 wherein the anti-scarring agent inhibits adhesion between the device and a host into which the device is implanted. 2444. The device of item 2324 wherein the device delivers the anti-scarring agent locally to tissue proximate to the device.
2445. The device of item 2324, further comprising a coating, wherein the coating comprises the anti-scarring agent.
2446. The device of item 2324, further comprising a coating, wherein the coating is disposed on a surface of the device.
2447. The device of item 2324, further comprising a coating, wherein the coating directly contacts the device.
2448. The device of item 2324, further comprising a coating, wherein the coating indirectly contacts the device.
2449. The device of item 2324, further comprising a coating, wherein the coating partially covers the device.
2450. The device of item 2324, further comprising a coating, wherein the coating completely covers the device.
2451. The device of item 2324, further comprising a coating, wherein the coating is a uniform coating.
2452. The device of item 2324, further comprising a coating, wherein the coating is a non-uniform coating.
2453. The device of item 2324, further comprising a coating, wherein the coating is a discontinuous coating. 2454. The device of item 2324, further comprising a coating, wherein the coating is a patterned coating.
2455. The device of item 2324, further comprising a coating, wherein the coating has a thickness of 100 Dm or less.
2456. The device of item 2324, further comprising a coating, wherein the coating has a thickness of 10 Om or less.
2457. The device of item 2324, further comprising a coating, wherein the coating adheres to the surface of the device upon deployment of the device.
2458. The device of item 2324, further comprising a coating, wherein the coating is stable at room temperature for a period of 1 year.
2459. The device of item 2324, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 0.0001% to about 1% by weight.
2460. The device of item 2324, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 1 % to about 10% by weight.
2461. The device of item 2324, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 10% to about 25% by weight.
2462. The device of item 2324, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 25% to about 70% by weight. 2463. The device of item 2324, further comprising a coating, wherein the coating further comprises a polymer.
2464. The device of item 2324, further comprising a first coating having a first composition and the second coating having a second composition.
2465. The device of item 2324, further comprising a first coating having a first composition and the second coating having a second composition, wherein the first composition and the second composition are different.
2466. The device of item 2324, further comprising a polymeric carrier, wherein the polymeric carrier comprises a copolymer.
2467. The device of item 2324, further comprising a polymeric carrier, wherein the polymeric carrier comprises a block copolymer.
2468. The device of item 2324, further comprising a polymeric carrier, wherein the polymeric carrier comprises a random copolymer.
2469. The device of item 2324, further comprising a polymeric carrier, wherein the polymeric carrier comprises a biodegradable polymer.
2470. The device of item 2324, further comprising a polymeric carrier, wherein the polymeric carrier comprises a non-biodegradable polymer.
2471. The device of item 2324, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrophilic polymer. 2472. The device of item 2324, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrophobic polymer.
2473. The device of item 2324, further comprising a polymeric carrier, wherein the polymeric carrier comprises a polymer having hydrophilic domains.
2474. The device of item 2324, further comprising a polymeric carrier, wherein the polymeric carrier comprises a polymer having hydrophobic domains.
2475. The device of item 2324, further comprising a polymeric carrier, wherein the polymeric carrier comprises a non-conductive polymer.
2476. The device of item 2324, further comprising a polymeric jcarrier, wherein the polymeric. carrier comprises an elastomer.
2477. The device of item 2324, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrogel.
2478. The device of item 2324, further comprising a polymeric carrier, wherein the polymeric carrier comprises a silicone polymer.
2479. The device of item 2324, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrocarbon polymer.
2480. The device of item 2324, further comprising a polymeric carrier, wherein the polymeric carrier comprises a styrene-derived polymer.
2481. The device of item 2324, further comprising a polymeric carrier, wherein the polymeric carrier comprises a butadiene polymer. 2482. The device of item 2324, further comprising a polymeric carrier, wherein the polymeric carrier comprises a macromer.
2483. The device of item 2324, further comprising a polymeric carrier, wherein the polymeric carrier comprises a poly(ethylene glycol) polymer.
2484. The device of item 2324, further comprising a polymeric carrier, wherein the polymeric carrier comprises an amorphous polymer.
2485. The device of item 2324, further comprising a lubricious coating.
2486. The device of item 2324 wherein the anti-scarring agent is located within pores or holes of the device.
2487. The device of item 2324 wherein the anti-scarring agent is located within a channel, lumen, or divet of the device.
2488. The device of item 2324, further comprising a second pharmaceutically active agent.
2489. The device of item 2324, further comprising an antiinflammatory agent.
2490. The device of item 2324, further comprising an agent that inhibits infection.
2491. The device of item 2324, further comprising an agent that inhibits infection, wherein the agent is an anthracycline. 2492. The device of item 2324, further comprising an agent that inhibits infection, wherein the agent is doxorubicin.
2493. The device of item 2324, further comprising an agent that inhibits infection, wherein the agent is mitoxantrone.
2494. The device of item 2324, further comprising an agent that inhibits infection, wherein the agent is a fluoropyrimidine.
2495. The device of item 2324, further comprising an agent that inhibits infection, wherein the agent is 5-fluorouracil (5-FU).
2496. The device of item 2324, further comprising an agent that inhibits infection, wherein the agent is a folic acid antagonist.
2497. The device of item 2324, further comprising an agent that inhibits infection, wherein the agent is methotrexate.
2498. The device of item 2324, further comprising an agent that inhibits infection, wherein the agent is a podophylotoxin.
2499. The device of item 2324, further comprising an agent that inhibits infection, wherein the agent is etoposide.
2500. The device of item 2324, further comprising an agent that inhibits infection, wherein the agent is a camptothecin.
2501. The device of item 2324, further comprising an agent that inhibits infection, wherein the agent is a hydroxyurea. 2502. The device of item 2324, further comprising an agent that inhibits infection, wherein the agent is a platinum complex.
2503. The device of item 2324, further comprising an agent that inhibits infection, wherein the agent is cisplatin.
2504. The device of item 2324, further comprising an antithrombotic agent.
2505. The device of item 2324, further comprising a visualization agent.
2506. The device of item 2324, further comprising a visualization agent, wherein the visualization agent is a radiopaque material, wherein the radiopaque material comprises a metal, a halogenated compound, or a barium containing compound.
2507. The device of item 2324, further comprising a visualization agent, wherein the visualization agent is a radiopaque material, wherein the radiopaque material comprises barium, tantalum, or technetium.
2508. The device of item 2324, further comprising a visualization agent, wherein the visualization agent is a MRI responsive material.
2509. The device of item 2324, further comprising a visualization agent, wherein the visualization agent comprises a gadolinium chelate. 2510. The device of item 2324, further comprising a visualization agent, wherein the visualization agent comprises iron, magnesium, manganese, copper, or chromium.
2511. The device of item 2324, further comprising a visualization agent, wherein the visualization agent comprises an iron oxide compound.
2512. The device of item 2324, further comprising a visualization agent, wherein the visualization agent comprises a dye, pigment, or colorant.
2513. The device of item 2324, further comprising an echogenic material.
-2514. The device of item 2324, further comprising an echogenic material, wherein the echogenic material is in the form of a coating.
2515. The device of item 2324 wherein the device is sterile.
2516. The device of item 2324 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device.
2517. The device of item 2324 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is connective tissue. 2518. The device of item 2324 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is muscle tissue.
2519. The device of item 2324 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is nerve tissue.
2520. The device of item 2324 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is epithelium tissue.
2521. The device of item 2324 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from the time of deployment of the device to about 1 year.
2522. The device of item 2324 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from about 1 month to 6 months.
2523. The device of item 2324 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from about 1 - 90 days.
2524. The device of item 2324 wherein the anti-scarring agent is released in effective concentrations from the device at a constant rate.
2525. The device of item 2324 wherein the anti-scarring agent is released in effective concentrations from the device at an increasing rate. 2526. The device of item 2324 wherein the anti-scarring agent is released in effective concentrations from the device at a decreasing rate.
2527. The device of item 2324 wherein the anti-scarring agent is released in effective concentrations from the composition comprising the anti-scarring agent by diffusion over a period ranging from the time of deployment of the device to about 90 days.
2528. The device of item 2324 wherein the anti-scarring agent is released in effective concentrations from the composition comprising the anti-scarring agent by erosion of the composition over a period ranging from the time of deployment of the device to about 90 days.
2529. The device of item 2324 wherein the device comprises about 0.01 Dg to about 10 Dg of the anti-scarring agent.
2530. The device of item 2324 wherein the device comprises about 10 Dg to about 10 mg of the anti-scarring agent.
2531. The device of item 2324 wherein the device comprises about 10 mg to about 250 mg of the anti-scarring agent.
2532. The device of item 2324 wherein the device comprises about 250 mg to about 1000 mg of the anti-scarring agent.
2533. The device of item 2324 wherein the device comprises about 1000 mg to about 2500 mg of the anti-scarring agent.
2534. The device of item 2324 wherein a surface of the device comprises less than 0.01 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied. 2535. The device of item 2324 wherein a surface of the device comprises about 0.01 Dg to about 1 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
2536. The device of item 2324 wherein a surface of the device comprises about 1 Dg to about 10 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
2537. The device of item 2324 wherein a surface of the device comprises about 10 Dg to about 250 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
2538. The device of item 2324 wherein a surface of the device comprises about 250 Dg to about 1000 Dg of the anti-scarring agent of anti- scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
2539. The device of item 2324 wherein a surface of the device comprises about 1000 Dg to about 2500 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
2540. A method as in any one of items 2324-2539, wherein the device is an AV fistula graft.
2541. A method as in any one of items 2324-2539, wherein the device is an AV access graft.
2542. A method as in any one of items 2324-2539, wherein the device is a venous catheter. 2543. A method as in any one of items 2324-2539, wherein the device is a vascular graft.
2544. A method as in any one of items 2324-2539, wherein the device is an implantable port.
2545. A method as in any one of items 2324-2539, wherein the device is an AV shunt.
2546. A device, comprising a medical device comprising a film or a mesh (e.g., an implant) and an anti-scarring agent or a composition comprising an anti-scarring agent, wherein the agent inhibits scarring between the device and a host into which the device is implanted.
2547. The device of item 2546 wherein the agent is an adensosine A2A receptor antagonist,
2548. The device of item 2546 wherein the agent is an AKT inhibitor.
2549. The device of item 2546 wherein the agent is an alpha 2 integrin antagonist, wherein the alpha 2 integrin antagonist is Pharmaprojects No. 5754 (Merck KgaA).
2550. The device of item 2546 wherein the agent an alpha 4 integrin antagonist.
2551. The device of item 2546 wherein the agent is an alpha 7 nicotinic receptor agonist. 2552. The device of item 2546 wherein the agent is an angiogenesis inhibitor selected from the group consisting of AG-12,958 (Pfizer), ATN-161 (Attention LLC), neovastat, an angiogenesis inhibitor from Jerina AG (Germany), NM-3 (Mercian), VGA-1155 (Taisho), FCE-26644 (Pfizer), FCE-26950 (Pfizer), FPMA (Meiji Daries), FR-111142 (Fujisawa), GGTI-298, GM-1306 (Ligand), GPA-1734 (Novartis), NNC-47-0011 (Novo Nordisk), herbamycin (Nippon Kayaku), lenalidomide (Celegene), IP-10 (NIH), ABT-828 (Abbott), KIN-841 (Tokushima University, Japan), SF-1126 (Semafore Pharmaceuticals), laminin technology (NIH), CHIR-258 (Chiron), NVP-AEW541 (Novartis), NVP-AEW541 (Novartis), Vt16907 (Alchemia), OXI-8007 (Oxigene), EG-3306 (Ark Therapeutics), Maspin (Arriva), ABT- 567 (Abbott), PPI-2458 (Praecis Pharmaceuticals), CC-5079, CC-4089 (Celgene), HIF-1 alpha inhibitors (Xenova), S-247 (Pfizer), AP-23573 (Ariad), AZD-9935 (Astra Zeneca), mebendazole (Introgen Therapeutics), MetAP-2 inhibitors (GlaxoSmithKline), AG-615 (Angiogene Pharmaceuticals), Tie-2 antagonists (Hybrigenics), NC-381, CYC-381, NC-169, NCτ219, NC-383, NC-384, NC-407 (Lorus Therapeutics), ATN-224 (Attenuon), ON-01370 (Onconova), Vitronectin antagonists (Amgen), SDX-103 (Salmedix), Vitronectin antagonists (Shire), CHP (Riemser), TEK (Amgen), Anecortave acetate (Alcon), T46.2 (Matrix Therapeutics), HG-2 (Heptagen), TEM antagonists (Genzyme), Oxi-4500 (Oxigene), ATN-161 (Attenuon), WX-293 (Wilex), M-2025 (Metris Therapeutics), Alphastatin (BioActa), YH-16 (Yantai Rongchang), BIBF-1120 (Boehringer Ingelheim), BAY-57-9352 (Bayer), AS- 1404 (Cancer Research Technology), SC-77964 (Pfizer), glycomimetics (BioTie Therapies), TIE-2 Inhibitors (Ontogen), DIMI, Octamer (Octamer), ABR-215050 (Active Biotech), ABT-518 (Abbott), KDR inhibitors (Abbott), BSF-466895 (Abbott), SCH-221153 (Schering-Plough), DAC:antiangiogenic (ConjuChem), TFPI (EntreMed), AZD-2171 (Astra-Zenaca), CDC-394 (Celgene), LY290293 (EIi Lilly), IDN-5390 (Indena), Kdr Kinase Inhibitors (Merck), CT-113020, CT-116433, CT-116563, CT-31890, CT-32228) (Cell Therapeutics), A-299620 (Abbott), TWEAK Inhibitor (Amgen), VEGF modulators (Johnson and Johnson), Tum-N53, tumstatin (Genzyme), Thios- 1 , Thios-2 (Thios Pharmaceuticals), MV-6401 (Miravant Medical Technologies), Spisulosine (PharmaMar), CEP-7055 (Cephalon), AUV-201 (Auvation), LM-609 (EIi Lilly), SKF-106615 (AnorMED), Oglufanide disodium (Cytran), BW-114 (Pharminox), Calreticulin (NIH), WX-678 (Wilex), SD-7784 (Pfizer), WX-UK1 (Wilex), SH-268 (Schering AG), 2-Me-PGA (Celgene), S- 137 (Pfizer), ZD-6126 (Angiogene Pharmaceuticals), SG-292 (SignalGen), Benefin (Lane Labs), A6, A36 (Angstrom), SB-2723005 (GlaxoSmithKline), SC-7 (Cell Therapeutics), ZEN-014 (AEterna Zentaris), 2-methoxyestradiol (EntreMed), NK-130119 (Nippon Kayaku), CC-10004 (Celgene), AVE- 8062A (Ajinomoto), Tacedinaline (Pfizer), Actinonin (Tokyo Metropolitan Institute of Medical Science), Lenalidomide (Celgene), VGA-1155, BTO-956 (SRI International), ER-68203-00 (Eisai), CT-6685 (UCB), JKC-362 (Phoenix Pharmaceuticals), DMI-3798 (DMI Biosciences, Angiomate (Ipsen), ZD-6474 (AstraZeneca), CEP-5214 (Cephalon), Canstatin (Genzyme) r NM-3 (Mercian), Oridigm (MediQuest Therapeutics), Exherin (Adherex), BLS-0597 (Boston Life Sciences), PTC-299 (PTC Therapeutics), NPI-2358 (Nereus Pharmaceuticals), CGP-79787 (Novartis), AEE-788 (Novartis), CKD-732 (Chong Kun Dang), CP-564959 (OSI Pharmaceuticals), CM-101 (CarboMed), CT-2584, CT3501 (Cell Therapeutics), combretastatin and analogues and derivatives thereof (Oxigene), Rebimastat (Bristol-Meyers Squibb), Dextrin 2-sulfate (ML Laboratories), Cilengitide (Merk KGaA), NSC-706704 (Pharminox), KRN- 951 (Kirin Brewery), Ukrain, NSC-631570 (Nowicky Pharma), Tecogalan sodium (Daiichi Pharmaceutical), Tz-93 (Tsumura), TBC-1635 (Encysive Pharmaceuticals), TAN-1120 (Takeda), Semaxanib (Pfizer), BDI-7800 (Biopharmacopae), SD-186, SD-983 (Bristol-Meyers Squibb), SB-223245 (GlaxoSmithKline), SC-236 (Pfizer), RWJ-590973 (Johnson and Johnson), ILX-1850 (Genzyme), SC-68488, S-836 (Pfizer), CG-55069-11 (CuraGen), Ki-23057 (Kirin Brewery), CCX-700 (Chemoentryx), Pegaptanib octasodium (Giled Sciences), ANGIOCOL (available from Biostratum Inc.), or an analogue or derivative thereof.
2553. The device of item 2546 wherein the agent is an apoptosis antagonist.
2554. The device of item 2546 wherein the agent is an apoptosis activator.
2555. The device of item 2546 wherein the agent is a beta 1 integrin antagonist.
2556. The device of item 2546 wherein the agent is a beta tubulin inhibitor.
2557. The device of item 2546 wherein the agent is a blocker of enzyme production in Hepatitis C.
2558. The device of item 2546 wherein the agent is a Bruton's tyrosine kinase inhibitor.
2559. The device of item 2546 wherein the agent is a calcineurin inhibitor.
2560. The device of item 2546 wherein the agent is a caspase 3 inhibitor.
2561. The device of item 2546 wherein the agent is a CC chemokine receptor antagonist. 2562. The device of item 2546 wherein the agent is a ceil cycle inhibitor selected from the group consisting of SNS-595 (Sunesis), synthadotin, KRX-0403, and an analogue or derivative thereof.
2563. The device of item 2546 wherein the agent is a cathepsin B inhibitor.
2564. The device of item 2546 wherein the agent is a cathepsin K inhibitor, wherein the cathepsin K inhibitor is 462795 (GlaxoSmithKline), INPL-022-D6 (Amura Therapeutics), an analogue or derivative thereof).
2565. The device of item 2546 wherein the agent is a cathepsin L inhibitor.
2566. The device of item 2546 wherein the agent is a CD40 antagonist.
2567. The device of item 2546 wherein the agent is a chemokine receptor agonist.
2568. The device of item 2546 wherein the agent is a chymase inhibitor.
2569. The device of item 2546 wherein the agent is a collagenase antagonist.
2570. The device of item 2546 wherein the agent is a CXCR antagonist. 2571. The device of item 2546 wherein the agent is a cyclin dependent kinase inhibitor selected from the group consisting of a CDK-1 inhibitor, a CDK-2 inhibitor, a CDK- 4 inhibitor, a CDK-6 inhibitor, a CAK1 inhibitor from GPC Biotech and Bristol-Myers Squibb, RGB-286199 (GPC Biotech), an anticancer agent from Astex Technology, a CAK1 inhibitor from GPC Biotech, a CDK inhibitor from Sanofi-Aventis, a CDK1/CDK2 inhibitor from Amgen, a CDK2 inhibitor from SUGEN-2 (Pfizer), a hearing loss therapy agent (Sound Pharmaceuticals), PD-0332991 (Pfizer), RGB-286199 (GPC Biotech), Ro-0505124 (Hoffmann-La Roche), a Ser/Thr kinase inhibitor from Lilly (EIi Lilly), CVT-2584 (CAS No. 199986-75-9) (CV Therapeutics), and an analogue or derivative thereof.
2572. The device of item 2546 wherein the agent is a cyclooxygenase 1 inhibitor.
-2573, The device of item 2546 wherein the agent is a DHFR inhibitor.
2574. The device of item 2546 wherein the agent is a dual integrin inhibitor.
2575. The device of item 2546 wherein the agent is an elastase inhibitor.
2576. The device of item 2546 wherein the agent is an elongation factor-1 alpha inhibitor.
2577. The device of item 2546 wherein the agent is an endothelial growth factor antagonist. 2578. The device of item 2546 wherein the agent is an endothelial growth factor receptor kinase inhibitor selected from the group consisting of sorafenib tosylate (Bayer), AAL-993 (Novartis), ABP-309 (Novartis), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), E- 7080 (Eisai), EG-3306 (Ark Therapeutics), EXEL-2880 (Exelixis), GW- 654652 (GlaxoSmithKline), a KDR inhibitor from LG Life Sciences, CT-6685 and CT-6729 (UCB), KRN-633 and KRN-951 (Kirin Brewery), OSI-930 (OSI Pharmaceuticals), SP-5.2 (Supratek Pharma), SU-11657 (Pfizer), a Tie-2 antagonist (Hybrigenics), a VEGFR-2 kinase inhibitor (Bristol-Myers Squibb), XL-647 (Exelixis), a KDR inhibitor from Abbott Laboratories, sorafenib tosylate, and an analogue or derivative thereof.
2579. The device of item 2546 wherein the agent is an endotoxin antagonist.
2580. The device of item 2546 wherein the agent is an epothilone and tubulin binder.
2581. The device of item 2546 wherein the agent is an estrogen receptor antagonist.
2582. The device of item 2546 wherein the agent is an FGF inhibitor.
2583. The device of item 2546 wherein the agent is a farnexyl transferase inhibitor.
2584. The device of item 2546 wherein the agent is farnesyltransferase inhibitor selected from the group of A-197574 (Abbott), a famesyltransferase inhibitor from Servier, FPTIII (Strathclyde Institute for Drug R), LB-42908 (LG Life Sciences), Pharmaprojects No. 5063 (Genzyme), Pharmaprojects No. 5597 (Ipsen), Yissum Project No. B-1055 (Yissum), and an analogue or derivative thereof.
2585. The device of item 2546 wherein the agent is an FLT-3 kinase inhibitor.
2586. The device of item 2546 wherein the agent is an FGF receptor kinase inhibitor.
2587. The device of item 2546 wherein the agent is a fibrinogen antagonist selected from the group consisting of AUV-201 (Auvation), MG-13926 (Sanofi-Aventis), plasminogen activator (CAS No. 105913-11-9) (from Sanofi-Aventis or UCB), plasminogen activator-2 (tPA- 2) (Sanofi-Aventis), pro-urokinase (CAS No. 82657-92-9) (Sanofi-Aventis), and an analogue or derivative thereof.
2588. The device of item 2546 wherein the agent is a heat shock protein 90 antagonist selected from the group consisting of SRN-005 (Sirenade), geldanamycin, NSC-33050 (17-allylaminogeldanamycin; 17- AAG), 17-dimethylaminoethylamino-17-demethoxy-geldanamycin (17- DMAG), rifabutin (rifamycin XIV, 1\4-didehydro-1-deoxy-1 ,4-dihydro-5'-(2- methylpropyl)-1-oxo-), and an analogue or derivative thereof.
2589. The device of item 2546 wherein the agent is a histone deacetylase inhibitor.
2590. The device of item 2546 wherein the agent is an HMGCoA reductase inhibitor selected from the group consisting of an atherosclerosis therapeutic from Lipid Sciences, ATI-16000 (ARYx Therapeutics), KS-01-019 (Kos Pharmaceuticals), Pharmaprojects No. 2197 (Sanofi-Aventi), RP 61969 (Sanofi-Aventis), and an analogue or derivative thereof.
2591. The device of item 2546 wherein the agent is an ICAM inhibitor.
2592. The device of item 2546 wherein the agent is an IL, ICE and IRAK antagonist, wherein the antagonist is a CJ-14877, CP-424174 (Pfizer), NF-61 (Negma-Lerads), and an analogue or derivative thereof.
2593. The device of item 2546 wherein the agent is an IL-2 inhibitor.
2594. The device of item 2546 wherein the agent is an immunosuppressant selected from the group consisting of teriflunomide (Sanofi Aventis), chlorsulfaquinoxalone (NSC-339004), chlorsulfaquinoxalone sulfate, CS-712 (Sankyo), ismomultin alfa (CAS No. 457913-93-8) (Akzo Nobel), antiallergics from GenPat77, antiinflammatories or AT-005 (Androclus Therapeutics), autoimmune disease therapeutics from EpiVax, BN-007 (Bone), budesonide (CAS No. 51333-22- 3) (MAP Pharmaceuticals), CO-14 (Genzyme), edratide (CAS No. 433922- 67-9) (Teva), EP-314 (Enanta), eprovafen (CAS No. 101335-99-3) (Sanofi- Aventis), HWA-131 (CAS No. 118788-41-3) (Sanofi-Aventis), immunomodulators from MerLion Pharmaceuticals, immunosuppressives from Alchemia, IPL-12 (Inflazyme), MDL-9563 (CAS No. 27086-86-8) (Sanofi-Aventis), Pharmaprojects No. 2330 (Sanofi-Aventis), Pharmaprojects No. 6426 (Abgenix), PXS-25 (Pharmaxis), rosmarinic acid (CAS No. 20283-92-5) (Sanofi-Aventis), RP 42927 or RP 54745 (CAS No. 135330-08-4) (Sanofi-Aventis), SGN-35 (Seattle Genetics), ST-1959 (Sigma-Tau), type I diabetes therapy from SYNX Pharma, UNIL-88 (Debiopharm), VP-025 (Vasogen), VR-694 (Vectura), PRTX-001 (Protalex), and an analogue or derivative thereof.
2595. The device of item 2546 wherein the agent is an IMPDH (inosine monophosphate).
2596. The device of item 2546 wherein the agent is an integrin antagonist.
2597. The device of item 2546 wherein the agent is an interleukin antagonist.
2598. The device of item 2546 wherein the agent is an inhibitor of type III receptor tyrosine kinase.
2599. The device of item 2546 wherein the agent is an irreversible inhibitor of enzyme methionine aminopeptidase type 2.
2600. The device of item 2546 wherein the agent is an isozyme selective delta protein kinase C inhibitor.
2601. The device of item 2546 wherein the agent a JAK3 enzyme inhibitor.
2602. The device of item 2546 wherein the agent is a JNK inhibitor.
2603. The device of item 2546 wherein the agent is a kinase inhibitor. 2604. The device of item 2546 wherein the agent is kinesin antagonist.
2605. The device of item 2546 wherein the agent is a kinesin antagonist.
2606. The device of item 2546 wherein the agent is a leukotriene inhibitor and antagonist selected from the group consisting of ambicromil (CAS No. 58805-38-2) (Sanofi-Aventis), amelubant (CAS No. 346735-24-8) (Boehringer Ingelheim), DW-1141 (Dong Wha), ebselen (Daiichi Pharmaceutical), ibudilast (Kyorin), leucotriene inhibitors from Sanofi-Aventis, lymphotoxin-beta receptor (LT-D) from Biogen Idee, Pharmaprojects No. 1535 and 2728 (CAS No. 119340-33-9) (Sanofi- Aventis), R-112 (Rigel), Rev-5367 (CAS No. 92532-05-3) (Sanofi-Aventis), RG-14893 (CAS No. 141835-49-6) (Sanofi-Aventis), RG-5901-A (CAS No. 101910-24-1), 92532-23-5, RP 66153 (CAS No. 142422-79-5), RP 66364 (CAS No.186916-92-5), RP 69698 (CAS No. 141748-00-7) (Sanofi-Aventis), SC-411930 (Pfizer), SC-41930 (CAS No. 120072-59-5) (Pfizer), SC-50605 (CAS No. 138828-39-4) (Pfizer), SC-51146 (CAS No. 141059-52-1), SC- 53228 (CAS No. 153633-01-3) (Pfizer), spaglumic acid (ZY-15106) (CAS No.3106-85-2), 80619-64-3 (Novartis), tipredane (CAS No. 85197-77-9) (Bristol-Myers Squibb), U-75302 (CAS No. 119477-85-9) (Pfizer), and analogue or derivative thereof.
2607. The device of item 2546 wherein the agent is an MAP kinase inhibitor.
2608. The device of item 2546 wherein the agent is a matrix metalloproteinase inhibitor. 2609. The device of item 2546 wherein the agent is an MCP- CCR2 inhibitor.
2610. The device of item 2546 wherein the agent is an mTOR inhibitor.
2611. The device of item 2546 wherein the agent is an mTOR kinase inhibitor.
2612. The device of item 2546 wherein the agent is a microtubule inhibitor selected from the group consisting of antibody- maytansinoid conjugates from Biogen Idee, colchicines (MantiCore Pharmaceuticals), anticancer immunoconjugates from Johnson & Johnson, DIME from Octamer, gni-1f (GNI), huC242-DM4, huMy9-6-DM1 (ImmunoGen), lDN-5404 (Indena), IMO-098, IMOderm (Imotep), mebendazole (Introgen Therapeutics), microtubule poisons from Cambridge Enterprise, paclitaxel such as LOTAX from Aphios (CAS No. 33069-62-4), Genexol-PM from Samyang, Pharmaprojects No. 6383 (Tapestry Pharmaceuticals), RPR-112378 (Sanofi-Aventis), SGN-75 (Seattle Genetics), SPL-7435 (Starpharma), SSR-250411 (Sanofi-Aventis), trastuzumab-DM1 (Genentech), vinorelbine, and an analogue or derivative thereof.
2613. The device of item 2546 wherein the agent is an MIF inhibitor.
2614. The device of item 2546 wherein the agent is an MMP inhibitor.
2615. The device of item 2546 wherein the agent is a neurokinin (NK) antagonist selected from the group consisting of anthrotainin (CAS No. 148084-40-6) (Sanofi-Aventis), an IBS thereapeutic from ArQuIe, MDL-105212A (CAS No. 167261-60-1) (Ssanofi-Aventis), Pharmaprojects No. 2744, 3258 (CAS No. 139167-47-8) 4006, 4201, or 5986 (Sanofi-Aventis), RP 67580 (CAS No. 135911-02-3), SR-144190 (CAS No. 201152-86-5), SSR-240600, SSR-241586 (Sanofi-Aventis), TKA-457 (Novartis), vestipitant mesylate (CAS No. 334476-64-1) (GlaxoSmithKline), Win-64821 (Sanofi-Aventis), PRX-96026 (Predix Pharmaceuticals), and an analogue or derivative thereof.
2616. The device of item 2546 wherein the agent is an NF kappa B inhibitor selected from the group consisting of AVE-0545 or AVE- 0547 (Sanofi-Aventis), bortezomib (CAS No. 179324-69-7) (Millennium Pharmaceuticals), dexanabinol (CAS No. 112924-45-5) (Pharmos), dexlipotam (Viatris), Pharmaprojects No. 6283 (INDRA) (OXiGENE), IPL- 576092 (CAS No. 137571-30-3) (Inflazyme), NFKB decoy (Corgentech), NFKB decoy oligo (AnGes MG), NFKB's from Ariad, osteoporosis treatments or S5 (F005) from Fulcrum Pharmaceuticals, P61 (Phytopharm), R-flurbiprofen (CAS No. 5104-49-4) (Encore Pharmaceuticals), and an analogue or derivative thereof.
2617. The device of item 2546 wherein the agent is a nitric oxide agonist.
2618. The device of item 2546 wherein the agent is an ornithine decarboxylase inhibitor.
2619. The device of item 2546 wherein the agent is a p38 MAP kinase inhibitor selected from the group consisting of AZD-6703 (AstraZeneca), JX-401 (Jexys Pharmaceuticals), BMS-2 (Bristol-Myers Squibb), a p38 MAP kinase inhibitor from Novartis, a p38-alpha MAP kinase inhibitor from Amphora, Pharmaprojects No. 5704 (Pharmacopeia), RPR- 200765A (Sanofi-Aventis), SD-282 (Johnson & Johnson), TAK-715 (Takeda), and an analogue or derivative thereof.
2620. The device of item 2546 wherein the agent is a palmitoyl-protein thioesterase inhibitor.
2621. The device of item 2546 wherein the agent is a PDGF receptor kinase inhibitor selected from the group consisting of AAL-993, AMN-107, or ABP-309 (Novartis), AMG-706 (Amgen), BAY-57-9352 (Bayer), CDP-860 (UCB), E-7080 (Eisai), imatinib (CAS No. 152459-95-5) (Novartis), OSI-930 (OSI Pharmaceuticals), RPR-127963E (Sanofi-Aventis), RWJ-540973 (Johnson & Johnson), sorafenib tosylate (Bayer), SU-11657 (Pfizer), tandutinib (CAS No. 387867-13-2) (Millennium Pharmaceuticals), vatalanib (Novartis), ZK-CDK (Schering AG), and an analogue or derivative thereof.
2622. The device of item 2546 wherein the agent is (-)- halofenate (Metabolex), AMG-131 (Amgen), antidiabetics from Japan Tobacco, AZD-4619, AZD-8450, AZD-8677 (AstraZeneca), DRF-10945, balaglitazone (Dr Reddy's), CS-00088, CS-00098 (Chipscreen Biosciences), E-3030 (Eisai), etalocib (CAS No. 161172-51-6) (EIi Lilly), GSK-641597 (Ligand), GSK-677954 (GlaxoSmithKline), GW-409544 (Ligand), GW- 590735 (GlaxoSmithKline), K-111 (Hoffmann-La Roche), LY-518674 (EIi Lilly), LY-674 (Ligand), LY-929 (Ligand), MC-3001, MC-3002 (MaxoCore Pharmaceuticals), metformin HCI + pioglitazone (CAS No. 1115-70-4 and
112529-15-4), ACTOPLUS MET from Andrx), muraglitazar (CAS No. 331741-94-7) (Bristol-Myers Squibb), naveglitazar (Ligand), oleoylethanolamide (Kadmus Pharmaceuticals), ONO-5129, pioglitazone hydrochloride (CAS No. 111025-46-8 and 112529-15-4) (Takeda), PLX-204 (Plexxikon), PPAR agonists from Genfit, PPAR delta agonists from EIi Lilly, PPAR-alpha agonists from CrystalGenomics, PPAR-gamma modulators and PPAR-β modulators from CareX, rosigiitazone maleate (CAS No. 122324- 73-4 or 155141-29-0) (GlaxoSmithKline), rosigiitazone maleate/glimepir (CAS No. 155141-29-0 and 93479-97-1), AVANDARYL, rosigiitazone maleate/metformin extend (CAS No. 155141-29-0 and 657-24-9), AVANDAMET, rosigiitazone maleate+metformin, AVANDAMET (GlaxoSmithKline), tesaglitazar (AstraZeneca), and an analogue or derivative thereof.
2623. The device of item 2546 wherein the agent is a phosphatase inhibitor.
2624. The device of item 2546 wherein the agent is a phosphodiesterase (PDE) inhibitor selected from the group consisting of avanafil (Tanabe Seiyaku), dasantafil (CAS No. 569351-91-3) (Schering- Plough), A-906119 (CAS No. 134072-58-5), DL-850 (Sanofi-Aventis), GRC- 3015, GRC-3566, GRC-3886 (Glenmark), HWA-153 (CAS No. 56395-66-5) (Sanofi-Aventis), hydroxypumafentrine (Altana), IBFB-130011 , IBFB-14-016, IBFB-140301 , IBFB-150007, IBFB-211913 (IBFB Pharma), L-826141 (Merck & Co), medorinone (CAS No. 88296-61-1) (Sanofi-Aventis), MEM- 1917 (Memory Pharmaceuticals), ND-1251 (Neuro3d), PDE inhibitors from ICOS, PDE IV inhibitors from Memory Pharmaceuticals and CrystalGenomics, Pharmaprojects No. 2742 and 6141 (Sanofi-Aventis), QAD-171 (Novartis), RHC-2963 (CAS No. 76993-12-9 and 76993-14-1), RPR-117658, RPR-122818 derivatives, SR-24870 , and RPR-132294 (Sanofi-Aventis), SK-350 (ln2Gen), stroke therapy agents from deCODE Genetics, TAS-203 (Taiho), tofimilast (CAS No. 185954-27-2) (Pfizer), UK- 371800 (Pfizer), WIN-65579 (CAS No. 158020-82-7) (Sanofi-Aventis), IBFB- 130020 (IBFB Pharma), OPC-6535 (CAS No. 145739-56-6) (Otsuka), a phosphodiesterase III inhibitor, enoximone, a phosphodiesterase IV inhibitor, fosfosal, Atopik (Barrier Therapeutics), triflusal, a phosphodiesterase V inhibitor, and an analogue or derivative thereof. 2625. The device of item 2546 wherein the agent is a PKC inhibitor.
2626. The device of item 2546 wherein the agent is a platelet activating factor antagonist.
2627. The device of item 2546 wherein the agent is a platelet- derived growth factor receptor kinase inhibitor.
2628. The device of item 2546 wherein the agent is a prolyl hydroxylase inhibitor.
2629. The device of item 2546 wherein the agent is a polymorphonuclear neutrophil inhibitor.
2630. The device of jtem 2546 wherein the agent is a protein kinase B inhibitor.
2631. The device of item 2546 wherein the agent is a protein kinase C stimulant.
2632. The device of item 2546 wherein the agent is a purine nucleoside analogue.
2633. The device of item 2546 wherein the agent is a purinoreceptor P2X antagonist.
2634. The device of item 2546 wherein the agent is a Raf kinase inhibitor. 2635. The device of item 2546 wherein the agent is a reversible inhibitor of ErbB1 and ErbB2.
2636. The device of item 2546 wherein the agent is a ribonucleoside triphosphate reductase inhibitor.
2637. The device of item 2546 wherein the agent is an SDF-1 antagonist.
2638. The device of item 2546 wherein the agent is a sheddase inhibitor.
2639. The device of item 2546 wherein the agent is an SRC inhibitor.
2640. The device, of jtem 2546 wherein^ the agent is a stromelysin inhibitor.
2641. The device of item 2546 wherein the agent is an Syk kinase inhibitor.
2642. The device of item 2546 wherein the agent is a telomerase inhibitor.
2643. The device of item 2546 wherein the agent is a TGF beta inhibitor selected from the group consisting of pirfenidone (CAS No. 53179-13-8) (MARNAC), tranilast (CAS No. 53902-12-8) (Kissei), IN-1130 (ln2Gen), mannose-6-phosphate (BTG), TGF-β antagonists from Inflazyme (Pharmaprojects No. 6075), TGF-β antagonists from Sydney, non-industrial source), TGF-β I receptor kinase inhibitors from EIi Lilly, TGF-β receptor inhibitors from Johnson & Johnson, and an analogue or derivative thereof. 2644. The device of item 2546 wherein the agent is a TNFα antagonist or TACE inhibitor selected from the group consisting of adalimumab (CAS No. 331731-18-1) (Cambridge Antibody Technology), AGIX-4207 (AtheroGenics), AGT-1 (Advanced Biotherapy), an antiinflammatory from Borean Pharma, Cellzome, or Paradigm Therapeutics, anti-inflammatory vaccine (TNF-alpha kinoid) from Neovacs, humanized anti-TNF antibody or an anti-TNF MAb (CB0006) Celltech (UCB), apratastat (CAS No. 287445-51-0) (Wyeth), BMS-561392 (Bristol-Myers Squibb), BN- 006 (Bone), certolizumab pegol (CAS No. 428863-50-7 or CH-138 (UCB), cilomilast (CAS No. 153259-65-5) (GlaxoSmithKline), CR-1 (Nuada Pharmaceuticals), CRx-119 (CombinatoRx), D-5410 (UCB), dacopafant (CAS No. 125372-33-0) (Sanofi-Aventis), dersalazine (CAS No. 188913-57- 7/188913-58-8) (Uriach), etanercept (CAS No. 185243-69-0) (Amgen), ethyl pyruvate (Critical (Critical Therapeutics), golimumab (CAS No. 476181-74-5) (Johnson & Johnson), hormono-immunotherapy from Ipsen, CDP571 (e.g., Humicade from JJCB), IC-485 (ICOS), infliximab (CAS No. 170277-31-3) (Johnson & Johnson), IP-751 (Manhattan Pharmaceuticals), ISIS-104838 (CAS No. 250755-32-9) (ISIS Pharmaceuticals), lenalidomide (CAS No. 191732-72-6) (Celgene), lentinan (CAS No. 37339-90-5) (Ajinomoto), MDL- 201112 (CAS No. 142130-73-2) (Sanofi-Aventis), medroxyprogesterone (CAS No. 520-85-4) (InKine Pharmaceutical), N-acetylcysteine (CAS No. 616-91-1) (Zambon), NBE-P2 (DIREVO Biotech), nerelimomab (CAS No. 162774-06-3) (Chiron), OM-294DP (OM PHARMA), onercept (CAS No. 199685-57-9) (Yeda), PASSTNF-alpha (Verigen), pentoxifylline or oxypentifylline (Sanofi-Aventis), Pharmaprojects No. 4091, 4241, 4295, or 4488 (Sanofi-Aventis), Pharmaprojects No. 5480 (Amgen), Pharmaprojects No. 6749 (Cengent), pirfenidone (CAS No. 53179-13-8) (MARNAC), RPR- 132294 (Sanofi-Aventis), S5 (F002) (Fulcrum Pharmaceuticals), simvastatin (CAS No. 79902-63-9) (Merck & Co), STA-6292 (Synta Pharmaceuticals), tacrolimus (CAS No. 104987-11-3) (Fujisawa LifeCycle Pharma), talactoferrin alfa (CAS No. 308240-58-6) (Agennix), thalidomide (CAS No. 50-35-1) (Celgene), TNF antagonists form ProStrakan, and Synergen, TNF inhibitors (Amgen), TNF-alpha antagonists from Dynavax Technologies and Jerina AG (Germany), TNF-alpha inhibitors from IBFB Pharma and Xencor (Xencor), torbafylline (CAS No. 105102-21-4) (Sanofi-Aventis), UR-1505 (Uriach), VT-346 (Viron Therapeutics), YSIL6 (Y1S Therapeutics), YSTH2 (Y's Therapeutics), NPI-1302a-3 (Nereus Pharmaceuticals, a TNF antagonist from Jerina AG (Germany), dersalazine, and an analogue or derivative thereof.
2645. The device of item 2546 wherein the agent is a Toll receptor inhibitor.
2646. The device of item 2546 wherein the agent is a tubulin antagonist.
2647. The device of item 2546 wherein the agent is a tyrosine kinase inhibitor selected from the group consisting of SU-011248, SUTENT from Pfizer Inc. (New York, NY), BMS-354825, PN-355 (Paracelsian Pharmaceuticals), AGN-199659 (Allergan), AAL-993 or ABP-309 (Novartis), adaphostin (NIH), AEE-788 (Novartis), AG-013736 (OSI Pharmaceuticals), AG-13736 (Pfizer), ALT-110 (Alteris Therapeutics), AMG-706 (Amgen), anticancer MAbs from Xencor, anti-EGFrvlll MAbs from Abgenix, anti-HER2 MAb from Abiogen, AZD-2171 or AZD-9935 (AstraZeneca), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), CEP-5214 (Cephalon), CEP- 7055 (Cephalon), cetuximab (ImClone Systems), CHIR-200131 and CHIR- 258 (Chiron), CP-547632 (OSI Pharmaceuticals), CP-724714 (Pfizer), CT- 301 (Creabilis Therapeutics), D-69491 (Baxter International), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EGFR/ErbB2 inhibitors from Array BioPharma, erlotinib (CAS No. 183319-69-9) (OSI Pharmaceuticals), EXEL-2880 (Exelixis), FK-778 (Sanofi-Aventis) , gefitinib (CAS No. 184475-35-2) (AstraZeneca), GW-2286 or GW-654652 (GlaxoSmithKline), her2/neu antigen from AlphaVax, HER-2/neu inhibitor from Generex, Herzyme (Medipad) (Sirna Therapeutics), HKI-272 (Wyeth), HuMax-EGFr (Genmab), idronoxil (CAS No. 81267-65-4) (Novogen), IGF-1 inhibitors from Ontogen, IMC-11 F8 (ImClone Systems), kahalalide F (CAS No. 149204-42-2) (PharmaMar), KDR inhibitor from LG Life Sciences, KDR inhibitors from Abbott Laboratories, KDR kinase inhibitors (UCB), Kdr kinase inhibitors from Merck & Co, KRN-633 and KRN-951 (Kirin Brewery), KSB-102 (Xenova), lapatinib ditosylate (CAS No. 388082-78-8) (GlaxoSmithKline), matuzumab (Merck KGaA), MDX-214 (Medarex), ME-103 (Pharmexa), MED-A300 (Gerolymatos), MNAC-13 (Lay Line Genomics), nimotuzumab (Center of Molecular Immunology), NSC-330507 or NSC-707545 (NIH), NV-50 (Novogen), OSI-930 (OSI Pharmaceuticals), panitumumab (Abgenix), pelitinib (CAS No. 287933-82-7) (Wyeth), pertuzumab (CAS No. 380610-27- 5) (Genentech), Pharmaprojects No. 3985 (Sanofi-Aventis), prostate cancer therapeutics from Sequenom (SQPC35, SQPC36, SQPC90), removab and remoxab (Trion Pharma), RG-13022 (CAS No. 136831-48-6), RG-13291 (CAS No. 138989-50-1), or RG-14620 (CAS No. 136831-49-7) (Sanofi- Aventis), RM-6427 (Romark), RNAi breast cancer therapy from Benitec, RP 53801 (CAS No. 125882-88-4) (Sanofi-Aventis), sorafenib tosylate (Bayer), SU-11657 (Pfizer), Tie-2 antagonists from Semaia (Hybrigenics), Tie-2 inhibitors from Ontogen, trastuzumab (CAS No. 180288-69-1) (Genentech), tyrosine kinase inhibitors from Sanofi-Aventis, U3-1287, U3-1565, U3-1784, U3-1800 (U3 Pharma), vatalanib (Novartis), VEGFR-2 kinase inhibitor from Bristol-Myers Squibb, XL-647 (Exelixis), ZD-6474 (AstraZeneca), ZK-CDK (Schering AG), an EGFR tyrosine kinase inhibitor, EKB-569 (Wyeth), and an analogue or derivative thereof.
2648. The device of item 2546 wherein the agent is a VEGF inhibitor. 2649. The device of item 2546 wherein the agent is a vitamin D receptor agonist.
2650. The device of item 2546 wherein the agent is ZD-6474 (an angiogenesis inhibitor).
2651. The device of item 2546 wherein the agent is AP-23573 (an mTOR inhibitor).
2652. The device of item 2546 wherein the agent is synthadotin (a tubulin antagonist).
2653. The device of item 2546 wherein the agent is S-0885 (a collagenase inhibitor).
2654^ The device otitem 2546 wherein the agent is aplidine (an elongation factor-1 alpha inhibitor).
2655. The device of item 2546 wherein the agent is ixabepilone (an epithilone).
2656. The device of item 2546 wherein the agent is IDN-5390 (an angiogenesis inhibitor).
2657. The device of item 2546 wherein the agent is SB- 2723005 (an angiogenesis inhibitor).
2658. The device of item 2546 wherein the agent is ABT-518 (an angiogenesis inhibitor). 2659. The device of item 2546 wherein the agent is combretastatin (an angiogenesis inhibitor).
2660. The device of item 2546 wherein the agent is anecortave acetate (an angiogenesis inhibitor).
2661. The device of item 2546 wherein the agent is SB- 715992 (a kinesin antagonist).
2662. The device of item 2546 wherein the agent is temsirolimus (an mTOR inhibitor).
2663. The device of item 2546 wherein the agent is adalimumab (a TNFα antagonist).
2664. The device of item 2546, further comprising a polymer.
2665. The device of item 2546 wherein the anti-scarring agent inhibits adhesion between the device and a host into which the device is implanted.
2666. The device of item 2546 wherein the device delivers the anti-scarring agent locally to tissue proximate to the device.
2667. The device of item 2546, further comprising a coating, wherein the coating comprises the anti-scarring agent.
2668. The device of item 2546, further comprising a coating, wherein the coating is disposed on a surface of the device. 2669. The device of item 2546, further comprising a coating, wherein the coating directly contacts the device.
2670. The device of item 2546, further comprising a coating, wherein the coating indirectly contacts the device.
2671. The device of item 2546, further comprising a coating, wherein the coating partially covers the device.
2672. The device of item 2546, further comprising a coating, wherein the coating completely covers the device.
2673. The device of item 2546, further comprising a coating, wherein the coating is a uniform coating.
. 2674. The device of item 2546, further comprising a coating, wherein the coating is a non-uniform coating.
2675. The device of item 2546, further comprising a coating, wherein the coating is a discontinuous coating.
2676. The device of item 2546, further comprising a coating, wherein the coating is a patterned coating.
2677. The device of item 2546, further comprising a coating, wherein the coating has a thickness of 100 Dm or less.
2678. The device of item 2546, further comprising a coating, wherein the coating has a thickness of 10 Dm or less. 2679. The device of item 2546, further comprising a coating, wherein the coating adheres to the surface of the device upon deployment of the device.
2680. The device of item 2546, further comprising a coating, wherein the coating is stable at room temperature for a period of 1 year.
2681. The device of item 2546, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 0.0001% to about 1% by weight.
2682. The device of item 2546, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 1 % to about 10% by weight.
2683. The device of item 2546, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 10% to about 25% by weight.
2684. The device of item 2546, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 25% to about 70% by weight.
2685. The device of item 2546, further comprising a coating, wherein the coating further comprises a polymer.
2686. The device of item 2546, further comprising a first coating having a first composition and the second coating having a second composition. 2687. The device of item 2546, further comprising a first coating having a first composition and the second coating having a second composition, wherein the first composition and the second composition are different.
2688. The device of item 2546, further comprising a polymeric carrier, wherein the polymeric carrier comprises a copolymer.
2689. The device of item 2546, further comprising a polymeric carrier, wherein the polymeric carrier comprises a block copolymer.
2690. The device of item 2546, further comprising a polymeric carrier, wherein the polymeric carrier comprises a random copolymer.
2691. The device of item 2546, further comprising a polymeric carrier, wherein the polymeric carrier comprises a biodegradable polymer.
2692. The device of item 2546, further comprising a polymeric carrier, wherein the polymeric carrier comprises a non-biodegradable polymer.
2693. The device of item 2546, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrophilic polymer.
2694. The device of item 2546, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrophobic polymer.
2695. The device of item 2546, further comprising a polymeric carrier, wherein the polymeric carrier comprises a polymer having hydrophilic domains. 2696. The device of item 2546, further comprising a polymeric carrier, wherein the polymeric carrier comprises a polymer having hydrophobic domains.
2697. The device of item 2546, further comprising a polymeric carrier, wherein the polymeric carrier comprises a non-conductive polymer.
2698. The device of item 2546, further comprising a polymeric carrier, wherein the polymeric carrier comprises an elastomer.
2699. The device of item 2546, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrogel.
2700. The device of item 2546, further comprising a polymeric carrier, wherein the polymeric carrier comprises a silicone polymer.
2701. The device of item 2546, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrocarbon polymer.
2702. The device of item 2546, further comprising a polymeric carrier, wherein the polymeric carrier comprises a styrene-derived polymer.
2703. The device of item 2546, further comprising a polymeric carrier, wherein the polymeric carrier comprises a butadiene polymer.
2704. The device of item 2546, further comprising a polymeric carrier, wherein the polymeric carrier comprises a macromer.
2705. The device of item 2546, further comprising a polymeric carrier, wherein the polymeric carrier comprises a poly(ethylene glycol) polymer. 2706. The device of item 2546, further comprising a polymeric carrier, wherein the polymeric carrier comprises an amorphous polymer.
2707. The device of item 2546, further comprising a lubricious coating.
2708. The device of item 2546 wherein the anti-scarring agent is located within pores or holes of the device.
2709. The device of item 2546 wherein the anti-scarring agent is located within a channel, lumen, or divet of the device.
2710. The device of item 2546, further comprising a second pharmaceutically active agent.
2711. The device of item 2546, further comprising an anti- - inflammatory agent.
2712. The device of item 2546, further comprising an agent that inhibits infection.
2713. The device of item 2546, further comprising an agent that inhibits infection, wherein the agent is an anthracycline.
2714. The device of item 2546, further comprising an agent that inhibits infection, wherein the agent is doxorubicin.
2715. The device of item 2546, further comprising an agent that inhibits infection, wherein the agent is mitoxantrone. 2716. The device of item 2546, further comprising an agent that inhibits infection, wherein the agent is a fluoropyrimidine.
2717. The device of item 2546, further comprising an agent that inhibits infection, wherein the agent is 5-fluorouracil (5-FU).
2718. The device of item 2546, further comprising an agent that inhibits infection, wherein the agent is a folic acid antagonist.
2719. The device of item 2546, further comprising an agent that inhibits infection, wherein the agent is methotrexate.
2720. The device of item 2546, further comprising an agent that inhibits infection, wherein the agent is a podophylotoxin.
2721. The device of item 2546, further comprising an agent that inhibits infection, wherein the agent is etoposide.
2722. The device of item 2546, further comprising an agent that inhibits infection, wherein the agent is a camptothecin.
2723. The device of item 2546, further comprising an agent that inhibits infection, wherein the agent is a hydroxyurea.
2724. The device of item 2546, further comprising an agent that inhibits infection, wherein the agent is a platinum complex.
2725. The device of item 2546, further comprising an agent that inhibits infection, wherein the agent is cisplatin. 2726. The device of item 2546, further comprising an antithrombotic agent.
2727. The device of item 2546, further comprising a visualization agent.
2728. The device of item 2546, further comprising a visualization agent, wherein the visualization agent is a radiopaque material, wherein the radiopaque material comprises a metal, a halogenated compound, or a barium containing compound.
2729. The device of item 2546, further comprising a visualization agent, wherein the visualization agent is a radiopaque material, wherein the radiopaque material comprises barium, tantalum, or technetium.
2730. The device of item 2546, further comprising a visualization agent, wherein the visualization agent is a MRI responsive material.
2731. The device of item 2546, further comprising a visualization agent, wherein the visualization agent comprises a gadolinium chelate.
2732. The device of item 2546, further comprising a visualization agent, wherein the visualization agent comprises iron, magnesium, manganese, copper, or chromium.
2733. The device of item 2546, further comprising a visualization agent, wherein the visualization agent comprises an iron oxide compound. 2734. The device of item 2546, further comprising a visualization agent, wherein the visualization agent comprises a dye, pigment, or colorant.
2735. The device of item 2546, further comprising an echogenic material.
2736. The device of item 2546, further comprising an echogenic material, wherein the echogenic material is in the form of a coating.
2737. The device of item 2546 wherein the device is sterile.
2738. The device of item 2546 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device ... - . . -
2739. The device of item 2546 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is connective tissue.
2740. The device of item 2546 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is muscle tissue.
2741. The device of item 2546 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is nerve tissue. 2742. The device of item 2546 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is epithelium tissue.
2743. The device of item 2546 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from the time of deployment of the device to about 1 year.
2744. The device of item 2546 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from about 1 month to 6 months.
2745. The device of item 2546 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from about 1 - 90 days.
2746. The device of item 2546 wherein the anti-scarring agent is released in effective concentrations from the device at a constant rate.
2747. The device of item 2546 wherein the anti-scarring agent is released in effective concentrations from the device at an increasing rate.
2748. The device of item 2546 wherein the anti-scarring agent is released in effective concentrations from the device at a decreasing rate.
2749. The device of item 2546 wherein the anti-scarring agent is released in effective concentrations from the composition comprising the anti-scarring agent by diffusion over a period ranging from the time of deployment of the device to about 90 days. 2750. The device of item 2546 wherein the anti-scarring agent is released in effective concentrations from the composition comprising the anti-scarring agent by erosion of the composition over a period ranging from the time of deployment of the device to about 90 days.
2751. The device of item 2546 wherein the device comprises about 0.01 Dg to about 10 Dg of the anti-scarring agent.
2752. The device of item 2546 wherein the device comprises about 10 Dg to about 10 mg of the anti-scarring agent.
2753. The device of item 2546 wherein the device comprises about 10 mg to about 250 mg of the anti-scarring agent.
2754. The device of item 2546 wherein the device comprises about 250 mg to about 1000 mg of the anti-scarring agent. - - - -
2755. The device of item 2546 wherein the device comprises about 1000 mg to about 2500 mg of the anti-scarring agent.
2756. The device of item 2546 wherein a surface of the device comprises less than 0.01 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
2757. The device of item 2546 wherein a surface of the device comprises about 0.01 Dg to about 1 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
2758. The device of item 2546 wherein a surface of the device comprises about 1 Dg to about 10 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied. 2759. The device of item 2546 wherein a surface of the device comprises about 10 Gg to about 250 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
2760. The device of item 2546 wherein a surface of the device comprises about 250 Dg to about 1000 Dg of the anti-scarring agent of anti- scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
2761. The device of item 2546 wherein a surface of the device comprises about 1000 Dg to about 2500 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
2762. A method as in any one of items 2546-2761, wherein the device is a surgical barrier.
2763. A method as in any one of items 2546-2761 , wherein the device is a surgical adhesion barrier.
2764. A method as in any one of items 2546-2761, wherein the device is a surgical sheet.
2765. A method as in any one of items 2546-2761 , wherein the device is a surgical patch.
2766. A method as in any one of items 2546-2761 , wherein the device is a surgical wrap.
2767. A method as in any one of items 2546-2761 , wherein the device is a vascular wrap. 2768. A method as in any one of items 2546-2761 , wherein the device is a perivascular wrap.
2769. A method as in any one of items 2546-2761 , wherein the device is a adventitial wrap.
2770. A method as in any one of items 2546-2761 , wherein the device is a periadventitial wrap.
2771. A method as in any one of items 2546-2761 , wherein the device is an adventitial sheet.
2772. A method as in any one of items 2546-2761 , wherein the device is a perivascular mesh.
.. _ 2773. A.method as. in any one of items 2546-276-1 , wherein the device is a bandage.
2774. A method as in any one of items 2546-2761 , wherein the device is a liquid bandage.
2775. A method as in any one of items 2546-2761, wherein the device is a surgical dressing.
2776. A method as in any one of items 2546-2761 , wherein the device is a gauze.
2777. A method as in any one of items 2546-2761 , wherein the device is a fabric. 2778. A method as in any one of items 2546-2761 , wherein the device is a tape.
2779. A method as in any one of items 2546-2761, wherein the device is a surgical membrane.
2780. A method as in any one of items 2546-2761 , wherein the device is a polymer matrix.
2781. A method as in any one of items 2546-2761 , wherein the device is a tissue covering.
2782. A method as in any one of items 2546-2761, wherein the device is a surgical matrix.
2783. A method as in any one of items 2546-2761 , wherein the device is a envelope.
2784. A method as in any one of items 2546-2761 , wherein the device is a tissue covering.
2785. A device, comprising a glaucoma drainage device (e.g., an implant) and an anti-scarring agent or a composition comprising an anti- scarring agent, wherein the agent inhibits scarring between the device and a host into which the device is implanted.
2786. The device of item 2785 wherein the agent is an adensosine A2A receptor antagonist.
2787. The device of item 2785 wherein the agent is an AKT inhibitor. 2788. The device of item 2785 wherein the agent is an alpha 2 integrin antagonist, wherein the alpha 2 integrin antagonist is Pharrnaprojects No. 5754 (Merck KgaA).
2789. The device of item 2785 wherein the agent an alpha 4 integrin antagonist.
2790. The device of item 2785 wherein the agent is an alpha 7 nicotinic receptor agonist.
2791. The device of item 2785 wherein the agent is an angiogenesis inhibitor selected from the group consisting of AG-12,958 (Pfizer), ATN-161 (Attenuon LLC), neovastat, an angiogenesis inhibitor from Jerina AG (Germany), NM-3 (Mercian), VGA-1155 (Taisho), FCE-26644 (Pfizer), FCE-26950 (Pfizer), FPMA (Meiji Daries), FR-111142 (Fujisawa), GGTI-298, GM-13Q6 (Ligand), GPA- 1734 (Novartis), NNC-47-0011 (Novo Nordisk), herbamycin (Nippon Kayaku), lenalidomide (Celegene), IP-10 (NIH), ABT-828 (Abbott), KIN-841 (Tokushima University, Japan), SF-1126 (Semafore Pharmaceuticals), laminin technology (NIH), CHIR-258 (Chiron), NVP-AEW541 (Novartis), NVP-AEW541 (Novartis), Vt16907 (Alchemia), OXI-8007 (Oxigene), EG-3306 (Ark Therapeutics), Maspin (Arriva), ABT- 567 (Abbott), PPI-2458 (Praecis Pharmaceuticals), CC-5079, CC-4089 (Celgene), HIF-1 alpha inhibitors (Xenova), S-247 (Pfizer), AP-23573 (Ariad), AZD-9935 (Astra Zeneca), mebendazole (Introgen Therapeutics), MetAP-2 inhibitors (GlaxoSmithKline), AG-615 (Angiogene Pharmaceuticals), Tie-2 antagonists (Hybrigenics), NC-381 , CYC-381, NC-169, NC-219, NC-383, NC-384, NC-407 (Lorus Therapeutics), ATN-224 (Attenuon), ON-01370 (Onconova), Vitronectin antagonists (Amgen), SDX-103 (Salmedix), Vitronectin antagonists (Shire), CHP (Riemser), TEK (Amgen), Anecortave acetate (Alcon), T46.2 (Matrix Therapeutics), HG-2 (Heptagen), TEM antagonists (Genzyme), Oxi-4500 (Oxigene), ATN-161 (Attenuon), WX-293 (Wilex), M-2025 (Metris Therapeutics), Alphastatin (BioActa), YH-16 (Yantai Rongchang), BIBF-1120 (Boehringer Ingelheim), BAY-57-9352 (Bayer), AS- 1404 (Cancer Research Technology), SC-77964 (Pfizer), glycomimetics (BioTie Therapies), TIE-2 Inhibitors (Ontogen), DIMI, Octamer (Octamer), ABR-215050 (Active Biotech), ABT-518 (Abbott), KDR inhibitors (Abbott), BSF-466895 (Abbott), SCH-221153 (Schering-Plough), DAC:antiangiogenic (ConjuChem), TFPI (EntreMed), AZD-2171 (Astra-Zenaca), CDC-394 (Celgene), LY290293 (EIi Lilly), IDN-5390 (Indena), Kdr Kinase Inhibitors (Merck), CT-113020, CT-116433, CT-116563, CT-31890, CT-32228) (Cell Therapeutics), A-299620 (Abbott), TWEAK Inhibitor (Amgen), VEGF modulators (Johnson and Johnson), Tum-N53, tumstatin (Genzyme), Thios- 1 , Thios-2 (Thios Pharmaceuticals), MV-6401 (Miravant Medical Technologies), Spisulosine (PharmaMar), CEP-7055 (Cephalon), AUV-201 (Auvation), LM-609 (EIi Lilly), SKF-106615 (AnorMED), Oglufanide disodium (Cytran), BW-114 (Pharminox), Calreticulin (NIH), WX-678 (Wilex), SD-7784 (Pfizer), WX-UKt (Wilex), SH-268 (Schering AG), 2-Me-PGA (Celgene), S- 137 (Pfizer), ZD-6126 (Angiogene Pharmaceuticals), SG-292 (SignalGen), Benefin (Lane Labs), A6, A36 (Angstrom), SB-2723005 (GlaxoSmithKline), SC-7 (Cell Therapeutics), ZEN-014 (AEterna Zentaris), 2-methoxyestradiol (EntreMed), NK-130119 (Nippon Kayaku), CC-10004 (Celgene), AVE- 8062A (Ajinomoto), Tacedinaline (Pfizer), Actinonin (Tokyo Metropolitan Institute of Medical Science), Lenalidomide (Celgene), VGA-1155, BTO-956 (SRI International), ER-68203-00 (Eisai), CT-6685 (UCB), JKC-362 (Phoenix Pharmaceuticals), DMI-3798 (DMI Biosciences, Angiomate (Ipsen), ZD-6474 (AstraZeneca), CEP-5214 (Cephalon), Canstatin (Genzyme), NM-3 (Mercian), Oridigm (MediQuest Therapeutics), Exherin (Adherex), BLS-0597 (Boston Life Sciences), PTC-299 (PTC Therapeutics), NPI-2358 (Nereus Pharmaceuticals), CGP-79787 (Novartis), AEE-788 (Novartis), CKD-732 (Chong Kun Dang), CP-564959 (OSI Pharmaceuticals), CM-101 (CarboMed), CT-2584, CT3501 (Cell Therapeutics), combretastatin and analogues and derivatives thereof (Oxigene), Rebimastat (Bristol-Meyers Squibb), Dextrin 2-sulfate (ML Laboratories), Cilengitide (Merk KGaA), NSC-706704 (Pharminox), KRN- 951 (Kirin Brewery), Ukrain, NSC-631570 (Nowicky Pharma), Tecogalan sodium (Daiichi Pharmaceutical), Tz-93 (Tsumura), TBC-1635 (Encysive Pharmaceuticals), TAN-1120 (Takeda), Semaxanib (Pfizer), BDI-7800 (Biopharmacopae), SD-186, SD-983 (Bristol-Meyers Squibb), SB-223245 (GlaxoSmithKline), SC-236 (Pfizer), RWJ-590973 (Johnson and Johnson), ILX-1850 (Genzyme), SC-68488, S-836 (Pfizer), CG-55069-11 (CuraGen), Ki-23057 (Kirin Brewery), CCX-700 (Chemoentryx), Pegaptanib octasodium (Giled Sciences), ANGIOCOL (available from Biostratum Inc.), or an analogue or derivative thereof.
2792. The device of item 2785 wherein the agent is an apoptosis antagonist.
2793. The device of item 2785 wherein the agent is an apoptosis activator.
2794. The device of item 2785 wherein the agent is a beta 1 integrin antagonist.
2795. The device of item 2785 wherein the agent is a beta tubulin inhibitor.
2796. The device of item 2785 wherein the agent is a blocker of enzyme production in Hepatitis C.
2797. The device of item 2785 wherein the agent is a Bruton's tyrosine kinase inhibitor. 2798. The device of item 2785 wherein the agent is a calcineurin inhibitor.
2799. The device of item 2785 wherein the agent is a caspase 3 inhibitor.
2800. The device of item 2785 wherein the agent is a CC chemokine receptor antagonist.
2801. The device of item 2785 wherein the agent is a cell cycle inhibitor selected from the group consisting of SNS-595 (Sunesis), synthadotin, KRX-0403, and an analogue or derivative thereof.
2802. The device of item 2785 wherein the agent is a cathepsin B inhibitor.
2803. The device of item 2785 wherein the agent is a cathepsin K inhibitor, wherein the cathepsin K inhibitor is 462795 (GlaxoSmithKline), INPL-022-D6 (Amura Therapeutics), an analogue or derivative thereof).
2804. The device of item 2785 wherein the agent is a cathepsin L inhibitor.
2805. The device of item 2785 wherein the agent is a CD40 antagonist.
2806. The device of item 2785 wherein the agent is a chemokine receptor agonist. 2807. The device of item 2785 wherein the agent is a chymase inhibitor.
2808. The device of item 2785 wherein the agent is a collagenase antagonist.
2809. The device of item 2785 wherein the agent is a CXCR antagonist.
2810. The device of item 2785 wherein the agent is a cyclin dependent kinase inhibitor selected from the group consisting of a CDK-1 inhibitor, a CDK-2 inhibitor, a CDK- 4 inhibitor, a CDK-6 inhibitor, a CAK1 inhibitor from GPC Biotech and Bristol-Myers Squibb, RGB-286199 (GPC Biotech), an anticancer agent from Astex Technology, a CAK1 inhibitor from GPC Biotech, a CDK inhibitor from Sanofi-Aventis, a CDK1/CDK2 inhibitor from Amgen, a CDK2 inhibitor from SUGEN-2 (Pfizer), a hearing loss therapy agent (Sound Pharmaceuticals), PD-0332991 (Pfizer), RGB-286199 (GPC Biotech), Ro-0505124 (Hoffmann-La Roche), a Ser/Thr kinase inhibitor from Lilly (EIi Lilly), CVT-2584 (CAS No. 199986-75-9) (CV Therapeutics), and an analogue or derivative thereof.
2811. The device of item 2785 wherein the agent is a cyclooxygenase 1 inhibitor.
2812. The device of item 2785 wherein the agent is a DHFR inhibitor.
2813. The device of item 2785 wherein the agent is a dual integrin inhibitor. 2814. The device of item 2785 wherein the agent is an elastase inhibitor.
2815. The device of item 2785 wherein the agent is an elongation factor-1 alpha inhibitor.
2816. The device of item 2785 wherein the agent is an endothelial growth factor antagonist.
2817. The device of item 2785 wherein the agent is an endothelial growth factor receptor kinase inhibitor selected from the group consisting of sorafenib tosylate (Bayer), AAL-993 (Novartis), ABP-309 (Novartis), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), E- 7080 (Eisai), EG-3306 (Ark Therapeutics), EXEL-2880 (Exelixis), GW- 654652 (GlaxoSmithKline), a KDR inhibitor from LG Life Sciences, CT-6685 and CT-6729 (UCB), KRN-633 and KRN-951 (Kirin Brewery), OSI-930 (OS! Pharmaceuticals), SP-5.2 (Supratek Pharma), SU-11657 (Pfizer), a Tie-2 antagonist (Hybrigenics), a VEGFR-2 kinase inhibitor (Bristol-Myers Squibb), XL-647 (Exelixis), a KDR inhibitor from Abbott Laboratories, sorafenib tosylate, and an analogue or derivative thereof.
2818. The device of item 2785 wherein the agent is an endotoxin antagonist.
2819. The device of item 2785 wherein the agent is an epothilone and tubulin binder.
2820. The device of item 2785 wherein the agent is an estrogen receptor antagonist. 2821. The device of item 2785 wherein the agent is an FGF inhibitor.
2822. The device of item 2785 wherein the agent is a farnexyl transferase inhibitor.
2823. The device of item 2785 wherein the agent is farnesyltransferase inhibitor selected from the group of A-197574 (Abbott), a farnesyltransferase inhibitor from Servier, FPTIII (Strathclyde Institute for Drug R), LB-42908 (LG Life Sciences), Pharmaprojects No. 5063 (Genzyme), Pharmaprojects No. 5597 (Ipsen), Yissum Project No. B-1055 (Yissum), and an analogue or derivative thereof.
2824. The device of item 2785 wherein the agent is an FLT-3 kinase inhibitor.
2825. The device of item 2785 wherein the agent is an FGF receptor kinase inhibitor.
2826. The device of item 2785 wherein the agent is a fibrinogen antagonist selected from the group consisting of AUV-201 (Auvation), MG-13926 (Sanofi-Aventis), plasminogen activator (CAS No. 105913-11-9) (from Sanofi-Aventis or UCB), plasminogen activator-2 (tPA- 2) (Sanofi-Aventis), pro-urokinase (CAS No. 82657-92-9) (Sanofi-Aventis), and an analogue or derivative thereof.
2827. The device of item 2785 wherein the agent is a heat shock protein 90 antagonist selected from the group consisting of SRN-005 (Sirenade), geldanamycin, NSC-33050 (17-allylaminogeldanamycin; 17- AAG), 17-dimethylaminoethylamino-17-demethoxy-geldanamycin (17- DMAG), rifabutin (rifamycin XIV, 1',4-didehydro-1-deoxy-1 ,4-dihydro-5'-(2- methylpropyl)-1-oxo-), and an analogue or derivative thereof.
2828. The device of item 2785 wherein the agent is a histone deacetylase inhibitor.
2829. The device of item 2785 wherein the agent is an HMGCoA reductase inhibitor selected from the group consisting of an atherosclerosis therapeutic from Lipid Sciences, ATI-16000 (ARYx Therapeutics), KS-01-019 (Kos Pharmaceuticals), Pharmaprojects No. 2197 (Sanofi-Aventi), RP 61969 (Sanofi-Aventis), and an analogue or derivative thereof.
2830. The device of item 2785 wherein the agent is an ICAM inhibitor.
2831. The device of item 2785 wherein the agent is an IL, ICE and IRAK antagonist, wherein the antagonist is a CJ-14877, CP-424174 (Pfizer), NF-61 (Negma-Lerads), and an analogue or derivative thereof.
2832. The device of item 2785 wherein the agent is an IL-2 inhibitor.
2833. The device of item 2785 wherein the agent is an immunosuppressant selected from the group consisting of teriflunomide (Sanofi Aventis), chlorsulfaquinoxalone (NSC-339004), chlorsulfaquinoxalone sulfate, CS-712 (Sankyo), ismomultin alfa (CAS No. 457913-93-8) (Akzo Nobel), antiallergics from GenPat77, antiinflammatories or AT-005 (Androclus Therapeutics), autoimmune disease therapeutics from EpiVax, BN-007 (Bone), budesonide (CAS No. 51333-22- 3) (MAP Pharmaceuticals), CO-14 (Genzyme), edratide (CAS No. 433922- 67-9) (Teva), EP-314 (Enanta), eprovafen (CAS No. 101335-99-3) (Sanofi- Aventis), HWA-131 (CAS No. 118788-41-3) (Sanofi-Aventis), immυnomodulators from MerLion Pharmaceuticals, immunosuppressives from Alchemia, IPL-12 (Inflazyme), MDL-9563 (CAS No. 27086-86-8) (Sanofi-Aventis), Pharmaprojects No. 2330 (Sanofi-Aventis), Pharmaprojects No. 6426 (Abgenix), PXS-25 (Pharmaxis), rosmarinic acid (CAS No. 20283-92-5) (Sanofi-Aventis), RP 42927 or RP 54745 (CAS No. 135330-08-4) (Sanofi-Aventis), SGN-35 (Seattle Genetics), ST-1959 (Sigma-Tau), type I diabetes therapy from SYNX Pharma, UNIL-88 (Debiopharm), VP-025 (Vasogen), VR-694 (Vectura), PRTX-001 (Protalex), and an analogue or derivative thereof.
2834. The device of item 2785 wherein the agent is an IMPDH (inosine monophosphate).
2835. The device of item 2785 wherein the agent is an integrin antagonist.
2836. The device of item 2785 wherein the agent is an interleukin antagonist.
2837. The device of item 2785 wherein the agent is an inhibitor of type III receptor tyrosine kinase.
2838. The device of item 2785 wherein the agent is an irreversible inhibitor of enzyme methionine aminopeptidase type 2.
2839. The device of item 2785 wherein the agent is an isozyme selective delta protein kinase C inhibitor. 2840. The device of item 2785 wherein the agent a JAK3 enzyme inhibitor.
2841. The device of item 2785 wherein the agent is a JNK inhibitor.
2842. The device of item 2785 wherein the agent is a kinase inhibitor.
2843. The device of item 2785 wherein the agent is kinesin antagonist.
2844. The device of item 2785 wherein the agent is a kinesin antagonist.
2845. The device of item 2785 wherein the agent is a leukotriene inhibitor and antagonist selected from the group consisting of ambicromil (CAS No. 58805-38-2) (Sanofi-Aventis), amelubant (CAS No. 346735-24-8) (Boehringer Ingelheim), DW-1141 (Dong Wha), ebselen (Daiichi Pharmaceutical), ibudilast (Kyorin), leucotriene inhibitors from Sanofi-Aventis, lymphotoxin-beta receptor (LT-D) from Biogen Idee, Pharmaprojects No. 1535 and 2728 (CAS No. 119340-33-9) (Sanofi- Aventis), R-112 (Rigel), Rev-5367 (CAS No. 92532-05-3) (Sanofi-Aventis), RG-14893 (CAS No. 141835-49-6) (Sanofi-Aventis), RG-5901-A (CAS No. 101910-24-1), 92532-23-5, RP 66153 (CAS No. 142422-79-5), RP 66364 (CAS No.186916-92-5), RP 69698 (CAS No. 141748-00-7) (Sanofi-Aventis), SC-411930 (Pfizer), SC-41930 (CAS No. 120072-59-5) (Pfizer), SC-50605 (CAS No. 138828-39-4) (Pfizer), SC-51146 (CAS No. 141059-52-1), SC- 53228 (CAS No. 153633-01-3) (Pfizer), spaglumic acid (ZY-15106) (CAS No.3106-85-2), 80619-64-3 (Novartis), tipredane (CAS No. 85197-77-9) (Bristol-Myers Squibb), U-75302 (CAS No. 119477-85-9) (Pfizer), and analogue or derivative thereof.
2846. The device of item 2785 wherein the agent is an MAP kinase inhibitor.
2847. The device of item 2785 wherein the agent is a matrix metalloproteinase inhibitor.
2848. The device of item 2785 wherein the agent is an MCP- CCR2 inhibitor.
2849. The device of item 2785 wherein the agent is an mTOR inhibitor.
2850. The device- of item 2785 wherein the agent is an mTOR kinase inhibitor.
2851. The device of item 2785 wherein the agent is a microtubule inhibitor selected from the group consisting of antibody- maytansinoid conjugates from Biogen Idee, colchicines (MantiCore Pharmaceuticals), anticancer immunoconjugates from Johnson & Johnson, DIME from Octamer, gni-1f (GNI), huC242-DM4, huMy9-6-DM1 (ImmunoGen), IDN-5404 (Indena), IMO-098, IMOderm (Imotep), mebendazole (Introgen Therapeutics), microtubule poisons from Cambridge Enterprise, paclitaxel such as LOTAX from Aphios (CAS No. 33069-62-4), Genexol-PM from Samyang, Pharmaprojects No. 6383 (Tapestry Pharmaceuticals), RPR-112378 (Sanofi-Aventis), SGN-75 (Seattle Genetics), SPL-7435 (Starpharma), SSR-250411 (Sanofi-Aventis), trastuzumab-DM1 (Genentech), vinorelbine, and an analogue or derivative thereof. 2852. The device of item 2785 wherein the agent is an MIF inhibitor.
2853. The device of item 2785 wherein the agent is an MMP inhibitor.
2854. The device of item 2785 wherein the agent is a neurokinin (NK) antagonist selected from the group consisting of anthrotainin (CAS No. 148084-40-6) (Sanofi-Aventis), an IBS thereapeutic from ArQuIe, MDL-105212A (CAS No. 167261-60-1) (Ssanofi-Aventis), Pharmaprojects No. 2744, 3258 (CAS No. 139167-47-8) 4006, 4201 , or 5986 (Sanofi-Aventis), RP 67580 (CAS No. 135911-02-3), SR-144190 (CAS No. 201152-86-5), SSR-240600, SSR-241586 (Sanofi-Aventis), TKA-457 (Novartis), vestipitant mesylate (CAS No. 334476-64-1) (GlaxoSmithKline), Win-64821 (Sanofi-Aventis), PRX-96026 (Predix Pharmaceuticals), and an analogue or derivative thereof.
2855. The device of item 2785 wherein the agent is an NF kappa B inhibitor selected from the group consisting of AVE-0545 or AVE- 0547 (Sanofi-Aventis), bortezomib (CAS No. 179324-69-7) (Millennium Pharmaceuticals), dexanabinol (CAS No. 112924-45-5) (Pharmos), dexlipotam (Viatris), Pharmaprojects No. 6283 (INDRA) (OXiGENE), IPL- 576092 (CAS No. 137571-30-3) (Inflazyme), NFKB decoy (Corgentech), NFKB decoy oligo (AnGes MG), NFKB's from Ariad, osteoporosis treatments or S5 (F005) from Fulcrum Pharmaceuticals, P61 (Phytopharm), R-fturbiprofen (CAS No. 5104-49-4) (Encore Pharmaceuticals), and an analogue or derivative thereof.
2856. The device of item 2785 wherein the agent is a nitric oxide agonist. 2857. The device of item 2785 wherein the agent is an ornithine decarboxylase inhibitor.
2858. The device of item 2785 wherein the agent is a p38 MAP kinase inhibitor selected from the group consisting of AZD-6703 (AstraZeneca), JX-401 (Jexys Pharmaceuticals), BMS-2 (Bristol-Myers Squibb), a p38 MAP kinase inhibitor from Novartis, a p38-alpha MAP kinase inhibitor from Amphora, Pharmaprojects No. 5704 (Pharmacopeia), RPR- 200765A (Sanofi-Aventis), SD-282 (Johnson & Johnson), TAK-715 (Takeda), and an analogue or derivative thereof.
2859. The device of item 2785 wherein the agent is a palmitoyl-protein thioesterase inhibitor.
2860. The device of item 2785 wherein the agent is a PDGF receptor kinase inhibitor selected- from thagroup consisting of AAL-993, AMN-107, or ABP-309 (Novartis), AMG-706 (Amgen), BAY-57-9352 (Bayer), CDP-860 (UCB), E-7080 (Eisai), imatinib (CAS No. 152459-95-5) (Novartis), OSI-930 (OSI Pharmaceuticals), RPR-127963E (Sanofi-Aventis), RWJ-540973 (Johnson & Johnson), sorafenib tosylate (Bayer), SU-11657 (Pfizer), tandutinib (CAS No. 387867-13-2) (Millennium Pharmaceuticals), vatalanib (Novartis), ZK-CDK (Schering AG), and an analogue or derivative thereof.
2861. The device of item 2785 wherein the agent is (-)- halofenate (Metabolex), AMG-131 (Amgen), antidiabetics from Japan Tobacco, AZD-4619, AZD-8450, AZD-8677 (AstraZeneca), DRF-10945, balaglitazone (Dr Reddy's), CS-00088, CS-00098 (Chipscreen Biosciences), E-3030 (Eisai), etalocib (CAS No. 161172-51-6) (EIi Lilly), GSK-641597 (Ligand), GSK-677954 (GlaxoSmithKline), GW-409544 (Ligand), GW- 590735 (GlaxoSmithKline), K-111 (Hoffmann-La Roche), LY-518674 (EIi Lilly), LY-674 (Ligand), LY-929 (Ligand), MC-3001 , MC-3002 (MaxoCore Pharmaceuticals), metformin HCl + pioglitazone (CAS No. 1115-70-4 and 112529-15-4), ACTOPLUS MET from Andrx), muraglitazar (CAS No. 331741-94-7) (Bristol-Myers Squibb), naveglitazar (Ligand), oleoylethanolamide (Kadmus Pharmaceuticals), ONO-5129, pioglitazone hydrochloride (CAS No. 111025-46-8 and 112529-15-4) (Takeda), PLX-204 (Plexxikon), PPAR agonists from Genfit, PPAR delta agonists from EIi Lilly, PPAR-alpha agonists from CrystalGenomics, PPAR-gamma modulators and PPAR-β modulators from CareX, rosiglitazone maleate (CAS No. 122324- 73-4 or 155141-29-0) (GlaxoSmithKline), rosiglitazone maleate/glimepir (CAS No. 155141-29-0 and 93479-97-1), AVANDARYL, rosiglitazone maleate/metformin extend (CAS No. 155141-29-0 and 657-24-9), AVANDAMET, rosiglitazone maleate+metformin, AVANDAMET (GlaxoSmithKline), tesaglitazar (AstraZeneca), and an analogue or derivative thereof.
2862. The device of item 2785 wherein the agent is a phosphatase inhibitor.
2863. The device of item 2785 wherein the agent is a phosphodiesterase (PDE) inhibitor selected from the group consisting of avanafil (Tanabe Seiyaku), dasantafil (CAS No. 569351-91-3) (Schering- Plough), A-906119 (CAS No. 134072-58-5), DL-850 (Sanofi-Aventis), GRC- 3015, GRC-3566, GRC-3886 (Glenmark), HWA-153 (CAS No. 56395-66-5) (Sanofi-Aventis), hydroxypumafentrine (Altana), IBFB-130011, IBFB-14-016, IBFB-140301 , IBFB-150007, IBFB-211913 (IBFB Pharma), L-826141 (Merck & Co), medorinone (CAS No. 88296-61-1) (Sanofi-Aventis), MEM- 1917 (Memory Pharmaceuticals), ND-1251 (Neuro3d), PDE inhibitors from ICOS1 PDE IV inhibitors from Memory Pharmaceuticals and CrystalGenomics, Pharmaprojects No. 2742 and 6141 (Sanofi-Aventis), QAD-171 (Novartis), RHC-2963 (CAS No. 76993-12-9 and 76993-14-1), RPR-117658, RPR-122818 derivatives, SR-24870 , and RPR-132294 (Sanofi-Aventis), SK-350 (ln2Gen), stroke therapy agents from deCODE Genetics, TAS-203 (Taiho), tofimilast (CAS No. 185954-27-2) (Pfizer), UK- 371800 (Pfizer), WIN-65579 (CAS No. 158020-82-7) (Sanofi-Aventis), IBFB- 130020 (IBFB Pharma), OPC-6535 (CAS No. 145739-56-6) (Otsuka), a phosphodiesterase III inhibitor, enoximone, a phosphodiesterase IV inhibitor, fosfosal, Atopik (Barrier Therapeutics), triflusal, a phosphodiesterase V inhibitor, and an analogue or derivative thereof.
2864. The device of item 2785 wherein the agent is a PKC inhibitor.
2865. The device of item 2785 wherein the agent is a platelet activating factor antagonist.
~ - 2866. The device of item 2785 wherein the agent is a platelet- derived growth factor receptor kinase inhibitor.
2867. The device of item 2785 wherein the agent is a prolyl hydroxylase inhibitor.
2868. The device of item 2785 wherein the agent is a polymorphonuclear neutrophil inhibitor.
2869. The device of item 2785 wherein the agent is a protein kinase B inhibitor.
2870. The device of item 2785 wherein the agent is a protein kinase C stimulant. 2871. The device of item 2785 wherein the agent is a purine nucleoside analogue.
2872. The device of item 2785 wherein the agent is a purinoreceptor P2X antagonist.
2873. The device of item 2785 wherein the agent is a Raf kinase inhibitor.
2874. The device of item 2785 wherein the agent is a reversible inhibitor of ErbB1 and ErbB2.
2875. The device of item 2785 wherein the agent is a ribonucleoside triphosphate reductase inhibitor.
2876. The device of iteiu 2785 wherein the agent is an SDF- 1 antagonist.
2877. The device of item 2785 wherein the agent is a sheddase inhibitor.
2878. The device of item 2785 wherein the agent is an SRC inhibitor.
2879. The device of item 2785 wherein the agent is a stromelysin inhibitor.
2880. The device of item 2785 wherein the agent is an Syk kinase inhibitor. 2881. The device of item 2785 wherein the agent is a telomerase inhibitor.
2882. The device of item 2785 wherein the agent is a TGF beta inhibitor selected from the group consisting of pirfenidone (CAS No. 53179-13-8) (MARNAC), tranilast (CAS No. 53902-12-8) (Kissei), IN-1130 (ln2Gen), mannose-6-phosphate (BTG), TGF-β antagonists from Inflazyme (Pharmaprojects No. 6075), TGF-β antagonists from Sydney, non-industrial source), TGF-β I receptor kinase inhibitors from EIi Lilly, TGF-β receptor inhibitors from Johnson & Johnson, and an analogue or derivative thereof.
2883. The device of item 2785 wherein the agent is a TNFα antagonist or TACE inhibitor selected from the group consisting of adalimumab (CAS No. 331731-18-1) (Cambridge Antibody Technology), AGIX-4207 (AtheroGenics), AGT-1 (Advanced Biotherapy), an antiinflammatory from Borean Pharma, Cellzome, or Paradigm Therapeutics, anti-inflammatory vaccine (TNF-alpha kinoid) from Neovacs, humanized anti-TNF antibody or an anti-TNF MAb (CB0006) Celltech (UCB), apratastat (CAS No. 287445-51-0) (Wyeth), BMS-561392 (Bristol-Myers Squibb), BN- 006 (Bone), certolizumab pegol (CAS No. 428863-50-7 or CH-138 (UCB), cilomilast (CAS No. 153259-65-5) (GlaxoSmithKline), CR-1 (Nuada Pharmaceuticals), CRx-119 (CombinatoRx), D-5410 (UCB), dacopafant (CAS No. 125372-33-0) (Sanofi-Aventis), dersalazine (CAS No. 188913-57- 7/188913-58-8) (Uriach), etanercept (CAS No. 185243-69-0) (Amgen), ethyl pyruvate (Critical (Critical Therapeutics), golimumab (CAS No. 476181-74-5) (Johnson & Johnson), hormono-immunotherapy from Ipsen, CDP571 (e.g., Humicade from UCB), IC-485 (ICOS), infliximab (CAS No. 170277-31-3) (Johnson & Johnson), IP-751 (Manhattan Pharmaceuticals), ISIS-104838 (CAS No. 250755-32-9) (ISIS Pharmaceuticals), lenalidomide (CAS No. 191732-72-6) (Celgene), lentinan (CAS No. 37339-90-5) (Ajinomoto), MDL- 201112 (CAS No. 142130-73-2) (Sanofi-Aventis), medroxyprogesterone (CAS No. 520-85-4) (InKine Pharmaceutical), N-acetylcysteine (CAS No. 616-91-1) (Zambon), NBE-P2 (DIREVO Biotech), nerelimomab (CAS No. 162774-06-3) (Chiron), OM-294DP (OM PHARMA), onercept (CAS No. 199685-57-9) (Yeda), PASSTNF-alpha (Verigen), pentoxifylline or oxypentifylline (Sanofi-Aventis), Pharmaprojects No. 4091 , 4241 , 4295, or 4488 (Sanofi-Aventis), Pharmaprojects No. 5480 (Amgen), Pharmaprojects No. 6749 (Cengent), pirfenidone (CAS No. 53179-13-8) (MARNAC), RPR- 132294 (Sanofi-Aventis), S5 (F002) (Fulcrum Pharmaceuticals), simvastatin (CAS No. 79902-63-9) (Merck & Co), STA-6292 (Synta Pharmaceuticals), tacrolimus (CAS No. 104987-11-3) (Fujisawa LifeCycle Pharma), talactoferrin alfa (CAS No. 308240-58-6) (Agennix), thalidomide (CAS No. 50-35-1) (Celgene), TNF antagonists form ProStrakan, and Synergen, TNF inhibitors (Amgen), TNF-alpha antagonists from Dynavax Technologies and Jerina AG (Germany), TNF-alpha inhibitors from IBFB Pharma and Xencor (Xencor), torbafylline (CAS No. 105102-21-4) (Sanofi-Aventis), UR-1505 (Uriach), VT-346 (Viron Therapeutics), YSIL6 (Y's Therapeutics), YSTH2 (Y's Therapeutics), NPI-1302a-3 (Nereus Pharmaceuticals, a TNF antagonist from Jerina AG (Germany), dersalazine, and an analogue or derivative thereof.
2884. The device of item 2785 wherein the agent is a Toll receptor inhibitor.
2885. The device of item 2785 wherein the agent is a tubulin antagonist.
2886. The device of item 2785 wherein the agent is a tyrosine kinase inhibitor selected from the group consisting of SU-011248, SUTENT from Pfizer Inc. (New York, NY), BMS-354825, PN-355 (Paracelsian Pharmaceuticals), AGN-199659 (Allergan), AAL-993 or ABP-309 (Novartis), adaphostin (NIH), AEE-788 (Novartis), AG-013736 (OSI Pharmaceuticals), AG-13736 (Pfizer), ALT-110 (Alteris Therapeutics), AMG-706 (Amgen), anticancer MAbs from Xencor, anti-EGFrvlll MAbs from Abgenix, anti-HER2 MAb from Abiogen, AZD-2171 or AZD-9935 (AstraZeneca), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), CEP-5214 (Cephalon), CEP- 7055 (Cephalon), cetuximab (ImCIone Systems), CHIR-200131 and CHIR- 258 (Chiron), CP-547632 (OSI Pharmaceuticals), CP-724714 (Pfizer), CT- 301 (Creabilis Therapeutics), D-69491 (Baxter International), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EGFR/ErbB2 inhibitors from Array BioPharma, erlotinib (CAS No. 183319-69-9) (OSI Pharmaceuticals), EXEL-2880 (Exelixis), FK-778 (Sanofi-Aventis) , gefitinib (CAS No. 184475-35-2) (AstraZeneca), GW-2286 or GW-654652 (GlaxoSmithKline), her2/neu antigen from AlphaVax, HER-2/neu inhibitor from Generex, Herzyme (Medipad) (Sima Therapeutics), HKI-272 (Wyeth), HuMax-EGFr (Genmab), idronoxil (CAS No. 81267-65-4) (Novogen), IGF-1 inhibitors from Ontogen, IMC-11F8 (ImCIone Systems), kahalalide F (CAS No. 149204-42-2) (PharmaMar), KDR. inhibitor from LG Life Sciences, KDR inhibitors from Abbott Laboratories, KDR kinase inhibitors (UCB), Kdr kinase inhibitors from Merck & Co, KRN-633 and KRN-951 (Kirin Brewery), KSB-102 (Xenova), lapatinib ditosylate (CAS No. 388082-78-8) (GlaxoSmithKline), matuzumab (Merck KGaA), MDX-214 (Medarex), ME-103 (Pharmexa), MED-A300 (Gerolymatos), MNAC-13 (Lay Line Genomics), nimotuzumab (Center of Molecular Immunology), NSC-330507 or NSC-707545 (NIH), NV-50 (Novogen), OSI-930 (OSI Pharmaceuticals), panitumumab (Abgenix), pelitinib (CAS No. 287933-82-7) (Wyeth), pertuzumab (CAS No. 380610-27- 5) (Genentech), Pharmaprojects No. 3985 (Sanofi-Aventis), prostate cancer therapeutics from Sequenom (SQPC35, SQPC36, SQPC90), removab and remoxab (Trion Pharma), RG-13022 (CAS No. 136831-48-6), RG-13291 (CAS No. 138989-50-1), or RG-14620 (CAS No. 136831-49-7) (Sanofi- Aventis), RM-6427 (Romark), RNAi breast cancer therapy from Benitec, RP 53801 (CAS No. 125882-88-4) (Sanofi-Aventis), sorafenib tosylate (Bayer), SU-11657 (Pfizer), Tie-2 antagonists from Semaia (Hybrigenics), Tie-2 inhibitors from Ontogen, trastuzumab (CAS No. 180288-69-1) (Genentech), tyrosine kinase inhibitors from Sanofi-Aventis, U3-1287, U3-1565, U3-1784, U3-1800 (U3 Pharma), vatalanib (Novartis), VEGFR-2 kinase inhibitor from Bristol-Myers Squibb, XL-647 (Exeiixis), ZD-6474 (AstraZeneca), ZK-CDK (Schering AG), an EGFR tyrosine kinase inhibitor, EKB-569 (Wyeth), and an analogue or derivative thereof.
2887. The device of item 2785 wherein the agent is a VEGF inhibitor.
2888. The device of item 2785 wherein the agent is a vitamin D receptor agonist.
2889. The device of item 2785 wherein the agent is ZD-6474 (an angiogenesis inhibitor).
2890. The device of item 2785 wherein the agent is AP-23573 (an mTOR inhibitor).
2891. The device of item 2785 wherein the agent is synthadotin (a tubulin antagonist).
2892. The device of item 2785 wherein the agent is S-0885 (a collagenase inhibitor).
2893. The device of item 2785 wherein the agent is aplidine (an elongation factor-1 alpha inhibitor).
2894. The device of item 2785 wherein the agent is ixabepilone (an epithilone). 2895. The device of item 2785 wherein the agent is IDN-5390 (an angiogenesis inhibitor).
2896. The device of item 2785 wherein the agent is SB- 2723005 (an angiogenesis inhibitor).
2897. The device of item 2785 wherein the agent is ABT-518 (an angiogenesis inhibitor).
2898. The device of item 2785 wherein the agent is combretastatin (an angiogenesis inhibitor).
2899. The device of item 2785 wherein the agent is anecortave acetate (an angiogenesis inhibitor).
2900. The device of item 2785 wherein the agent is SB- 715992 (a kinesin antagonist).
2901. The device of item 2785 wherein the agent is temsirolimus (an mTOR inhibitor).
2902. The device of item 2785 wherein the agent is adalimumab (a TNFα antagonist).
2903. The device of item 2785, further comprising a polymer.
2904. The device of item 2785 wherein the anti-scarring agent inhibits adhesion between the device and a host into which the device is implanted. 2905. The device of item 2785 wherein the device delivers the anti-scarring agent locally to tissue proximate to the device.
2906. The device of item 2785, further comprising a coating, wherein the coating comprises the anti-scarring agent.
2907. The device of item 2785, further comprising a coating, wherein the coating is disposed on a surface of the device.
2908. The device of item 2785, further comprising a coating, wherein the coating directly contacts the device.
2909. The device of item 2785, further comprising a coating, wherein the coating indirectly contacts the device.
2910. The device of item 2785,-further comprising a coating, wherein the coating partially covers the device.
2911. The device of item 2785, further comprising a coating, wherein the coating completely covers the device.
2912. The device of item 2785, further comprising a coating, wherein the coating is a uniform coating.
2913. The device of item 2785, further comprising a coating, wherein the coating is a non-uniform coating.
2914. The device of item 2785, further comprising a coating, wherein the coating is a discontinuous coating. 2915. The device of item 2785, further comprising a coating, wherein the coating is a patterned coating.
2916. The device of item 2785, further comprising a coating, wherein the coating has a thickness of 100 Dm or less.
2917. The device of item 2785, further comprising a coating, wherein the coating has a thickness of 10 Dm or less.
2918. The device of item 2785, further comprising a coating, wherein the coating adheres to the surface of the device upon deployment of the device.
2919. The device of item 2785, further comprising a coating, wherein the coating is stable at room temperature for a period of 1 year.
2920. The device of item 2785, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 0.0001% to about 1% by weight.
2921. The device of item 2785, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 1% to about 10% by weight.
2922. The device of item 2785, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 10% to about 25% by weight.
2923. The device of item 2785, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 25% to about 70% by weight. 2924. The device of item 2785, further comprising a coating, wherein the coating further comprises a polymer.
2925. The device of item 2785, further comprising a first coating having a first composition and the second coating having a second composition.
2926. The device of item 2785, further comprising a first coating having a first composition and the second coating having a second composition, wherein the first composition and the second composition are different.
2927. The device of item 2785, further comprising a polymeric carrier, wherein the polymeric carrier comprises a copolymer.
2928. The device of item 2785, further comprising a polymeric carrier, wherein the polymeric carrier comprises a block copolymer.
2929. The device of item 2785, further comprising a polymeric carrier, wherein the polymeric carrier comprises a random copolymer.
2930. The device of item 2785, further comprising a polymeric carrier, wherein the polymeric carrier comprises a biodegradable polymer.
2931. The device of item 2785, further comprising a polymeric carrier, wherein the polymeric carrier comprises a non-biodegradable polymer.
2932. The device of item 2785, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrophilic polymer. 2933. The device of item 2785, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrophobic polymer.
2934. The device of item 2785, further comprising a polymeric carrier, wherein the polymeric carrier comprises a polymer having hydrophilic domains.
2935. The device of item 2785, further comprising a polymeric carrier, wherein the polymeric carrier comprises a polymer having hydrophobic domains.
2936. The device of item 2785, further comprising a polymeric carrier, wherein the polymeric carrier comprises a non-conductive polymer.
2937. The device of item 2785, further comprising a polymeric carrier, wherein the polymeric carrier comprises an elastomer.
2938. The device of item 2785, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrogel.
2939. The device of item 2785, further comprising a polymeric carrier, wherein the polymeric carrier comprises a silicone polymer.
2940. The device of item 2785, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrocarbon polymer.
2941. The device of item 2785, further comprising a polymeric carrier, wherein the polymeric carrier comprises a styrene-derived polymer.
2942. The device of item 2785, further comprising a polymeric carrier, wherein the polymeric carrier comprises a butadiene polymer. 2943. The device of item 2785, further comprising a polymeric carrier, wherein the polymeric carrier comprises a macromer.
2944. The device of item 2785, further comprising a polymeric carrier, wherein the polymeric carrier comprises a poly(ethylene glycol) polymer.
2945. The device of item 2785, further comprising a polymeric carrier, wherein the polymeric carrier comprises an amorphous polymer.
2946. The device of item 2785, further comprising a lubricious coating.
2947. The device of item 2785 wherein the anti-scarring agent is located within pores or holes of the device.
2948. The device of item 2785 wherein the anti-scarring agent is located within a channel, lumen, or divet of the device.
2949. The device of item 2785, further comprising a second pharmaceutically active agent.
2950. The device of item 2785, further comprising an antiinflammatory agent.
2951. The device of item 2785, further comprising an agent that inhibits infection.
2952. The device of item 2785, further comprising an agent that inhibits infection, wherein the agent is an anthracycline. 2953. The device of item 2785, further comprising an agent that inhibits infection, wherein the agent is doxorubicin.
2954. The device of item 2785, further comprising an agent that inhibits infection, wherein the agent is mitoxantrone.
2955. The device of item 2785, further comprising an agent that inhibits infection, wherein the agent is a fluoropyrimidine.
2956. The device of item 2785, further comprising an agent that inhibits infection, wherein the agent is 5-fluorouracil (5-FU).
2957. The device of item 2785, further comprising an agent that inhibits infection, wherein the agent is a folic acid antagonist.
2958. The device of item 2785, further comprising an agent that inhibits infection, wherein the agent is methotrexate.
2959. The device of item 2785, further comprising an agent that inhibits infection, wherein the agent is a podophylotoxin.
2960. The device of item 2785, further comprising an agent that inhibits infection, wherein the agent is etoposide.
2961. The device of item 2785, further comprising an agent that inhibits infection, wherein the agent is a camptothecin.
2962. The device of item 2785, further comprising an agent that inhibits infection, wherein the agent is a hydroxyurea. 2963. The device of item 2785, further comprising an agent that inhibits infection, wherein the agent is a platinum complex.
2964. The device of item 2785, further comprising an agent that inhibits infection, wherein the agent is cisplatin.
2965. The device of item 2785, further comprising an antithrombotic agent.
2966. The device of item 2785, further comprising a visualization agent.
2967. The device of item 2785, further comprising a visualization agent, wherein the visualization agent is a radiopaque material, wherein the radiopaque material comprises a metal, a halogenated ^compound, or a barium containing compound.
2968. The device of item 2785, further comprising a visualization agent, wherein the visualization agent is a radiopaque material, wherein the radiopaque material comprises barium, tantalum, or technetium.
2969. The device of item 2785, further comprising a visualization agent, wherein the visualization agent is a MRI responsive material.
2970. The device of item 2785, further comprising a visualization agent, wherein the visualization agent comprises a gadolinium chelate. 2971. The device of item 2785, further comprising a visualization agent, wherein the visualization agent comprises iron, magnesium, manganese, copper, or chromium.
2972. The device of item 2785, further comprising a visualization agent, wherein the visualization agent comprises an iron oxide compound.
2973. The device of item 2785, further comprising a visualization agent, wherein the visualization agent comprises a dye, pigment, or colorant.
2974. The device of item 2785, further comprising an echogenic material.
._. 2975. The device of item 2785, further comprising an echogenic material, wherein the echogenic material is in the form of a coating.
2976. The device of item 2785 wherein the device is sterile.
2977. The device of item 2785 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device.
2978. The device of item 2785 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is connective tissue. 2979. The device of item 2785 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is muscle tissue.
2980. The device of item 2785 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is nerve tissue.
2981. The device of item 2785 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is epithelium tissue.
2982. The device of item 2785 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from the time of deployment of the device to about 1 year.
2983. The device of item 2785 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from about 1 month to 6 months.
2984. The device of item 2785 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from about 1 - 90 days.
2985. The device of item 2785 wherein the anti-scarring agent is released in effective concentrations from the device at a constant rate.
2986. The device of item 2785 wherein the anti-scarring agent is released in effective concentrations from the device at an increasing rate. 2987. The device of item 2785 wherein the anti-scarring agent is released in effective concentrations from the device at a decreasing rate.
2988. The device of item 2785 wherein the anti-scarring agent is released in effective concentrations from the composition comprising the anti-scarring agent by diffusion over a period ranging from the time of deployment of the device to about 90 days.
2989. The device of item 2785 wherein the anti-scarring agent is released in effective concentrations from the composition comprising the anti-scarring agent by erosion of the composition over a period ranging from the time of deployment of the device to about 90 days.
2990. The device of item 2785 wherein the device comprises about 0.01 Dg to about 10 Dg of the anti-scarring agent.
2991. The device of item 2785 wherein the device comprises about 10 Dg to about 10 mg of the anti-scarring agent.
2992. The device of item 2785 wherein the device comprises about 10 mg to about 250 mg of the anti-scarring agent.
2993. The device of item 2785 wherein the device comprises about 250 mg to about 1000 mg of the anti-scarring agent.
2994. The device of item 2785 wherein the device comprises about 1000 mg to about 2500 mg of the anti-scarring agent.
2995. The device of item 2785 wherein a surface of the device comprises less than 0.01 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied. 2996. The device of item 2785 wherein a surface of the device comprises about 0.01 Dg to about 1 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
2997. The device of item 2785 wherein a surface of the device comprises about 1 Dg to about 10 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
2998. The device of item 2785 wherein a surface of the device comprises about 10 Dg to about 250 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
2999. The device of item 2785 wherein a surface of the device comprises about 250 Dg to about 1000 Dg of the anti-scarring agent of anti- scarring agent per mm2 of device surface to which the anti-scarring agent is applied. - - . . . .. . . . .
3000. The device of item 2785 wherein a surface of the device comprises about 1000 Dg to about 2500 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
3001. A method as in any one of items 2785-3000, wherein the device is a glaucoma drainage device comprising a plate and a tube.
3002. A device, comprising a prosthetic heart valve or component thereof (e.g., an implant) and an anti-scarring agent or a composition comprising an anti-scarring agent, wherein the agent inhibits scarring between the device and a host into which the device is implanted.
3003. The device of item 3002 wherein the agent is an adensosine A2A receptor antagonist. 3004. The device of item 3002 wherein the agent is an AKT inhibitor.
3005. The device of item 3002 wherein the agent is an alpha 2 integrin antagonist, wherein the alpha 2 integrin antagonist is Pharmaprojects No. 5754 (Merck KgaA).
3006. The device of item 3002 wherein the agent an alpha 4 integrin antagonist.
3007. The device of item 3002 wherein the agent is an alpha 7 nicotinic receptor agonist.
3008. The device of item 3002 wherein the agent is an angiogenesis inhibitor selected from the group consisting of AG-12,958 (Pfizer), ATN-161 (Attenuon LLC), neovastat, an angiogenesis inhibitor from Jerina AG (Germany), NM-3 (Mercian), VGA-1155 (Taisho), FCE-26644 (Pfizer), FCE-26950 (Pfizer), FPMA (Meiji Daries), FR-111142 (Fujisawa), GGTI-298, GM-1306 (Ligand), GPA-1734 (Novartis), NNC-47-0011 (Novo Nordisk), herbamycin (Nippon Kayaku), lenalidomide (Celegene), IP-10 (NIH), ABT-828 (Abbott), KIN-841 (Tokushima University, Japan), SF-1126 (Semafore Pharmaceuticals), laminin technology (NIH), CHIR-258 (Chiron), NVP-AEW541 (Novartis), NVP-AEW541 (Novartis), Vt16907 (Alchemia), OXI-8007 (Oxigene), EG-3306 (Ark Therapeutics), Maspin (Arriva), ABT- 567 (Abbott), PPI-2458 (Praecis Pharmaceuticals), CC-5079, CC-4089 (Celgene), HIF-1 alpha inhibitors (Xenova), S-247 (Pfizer), AP-23573 (Ariad), AZD-9935 (Astra Zeneca), mebendazole (Introgen Therapeutics), MetAP-2 inhibitors (GlaxoSmithKline), AG-615 (Angiogene Pharmaceuticals), Tie-2 antagonists (Hybrigenics), NC-381 , CYC-381, NC-169, NC-219, NC-383, NC-384, NC-407 (Lorus Therapeutics), ATN-224 (Attenuon), ON-01370 (Onconova), Vitronectin antagonists (Amgen), SDX-103 (Salmedix), Vitronectin antagonists (Shire), CHP (Riemser), TEK (Amgen), Anecortave acetate (Alcon), T46.2 (Matrix Therapeutics), HG-2 (Heptagen), TEM antagonists (Genzyme), Oxi-4500 (Oxigene), ATN-161 (Attenuon), WX-293 (Wilex), M-2025 (Metris Therapeutics), Alphastatin (BioActa), YH-16 (Yantai Rongchang), BIBF-1120 (Boehringer Ingelheim), BAY-57-9352 (Bayer), AS- 1404 (Cancer Research Technology), SC-77964 (Pfizer), glycomimetics (BioTie Therapies), TIE-2 Inhibitors (Ontogen), DIMI, Octamer (Octamer), ABR-215050 (Active Biotech), ABT-518 (Abbott), KDR inhibitors (Abbott), BSF-466895 (Abbott), SCH-221153 (Schering-Plough), DAC:antiangiogenic (ConjuChem), TFPI (EntreMed), AZD-2171 (Astra-Zenaca), CDC-394 (Celgene), LY290293 (EIi Lilly), IDN-5390 (Indena), Kdr Kinase Inhibitors (Merck), CT-113020, CT-116433, CT-116563, CT-31890, CT-32228) (Cell Therapeutics), A-299620 (Abbott), TWEAK Inhibitor (Amgen), VEGF modulators (Johnson and Johnson), Tum-N53, tumstatin (Genzyme), Thios- 1 , Thios-2 (Thios Pharmaceuticals), MV-6401 (Miravant Medical Technologies), Spisulosine (PharmaMar),_CEP-7055 (Cephalon), AUV-20-1 (Auvation), LM-609 (EIi Lilly), SKF-106615 (AnorMED), Oglufanide disodium (Cytran), BW-114 (Pharminox), Calreticulin (NIH), WX-678 (Wilex), SD-7784 (Pfizer), WX-UK1 (Wilex), SH-268 (Schering AG), 2-Me-PGA (Celgene), S- 137 (Pfizer), ZD-6126 (Angiogene Pharmaceuticals), SG-292 (SignalGen), Benefin (Lane Labs), A6, A36 (Angstrom), SB-2723005 (GlaxoSmithKline), SC-7 (Cell Therapeutics), ZEN-014 (AEterna Zentaris), 2-methoxyestradiol (EntreMed), NK-130119 (Nippon Kayaku), CC-10004 (Celgene), AVE- 8062A (Ajinomoto), Tacedinaline (Pfizer), Actinonin (Tokyo Metropolitan Institute of Medical Science), Lenalidomide (Celgene), VGA-1155, BTO-956 (SRI International), ER-68203-00 (Eisai), CT-6685 (UCB), JKC-362 (Phoenix Pharmaceuticals), DMI-3798 (DMI Biosciences, Angiomate (Ipsen), ZD-6474 (AstraZeneca), CEP-5214 (Cephalon), Canstatin (Genzyme), NM-3 (Mercian), Oridigm (MediQuest Therapeutics), Exherin (Adherex), BLS-0597 (Boston Life Sciences), PTC-299 (PTC Therapeutics), NPI-2358 (Nereus Pharmaceuticals), CGP-79787 (Novartis), AEE-788 (Novartis), CKD-732 (Chong Kun Dang), CP-564959 (OSI Pharmaceuticals), CM-101 (CarboMed), CT-2584, CT3501 (Cell Therapeutics), combretastatin and analogues and derivatives thereof (Oxigene), Rebimastat (Bristol-Meyers Squibb), Dextrin 2-sulfate (ML Laboratories), Cilengitide (Merk KGaA), NSC-706704 (Pharminox), KRN- 951 (Kirin Brewery), Ukrain, NSC-631570 (Nowicky Pharma), Tecogalan sodium (Daiichi Pharmaceutical), Tz-93 (Tsumura), TBC-1635 (Encysive Pharmaceuticals), TAN-1120 (Takeda), Semaxanib (Pfizer), BDI-7800 (Biopharmacopae), SD-186, SD-983 (Bristol-Meyers Squibb), SB-223245 (GlaxoSmithKline), SC-236 (Pfizer), RWJ-590973 (Johnson and Johnson), ILX-1850 (Genzyme), SC-68488, S-836 (Pfizer), CG-55069-11 (CuraGen), Ki-23057 (Kirin Brewery), CCX-700 (Chemoentryx), Pegaptanib octasodium (Giled Sciences), ANGIOCOL (available from Biostratum Inc.), or an analogue or derivative thereof.
3009. The device of item 3002 wherein the agent is an apoptosis antagonist.
3010. The device of item 3002 wherein the agent is an apoptosis activator.
3011. The device of item 3002 wherein the agent is a beta 1 integrin antagonist.
3012. The device of item 3002 wherein the agent is a beta tubulin inhibitor.
3013. The device of item 3002 wherein the agent is a blocker of enzyme production in Hepatitis C. 3014. The device of item 3002 wherein the agent is a Bruton's tyrosine kinase inhibitor.
3015. The device of item 3002 wherein the agent is a calcineurin inhibitor.
3016. The device of item 3002 wherein the agent is a caspase 3 inhibitor.
3017. The device of item 3002 wherein the agent is a CC chemokine receptor antagonist.
3018. The device of item 3002 wherein the agent is a cell cycle inhibitor selected from the group consisting of SNS-595 (Sunesis), synthadotin, KRX-0403, and an analogue or derivative thereof.
3019. The device of item 3002 wherein the agent is a cathepsin B inhibitor.
3020. The device of item 3002 wherein the agent is a cathepsin K inhibitor, wherein the cathepsin K inhibitor is 462795 (GlaxoSmithKline), INPL-022-D6 (Amura Therapeutics), an analogue or derivative thereof).
3021. The device of item 3002 wherein the agent is a cathepsin L inhibitor.
3022. The device of item 3002 wherein the agent is a CD40 antagonist. 3023. The device of item 3002 wherein the agent is a chemokine receptor agonist.
3024. The device of item 3002 wherein the agent is a chymase inhibitor.
3025. The device of item 3002 wherein the agent is a collagenase antagonist.
3026. The device of item 3002 wherein the agent is a CXCR antagonist.
3027. The device of item 3002 wherein the agent is a cyclin dependent kinase inhibitor selected from the group consisting of a CDK-1 inhibitor, a CDK-2 inhibitor, a CDK- 4 inhibitor, a CDK-6 inhibitor, a CAK1 inhibitor from GPC Biotech and Bristol-Myers Squibb, RGB-286199 (GPC Biotech), an anticancer agent from Astex Technology, a CAK1 inhibitor from GPC Biotech, a CDK inhibitor from Sanofi-Aventis, a CDK1/CDK2 inhibitor from Amgen, a CDK2 inhibitor from SUGEN-2 (Pfizer), a hearing loss therapy agent (Sound Pharmaceuticals), PD-0332991 (Pfizer), RGB-286199 (GPC Biotech), Ro-0505124 (Hoffmann-La Roche), a Ser/Thr kinase inhibitor from Lilly (EIi Lilly), CVT-2584 (CAS No. 199986-75-9) (CV Therapeutics), and an analogue or derivative thereof.
3028. The device of item 3002 wherein the agent is a cyclooxygenase 1 inhibitor.
3029. The device of item 3002 wherein the agent is a DHFR inhibitor. 3030. The device of item 3002 wherein the agent is a dual integrin inhibitor.
3031. The device of item 3002 wherein the agent is an elastase inhibitor.
3032. The device of item 3002 wherein the agent is an elongation factor-1 alpha inhibitor.
3033. The device of item 3002 wherein the agent is an endothelial growth factor antagonist.
3034. The device of item 3002 wherein the agent is an endothelial growth factor receptor kinase inhibitor selected from the group consisting of sorafenib tosylate (Bayer), AAL-993 (Novartis), ABP-309 (Novartis), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), E- 7080 (Eisai), EG-3306 (Ark Therapeutics), EXEL-2880 (Exelixis), GW- 654652 (GlaxoSmithKline), a KDR inhibitor from LG Life Sciences, CT-6685 and CT-6729 (UCB), KRN-633 and KRN-951 (Kirin Brewery), OSI-930 (OSI Pharmaceuticals), SP-5.2 (Supratek Pharma), SU-11657 (Pfizer), a Tie-2 antagonist (Hybrigenics), a VEGFR-2 kinase inhibitor (Bristol-Myers Squibb), XL-647 (Exelixis), a KDR inhibitor from Abbott Laboratories, sorafenib tosylate, and an analogue or derivative thereof.
3035. The device of item 3002 wherein the agent is an endotoxin antagonist.
3036. The device of item 3002 wherein the agent is an epothilone and tubulin binder. 3037. The device of item 3002 wherein the agent is an estrogen receptor antagonist.
3038. The device of item 3002 wherein the agent is an FGF inhibitor.
3039. The device of item 3002 wherein the agent is a famexyl transferase inhibitor.
3040. The device of item 3002 wherein the agent is farnesyltransferase inhibitor selected from the group of A-197574 (Abbott), a famesyltransferase inhibitor from Servier, FPTIII (Strathclyde Institute for Drug R), LB-42908 (LG Life Sciences), Pharmaprojects No. 5063 (Genzyme), Pharmaprojects No. 5597 (Ipsen), Yissum Project No. B-1055 (Yissum), and an analogue or derivative thereof.
3041. The device of item 3002 wherein the agent is an FLT-3 kinase inhibitor.
3042. The device of item 3002 wherein the agent is an FGF receptor kinase inhibitor.
3043. The device of item 3002 wherein the agent is a fibrinogen antagonist selected from the group consisting of AUV-201 (Auvation), MG-13926 (Sanofi-Aventis), plasminogen activator (CAS No. 105913-11-9) (from Sanofi-Aventis or UCB), plasminogen activator-2 (tPA- 2) (Sanofi-Aventis), pro-urokinase (CAS No. 82657-92-9) (Sanofi-Aventis), and an analogue or derivative thereof.
3044. The device of item 3002 wherein the agent is a heat shock protein 90 antagonist selected from the group consisting of SRN-005 (Sirenade), geldanamycin, NSC-33050 (17-aIlylaminogeldanamycin; 17- AAG), ^-dimethylaminoethylamino-^-demethoxy-geldanamycin (17- DMAG), rifabutin (rifamycin XIV, 1',4-didehydro-1-deoxy-1 ,4-dihydro-5'-(2- methylpropyl)-1-oxo-), and an analogue or derivative thereof.
3045. The device of item 3002 wherein the agent is a histone deacetylase inhibitor.
3046. The device of item 3002 wherein the agent is an HMGCoA reductase inhibitor selected from the group consisting of an atherosclerosis therapeutic from Lipid Sciences, ATI-16000 (ARYx Therapeutics), KS-01-019 (Kos Pharmaceuticals), Pharmaprojects No. 2197 (Sanofi-Aventi), RP 61969 (Sanofi-Aventis), and an analogue or derivative thereof.
3047. The device of item 3002 wherein the agent is an ICAM inhibitor.
3048. The device of item 3002 wherein the agent is an IL, ICE and IRAK antagonist, wherein the antagonist is a CJ-14877, CP-424174 (Pfizer), NF-61 (Negma-Lerads), and an analogue or derivative thereof.
3049. The device of item 3002 wherein the agent is an IL-2 inhibitor.
3050. The device of item 3002 wherein the agent is an immunosuppressant selected from the group consisting of teriflunomide (Sanofi Aventis), chlorsulfaquinoxalone (NSC-339004), chlorsulfaquinoxalone sulfate, CS-712 (Sankyo), ismomultin alfa (CAS No. 457913-93-8) (Akzo Nobel), antiallergics from GenPat77, antiinflammatories or AT-005 (Androcius Therapeutics), autoimmune disease therapeutics from EpiVax, BN-007 (Bone), budesonide (CAS No. 51333-22- 3) (MAP Pharmaceuticals), CO-14 (Genzyme), edratide (CAS No. 433922- 67-9) (Teva), EP-314 (Enanta), eprovafen (CAS No. 101335-99-3) (Sanofi- Aventis), HWA-131 (CAS No. 118788-41-3) (Sanofi-Aventis), immunomodulators from MerLion Pharmaceuticals, immunosuppressives from Alchemia, IPL-12 (Inflazyme), MDL-9563 (CAS No. 27086-86-8) (Sanofi-Aventis), Pharmaprojects No. 2330 (Sanofi-Aventis), Pharmaprojects No. 6426 (Abgenix), PXS-25 (Pharmaxis), rosmarinic acid (CAS No. 20283-92-5) (Sanofi-Aventis), RP 42927 or RP 54745 (CAS No. 135330-08-4) (Sanofi-Aventis), SGN-35 (Seattle Genetics), ST-1959 (Sigma-Tau), type I diabetes therapy from SYNX Pharma, UNIL-88 (Debiopharm), VP-025 (Vasogen), VR-694 (Vectura), PRTX-001 (Protalex), and an analogue or derivative thereof.
3051. The device of item 3002 wherein the agent is an IMPDH (inosine monophosphate), - -- -
3052. The device of item 3002 wherein the agent is an integrin antagonist.
3053. The device of item 3002 wherein the agent is an interleukin antagonist.
3054. The device of item 3002 wherein the agent is an inhibitor of type III receptor tyrosine kinase.
3055. The device of item 3002 wherein the agent is an irreversible inhibitor of enzyme methionine aminopeptidase type 2.
3056. The device of item 3002 wherein the agent is an isozyme selective delta protein kinase C inhibitor. 3057. The device of item 3002 wherein the agent a JAK3 enzyme inhibitor.
3058. The device of item 3002 wherein the agent is a JNK inhibitor.
3059. The device of item 3002 wherein the agent is a kinase inhibitor.
3060. The device of item 3002 wherein the agent is kinesin antagonist.
3061. The device of item 3002 wherein the agent is a kinesin antagonist.
3062. The device of item 3002 wherein the agent is a leukotriene inhibitor and antagonist selected from the group consisting of ambicromil (CAS No. 58805-38-2) (Sanofi-Aventis), amelubant (CAS No. 346735-24-8) (Boehringer Ingelheim), DW-1141 (Dong Wha), ebselen (Daiichi Pharmaceutical), ibudilast (Kyorin), leucotriene inhibitors from Sanofi-Aventis, lymphotoxin-beta receptor (LT-D) from Biogen Idee, Pharmaprojects No. 1535 and 2728 (CAS No. 119340-33-9) (Sanofi- Aventis), R-112 (Rigel), Rev-5367 (CAS No. 92532-05-3) (Sanofi-Aventis), RG-14893 (CAS No. 141835-49-6) (Sanofi-Aventis), RG-5901-A (CAS No. 101910-24-1), 92532-23-5, RP 66153 (CAS No. 142422-79-5), RP 66364 (CAS No.186916-92-5), RP 69698 (CAS No. 141748-00-7) (Sanofi-Aventis), SC-411930 (Pfizer), SC-41930 (CAS No. 120072-59-5) (Pfizer), SC-50605 (CAS No. 138828-39-4) (Pfizer), SC-51146 (CAS No. 141059-52-1), SC- 53228 (CAS No. 153633-01-3) (Pfizer), spaglumic acid (ZY-15106) (CAS No.3106-85-2), 80619-64-3 (Novartis), tipredane (CAS No. 85197-77-9) (Bristol-Myers Squibb), U-75302 (CAS No. 119477-85-9) (Pfizer), and analogue or derivative thereof.
3063. The device of item 3002 wherein the agent is an MAP kinase inhibitor.
3064. The device of item 3002 wherein the agent is a matrix metalloproteinase inhibitor.
3065. The device of item 3002 wherein the agent is an MCP- CCR2 inhibitor.
3066. The device of item 3002 wherein the agent is an mTOR inhibitor.
3067. Thexlevice of item 3002 wherein the agent is an mTOR kinase inhibitor.
3068. The device of item 3002 wherein the agent is a microtubule inhibitor selected from the group consisting of antibody- maytansinoid conjugates from Biogen Idee, colchicines (MantiCore Pharmaceuticals), anticancer immunoconjugates from Johnson & Johnson, DIME from Octamer, gni-1f (GNI), huC242-DM4, huMy9-6-DM1 (ImmunoGen), IDN-5404 (Indena), IMO-098, IMOderm (Imotep), mebendazole (Introgen Therapeutics), microtubule poisons from Cambridge Enterprise, paclitaxel such as LOTAX from Aphios (CAS No. 33069-62-4), Genexol-PM from Samyang, Pharmaprojects No. 6383 (Tapestry Pharmaceuticals), RPR-112378 (Sanofi-Aventis), SGN-75 (Seattle Genetics), SPL-7435 (Starpharma), SSR-250411 (Sanofi-Aventis), trastuzumab-DM1 (Genentech), vinorelbine, and an analogue or derivative thereof. 3069. The device of item 3002 wherein the agent is an MIF inhibitor.
3070. The device of item 3002 wherein the agent is an MMP inhibitor.
3071. The device of item 3002 wherein the agent is a neurokinin (NK) antagonist selected from the group consisting of anthrotainin (CAS No. 148084-40-6) (Sanofi-Aventis), an IBS thereapeutic from ArQuIe, MDL-105212A (CAS No. 167261-60-1) (Ssanofi-Aventis), Pharmaprojects No. 2744, 3258 (CAS No. 139167-47-8) 4006, 4201, or 5986 (Sanofi-Aventis), RP 67580 (CAS No. 135911-02-3), SR-144190 (CAS No. 201152-86-5), SSR-240600, SSR-241586 (Sanofi-Aventis), TKA-457 (Novartis), vestipitant mesylate (CAS No. 334476-64-1) (GlaxoSmithKline), Win-64821 (Sanofi-Aventis), PRX-96026 (Predix Pharmaceuticals), and an analogue or derivative thereof.
3072. The device of item 3002 wherein the agent is an NF kappa B inhibitor selected from the group consisting of AVE-0545 or AVE- 0547 (Sanofi-Aventis), bortezomib (CAS No. 179324-69-7) (Millennium Pharmaceuticals), dexanabinol (CAS No. 112924-45-5) (Pharmos), dexlipotam (Viatris), Pharmaprojects No. 6283 (INDRA) (OXiGENE), IPL- 576092 (CAS No. 137571-30-3) (Inflazyme), NFKB decoy (Corgentech), NFKB decoy oligo (AnGes MG), NFKB's from Ariad, osteoporosis treatments or S5 (F005) from Fulcrum Pharmaceuticals, P61 (Phytopharm), R-flurbiprofen (CAS No. 5104-49-4) (Encore Pharmaceuticals), and an analogue or derivative thereof.
3073. The device of item 3002 wherein the agent is a nitric oxide agonist. 3074. The device of item 3002 wherein the agent is an ornithine decarboxylase inhibitor.
3075. The device of item 3002 wherein the agent is a p38 MAP kinase inhibitor selected from the group consisting of AZD-6703 (AstraZeneca), JX-401 (Jexys Pharmaceuticals), BMS-2 (Bristol-Myers Squibb), a p38 MAP kinase inhibitor from Novartis, a p38-alpha MAP kinase inhibitor from Amphora, Pharmaprojects No. 5704 (Pharmacopeia), RPR- 200765A (Sanofi-Aventis), SD-282 (Johnson & Johnson), TAK-715 (Takeda), and an analogue or derivative thereof.
3076. The device of item 3002 wherein the agent is a palmitoyl-protein thioesterase inhibitor.
3077. The device of item 3002 wherein the agent is a PDGF receptor kinase inhibitor selected from the group consisting of AAL-993, AMN-107, or ABP-309 (Novartis), AMG-706 (Amgen), BAY-57-9352 (Bayer), CDP-860 (UCB), E-7080 (Eisai), imatinib (CAS No. 152459-95-5) (Novartis), OSI-930 (OSI Pharmaceuticals), RPR-127963E (Sanofi-Aventis), RWJ-540973 (Johnson & Johnson), sorafenib tosylate (Bayer), SU-11657 (Pfizer), tandutinib (CAS No. 387867-13-2) (Millennium Pharmaceuticals), vatalanib (Novartis), ZK-CDK (Schering AG), and an analogue or derivative thereof.
3078. The device of item 3002 wherein the agent is (-)- halofenate (Metabolex), AMG-131 (Amgen), antidiabetics from Japan Tobacco, AZD-4619, AZD-8450, AZD-8677 (AstraZeneca), DRF-10945, balaglitazone (Dr Reddy's), CS-00088, CS-00098 (Chipscreen Biosciences), E-3030 (Eisai), etalocib (CAS No. 161172-51-6) (EIi Lilly), GSK-641597 (Ligand), GSK-677954 (GlaxoSmithKline), GW-409544 (Ligand), GW- 590735 (GlaxoSmithKline), K-111 (Hoffmann-La Roche), LY-518674 (EIi Lilly), LY-674 (Ligand), LY-929 (Ligand), MC-3001 , MC-3002 (MaxoCore Pharmaceuticals), metformin HCI + pioglitazone (CAS No. 1115-70-4 and 112529-15-4), ACTOPLUS MET from Andrx), muraglitazar (CAS No. 331741-94-7) (Bristol-Myers Squibb), naveglitazar (Ligand), oleoylethanolamide (Kadmus Pharmaceuticals), ONO-5129, pioglitazone hydrochloride (CAS No. 111025-46-8 and 112529-15-4) (Takeda), PLX-204 (Plexxikon), PPAR agonists from Genfit, PPAR delta agonists from EIi Lilly, PPAR-alpha agonists from CrystalGenomics, PPAR-gamma modulators and PPAR-β modulators from CareX, rosiglitazone maleate (CAS No. 122324- 73-4 or 155141-29-0) (GlaxoSmithKline), rosiglitazone maleate/glimepir (CAS No. 155141-29-0 and 93479-97-1), AVANDARYL, rosiglitazone maleate/metformin extend (CAS No. 155141-29-0 and 657-24-9), AVANDAMET, rosiglitazone maleate+metformin, AVANDAMET (GlaxoSmithKline), tesaglitazar (AstraZeneca), and an analogue or derivative thereof.
3079. The device of item 3002 wherein the agent is a phosphatase inhibitor.
3080. The device of item 3002 wherein the agent is a phosphodiesterase (PDE) inhibitor selected from the group consisting of avanafil (Tanabe Seiyaku), dasantafil (CAS No. 569351-91-3) (Schering- Plough), A-906119 (CAS No. 134072-58-5), DL-850 (Sanofi-Aventis), GRC- 3015, GRC-3566, GRC-3886 (Glenmark), HWA-153 (CAS No. 56395-66-5) (Sanofi-Aventis), hydroxypumafentrine (Altana), IBFB-130011 , IBFB-14-016, IBFB-140301 , IBFB-150007, IBFB-211913 (IBFB Pharma), L-826141 (Merck & Co), medorinone (CAS No. 88296-61-1) (Sanofi-Aventis), MEM- 1917 (Memory Pharmaceuticals), ND-1251 (Neuro3d), PDE inhibitors from ICOS, PDE IV inhibitors from Memory Pharmaceuticals and CrystalGenomics, Pharmaprojects No. 2742 and 6141 (Sanofi-Aventis), QAD-171 (Novartis), RHC-2963 (CAS No. 76993-12-9 and 76993-14-1), RPR-117658, RPR-122818 derivatives, SR-24870 , and RPR-132294 (Sanofi-Aventis), SK-350 (ln2Gen), stroke therapy agents from deCODE Genetics, TAS-203 (Taiho), tofimilast (CAS No. 185954-27-2) (Pfizer), UK- 371800 (Pfizer), WIN-65579 (CAS No. 158020-82-7) (Sanofi-Aventis), IBFB- 130020 (IBFB Pharma), OPC-6535 (CAS No. 145739-56-6) (Otsuka), a phosphodiesterase III inhibitor, enoximone, a phosphodiesterase IV inhibitor, fosfosal, Atopik (Barrier Therapeutics), triflusal, a phosphodiesterase V inhibitor, and an analogue or derivative thereof.
3081. The device of item 3002 wherein the agent is a PKC inhibitor.
3082. The device of item 3002 wherein the agent is a platelet activating factor antagonist.
- - 3083. The device of item 3002 wherein the agent is a platelet- derived growth factor receptor kinase inhibitor.
3084. The device of item 3002 wherein the agent is a prolyl hydroxylase inhibitor.
3085. The device of item 3002 wherein the agent is a polymorphonuclear neutrophil inhibitor.
3086. The device of item 3002 wherein the agent is a protein kinase B inhibitor.
3087. The device of item 3002 wherein the agent is a protein kinase C stimulant. 3088. The device of item 3002 wherein the agent is a purine nucleoside analogue.
3089. The device of item 3002 wherein the agent is a purinoreceptor P2X antagonist.
3090. The device of item 3002 wherein the agent is a Raf kinase inhibitor.
3091. The device of item 3002 wherein the agent is a reversible inhibitor of ErbB1 and ErbB2.
3092. The device of item 3002 wherein the agent is a ribonucleoside triphosphate reductase inhibitor.
3093. The device of-item 3002 wherein the agent is an SDF-1 antagonist.
3094. The device of item 3002 wherein the agent is a sheddase inhibitor.
3095. The device of item 3002 wherein the agent is an SRC inhibitor.
3096. The device of item 3002 wherein the agent is a stromelysin inhibitor.
3097. The device of item 3002 wherein the agent is an Syk kinase inhibitor. 3098. The device of item 3002 wherein the agent is a telomerase inhibitor.
3099. The device of item 3002 wherein the agent is a TGF beta inhibitor selected from the group consisting of pirfenidone (CAS No. 53179-13-8) (MARNAC), tranilast (CAS No. 53902-12-8) (Kissei), IN-1130 (ln2Gen), mannose-6-phosphate (BTG), TGF-β antagonists from Inflazyme (Pharmaprojects No. 6075), TGF-β antagonists from Sydney, non-industrial source), TGF-β I receptor kinase inhibitors from EIi Lilly, TGF-β receptor inhibitors from Johnson & Johnson, and an analogue or derivative thereof.
3100. The device of item 3002 wherein the agent is a TNFα antagonist or TACE inhibitor selected from the group consisting of adalimumab (CAS No. 331731-18-1) (Cambridge Antibody Technology), AGIX-4207 (AtheroGenics), AGT-1 (Advanced Biotherapy), an antiinflammatory from Borean-Pharma, Ceilzome, or Paradigm Therapeutics, anti-inflammatory vaccine (TNF-alpha kinoid) from Neovacs, humanized anti-TNF antibody or an anti-TNF MAb (CB0006) Celltech (UCB), apratastat (CAS No. 287445-51-0) (Wyeth), BMS-561392 (Bristol-Myers Squibb), BN- 006 (Bone), certolizumab pegol (CAS No. 428863-50-7 or CH-138 (UCB), cilomilast (CAS No. 153259-65-5) (GlaxoSmithKline), CR-1 (Nuada Pharmaceuticals), CRx-119 (CombinatoRx), D-5410 (UCB), dacopafant (CAS No. 125372-33-0) (Sanofi-Aventis), dersalazine (CAS No. 188913-57- 7/188913-58-8) (Uriach), etanercept (CAS No. 185243-69-0) (Amgen), ethyl pyruvate (Critical (Critical Therapeutics), golimumab (CAS No. 476181-74-5) (Johnson & Johnson), hormono-immunotherapy from Ipsen, CDP571 (e.g., Humicade from UCB), IC-485 (ICOS), infliximab (CAS No. 170277-31-3) (Johnson & Johnson), IP-751 (Manhattan Pharmaceuticals), ISIS-104838 (CAS No. 250755-32-9) (ISIS Pharmaceuticals), lenalidomide (CAS No. 191732-72-6) (Celgene), lentinan (CAS No. 37339-90-5) (Ajinomoto), MDL- 201112 (CAS No. 142130-73-2) (Sanofi-Aventis), medroxyprogesterone (CAS No. 520-85-4) (InKine Pharmaceutical), N-acetylcysteine (CAS No. 616-91-1) (Zambon), NBE-P2 (DIREVO Biotech), nerelimomab (CAS No. 162774-06-3) (Chiron), OM-294DP (OM PHARMA), onercept (CAS No. 199685-57-9) (Yeda), PASSTNF-alpha (Verigen), pentoxifylline or oxypentifylline (Sanofi-Aventis), Pharmaprojects No. 4091 , 4241 , 4295, or 4488 (Sanofi-Aventis), Pharmaprojects No. 5480 (Amgen), Pharmaprojects No. 6749 (Cengent), pirfenidone (CAS No. 53179-13-8) (MARNAC), RPR- 132294 (Sanofi-Aventis), S5 (F002) (Fulcrum Pharmaceuticals), simvastatin (CAS No. 79902-63-9) (Merck & Co), STA-6292 (Synta Pharmaceuticals), tacrolimus (CAS No. 104987-11-3) (Fujisawa LifeCycle Pharma), talactoferrin alfa (CAS No. 308240-58-6) (Agennix), thalidomide (CAS No. 50-35-1) (Celgene), TNF antagonists form ProStrakan, and Synergen, TNF inhibitors (Amgen), TNF-alpha antagonists from Dynavax Technologies and Jerina AG (Germany), TNF-alpha inhibitors from IBFB Pharma and Xencor (Xencor), torbafylline (CAS No. 105102-21-4) (Sanofi-Aventis), UR-1505 (Uriach), VT-346 (Viron Therapeutics), YSIL6 (Y1S Therapeutics), YSTH2 (Y's Therapeutics), NPI-1302a-3 (Nereus Pharmaceuticals, a TNF antagonist from Jerina AG (Germany), dersalazine, and an analogue or derivative thereof.
3101. The device of item 3002 wherein the agent is a Toll receptor inhibitor.
3102. The device of item 3002 wherein the agent is a tubulin antagonist.
3103. The device of item 3002 wherein the agent is a tyrosine kinase inhibitor selected from the group consisting of SU-011248, SUTENT from Pfizer Inc. (New York, NY), BMS-354825, PN-355 (Paracelsian Pharmaceuticals), AGN-199659 (Allergan), AAL-993 or ABP-309 (Novartis), adaphostin (NIH), AEE-788 (Novartis), AG-013736 (OSI Pharmaceuticals), AG-13736 (Pfizer), ALT-110 (Alteris Therapeutics), AMG-706 (Amgen), anticancer MAbs from Xencor, anti-EGFrvlll MAbs from Abgenix, anti-HER2 MAb from Abiogen, AZD-2171 or AZD-9935 (AstraZeneca), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), CEP-5214 (Cephalon), CEP- 7055 (Cephalon), cetuximab (ImCIone Systems), CHIR-200131 and CHIR- 258 (Chiron), CP-547632 (OSI Pharmaceuticals), CP-724714 (Pfizer), CT- 301 (Creabilis Therapeutics), D-69491 (Baxter International), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EGFR/ErbB2 inhibitors from Array BioPharma, erlotinib (CAS No. 183319-69-9) (OSI Pharmaceuticals), EXEL-2880 (Exelixis), FK-778 (Sanofi-Aventis) , gefitinib (CAS No. 184475-35-2) (AstraZeneca), GW-2286 or GW-654652 (GlaxoSmithKline), her2/neu antigen from AlphaVax, HER-2/neu inhibitor from Generex, Herzyme (Medipad) (Sima Therapeutics), HKI-272 (Wyeth), HuMax-EGFr (Genmab), idronoxil (CAS No. 81267-65-4) (Novogen), IGF-1 inhibitors from Ontogen, IMC-11 F8 (ImCIone Systems), kahalalide F (CAS No. 149204-42-2) (PharmaMar); KDR inhibitor from LG Life Sciences, KDR inhibitors from Abbott Laboratories, KDR kinase inhibitors (UCB), Kdr kinase inhibitors from Merck & Co, KRN-633 and KRN-951 (Kirin Brewery), KSB-102 (Xenova), lapatinib ditosylate (CAS No. 388082-78-8) (GlaxoSmithKline), matuzumab (Merck KGaA), MDX-214 (Medarex), ME-103 (Pharmexa), MED-A300 (Gerolymatos), MNAC-13 (Lay Line Genomics), nimotuzumab (Center of Molecular Immunology), NSC-330507 or NSC-707545 (NIH), NV-50 (Novogen), OSI-930 (OSI Pharmaceuticals), panitumumab (Abgenix), pelitinib (CAS No. 287933-82-7) (Wyeth), pertuzumab (CAS No. 380610-27- 5) (Genentech), Pharmaprojects No. 3985 (Sanofi-Aventis), prostate cancer therapeutics from Sequenom (SQPC35, SQPC36, SQPC90), removab and remoxab (Trion Pharma), RG-13022 (CAS No. 136831-48-6), RG-13291 (CAS No. 138989-50-1), or RG-14620 (CAS No. 136831-49-7) (Sanofi- Aventis), RM-6427 (Romark), RNAi breast cancer therapy from Benitec, RP 53801 (CAS No. 125882-88-4) (Sanofi-Aventis), sorafenib tosylate (Bayer), SU-11657 (Pfizer), Tie-2 antagonists from Semaia (Hybrigenics), Tie-2 inhibitors from Ontogen, trastuzumab (CAS No. 180288-69-1) (Genentech), tyrosine kinase inhibitors from Sanofi-Aventis, U3-1287, U3-1565, U3-1784, U3-1800 (U3 Pharma), vatalanib (Novartis), VEGFR-2 kinase inhibitor from Bristol-Myers Squibb, XL-647 (Exelixis), ZD-6474 (AstraZeneca), ZK-CDK (Schering AG), an EGFR tyrosine kinase inhibitor, EKB-569 (Wyeth), and an analogue or derivative thereof.
3104. The device of item 3002 wherein the agent is a VEGF inhibitor.
3105. The device of item 3002 wherein the agent is a vitamin D receptor agonist.
3106. The device of item 3002 wherein the agent is ZD-6474 (an angiogenesis inhibitor).
3107. The device of item 3002 wherein the agent is AP-23573 (an mTOR inhibitor).
3108. The device of item 3002 wherein the agent is synthadotin (a tubulin antagonist).
3109. The device of item 3002 wherein the agent is S-0885 (a collagenase inhibitor).
3110. The device of item 3002 wherein the agent is aplidine (an elongation factor-1 alpha inhibitor).
3111. The device of item 3002 wherein the agent is ixabepilone (an epithilone). 3112. The device of item 3002 wherein the agent is IDN-5390 (an angiogenesis inhibitor).
3113. The device of item 3002 wherein the agent is SB- 2723005 (an angiogenesis inhibitor).
3114. The device of item 3002 wherein the agent is ABT-518 (an angiogenesis inhibitor).
3115. The device of item 3002 wherein the agent is combretastatin (an angiogenesis inhibitor).
3116. The device of item 3002 wherein the agent is anecortave acetate (an angiogenesis inhibitor).
3117. The device of item 3002 wherein the-agent is SB- 715992 (a kinesin antagonist).
3118. The device of item 3002 wherein the agent is temsirolimus (an mTOR inhibitor).
3119. The device of item 3002 wherein the agent is adalimumab (a TNFα antagonist).
3120. The device of item 3002, further comprising a polymer.
3121. The device of item 3002 wherein the anti-scarring agent inhibits adhesion between the device and a host into which the device is implanted. 3122. The device of item 3002 wherein the device delivers the anti-scarring agent locally to tissue proximate to the device.
3123. The device of item 3002, further comprising a coating, wherein the coating comprises the anti-scarring agent.
3124. The device of item 3002, further comprising a coating, wherein the coating is disposed on a surface of the device.
3125. The device of item 3002, further comprising a coating, wherein the coating directly contacts the device.
3126. The device of item 3002, further comprising a coating, wherein the coating indirectly contacts the device.
3127. The device of item 3002, further comprising a coating, wherein the coating partially covers the device.
3128. The device of item 3002, further comprising a coating, wherein the coating completely covers the device.
3129. The device of item 3002, further comprising a coating, wherein the coating is a uniform coating.
3130. The device of item 3002, further comprising a coating, wherein the coating is a non-uniform coating.
3131. The device of item 3002, further comprising a coating, wherein the coating is a discontinuous coating. 3132. The device of item 3002, further comprising a coating, wherein the coating is a patterned coating.
3133. The device of item 3002, further comprising a coating, wherein the coating has a thickness of 100 Dm or less.
3134. The device of item 3002, further comprising a coating, wherein the coating has a thickness of 10 Dm or less.
3135. The device of item 3002, further comprising a coating, wherein the coating adheres to the surface of the device upon deployment of the device.
3136. The device of item 3002, further comprising a coating, wherein the coating is stable at room temperature for a period of 1 year.
3137. The device of item 3002, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 0.0001% to about 1% by weight.
3138. The device of item 3002, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 1% to about 10% by weight.
3139. The device of item 3002, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 10% to about 25% by weight.
3140. The device of item 3002, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 25% to about 70% by weight. 3141. The device of item 3002, further comprising a coating, wherein the coating further comprises a polymer.
3142. The device of item 3002, further comprising a first coating having a first composition and the second coating having a second composition.
3143. The device of item 3002, further comprising a first coating having a first composition and the second coating having a second composition, wherein the first composition and the second composition are different.
3144. The device of item 3002, further comprising a polymeric carrier, wherein the polymeric carrier comprises a copolymer.
- _. _ .- - 3145. The device of item 3002, further comprising a polymeric carrier, wherein the polymeric carrier comprises a block copolymer.
3146. The device of item 3002, further comprising a polymeric carrier, wherein the polymeric carrier comprises a random copolymer.
3147. The device of item 3002, further comprising a polymeric carrier, wherein the polymeric carrier comprises a biodegradable polymer.
3148. The device of item 3002, further comprising a polymeric carrier, wherein the polymeric carrier comprises a non-biodegradable polymer.
3149. The device of item 3002, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrophilic polymer. 3150. The device of item 3002, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrophobic polymer.
3151. The device of item 3002, further comprising a polymeric carrier, wherein the polymeric carrier comprises a polymer having hydrophilic domains.
3152. The device of item 3002, further comprising a polymeric carrier, wherein the polymeric carrier comprises a polymer having hydrophobic domains.
3153. The device of item 3002, further comprising a polymeric carrier, wherein the polymeric carrier comprises a non-conductive polymer.
3154. The device of item 3002, further comprising a polymeric carrier, wherein the polymeric carrier comprises an elastomer.
3155. The device of item 3002, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrogel.
3156. The device of item 3002, further comprising a polymeric carrier, wherein the polymeric carrier comprises a silicone polymer.
3157. The device of item 3002, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrocarbon polymer.
3158. The device of item 3002, further comprising a polymeric carrier, wherein the polymeric carrier comprises a styrene-derived polymer.
3159. The device of item 3002, further comprising a polymeric carrier, wherein the polymeric carrier comprises a butadiene polymer. 3160. The device of item 3002, further comprising a polymeric carrier, wherein the polymeric carrier comprises a macromer.
3161. The device of item 3002, further comprising a polymeric carrier, wherein the polymeric carrier comprises a poly(ethylene glycol) polymer.
3162. The device of item 3002, further comprising a polymeric carrier, wherein the polymeric carrier comprises an amorphous polymer.
3163. The device of item 3002, further comprising a lubricious coating.
3164. The device of item 3002 wherein the anti-scarring agent is located within pores or holes of the device.
3165. The device of item 3002 wherein the anti-scarring agent is located within a channel, lumen, or divet of the device.
3166. The device of item 3002, further comprising a second pharmaceutically active agent.
3167. The device of item 3002, further comprising an antiinflammatory agent.
3168. The device of item 3002, further comprising an agent that inhibits infection.
3169. The device of item 3002, further comprising an agent that inhibits infection, wherein the agent is an anthracycline. 3170. The device of item 3002, further comprising an agent that inhibits infection, wherein the agent is doxorubicin.
3171. The device of item 3002, further comprising an agent that inhibits infection, wherein the agent is mitoxantrone.
3172. The device of item 3002, further comprising an agent that inhibits infection, wherein the agent is a fluoropyrimidine.
3173. The device of item 3002, further comprising an agent that inhibits infection, wherein the agent is 5-fluorouracil (5-FU).
3174. The device of item 3002, further comprising an agent that inhibits infection, wherein the agent is a folic acid antagonist.
3175. The device of item 3002, further comprising an agent that inhibits infection, wherein the agent is methotrexate.
3176. The device of item 3002, further comprising an agent that inhibits infection, wherein the agent is a podophylotoxin.
3177. The device of item 3002, further comprising an agent that inhibits infection, wherein the agent is etoposide.
3178. The device of item 3002, further comprising an agent that inhibits infection, wherein the agent is a camptothecin.
3179. The device of item 3002, further comprising an agent that inhibits infection, wherein the agent is a hydroxyurea. 3180. The device of item 3002, further comprising an agent that inhibits infection, wherein the agent is a platinum complex.
3181. The device of item 3002, further comprising an agent that inhibits infection, wherein the agent is cisplatin.
3182. The device of item 3002, further comprising an antithrombotic agent.
3183. The device of item 3002, further comprising a visualization agent. '
3184. The device of item 3002, further comprising a visualization agent, wherein the visualization agent is a radiopaque material, wherein the radiopaque material comprises a metal, a halogenated compound, or a barium containing compound.
3185. The device of item 3002, further comprising a visualization agent, wherein the visualization agent is a radiopaque material, wherein the radiopaque material comprises barium, tantalum, or technetium.
3186. The device of item 3002, further comprising a visualization agent, wherein the visualization agent is a MRI responsive material.
3187. The device of item 3002, further comprising a visualization agent, wherein the visualization agent comprises a gadolinium chelate. 3188. The device of item 3002, further comprising a visualization agent, wherein the visualization agent comprises iron, magnesium, manganese, copper, or chromium.
3189. The device of item 3002, further comprising a visualization agent, wherein the visualization agent comprises an iron oxide compound.
3190. The device of item 3002, further comprising a visualization agent, wherein the visualization agent comprises a dye, pigment, or colorant.
3191. The device of item 3002, further comprising an echogenic material.
3192. The device of item 3002, further comprising an echogenic material, wherein the echogenic material is in the form of a coating.
3193. The device of item 3002 wherein the device is sterile.
3194. The device of item 3002 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device.
3195. The device of item 3002 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is connective tissue. 3196. The device of item 3002 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is muscle tissue.
3197. The device of item 3002 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is nerve tissue.
3198. The device of item 3002 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is epithelium tissue.
3199. The device of item 3002 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from the time of deployment of the device to about 1 year.
3200. The device of item 3002 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from about 1 month to 6 months.
3201. The device of item 3002 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from about 1 - 90 days.
3202. The device of item 3002 wherein the anti-scarring agent is released in effective concentrations from the device at a constant rate.
3203. The device of item 3002 wherein the anti-scarring agent is released in effective concentrations from the device at an increasing rate. 3204. The device of item 3002 wherein the anti-scarring agent is released in effective concentrations from the device at a decreasing rate.
3205. The device of item 3002 wherein the anti-scarring agent is released in effective concentrations from the composition comprising the anti-scarring agent by diffusion over a period ranging from the time of deployment of the device to about 90 days.
3206. The device of item 3002 wherein the anti-scarring agent is released in effective concentrations from the composition comprising the anti-scarring agent by erosion of the composition over a period ranging from the time of deployment of the device to about 90 days.
3207. The device of item 3002 wherein the device comprises about 0.01 Dg to about 10 Dg of the anti-scarring agent.
3208. The device of item 3002 wherein the device comprises about 10 Dg to about 10 mg of the anti-scarring agent.
3209. The device of item 3002 wherein the device comprises about 10 mg to about 250 mg of the anti-scarring agent.
3210. The device of item 3002 wherein the device comprises about 250 mg to about 1000 mg of the anti-scarring agent.
3211. The device of item 3002 wherein the device comprises about 1000 mg to about 2500 mg of the anti-scarring agent.
3212. The device of item 3002 wherein a surface of the device comprises less than 0.01 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied. 3213. The device of item 3002 wherein a surface of the device comprises about 0.01 Dg to about 1 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
3214. The device of item 3002 wherein a surface of the device comprises about 1 Dg to about 10 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
3215. The device of item 3002 wherein a surface of the device comprises about 10 Dg to about 250 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
3216. The device of item 3002 wherein a surface of the device comprises about 250 Dg to about 1000 Dg of the anti-scarring agent of anti- scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
3217. The device of item 3002 wherein a surface of the device comprises about 1000 Dg to about 2500 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
3218. A method as in any one of items 3002-3217, wherein the device is a mechanical prosthetic heart valve.
3219. A method as in any one of items 3002-3217, wherein the device is a bioprosthetic heart valve.
3220. A method as in any one of items 3002-3217, wherein the device is an implantable annular ring for receiving a prosthetic heart valve. 3221. A method as in any one of items 3002-3217, wherein the device is a suture ring having an outer peripheral tapered thread for attaching a heart valve prosthesis.
3222. A method as in any one of items 3002-3217, wherein the device is a suture ring for a mechanical heart valve.
3223. A device, comprising a penile implant and an anti- scarring agent or a composition comprising an anti-scarring agent, wherein the agent inhibits scarring between the device and a host into which the device is implanted.
3224. The device of item 3223 wherein the agent is an adensosine A2A receptor antagonist.
3225. The device of item 3223 wherein the agent is an AKT inhibitor. _ „ „ _ . _ _ _
3226. The device of item 3223 wherein the agent is an alpha 2 integrin antagonist, wherein the alpha 2 integrin antagonist is Pharmaprojects No. 5754 (Merck KgaA).
3227. The device of item 3223 wherein the agent an alpha 4 integrin antagonist.
3228. The device of item 3223 wherein the agent is an alpha 7 nicotinic receptor agonist.
3229. The device of item 3223 wherein the agent is an angiogenesis inhibitor selected from the group consisting of AG-12,958 (Pfizer), ATN-161 (Attenuon LLC), neovastat, an angiogenesis inhibitor from Jerina AG (Germany), NM-3 (Mercian), VGA-1155 (Taisho), FCE-26644 (Pfizer), FCE-26950 (Pfizer), FPMA (Meiji Daries), FFM 11142 (Fujisawa), GGTI-298, GM-1306 (Ligand), GPA-1734 (Novartis), NNC-47-0011 (Novo Nordisk), herbamycin (Nippon Kayaku), lenalidomide (Celegene), IP-10 (NIH), ABT-828 (Abbott), KIN-841 (Tokushima University, Japan), SF-1126 (Semafore Pharmaceuticals), laminin technology (NIH), CHIR-258 (Chiron), NVP-AEW541 (Novartis), NVP-AEW541 (Novartis), Vt16907 (Alchemia), OXI-8007 (Oxigene), EG-3306 (Ark Therapeutics), Maspin (Arriva), ABT- 567 (Abbott), PPI-2458 (Praecis Pharmaceuticals), CC-5079, CC-4089 (Celgene), HIF-1 alpha inhibitors (Xenova), S-247 (Pfizer), AP-23573 (Ariad), AZD-9935 (Astra Zeneca), mebendazole (Introgen Therapeutics), MetAP-2 inhibitors (GlaxoSmithKline), AG-615 (Angiogene Pharmaceuticals), Tie-2 antagonists (Hybrigenics), NC-381 , CYC-381, NC-169, NC-219, NC-383, NC-384, NC-407 (Lorus Therapeutics), ATN-224 (Attenuon), ON-01370 (Onconova), Vitronectin antagonists (Amgen), SDX-103 (Salmedix), Vitronectin antagonists (Shire), CHP (Riemser), TEK (Amgen), Anecortave acetate (Alcon), T46.2 (Matrix Therapeutics), HG-2 (Heptagen), TEM antagonists (Genzyme), Oxi-4500 (Oxigene), ATN-161 (Attenuon), WX-293 (Wilex), M-2025 (Metris Therapeutics), Alphastatin (BioActa), YH-16 (Yantai Rongchang), BIBF-1120 (Boehringer Ingelheim), BAY-57-9352 (Bayer), AS- 1404 (Cancer Research Technology), SC-77964 (Pfizer), glycomimetics (BioTie Therapies), TIE-2 Inhibitors (Ontogen), DIMI, Octamer (Octamer), ABR-215050 (Active Biotech), ABT-518 (Abbott), KDR inhibitors (Abbott), BSF-466895 (Abbott), SCH-221153 (Schering-Plough), DAC:antiangiogenic (ConjuChem), TFPI (EntreMed), AZD-2171 (Astra-Zenaca), CDC-394 (Celgene), LY290293 (EIi Lilly), IDN-5390 (Indena), Kdr Kinase Inhibitors (Merck), CT-113020, CT-116433, CT-116563, CT-31890, CT-32228) (Cell Therapeutics), A-299620 (Abbott), TWEAK Inhibitor (Amgen), VEGF modulators (Johnson and Johnson), Tum-N53, tumstatin (Genzyme), Thios- 1 , Thios-2 (Thios Pharmaceuticals), MV-6401 (Miravant Medical Technologies), Spisulosine (PharmaMar), CEP-7055 (Cephalon), AUV-201 (Auvation), LM-609 (EIi Lilly), SKF-106615 (AnorMED), Oglufanide disodium (Cytran), BW-114 (Pharminox), Calreticulin (NIH)1 WX-678 (Wilex), SD-7784 (Pfizer), WX-UK1 (Wilex), SH-268 (Schering AG), 2-Me-PGA (Celgene), S- 137 (Pfizer), ZD-6126 (Angiogene Pharmaceuticals), SG-292 (SignalGen), Benefin (Lane Labs), A6, A36 (Angstrom), SB-2723005 (GlaxoSmithKline), SC-7 (Cell Therapeutics), ZEN-014 (AEterna Zentaris), 2-methoxyestradiol (EntreMed), NK-130119 (Nippon Kayaku), CC-10004 (Celgene), AVE- 8062A (Ajinomoto), Tacedinaline (Pfizer), Actinonin (Tokyo Metropolitan Institute of Medical Science), Lenalidomide (Celgene), VGA-1155, BTO-956 (SRI International), ER-68203-00 (Eisai), CT-6685 (UCB), JKC-362 (Phoenix Pharmaceuticals), DMI-3798 (DMI Biosciences, Angiomate (Ipsen), ZD-6474 (AstraZeneca), CEP-5214 (Cephalon), Canstatin (Genzyme), NM-3 (Mercian), Oridigm (MediQuest Therapeutics), Exherin (Adherex), BLS-0597 (Boston Life Sciences), PTC-299 (PTC Therapeutics), NPI-2358 (Nereus Pharmaceuticals), CGP-79787 (Novartis), AEE-788 (Novartis), CKD-732 (Chong Kun Dang), CP-564959 (OSI Pharmaceuticals), CM-101 (CarboMed), CT-2584, CT3501 (Cell Therapeutics), combretastatin and analogues and derivatives thereof (Oxigene), Rebimastat (Bristol-Meyers Squibb), Dextrin 2-sulfate (ML Laboratories), Cilengitide (Merk KGaA), NSC-706704 (Pharminox), KRN- 951 (Kirin Brewery), Ukrain, NSC-631570 (Nowicky Pharma), Tecogalan sodium (Daiichi Pharmaceutical), Tz-93 (Tsumura), TBC-1635 (Encysive Pharmaceuticals), TAN-1120 (Takeda), Semaxanib (Pfizer), BDI-7800 (Biopharmacopae), SD-186, SD-983 (Bristol-Meyers Squibb), SB-223245 (GlaxoSmithKline), SC-236 (Pfizer), RWJ-590973 (Johnson and Johnson), ILX-1850 (Genzyme), SC-68488, S-836 (Pfizer), CG-55069-11 (CuraGen), Ki-23057 (Kirin Brewery), CCX-700 (Chemoentryx), Pegaptanib octasodium (Giled Sciences), ANGIOCOL (available from Biostratum Inc.), or an analogue or derivative thereof.
3230. The device of item 3223 wherein the agent is an apoptosis antagonist. 3231. The device of item 3223 wherein the agent is an apoptosis activator.
3232. The device of item 3223 wherein the agent is a beta 1 integrin antagonist.
3233. The device of item 3223 wherein the agent is a beta tubulin inhibitor.
3234. The device of item 3223 wherein the agent is a blocker of enzyme production in Hepatitis C.
3235. The device of item 3223 wherein the agent is a Bruton's tyrosine kinase inhibitor.
3236. The device of item 3223 wherein the agent is a calcineurin inhibitor.
3237. The device of item 3223 wherein the agent is a caspase 3 inhibitor.
3238. The device of item 3223 wherein the agent is a CC chemokine receptor antagonist.
3239. The device of item 3223 wherein the agent is a cell cycle inhibitor selected from the group consisting of SNS-595 (Sunesis), synthadotin, KRX-0403, and an analogue or derivative thereof.
3240. The device of item 3223 wherein the agent is a cathepsin B inhibitor. 3241. The device of item 3223 wherein the agent is a cathepsin K inhibitor, wherein the cathepsin K inhibitor is 462795 (GlaxoSmithKline), INPL-022-D6 (Amura Therapeutics), an analogue or derivative thereof).
3242. The device of item 3223 wherein the agent is a cathepsin L inhibitor.
3243. The device of item 3223 wherein the agent is a CD40 antagonist.
3244. The device of item 3223 wherein the agent is a chemokine receptor agonist.
3245. The device of item 3223 wherein the agent is a chymase inhibitor.
3246. The device of item 3223 wherein the agent is a collagenase antagonist.
3247. The device of item 3223 wherein the agent is a CXCR antagonist.
3248. The device of item 3223 wherein the agent is a cyclin dependent kinase inhibitor selected from the group consisting of a CDK-1 inhibitor, a CDK-2 inhibitor, a CDK- 4 inhibitor, a CDK-6 inhibitor, a CAK1 inhibitor from GPC Biotech and Bristol-Myers Squibb, RGB-286199 (GPC Biotech), an anticancer agent from Astex Technology, a CAK1 inhibitor from GPC Biotech, a CDK inhibitor from Sanofi-Aventis, a CDK1/CDK2 inhibitor from Amgen, a CDK2 inhibitor from SUGEN-2 (Pfizer), a hearing loss therapy agent (Sound Pharmaceuticals), PD-0332991 (Pfizer), RGB-286199 (GPC Biotech), Ro-0505124 (Hoffmann- La Roche), a Ser/Thr kinase inhibitor from Lilly (EIi Lilly), CVT-2584 (CAS No. 199986-75-9) (CV Therapeutics), and an analogue or derivative thereof.
3249. The device of item 3223 wherein the agent is a cyclooxygenase 1 inhibitor.
3250. The device of item 3223 wherein the agent is a DHFR inhibitor.
3251. The device of item 3223 wherein the agent is a dual integrin inhibitor.
3252. The device of item 3223 wherein the agent is an elastase inhibitor.
3253. The device of item 3223 wherein the agent is an elongation factor-1 alpha inhibitor.
3254. The device of item 3223 wherein the agent is an endothelial growth factor antagonist.
3255. The device of item 3223 wherein the agent is an endothelial growth factor receptor kinase inhibitor selected from the group consisting of sorafenib tosylate (Bayer), AAL-993 (Novartis), ABP-309 (Novartis), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), E- 7080 (Eisai), EG-3306 (Ark Therapeutics), EXEL-2880 (Exelixis), GW- 654652 (GlaxoSmithKline), a KDR inhibitor from LG Life Sciences, CT-6685 and CT-6729 (UCB), KRN-633 and KRN-951 (Kirin Brewery), OSI-930 (OSI Pharmaceuticals), SP-5.2 (Supratek Pharma), SU-11657 (Pfizer), a Tie-2 antagonist (Hybrigenics), a VEGFR-2 kinase inhibitor (Bristol-Myers Squibb), XL-647 (Exelixis), a KDR inhibitor from Abbott Laboratories, sorafenib tosylate, and an analogue or derivative thereof.
3256. The device of item 3223 wherein the agent is an endotoxin antagonist.
3257. The device of item 3223 wherein the agent is an epothilone and tubulin binder.
3258. The device of item 3223 wherein the agent is an estrogen receptor antagonist.
3259. The device of item 3223 wherein the agent is an FGF inhibitor.
3260. The device of item 3223 wherein the agent is a farnexyl transferase inhibitor.
3261. The device of item 3223 wherein the agent is farnesyltransferase inhibitor selected from the group of A-197574 (Abbott), a farnesyltransferase inhibitor from Servier, FPTIII (Strathclyde Institute for Drug R), LB-42908 (LG Life Sciences), Pharmaprojects No. 5063 (Genzyme), Pharmaprojects No. 5597 (Ipsen), Yissum Project No. B-1055 (Yissum), and an analogue or derivative thereof.
3262. The device of item 3223 wherein the agent is an FLT-3 kinase inhibitor.
3263. The device of item 3223 wherein the agent is an FGF receptor kinase inhibitor. 3264. The device of item 3223 wherein the agent is a fibrinogen antagonist selected from the group consisting of AUV-201 (Auvation), MG-13926 (Sanofi-Aventis), plasminogen activator (CAS No. 105913-11-9) (from Sanofi-Aventis or UCB), plasminogen activator-2 (tPA- 2) (Sanofi-Aventis), pro-urokinase (CAS No. 82657-92-9) (Sanofi-Aventis), and an analogue or derivative thereof.
3265. The device of item 3223 wherein the agent is a heat shock protein 90 antagonist selected from the group consisting of SRN-005 (Sirenade), geldanamycin, NSC-33050 (17-allylaminogeldanamycin; 17- AAG), 17-dimethylaminoethylamino-17-demethoxy-geldanamycin (17- DMAG), rifabutin (rifamycin XIV, 1',4-didehydro-1-deoxy-1,4-dihydro-5'-(2- methylpropyl)-1-oxo-), and an analogue or derivative thereof.
3266. The device of item 3223 wherein the agent is a histone deacetylase inhibitor.
3267. The device of item 3223 wherein the agent is an HMGCoA reductase inhibitor selected from the group consisting of an atherosclerosis therapeutic from Lipid Sciences, ATI-16000 (ARYx Therapeutics), KS-01-019 (Kos Pharmaceuticals), Pharmaprojects No. 2197 (Sanofi-Aventi), RP 61969 (Sanofi-Aventis), and an analogue or derivative thereof.
3268. The device of item 3223 wherein the agent is an ICAM inhibitor.
3269. The device of item 3223 wherein the agent is an IL, ICE and IRAK antagonist, wherein the antagonist is a CJ-14877, CP-424174 (Pfizer), NF-61 (Negma-Lerads), and an analogue or derivative thereof. 3270. The device of item 3223 wherein the agent is an IL-2 inhibitor.
3271. The device of item 3223 wherein the agent is an immunosuppressant selected from the group consisting of teriflunomide (Sanofi Aventis), chlorsulfaquinoxalone (NSC-339004), chlorsulfaquinoxalone sulfate, CS-712 (Sankyo), ismomultin alfa (CAS No. 457913-93-8) (Akzo Nobel), antiallergics from GenPat77, antiinflammatories or AT-005 (Androclus Therapeutics), autoimmune disease therapeutics from EpiVax, BN-007 (Bone), budesonide (CAS No. 51333-22- 3) (MAP Pharmaceuticals), CO-14 (Genzyme), edratide (CAS No. 433922- 67-9) (Teva), EP-314 (Enanta), eprovafen (CAS No. 101335-99-3) (Sanofi- Aventis), HWA-131 (CAS No. 118788-41-3) (Sanofi-Aventis), immunomodulators from MerLion Pharmaceuticals, immunosuppressives from Alchemia, IPL-12 (Inflazyme), MDL-9563 (CAS No. 27086-86-8) (Sanofi-Aventis), Pharmaprojects No. 2330 (SanqfhAventis), Pharmaprojects No. 6426 (Abgenix), PXS-25 (Pharmaxis), rosmarinic acid (CAS No. 20283-92-5) (Sanofi-Aventis), RP 42927 or RP 54745 (CAS No. 135330-08-4) (Sanofi-Aventis), SGN-35 (Seattle Genetics), ST-1959 (Sigma-Tau), type I diabetes therapy from SYNX Pharma, UNIL-88 (Debiopharm), VP-025 (Vasogen), VR-694 (Vectura), PRTX-001 (Protalex), and an analogue or derivative thereof.
3272. The device of item 3223 wherein the agent is an IMPDH (inosine monophosphate).
3273. The device of item 3223 wherein the agent is an integrin antagonist.
3274. The device of item 3223 wherein the agent is an interleukin antagonist. 3275. The device of item 3223 wherein the agent is an inhibitor of type III receptor tyrosine kinase.
3276. The device of item 3223 wherein the agent is an irreversible inhibitor of enzyme methionine aminopeptidase type 2.
3277. The device of item 3223 wherein the agent is an isozyme selective delta protein kinase C inhibitor.
3278. The device of item 3223 wherein the agent a JAK3 enzyme inhibitor.
3279. The device of item 3223 wherein the agent is a JNK inhibitor.
3280. The device of item 3223 wherein the agent is a kinase inhibitor.
3281. The device of item 3223 wherein the agent is kinesin antagonist.
3282. The device of item 3223 wherein the agent is a kinesin antagonist.
3283. The device of item 3223 wherein the agent is a leukotriene inhibitor and antagonist selected from the group consisting of ambicromil (CAS No. 58805-38-2) (Sanofi-Aventis), amelubant (CAS No. 346735-24-8) (Boehringer Ingelheim), DW-1141 (Dong Wha), ebselen (Daiichi Pharmaceutical), ibudilast (Kyorin), leucotriene inhibitors from Sanofi-Aventis, lymphotoxin-beta receptor (LT-D) from Biogen Idee, Pharmaprojects No. 1535 and 2728 (CAS No. 119340-33-9) (Sanofi- Aventis), R-112 (Rigel), Rev-5367 (CAS No. 92532-05-3) (Sanofi-Aventis), RG-14893 (CAS No. 141835-49-6) (Sanofi-Aventis), RG-5901-A (CAS No. 101910-24-1), 92532-23-5, RP 66153 (CAS No. 142422-79-5), RP 66364 (CAS No.186916-92-5), RP 69698 (CAS No. 141748-00-7) (Sanofi-Aventis), SC-411930 (Pfizer), SC-41930 (CAS No. 120072-59-5) (Pfizer), SC-50605 (CAS No. 138828-39-4) (Pfizer), SC-51146 (CAS No. 141059-52-1), SC- 53228 (CAS No. 153633-01-3) (Pfizer), spaglumic acid (ZY-15106) (CAS No.3106-85-2), 80619-64-3 (Novartis), tipredane (CAS No. 85197-77-9) (Bristol-Myers Squibb), U-75302 (CAS No. 119477-85-9) (Pfizer), and analogue or derivative thereof.
3284. The device of item 3223 wherein the agent is an MAP kinase inhibitor.
3285. The device of item 3223 wherein the agent is a matrix metalloproteinase inhibitor.
3286. The device of item 3223 wherein the agent is an MCP- CCR2 inhibitor.
3287. The device of item 3223 wherein the agent is an mTOR inhibitor.
3288. The device of item 3223 wherein the agent is an mTOR kinase inhibitor.
3289. The device of item 3223 wherein the agent is a microtubule inhibitor selected from the group consisting of antibody- maytansinoid conjugates from Biogen Idee, colchicines (MantiCore Pharmaceuticals), anticancer immunoconjugates from Johnson & Johnson, DIME from Octamer, gni-1f (GNI)1 huC242-DM4, huMy9-6-DM1 (ImmunoGen), IDN-5404 (Indena), IMO-098, IMOderm (Imotep), mebendazole (Introgen Therapeutics), microtubule poisons from Cambridge Enterprise, paclitaxel such as LOTAX from Aphios (CAS No. 33069-62-4), Genexol-PM from Samyang, Pharmaprojects No. 6383 (Tapestry Pharmaceuticals), RPR-112378 (Sanofi-Aventis), SGN-75 (Seattle Genetics), SPL-7435 (Starpharma), SSR-250411 (Sanofi-Aventis), trastuzumab-DM1 (Genentech), vinorelbine, and an analogue or derivative thereof.
3290. The device of item 3223 wherein the agent is an MIF inhibitor.
3291. The device of item 3223 wherein the agent is an MMP inhibitor.
3292. The device of item 3223 wherein the agent is a neurokinin (NK) antagonist selected from the group consisting of anthrotainin (CAS No. 148084-40-6) (Sanofi-Aventis), an IBS thereapeutic from ArQuIe, MDL-105212A (CAS No. 167261-60-1) (Ssanofi-Aventis), Pharmaprojects No. 2744, 3258 (CAS No. 139167-47-8) 4006, 4201 , or 5986 (Sanofi-Aventis), RP 67580 (CAS No. 135911-02-3), SR-144190 (CAS No. 201152-86-5), SSR-240600, SSR-241586 (Sanofi-Aventis), TKA-457 (Novartis), vestipitant mesylate (CAS No. 334476-64-1) (GlaxoSmithKline), Win-64821 (Sanofi-Aventis), PRX-96026 (Predix Pharmaceuticals), and an analogue or derivative thereof.
3293. The device of item 3223 wherein the agent is an NF kappa B inhibitor selected from the group consisting of AVE-0545 or AVE- 0547 (Sanofi-Aventis), bortezomib (CAS No. 179324-69-7) (Millennium Pharmaceuticals), dexanabinol (CAS No. 112924-45-5) (Pharmos), dexlipotam (Viatris), Pharmaprojects No. 6283 (INDRA) (OXiGENE), IPL- 576092 (CAS No. 137571-30-3) (Inflazyme), NFKB decoy (Corgentech), NFKB decoy oligo (AnGes MG), NFKB's from Ariad, osteoporosis treatments or S5 (F005) from Fulcrum Pharmaceuticals, P61 (Phytopharm), R-flurbiprofen (CAS No. 5104-49-4) (Encore Pharmaceuticals), and an analogue or derivative thereof.
3294. The device of item 3223 wherein the agent is a nitric oxide agonist.
3295. The device of item 3223 wherein the agent is an ornithine decarboxylase inhibitor.
3296. The device of item 3223 wherein the agent is a p38 MAP kinase inhibitor selected from the group consisting of AZD-6703 (AstraZeneca), JX-401 (Jexys Pharmaceuticals), BMS-2 (Bristol-Myers Squibb), a p38 MAP kinase inhibitor from Novartis, a p3O-alpha MAP kinase inhibitor from Amphora, Pharmaprojects No. 5704 (Pharmacopeia), RPR- 200765A (Sanofi-Aventis), SD-282 (Johnson & Johnson), TAK-715 (Takeda), and an analogue or derivative thereof.
3297. The device of item 3223 wherein the agent is a palmitoyl-protein thioesterase inhibitor.
3298. The device of item 3223 wherein the agent is a PDGF receptor kinase inhibitor selected from the group consisting of AAL-993, AMN-107, or ABP-309 (Novartis), AMG-706 (Amgen), BAY-57-9352 (Bayer), CDP-860 (UCB), E-7080 (Eisai), imatinib (CAS No. 152459-95-5) (Novartis), OSI-930 (OSI Pharmaceuticals), RPR-127963E (Sanofi-Aventis), RWJ-540973 (Johnson & Johnson), sorafenib tosylate (Bayer), SU-11657 (Pfizer), tandutinib (CAS No. 387867-13-2) (Millennium Pharmaceuticals), vatalanib (Novartis), ZK-CDK (Schering AG), and an analogue or derivative thereof.
3299. The device of item 3223 wherein the agent is (-)- halofenate (Metabolex), AMG-131 (Amgen), antidiabetics from Japan Tobacco, AZD-4619, AZD-8450, AZD-8677 (AstraZeneca), DRF-10945, balaglitazone (Dr Reddy's), CS-00088, CS-00098 (Chipscreen Biosciences), E-3030 (Eisai), etalocib (CAS No. 161172-51-6) (EIi Lilly), GSK-641597 (Ligand), GSK-677954 (GlaxoSmithKline), GW-409544 (Ligand), GW- 590735 (GlaxoSmithKline), K-111 (Hoffmann-La Roche), LY-518674 (EIi Lilly), LY-674 (Ligand), LY-929 (Ligand), MC-3001 , MC-3002 (MaxoCore Pharmaceuticals), metformin HCI + pioglitazone (CAS No. 1115-70-4 and
112529-15-4), ACTOPLUS MET from Andrx), muraglitazar (CAS No. 331741-94-7) (Bristol-Myers Squibb), naveglitazar (Ligand), oleoylethanolamide (Kadmus Pharmaceuticals), ONO-5129, pioglitazone hydrochloride (CAS No. 111025-46-8 and I 112529-15-4) (Takeda), PLX-204 (Plexxikon), PPAR agonists from Genfit, PPAR delta agonists from EIi Lilly, PPAR-alpha agonists from CrystalGenomics, PPAR-gamma modulators and PPAR-β modulators from CareX, rosiglitazone maleate (CAS No. 122324- 73-4 or 155141-29-0) (GlaxoSmithKline), rosiglitazone maleate/glimepir (CAS No. 155141-29-0 and 93479-97-1), AVANDARYL, rosiglitazone maleate/metformin extend (CAS No. 155141-29-0 and 657-24-9), AVANDAMET, rosiglitazone maleate+metformin, AVANDAMET (GlaxoSmithKline), tesaglitazar (AstraZeneca), and an analogue or derivative thereof.
3300. The device of item 3223 wherein the agent is a phosphatase inhibitor.
3301. The device of item 3223 wherein the agent is a phosphodiesterase (PDE) inhibitor selected from the group consisting of avanafil (Tanabe Seiyaku), dasantafil (CAS No. 569351-91-3) (Schering- Plough), A-906119 (CAS No. 134072-58-5), DL-850 (Sanofi-Aventis), GRC- 3015, GRC-3566, GRC-3886 (Glenmark), HWA-153 (CAS No. 56395-66-5) (Sanofi-Aventis), hydroxypumafentrine (Altana), IBFB-130011 , IBFB-14-016, IBFB-140301, IBFB-150007, IBFB-211913 (IBFB Pharma), L-826141 (Merck & Co), medorinone (CAS No. 88296-61-1) (Sanofi-Aventis), MEM- 1917 (Memory Pharmaceuticals), ND-1251 (Neuro3d), PDE inhibitors from ICOS, PDE IV inhibitors from Memory Pharmaceuticals and CrystalGenomics, Pharmaprojects No. 2742 and 6141 (Sanofi-Aventis), QAD-171 (Novartis), RHC-2963 (CAS No. 76993-12-9 and 76993-14-1), RPR-117658, RPR-122818 derivatives, SR-24870 , and RPR-132294 (Sanofi-Aventis), SK-350 (ln2Gen), stroke therapy agents from deCODE Genetics, TAS-203 (Taiho), tofimilast (CAS No. 185954-27-2) (Pfizer), UK- 371800 (Pfizer), WIN-65579 (CAS No. 158020-82-7) (Sanofi-Aventis), IBFB- 130020 (IBFB Pharma), OPC-6535 (CAS No. 145739-56-6) (Otsuka), a phosphodiesterase lljjnhibitor, enoximone, a phosphodiesterase IV inhibitor, fosfosal, Atopik (Barrier Therapeutics), triflusal, a phosphodiesterase V inhibitor, and an analogue or derivative thereof.
3302. The device of item 3223 wherein the agent is a PKC inhibitor.
3303. The device of item 3223 wherein the agent is a platelet activating factor antagonist.
3304. The device of item 3223 wherein the agent is a platelet- derived growth factor receptor kinase inhibitor.
3305. The device of item 3223 wherein the agent is a prolyl hydroxylase inhibitor. 3306. The device of item 3223 wherein the agent is a polymorphonuclear neutrophil inhibitor.
3307. The device of item 3223 wherein the agent is a protein kinase B inhibitor.
3308. The device of item 3223 wherein the agent is a protein kinase C stimulant.
3309. The device of item 3223 wherein the agent is a purine nucleoside analogue.
3310. The device of item 3223 wherein the agent is a purinoreceptor P2X antagonist.
3311. The device of item 3223 wherein the .agent is a Raf . kinase inhibitor.
3312. The device of item 3223 wherein the agent is a reversible inhibitor of ErbB1 and ErbB2.
3313. The device of item 3223 wherein the agent is a ribonucleoside triphosphate reductase inhibitor.
3314. The device of item 3223 wherein the agent is an SDF-1 antagonist.
3315. The device of item 3223 wherein the agent is a sheddase inhibitor. 3316. The device of item 3223 wherein the agent is an SRC inhibitor.
3317. The device of item 3223 wherein the agent is a stromelysin inhibitor.
3318. The device of item 3223 wherein the agent is an Syk kinase inhibitor.
3319. The device of item 3223 wherein the agent is a telomerase inhibitor.
3320. The device of item 3223 wherein the agent is a TGF beta inhibitor selected from the group consisting of pirfenidone (CAS No. 53179-13-8) (MARNAC), tranilast (CAS No. 53902-12-8) (Kissei), IN-1130 (ln2Gen), mannose-6-phospjiateXBTG), TGF-β antagonists from Inflazyme (Pharmaprojects No. 6075), TGF-β antagonists from Sydney, non-industrial source), TGF-β I receptor kinase inhibitors from EIi Lilly, TGF-β receptor inhibitors from Johnson & Johnson, and an analogue or derivative thereof.
3321. The device of item 3223 wherein the agent is a TN Fa antagonist or TACE inhibitor selected from the group consisting of adalimumab (CAS No. 331731-18-1) (Cambridge Antibody Technology), AGIX-4207 (AtheroGenics), AGT-1 (Advanced Biotherapy), an antiinflammatory from Borean Pharma, Cellzome, or Paradigm Therapeutics, anti-inflammatory vaccine (TNF-alpha kinoid) from Neovacs, humanized anti-TNF antibody or an anti-TNF MAb (CB0006) Celltech (UCB), apratastat (CAS No. 287445-51-0) (Wyeth), BMS-561392 (Bristol-Myers Squibb), BN- 006 (Bone), certolizumab pegol (CAS No. 428863-50-7 or CH-138 (UCB), cilomilast (CAS No. 153259-65-5) (GlaxoSmithKline), CR-1 (Nuada Pharmaceuticals), CRx-119 (CombinatoRx), D-5410 (UCB), dacopafant (CAS No. 125372-33-0) (Sanofi-Aventis), dersalazine (CAS No. 188913-57- 7/188913-58-8) (Uriach), etanercept (CAS No. 185243-69-0) (Amgen), ethyl pyruvate (Critical (Critical Therapeutics), golimumab (CAS No. 476181-74-5) (Johnson & Johnson), hormono-immunotherapy from Ipsen, CDP571 (e.g., Humicade from UCB), IC-485 (ICOS), infliximab (CAS No. 170277-31-3) (Johnson & Johnson), IP-751 (Manhattan Pharmaceuticals), ISIS-104838 (CAS No. 250755-32-9) (ISIS Pharmaceuticals), lenalidomide (CAS No. 191732-72-6) (Celgene), lentinan (CAS No. 37339-90-5) (Ajinomoto), MDL- 201112 (CAS No. 142130-73-2) (Sanofi-Aventis), medroxyprogesterone (CAS No. 520-85-4) (InKine Pharmaceutical), N-acetylcysteine (CAS No. 616-91-1) (Zambon), NBE-P2 (DIREVO Biotech), nerelimomab (CAS No. 162774-06-3) (Chiron), OM-294DP (OM PHARMA), onercept (CAS No. 199685-57-9) (Yeda), PASSTNF-alpha (Verigen), pentoxifylline or oxypentifylline (Sanofi-Aventis), Pharmaprojects No. 4091 , 4241 , 4295, or 4488 (Sanofi-Aventis), Pharmaprojects No. 5480 (Amgen), Pharmaprojects No. 6749 (Cengentl, pirfenidone (CAS No, 53179-13-8) (MARNAC), RPR- 132294 (Sanofi-Aventis), S5 (F002) (Fulcrum Pharmaceuticals), simvastatin (CAS No. 79902-63-9) (Merck & Co), STA-6292 (Synta Pharmaceuticals), tacrolimus (CAS No. 104987-11-3) (Fujisawa LifeCycle Pharma), talactoferrin alfa (CAS No. 308240-58-6) (Agennix), thalidomide (CAS No. 50-35-1) (Celgene), TNF antagonists form ProStrakan, and Synergen, TNF inhibitors (Amgen), TNF-alpha antagonists from Dynavax Technologies and Jerina AG (Germany), TNF-alpha inhibitors from IBFB Pharma and Xencor (Xencor), torbafylline (CAS No. 105102-21-4) (Sanofi-Aventis), UR-1505 (Uriach), VT-346 (Viron Therapeutics), YSIL6 (Y's Therapeutics), YSTH2 (Y's Therapeutics), NPI-1302a-3 (Nereus Pharmaceuticals, a TNF antagonist from Jerina AG (Germany), dersalazine, and an analogue or derivative thereof.
3322. The device of item 3223 wherein the agent is a Toll receptor inhibitor. 3323. The device of item 3223 wherein the agent is a tubulin antagonist.
3324. The device of item 3223 wherein the agent is a tyrosine kinase inhibitor selected from the group consisting of SU-011248, SUTENT from Pfizer Inc. (New York, NY), BMS-354825, PN-355 (Paracelsian Pharmaceuticals), AGN-199659 (Allergan), AAL-993 or ABP-309 (Novartis), adaphostin (NIH), AEE-788 (Novartis), AG-013736 (OSI Pharmaceuticals), AG-13736 (Pfizer), ALT-110 (Alteris Therapeutics), AMG-706 (Amgen), anticancer MAbs from Xencor, anti-EGFrvlll MAbs from Abgenix, anti-HER2 MAb from Abiogen, AZD-2171 or AZD-9935 (AstraZeneca), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), CEP-5214 (Cephalon), CEP- 7055 (Cephalon), cetuximab (ImClone Systems), CHIR-200131 and CHIR- 258 (Chiron), CP-547632 (OSI Pharmaceuticals), CP-724714 (Pfizer), CT- 301 (Creabilis Therapeutics), D-69491 (Baxter International), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EGFR/ErbB2 inhibitors from Array BioPharma, erlotinib (CAS No. 183319-69-9) (OSI Pharmaceuticals), EXEL-2880 (Exelixis), FK-778 (Sanofi-Aventis) , gefitinib (CAS No. 184475-35-2) (AstraZeneca), GW-2286 or GW-654652 (GlaxoSmithKline), her2/neu antigen from AlphaVax, HER-2/neu inhibitor from Generex, Herzyme (Medipad) (Sirna Therapeutics), HKI-272 (Wyeth), HuMax-EGFr (Genmab), idronoxil (CAS No. 81267-65-4) (Novogen), IGF-1 inhibitors from Ontogen, IMC-11F8 (ImClone Systems), kahalalide F (CAS No. 149204-42-2) (PharmaMar), KDR inhibitor from LG Life Sciences, KDR inhibitors from Abbott Laboratories, KDR kinase inhibitors (UCB), Kdr kinase inhibitors from Merck & Co, KRN-633 and KRN-951 (Kirin Brewery), KSB-102 (Xenova), lapatinib ditosylate (CAS No. 388082-78-8) (GlaxoSmithKline), matuzumab (Merck KGaA), MDX-214 (Medarex), ME-103 (Pharmexa), MED-A300 (Gerolymatos), MNAC-13 (Lay Line Genomics), nimotuzumab (Center of Molecular Immunology), NSC-330507 or NSC-707545 (NIH), NV-50 (Novogen), OSI-930 (OSI Pharmaceuticals), panitumumab (Abgenix), pelitinib (CAS No. 287933-82-7) (Wyeth), pertuzumab (CAS No. 380610-27- 5) (Genentech), Pharmaprojects No. 3985 (Sanofi-Aventis), prostate cancer therapeutics from Sequenom (SQPC35, SQPC36, SQPC90), removab and remoxab (Trion Pharma), RG-13022 (CAS No. 136831-48-6), RG-13291 (CAS No. 138989-50-1), or RG-14620 (CAS No. 136831-49-7) (Sanofi- Aventis), RM-6427 (Romark), RNAi breast cancer therapy from Benitec, RP 53801 (CAS No. 125882-88-4) (Sanofi-Aventis), sorafenib tosylate (Bayer), SU-11657 (Pfizer), Tie-2 antagonists from Semaia (Hybrigenics), Tie-2 inhibitors from Ontogen, trastuzumab (CAS No. 180288-69-1) (Genentech), tyrosine kinase inhibitors from Sanofi-Aventis, U3-1287, U3-1565, U3-1784, U3-1800 (U3 Pharma), vatalanib (Novartis), VEGFR-2 kinase inhibitor from Bristol-Myers Squibb, XL-647 (Exelixis), ZD-6474 (AstraZeneca), ZK-CDK (Schering AG), an EGFR tyrosine kinase inhibitor, EKB-569 (Wyeth), and an analogue or derivative thereof.
3325. The device of item 3223 wherein the agent is a VEGF inhibitor.
3326. The device of item 3223 wherein the agent is a vitamin D receptor agonist.
3327. The device of item 3223 wherein the agent is ZD-6474 (an angiogenesis inhibitor).
3328. The device of item 3223 wherein the agent is AP-23573 (an mTOR inhibitor).
3329. The device of item 3223 wherein the agent is synthadotin (a tubulin antagonist). 3330. The device of item 3223 wherein the agent is S-0885 (a collagenase inhibitor).
3331. The device of item 3223 wherein the agent is aplidine (an elongation factor-1 alpha inhibitor).
3332. The device of item 3223 wherein the agent is ixabepilone (an epithilone).
3333. The device of item 3223 wherein the agent is IDN-5390 (an angiogenesis inhibitor).
3334. The device of item 3223 wherein the agent is SB- 2723005 (an angiogenesis inhibitor).
_ . . . -3335. The device of item 3223 wherein the agent is ABTτ.518 (an angiogenesis inhibitor).
3336. The device of item 3223 wherein the agent is combretastatin (an angiogenesis inhibitor).
3337. The device of item 3223 wherein the agent is anecortave acetate (an angiogenesis inhibitor).
3338. The device of item 3223 wherein the agent is SB- 715992 (a kinesin antagonist).
3339. The device of item 3223 wherein the agent is temsirolimus (an mTOR inhibitor). 3340. The device of item 3223 wherein the agent is adalimumab (a TNFα antagonist).
3341. The device of item 3223, further comprising a polymer.
3342. The device of item 3223 wherein the anti-scarring agent inhibits adhesion between the device and a host into which the device is implanted.
3343. The device of item 3223 wherein the device delivers the anti-scarring agent locally to tissue proximate to the device.
3344. The device of item 3223, further comprising a coating, wherein the coating comprises the anti-scarring agent.
3345. The device of item 3223, further comprising a coating,^ wherein the coating is disposed on a surface of the device.
3346. The device of item 3223, further comprising a coating, wherein the coating directly contacts the device.
3347. The device of item 3223, further comprising a coating, wherein the coating indirectly contacts the device.
3348. The device of item 3223, further comprising a coating, wherein the coating partially covers the device.
3349. The device of item 3223, further comprising a coating, wherein the coating completely covers the device. 3350. The device of item 3223, further comprising a coating, wherein the coating is a uniform coating.
3351. The device of item 3223, further comprising a coating, wherein the coating is a non-uniform coating.
3352. The device of item 3223, further comprising a coating, wherein the coating is a discontinuous coating.
3353. The device of item 3223, further comprising a coating, wherein the coating is a patterned coating.
3354. The device of item 3223, further comprising a coating, wherein the coating has a thickness of 100 Dm or less.
3355. The device. of item 3223, further comprising a coating, wherein the coating has a thickness of 10 Dm or less.
3356. The device of item 3223, further comprising a coating, wherein the coating adheres to the surface of the device upon deployment of the device.
3357. The device of item 3223, further comprising a coating, wherein the coating is stable at room temperature for a period of 1 year.
3358. The device of item 3223, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 0.0001% to about 1% by weight. 3359. The device of item 3223, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 1% to about 10% by weight.
3360. The device of item 3223, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 10% to about 25% by weight.
3361. The device of item 3223, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 25% to about 70% by weight.
3362. The device of item 3223, further comprising a coating, wherein the coating further comprises a polymer.
„ 3363. The device of item 3223, further comprising a first coating having a first composition and the second coating having a second composition.
3364. The device of item 3223, further comprising a first coating having a first composition and the second coating having a second composition, wherein the first composition and the second composition are different.
3365. The device of item 3223, further comprising a polymeric carrier, wherein the polymeric carrier comprises a copolymer.
3366. The device of item 3223, further comprising a polymeric carrier, wherein the polymeric carrier comprises a block copolymer. 3367. The device of item 3223, further comprising a polymeric carrier, wherein the polymeric carrier comprises a random copolymer.
3368. The device of item 3223, further comprising a polymeric carrier, wherein the polymeric carrier comprises a biodegradable polymer.
3369. The device of item 3223, further comprising a polymeric carrier, wherein the polymeric carrier comprises a non-biodegradable polymer.
3370. The device of item 3223, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrophilic polymer.
3371. The device of item 3223, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrophobic polymer.
3372. The device of item 3223, further comprising a polymeric carrier, wherein the polymeric carrier comprises a polymer having hydrophilic domains.
3373. The device of item 3223, further comprising a polymeric carrier, wherein the polymeric carrier comprises a polymer having hydrophobic domains.
3374. The device of item 3223, further comprising a polymeric carrier, wherein the polymeric carrier comprises a non-conductive polymer.
3375. The device of item 3223, further comprising a polymeric carrier, wherein the polymeric carrier comprises an elastomer. 3376. The device of item 3223, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrogel.
3377. The device of item 3223, further comprising a polymeric carrier, wherein the polymeric carrier comprises a silicone polymer.
3378. The device of item 3223, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrocarbon polymer.
3379. The device of item 3223, further comprising a polymeric carrier, wherein the polymeric carrier comprises a styrene-derived polymer.
3380. The device of item 3223, further comprising a polymeric carrier, wherein the polymeric carrier comprises a butadiene polymer.
3381. The device of item 3223, further comprising a polymeric carrier, wherein the polymeric carrier comprises a macromer.
3382. The device of item 3223, further comprising a polymeric carrier, wherein the polymeric carrier comprises a poly(ethylene glycol) polymer.
3383. The device of item 3223, further comprising a polymeric carrier, wherein the polymeric carrier comprises an amorphous polymer.
3384. The device of item 3223, further comprising a lubricious coating.
3385. The device of item 3223 wherein the anti-scarring agent is located within pores or holes of the device. 3386. The device of item 3223 wherein the anti-scarring agent is located within a channel, lumen, or divet of the device.
3387. The device of item 3223, further comprising a second pharmaceutically active agent.
3388. The device of item 3223, further comprising an antiinflammatory agent.
3389. The device of item 3223, further comprising an agent that inhibits infection.
3390. The device of item 3223, further comprising an agent that inhibits infection, wherein the agent is an anthracycline.
3391. The device of item 3223, further_cpmprlsing.an agent that inhibits infection, wherein the agent is doxorubicin.
3392. The device of item 3223, further comprising an agent that inhibits infection, wherein the agent is mitoxantrone.
3393. The device of item 3223, further comprising an agent that inhibits infection, wherein the agent is a fluoropyrimidine.
3394. The device of item 3223, further comprising an agent that inhibits infection, wherein the agent is 5-fluorouracil (5-FU).
3395. The device of item 3223, further comprising an agent that inhibits infection, wherein the agent is a folic acid antagonist. 3396. The device of item 3223, further comprising an agent that inhibits infection, wherein the agent is methotrexate.
3397. The device of item 3223, further comprising an agent that inhibits infection, wherein the agent is a podophylotoxin.
3398. The device of item 3223, further comprising an agent that inhibits infection, wherein the agent is etoposide.
3399. The device of item 3223, further comprising an agent that inhibits infection, wherein the agent is a camptothecin.
3400. The device of item 3223, further comprising an agent that inhibits infection, wherein the agent is a hydroxyurea.
3401. The device of item 3223, further comprising an agent that inhibits infection, wherein the agent is a platinum complex.
3402. The device of item 3223, further comprising an agent that inhibits infection, wherein the agent is cisplatin.
3403. The device of item 3223, further comprising an antithrombotic agent.
3404. The device of item 3223, further comprising a visualization agent.
3405. The device of item 3223, further comprising a visualization agent, wherein the visualization agent is a radiopaque material, wherein the radiopaque material comprises a metal, a halogenated compound, or a barium containing compound. 3406. The device of item 3223, further comprising a visualization agent, wherein the visualization agent is a radiopaque material, wherein the radiopaque material comprises barium, tantalum, or technetium.
3407. The device of item 3223, further comprising a visualization agent, wherein the visualization agent is a MRI responsive material.
3408. The device of item 3223, further comprising a visualization agent, wherein the visualization agent comprises a gadolinium chelate.
3409. The device of item 3223, further comprising a visualization agent, wherein the visualization agent comprises iron, magnesium, manganese, copper, or chromium.
3410. The device of item 3223, further comprising a visualization agent, wherein the visualization agent comprises an iron oxide compound.
3411. The device of item 3223, further comprising a visualization agent, wherein the visualization agent comprises a dye, pigment, or colorant.
3412. The device of item 3223, further comprising an echogenic material.
3413. The device of item 3223, further comprising an echogenic material, wherein the echogenic material is in the form of a coating. 3414. The device of item 3223 wherein the device is sterile.
3415. The device of item 3223 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device.
3416. The device of item 3223 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is connective tissue.
3417. The device of item 3223 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is muscle tissue.
3418. The device of item 3223 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is nerve tissue.
3419. The device of item 3223 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is epithelium tissue.
3420. The device of item 3223 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from the time of deployment of the device to about 1 year.
3421. The device of item 3223 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from about 1 month to 6 months. 3422. The device of item 3223 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from about 1 - 90 days.
3423. The device of item 3223 wherein the anti-scarring agent is released in effective concentrations from the device at a constant rate.
3424. The device of item 3223 wherein the anti-scarring agent is released in effective concentrations from the device at an increasing rate.
3425. The device of item 3223 wherein the anti-scarring agent is released in effective concentrations from the device at a decreasing rate.
3426. The device of item 3223 wherein the anti-scarring agent is released in effective concentrations from the composition comprising the anti-scarring agent by diffusion over a period ranging from the time of deployment of the device to about 90 days.
3427. The device of item 3223 wherein the anti-scarring agent is released in effective concentrations from the composition comprising the anti-scarring agent by erosion of the composition over a period ranging from the time of deployment of the device to about 90 days.
3428. The device of item 3223 wherein the device comprises about 0.01 Dg to about 10 Dg of the anti-scarring agent.
3429. The device of item 3223 wherein the device comprises about 10 Dg to about 10 mg of the anti-scarring agent.
3430. The device of item 3223 wherein the device comprises about 10 mg to about 250 mg of the anti-scarring agent. 3431. The device of item 3223 wherein the device comprises about 250 mg to about 1000 mg of the anti-scarring agent.
3432. The device of item 3223 wherein the device comprises about 1000 mg to about 2500 mg of the anti-scarring agent.
3433. The device of item 3223 wherein a surface of the device comprises less than 0.01 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
3434. The device of item 3223 wherein a surface of the device comprises about 0.01 Dg to about 1 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
3435. The device of item 3223 wherein a surface of the device comprises about 1 Dg to about 10 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
3436. The device of item 3223 wherein a surface of the device comprises about 10 Dg to about 250 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
3437. The device of item 3223 wherein a surface of the device comprises about 250 Dg to about 1000 Dg of the anti-scarring agent of anti- scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
3438. The device of item 3223 wherein a surface of the device comprises about 1000 Dg to about 2500 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied. 3439. A method as in any one of items 3223-3438, wherein the device is a penile implant that is a flexible rod.
3440. A method as in any one of items 3223-3438, wherein the device is a penile implant that is a hinged rod.
3441. A method as in any one of items 3223-3438, wherein the device is a penile implant that is an inflatable device with a pump.
3442. A device, comprising an endotracheal or tracheostomy tube (e.g., an implant) and an anti-scarring agent or a composition comprising an anti-searing agent, wherein the agent inhibits scarring between the device and a host into which the device is implanted.
3443. The device of item 3442 wherein the agent is an adensosine A2A receptor antagonist.
3444. The device of item 3442 wherein the agent is an AKT inhibitor.
3445. The device of item 3442 wherein the agent is an alpha 2 integrin antagonist, wherein the alpha 2 integrin antagonist is Pharmaprojects No. 5754 (Merck KgaA).
3446. The device of item 3442 wherein the agent an alpha 4 integrin antagonist.
3447. The device of item 3442 wherein the agent is an alpha 7 nicotinic receptor agonist.
3448. The device of item 3442 wherein the agent is an angiogenesis inhibitor selected from the group consisting of AG-12,958 (Pfizer), ATN-161 (Attention LLC), neovastat, an angiogenesis inhibitor from Jerina AG (Germany), NM-3 (Mercian), VGA-1155 (Taisho), FCE-26644 (Pfizer), FCE-26950 (Pfizer), FPMA (Meiji Daries), FR-111142 (Fujisawa), GGTI-298, GM-1306 (Ligand), GPA-1734 (Novartis), NNC-47-0011 (Novo Nordisk), herbamycin (Nippon Kayaku), lenalidomide (Celegene), IP-10 (NIH), ABT-828 (Abbott), KIN-841 (Tokushima University, Japan), SF-1126 (Semafore Pharmaceuticals), laminin technology (NIH), CHIR-258 (Chiron), NVP-AEW541 (Novartis), NVP-AEW541 (Novartis), Vt16907 (Alchemia), OXI-8007 (Oxigene), EG-3306 (Ark Therapeutics), Maspin (Arriva), ABT- 567 (Abbott), PPI-2458 (Praecis Pharmaceuticals), CC-5079, CC-4089 (Celgene), HIF-1 alpha inhibitors (Xenova), S-247 (Pfizer), AP-23573 (Ariad), AZD-9935 (Astra Zeneca), mebendazole (Introgen Therapeutics), MetAP-2 inhibitors (GlaxoSmithKline), AG-615 (Angiogene Pharmaceuticals), Tie-2 antagonists (Hybrigenics), NC-381 , CYC-381, NC-169, NC-219, NC-383, NC-384, NC-407 (Lorus Therapeutics), ATN-224 (Attenuon), ON-01370 (Onconova), Vitronectin antagonists (Amgen), SDX-103 (Salmedix), Vitronectin antagonists (Shire), CHP (Riemser), TEK (Amgen), Anecortave acetate (Alcon), T46.2 (Matrix Therapeutics), HG-2 (Heptagen), TEM antagonists (Genzyme), Oxi-4500 (Oxigene), ATN-161 (Attenuon), WX-293 (Wilex), M-2025 (Metris Therapeutics), Alphastatin (BioActa), YH-16 (Yantai Rongchang), BIBF-1120 (Boehringer Ingelheim), BAY-57-9352 (Bayer), AS- 1404 (Cancer Research Technology), SC-77964 (Pfizer), glycomimetics (BioTie Therapies), TIE-2 Inhibitors (Ontogen), DIMI, Octamer (Octamer), ABR-215050 (Active Biotech), ABT-518 (Abbott), KDR inhibitors (Abbott), BSF-466895 (Abbott), SCH-221153 (Schering-Plough), DAC:antiangiogenic (ConjuChem), TFPI (EntreMed), AZD-2171 (Astra-Zenaca), CDC-394 (Celgene), LY290293 (EIi Lilly), IDN-5390 (Indena), Kdr Kinase Inhibitors (Merck), CT-113020, CT-116433, CT-116563, CT-31890, CT-32228) (Cell Therapeutics), A-299620 (Abbott), TWEAK Inhibitor (Amgen), VEGF modulators (Johnson and Johnson), Tum-N53, tumstatin (Genzyme), Thios- 1 , Thios-2 (Thios Pharmaceuticals), MV-6401 (Miravant Medical Technologies), Spisulosine (PharmaMar), CEP-7055 (Cephalon), AUV-201 (Auvation), LM-609 (EIi Lilly), SKF-106615 (AnorMED), Oglufanide disodium (Cytran), BW-114 (Pharminox), Calreticulin (NIH), WX-678 (Wilex), SD-7784 (Pfizer), WX-UK1 (Wilex), SH-268 (Schering AG), 2-Me-PGA (Celgene), S- 137 (Pfizer), ZD-6126 (Angiogene Pharmaceuticals), SG-292 (SignalGen), Benefin (Lane Labs), A6, A36 (Angstrom), SB-2723005 (GlaxoSmithKline), SC-7 (Cell Therapeutics), ZEN-014 (AEterna Zentaris), 2-methoxyestradiol (EntreMed), NK-130119 (Nippon Kayaku), CC-10004 (Celgene), AVE- 8062A (Ajinomoto), Tacedinaline (Pfizer), Actinonin (Tokyo Metropolitan Institute of Medical Science), Lenalidomide (Celgene), VGA-1155, BTO-956 (SRI International), ER-68203-00 (Eisai), CT-6685 (UCB), JKC-362 (Phoenix Pharmaceuticals), DMI-3798 (DMI Biosciences, Angiomate (Ipsen), ZD-6474 (AstraZeneca), CEP-5214 (Cephalon), Canstatin (Genzyme), NM-3 (Mercian), Oridigm (MediQuest Therapeutics), Exherin (Adherex), BLS-0597 (Boston Life Sciences), PTC-299 (PTC Therapeutics), NPI-2358-(Nereus Pharmaceuticals), CGP-79787 (Novartis), AEE-788. (Novartis), CKD-732 (Chong Kun Dang), CP-564959 (OSI Pharmaceuticals), CM-101 (CarboMed), CT-2584, CT3501 (Cell Therapeutics), combretastatin and analogues and derivatives thereof (Oxigene), Rebimastat (Bristol-Meyers Squibb), Dextrin 2-sulfate (ML Laboratories), Cilengitide (Merk KGaA), NSC-706704 (Pharminox), KRN- 951 (Kirin Brewery), Ukrain, NSC-631570 (Nowicky Pharma), Tecogalan sodium (Daiichi Pharmaceutical), Tz-93 (Tsumura), TBC-1635 (Encysive Pharmaceuticals), TAN-1120 (Takeda), Semaxanib (Pfizer), BDI-7800 (Biopharmacopae), SD-186, SD-983 (Bristol-Meyers Squibb), SB-223245 (GlaxoSmithKline), SC-236 (Pfizer), RWJ-590973 (Johnson and Johnson), ILX-1850 (Genzyme), SC-68488, S-836 (Pfizer), CG-55069-11 (CuraGen), Ki-23057 (Kirin Brewery), CCX-700 (Chemoentryx), Pegaptanib octasodium (Giled Sciences), ANGIOCOL (available from Biostratum Inc.), or an analogue or derivative thereof. 3449. The device of item 3442 wherein the agent is an apoptosis antagonist.
3450. The device of item 3442 wherein the agent is an apoptosis activator.
3451. The device of item 3442 wherein the agent is a beta 1 integrin antagonist.
3452. The device of item 3442 wherein the agent is a beta tubulin inhibitor.
3453. The device of item 3442 wherein the agent is a blocker of enzyme production in Hepatitis C.
3454. The device of jtem_3442 wherein the agent is a Bruton's- tyrosine kinase inhibitor.
3455. The device of item 3442 wherein the agent is a calcineurin inhibitor.
3456. The device of item 3442 wherein the agent is a caspase 3 inhibitor.
3457. The device of item 3442 wherein the agent is a CC chemokine receptor antagonist.
3458. The device of item 3442 wherein the agent is a cell cycle inhibitor selected from the group consisting of SNS-595 (Sunesis), synthadotin, KRX-0403, and an analogue or derivative thereof. 3459. The device of item 3442 wherein the agent is a cathepsin B inhibitor.
3460. The device of item 3442 wherein the agent is a cathepsin K inhibitor, wherein the cathepsin K inhibitor is 462795 (GlaxoSmithKline), INPL-022-D6 (Amura Therapeutics), an analogue or derivative thereof).
3461. The device of item 3442 wherein the agent is a cathepsin L inhibitor.
3462. The device of item 3442 wherein the agent is a CD40 antagonist.
3463. The device of item 3442 wherein the agent is a chemokine receptor agonist.
3464. The device of item 3442 wherein the agent is a chymase inhibitor.
3465. The device of item 3442 wherein the agent is a collagenase antagonist.
3466. The device of item 3442 wherein the agent is a CXCR antagonist.
3467. The device of item 3442 wherein the agent is a cyclin dependent kinase inhibitor selected from the group consisting of a CDK-1 inhibitor, a CDK-2 inhibitor, a CDK- 4 inhibitor, a CDK-6 inhibitor, a CAK1 inhibitor from GPC Biotech and Bristol-Myers Squibb, RGB-286199 (GPC Biotech), an anticancer agent from Astex Technology, a CAK1 inhibitor from GPC Biotech, a CDK inhibitor from Sanofi-Aventis, a CDK1/CDK2 inhibitor from Amgen, a CDK2 inhibitor from SUGEN-2 (Pfizer), a hearing loss therapy agent (Sound Pharmaceuticals), PD-0332991 (Pfizer), RGB-286199 (GPC Biotech), Ro-0505124 (Hoffmann-La Roche), a Ser/Thr kinase inhibitor from Lilly (EIi Lilly), CVT-2584 (CAS No. 199986-75-9) (CV Therapeutics), and an analogue or derivative thereof.
3468. The device of item 3442 wherein the agent is a cyclooxygenase 1 inhibitor.
3469. The device of item 3442 wherein the agent is a DHFR inhibitor.
3470. The device of item 3442 wherein the agent is a dual integrin inhibitor.
3471. The device of item 3442 wherein the agent is an elastase inhibitor.
3472. The device of item 3442 wherein the agent is an elongation factor-1 alpha inhibitor.
3473. The device of item 3442 wherein the agent is an endothelial growth factor antagonist.
3474. The device of item 3442 wherein the agent is an endothelial growth factor receptor kinase inhibitor selected from the group consisting of sorafenib tosylate (Bayer), AAL-993 (Novartis), ABP-309 (Novartis), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Jngelheim), E- 7080 (Eisai), EG-3306 (Ark Therapeutics), EXEL-2880 (Exelixis), GW- 654652 (GlaxoSmithKline), a KDR inhibitor from LG Life Sciences, CT-6685 and CT-6729 (UCB), KRN-633 and KRN-951 (Kirin Brewery), OSI-930 (OSI Pharmaceuticals), SP-5.2 (Supratek Pharma), SU-11657 (Pfizer), a Tie-2 antagonist (Hybrigenics), a VEGFR-2 kinase inhibitor (Bristol-Myers Squibb), XL-647 (Exelixis), a KDR inhibitor from Abbott Laboratories, sorafenib tosylate, and an analogue or derivative thereof.
3475. The device of item 3442 wherein the agent is an endotoxin antagonist.
3476. The device of item 3442 wherein the agent is an epothilone and tubulin binder.
3477. The device of item 3442 wherein the agent is an estrogen receptor antagonist.
- 3478. The device of item 3442 wherein the agent is an FGF inhibitor.
3479. The device of item 3442 wherein the agent is a farnexyl transferase inhibitor.
3480. The device of item 3442 wherein the agent is farnesyltransferase inhibitor selected from the group of A-197574 (Abbott), a farnesyltransferase inhibitor from Servier, FPTIII (Strathclyde Institute for Drug R), LB-42908 (LG Life Sciences), Pharmaprojects No. 5063 (Genzyme), Pharmaprojects No. 5597 (Ipsen), Yissum Project No. B-1055 (Yissum), and an analogue or derivative thereof.
3481. The device of item 3442 wherein the agent is an FLT-3 kinase inhibitor. 3482. The device of item 3442 wherein the agent is an FGF receptor kinase inhibitor.
3483. The device of item 3442 wherein the agent is a fibrinogen antagonist selected from the group consisting of AUV-201 (Auvation), MG-13926 (Sanofi-Aventis), plasminogen activator (CAS No. 105913-11-9) (from Sanofi-Aventis or UCB), plasminogen activator-2 (tPA- 2) (Sanofi-Aventis), pro-urokinase (CAS No. 82657-92-9) (Sanofi-Aventis), and an analogue or derivative thereof.
3484. The device of item 3442 wherein the agent is a heat shock protein 90 antagonist selected from the group consisting of SRN-005 (Sirenade), geldanamycin, NSC-33050 (17-allylaminogeldanamycin; 17- AAG), 17-dimethylaminoethylamino-17-demethoxy-geldanamycin (17- DMAG), rifabutin (rifamycin XIV, 1',4-didehydro-1-deoxy-1 ,4-dihydro-5'-(2- methylpropyl)-1-oxo-), and an analogue or derivative thereof..
3485. The device of item 3442 wherein the agent is a histone deacetylase inhibitor.
3486. The device of item 3442 wherein the agent is an HMGCoA reductase inhibitor selected from the group consisting of an atherosclerosis therapeutic from Lipid Sciences, ATI-16000 (ARYx Therapeutics), KS-01-019 (Kos Pharmaceuticals), Pharmaprojects No. 2197 (Sanofi-Aventi), RP 61969 (Sanofi-Aventis), and an analogue or derivative thereof.
3487. The device of item 3442 wherein the agent is an ICAM inhibitor. 3488. The device of item 3442 wherein the agent is an IL, ICE and IRAK antagonist, wherein the antagonist is a CJ-14877, CP-424174 (Pfizer), NF-61 (Negma-Lerads), and an analogue or derivative thereof.
3489. The device of item 3442 wherein the agent is an IL-2 inhibitor.
3490. The device of item 3442 wherein the agent is an immunosuppressant selected from the group consisting of teriflunomide (Sanofi Aventis), chlorsulfaquinoxalone (NSC-339004), chlorsulfaquinoxalone sulfate, CS-712 (Sankyo), ismomultin alfa (CAS No. 457913-93-8) (Akzo Nobel), antiallergics from GenPat77, antiinflammatories or AT-005 (Androclus Therapeutics), autoimmune disease therapeutics from EpiVax, BN-007 (Bone), budesonide (CAS No. 51333-22- 3) (MAP Pharmaceuticals), CO-14 (Genzyme), edratide (CAS No. 433922- 67-9) (Teva), EP-314 (Enanta), eprovafen (CAS No. 101335-99-3) (Sanofi- Aventis), HWA-131 (CAS No. 118788-41-3) (Sanofi-Aventis), immunomodulators from MerLion Pharmaceuticals, immunosuppressives from Alchemia, IPL-12 (Inflazyme), MDL-9563 (CAS No. 27086-86-8) (Sanofi-Aventis), Pharmaprojects No. 2330 (Sanofi-Aventis), Pharmaprojects No. 6426 (Abgenix), PXS-25 (Pharmaxis), rosmarinic acid (CAS No. 20283-92-5) (Sanofi-Aventis), RP 42927 or RP 54745 (CAS No. 135330-08-4) (Sanofi-Aventis), SGN-35 (Seattle Genetics), ST-1959 (Sigma-Tau), type I diabetes therapy from SYNX Pharma, UNIL-88 (Debiopharm), VP-025 (Vasogen), VR-694 (Vectura), PRTX-001 (Protalex), and an analogue or derivative thereof.
3491. The device of item 3442 wherein the agent is an IMPDH (inosine monophosphate). 3492. The device of item 3442 wherein the agent is an integrin antagonist.
3493. The device of item 3442 wherein the agent is an interleukin antagonist.
3494. The device of item 3442 wherein the agent is an inhibitor of type III receptor tyrosine kinase.
3495. The device of item 3442 wherein the agent is an irreversible inhibitor of enzyme methionine aminopeptidase type 2.
3496. The device of item 3442 wherein the agent is an isozyme selective delta protein kinase C inhibitor.
„ 3497., The. device of item 3442 wherein the agent a JAK3 enzyme inhibitor.
3498. The device of item 3442 wherein the agent is a JNK inhibitor.
3499. The device of item 3442 wherein the agent is a kinase inhibitor.
3500. The device of item 3442 wherein the agent is kinesin antagonist.
3501. The device of item 3442 wherein the agent is a kinesin antagonist. 3502. The device of item 3442 wherein the agent is a leukotriene inhibitor and antagonist selected from the group consisting of ambicromil (CAS No. 58805-38-2) (Sanofi-Aventis), amelubant (CAS No. 346735-24-8) (Boehringer Ingeiheim), DW-1141 (Dong Wha), ebselen (Daiichi Pharmaceutical), ibudilast (Kyorin), leucotriene inhibitors from Sanofi-Aventis, lymphotoxin-beta receptor (LT-D) from Biogen Idee, Pharmaprojects No. 1535 and 2728 (CAS No. 119340-33-9) (Sanofi- Aventis), R-112 (Rigel), Rev-5367 (CAS No. 92532-05-3) (Sanofi-Aventis), RG-14893 (CAS No. 141835-49-6) (Sanofi-Aventis), RG-5901-A (CAS No. 101910-24-1), 92532-23-5, RP 66153 (CAS No. 142422-79-5), RP 66364 (CAS No.186916-92-5), RP 69698 (CAS No. 141748-00-7) (Sanofi-Aventis), SC-411930 (Pfizer), SC-41930 (CAS No. 120072-59-5) (Pfizer), SC-50605 (CAS No. 138828-39-4) (Pfizer), SC-51146 (CAS No. 141059-52-1), SC- 53228 (CAS No. 153633-01-3) (Pfizer), spaglumic acid (ZY-15106) (CAS No.3106-85-2), 80619-64-3 (Novartis), tipredane (CAS No. 85197-77-9) (Bristol-Myers Squibb), U-75302 (CAS No. 119477-85-9) (Pfizer), and analogue or derivative thereof.
3503. The device of item 3442 wherein the agent is an MAP kinase inhibitor.
3504. The device of item 3442 wherein the agent is a matrix metalloproteinase inhibitor.
3505. The device of item 3442 wherein the agent is an MCP- CCR2 inhibitor.
3506. The device of item 3442 wherein the agent is an mTOR inhibitor. 3507. The device of item 3442 wherein the agent is an mTOR kinase inhibitor.
3508. The device of item 3442 wherein the agent is a microtubule inhibitor selected from the group consisting of antibody- maytansinoid conjugates from Biogen Idee, colchicines (MantiCore Pharmaceuticals), anticancer immunoconjugates from Johnson & Johnson, DIME from Octamer, gni-1f (GNI), huC242-DM4, huMy9-6-DM1 (ImmunoGen), IDN-5404 (Indena), IMO-098, IMOderm (Imotep), mebendazole (Introgen Therapeutics), microtubule poisons from Cambridge Enterprise, paclitaxel such as LOTAX from Aphios (CAS No. 33069-62-4), Genexol-PM from Samyang, Pharmaprojects No. 6383 (Tapestry Pharmaceuticals), RPR-112378 (Sanofi-Aventis), SGN-75 (Seattle Genetics), SPL-7435 (Starpharma), SSR-250411 (Sanofi-Aventis), trastuzumab-DM1 (Genentech), vinorelbine, and an analogue or derivative
-thereoL . . . . ._ .
3509. The device of item 3442 wherein the agent is an MIF inhibitor.
3510. The device of item 3442 wherein the agent is an MMP inhibitor.
3511. The device of item 3442 wherein the agent is a neurokinin (NK) antagonist selected from the group consisting of anthrotainin (CAS No. 148084-40-6) (Sanofi-Aventis), an IBS thereapeutic from ArQuIe, MDL-105212A (CAS No. 167261-60-1) (Ssanofi-Aventis), Pharmaprojects No. 2744, 3258 (CAS No. 139167-47-8) 4006, 4201, or 5986 (Sanofi-Aventis), RP 67580 (CAS No. 135911-02-3), SR-144190 (CAS No. 201152-86-5), SSR-240600, SSR-241586 (Sanofi-Aventis), TKA-457 (Novartis), vestipitant mesylate (CAS No. 334476-64-1) (GlaxoSmithKline), Win-64821 (Sanofi-Aventis), PRX-96026 (Predix Pharmaceuticals), and an analogue or derivative thereof.
3512. The device of item 3442 wherein the agent is an NF kappa B inhibitor selected from the group consisting of AVE-0545 or AVE- 0547 (Sanofi-Aventis), bortezomib (CAS No. 179324-69-7) (Millennium Pharmaceuticals), dexanabinol (CAS No. 112924-45-5) (Pharmos), dexlipotam (Viatris), Pharmaprojects No. 6283 (INDRA) (OXiGENE), IPL- 576092 (CAS No. 137571-30-3) (Inflazyme), NFKB decoy (Corgentech), NFKB decoy oligo (AnGes MG), NFKB's from Ariad, osteoporosis treatments or S5 (F005) from Fulcrum Pharmaceuticals, P61 (Phytopharm), R-flurbiprofen (CAS No. 5104-49-4) (Encore Pharmaceuticals), and an analogue or derivative thereof.
3513. The device of item 3442 wherein the agent is a nitric oxide agonist.
3514. The device of item 3442 wherein the agent is an ornithine decarboxylase inhibitor.
3515. The device of item 3442 wherein the agent is a p38 MAP kinase inhibitor selected from the group consisting of AZD-6703 (AstraZeneca), JX-401 (Jexys Pharmaceuticals), BMS-2 (Bristol-Myers Squibb), a p38 MAP kinase inhibitor from Novartis, a p38-alpha MAP kinase inhibitor from Amphora, Pharmaprojects No. 5704 (Pharmacopeia), RPR- 200765A (Sanofi-Aventis), SD-282 (Johnson & Johnson), TAK-715 (Takeda), and an analogue or derivative thereof.
3516. The device of item 3442 wherein the agent is a palmitoyl-protein thioesterase inhibitor. 3517. The device of item 3442 wherein the agent is a PDGF receptor kinase inhibitor selected from the group consisting of AAL-993, AMN-107, or ABP-309 (Novartis), AMG-706 (Amgen), BAY-57-9352 (Bayer), CDP-860 (UCB), E-7080 (Eisai), imatinib (CAS No. 152459-95-5) (Novartis), OSI-930 (OSI Pharmaceuticals), RPR-127963E (Sanofi-Aventis), RWJ-540973 (Johnson & Johnson), sorafenib tosylate (Bayer), SU-11657 (Pfizer), tandutinib (CAS No. 387867-13-2) (Millennium Pharmaceuticals), vatalanib (Novartis), ZK-CDK (Schering AG), and an analogue or derivative thereof.
3518. The device of item 3442 wherein the agent is (-)- halofenate (Metabolex), AMG-131 (Amgen), antidiabetics from Japan Tobacco, AZD-4619, AZD-8450, AZD-8677 (AstraZeneca), DRF-10945, balaglitazone (Dr Reddy's), CS-00088, CS-00098 (Chipscreen Biosciences), E-3030 (Eisai), etalocib (CAS No. 161172-51-6) (EIi Lilly), GSK-641597 (Ligand),-GSK-677954 (GlaxoSmithKline), GW-409544 (Ligand), GW- 590735 (GlaxoSmithKline), K-111 (Hoffmann-La Roche), LY-518674 (EIi Lilly), LY-674 (Ligand), LY-929 (Ligand), MC-3001 , MC-3002 (MaxoCore Pharmaceuticals), metformin HCI + pioglitazone (CAS No. 1115-70-4 and
112529-15-4), ACTOPLUS MET from Andrx), muraglitazar (CAS No. 331741-94-7) (Bristol-Myers Squibb), naveglitazar (Ligand), oleoylethanolamide (Kadmus Pharmaceuticals), ONO-5129, pioglitazone hydrochloride (CAS No. 111025-46-8 and 112529-15-4) (Takeda), PLX-204 (Plexxikon), PPAR agonists from Genfit, PPAR delta agonists from EIi Lilly, PPAR-alpha agonists from CrystalGenomics, PPAR-gamma modulators and PPAR-β modulators from CareX, rosiglitazone maleate (CAS No. 122324- 73-4 or 155141-29-0) (GlaxoSmithKline), rosiglitazone maleate/glimepir (CAS No. 155141-29-0 and 93479-97-1), AVANDARYL, rosiglitazone maleate/metformin extend (CAS No. 155141-29-0 and 657-24-9), AVANDAMET, rosiglitazone maleate+metformin, AVANDAMET (GlaxoSmithKline), tesaglitazar (AstraZeneca), and an analogue or derivative thereof.
3519. The device of item 3442 wherein the agent is a phosphatase inhibitor.
3520. The device of item 3442 wherein the agent is a phosphodiesterase (PDE) inhibitor selected from the group consisting of avanafil (Tanabe Seiyaku), dasantafil (CAS No. 569351-91-3) (Schering- Plough), A-906119 (CAS No. 134072-58-5), DL-850 (Sanofi-Aventis), GRC- 3015, GRC-3566, GRC-3886 (Glenmark), HWA-153 (CAS No. 56395-66-5) (Sanofi-Aventis), hydroxypumafentrine (Altana), IBFB-130011 , IBFB-14-016, IBFB-140301 , IBFB-150007, IBFB-211913 (IBFB Pharma), L-826141 (Merck & Co), medorinone (CAS No. 88296-61-1) (Sanofi-Aventis), MEM- 1917 (Memory Pharmaceuticals), ND-1251 (Neuro3d), PDE inhibitors from ICOS, PDE IV inhibitors from Memory Pharmaceuticals and CrystalGenomics, Pharmaprojects No. 2742 and 6141 (Sanofi-Aventis), QAD-171 (Novartis), RHC-2963 (CAS No. 76993-12-9 and 76993-14-1), RPR-117658, RPR-122818 derivatives, SR-24870 , and RPR-132294 (Sanofi-Aventis), SK-350 (ln2Gen), stroke therapy agents from deCODE Genetics, TAS-203 (Taiho), tofimilast (CAS No. 185954-27-2) (Pfizer), UK- 371800 (Pfizer), WIN-65579 (CAS No. 158020-82-7) (Sanofi-Aventis), IBFB- 130020 (IBFB Pharma), OPC-6535 (CAS No. 145739-56-6) (Otsuka), a phosphodiesterase III inhibitor, enoximone, a phosphodiesterase IV inhibitor, fosfosal, Atopik (Barrier Therapeutics), triflusal, a phosphodiesterase V inhibitor, and an analogue or derivative thereof.
3521. The device of item 3442 wherein the agent is a PKC inhibitor. 3522. The device of item 3442 wherein the agent is a platelet activating factor antagonist.
3523. The device of item 3442 wherein the agent is a platelet- derived growth factor receptor kinase inhibitor.
3524. The device of item 3442 wherein the agent is a prolyl hydroxylase inhibitor.
3525. The device of item 3442 wherein the agent is a polymorphonuclear neutrophil inhibitor.
3526. The device of item 3442 wherein the agent is a protein kinase B inhibitor.
3527. The device of item 3442 wherein the agent is a protein kinase C stimulant
3528. The device of item 3442 wherein the agent is a purine nucleoside analogue.
3529. The device of item 3442 wherein the agent is a purinoreceptor P2X antagonist.
3530. The device of item 3442 wherein the agent is a Raf kinase inhibitor.
3531. The device of item 3442 wherein the agent is a reversible inhibitor of ErbB1 and ErbB2. 3532. The device of item 3442 wherein the agent is a ribonucleoside triphosphate reductase inhibitor.
3533. The device of item 3442 wherein the agent is an SDF-1 antagonist.
3534. The device of item 3442 wherein the agent is a sheddase inhibitor.
3535. The device of item 3442 wherein the agent is an SRC inhibitor.
3536. The device of item 3442 wherein the agent is a stromelysin inhibitor.
. . _. . ._ 3537. The device of item 3442 wherein the agent is an Syk kinase inhibitor.
3538. The device of item 3442 wherein the agent is a telomerase inhibitor.
3539. The device of item 3442 wherein the agent is a TGF beta inhibitor selected from the group consisting of pirfenidone (CAS No. 53179-13-8) (MARNAC), tranilast (CAS No. 53902-12-8) (Kissei), IN-1130 (ln2Gen), mannose-6-phosphate (BTG), TGF-β antagonists from Inflazyme (Pharmaprojects No. 6075), TGF-β antagonists from Sydney, non-industrial source), TGF-β I receptor kinase inhibitors from EIi Lilly, TGF-β receptor inhibitors from Johnson & Johnson, and an analogue or derivative thereof.
3540. The device of item 3442 wherein the agent is a TN Fa antagonist or TACE inhibitor selected from the group consisting of adalimumab (CAS No. 331731-18-1) (Cambridge Antibody Technology), AGIX-4207 (AtheroGenics), AGT-1 (Advanced Biotherapy), an antiinflammatory from Borean Pharma, Cellzome, or Paradigm Therapeutics, anti-inflammatory vaccine (TNF-alpha kinoid) from Neovacs, humanized anti-TNF antibody or an anti-TNF MAb (CB0006) Celltech (UCB), apratastat (CAS No. 287445-51-0) (Wyeth), BMS-561392 (Bristol-Myers Squibb), BN- 006 (Bone), certolizumab pegol (CAS No. 428863-50-7 or CH-138 (UCB), cilomilast (CAS No. 153259-65-5) (GlaxoSmithKline), CR-1 (Nuada Pharmaceuticals), CRx-119 (CombinatoRx), D-5410 (UCB), dacopafant (CAS No. 125372-33-0) (Sanofi-Aventis), dersalazine (CAS No. 188913-57- 7/188913-58-8) (Uriach), etanercept (CAS No. 185243-69-0) (Amgen), ethyl pyruvate (Critical (Critical Therapeutics), golimumab (CAS No. 476181-74-5) (Johnson & Johnson), hormono-immunotherapy from Ipsen, CDP571 (e.g., Humicade from UCB), IC-485 (ICOS), infliximab (CAS No. 170277-31-3) (Johnson & Johnson), IP-751 (Manhattan Pharmaceuticals), ISIS-104838 (CAS No. 250755-32-9) (ISIS Pharmaceuticals), lenalidomide (CAS No. 191732-72-6) (Celgene), lentinan (CAS No. 37339-90-5) (Ajinomoto), MDL- 201112 (CAS No. 142130-73-2) (Sanofi-Aventis), medroxyprogesterone (CAS No. 520-85-4) (InKine Pharmaceutical), N-acetylcysteine (CAS No. 616-91-1) (Zambon), NBE-P2 (DIREVO Biotech), nerelimomab (CAS No. 162774-06-3) (Chiron), OM-294DP (OM PHARMA), onercept (CAS No. 199685-57-9) (Yeda), PASSTNF-alpha (Verigen), pentoxifylline or oxypentifylline (Sanofi-Aventis), Pharmaprojects No. 4091 , 4241, 4295, or 4488 (Sanofi-Aventis), Pharmaprojects No. 5480 (Amgen), Pharmaprojects No. 6749 (Cengent), pirfenidone (CAS No. 53179-13-8) (MARNAC), RPR- 132294 (Sanofi-Aventis), S5 (F002) (Fulcrum Pharmaceuticals), simvastatin (CAS No. 79902-63-9) (Merck & Co), STA-6292 (Synta Pharmaceuticals), tacrolimus (CAS No. 104987-11-3) (Fujisawa LifeCycle Pharma), talactoferrin alfa (CAS No. 308240-58-6) (Agennix), thalidomide (CAS No. 50-35-1) (Celgene), TNF antagonists form ProStrakan, and Synergen, TNF inhibitors (Amgen), TNF-alpha antagonists from Dynavax Technologies and Jerina AG (Germany), TNF-alpha inhibitors from IBFB Pharma and Xencor (Xencor), torbafylline (CAS No. 105102-21-4) (Sanofi-Aventis), UR-1505 (Uriach), VT-346 (Viron Therapeutics), YSIL6 (Y's Therapeutics), YSTH2 (Y's Therapeutics), NPI-1302a-3 (Nereus Pharmaceuticals, a TNF antagonist from Jerina AG (Germany), dersalazine, and an analogue or derivative thereof.
3541. The device of item 3442 wherein the agent is a Toll receptor inhibitor.
3542. The device of item 3442 wherein the agent is a tubulin antagonist.
3543. The device of item 3442 wherein the agent is a tyrosine kinase inhibitor selected from the group consisting of SU-011248, SUTENT from Pfizer Inc. (New York, NY), BMS-354825, PN-355 (Paracelsian Pharmaceuticals), AGN-199659 (Allergan), AAL-993 or ABP-309 (Novartis), adaphostin (NIH), AEE-788 (Novartis), AG-013736 (OSI Pharmaceuticals), AG-13736 (Pfizer), ALT-110 (Alteris Therapeutics), AMG-706 (Amgen), anticancer MAbs from Xencor, anti-EGFrvlll MAbs from Abgenix, anti-HER2 MAb from Abiogen, AZD-2171 or AZD-9935 (AstraZeneca), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), CEP-5214 (Cephalon), CEP- 7055 (Cephalon), cetuximab (ImClone Systems), CHIR-200131 and CHIR- 258 (Chiron), CP-547632 (OSI Pharmaceuticals), CP-724714 (Pfizer), CT- 301 (Creabilis Therapeutics), D-69491 (Baxter International), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EGFR/ErbB2 inhibitors from Array BioPharma, erlotinib (CAS No. 183319-69-9) (OSI Pharmaceuticals), EXEL-2880 (Exelixis), FK-778 (Sanofi-Aventis) , gefitinib (CAS No. 184475-35-2) (AstraZeneca), GW-2286 or GW-654652 (GlaxoSmithKline), her2/neu antigen from AlphaVax, HER-2/neu inhibitor from Generex, Herzyme (Medipad) (Sirna Therapeutics), HKI-272 (Wyeth), HuMax-EGFr (Genmab), idronoxil (CAS No. 81267-65-4) (Novogen), IGF-1 inhibitors from Ontogen, IMC-11 F8 (ImCIone Systems), kahalalide F (CAS No. 149204-42-2) (PharmaMar), KDR inhibitor from LG Life Sciences, KDR inhibitors from Abbott Laboratories, KDR kinase inhibitors (UCB), Kdr kinase inhibitors from Merck & Co, KRN-633 and KRN-951 (Kirin Brewery), KSB-102 (Xenova), lapatinib ditosylate (CAS No. 388082-78-8) (GlaxoSmithKline), matuzumab (Merck KGaA), MDX-214 (Medarex), ME-103 (Pharmexa), MED-A300 (Gerolymatos), MNAC-13 (Lay Line Genomics), nimotuzumab (Center of Molecular Immunology), NSC-330507 or NSC-707545 (NIH), NV-50 (Novogen), OSI-930 (OSI Pharmaceuticals), panitumumab (Abgenix), pelitinib (CAS No. 287933-82-7) (Wyeth), pertuzumab (CAS No. 380610-27- 5) (Genentech), Pharmaprojects No. 3985 (Sanofi-Aventis), prostate cancer therapeutics from Sequenom (SQPC35, SQPC36, SQPC90), removab and remoxab (Trion Pharma), RG-13022 (CAS No. 136831-48-6), RG-13291 (CAS No. 138989-50-1), or RG-14620 (CAS No. 136831-49-7) (Sanofi- Aventis), RM-6427 (Romark), RNAi breast cancer therapy from Benitec, RP 53801 (CAS No. 125882-88-4) (Sanofi-Aventis), sorafenib tosylate (Bayer), SU-11657 (Pfizer), Tie-2 antagonists from Semaia (Hybrigenics), Tie-2 inhibitors from Ontogen, trastuzumab (CAS No. 180288-69-1) (Genentech), tyrosine kinase inhibitors from Sanofi-Aventis, U3-1287, U3-1565, U3-1784, U3-1800 (U3 Pharma), vatalanib (Novartis), VEGFR-2 kinase inhibitor from Bristol-Myers Squibb, XL-647 (Exelixis), ZD-6474 (AstraZeneca), ZK-CDK (Schering AG), an EGFR tyrosine kinase inhibitor, EKB-569 (Wyeth), and an analogue or derivative thereof.
3544. The device of item 3442 wherein the agent is a VEGF inhibitor.
3545. The device of item 3442 wherein the agent is a vitamin D receptor agonist. 3546. The device of item 3442 wherein the agent is ZD-6474 (an angiogenesis inhibitor).
3547. The device of item 3442 wherein the agent is AP-23573 (an mTOR inhibitor).
3548. The device of item 3442 wherein the agent is synthadotin (a tubulin antagonist).
3549. The device of item 3442 wherein the agent is S-0885 (a collagenase inhibitor).
3550. The device of item 3442 wherein the agent is aplidine (an elongation factor-1 alpha inhibitor).
3551. The device of item 3442 wherein the agent is ixabepilone (an epithilone).
3552. The device of item 3442 wherein the agent is IDN-5390 (an angiogenesis inhibitor).
3553. The device of item 3442 wherein the agent is SB- 2723005 (an angiogenesis inhibitor).
3554. The device of item 3442 wherein the agent is ABT-518 (an angiogenesis inhibitor).
3555. The device of item 3442 wherein the agent is combretastatin (an angiogenesis inhibitor). 3556. The device of item 3442 wherein the agent is anecortave acetate (an angiogenesis inhibitor).
3557. The device of item 3442 wherein the agent is SB- 715992 (a kinesin antagonist).
3558. The device of item 3442 wherein the agent is temsirolimus (an mTOR inhibitor).
3559. The device of item 3442 wherein the agent is adalimumab (a TNFα antagonist).
3560. The device of item 3442, further comprising a polymer.
3561. The device of item 3442 wherein the anti-scarring agent inhibits adhesion between the device and a host into which the device is implanted.
3562. The device of item 3442 wherein the device delivers the anti-scarring agent locally to tissue proximate to the device.
3563. The device of item 3442, further comprising a coating, wherein the coating comprises the anti-scarring agent.
3564. The device of item 3442, further comprising a coating, wherein the coating is disposed on a surface of the device.
3565. The device of item 3442, further comprising a coating, wherein the coating directly contacts the device. 3566. The device of item 3442, further comprising a coating, wherein the coating indirectly contacts the device.
3567. The device of item 3442, further comprising a coating, wherein the coating partially covers the device.
3568. The device of item 3442, further comprising a coating, wherein the coating completely covers the device.
3569. The device of item 3442, further comprising a coating, wherein the coating is a uniform coating.
3570. The device of item 3442, further comprising a coating, wherein the coating is a non-uniform coating.
3571. The device of item 3442, -further comprising a coating, wherein the coating is a discontinuous coating.
3572. The device of item 3442, further comprising a coating, wherein the coating is a patterned coating.
3573. The device of item 3442, further comprising a coating, wherein the coating has a thickness of 100 Dm or less.
3574. The device of item 3442, further comprising a coating, wherein the coating has a thickness of 10 Dm or less.
3575. The device of item 3442, further comprising a coating, wherein the coating adheres to the surface of the device upon deployment of the device. 3576. The device of item 3442, further comprising a coating, wherein the coating is stable at room temperature for a period of 1 year.
3577. The device of item 3442, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 0.0001% to about 1% by weight.
3578. The device of item 3442, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 1% to about 10% by weight.
3579. The device of item 3442, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 10% to about 25% by weight.
- 3580. The device of item 3442, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 25% to about 70% by weight.
3581. The device of item 3442, further comprising a coating, wherein the coating further comprises a polymer.
3582. The device of item 3442, further comprising a first coating having a first composition and the second coating having a second composition.
3583. The device of item 3442, further comprising a first coating having a first composition and the second coating having a second composition, wherein the first composition and the second composition are different. 3584. The device of item 3442, further comprising a polymeric carrier, wherein the polymeric carrier comprises a copolymer.
3585. The device of item 3442, further comprising a polymeric carrier, wherein the polymeric carrier comprises a block copolymer.
3586. The device of item 3442, further comprising a polymeric carrier, wherein the polymeric carrier comprises a random copolymer.
3587. The device of item 3442, further comprising a polymeric carrier, wherein the polymeric carrier comprises a biodegradable polymer.
3588. The device of item 3442, further comprising a polymeric carrier, wherein the polymeric carrier comprises a non-biodegradable polymer.
3589. The device of item 3442, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrophilic polymer.
3590. The device of item 3442, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrophobic polymer.
3591. The device of item 3442, further comprising a polymeric carrier, wherein the polymeric carrier comprises a polymer having hydrophilic domains.
3592. The device of item 3442, further comprising a polymeric carrier, wherein the polymeric carrier comprises a polymer having hydrophobic domains. 3593. The device of item 3442, further comprising a polymeric carrier, wherein the polymeric carrier comprises a non-conductive polymer.
3594. The device of item 3442, further comprising a polymeric carrier, wherein the polymeric carrier comprises an elastomer.
3595. The device of item 3442, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrogel.
3596. The device of item 3442, further comprising a polymeric carrier, wherein the polymeric carrier comprises a silicone polymer.
3597. The device of item 3442, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrocarbon polymer.
3598. The device of item 3442, -further comprising a polymeric carrier, wherein the polymeric carrier comprises a styrene-derived polymer.
3599. The device of item 3442, further comprising a polymeric carrier, wherein the polymeric carrier comprises a butadiene polymer.
3600. The device of item 3442, further comprising a polymeric carrier, wherein the polymeric carrier comprises a macromer.
3601. The device of item 3442, further comprising a polymeric carrier, wherein the polymeric carrier comprises a poly(ethylene glycol) polymer.
3602. The device of item 3442, further comprising a polymeric carrier, wherein the polymeric carrier comprises an amorphous polymer. 3603. The device of item 3442, further comprising a lubricious coating.
3604. The device of item 3442 wherein the anti-scarring agent is located within pores or holes of the device.
3605. The device of item 3442 wherein the anti-scarring agent is located within a channel, lumen, or divet of the device.
3606. The device of item 3442, further comprising a second pharmaceutically active agent.
3607. The device of item 3442, further comprising an antiinflammatory agent.
3608. The device of item 3442, further comprising an agent that inhibits infection.
3609. The device of item 3442, further comprising an agent that inhibits infection, wherein the agent is an anthracycline.
3610. The device of item 3442, further comprising an agent that inhibits infection, wherein the agent is doxorubicin.
3611. The device of item 3442, further comprising an agent that inhibits infection, wherein the agent is mitoxantrone.
3612. The device of item 3442, further comprising an agent that inhibits infection, wherein the agent is a fluoropyrimidine. 3613. The device of item 3442, further comprising an agent that inhibits infection, wherein the agent is 5-fluorouracil (5-FU).
3614. The device of item 3442, further comprising an agent that inhibits infection, wherein the agent is a folic acid antagonist.
3615. The device of item 3442, further comprising an agent that inhibits infection, wherein the agent is methotrexate.
3616. The device of item 3442, further comprising an agent that inhibits infection, wherein the agent is a podophylotoxin.
3617. The device of item 3442, further comprising an agent that inhibits infection, wherein the agent is etoposide.
3618. The device of item 3442, further comprising an agent that inhibits infection, wherein the agent is a camptothecin.
3619. The device of item 3442, further comprising an agent that inhibits infection, wherein the agent is a hydroxyurea.
3620. The device of item 3442, further comprising an agent that inhibits infection, wherein the agent is a platinum complex.
3621. The device of item 3442, further comprising an agent that inhibits infection, wherein the agent is cisplatin.
3622. The device of item 3442, further comprising an antithrombotic agent. 3623. The device of item 3442, further comprising a visualization agent.
3624. The device of item 3442, further comprising a visualization agent, wherein the visualization agent is a radiopaque material, wherein the radiopaque material comprises a metal, a halogenated compound, or a barium containing compound.
3625. The device of item 3442, further comprising a visualization agent, wherein the visualization agent is a radiopaque material, wherein the radiopaque material comprises barium, tantalum, or technetium.
3626. The device of item 3442, further comprising a visualization agent, wherein the visualization agent is a MRI responsive material.
3627. The device of item 3442, further comprising a visualization agent, wherein the visualization agent comprises a gadolinium chelate.
3628. The device of item 3442, further comprising a visualization agent, wherein the visualization agent comprises iron, magnesium, manganese, copper, or chromium.
3629. The device of item 3442, further comprising a visualization agent, wherein the visualization agent comprises an iron oxide compound.
3630. The device of item 3442, further comprising a visualization agent, wherein the visualization agent comprises a dye, pigment, or colorant. 3631. The device of item 3442, further comprising an echogenic material.
3632. The device of item 3442, further comprising an echogenic material, wherein the echogenic material is in the form of a coating.
3633. The device of item 3442 wherein the device is sterile.
3634. The device of item 3442 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device.
3635. The device of item 3442 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein-the tissue is connective-tissue.
3636. The device of item 3442 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is muscle tissue.
3637. The device of item 3442 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is nerve tissue.
3638. The device of item 3442 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is epithelium tissue. 3639. The device of item 3442 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from the time of deployment of the device to about 1 year.
3640. The device of item 3442 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from about 1 month to 6 months.
3641. The device of item 3442 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from about 1 - 90 days.
3642. The device of item 3442 wherein the anti-scarring agent is released in effective concentrations from the device at a constant rate.
3643. The device of item 3442 wherein the anti-scarring agent is released in effective concentrations from the device at an increasing rate.
3644. The device of item 3442 wherein the anti-scarring agent is released in effective concentrations from the device at a decreasing rate.
3645. The device of item 3442 wherein the anti-scarring agent is released in effective concentrations from the composition comprising the anti-scarring agent by diffusion over a period ranging from the time of deployment of the device to about 90 days.
3646. The device of item 3442 wherein the anti-scarring agent is released in effective concentrations from the composition comprising the anti-scarring agent by erosion of the composition over a period ranging from the time of deployment of the device to about 90 days. 3647. The device of item 3442 wherein the device comprises about 0.01 Dg to about 10 Dg of the anti-scarring agent.
3648. The device of item 3442 wherein the device comprises about 10 Dg to about 10 mg of the anti-scarring agent.
3649. The device of item 3442 wherein the device comprises about 10 mg to about 250 mg of the anti-scarring agent.
3650. The device of item 3442 wherein the device comprises about 250 mg to about 1000 mg of the anti-scarring agent.
3651. The device of item 3442 wherein the device comprises about 1000 mg to about 2500 mg of the anti-scarring agent.
3652. The device of item 3442 wherein-a surface of the device comprises less than 0.01 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
3653. The device of item 3442 wherein a surface of the device comprises about 0.01 Dg to about 1 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
3654. The device of item 3442 wherein a surface of the device comprises about 1 Dg to about 10 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
3655. The device of item 3442 wherein a surface of the device comprises about 10 Dg to about 250 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied. 3656. The device of item 3442 wherein a surface of the device comprises about 250 Dg to about 1000 Dg of the anti-scarring agent of anti- scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
3657. The device of item 3442 wherein a surface of the device comprises about 1000 Dg to about 2500 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
3658. A method as in any one of items 3442-3657, wherein the device is an endotracheal tube.
3659. A method as in any one of items 3442-3657, wherein the device is an endotracheal tube with a single lumen.
3660. A method as in any one of items 3442-3657, wherein the device is an endotracheal tube with double lumens.
3661. A method as in any one of items 3442-3657, wherein the device is a tracheostomy tube.
3662. A device, comprising a peritoneal dialysis catheter (e.g., an implant) and an anti-scarring agent or a composition comprising an anti- scarring agent, wherein the agent inhibits scarring between the device and a host into which the device is implanted.
3663. The device of item 3662 wherein the agent is an adensosine A2A receptor antagonist.
3664. The device of item 3662 wherein the agent is an AKT inhibitor. 3665. The device of item 3662 wherein the agent is an alpha 2 integrin antagonist, wherein the alpha 2 integrin antagonist is Pharmaprojects No. 5754 (Merck KgaA).
3666. The device of item 3662 wherein the agent an alpha 4 integrin antagonist.
3667. The device of item 3662 wherein the agent is an alpha 7 nicotinic receptor agonist.
3668. The device of item 3662 wherein the agent is an angiogenesis inhibitor selected from the group consisting of AG-12,958 (Pfizer), ATN-161 (Attention LLC), neovastat, an angiogenesis inhibitor from Jerina AG (Germany), NM-3 (Mercian), VGA-1155 (Taisho), FCE-26644 (Pfizer), FCE-26950 (Pfizer), FPMA (Meiji Daries), FR-111142 (Fujisawa), GGTI-298, GM-1306 (Ligand), GPA-1734 (Novartis}, NNC-47-001-1 (Novo Nordisk), herbamycin (Nippon Kayaku), lenalidomide (Celegene), IP-10 (NIH), ABT-828 (Abbott), KIN-841 (Tokushima University, Japan), SF-1126 (Semafore Pharmaceuticals), laminin technology (NIH), CHIR-258 (Chiron), NVP-AEW541 (Novartis), NVP-AEW541 (Novartis), Vt16907 (Alchemia), OXI-8007 (Oxigene), EG-3306 (Ark Therapeutics), Maspin (Arriva), ABT- 567 (Abbott), PPI-2458 (Praecis Pharmaceuticals), CC-5079, CC-4089 (Celgene), HIF-1 alpha inhibitors (Xenova), S-247 (Pfizer), AP-23573 (Ariad), AZD-9935 (Astra Zeneca), mebendazole (Introgen Therapeutics), MetAP-2 inhibitors (GlaxoSmithKline), AG-615 (Angiogene Pharmaceuticals), Tie-2 antagonists (Hybrigenics), NC-381 , CYC-381 , NC-169, NC-219, NC-383, NC-384, NC-407 (Lorus Therapeutics), ATN-224 (Attenuon), ON-01370 (Onconova), Vitronectin antagonists (Amgen), SDX-103 (Salmedix), Vitronectin antagonists (Shire), CHP (Riemser), TEK (Amgen), Anecortave acetate (Alcon), T46.2 (Matrix Therapeutics), HG-2 (Heptagen), TEM antagonists (Genzyme), Oxϊ-4500 (Oxigene), ATN-161 (Attenuon), WX-293 (Wilex), M-2025 (Metris Therapeutics), Alphastatin (BioActa), YH-16 (Yantai Rongchang), BIBF-1120 (Boehringer Ingelheim), BAY-57-9352 (Bayer), AS- 1404 (Cancer Research Technology), SC-77964 (Pfizer), glycomimetics (BioTie Therapies), TIE-2 Inhibitors (Ontogen), DIMI, Octamer (Octamer), ABR-215050 (Active Biotech), ABT-518 (Abbott), KDR inhibitors (Abbott), BSF-466895 (Abbott), SCH-221153 (Schering-Plough), DAC:antiangiogenic (ConjuChem), TFPI (EntreMed), AZD-2171 (Astra-Zenaca), CDC-394 (Celgene), LY290293 (EIi Lilly), IDN-5390 (Indena), Kdr Kinase Inhibitors (Merck), CT-113020, CT-116433, CT-116563, CT-31890, CT-32228) (Cell Therapeutics), A-299620 (Abbott), TWEAK Inhibitor (Amgen), VEGF modulators (Johnson and Johnson), Tum-N53, tumstatin (Genzyme), Thios- 1 , Thios-2 (Thios Pharmaceuticals), MV-6401 (Miravant Medical Technologies), Spisulosine (PharmaMar), CEP-7055 (Cephalon), AUV-201 (Auvation), LM-609 (EIi Lilly), SKF-106615 (AnorMED), Oglufanide disodium (Cytran), BW-114 (Pharminox), Calreticulin (NIH), WX-678 (Wilex), SD-7784 (Pfizer), WX-UK1 (Wilex), SH-268 (Schering AG), 2-Me-PGA (Celgene), S- 137 (Pfizer), ZD-6126 (Angiogene Pharmaceuticals), SG-292 (SignalGen), Benefin (Lane Labs), A6, A36 (Angstrom), SB-2723005 (GlaxoSmithKline), SC-7 (Cell Therapeutics), ZEN-014 (AEterna Zentaris), 2-methoxyestradiol (EntreMed), NK-130119 (Nippon Kayaku), CC-10004 (Celgene), AVE- 8062A (Ajinomoto), Tacedinaline (Pfizer), Actinonin (Tokyo Metropolitan Institute of Medical Science), Lenalidomide (Celgene), VGA-1155, BTO-956 (SRI International), ER-68203-00 (Eisai), CT-6685 (UCB), JKC-362 (Phoenix Pharmaceuticals), DMI-3798 (DMI Biosciences, Angiomate (Ipsen), ZD-6474 (AstraZeneca), CEP-5214 (Cephalon), Canstatin (Genzyme), NM-3 (Mercian), Oridigm (MediQuest Therapeutics), Exherin (Adherex), BLS-0597 (Boston Life Sciences), PTC-299 (PTC Therapeutics), NPI-2358 (Nereus Pharmaceuticals), CGP-79787 (Novartis), AEE-788 (Novartis), CKD-732 (Chong Kun Dang), CP-564959 (OSI Pharmaceuticals), CM-101 (CarboMed), CT-2584, CT3501 (Cell Therapeutics), combretastatin and analogues and derivatives thereof (Oxigene), Rebimastat (Bristol-Meyers Squibb), Dextrin 2-sulfate (ML Laboratories), Cilengitide (Merk KGaA), NSC-706704 (Pharminox), KRN- 951 (Kirin Brewery), Ukrain, NSC-631570 (Nowicky Pharma), Tecogalan sodium (Daiichi Pharmaceutical), Tz-93 (Tsumura), TBC-1635 (Encysive Pharmaceuticals), TAN-1120 (Takeda), Semaxanib (Pfizer), BDI-7800 (Biopharmacopae), SD-186, SD-983 (Bristol-Meyers Squibb), SB-223245 (GlaxoSmithKline), SC-236 (Pfizer), RWJ-590973 (Johnson and Johnson), ILX-1850 (Genzyme), SC-68488, S-836 (Pfizer), CG-55069-11 (CuraGen), Ki-23057 (Kirin Brewery), CCX-700 (Chemoentryx), Pegaptanib octasodium (Giled Sciences), ANGIOCOL (available from Biostratum Inc.), or an analogue or derivative thereof.
3669. The device of item 3662 wherein the agent is an apoptosis antagonist.
3670. The device of item 3662 wherein the agent is an apoptosis activator.
3671. The device of item 3662 wherein the agent is a beta 1 integrin antagonist.
3672. The device of item 3662 wherein the agent is a beta tubulin inhibitor.
3673. The device of item 3662 wherein the agent is a blocker of enzyme production in Hepatitis C.
3674. The device of item 3662 wherein the agent is a Bruton's tyrosine kinase inhibitor. 3675. The device of item 3662 wherein the agent is a calcineurin inhibitor.
3676. The device of item 3662 wherein the agent is a caspase 3 inhibitor.
3677. The device of item 3662 wherein the agent is a CC chemokine receptor antagonist.
3678. The device of item 3662 wherein the agent is a cell cycle inhibitor selected from the group consisting of SNS-595 (Sunesis), synthadotin, KRX-0403, and an analogue or derivative thereof.
3679. The device of item 3662 wherein the agent is a cathepsin B inhibitor.
3680. The device of item 3662 wherein the agent is a cathepsin K inhibitor, wherein the cathepsin K inhibitor is 462795 (GlaxoSmithKline), INPL-022-D6 (Amura Therapeutics), an analogue or derivative thereof).
3681. The device of item 3662 wherein the agent is a cathepsin L inhibitor.
3682. The device of item 3662 wherein the agent is a CD40 antagonist.
3683. The device of item 3662 wherein the agent is a chemokine receptor agonist. 3684. The device of item 3662 wherein the agent is a chymase inhibitor.
3685. The device of item 3662 wherein the agent is a collagenase antagonist.
3686. The device of item 3662 wherein the agent is a CXCR antagonist.
3687. The device of item 3662 wherein the agent is a cyclin dependent kinase inhibitor selected from the group consisting of a CDK-1 inhibitor, a CDK-2 inhibitor, a CDK- 4 inhibitor, a CDK-6 inhibitor, a CAK1 inhibitor from GPC Biotech and Bristol-Myers Squibb, RGB-286199 (GPC Biotech), an anticancer agent from Astex Technology, a CAK1 inhibitor from GPC Biotech, a CDK inhibitor from Sanofi-Aventis, a CDK1/CDK2 inhibitor from Amgen, a CDK2 inhibitor from SUGEN-2 (Pfizer), a hearing loss therapy agent (Sound Pharmaceuticals), PD-0332991 (Pfizer), RGB-286199 (GPC Biotech), Ro-0505124 (Hoffmann-La Roche), a Ser/Thr kinase inhibitor from Lilly (EIi Lilly), CVT-2584 (CAS No. 199986-75-9) (CV Therapeutics), and an analogue or derivative thereof.
3688. The device of item 3662 wherein the agent is a cyclooxygenase 1 inhibitor.
3689. The device of item 3662 wherein the agent is a DHFR inhibitor.
3690. The device of item 3662 wherein the agent is a dual integrin inhibitor. 3691. The device of item 3662 wherein the agent is an elastase inhibitor.
3692. The device of item 3662 wherein the agent is an elongation factor-1 alpha inhibitor.
3693. The device of item 3662 wherein the agent is an endothelial growth factor antagonist.
3694. The device of item 3662 wherein the agent is an endothelial growth factor receptor kinase inhibitor selected from the group consisting of sorafenib tosylate (Bayer), AAL-993 (Novartis), ABP-309 (Novartis), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), E- 7080 (Eisai), EG-3306 (Ark Therapeutics), EXEL-2880 (Exelixis), GW- 654652 (GlaxoSmithKline), a KDR inhibitor from LG Life Sciences, CT-6685 and CT-6729 (UCB), KRN-633 and KRN-951 (Kirin Brewery), OSI-930 (OSI Pharmaceuticals), SP-5.2 (Supratek Pharma), SU-11657 (Pfizer), a Tie-2 antagonist (Hybrigenics), a VEGFR-2 kinase inhibitor (Bristol-Myers Squibb), XL-647 (Exelixis), a KDR inhibitor from Abbott Laboratories, sorafenib tosylate, and an analogue or derivative thereof.
3695. The device of item 3662 wherein the agent is an endotoxin antagonist.
3696. The device of item 3662 wherein the agent is an epothilone and tubulin binder.
3697. The device of item 3662 wherein the agent is an estrogen receptor antagonist. 3698. The device of item 3662 wherein the agent is an FGF inhibitor.
3699. The device of item 3662 wherein the agent is a farnexyl transferase inhibitor.
3700. The device of item 3662 wherein the agent is farnesyltransferase inhibitor selected from the group of A-197574 (Abbott), a famesyltransferase inhibitor from Servier, FPTIII (Strathclyde Institute for Drug R), LB-42908 (LG Life Sciences), Pharmaprojects No. 5063 (Genzyme), Pharmaprojects No. 5597 (Ipsen), Yissum Project No. B-1055 (Yissum), and an analogue or derivative thereof.
3701. The device of item 3662 wherein the agent is an FLT-3 kinase inhibitor.
3702. The device of item 3662 wherein the agent is an FGF receptor kinase inhibitor.
3703. The device of item 3662 wherein the agent is a fibrinogen antagonist selected from the group consisting of AUV-201 (Auvation), MG-13926 (Sanofi-Aventis), plasminogen activator (CAS No. 105913-11-9) (from Sanofi-Aventis or UCB), plasminogen activator-2 (tPA- 2) (Sanofi-Aventis), pro-urokinase (CAS No. 82657-92-9) (Sanofi-Aventis), and an analogue or derivative thereof.
3704. The device of item 3662 wherein the agent is a heat shock protein 90 antagonist selected from the group consisting of SRN-005 (Sirenade), geldanamycin, NSC-33050 (17-allylaminogeldanamycin; 17- AAG), 17-dimethylaminoethylamino-17-demethoxy-geldanamycin (17- DMAG), rifabutin (rifamycin XIV1 1\4-didehydro-1-deoxy-1 ,4-dihydro-5'-(2- methylpropyl)-1-oxo-), and an analogue or derivative thereof.
3705. The device of item 3662 wherein the agent is a histone deacetylase inhibitor.
3706. The device of item 3662 wherein the agent is an HMGCoA reductase inhibitor selected from the group consisting of an atherosclerosis therapeutic from Lipid Sciences, ATI-16000 (ARYx Therapeutics), KS-01-019 (Kos Pharmaceuticals), Pharmaprojects No. 2197 (Sanofi-Aventi), RP 61969 (Sanofi-Aventis), and an analogue or derivative thereof.
3707. The device of item 3662 wherein the agent is an ICAM inhibitor.
3708. The device of item 3662 wherein the agent is an IL, ICE and IRAK antagonist, wherein the antagonist is a CJ-14877, CP-424174 (Pfizer), NF-61 (Negma-Lerads), and an analogue or derivative thereof.
3709. The device of item 3662 wherein the agent is an IL-2 inhibitor.
3710. The device of item 3662 wherein the agent is an immunosuppressant selected from the group consisting of teriflunomide (Sanofi Aventis), chlorsulfaquinoxalone (NSC-339004), chlorsulfaquinoxalone sulfate, CS-712 (Sankyo), ismomultin alfa (CAS No. 457913-93-8) (Akzo Nobel), antiallergics from GenPat77, antiinflammatories or AT-005 (Androclus Therapeutics), autoimmune disease therapeutics from EpiVax, BN-007 (Bone), budesonide (CAS No. 51333-22- 3) (MAP Pharmaceuticals), CO-14 (Genzyme), edratide (CAS No. 433922- 67-9) (Teva), EP-314 (Enanta), eprovafen (CAS No. 101335-99-3) (Sanofi- Aventis), HWA-131 (CAS No. 118788-41-3) (Sanofi-Aventis), immunomodulators from MerLion Pharmaceuticals, immunosuppressives from Alchemia, IPL-12 (Inflazyme), MDL-9563 (CAS No. 27086-86-8) (Sanofi-Aventis), Pharmaprojects No. 2330 (Sanofi-Aventis), Pharmaprojects No. 6426 (Abgenix), PXS-25 (Pharmaxis), rosmarinic acid (CAS No. 20283-92-5) (Sanofi-Aventis), RP 42927 or RP 54745 (CAS No. 135330-08-4) (Sanofi-Aventis), SGN-35 (Seattle Genetics), ST-1959 (Sigma-Tau), type I diabetes therapy from SYNX Pharma, UNIL-88 (Debiopharm), VP-025 (Vasogen), VR-694 (Vectura), PRTX-001 (Protalex), and an analogue or derivative thereof.
3711. The device of item 3662 wherein the agent is an IMPDH (inosine monophosphate).
- - - -3712. The device of item 3662 wherein the agent is an integrin antagonist.
3713. The device of item 3662 wherein the agent is an interleukin antagonist.
3714. The device of item 3662 wherein the agent is an inhibitor of type III receptor tyrosine kinase.
3715. The device of item 3662 wherein the agent is an irreversible inhibitor of enzyme methionine aminopeptidase type 2.
3716. The device of item 3662 wherein the agent is an isozyme selective delta protein kinase C inhibitor. 3717. The device of item 3662 wherein the agent a JAK3 enzyme inhibitor.
3718. The device of item 3662 wherein the agent is a JNK inhibitor.
3719. The device of item 3662 wherein the agent is a kinase inhibitor.
3720. The device of item 3662 wherein the agent is kinesin antagonist.
3721. The device of item 3662 wherein the agent is a kinesin antagonist.
3722. The device of item 3662-wherein the-agent is a leukotriene inhibitor and antagonist selected from the group consisting of ambicromil (CAS No. 58805-38-2) (Sanofi-Aventis), amelubant (CAS No. 346735-24-8) (Boehringer Ingelheim), DW-1141 (Dong Wha), ebselen (Daiichi Pharmaceutical), ibudilast (Kyorin), leucotriene inhibitors from Sanofi-Aventis, lymphotoxin-beta receptor (LT-D) from Biogen Idee, Pharmaprojects No. 1535 and 2728 (CAS No. 119340-33-9) (Sanofi- Aventis), R-112 (Rigel), Rev-5367 (CAS No. 92532-05-3) (Sanofi-Aventis), RG-14893 (CAS No. 141835-49-6) (Sanofi-Aventis), RG-5901-A (CAS No. 101910-24-1), 92532-23-5, RP 66153 (CAS No. 142422-79-5), RP 66364 (CAS No.186916-92-5), RP 69698 (CAS No. 141748-00-7) (Sanofi-Aventis), SC-411930 (Pfizer), SC-41930 (CAS No. 120072-59-5) (Pfizer), SC-50605 (CAS No. 138828-39-4) (Pfizer), SC-51146 (CAS No. 141059-52-1), SC- 53228 (CAS No. 153633-01-3) (Pfizer), spaglumic acid (ZY-15106) (CAS No.3106-85-2), 80619-64-3 (Novartis), tipredane (CAS No. 85197-77-9) (Bristol-Myers Squibb), U-75302 (CAS No. 119477-85-9) (Pfizer), and analogue or derivative thereof.
3723. The device of item 3662 wherein the agent is an MAP kinase inhibitor.
3724. The device of item 3662 wherein the agent is a matrix metalloproteinase inhibitor.
3725. The device of item 3662 wherein the agent is an MCP- CCR2 inhibitor.
3726. The device of item 3662 wherein the agent is an mTOR inhibitor.
3727. The device of item 3662 wherein the agent is an mTOR kinase inhibitor.
3728. The device of item 3662 wherein the agent is a microtubule inhibitor selected from the group consisting of antibody- maytansinoid conjugates from Biogen Idee, colchicines (MantiCore Pharmaceuticals), anticancer immunoconjugates from Johnson & Johnson, DIME from Octamer, gni-1f (GNI), huC242-DM4, huMy9-6-DM1 (ImmunoGen), IDN-5404 (Indena), IMO-098, IMOderm (Imotep), mebendazole (Introgen Therapeutics), microtubule poisons from Cambridge Enterprise, paclitaxel such as LOTAX from Aphios (CAS No. 33069-62-4), Genexol-PM from Samyang, Pharmaprojects No. 6383 (Tapestry Pharmaceuticals), RPR-112378 (Sanofi-Aventis), SGN-75 (Seattle Genetics), SPL-7435 (Starpharma), SSR-250411 (Sanofi-Aventis), trastuzumab-DM1 (Genentech), vinorelbine, and an analogue or derivative thereof. 3729. The device of item 3662 wherein the agent is an MIF inhibitor.
3730. The device of item 3662 wherein the agent is an MMP inhibitor.
3731. The device of item 3662 wherein the agent is a neurokinin (NK) antagonist selected from the group consisting of anthrotainin (CAS No. 148084-40-6) (Sanofi-Aventis), an IBS thereapeutic from ArQuIe, MDL-105212A (CAS No. 167261-60-1) (Ssanofi-Aventis), Pharmaprojects No. 2744, 3258 (CAS No. 139167-47-8) 4006, 4201 , or 5986 (Sanofi-Aventis), RP 67580 (CAS No. 135911-02-3), SR-144190 (CAS No. 201152-86-5), SSR-240600, SSR-241586 (Sanofi-Aventis), TKA-457 (Novartis), vestipitant mesylate (CAS No. 334476-64-1) (GlaxoSmithKline), Win-64821 (Sanofi-Aventis), PRX-96026 (Predix Pharmaceuticals), and an analogue or derivative thereof. - - -
3732. The device of item 3662 wherein the agent is an NF kappa B inhibitor selected from the group consisting of AVE-0545 or AVE- 0547 (Sanofi-Aventis), bortezomib (CAS No. 179324-69-7) (Millennium Pharmaceuticals), dexanabinol (CAS No. 112924-45-5) (Pharmos), dexlipotam (Viatris), Pharmaprojects No. 6283 (INDRA) (OXiGENE), IPL- 576092 (CAS No. 137571-30-3) (Inflazyme), NFKB decoy (Corgentech), NFKB decoy oligo (AnGes MG), NFKB's from Ariad, osteoporosis treatments or S5 (F005) from Fulcrum Pharmaceuticals, P61 (Phytopharm), R-flurbiprofen (CAS No. 5104-49-4) (Encore Pharmaceuticals), and an analogue or derivative thereof.
3733. The device of item 3662 wherein the agent is a nitric oxide agonist. 3734. The device of item 3662 wherein the agent is an ornithine decarboxylase inhibitor.
3735. The device of item 3662 wherein the agent is a p38 MAP kinase inhibitor selected from the group consisting of AZD-6703 (AstraZeneca), JX-401 (Jexys Pharmaceuticals), BMS-2 (Bristol-Myers Squibb), a p38 MAP kinase inhibitor from Novartis, a p38-alpha MAP kinase inhibitor from Amphora, Pharmaprojects No. 5704 (Pharmacopeia), RPR- 200765A (Sanofi-Aventis), SD-282 (Johnson & Johnson), TAK-715 (Takeda), and an analogue or derivative thereof.
3736. The device of item 3662 wherein the agent is a palmitoyl-protein thioesterase inhibitor.
3737. The device of item 3662 wherein the agent is a PDGF receptor kinase inhibitor selected from the group consisting of AAL-993, AMN-107, or ABP-309 (Novartis), AMG-706 (Amgen), BAY-57-9352 (Bayer), CDP-860 (UCB), E-7080 (Eisai), imatinib (CAS No. 152459-95-5) (Novartis), OSI-930 (OSI Pharmaceuticals), RPR-127963E (Sanofi-Aventis), RWJ-540973 (Johnson & Johnson), sorafenib tosylate (Bayer), SU-11657 (Pfizer), tandutinib (CAS No. 387867-13-2) (Millennium Pharmaceuticals), vatalanib (Novartis), ZK-CDK (Schering AG), and an analogue or derivative thereof.
3738. The device of item 3662 wherein the agent is (-)- halofenate (Metabolex), AMG-131 (Amgen), antidiabetics from Japan Tobacco, AZD-4619, AZD-8450, AZD-8677 (AstraZeneca), DRF-10945, balaglitazone (Dr Reddy's), CS-00088, CS-00098 (Chipscreen Biosciences), E-3030 (Eisai), etalocib (CAS No. 161172-51-6) (EIi Lilly), GSK-641597 (Ligand), GSK-677954 (GlaxoSmithKline), GW-409544 (Ligand), GW- 590735 (GlaxoSmithKline), K-111 (Hoffmann-La Roche), LY-518674 (EIi Lilly), LY-674 (Ligand), LY-929 (Ligand), MC-3001 , MC-3002 (MaxoCore Pharmaceuticals), metformin HCI + pioglitazone (CAS No. 1115-70-4 and 112529-15-4), ACTOPLUS MET from Andrx), muraglitazar (CAS No. 331741-94-7) (Bristol-Myers Squibb), naveglitazar (Ligand), oleoylethanolamide (Kadmus Pharmaceuticals), ONO-5129, pioglitazone hydrochloride (CAS No. 111025-46-8 and 112529-15-4) (Takeda), PLX-204 (Plexxikon), PPAR agonists from Genfit, PPAR delta agonists from EIi Lilly, PPAR-alpha agonists from CrystalGenomics, PPAR-gamma modulators and PPAR-β modulators from CareX, rosiglitazone maleate (CAS No. 122324- 73-4 or 155141-29-0) (GlaxoSmithKline), rosiglitazone maleate/glimepir (CAS No. 155141-29-0 and 93479-97-1), AVANDARYL, rosiglitazone maleate/metformin extend (CAS No. 155141-29-0 and 657-24-9), AVANDAMET, rosiglitazone maleate+metformin, AVANDAMET (GlaxoSmithKline), tesaglitazar (AstraZeneca), and an analogue or derivative thereof.
3739. The device of item 3662 wherein the agent is a phosphatase inhibitor.
3740. The device of item 3662 wherein the agent is a phosphodiesterase (PDE) inhibitor selected from the group consisting of avanafil (Tanabe Seiyaku), dasantafil (CAS No. 569351-91-3) (Schering- Plough), A-906119 (CAS No. 134072-58-5), DL-850 (Sanofi-Aventis), GRC- 3015, GRC-3566, GRC-3886 (Glenmark), HWA-153 (CAS No. 56395-66-5) (Sanofi-Aventis), hydroxypumafentrine (Altana), IBFB-130011 , IBFB-14-016, IBFB-140301, IBFB-150007, IBFB-211913 (IBFB Pharma), L-826141 (Merck & Co), medorinone (CAS No. 88296-61-1) (Sanofi-Aventis), MEM- 1917 (Memory Pharmaceuticals), ND-1251 (Neuro3d), PDE inhibitors from ICOS, PDE IV inhibitors from Memory Pharmaceuticals and CrystalGenomics, Pharmaprojects No. 2742 and 6141 (Sanofi-Aventis), QAD-171 (Novartis), RHC-2963 (CAS No. 76993-12-9 and 76993-14-1), RPR-117658, RPR-122818 derivatives, SR-24870 , and RPR-132294 (Sanofi-Aventis), SK-350 (ln2Gen), stroke therapy agents from deCODE Genetics, TAS-203 (Taiho), tofimilast (CAS No. 185954-27-2) (Pfizer), UK- 371800 (Pfizer), WIN-65579 (CAS No. 158020-82-7) (Sanofi-Aventis), IBFB- 130020 (IBFB Pharma), OPC-6535 (CAS No. 145739-56-6) (Otsuka), a phosphodiesterase III inhibitor, enoximone, a phosphodiesterase IV inhibitor, fosfosal, Atopik (Barrier Therapeutics), triflusal, a phosphodiesterase V inhibitor, and an analogue or derivative thereof.
3741. The device of item 3662 wherein the agent is a PKC inhibitor.
3742. The device of item 3662 wherein the agent is a platelet activating factor antagonist.
- - 3743. The device of item 3662 wherein the agent is a platelet- derived growth factor receptor kinase inhibitor.
3744. The device of item 3662 wherein the agent is a prolyl hydroxylase inhibitor.
3745. The device of item 3662 wherein the agent is a polymorphonuclear neutrophil inhibitor.
3746. The device of item 3662 wherein the agent is a protein kinase B inhibitor.
3747. The device of item 3662 wherein the agent is a protein kinase C stimulant. 3748. The device of item 3662 wherein the agent is a purine nucleoside analogue.
3749. The device of item 3662 wherein the agent is a purinoreceptor P2X antagonist.
3750. The device of item 3662 wherein the agent is a Raf kinase inhibitor.
3751. The device of item 3662 wherein the agent is a reversible inhibitor of ErbB1 and ErbB2.
3752. The device of item 3662 wherein the agent is a ribonucleoside triphosphate reductase inhibitor.
3753. The device of item 3662 wherein the agent is an SDF-1 antagonist.
3754. The device of item 3662 wherein the agent is a sheddase inhibitor.
3755. The device of item 3662 wherein the agent is an SRC inhibitor.
3756. The device of item 3662 wherein the agent is a stromelysin inhibitor.
3757. The device of item 3662 wherein the agent is an Syk kinase inhibitor. 3758. The device of item 3662 wherein the agent is a telomerase inhibitor.
3759. The device of item 3662 wherein the agent is a TGF beta inhibitor selected from the group consisting of pirfenidone (CAS No. 53179-13-8) (MARNAC), tranilast (CAS No. 53902-12-8) (Kissei), IN-1130 (ln2Gen), mannose-6-phosphate (BTG), TGF-β antagonists from Inflazyme (Pharmaprojects No. 6075), TGF-β antagonists from Sydney, non-industrial source), TGF-β I receptor kinase inhibitors from EIi Lilly, TGF-β receptor inhibitors from Johnson & Johnson, and an analogue or derivative thereof.
3760. The device of item 3662 wherein the agent is a TN Fa antagonist or TACE inhibitor selected from the group consisting of adalimumab (CAS No. 331731-18-1) (Cambridge Antibody Technology), AGIX-4207 (AtheroGenics), AGT-1 (Advanced Biotherapy), an anti- inflammatoiy from Borean Pharma, Cellzome, or Paradigm Therapeutics, anti-inflammatory vaccine (TNF-alpha kinoid) from Neovacs, humanized anti-TNF antibody or an anti-TNF MAb (CB0006) Celltech (UCB), apratastat (CAS No. 287445-51-0) (Wyeth), BMS-561392 (Bristol-Myers Squibb), BN- 006 (Bone), certolizumab pegol (CAS No. 428863-50-7 or CH-138 (UCB), cilomilast (CAS No. 153259-65-5) (GlaxoSmithKline), CR-1 (Nuada Pharmaceuticals), CRx-119 (CombinatoRx), D-5410 (UCB), dacopafant (CAS No. 125372-33-0) (Sanofi-Aventis), dersalazine (CAS No. 188913-57- 7/188913-58-8) (Uriach), etanercept (CAS No. 185243-69-0) (Amgen), ethyl pyruvate (Critical (Critical Therapeutics), golimumab (CAS No. 476181-74-5) (Johnson & Johnson), hormono-immunotherapy from Ipsen, CDP571 (e.g., Humicade from UCB), IC-485 (ICOS), infliximab (CAS No. 170277-31-3) (Johnson & Johnson), IP-751 (Manhattan Pharmaceuticals), ISIS-104838 (CAS No. 250755-32-9) (ISIS Pharmaceuticals), lenalidomide (CAS No. 191732-72-6) (Celgene), lentinan (CAS No. 37339-90-5) (Ajinomoto), MDL- 201112 (CAS No. 142130-73-2) (Sanofi-Aventis), medroxyprogesterone (CAS No. 520-85-4) (InKine Pharmaceutical), N-acetylcysteine (CAS No. 616-91-1) (Zambon), NBE-P2 (DIREVO Biotech), nerelimomab (CAS No. 162774-06-3) (Chiron), OM-294DP (OM PHARMA), onercept (CAS No. 199685-57-9) (Yeda), PASSTNF-alpha (Verigen), pentoxifylline or oxypentifylline (Sanofi-Aventis), Pharmaprojects No. 4091 , 4241 , 4295, or 4488 (Sanofi-Aventis), Pharmaprojects No. 5480 (Amgen), Pharmaprojects No. 6749 (Cengent), pirfenidone (CAS No. 53179-13-8) (MARNAC), RPR- 132294 (Sanofi-Aventis), S5 (F002) (Fulcrum Pharmaceuticals), simvastatin (CAS No. 79902-63-9) (Merck & Co), STA-6292 (Synta Pharmaceuticals), tacrolimus (CAS No. 104987-11-3) (Fujisawa LifeCycle Pharma), talactoferrin alfa (CAS No. 308240-58-6) (Agennix), thalidomide (CAS No. 50-35-1) (Celgene), TNF antagonists form ProStrakan, and Synergen, TNF inhibitors (Amgen), TNF-alpha antagonists from Dynavax Technologies and Jerina AG (Germany), TNF-alpha inhibitors from IBFB Pharma and Xencor (Xencor), torbafylline (CAS No. 105102-21-4) (Sanofi-Aventis), UR-1505 (Uriach), VT-346 (Viron Therapeutics), YSIL6 (Y's Therapeutics), -YSTH2 (Y's Therapeutics), NPI-1302a-3 (Nereus Pharmaceuticals, a TNF antagonist from Jerina AG (Germany), dersalazine, and an analogue or derivative thereof.
3761. The device of item 3662 wherein the agent is a Toll receptor inhibitor.
3762. The device of item 3662 wherein the agent is a tubulin antagonist.
3763. The device of item 3662 wherein the agent is a tyrosine kinase inhibitor selected from the group consisting of SU-011248, SUTENT from Pfizer Inc. (New York, NY), BMS-354825, PN-355 (Paracelsian Pharmaceuticals), AGN-199659 (Allergan), AAL-993 or ABP-309 (Novartis), adaphostin (NIH), AEE-788 (Novartis), AG-013736 (OSI Pharmaceuticals), AG-13736 (Pfizer), ALT-110 (Alteris Therapeutics), AMG-706 (Amgen), anticancer MAbs from Xencor, anti-EGFrvlll MAbs from Abgenix, anti-HER2 MAb from Abiogen, AZD-2171 or AZD-9935 (AstraZeneca), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), CEP-5214 (Cephalon), CEP- 7055 (Cephalon), cetuximab (ImCIone Systems), CHIR-200131 and CHIR- 258 (Chiron), CP-547632 (OSI Pharmaceuticals), CP-724714 (Pfizer), CT- 301 (Creabilis Therapeutics), D-69491 (Baxter International), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EGFR/ErbB2 inhibitors from Array BioPharma, erlotinib (CAS No. 183319-69-9) (OSI Pharmaceuticals), EXEL-2880 (Exelixis), FK-778 (Sanofi-Aventis) , gefitinib (CAS No. 184475-35-2) (AstraZeneca), GW-2286 or GW-654652 (GlaxoSmithKline), her2/neu antigen from AlphaVax, HER-2/neu inhibitor from Generex, Herzyme (Medipad) (Sirna Therapeutics), HKI-272 (Wyeth), HuMax-EGFr (Genmab), idronoxil (CAS No. 81267-65-4) (Novogen), IGF-1 inhibitors from Ontogen, IMC-11 F8 (ImCIone Systems), kahalalide F (CAS No. 149204-42-2) (PharmaMar), KDR inhibitor from LG Life Sciences, KDR inhibitors from Abbott Laboratories, KDR kinase inhibitors (UCB), Kdr kinase inhibitors from Merck & Co, KRN-633 and KRN-951 (Kirin Brewery), KSB-102 (Xenova), lapatinib ditosylate (CAS No. 388082-78-8) (GlaxoSmithKline), matuzumab (Merck KGaA), MDX-214 (Medarex), ME-103 (Pharmexa), MED-A300 (Gerolymatos), MNAC-13 (Lay Line Genomics), nimotuzumab (Center of Molecular Immunology), NSC-330507 or NSC-707545 (NIH), NV-50 (Novogen), OSI-930 (OSI Pharmaceuticals), panitumumab (Abgenix), pelitinib (CAS No. 287933-82-7) (Wyeth), pertuzumab (CAS No. 380610-27- 5) (Genentech), Pharmaprojects No. 3985 (Sanofi-Aventis), prostate cancer therapeutics from Sequenom (SQPC35, SQPC36, SQPC90), removab and remoxab (Trion Pharma), RG-13022 (CAS No. 136831-48-6), RG-13291 (CAS No. 138989-50-1), or RG-14620 (CAS No. 136831-49-7) (Sanofi- Aventis), RM-6427 (Romark), RNAi breast cancer therapy from Benitec, RP 53801 (CAS No. 125882-88-4) (Sanofi-Aventis), sorafenib tosylate (Bayer), SU-11657 (Pfizer), Tie-2 antagonists from Semaia (Hybrigenics), Tie-2 inhibitors from Ontogen, trastuzumab (CAS No. 180288-69-1) (Genentech), tyrosine kinase inhibitors from Sanofi-Aventis, U3-1287, U3-1565, U3-1784, U3-1800 (U3 Pharma), vatalanib (Novartis), VEGFR-2 kinase inhibitor from Bristol-Myers Squibb, XL-647 (Exelixis), ZD-6474 (AstraZeneca), ZK-CDK (Schering AG), an EGFR tyrosine kinase inhibitor, EKB-569 (Wyeth), and an analogue or derivative thereof.
3764. The device of item 3662 wherein the agent is a VEGF inhibitor.
3765. The device of item 3662 wherein the agent is a vitamin D receptor agonist.
3766. The device of item 3662 wherein the agent is ZD-6474 (an angiogenesis inhibitor).
3767. The device of item 3662 wherein the agent is AP-23573 (an mTOR inhibitor).
3768. The device of item 3662 wherein the agent is synthadotin (a tubulin antagonist).
3769. The device of item 3662 wherein the agent is S-0885 (a collagenase inhibitor).
3770. The device of item 3662 wherein the agent is aplidine (an elongation factor-1 alpha inhibitor).
3771. The device of item 3662 wherein the agent is ixabepilone (an epithilone). 3772. The device of item 3662 wherein the agent is IDN-5390 (an angiogenesis inhibitor).
3773. The device of item 3662 wherein the agent is SB- 2723005 (an angiogenesis inhibitor).
3774. The device of item 3662 wherein the agent is ABT-518 (an angiogenesis inhibitor).
3775. The device of item 3662 wherein the agent is combretastatin (an angiogenesis inhibitor).
3776. The device of item 3662 wherein the agent is anecortave acetate (an angiogenesis inhibitor).
3777. The device of item 3662 wherein the agent is SB- 715992 (a kinesin antagonist).
3778. The device of item 3662 wherein the agent is temsirolimus (an mTOR inhibitor).
3779. The device of item 3662 wherein the agent is adalimumab (a TNFα antagonist).
3780. The device of item 3662, further comprising a polymer.
3781. The device of item 3662 wherein the anti-scarring agent inhibits adhesion between the device and a host into which the device is implanted. 3782. The device of item 3662 wherein the device delivers the anti-scarring agent locally to tissue proximate to the device.
3783. The device of item 3662, further comprising a coating, wherein the coating comprises the anti-scarring agent.
3784. The device of item 3662, further comprising a coating, wherein the coating is disposed on a surface of the device.
3785. The device of item 3662, further comprising a coating, wherein the coating directly contacts the device.
3786. The device of item 3662, further comprising a coating, wherein the coating indirectly contacts the device.
3787. The device of item 3662, further comprising a coating, wherein the coating partially covers the device.
3788. The device of item 3662, further comprising a coating, wherein the coating completely covers the device.
3789. The device of item 3662, further comprising a coating, wherein the coating is a uniform coating.
3790. The device of item 3662, further comprising a coating, wherein the coating is a non-uniform coating.
3791. The device of item 3662, further comprising a coating, wherein the coating is a discontinuous coating. 3792. The device of item 3662, further comprising a coating, wherein the coating is a patterned coating.
3793. The device of item 3662, further comprising a coating, wherein the coating has a thickness of 100 Dm or less.
3794. The device of item 3662, further comprising a coating, wherein the coating has a thickness of 10 Dm or less.
3795. The device of item 3662, further comprising a coating, wherein the coating adheres to the surface of the device upon deployment of the device.
3796. The device of item 3662, further comprising a coating, wherein the coating is stable at room temperature for a period of 1 year.
3797. The device of item 3662, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 0.0001% to about 1% by weight.
3798. The device of item 3662, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 1% to about 10% by weight.
3799. The device of item 3662, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 10% to about 25% by weight.
3800. The device of item 3662, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 25% to about 70% by weight. 3801. The device of item 3662, further comprising a coating, wherein the coating further comprises a polymer.
3802. The device of item 3662, further comprising a first coating having a first composition and the second coating having a second composition.
3803. The device of item 3662, further comprising a first coating having a first composition and the second coating having a second composition, wherein the first composition and the second composition are different.
3804. The device of item 3662, further comprising a polymeric carrier, wherein the polymeric carrier comprises a copolymer.
3805r The device of item 3662, further comprising a polymeric carrier, wherein the polymeric carrier comprises a block copolymer.
3806. The device of item 3662, further comprising a polymeric carrier, wherein the polymeric carrier comprises a random copolymer.
3807. The device of item 3662, further comprising a polymeric carrier, wherein the polymeric carrier comprises a biodegradable polymer.
3808. The device of item 3662, further comprising a polymeric carrier, wherein the polymeric carrier comprises a non-biodegradable polymer.
3809. The device of item 3662, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrophilic polymer. 3810. The device of item 3662, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrophobic polymer.
3811. The device of item 3662, further comprising a polymeric carrier, wherein the polymeric carrier comprises a polymer having hydrophilic domains.
3812. The device of item 3662, further comprising a polymeric carrier, wherein the polymeric carrier comprises a polymer having hydrophobic domains.
3813. The device of item 3662, further comprising a polymeric carrier, wherein the polymeric carrier comprises a non-conductive polymer.
3814. The device of item 3662, further comprising a polymeric carrier, wherein the polymeric carrier comprises an elastomer.
3815. The device of item 3662, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrogel.
3816. The device of item 3662, further comprising a polymeric carrier, wherein the polymeric carrier comprises a silicone polymer.
3817. The device of item 3662, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrocarbon polymer.
3818. The device of item 3662, further comprising a polymeric carrier, wherein the polymeric carrier comprises a styrene-derived polymer.
3819. The device of item 3662, further comprising a polymeric carrier, wherein the polymeric carrier comprises a butadiene polymer. 3820. The device of item 3662, further comprising a polymeric carrier, wherein the polymeric carrier comprises a macromer.
3821. The device of item 3662, further comprising a polymeric carrier, wherein the polymeric carrier comprises a poly(ethylene glycol) polymer.
3822. The device of item 3662, further comprising a polymeric carrier, wherein the polymeric carrier comprises an amorphous polymer.
3823. The device of item 3662,- further comprising a lubricious coating.
3824. The device of item 3662 wherein the anti-scarring agent is located within pores or holes of the device.
3825. The device of item 3662 wherein the anti-scarring agent is located within a channel, lumen, or divet of the device.
3826. The device of item 3662, further comprising a second pharmaceutically active agent.
3827. The device of item 3662, further comprising an antiinflammatory agent.
3828. The device of item 3662, further comprising an agent that inhibits infection.
3829. The device of item 3662, further comprising an agent that inhibits infection, wherein the agent is an anthracycline. 3830. The device of item 3662, further comprising an agent that inhibits infection, wherein the agent is doxorubicin.
3831. The device of item 3662, further comprising an agent that inhibits infection, wherein the agent is mitoxantrone.
3832. The device of item 3662, further comprising an agent that inhibits infection, wherein the agent is a fluoropyrimidine.
3833. The device of item 3662, further comprising an agent that inhibits infection, wherein the agent is 5-fIuorouracil (5-FU).
3834. The device of item 3662, further comprising an agent that inhibits infection, wherein the agent is a folic acid antagonist.
3835. The device of item 3662, further comprising an agent that inhibits infection, wherein the agent is methotrexate.
3836. The device of item 3662, further comprising an agent that inhibits infection, wherein the agent is a podophylotoxin.
3837. The device of item 3662, further comprising an agent that inhibits infection, wherein the agent is etoposide.
3838. The device of item 3662, further comprising an agent that inhibits infection, wherein the agent is a camptothecin.
3839. The device of item 3662, further comprising an agent that inhibits infection, wherein the agent is a hydroxyurea. 3840. The device of item 3662, further comprising an agent that inhibits infection, wherein the agent is a platinum complex.
3841. The device of item 3662, further comprising an agent that inhibits infection, wherein the agent is cisplatin.
3842. The device of item 3662, further comprising an antithrombotic agent.
3843. The device of item 3662, further comprising a visualization agent.
3844. The device of item 3662, further comprising a visualization agent, wherein the visualization agent is a radiopaque material, wherein the radiopaque material comprises a metal, a halogenated compound, or a barium containing compound. - - - - - - - - -
3845. The device of item 3662, further comprising a visualization agent, wherein the visualization agent is a radiopaque material, wherein the radiopaque material comprises barium, tantalum, or technetium.
3846. The device of item 3662, further comprising a visualization agent, wherein the visualization agent is a MRI responsive material.
3847. The device of item 3662, further comprising a visualization agent, wherein the visualization agent comprises a gadolinium chelate. 3848. The device of item 3662, further comprising a visualization agent, wherein the visualization agent comprises iron, magnesium, manganese, copper, or chromium.
3849. The device of item 3662, further comprising a visualization agent, wherein the visualization agent comprises an iron oxide compound.
3850. The device of item 3662, further comprising a visualization agent, wherein the visualization agent comprises a dye, pigment, or colorant.
3851. The device of item 3662, further comprising an echogenic material.
3852.- The device of item 3662, further comprising an echogenic material, wherein the echogenic material is in the form of a coating.
3853. The device of item 3662 wherein the device is sterile.
3854. The device of item 3662 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device.
3855. The device of item 3662 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is connective tissue. 3856. The device of item 3662 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is muscle tissue.
3857. The device of item 3662 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is nerve tissue.
3858. The device of item 3662 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is epithelium tissue.
3859. The device of item 3662 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from the time of deployment of the device to about 1 year.
3860. The device of item 3662 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from about 1 month to 6 months.
3861. The device of item 3662 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from about 1 - 90 days.
3862. The device of item 3662 wherein the anti-scarring agent is released in effective concentrations from the device at a constant rate.
3863. The device of item 3662 wherein the anti-scarring agent is released in effective concentrations from the device at an increasing rate. 3864. The device of item 3662 wherein the anti-scarring agent is released in effective concentrations from the device at a decreasing rate.
3865.. The device of item 3662 wherein the anti-scarring agent is released in effective concentrations from the composition comprising the anti-scarring agent by diffusion over a period ranging from the time of deployment of the device to about 90 days.
3866. The device of item 3662 wherein the anti-scarring agent is released in effective concentrations from the composition comprising the anti-scarring agent by erosion of the composition over a period ranging from the time of deployment of the device to about 90 days.
3867. The device of item 3662 wherein the device comprises about 0.01 Dg to about 10 Dg of the anti-scarring agent.
3868. The device of item 3662 wherein the device comprises about 10 Dg to about 10 mg of the anti-scarring agent.
3869. The device of item 3662 wherein the device comprises about 10 mg to about 250 mg of the anti-scarring agent.
3870. The device of item 3662 wherein the device comprises about 250 mg to about 1000 mg of the anti-scarring agent.
3871. The device of item 3662 wherein the device comprises about 1000 mg to about 2500 mg of the anti-scarring agent.
3872. The device of item 3662 wherein a surface of the device comprises less than 0.01 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied. 3873. The device of item 3662 wherein a surface of the device comprises about 0.01 Dg to about 1 Og of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
3874. The device of item 3662 wherein a surface of the device comprises about 1 Dg to about 10 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
3875. The device of item 3662 wherein a surface of the device comprises about 10 Dg to about 250 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
3876. The device of item 3662 wherein a surface of the device comprises about 250 Dg to about 1000 Dg of the anti-scarring agent of anti- scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
3877. The device of item 3662 wherein a surface of the device comprises about 1000 Dg to about 2500 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
3878. A method as in any one of items 3662-3877, wherein the device is a peritoneal dialysis catheter adapted for delivering a drug to the peritoneum.
3879. A device, comprising a central nervous system shunt or pressure monitor device (e.g., an implant) and an anti-scarring agent or a composition comprising an anti-scarring agent, wherein the agent inhibits scarring between the device and a host into which the device is implanted.
3880. The device of item 3879 wherein the agent is an adensosine A2A receptor antagonist. 3881. The device of item 3879 wherein the agent is an AKT inhibitor.
3882. The device of item 3879 wherein the agent is an alpha 2 integrin antagonist, wherein the alpha 2 integrin antagonist is Pharmaprojects No. 5754 (Merck KgaA).
3883. The device of item 3879 wherein the agent an alpha 4 integrin antagonist.
3884. The device of item 3879 wherein the agent is an alpha 7 nicotinic receptor agonist.
3885. The device of item 3879 wherein the agent is an angiogenesis inhibitor selected from the group consisting of AG-12,958 (Pfizer), ATN-161 (Attention LLC), neovastat, an angiogenesis inhibitor from Jerina AG (Germany), NM-3 (Mercian), VGA-1155 (Taisho), FCE-26644 (Pfizer), FCE-26950 (Pfizer), FPMA (Meiji Daries), FR-111142 (Fujisawa), GGTI-298, GM-1306 (Ligand), GPA-1734 (Novartis), NNC-47-0011 (Novo Nordisk), herbamycin (Nippon Kayaku), lenalidomide (Celegene), IP-10 (NIH), ABT-828 (Abbott), KIN-841 (Tokushima University, Japan), SF-1126 (Semafore Pharmaceuticals), laminin technology (NIH), CHIR-258 (Chiron), NVP-AEW541 (Novartis), NVP-AEW541 (Novartis), Vt16907 (Alchemia), OXI-8007 (Oxigene), EG-3306 (Ark Therapeutics), Maspin (Arriva), ABT- 567 (Abbott), PPI-2458 (Praecis Pharmaceuticals), CC-5079, CC-4089 (Celgene), HIF-1 alpha inhibitors (Xenova), S-247 (Pfizer), AP-23573 (Ariad), AZD-9935 (Astra Zeneca), mebendazole (Introgen Therapeutics), MetAP-2 inhibitors (GlaxoSmithKline), AG-615 (Angiogene Pharmaceuticals), Tie-2 antagonists (Hybrigenics), NC-381, CYC-381, NC-169, NC-219, NC-383, NC-384, NC-407 (Lorus Therapeutics), ATN-224 (Attenuon), ON-01370 (Onconova), Vitronectin antagonists (Amgen), SDX-103 (Salmedix), Vitronectin antagonists (Shire), CHP (Riemser), TEK (Amgen), Anecortave acetate (Aicon), T46.2 (Matrix Therapeutics), HG-2 (Heptagen), TEM antagonists (Genzyme), Oxi-4500 (Oxigene), ATN-161 (Attenuon), WX-293 (Wilex), M-2025 (Metris Therapeutics), Alphastatin (BioActa), YH-16 (Yantai Rongchang), BIBF-1120 (Boehringer Ingelheim), BAY-57-9352 (Bayer), AS- 1404 (Cancer Research Technology), SC-77964 (Pfizer), glycomimetics (BioTie Therapies), TIE-2 Inhibitors (Ontogen), DIMI, Octamer (Octamer), ABR-215050 (Active Biotech), ABT-518 (Abbott), KDR inhibitors (Abbott), BSF-466895 (Abbott), SCH-221153 (Schering-Plough), DAC:antiangiogenic (ConjuChem), TFPI (EntreMed), AZD-2171 (Astra-Zenaca), CDC-394 (Celgene), LY290293 (EIi Lilly), IDN-5390 (Indena), Kdr Kinase Inhibitors (Merck), CT-113020, CT-116433, CT-116563, CT-31890, CT-32228) (Cell Therapeutics), A-299620 (Abbott), TWEAK Inhibitor (Amgen), VEGF modulators (Johnson and Johnson), Tum-N53, tumstatin (Genzyme), Thios- 1 , Thios-2 (Thios Pharmaceuticals), MV-6401 (Miravant Medical Technologies), Spisulosine (PharmaMar), GEP-7055 (Gephalon), AUV-201 (Auvation), LM-609 (EIi Lilly), SKF-106615 (AnorMED), Oglufanide disodium (Cytran), BW-114 (Pharminox), Calreticulin (NIH), WX-678 (Wilex), SD-7784 (Pfizer), WX-UK1 (Wilex), SH-268 (Schering AG), 2-Me-PGA (Celgene), S- 137 (Pfizer), ZD-6126 (Angiogene Pharmaceuticals), SG-292 (SignalGen), Benefin (Lane Labs), A6, A36 (Angstrom), SB-2723005 (GlaxoSmithKline), SC-7 (Cell Therapeutics), ZEN-014 (AEterna Zentaris), 2-methoxyestradiol (EntreMed), NK-130119 (Nippon Kayaku), CC-10004 (Celgene), AVE- 8062A (Ajinomoto), Tacedinaline (Pfizer), Actinonin (Tokyo Metropolitan Institute of Medical Science), Lenalidomide (Celgene), VGA-1155, BTO-956 (SRI International), ER-68203-00 (Eisai), CT-6685 (UCB), JKC-362 (Phoenix Pharmaceuticals), DMI-3798 (DMI Biosciences, Angiomate (Ipsen), ZD-6474 (AstraZeneca), CEP-5214 (Cephalon), Canstatin (Genzyme), NM-3 (Mercian), Oridigm (MediQuest Therapeutics), Exherin (Adherex), BLS-0597 (Boston Life Sciences), PTC-299 (PTC Therapeutics), NPI-2358 (Nereus Pharmaceuticals), CGP-79787 (Novartis), AEE-788 (Novartis), CKD-732 (Chong Kun Dang), CP-564959 (OSI Pharmaceuticals), CM-101 (CarboMed), CT-2584, CT3501 (Cell Therapeutics), combretastatin and analogues and derivatives thereof (Oxigene), Rebimastat (Bristol-Meyers Squibb), Dextrin 2-sulfate (ML Laboratories), Cilengitide (Merk KGaA), NSC-706704 (Pharminox), KRN- 951 (Kirin Brewery), Ukrain, NSC-631570 (Nowicky Pharma), Tecogalan sodium (Daiichi Pharmaceutical), Tz-93 (Tsumura), TBC-1635 (Encysive Pharmaceuticals), TAN-1120 (Takeda), Semaxanib (Pfizer), BDI-7800 (Biopharmacopae), SD-186, SD-983 (Bristol-Meyers Squibb), SB-223245 (GlaxoSmithKline), SC-236 (Pfizer), RWJ-590973 (Johnson and Johnson), ILX-1850 (Genzyme), SC-68488, S-836 (Pfizer), CG-55069-11 (CuraGen), Ki-23057 (Kirin Brewery), CCX-700 (Chemoentryx), Pegaptanib octasodium (Giled Sciences), ANGIOCOL (available from Biostratum Inc.), or an analogue or derivative thereof.
3886. The device of item 3879 wherein the agent is an apoptosis antagonist.
3887. The device of item 3879 wherein the agent is an apoptosis activator.
3888. The device of item 3879 wherein the agent is a beta 1 integrin antagonist.
3889. The device of item 3879 wherein the agent is a beta tubulin inhibitor.
3890. The device of item 3879 wherein the agent is a blocker of enzyme production in Hepatitis C. 3891. The device of item 3879 wherein the agent is a Bruton's tyrosine kinase inhibitor.
3892. The device of item 3879 wherein the agent is a calcineurin inhibitor.
3893. The device of item 3879 wherein the agent is a caspase 3 inhibitor.
3894. The device of item 3879 wherein the agent is a CC chemokine receptor antagonist.
3895. The device of item 3879 wherein the agent is a cell cycle inhibitor selected from the group consisting of SNS-595 (Sunesis), synthadotin, KRX-0403, and an analogue or derivative thereof.
3896. The device of item 3879 wherein the agent is a cathepsin B inhibitor.
3897. The device of item 3879 wherein the agent is a cathepsin K inhibitor, wherein the cathepsin K inhibitor is 462795 (GlaxoSmithKline), INPL-022-D6 (Amura Therapeutics), an analogue or derivative thereof).
3898. The device of item 3879 wherein the agent is a cathepsin L inhibitor.
3899. The device of item 3879 wherein the agent is a CD40 antagonist. 3900. The device of item 3879 wherein the agent is a chemokine receptor agonist.
3901. The device of item 3879 wherein the agent is a chymase inhibitor.
3902. The device of item 3879 wherein the agent is a collagenase antagonist.
3903. The device of item 3879 wherein the agent is a CXCR antagonist.
3904. The device of item 3879 wherein the agent is a cyclin dependent kinase inhibitor selected from the group consisting of a CDK-1 inhibitor, a CDK-2 inhibitor, a CDK- 4 inhibitor, a CDK-6 inhibitor, a CAK1 inhibitor from GPC Biotech and Bristol-Myers Squibb, RGB-286199 (GPC Biotech), an anticancer agent from Astex Technology, a CAK1 inhibitor from GPC Biotech, a CDK inhibitor from Sanofi-Aventis, a CDK1/CDK2 inhibitor from Amgen, a CDK2 inhibitor from SUGEN-2 (Pfizer), a hearing loss therapy agent (Sound Pharmaceuticals), PD-0332991 (Pfizer), RGB-286199 (GPC Biotech), Ro-0505124 (Hoffmann-La Roche), a Ser/Thr kinase inhibitor from Lilly (EIi Lilly), CVT-2584 (CAS No. 199986-75-9) (CV Therapeutics), and an analogue or derivative thereof.
3905. The device of item 3879 wherein the agent is a cyclooxygenase 1 inhibitor.
3906. The device of item 3879 wherein the agent is a DHFR inhibitor. 3907. The device of item 3879 wherein the agent is a dual integrin inhibitor.
3908. The device of item 3879 wherein the agent is an elastase inhibitor.
3909. The device of item 3879 wherein the agent is an elongation factor-1 alpha inhibitor.
3910. The device of item 3879 wherein the agent is an endothelial growth factor antagonist.
3911. The device of item 3879 wherein the agent is an endothelial growth factor receptor kinase inhibitor selected from the group consisting of sorafenib tosylate (Bayer), AAL-993 (Novartis), ABP-309 (Novartis), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), E- 7080 (Eisai), EG-3306 (Ark Therapeutics), EXEL-2880 (Exelixis), GW- 654652 (GlaxoSmithKline), a KDR inhibitor from LG Life Sciences, CT-6685 and CT-6729 (UCB), KRN-633 and KRN-951 (Kirin Brewery), OSI-930 (OSI Pharmaceuticals), SP-5.2 (Supratek Pharma), SU-11657 (Pfizer), a Tie-2 antagonist (Hybrigenics), a VEGFR-2 kinase inhibitor (Bristol-Myers Squibb), XL-647 (Exelixis), a KDR inhibitor from Abbott Laboratories, sorafenib tosylate, and an analogue or derivative thereof.
3912. The device of item 3879 wherein the agent is an endotoxin antagonist.
3913. The device of item 3879 wherein the agent is an epothilone and tubulin binder. 3914. The device of item 3879 wherein the agent is an estrogen receptor antagonist.
3915. The device of item 3879 wherein the agent is an FGF inhibitor.
3916. The device of item 3879 wherein the agent is a farnexyl transferase inhibitor.
3917. The device of item 3879 wherein the agent is farnesyltransferase inhibitor selected from the group of A-197574 (Abbott), a farnesyltransferase inhibitor from Servier, FPTIII (Strathclyde Institute for Drug R)1 LB-42908 (LG Life Sciences), Pharmaprojects No. 5063 (Genzyme), Pharmaprojects No. 5597 (Ipsen), Yissum Project No. B-1055 (Yissum), and an analogue or derivative thereof.
3918. The device of item 3879 wherein the agent is an FLT-3 kinase inhibitor.
3919. The device of item 3879 wherein the agent is an FGF receptor kinase inhibitor.
3920. The device of item 3879 wherein the agent is a fibrinogen antagonist selected from the group consisting of AUV-201 (Auvation), MG-13926 (Sanofi-Aventis), plasminogen activator (CAS No. 105913-11-9) (from Sanofi-Aventis or UCB), plasminogen activator-2 (tPA- 2) (Sanofi-Aventis), pro-urokinase (CAS No. 82657-92-9) (Sanofi-Aventis), and an analogue or derivative thereof.
3921. The device of item 3879 wherein the agent is a heat shock protein 90 antagonist selected from the group consisting of SRN-005 (Sirenade), geldanamycin, NSC-33050 (17-allylaminogeldanamycin; 17- AAG), 17-dimethylaminoethylamino-17-demethoxy-geldanamycin (17- DMAG), rifabutin (rifamycin XIV, 1',4-didehydro-1-deoxy-1 ,4-dihydro-5'-(2- methylpropyl)-1-oxo-), and an analogue or derivative thereof.
3922. The device of item 3879 wherein the agent is a histone deacetylase inhibitor.
3923. The device of item 3879 wherein the agent is an HMGCoA reductase inhibitor selected from the group consisting of an atherosclerosis therapeutic from Lipid Sciences, ATI-16000 (ARYx Therapeutics), KS-01-019 (Kos Pharmaceuticals), Pharmaprojects No. 2197 (Sanofi-Aventi), RP 61969 (Sanofi-Aventis), and an analogue or derivative thereof.
_ .. _ 3924. The device of item 3879 wherein the agent is an ICAM inhibitor.
3925. The device of item 3879 wherein the agent is an IL, ICE and IRAK antagonist, wherein the antagonist is a CJ-14877, CP-424174 (Pfizer), NF-61 (Negma-Lerads), and an analogue or derivative thereof.
3926. The device of item 3879 wherein the agent is an IL-2 inhibitor.
3927. The device of item 3879 wherein the agent is an immunosuppressant selected from the group consisting of teriflunomide (Sanofi Aventis), chlorsulfaquinoxalone (NSC-339004), chlorsulfaquinoxalone sulfate, CS-712 (Sankyo), ismomultin alfa (CAS No. 457913-93-8) (Akzo Nobel), antiallergics from GenPat77, antiinflammatories or AT-005 (Androclus Therapeutics), autoimmune disease therapeutics from EpiVax, BN-007 (Bone), budesonide (CAS No. 51333-22- 3) (MAP Pharmaceuticals), CO-14 (Genzyme), edratide (CAS No. 433922- 67-9) (Teva), EP-314 (Enanta), eprovafen (CAS No. 101335-99-3) (Sanofi- Aventis), HWA-131 (CAS No. 118788-41-3) (Sanofi-Aventis), immunomodulators from MerLion Pharmaceuticals, immunosuppressives from Alchemia, IPL-12 (Inflazyme), MDL-9563 (CAS No. 27086-86-8) (Sanofi-Aventis), Pharmaprojects No. 2330 (Sanofi-Aventis), Pharmaprojects No. 6426 (Abgenix), PXS-25 (Pharmaxis), rosmarinic acid (CAS No. 20283-92-5) (Sanofi-Aventis), RP 42927 or RP 54745 (CAS No. 135330-08-4) (Sanofi-Aventis), SGN-35 (Seattle Genetics), ST-1959 , (Sigma-Tau), type I diabetes therapy from SYNX Pharma, UNIL-88 (Debiopharm), VP-025 (Vasogen), VR-694 (Vectura), PRTX-001 (Protalex), and an analogue or derivative thereof.
3928. The device of item 3879 wherein the agent is an IMPDH (inosine monophosphate).- -
3929. The device of item 3879 wherein the agent is an integrin antagonist.
3930. The device of item 3879 wherein the agent is an interleukin antagonist.
3931. The device of item 3879 wherein the agent is an inhibitor of type III receptor tyrosine kinase.
3932. The device of item 3879 wherein the agent is an irreversible inhibitor of enzyme methionine aminopeptidase type 2.
3933. The device of item 3879 wherein the agent is an isozyme selective delta protein kinase C inhibitor. 3934. The device of item 3879 wherein the agent a JAK3 enzyme inhibitor.
3935. The device of item 3879 wherein the agent is a JNK inhibitor.
3936. The device of item 3879 wherein the agent is a kinase inhibitor.
3937. The device of item 3879 wherein the agent is kinesin antagonist.
3938. The device of item 3879 wherein the agent is a kinesin antagonist.
3939. The device of item 3879 wherein the agent is a leukotriene inhibitor and antagonist selected from the group consisting of ambicromil (CAS No. 58805-38-2) (Sanofi-Aventis), amelubant (CAS No. 346735-24-8) (Boehringer Ingelheim), DW-1141 (Dong Wha), ebselen (Daiichi Pharmaceutical), ibudilast (Kyorin), leucotriene inhibitors from Sanofi-Aventis, lymphotoxin-beta receptor (LT-D) from Biogen Idee, Pharmaprojects No. 1535 and 2728 (CAS No. 119340-33-9) (Sanofi- Aventis), R-112 (Rigel), Rev-5367 (CAS No. 92532-05-3) (Sanofi-Aventis), RG-14893 (CAS No. 141835-49-6) (Sanofi-Aventis), RG-5901-A (CAS No. 101910-24-1), 92532-23-5, RP 66153 (CAS No. 142422-79-5), RP 66364 (CAS No.186916-92-5), RP 69698 (CAS No. 141748-00-7) (Sanofi-Aventis), SC-411930 (Pfizer), SC-41930 (CAS No. 120072-59-5) (Pfizer), SC-50605 (CAS No. 138828-39-4) (Pfizer), SC-51146 (CAS No. 141059-52-1), SC- 53228 (CAS No. 153633-01-3) (Pfizer), spaglumic acid (ZY-15106) (CAS No.3106-85-2), 80619-64-3 (Novartis), tipredane (CAS No. 85197-77-9) W
(Bristol-Myers Squibb), U-75302 (CAS No. 119477-85-9) (Pfizer), and analogue or derivative thereof.
3940. The device of item 3879 wherein the agent is an MAP kinase inhibitor.
3941. The device of item 3879 wherein the agent is a matrix metalloproteinase inhibitor.
3942. The device of item 3879 wherein the agent is an MCP- CCR2 inhibitor.
3943. The device of item 3879 wherein the agent is an mTOR inhibitor.
3944. The device of item 3879 wherein the agent is an mTOR kinase inhibitor.
3945. The device of item 3879 wherein the agent is a microtubule inhibitor selected from the group consisting of antibody- maytansinoid conjugates from Biogen Idee, colchicines (MantiCore Pharmaceuticals), anticancer immunoconjugates from Johnson & Johnson, DIME from Octamer, gni-1f (GNI), huC242-DM4, huMy9-6-DM1 (ImmunoGen), IDN-5404 (Indena), IMO-098, IMOderm (Imotep), mebendazole (Introgen Therapeutics), microtubule poisons from Cambridge Enterprise, paclitaxel such as LOTAX from Aphios (CAS No. 33069-62-4), Genexol-PM from Samyang, Pharmaprojects No. 6383 (Tapestry Pharmaceuticals), RPR-112378 (Sanofi-Aventis), SGN-75 (Seattle Genetics), SPL-7435 (Starpharma), SSR-250411 (Sanofi-Aventis), trastuzumab-DM1 (Genentech), vinorelbine, and an analogue or derivative thereof. 3946. The device of item 3879 wherein the agent is an MIF inhibitor.
3947. The device of item 3879 wherein the agent is an MMP inhibitor.
3948. The device of item 3879 wherein the agent is a neurokinin (NK) antagonist selected from the group consisting of anthrotainin (CAS No. 148084-40-6) (Sanofi-Aventis), an IBS thereapeutic from ArQuIe, MDL-105212A (CAS No. 167261-60-1) (Ssanofi-Aventis), Pharmaprojects No. 2744, 3258 (CAS No. 139167-47-8) 4006, 4201, or 5986 (Sanofi-Aventis), RP 67580 (CAS No. 135911-02-3), SR-144190 (CAS No. 201152-86-5), SSR-240600, SSR-241586 (Sanofi-Aventis), TKA-457 (Novartis), vestipitant mesylate (CAS No. 334476-64-1) (GlaxoSmithKline), Win-64821 (Sanofi-Aventis), PRX-96026 (Predix Pharmaceuticals), and an analogue or derivative thereof.
3949. The device of item 3879 wherein the agent is an NF kappa B inhibitor selected from the group consisting of AVE-0545 or AVE- 0547 (Sanofi-Aventis), bortezomib (CAS No. 179324-69-7) (Millennium Pharmaceuticals), dexanabinol (CAS No. 112924-45-5) (Pharmos), dexlipotam (Viatris), Pharmaprojects No. 6283 (INDRA) (OXiGENE), IPL- 576092 (CAS No. 137571-30-3) (Inflazyme), NFKB decoy (Corgentech), NFKB decoy oligo (AnGes MG), NFKB's from Ariad, osteoporosis treatments or S5 (F005) from Fulcrum Pharmaceuticals, P61 (Phytopharm), R-flurbiprofen (CAS No. 5104-49-4) (Encore Pharmaceuticals), and an analogue or derivative thereof.
3950. The device of item 3879 wherein the agent is a nitric oxide agonist. 3951. The device of item 3879 wherein the agent is an ornithine decarboxylase inhibitor.
3952. The device of item 3879 wherein the agent is a p38 MAP kinase inhibitor selected from the group consisting of AZD-6703 (AstraZeneca), JX-401 (Jexys Pharmaceuticals), BMS-2 (Bristol-Myers Squibb), a p38 MAP kinase inhibitor from Novartis, a p38-alpha MAP kinase inhibitor from Amphora, Pharmaprojects No. 5704 (Pharmacopeia), RPR- 200765A (Sanofi-Aventis), SD-282 (Johnson & Johnson), TAK-715 (Takeda), and an analogue or derivative thereof.
3953. The device of item 3879 wherein the agent is a palmitoyl-protein thioesterase inhibitor.
3954. The device of item 3879 wherein the agent is a PDGF receptor kinase inhibitor selected from the group consisting of AAL-993, AMN-107, or ABP-309 (Novartis), AMG-706 (Amgen), BAY-57-9352 (Bayer), CDP-860 (UCB), E-7080 (Eisai), imatinib (CAS No. 152459-95-5) (Novartis), OSI-930 (OSI Pharmaceuticals), RPR-127963E (Sanofi-Aventis), RWJ-540973 (Johnson & Johnson), sorafenib tosylate (Bayer), SU-11657 (Pfizer), tandutinib (CAS No. 387867-13-2) (Millennium Pharmaceuticals), vatalanib (Novartis), ZK-CDK (Schering AG), and an analogue or derivative thereof.
3955. The device of item 3879 wherein the agent is (-)- halofenate (Metabolex), AMG-131 (Amgen), antidiabetics from Japan Tobacco, AZD-4619, AZD-8450, AZD-8677 (AstraZeneca), DRF-10945, balaglitazone (Dr Reddy's), CS-00088, CS-00098 (Chipscreen Biosciences), E-3030 (Eisai), etalocib (CAS No. 161172-51-6) (EIi Lilly), GSK-641597 (Ligand), GSK-677954 (GlaxoSmithKline), GW-409544 (Ligand), GW- 590735 (GlaxoSmithKline), K-111 (Hoffmann-La Roche), LY-518674 (EIi Lilly), LY-674 (Ligand), LY-929 (Ligand), MC-3001 , MC-3002 (MaxoCore Pharmaceuticals), metformin HCI + pioglitazone (CAS No. 1115-70-4 and 112529-15-4), ACTOPLUS MET from Andrx), muraglitazar (CAS No. 331741-94-7) (Bristol-Myers Squibb), naveglitazar (Ligand), oleoylethanolamide (Kadmus Pharmaceuticals), ONO-5129, pioglitazone hydrochloride (CAS No. 111025-46-8 and 112529-15-4) (Takeda), PLX-204 (Plexxikon), PPAR agonists from Genfit, PPAR delta agonists from EIi Lilly, PPAR-alpha agonists from CrystalGenomics, PPAR-gamma modulators and PPAR-β modulators from CareX, rosiglitazone maleate (CAS No. 122324- 73-4 or 155141-29-0) (GlaxoSmithKline), rosiglitazone maleate/glimepir (CAS No. 155141-29-0 and 93479-97-1), AVANDARYL, rosiglitazone maleate/metformin extend (CAS No. 155141-29-0 and 657-24-9), AVANDAMET, rosiglitazone maleate+metformin, AVANDAMET (GlaxoSmithKline), tesaglitazar (AstraZeneca), and an analogue or derivative thereof.
3956. The device of item 3879 wherein the agent is a phosphatase inhibitor.
3957. The device of item 3879 wherein the agent is a phosphodiesterase (PDE) inhibitor selected from the group consisting of avanafil (Tanabe Seiyaku), dasantafil (CAS No. 569351-91-3) (Schering- Plough), A-906119 (CAS No. 134072-58-5), DL-850 (Sanofi-Aventis), GRC- 3015, GRC-3566, GRC-3886 (Glenmark), HWA-153 (CAS No. 56395-66-5) (Sanofi-Aventis), hydroxypumafentrine (Altana), IBFB-130011, IBFB-14-016, IBFB-140301 , IBFB-150007, IBFB-211913 (IBFB Pharma), L-826141 (Merck & Co), medorinone (CAS No. 88296-61-1) (Sanofi-Aventis), MEM- 1917 (Memory Pharmaceuticals), ND-1251 (Neuro3d), PDE inhibitors from ICOS, PDE IV inhibitors from Memory Pharmaceuticals and CrystalGenomics, Pharmaprojects No. 2742 and 6141 (Sanofi-Aventis), QAD-171 (Novartis), RHC-2963 (CAS No. 76993-12-9 and 76993-14-1), RPR-117658, RPR-122818 derivatives, SR-24870 , and RPR-132294 (Sanofi-Aventis), SK-350 (ln2Gen), stroke therapy agents from deCODE Genetics, TAS-203 (Taiho), tofimilast (CAS No. 185954-27-2) (Pfizer), UK- 371800 (Pfizer), WIN-65579 (CAS No. 158020-82-7) (Sanofi-Aventis), IBFB- 130020 (IBFB Pharma), OPC-6535 (CAS No. 145739-56-6) (Otsuka), a phosphodiesterase III inhibitor, enoximone, a phosphodiesterase IV inhibitor, fosfosal, Atopik (Barrier Therapeutics), triflusal, a phosphodiesterase V inhibitor, and an analogue or derivative thereof.
3958. The device of item 3879 wherein the agent is a PKC inhibitor.
3959. The device of item 3879 wherein the agent is a platelet activating factor antagonist.
396O._ The device of item 3879 wherein the agent is a platelet- derived growth factor receptor kinase inhibitor.
3961. The device of item 3879 wherein the agent is a prolyl hydroxylase inhibitor.
3962. The device of item 3879 wherein the agent is a polymorphonuclear neutrophil inhibitor.
3963. The device of item 3879 wherein the agent is a protein kinase B inhibitor.
3964. The device of item 3879 wherein the agent is a protein kinase C stimulant. 3965. The device of item 3879 wherein the agent is a purine nucleoside analogue.
3966. The device of item 3879 wherein the agent is a purinoreceptor P2X antagonist.
3967. The device of item 3879 wherein the agent is a Raf kinase inhibitor.
3968. The device of item 3879 wherein the agent is a reversible inhibitor of ErbB1 and ErbB2.
3969. The device of item 3879 wherein the agent is a ribonucleoside triphosphate reductase inhibitor.
3970. The device of item 3879 wherein the agent is an SDF-1 antagonist.
3971. The device of item 3879 wherein the agent is a sheddase inhibitor.
3972. The device of item 3879 wherein the agent is an SRC inhibitor.
3973. The device of item 3879 wherein the agent is a stromelysin inhibitor.
3974. The device of item 3879 wherein the agent is an Syk kinase inhibitor. 3975. The device of item 3879 wherein the agent is a telomerase inhibitor.
3976. The device of item 3879 wherein the agent is a TGF beta inhibitor selected from the group consisting of pirfenidone (CAS No. 53179-13-8) (MARNAC)1 tranilast (CAS No. 53902-12-8) (Kissei), IN-1130 (ln2Gen), mannose-6-phosphate (BTG), TGF-β antagonists from Inflazyme (Pharmaprojects No. 6075), TGF-β antagonists from Sydney, non-industrial source), TGF-β I receptor kinase inhibitors from EIi Lilly, TGF-β receptor inhibitors from Johnson & Johnson, and an analogue or derivative thereof.
3977. The device of item 3879 wherein the agent is a TNFα antagonist or TACE inhibitor selected from the group consisting of adalimumab (CAS No. 331731-18-1) (Cambridge Antibody Technology), AGIX-4207 (AtheroGenics), AGT-1 (Advanced Biotherapy), an antiinflammatory from Borean Pharma, Cellzome, or Paradigm Therapeutics, anti-inflammatory vaccine (TNF-alpha kinoid) from Neovacs, humanized anti-TNF antibody or an anti-TNF MAb (CB0006) Celltech (UCB), apratastat (CAS No. 287445-51-0) (Wyeth), BMS-561392 (Bristol-Myers Squibb), BN- 006 (Bone), certolizumab pegol (CAS No. 428863-50-7 or CH-138 (UCB), cilomilast (CAS No. 153259-65-5) (GlaxoSmithKline), CR-1 (Nuada Pharmaceuticals), CRx-119 (CombinatoRx), D-5410 (UCB)1 dacopafant (CAS No. 125372-33-0) (Sanofi-Aventis), dersalazine (CAS No. 188913-57- 7/188913-58-8) (Uriach), etanercept (CAS No. 185243-69-0) (Amgen), ethyl pyruvate (Critical (Critical Therapeutics), golimumab (CAS No. 476181-74-5) (Johnson & Johnson), hormono-immunotherapy from Ipsen, CDP571 (e.g., Humicade from UCB), IC-485 (ICOS), infliximab (CAS No. 170277-31-3) (Johnson & Johnson), IP-751 (Manhattan Pharmaceuticals), ISIS-104838 (CAS No. 250755-32-9) (ISIS Pharmaceuticals), lenalidomide (CAS No. 191732-72-6) (Celgene), lentinan (CAS No. 37339-90-5) (Ajinomoto), MDL- 201112 (CAS No. 142130-73-2) (Sanofi-Aventis), medroxyprogesterone (CAS No. 520-85-4) (InKine Pharmaceutical), N-acetylcysteine (CAS No. 616-91-1) (Zambon), NBE-P2 (DIREVO Biotech), nerelimomab (CAS No. 162774-06-3) (Chiron), OM-294DP (OM PHARMA), onercept (CAS No. 199685-57-9) (Yeda), PASSTNF-alpha (Verigen), pentoxifylline or oxypentifylline (Sanofi-Aventis), Pharmaprojects No. 4091 , 4241 , 4295, or 4488 (Sanofi-Aventis), Pharmaprojects No. 5480 (Amgen), Pharmaprojects No. 6749 (Cengent), pirfenidone (CAS No. 53179-13-8) (MARNAC), RPR- 132294 (Sanofi-Aventis), S5 (F002) (Fulcrum Pharmaceuticals), simvastatin (CAS No. 79902-63-9) (Merck & Co), STA-6292 (Synta Pharmaceuticals), tacrolimus (CAS No. 104987-11-3) (Fujisawa LifeCycle Pharma), talactoferrin alfa (CAS No. 308240-58-6) (Agennix), thalidomide (CAS No. 50-35-1) (Celgene), TNF antagonists form ProStrakan, and Synergen, TNF inhibitors (Amgen), TNF-alpha antagonists from Dynavax Technologies and Jerina AG (Germany), TNF-alpha inhibitors from IBFB Pharma and Xencor (Xencor), torbafylline (CAS No. 105102-21-4) (Sanofi-Aventis), UR-1505 (Uriach),_VT-346 (Viron Therapeutics), YSIL6 (Y's Therapeutics), YSTH2 (Y's Therapeutics), NPI-1302a-3 (Nereus Pharmaceuticals, a TNF antagonist from Jerina AG (Germany), dersalazine, and an analogue or derivative thereof.
3978. The device of item 3879 wherein the agent is a Toll receptor inhibitor.
3979. The device of item 3879 wherein the agent is a tubulin antagonist.
3980. The device of item 3879 wherein the agent is a tyrosine kinase inhibitor selected from the group consisting of SU-011248, SUTENT from Pfizer Inc. (New York, NY), BMS-354825, PN-355 (Paracelsian Pharmaceuticals), AGN-199659 (Allergan), AAL-993 or ABP-309 (Novartis), adaphostin (NIH), AEE-788 (Novartis), AG-013736 (OSI Pharmaceuticals), AG-13736 (Pfizer), ALT-110 (Alteris Therapeutics), AMG-706 (Amgen), anticancer MAbs from Xencor, anti-EGFrvlll MAbs from Abgenix, anti-HER2 MAb from Abiogen, AZD-2171 or AZD-9935 (AstraZeneca), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), CEP-5214 (Cephalon), CEP- 7055 (Cephalon), cetuximab (ImCIone Systems), CHIR-200131 and CHIR- 258 (Chiron), CP-547632 (OSI Pharmaceuticals), CP-724714 (Pfizer), CT- 301 (Creabilis Therapeutics), D-69491 (Baxter International), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EGFR/ErbB2 inhibitors from Array BioPharma, erlotinib (CAS No. 183319-69-9) (OSI Pharmaceuticals), EXEL-2880 (Exelixis), FK-778 (Sanofi-Aventis) , gefitinib (CAS No. 184475-35-2) (AstraZeneca), GW-2286 or GW-654652 (GlaxoSmithKline), her2/neu antigen from AlphaVax, HER-2/neu inhibitor from Generex, Herzyme (Medipad) (Sirna Therapeutics), HKI-272 (Wyeth), HuMax-EGFr (Genmab), idronoxil (CAS No. 81267-65-4) (Novogen), IGF-1 inhibitors from Ontogen, IMC-11F8 (ImCIone Systems), kahalalide F (CAS No. 149204-42-2) (PharmaMar), KDR inhibitor frqm LG LifeJSciences, KDR inhibitors from Abbott Laboratories, KDR kinase inhibitors (UCB), Kdr kinase inhibitors from Merck & Co, KRN-633 and KRN-951 (Kirin Brewery), KSB-102 (Xenova), lapatinib ditosylate (CAS No. 388082-78-8) (GlaxoSmithKline), matuzumab (Merck KGaA), MDX-214 (Medarex), ME-103 (Pharmexa), MED-A300 (Gerolymatos), MNAC-13 (Lay Line Genomics), nimotuzumab (Center of Molecular Immunology), NSC-330507 or NSC-707545 (NIH), NV-50 (Novogen), OSI-930 (OSI Pharmaceuticals), panitumumab (Abgenix), pelitinib (CAS No. 287933-82-7) (Wyeth), pertuzumab (CAS No. 380610-27- 5) (Genentech), Pharmaprojects No. 3985 (Sanofi-Aventis), prostate cancer therapeutics from Sequenom (SQPC35, SQPC36, SQPC90), removab and remoxab (Trion Pharma), RG-13022 (CAS No. 136831-48-6), RG-13291 (CAS No. 138989-50-1), or RG-14620 (CAS No. 136831-49-7) (Sanofi- Aventis), RM-6427 (Romark), RNAi breast cancer therapy from Benitec, RP 53801 (CAS No. 125882-88-4) (Sanofi-Aventis), sorafenib tosyiate (Bayer), SU-11657 (Pfizer), Tie-2 antagonists from Semaia (Hybrigenics), Tie-2 inhibitors from Ontogen, trastuzumab (CAS No. 180288-69-1) (Genentech), tyrosine kinase inhibitors from Sanofi-Aventis, U3-1287, U3-1565, U3-1784, U3-1800 (U3 Pharma), vatalanib (Novartis), VEGFR-2 kinase inhibitor from Bristol-Myers Squibb, XL-647 (Exelixis), ZD-6474 (AstraZeneca), ZK-CDK (Schering AG), an EGFR tyrosine kinase inhibitor, EKB-569 (Wyeth), and an analogue or derivative thereof.
3981. The device of item 3879 wherein the agent is a VEGF inhibitor.
3982. The device of item 3879 wherein the agent is a vitamin D receptor agonist.
3983. The device of item 3879 wherein the agent is ZD-6474 (an angiogenesis inhibitor).
3984. The device of item 3879 wherein the agent is AP-23573 (an mTOR inhibitor).
3985. The device of item 3879 wherein the agent is synthadotin (a tubulin antagonist).
3986. The device of item 3879 wherein the agent is S-0885 (a collagenase inhibitor).
3987. The device of item 3879 wherein the agent is aplidine (an elongation factor-1 alpha inhibitor).
3988. The device of item 3879 wherein the agent is ixabepilone (an epithilone). 3989. The device of item 3879 wherein the agent is IDN-5390 (an angiogenesis inhibitor).
3990. The device of item 3879 wherein the agent is SB- 2723005 (an angiogenesis inhibitor).
3991. The device of item 3879 wherein the agent is ABT-518 (an angiogenesis inhibitor).
3992. The device of item 3879 wherein the agent is combretastatin (an angiogenesis inhibitor).
3993. The device of item 3879 wherein the agent is anecortave acetate (an angiogenesis inhibitor).
_ . 3994. The.device of. item 3879 wherein the agent is SB- 715992 (a kinesin antagonist).
3995. The device of item 3879 wherein the agent is temsirolimus (an mTOR inhibitor).
3996. The device of item 3879 wherein the agent is adalimumab (a TNFα antagonist).
3997. The device of item 3879, further comprising a polymer.
3998. The device of item 3879 wherein the anti-scarring agent inhibits adhesion between the device and a host into which the device is implanted. 3999. The device of item 3879 wherein the device delivers the anti-scarring agent locally to tissue proximate to the device.
4000. The device of item 3879, further comprising a coating, wherein the coating comprises the anti-scarring agent.
4001. The device of item 3879, further comprising a coating, wherein the coating is disposed on a surface of the device.
4002. The device of item 3879, further comprising a coating, wherein the coating directly contacts the device.
4003. The device of item 3879, further comprising a coating, wherein the coating indirectly contacts the device.
4004. The device of item 3879, further comprising a coating, wherein the coating partially covers the device.
4005. The device of item 3879, further comprising a coating, wherein the coating completely covers the device.
4006. The device of item 3879, further comprising a coating, wherein the coating is a uniform coating.
4007. The device of item 3879, further comprising a coating, wherein the coating is a non-uniform coating.
4008. The device of item 3879, further comprising a coating, wherein the coating is a discontinuous coating. 4009. The device of item 3879, further comprising a coating, wherein the coating is a patterned coating.
4010. The device of item 3879, further comprising a coating, wherein the coating has a thickness of 100 Dm or less.
4011. The device of item 3879, further comprising a coating, wherein the coating has a thickness of 10 Dm or less.
4012. The device of item 3879, further comprising a coating, wherein the coating adheres to the surface of the device upon deployment of the device.
4013. The device of item 3879, further comprising a coating, wherein the coating is stable at room temperature for a period of 1 year.
4014. The device of item 3879, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 0.0001 % to about 1% by weight.
4015. The device of item 3879, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 1% to about 10% by weight.
4016. The device of item 3879, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 10% to about 25% by weight.
4017. The device of item 3879, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 25% to about 70% by weight. 4018. The device of item 3879, further comprising a coating, wherein the coating further comprises a polymer.
4019. The device of item 3879, further comprising a first coating having a first composition and the second coating having a second composition.
4020. The device of item 3879, further comprising a first coating having a first composition and the second coating having a second composition, wherein the first composition and the second composition are different.
4021. The device of item 3879, further comprising a polymeric carrier, wherein the polymeric carrier comprises a copolymer.
4022. The device of item 3879, further comprising a polymeric carrier, wherein the polymeric carrier comprises a block copolymer.
4023. The device of item 3879, further comprising a polymeric carrier, wherein the polymeric carrier comprises a random copolymer.
4024. The device of item 3879, further comprising a polymeric carrier, wherein the polymeric carrier comprises a biodegradable polymer.
4025. The device of item 3879, further comprising a polymeric carrier, wherein the polymeric carrier comprises a non-biodegradable polymer.
4026. The device of item 3879, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrophilic polymer. 4027. The device of item 3879, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrophobic polymer.
4028. The device of item 3879, further comprising a polymeric carrier, wherein the polymeric carrier comprises a polymer having hydrophilic domains.
4029. The device of item 3879, further comprising a polymeric carrier, wherein the polymeric carrier comprises a polymer having hydrophobic domains.
4030. The device of item 3879, further comprising a polymeric carrier, wherein the polymeric carrier comprises a non-conductive polymer.
4031. The device of item 3879, further comprising a polymeric carrier, wherein the polymeric carrier comprises an elastomer.
4032. The device of item 3879, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrogel.
4033. The device of item 3879, further comprising a polymeric carrier, wherein the polymeric carrier comprises a silicone polymer.
4034. The device of item 3879, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrocarbon polymer.
4035. The device of item 3879, further comprising a polymeric carrier, wherein the polymeric carrier comprises a styrene-derived polymer.
4036. The device of item 3879, further comprising a polymeric carrier, wherein the polymeric carrier comprises a butadiene polymer. 4037. The device of item 3879, further comprising a polymeric carrier, wherein the polymeric carrier comprises a macromer.
4038. The device of item 3879, further comprising a polymeric carrier, wherein the polymeric carrier comprises a poly(ethylene glycol) polymer.
4039. The device of item 3879, further comprising a polymeric carrier, wherein the polymeric carrier comprises an amorphous polymer.
4040. The device of item 3879, further comprising a lubricious coating.
4041. The device of item 3879 wherein the anti-scarring agent is located within pores or holes of the device.
4042. The device of item 3879 wherein the anti-scarring agent is located within a channel, lumen, or divet of the device.
4043. The device of item 3879, further comprising a second pharmaceutically active agent.
4044. The device of item 3879, further comprising an antiinflammatory agent.
4045. The device of item 3879, further comprising an agent that inhibits infection.
4046. The device of item 3879, further comprising an agent that inhibits infection, wherein the agent is an anthracycline. 4047. The device of item 3879, further comprising an agent that inhibits infection, wherein the agent is doxorubicin.
4048. The device of item 3879, further comprising an agent that inhibits infection, wherein the agent is mitoxantrone.
4049. The device of item 3879, further comprising an agent that inhibits infection, wherein the agent is a fluoropyrimidine.
4050. The device of item 3879, further comprising an agent that inhibits infection, wherein the agent is 5-fluorouracil (5-FU).
4051. The device of item 3879, further comprising an agent that inhibits infection, wherein the agent is a folic acid antagonist.
4052. The device of item 3879, further comprising an agent that inhibits infection, wherein the agent is methotrexate.
4053. The device of item 3879, further comprising an agent that inhibits infection, wherein the agent is a podophylotoxin.
4054. The device of item 3879, further comprising an agent that inhibits infection, wherein the agent is etoposide.
4055. The device of item 3879, further comprising an agent that inhibits infection, wherein the agent is a camptothecin.
4056. The device of item 3879, further comprising an agent that inhibits infection, wherein the agent is a hydroxyurea. 4057. The device of item 3879, further comprising an agent that inhibits infection, wherein the agent is a platinum complex.
4058. The device of item 3879, further comprising an agent that inhibits infection, wherein the agent is cisplatin.
4059. The device of item 3879, further comprising an antithrombotic agent.
4060. The device of item 3879, further comprising a visualization agent.
4061. The device of item 3879, further comprising a visualization agent, wherein the visualization agent is a radiopaque material, wherein the radiopaque material comprises a metal, a halogenated compound, or a barium containing compound, - - -
4062. The device of item 3879, further comprising a visualization agent, wherein the visualization agent is a radiopaque material, wherein the radiopaque material comprises barium, tantalum, or technetium.
4063. The device of item 3879, further comprising a visualization agent, wherein the visualization agent is a MRI responsive material.
4064. The device of item 3879, further comprising a visualization agent, wherein the visualization agent comprises a gadolinium chelate. 4065. The device of item 3879, furtheir comprising a visualization agent, wherein the visualization agent comprises iron, magnesium, manganese, copper, or chromium.
4066. The device of item 3879, further comprising a visualization agent, wherein the visualization agent comprises an iron oxide compound.
4067. The device of item 3879, further comprising a visualization agent, wherein the visualization agent comprises a dye, pigment, or colorant.
4068. The device of item 3879, further comprising an echogenic material.
4069. The device of item 3879, furtheir comprising an echogenic material, wherein the echogenic material is fin the form of a coating.
4070. The device of item 3879 wherein the device is sterile.
4071. The device of item 3879 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device.
4072. The device of item 3879 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is connective tissue. 4073. The device of item 3879 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is muscle tissue.
4074. The device of item 3879 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is nerve tissue.
4075. The device of item 3879 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is epithelium tissue.
4076. The device of item 3879 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from the time of deployment of the device to about 1 year.
4077. The device of item 3879 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from about 1 month to 6 months.
4078. The device of item 3879 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from about 1 - 90 days.
4079. The device of item 3879 wherein the anti-scarring agent is released in effective concentrations from the device at a constant rate.
4080. The device of item 3879 wherein the anti-scarring agent is released in effective concentrations from the device at an increasing rate. 4081. The device of item 3879 wherein the anti-scarring agent is released in effective concentrations from the device at a decreasing rate.
4082. The device of item 3879 wherein the anti-scarring agent is released in effective concentrations from the composition comprising the anti-scarring agent by diffusion over a period ranging from the time of deployment of the device to about 90 days.
4083. The device of item 3879 wherein the anti-scarring agent is released in effective concentrations from the composition comprising the anti-scarring agent by erosion of the composition over a period ranging from the time of deployment of the device to about 90 days.
4084. The device of item 3879 wherein the device comprises about 0.01 Dg to about 10 Dg of the anti-scarring agent.
4085. The device of item 3879 wherein the device comprises about 10 Dg to about 10 mg of the anti-scarring agent.
4086. The device of item 3879 wherein the device comprises about 10 mg to about 250 mg of the anti-scarring agent.
4087. The device of item 3879 wherein the device comprises about 250 mg to about 1000 mg of the anti-scarring agent.
4088. The device of item 3879 wherein the device comprises about 1000 mg to about 2500 mg of the anti-scarring agent.
4089. The device of item 3879 wherein a surface of the device comprises less than 0.01 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied. 4090. The device of item 3879 wherein a surface of the device comprises about 0.01 Dg to about 1 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
4091. The device of item 3879 wherein a surface of the device comprises about 1 Dg to about 10 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
4092. The device of item 3879 wherein a surface of the device comprises about 10 Dg to about 250 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
4093. The device of item 3879 wherein a surface of the device comprises about 250 Dg to about 1000 Dg of the anti-scarring agent of anti- scarring agent per mm2 of device surface to which the anti-scarring agent is applied. - - —
4094. The device of item 3879 wherein a surface of the device comprises about 1000 Dg to about 2500 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
4095. A method as in any one of items 3879-4094, wherein the device is a ventriculopleural shunt.
4096. A method as in any one of items 3879-4094, wherein the device is a jugular vein shunt.
4097. A method as in any one of items 3879-4094, wherein the device is a vena cava shunt.
4098. A method as in any one of items 3879-4094, wherein the device is a ventriculoperitoneal shunt. 4099. A method as in any one of items 3879-4094, wherein the device is a gallbladder shunt.
4100. A method as in any one of items 3879-4094, wherein the device is a peritoneum shunt.
4101. A method as in any one of items 3879-4094, wherein the device is an external ventricular drainage device.
4102. A method as in any one of items 3879-4094, wherein the device is an intracranial pressure monitoring device.
4103. A method as in any one of items 3879-4094, wherein the device is a dural patch.
4104. A method as in any one of items 3879-4094, wherein the device is an implant to prevent epidural fibrosis post-laminectomy.
4105. A method as in any one of items 3879-4094, wherein the device is a device for continuous subarachnoid infusion.
4106. A method as in any one of items 3879-4094, wherein the device is a drainage shunt useful for draining fluids in the brain.
4107. A method as in any one of items 3879-4094, wherein the device is a pressure monitoring device.
4108. A device, comprising an inferior vena cava filter (e.g., an implant) and an anti-scarring agent or a composition comprising an anti- scarring agent, wherein the agent inhibits scarring between the device and a host into which the device is implanted.
4109. The device of item 4108 wherein the agent is an adensosine A2A receptor antagonist. 4110. The device of item 4108 wherein the agent is an AKT inhibitor.
4111. The device of item 4108 wherein the agent is an alpha 2 integrin antagonist, wherein the alpha 2 integrin antagonist is Pharmaprojects No. 5754 (Merck KgaA).
4112. The device of item 4108 wherein the agent an alpha 4 integrin antagonist.
4113. The device of item 4108 wherein the agent is an alpha 7 nicotinic receptor agonist.
4114. The device of item 4108 wherein the agent is an angiogenesis inhibitor selected from the group consisting of AG-12,958 (Pfizer), ATN-161 (Attenuon LLC), neovastat, an angiogenesis inhibitor from Jerina AG (Germany), NM-3 (Mercian), VGA-1155 (Taisho), FCE-26644 (Pfizer), FCE-26950 (Pfizer), FPMA (Meiji Daries), FR-111142 (Fujisawa), GGTI-298, GM-1306 (Ligand), GPA-1734 (Novartis), NNC-47-0011 (Novo Nordisk), herbamycin (Nippon Kayaku), lenalidomide (Celegene), IP-10 (NIH), ABT-828 (Abbott), KIN-841 (Tokushima University, Japan), SF-1126 (Semafore Pharmaceuticals), laminin technology (NIH), CHIR-258 (Chiron), NVP-AEW541 (Novartis), NVP-AEW541 (Novartis), Vt16907 (Alchemia), OXI-8007 (Oxigene), EG-3306 (Ark Therapeutics), Maspin (Arriva), ABT- 567 (Abbott), PPI-2458 (Praecis Pharmaceuticals), CC-5079, CC-4089 (Celgene), HIF-1 alpha inhibitors (Xenova), S-247 (Pfizer), AP-23573 (Ariad), AZD-9935 (Astra Zeneca), mebendazole (Introgen Therapeutics), MetAP-2 inhibitors (GlaxoSmithKline), AG-615 (Angiogene Pharmaceuticals), Tie-2 antagonists (Hybrigenics), NC-381 , CYC-381, NC-169, NC-219, NC-383, NC-384, NC-407 (Lorus Therapeutics), ATN-224 (Attenuon), ON-01370 (Onconova), Vitronectin antagonists (Amgen), SDX-103 (Salmedix), Vitronectin antagonists (Shire), CHP (Riemser), TEK (Amgen), Anecortave acetate (Alcon), T46.2 (Matrix Therapeutics), HG-2 (Heptagen), TEM antagonists (Genzyme), Oxi-4500 (Oxigene), ATN-161 (Attenuon), WX-293 (Wilex), M-2025 (Metris Therapeutics), Alphastatin (BioActa), YH-16 (Yantai Rongchang), BIBF-1120 (Boehringer Ingelheim), BAY-57-9352 (Bayer), AS- 1404 (Cancer Research Technology), SC-77964 (Pfizer), glycomimetics (BioTie Therapies), TIE-2 Inhibitors (Ontogen), DIMI, Octamer (Octamer), ABR-215050 (Active Biotech), ABT-518 (Abbott), KDR inhibitors (Abbott), BSF-466895 (Abbott), SCH-221153 (Schering-Plough), DAC:antiangiogenic (ConjuChem), TFPI (EntreMed), AZD-2171 (Astra-Zenaca), CDC-394 (Celgene), LY290293 (EIi Lilly), IDN-5390 (Indena), Kdr Kinase Inhibitors (Merck), CT-113020, CT-116433, CT-116563, CT-31890, CT-32228) (Cell Therapeutics), A-299620 (Abbott), TWEAK Inhibitor (Amgen), VEGF modulators (Johnson and Johnson), Tum-N53, tumstatin (Genzyme), Thios- 1 , Thios-2 (Thios Pharmaceuticals), MV-6401 (Miravant Medical -Technologies), Spisulosine (PharmaMar), GEP-7055 (Cephalon), AUV-201 (Auvation), LM-609 (EIi Lilly), SKF-106615 (AnorMED), Oglufanide disodium (Cytran), BW-114 (Pharminox), Calreticulin (NIH), WX-678 (Wilex), SD-7784 (Pfizer), WX-UK1 (Wilex), SH-268 (Schering AG), 2-Me-PGA (Celgene), S- 137 (Pfizer), ZD-6126 (Angiogene Pharmaceuticals), SG-292 (SignalGen), Benefin (Lane Labs), A6, A36 (Angstrom), SB-2723005 (GlaxoSmithKline), SC-7 (Cell Therapeutics), ZEN-014 (AEterna Zentaris), 2-methoxyestradiol (EntreMed), NK-130119 (Nippon Kayaku), CC-10004 (Celgene), AVE- 8062A (Ajinomoto), Tacedinaline (Pfizer), Actinonin (Tokyo Metropolitan Institute of Medical Science), Lenalidomide (Celgene), VGA-1155, BTO-956 (SRI International), ER-68203-00 (Eisai), CT-6685 (UCB), JKC-362 (Phoenix Pharmaceuticals), DMI-3798 (DMI Biosciences, Angiomate (Ipsen), ZD-6474 (AstraZeneca), CEP-5214 (Cephalon), Canstatin (Genzyme), NM-3 (Mercian), Oridigm (MediQuest Therapeutics), Exherin (Adherex), BLS-0597 (Boston Life Sciences), PTC-299 (PTC Therapeutics), NPI-2358 (Nereus Pharmaceuticals), CGP-79787 (Novartis), AEE-788 (Novartis), CKD-732 (Chong Kun Dang), CP-564959 (OSI Pharmaceuticals), CM-101 (CarboMed), CT-2584, CT3501 (Cell Therapeutics), combretastatin and analogues and derivatives thereof (Oxigene), Rebimastat (Bristol-Meyers Squibb), Dextrin 2-sulfate (ML Laboratories), Cilengitide (Merk KGaA), NSC-706704 (Pharminox), KRN- 951 (Kirin Brewery), Ukrain, NSC-631570 (Nowicky Pharma), Tecogalan sodium (Daiichi Pharmaceutical), Tz-93 (Tsumura), TBC-1635 (Encysive Pharmaceuticals), TAN-1120 (Takeda), Semaxanib (Pfizer), BDI-7800 (Biopharmacopae), SD-186, SD-983 (Bristol-Meyers Squibb), SB-223245 (GlaxoSmithKline), SC-236 (Pfizer), RWJ-590973 (Johnson and Johnson), ILX-1850 (Genzyme), SC-68488, S-836 (Pfizer), CG-55069-11 (CuraGen), Ki-23057 (Kirin Brewery), CCX-700 (Chemoentryx), Pegaptanib octasodium (Giled Sciences), ANGIOCOL (available from Biostratum Inc.), or an analogue or derivative thereof.
4115. The device of item 4108 wherein the agent is an apoptosis antagonist.
4116. The device of item 4108 wherein the agent is an apoptosis activator.
4117. The device of item 4108 wherein the agent is a beta 1 integrin antagonist.
4118. The device of item 4108 wherein the agent is a beta tubulin inhibitor.
4119. The device of item 4108 wherein the agent is a blocker of enzyme production in Hepatitis C. 4120. The device of item 4108 wherein the agent is a Bruton's tyrosine kinase inhibitor.
4121. The device of item 4108 wherein the agent is a calcineurin inhibitor.
4122. The device of item 4108 wherein the agent is a caspase 3 inhibitor.
4123. The device of item 4108 wherein the agent is a CC chemokine receptor antagonist.
4124. The device of item 4108 wherein the agent is a cell cycle inhibitor selected from the group consisting of SNS-595 (Sunesis), synthadotin, KRX-0403, and an analogue or derivative thereof.
4125. The device of item 4108 wherein the agent is a cathepsin B inhibitor.
4126. The device of item^iOδ wherein the agent is a cathepsin K inhibitor, wherein the cathepsin K inhibitor is 462795 (GlaxoSmithKline), INPL-022-D6 (Amura Therapeutics), an analogue or derivative thereof).
4127. The device of item 4108 wherein the agent is a cathepsin L inhibitor.
4128. The device of item 4108 wherein the agent is a CD40 antagonist. 4129. The device of item 4108 wherein the agent is a chemokine receptor agonist.
4130. The device of item 4108 wherein the agent is a chymase inhibitor.
4131. The device of item 4108 wherein the agent is a collagenase antagonist.
4132. The device of item 4108 wherein the agent is a CXCR antagonist.
4133. The device of item 4108 wherein the agent is a cyclin dependent kinase inhibitor selected from the group consisting of a CDK-1 inhibitor, a CDK-2 inhibitor, a CDK- 4 inhibitor, a CDK-6 inhibitor, a CAK1 inhibitor from GPC Biotech and Bristol-Myers Squibb, RGB-286199 (GPC Biotech), an anticancer agent from Astex Technology, a CAK1 inhibitor from GPC Biotech, a CDK inhibitor from Sanofi-Aventis, a CDK1/CDK2 inhibitor from Amgen, a CDK2 inhibitor from SUGEN-2 (Pfizer), a hearing loss therapy agent (Sound Pharmaceuticals), PD-0332991 (Pfizer), RGB-286199 (GPC Biotech), Ro-0505124 (Hoffmann-La Roche), a Ser/Thr kinase inhibitor from Lilly (EIi Lilly), CVT-2584 (CAS No. 199986-75-9) (CV Therapeutics), and an analogue or derivative thereof.
4134. The device of item 4108 wherein the agent is a cyclooxygenase 1 inhibitor.
4135. The device of item 4108 wherein the agent is a DHFR inhibitor. 4136. The device of item 4108 wherein the agent is a dual integrin inhibitor.
4137. The device of item 4108 wherein the agent is an elastase inhibitor.
4138. The device of item 4108 wherein the agent is an elongation factor-1 alpha inhibitor.
4139. The device of item 4108 wherein the agent is an endothelial growth factor antagonist.
4140. The device of item 4108 wherein the agent is an endothelial growth factor receptor kinase inhibitor selected from the group consisting of sorafenib tosylate (Bayer), AAL-993 (Novartis), ABP-309 (Novartis), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), E- 7080 (Eisai), EG-3306 (Ark Therapeutics), EXEL-2880 (Exelixis), GW- 654652 (GlaxoSmithKline), a KDR inhibitor from LG Life Sciences, CT-6685 and CT-6729 (UCB), KRN-633 and KRN-951 (Kirin Brewery), OSI-930 (OSI Pharmaceuticals), SP-5.2 (Supratek Pharma), SU-11657 (Pfizer), a Tie-2 antagonist (Hybrigenics), a VEGFR-2 kinase inhibitor (Bristol-Myers Squibb), XL-647 (Exelixis), a KDR inhibitor from Abbott Laboratories, sorafenib tosylate, and an analogue or derivative thereof.
4141. The device of item 4108 wherein the agent is an endotoxin antagonist.
4142. The device of item 4108 wherein the agent is an epothilone and tubulin binder. 4143. The device of item 4108 wherein the agent is an estrogen receptor antagonist.
4144. The device of item 4108 wherein the agent is an FGF inhibitor.
4145. The device of item 4108 wherein the agent is a farnexyl transferase inhibitor.
4146. The device of item 4108 wherein the agent is famesyltransferase inhibitor selected from the group of A-197574 (Abbott), a farnesyltransferase inhibitor from Servier, FPTIII (Strathclyde Institute for Drug R), LB-42908 (LG Life Sciences), Pharmaprojects No. 5063 (Genzyme), Pharmaprojects No. 5597 (Ipsen), Yissum Project No. B-1055 (Yissum), and an analogue or derivative thereof.
4147. The device of item 4108 wherein the agent is an FLT-3 kinase inhibitor.
4148. The device of item 4108 wherein the agent is an FGF receptor kinase inhibitor.
4149. The device of item 4108 wherein the agent is a fibrinogen antagonist selected from the group consisting of AUV-201 (Auvation), MG-13926 (Sanofi-Aventis), plasminogen activator (CAS No. 105913-11-9) (from Sanofi-Aventis or UCB), plasminogen activator-2 (tPA- 2) (Sanofi-Aventis), pro-urokinase (CAS No. 82657-92-9) (Sanofi-Aventis), and an analogue or derivative thereof.
4150. The device of item 4108 wherein the agent is a heat shock protein 90 antagonist selected from the group consisting of SRN-005 (Sirenade), geldanamycin, NSC-33050 (17-allylaminogeldanamycin; 17- AAG), ^-dimethylaminoethylamino-^-demethoxy-geldanamycin (17- DMAG), rifabutin (rifamycin XIV, r,4-didehydro-1-deoxy-1 ,4-dihydro-5'-(2- methylpropyl)-1-oxo-), and an analogue or derivative thereof.
4151. The device of item 4108 wherein the agent is a histone deacetylase inhibitor.
4152. The device of item 4108 wherein the agent is an HMGCoA reductase inhibitor selected from the group consisting of an atherosclerosis therapeutic from Lipid Sciences, ATI-16000 (ARYx Therapeutics), KS-01-019 (Kos Pharmaceuticals), Pharmaprojects No. 2197 (Sanofi-Aventi), RP 61969 (Sanofi-Aventis), and an analogue or derivative thereof.
4153. The device of item 4108 wherein the agent is an ICAM inhibitor.
4154. The device of item 4108 wherein the agent is an IL, ICE and IRAK antagonist, wherein the antagonist is a CJ-14877, CP-424174 (Pfizer), NF-61 (Negma-Lerads), and an analogue or derivative thereof.
4155. The device of item 4108 wherein the agent is an IL-2 inhibitor.
4156. The device of item 4108 wherein the agent is an immunosuppressant selected from the group consisting of teriflunomide (Sanofi Aventis), chlorsulfaquinoxalone (NSC-339004), chlorsulfaquinoxalone sulfate, CS-712 (Sankyo), ismomultin alfa (CAS No. 457913-93-8) (Akzo Nobel), antiallergics from GenPat77, antiinflammatories or AT-005 (Androclus Therapeutics), autoimmune disease therapeutics from EpiVax, BN-007 (Bone), budesonide (CAS No. 51333-22- 3) (MAP Pharmaceuticals), CO-14 (Genzyme), edratide (CAS No. 433922- 67-9) (Teva), EP-314 (Enanta), eprovafen (CAS No. 101335-99-3) (Sanofi- Aventis), HWA-131 (CAS No. 118788-41-3) (Sanofi-Aventis), immunomodulators from MerLion Pharmaceuticals, immunosuppressives from Alchemia, IPL-12 (Inflazyme), MDL-9563 (CAS No. 27086-86-8) (Sanofi-Aventis), Pharmaprojects No. 2330 (Sanofi-Aventis), Pharmaprojects No. 6426 (Abgenix), PXS-25 (Pharmaxis), rosmarinic acid (CAS No. 20283-92-5) (Sanofi-Aventis), RP 42927 or RP 54745 (CAS No. 135330-08-4) (Sanofi-Aventis), SGN-35 (Seattle Genetics), ST-1959 (Sigma-Tau), type I diabetes therapy from SYNX Pharma, UNIL-88 (Debiopharm), VP-025 (Vasogen), VR-694 (Vectura), PRTX-001 (Protalex), and an analogue or derivative thereof.
4157. The device of item 4108 wherein the agent is an IMPDH (inosine monophosphate). - - - - - - - - -
4158. The device of item 4108 wherein the agent is an integrin antagonist.
4159. The device of item 4108 wherein the agent is an interleukin antagonist.
4160. The device of item 4108 wherein the agent is an inhibitor of type III receptor tyrosine kinase.
4161. The device of item 4108 wherein the agent is an irreversible inhibitor of enzyme methionine aminopeptidase type 2.
4162. The device of item 4108 wherein the agent is an isozyme selective delta protein kinase C inhibitor. 4163. The device of item 4108 wherein the agent a JAK3 enzyme inhibitor.
4164. The device of item 4108 wherein the agent is a JNK inhibitor.
4165. The device of item 4108 wherein the agent is a kinase inhibitor.
4166. The device of item 4108 wherein the agent is kinesin antagonist.
4167. The device of item 4108 wherein the agent is a kinesin antagonist.
4168. The device of item 4108 wherein the agent is a leukotriene inhibitor and antagonist selected from the group consisting of ambicromil (CAS No. 58805-38-2) (Sanofi-Aventis), amelubant (CAS No. 346735-24-8) (Boehringer Ingelheim), DW-1141 (Dong Wha), ebselen (Daiichi Pharmaceutical), ibudilast (Kyorin), leucotriene inhibitors from Sanofi-Aventis, lymphotoxin-beta receptor (LT-D) from Biogen Idee, Pharmaprojects No. 1535 and 2728 (CAS No. 119340-33-9) (Sanofi- Aventis), R-112 (Rigel), Rev-5367 (CAS No. 92532-05-3) (Sanofi-Aventis), RG-14893 (CAS No. 141835-49-6) (Sanofi-Aventis), RG-5901-A (CAS No. 101910-24-1), 92532-23-5, RP 66153 (CAS No. 142422-79-5), RP 66364 (CAS No.186916-92-5), RP 69698 (CAS No. 141748-00-7) (Sanofi-Aventis), SC-411930 (Pfizer), SC-41930 (CAS No. 120072-59-5) (Pfizer), SC-50605 (CAS No. 138828-39-4) (Pfizer), SC-51146 (CAS No. 141059-52-1), SC- 53228 (CAS No. 153633-01-3) (Pfizer), spaglumic acid (ZY-15106) (CAS No.3106-85-2), 80619-64-3 (Novartis), tipredane (CAS No. 85197-77-9) (Bristol-Myers Squibb), U-75302 (CAS No. 119477-85-9) (Pfizer), and analogue or derivative thereof.
4169. The device of item 4108 wherein the agent is an MAP kinase inhibitor.
4170. The device of item 4108 wherein the agent is a matrix metalloproteinase inhibitor.
4171. The device of item 4108 wherein the agent is an MCP- CCR2 inhibitor.
4172. The device of item 4108 wherein the agent is an mTOR inhibitor.
4173. The device of item 4108 wherein the agent is an mTOR kinase inhibitor.
4174. The device of item 4108 wherein the agent is a microtubule inhibitor selected from the group consisting of antibody- maytansinoid conjugates from Biogen Idee, colchicines (MantiCore Pharmaceuticals), anticancer immunoconjugates from Johnson & Johnson, DIME from Octamer, gni-1f (GNI), huC242-DM4, huMy9-6-DM1 (ImmunoGen), IDN-5404 (Indena), IMO-098, IMOderm (Imotep), mebendazole (Introgen Therapeutics), microtubule poisons from Cambridge Enterprise, paclitaxel such as LOTAX from Aphios (CAS No. 33069-62-4), Genexol-PM from Samyang, Pharmaprojects No. 6383 (Tapestry Pharmaceuticals), RPR-112378 (Sanofi-Aventis), SGN-75 (Seattle Genetics), SPL-7435 (Starpharma), SSR-250411 (Sanofi-Aventis), trastuzumab-DM1 (Genentech), vinorelbine, and an analogue or derivative thereof. 4175. The device of item 4108 wherein the agent is an MIF inhibitor.
4176. The device of item 4108 wherein the agent is an MMP inhibitor.
4177. The device of item 4108 wherein the agent is a neurokinin (NK) antagonist selected from the group consisting of anthrotainin (CAS No. 148084-40-6) (Sanofi-Aventis), an IBS thereapeutic from ArQuIe1 MDL-105212A (CAS No. 167261-60-1) (Ssanofi-Aventis), Pharmaprojects No. 2744, 3258 (CAS No. 139167-47-8) 4006, 4201 , or 5986 (Sanofi-Aventis), RP 67580 (CAS No. 135911-02-3), SR-144190 (CAS No. 201152-86-5), SSR-240600, SSR-241586 (Sanofi-Aventis), TKA-457 (Novartis), vestipitant mesylate (CAS No. 334476-64-1) (GlaxoSmithKline), Win-64821 (Sanofi-Aventis), PRX-96026 (Predix Pharmaceuticals), and an analogue or derivative thereof.
4178. The device of item 4108 wherein the agent is an NF kappa B inhibitor selected from the group consisting of AVE-0545 or AVE- 0547 (Sanofi-Aventis), bortezomib (CAS No. 179324-69-7) (Millennium Pharmaceuticals), dexanabinol (CAS No. 112924-45-5) (Pharmos), dexlipotam (Viatris), Pharmaprojects No. 6283 (INDRA) (OXiGENE), IPL- 576092 (CAS No. 137571-30-3) (Inflazyme), NFKB decoy (Corgentech), NFKB decoy oligo (AnGes MG), NFKB's from Ariad, osteoporosis treatments or S5 (F005) from Fulcrum Pharmaceuticals, P61 (Phytopharm), R-flurbiprofen (CAS No. 5104-49-4) (Encore Pharmaceuticals), and an analogue or derivative thereof.
4179. The device of item 4108 wherein the agent is a nitric oxide agonist. 4180. The device of item 4108 wherein the agent is an ornithine decarboxylase inhibitor.
4181. The device of item 4108 wherein the agent is a p38 MAP kinase inhibitor selected from the group consisting of AZD-6703 (AstraZeneca), JX-401 (Jexys Pharmaceuticals), BMS-2 (Bristol-Myers Squibb), a p38 MAP kinase inhibitor from Novartis, a p38-alpha MAP kinase inhibitor from Amphora, Pharmaprojects No. 5704 (Pharmacopeia), RPR- 200765A (Sanofi-Aventis), SD-282 (Johnson & Johnson), TAK-715 (Takeda), and an analogue or derivative thereof.
4182. The device of item 4108 wherein the agent is a palmitoyl-protein thioesterase inhibitor.
4183. The device of item 4108 wherein the agent is a PDGF receptorkinase inhibitor selected from the group consisting of AAL-993, AMN-107, or ABP-309 (Novartis), AMG-706 (Amgen), BAY-57-9352 (Bayer), CDP-860 (UCB), E-7080 (Eisai), imatinib (CAS No. 152459-95-5) (Novartis), OSI-930 (OSI Pharmaceuticals), RPR-127963E (Sanofi-Aventis), RWJ-540973 (Johnson & Johnson), sorafenib tosylate (Bayer), SU-11657 (Pfizer), tandutinib (CAS No. 387867-13-2) (Millennium Pharmaceuticals), vatalanib (Novartis), ZK-CDK (Schering AG), and an analogue or derivative thereof.
4184. The device of item 4108 wherein the agent is (-)- halofenate (Metabolex), AMG-131 (Amgen), antidiabetics from Japan Tobacco, AZD-4619, AZD-8450, AZD-8677 (AstraZeneca), DRF-10945, balaglitazone (Dr Reddy's), CS-00088, CS-00098 (Chipscreen Biosciences), E-3030 (Eisai), etalocib (CAS No. 161172-51-6) (EIi Lilly), GSK-641597 (Ligand), GSK-677954 (GlaxoSmithKline), GW-409544 (Ligand), GW- 590735 (GlaxoSmithKline), K-111 (Hoffmann-La Roche), LY-518674 (EIi Lilly), LY-674 (Ligand), LY-929 (Ligand), MC-3001 , MC-3002 (MaxoCore Pharmaceuticals), metformin HCI + pioglitazone (CAS No. 1115-70-4 and 112529-15-4), ACTOPLUS MET from Andrx), muraglitazar (CAS No. 331741-94-7) (Bristol-Myers Squibb), naveglitazar (Ligand), oleoylethanolamide (Kadmus Pharmaceuticals), ONO-5129, pioglitazone hydrochloride (CAS No. 111025-46-8 and 112529-15-4) (Takeda), PLX-204 (Plexxikon), PPAR agonists from Genfit, PPAR delta agonists from EIi Lilly, PPAR-alpha agonists from CrystalGenomics, PPAR-gamma modulators and PPAR-β modulators from CareX, rosiglitazone maleate (CAS No. 122324- 73-4 or 155141-29-0) (GlaxoSmithKline), rosiglitazone maleate/glimepir (CAS No. 155141-29-0 and 93479-97-1), AVANDARYL, rosiglitazone maleate/metformin extend (CAS No. 155141-29-0 and 657-24-9), AVANDAMET, rosiglitazone maleate+metformin, AVANDAMET (GlaxoSmithKline), tesaglitazar (AstraZeneca), and an analogue or derivative thereof.
4185. The device of item 4108 wherein the agent is a phosphatase inhibitor.
4186. The device of item 4108 wherein the agent is a phosphodiesterase (PDE) inhibitor selected from the group consisting of avanafil (Tanabe Seiyaku), dasantafil (CAS No. 569351-91-3) (Schering- Plough), A-906119 (CAS No. 134072-58-5), DL-850 (Sanofi-Aventis), GRC- 3015, GRC-3566, GRC-3886 (Glenmark), HWA-153 (CAS No. 56395-66-5) (Sanofi-Aventis), hydroxypumafentrine (Altana), IBFB-130011 , IBFB-14-016, IBFB-140301 , IBFB-150007, IBFB-211913 (IBFB Pharma), L-826141 (Merck & Co), medorinone (CAS No. 88296-61-1) (Sanofi-Aventis), MEM- 1917 (Memory Pharmaceuticals), ND-1251 (Neuro3d), PDE inhibitors from ICOS, PDE IV inhibitors from Memory Pharmaceuticals and CrystalGenomics, Pharmaprojects No. 2742 and 6141 (Sanofi-Aventis), QAD-171 (Novartis), RHC-2963 (CAS No. 76993-12-9 and 76993-14-1), RPR-117658, RPR-122818 derivatives, SR-24870 , and RPR-132294 (Sanofi-Aventis), SK-350 (ln2Gen), stroke therapy agents from deCODE Genetics, TAS-203 (Taiho), tofimilast (CAS No. 185954-27-2) (Pfizer), UK- 371800 (Pfizer), WIN-65579 (CAS No. 158020-82-7) (Sanofi-Aventis), IBFB- 130020 (IBFB Pharma), OPC-6535 (CAS No. 145739-56-6) (Otsuka), a phosphodiesterase III inhibitor, enoximone, a phosphodiesterase IV inhibitor, fosfosal, Atopik (Barrier Therapeutics), triflusal, a phosphodiesterase V inhibitor, and an analogue or derivative thereof.
4187. The device of item 4108 wherein the agent is a PKC inhibitor.
4188. The device of item 4108 wherein the agent is a platelet activating factor antagonist.
4189; The device of item 4108 wherein the agent is a platelet- derived growth factor receptor kinase inhibitor.
4190. The device of item 4108 wherein the agent is a prolyl hydroxylase inhibitor.
4191. The device of item 4108 wherein the agent is a polymorphonuclear neutrophil inhibitor.
4192. The device of item 4108 wherein the agent is a protein kinase B inhibitor.
4193. The device of item 4108 wherein the agent is a protein kinase C stimulant. 4194. The device of item 4108 wherein the agent is a purine nucleoside analogue.
4195. The device of item 4108 wherein the agent is a purinoreceptor P2X antagonist.
4196. The device of item 4108 wherein the agent is a Raf kinase inhibitor.
4197. The device of item 4108 wherein the agent is a reversible inhibitor of ErbB1 and ErbB2.
4198. The device of item 4108 wherein the agent is a ribonucleoside triphosphate reductase inhibitor.
4199r The device of item 4108 wherein the agent is an SDF-1 antagonist.
4200. The device of item 4108 wherein the agent is a sheddase inhibitor.
4201. The device of item 4108 wherein the agent is an SRC inhibitor.
4202. The device of item 4108 wherein the agent is a stromelysin inhibitor.
4203. The device of item 4108 wherein the agent is an Syk kinase inhibitor. 4204. The device of item 4108 wherein the agent is a telomerase inhibitor.
4205. The device of item 4108 wherein the agent is a TGF beta inhibitor selected from the group consisting of pirfenidone (CAS No. 53179-13-8) (MARNAC), tranilast (CAS No. 53902-12-8) (Kissei), IN-1130 (ln2Gen), mannose-6-phosphate (BTG), TGF-β antagonists from Inflazyme (Pharmaprojects No. 6075), TGF-β antagonists from Sydney, non-industrial source), TGF-β I receptor kinase inhibitors from EIi Lilly, TGF-β receptor inhibitors from Johnson & Johnson, and an analogue or derivative thereof.
4206. The device of item 4108 wherein the agent is a TNFα antagonist or TACE inhibitor selected from the group consisting of adalimumab (CAS No. 331731-18-1) (Cambridge Antibody Technology), AGIX-4207 (AtheroGenics), AGT-1 (Advanced Biotherapy), an antiinflammatory from Borean Pharma, Cellzomer or Paradigm Therapeutics, anti-inflammatory vaccine (TNF-alpha kinoid) from Neovacs, humanized anti-TNF antibody or an anti-TNF MAb (CB0006) Celltech (UCB), apratastat (CAS No. 287445-51-0) (Wyeth), BMS-561392 (Bristol-Myers Squibb), BN- 006 (Bone), certolizumab pegol (CAS No. 428863-50-7 or CH-138 (UCB), cilomilast (CAS No. 153259-65-5) (GlaxoSmithKline), CR-1 (Nuada Pharmaceuticals), CRx-119 (CombinatoRx), D-5410 (UCB), dacopafant (CAS No. 125372-33-0) (Sanofi-Aventis), dersalazine (CAS No. 188913-57- 7/188913-58-8) (Uriach), etanercept (CAS No. 185243-69-0) (Amgen), ethyl pyruvate (Critical (Critical Therapeutics), golimumab (CAS No. 476181-74-5) (Johnson & Johnson), hormono-immunotherapy from Ipsen, CDP571 (e.g., Humicade from UCB), IC-485 (ICOS), infliximab (CAS No. 170277-31-3) (Johnson & Johnson), IP-751 (Manhattan Pharmaceuticals), ISIS-104838 (CAS No. 250755-32-9) (ISIS Pharmaceuticals), lenalidomide (CAS No. 191732-72-6) (Celgene), lentinan (CAS No. 37339-90-5) (Ajinomoto), MDL- 201112 (CAS No. 142130-73-2) (Sanofi-Aventis), medroxyprogesterone (CAS No. 520-85-4) (InKine Pharmaceutical), N-acetylcysteine (CAS No. 616-91-1) (Zambon), NBE-P2 (DIREVO Biotech), nerelimomab (CAS No. 162774-06-3) (Chiron), OM-294DP (OM PHARMA)1 onercept (CAS No. 199685-57-9) (Yeda), PASSTNF-alpha (Verigen), pentoxifylline or oxypentifylline (Sanofi-Aventis), Pharmaprojects No. 4091 , 4241 , 4295, or 4488 (Sanofi-Aventis), Pharmaprojects No. 5480 (Amgen), Pharmaprojects No. 6749 (Cengent), pirfenidone (CAS No. 53179-13-8) (MARNAC), RPR- 132294 (Sanofi-Aventis), S5 (F002) (Fulcrum Pharmaceuticals), simvastatin (CAS No. 79902-63-9) (Merck & Co), STA-6292 (Synta Pharmaceuticals), tacrolimus (CAS No. 104987-11-3) (Fujisawa LifeCycle Pharma), talactoferrin alfa (CAS No. 308240-58-6) (Agennix), thalidomide (CAS No. 50-35-1) (Celgene), TNF antagonists form ProStrakan, and Synergen, TNF inhibitors (Amgen), TNF-alpha antagonists from Dynavax Technologies and Jerina AG (Germany), TNF-alpha inhibitors from IBFB Pharma and Xencor (Xencor), torbafylline (CAS No. 105102-21-4) (Sanofi-Aventis), UR-1505 (Uriach), VT-346 (Won Therapeutics), YSIL6χY's Therapeutics), YSTH2 (Y's Therapeutics), NPI-1302a-3 (Nereus Pharmaceuticals, a TNF antagonist from Jerina AG (Germany), dersalazine, and an analogue or derivative thereof.
4207. The device of item 4108 wherein the agent is a Toll receptor inhibitor.
4208. The device of item 4108 wherein the agent is a tubulin antagonist.
4209. The device of item 4108 wherein the agent is a tyrosine kinase inhibitor selected from the group consisting of SU-011248, SUTENT from Pfizer Inc. (New York, NY), BMS-354825, PN-355 (Paracelsian Pharmaceuticals), AGN-199659 (Allergan), AAL-993 or ABP-309 (Novartis), adaphostin (NIH), AEE-788 (Novartis), AG-013736 (OSI Pharmaceuticals), AG-13736 (Pfizer), ALT-110 (Alteris Therapeutics), AMG-706 (Amgen), anticancer MAbs from Xencor, anti-EGFrvlll MAbs from Abgenix, anti-HER2 MAb from Abiogen, AZD-2171 or AZD-9935 (AstraZeneca), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), CEP-5214 (Cephalon), CEP- 7055 (Cephalon), cetuximab (ImCIone Systems), CHIR-200131 and CHIR- 258 (Chiron), CP-547632 (OSI Pharmaceuticals), CP-724714 (Pfizer), CT- 301 (Creabilis Therapeutics), D-69491 (Baxter International), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EGFR/ErbB2 inhibitors from Array BioPharma, erlotinib (CAS No. 183319-69-9) (OSI Pharmaceuticals), EXEL-2880 (Exelixis), FK-778 (Sanofi-Aventis) , gefitinib (CAS No. 184475-35-2) (AstraZeneca), GW-2286 or GW-654652 (GlaxoSmithKline), her2/neu antigen from AlphaVax, HER-2/neu inhibitor from Generex, Herzyme (Medipad) (Sirna Therapeutics), HKI-272 (Wyeth), HuMax-EGFr (Genmab), idronoxil (CAS No. 81267-65-4) (Novogen), IGF-1 inhibitors from Ontogen, IMC-11 F8 (ImCIone Systems), kahalalide F (CAS No. 149204-42-2) (PharmaMar), KDR inhibitorfrom LG Life Sciences, KDR inhibitors from Abbott Laboratories, KDR kinase inhibitors (UCB), Kdr kinase inhibitors from Merck & Co, KRN-633 and KRN-951 (Kirin Brewery), KSB-102 (Xenova), lapatinib ditosylate (CAS No. 388082-78-8) (GlaxoSmithKline), matuzumab (Merck KGaA), MDX-214 (Medarex), ME-103 (Pharmexa), MED-A300 (Gerolymatos), MNAC-13 (Lay Line Genomics), nimotuzumab (Center of Molecular Immunology), NSC-330507 or NSC-707545 (NIH), NV-50 (Novogen), OSI-930 (OSI Pharmaceuticals), panitumumab (Abgenix), pelitinib (CAS No. 287933-82-7) (Wyeth), pertuzumab (CAS No. 380610-27- 5) (Genentech), Pharmaprojects No. 3985 (Sanofi-Aventis), prostate cancer therapeutics from Sequenom (SQPC35, SQPC36, SQPC90), removab and remoxab (Trion Pharma), RG-13022 (CAS No. 136831-48-6), RG-13291 (CAS No. 138989-50-1), or RG-14620 (CAS No. 136831-49-7) (Sanofi- Aventis), RM-6427 (Romark), RNAi breast cancer therapy from Benitec, RP 53801 (CAS No. 125882-88-4) (Sanofi-Aventis), sorafenib tosylate (Bayer), SU-11657 (Pfizer), Tie-2 antagonists from Semaia (Hybrigenics), Tie-2 inhibitors from Ontogen, trastuzumab (CAS No. 180288-69-1) (Genentech), tyrosine kinase inhibitors from Sanofi-Aventis, U3-1287, U3-1565, U3-1784, U3-1800 (U3 Pharma), vatalanib (Novartis), VEGFR-2 kinase inhibitor from Bristol-Myers Squibb, XL-647 (Exelixis), ZD-6474 (AstraZeneca), ZK-CDK (Schering AG), an EGFR tyrosine kinase inhibitor, EKB-569 (Wyeth), and an analogue or derivative thereof.
4210. The device of item 4108 wherein the agent is a VEGF inhibitor.
4211. The device of item 4108 wherein the agent is a vitamin D receptor agonist.
4212. The device of item 4108 wherein the agent is ZD-6474 (an angiogenesis inhibitor).
4213. The device of item 4108 wherein the agent is AP-23573 (an mTOR inhibitor).
4214. The device of item 4108 wherein the agent is synthadotin (a tubulin antagonist).
4215. The device of item 4108 wherein the agent is S-0885 (a collagenase inhibitor).
4216. The device of item 4108 wherein the agent is aplidine (an elongation factor-1 alpha inhibitor).
4217. The device of item 4108 wherein the agent is ixabepilone (an epithilone). 4218. The device of item 4108 wherein the agent is IDN-5390 (an angiogenesis inhibitor).
4219. The device of item 4108 wherein the agent is SB- 2723005 (an angiogenesis inhibitor).
4220. The device of item 4108 wherein the agent is ABT-518 (an angiogenesis inhibitor).
4221. The device of item 4108 wherein the agent is combretastatin (an angiogenesis inhibitor).
4222. The device of item 4108 wherein the agent is anecortave acetate (an angiogenesis inhibitor).
4223. The device of item 4108 wherein the agent is SB- 715992 (a kinesin antagonist).
4224. The device of item 4108 wherein the agent is temsirolimus (an mTOR inhibitor).
4225. The device of item 4108 wherein the agent is adalimumab (a TNFα antagonist).
4226. The device of item 4108, further comprising a polymer.
4227. The device of item 4108 wherein the anti-scarring agent inhibits adhesion between the device and a host into which the device is implanted. 4228. The device of item 4108 wherein the device delivers the anti-scarring agent locally to tissue proximate to the device.
4229. The device of item 4108, further comprising a coating, wherein the coating comprises the anti-scarring agent.
4230. The device of item 4108, further comprising a coating, wherein the coating is disposed on a surface of the device.
4231. The device of item 4108, further comprising a coating, wherein the coating directly contacts the device.
4232. The device of item 4108, further comprising a coating, wherein the coating indirectly contacts the device.
4233. The device of item 4108, further comprising a coating, wherein the coating partially covers the device.
4234. The device of item 4108, further comprising a coating, wherein the coating completely covers the device.
4235. The device of item 4108, further comprising a coating, wherein the coating is a uniform coating.
4236. The device of item 4108, further comprising a coating, wherein the coating is a non-uniform coating.
4237. The device of item 4108, further comprising a coating, wherein the coating is a discontinuous coating. 4238. The device of item 4108, further comprising a coating, wherein the coating is a patterned coating.
4239. The device of item 4108, further comprising a coating, wherein the coating has a thickness of 100 Dm or less.
4240. The device of item 4108, further comprising a coating, wherein the coating has a thickness of 10 Dm or less.
4241. The device of item 4108, further comprising a coating, wherein the coating adheres to the surface of the device upon deployment of the device.
4242. The device of item 4108, further comprising a coating, wherein the coating is stable at room temperature for a period of 1 year.
4243. The device of item 4108, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 0.0001% to about 1% by weight.
4244. The device of item 4108, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 1% to about 10% by weight.
4245. The device of item 4108, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 10% to about 25% by weight.
4246. The device of item 4108, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 25% to about 70% by weight. 4247. The device of item 4108, further comprising a coating, wherein the coating further comprises a polymer.
4248. The device of item 4108, further comprising a first coating having a first composition and the second coating having a second composition.
4249. The device of item 4108, further comprising a first coating having a first composition and the second coating having a second composition, wherein the first composition and the second composition are different.
4250. The device of item 4108, further comprising a polymeric carrier, wherein the polymeric carrier comprises a copolymer.
4251. The device of item 4108, further comprising a polymeric carrier, wherein the polymeric carrier comprises a block copolymer.
4252. The device of item 4108, further comprising a polymeric carrier, wherein the polymeric carrier comprises a random copolymer.
4253. The device of item 4108, further comprising a polymeric carrier, wherein the polymeric carrier comprises a biodegradable polymer.
4254. The device of item 4108, further comprising a polymeric carrier, wherein the polymeric carrier comprises a non-biodegradable polymer.
4255. The device of item 4108, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrophilic polymer. 4256. The device of item 4108, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrophobic polymer.
4257. The device of item 4108, further comprising a polymeric carrier, wherein the polymeric carrier comprises a polymer having hydrophilic domains.
4258. The device of item 4108, further comprising a polymeric carrier, wherein the polymeric carrier comprises a polymer having hydrophobic domains.
4259. The device of item 4108, further comprising a polymeric carrier, wherein the polymeric carrier comprises a non-conductive polymer.
4260. The device of item 4108, further comprising a polymeric carrier, wherein the polymeric carrier comprises an elastomer.
4261. The device of item 4108, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrogel.
4262. The device of item 4108, further comprising a polymeric carrier, wherein the polymeric carrier comprises a silicone polymer.
4263. The device of item 4108, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrocarbon polymer.
4264. The device of item 4108, further comprising a polymeric carrier, wherein the polymeric carrier comprises a styrene-derived polymer.
4265. The device of item 4108, further comprising a polymeric carrier, wherein the polymeric carrier comprises a butadiene polymer. 4266. The device of item 4108, further comprising a polymeric carrier, wherein the polymeric carrier comprises a macromer.
4267. The device of item 4108, further comprising a polymeric carrier, wherein the polymeric carrier comprises a poly(ethylene glycol) polymer.
4268. The device of item 4108, further comprising a polymeric carrier, wherein the polymeric carrier comprises an amorphous polymer.
4269. The device of item 4108, further comprising a lubricious coating.
4270. The device of item 4108 wherein the anti-scarring agent is located within pores or holes of the device.
4271. The device of item 4108 wherein the anti-scarring agent is located within a channel, lumen, or divet of the device.
4272. The device of item 4108, further comprising a second pharmaceutically active agent.
4273. The device of item 4108, further comprising an antiinflammatory agent.
4274. The device of item 4108, further comprising an agent that inhibits infection.
4275. The device of item 4108, further comprising an agent that inhibits infection, wherein the agent is an anthracycline. 4276. The device of item 4108, further comprising an agent that inhibits infection, wherein the agent is doxorubicin.
4277. The device of item 4108, further comprising an agent that inhibits infection, wherein the agent is mitoxantrone.
4278. The device of item 4108, further comprising an agent that inhibits infection, wherein the agent is a fluoropyrimidine.
4279. The device of item 4108, further comprising an agent that inhibits infection, wherein the agent is 5-fluorouracil (5-FU).
4280. The device of item 4108, further comprising an agent that inhibits infection, wherein the agent is a folic acid antagonist.
4281. The device of item 4108, further comprising an agent that inhibits infection, wherein the agent is methotrexate.
4282. The device of item 4108, further comprising an agent that inhibits infection, wherein the agent is a podophylotoxin.
4283. The device of item 4108, further comprising an agent that inhibits infection, wherein the agent is etoposide.
4284. The device of item 4108, further comprising an agent that inhibits infection, wherein the agent is a camptothecin.
4285. The device of item 4108, further comprising an agent that inhibits infection, wherein the agent is a hydroxyurea. 4286. The device of item 4108, further comprising an agent that inhibits infection, wherein the agent is a platinum complex.
4287. The device of item 4108, further comprising an agent that inhibits infection, wherein the agent is cisplatin.
4288. The device of item 4108, further comprising an antithrombotic agent.
4289. The device of item 4108, further comprising a visualization agent.
4290. The device of item 4108, further comprising a visualization agent, wherein the visualization agent is a radiopaque material, wherein the radiopaque material comprises a metal, a halogenated compound, or a barium containing compound:
4291. The device of item 4108, further comprising a visualization agent, wherein the visualization agent is a radiopaque material, wherein the radiopaque material comprises barium, tantalum, or technetium.
4292. The device of item 4108, further comprising a visualization agent, wherein the visualization agent is a MRI responsive material.
4293. The device of item 4108, further comprising a visualization agent, wherein the visualization agent comprises a gadolinium chelate. 4294. The device of item 4108, further comprising a visualization agent, wherein the visualization agent comprises iron, magnesium, manganese, copper, or chromium.
4295. The device of item 4108, further comprising a visualization agent, wherein the visualization agent comprises an iron oxide compound.
4296. The device of item 4108, further comprising a visualization agent, wherein the visualization agent comprises a dye, pigment, or colorant.
4297. The device of item 4108, further comprising an echogenic material.
- 4298. The device of item 4108, further comprising an echogenic material, wherein the echogenic material is in the form of a coating.
4299. The device of item 4108 wherein the device is sterile.
4300. The device of item 4108 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device.
4301. The device of item 4108 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is connective tissue. 4302. The device of item 4108 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is muscle tissue.
4303. The device of item 4108 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is nerve tissue.
4304. The device of item 4108 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is epithelium tissue.
4305. The device of item 4108 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from the time of deployment of the device to about 1 year.
4306. The device of item 4108 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from about 1 month to 6 months.
4307. The device of item 4108 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from about 1 - 90 days.
4308. The device of item 4108 wherein the anti-scarring agent is released in effective concentrations from the device at a constant rate.
4309. The device of item 4108 wherein the anti-scarring agent is released in effective concentrations from the device at an increasing rate. 4310. The device of item 4108 wherein the anti-scarring agent is released in effective concentrations from the device at a decreasing rate.
4311. The device of item 4108 wherein the anti-scarring agent is released in effective concentrations from the composition comprising the anti-scarring agent by diffusion over a period ranging from the time of deployment of the device to about 90 days.
4312. The device of item 4108 wherein the anti-scarring agent is released in effective concentrations from the composition comprising the anti-scarring agent by erosion of the composition over a period ranging from the time of deployment of the device to about 90 days.
4313. The device of item 4108 wherein the device comprises about 0.01 Dg to about 10 Dg of the anti-scarring agent.
4314. The device of item 4108 wherein the device comprises about 10 Dg to about 10 mg of the anti-scarring agent.
4315. The device of item 4108 wherein the device comprises about 10 mg to about 250 mg of the anti-scarring agent.
4316. The device of item 4108 wherein the device comprises about 250 mg to about 1000 mg of the anti-scarring agent.
4317. The device of item 4108 wherein the device comprises about 1000 mg to about 2500 mg of the anti-scarring agent.
4318. The device of item 4108 wherein a surface of the device comprises less than 0.01 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied. 4319. The device of item 4108 wherein a surface of the device comprises about 0.01 Dg to about 1 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
4320. The device of item 4108 wherein a surface of the device comprises about 1 Dg to about 10 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
4321. The device of item 4108 wherein a surface of the device comprises about 10 Dg to about 250 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
4322. The device of item 4108 wherein a surface of the device comprises about 250 Dg to about 1000 Dg of the anti-scarring agent of anti- scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
4323. The device of item 4108 wherein a surface of the device comprises about 1000 Dg to about 2500 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
4324. A method as in any one of items 4108-4323, wherein the device is a vascular filter.
4325. A method as in any one of items 4108-4323, wherein the device is a blood filter.
4326. A method as in any one of items 4108-4323, wherein the device is a caval filter.
4327. A method as in any one of items 4108-4323, wherein the device is a vena cava filter. 4328. A method as in any one of items 4108-4323, wherein the device is a thrombus filter.
4329. A method as in any one of items 4108-4323, wherein the device is an antimigration filter.
4330. A method as in any one of items 4108-4323, wherein the device is a percutaneous filter system.
4331. A method as in any one of items 4108-4323, wherein the device is an intravascular step.
4332. A method as in any one of items 4108-4323, wherein the device is an intravascular filter.
4333. A method as in any one of items 4108-4323, wherein the device is a clot filter.
4334. A method as in any one of items 4108-4323, wherein the device is a vein filter.
4335. A method as in any one of items 4108-4323, wherein the device is a body vessel filter.
4336. A device, comprising a gastrointestinal device (e.g., an implant) and an anti-scarring agent or a composition comprising an anti- scarring agent, wherein the agent inhibits scarring between the device and a host into which the device is implanted.
4337. The device of item 4336 wherein the agent is an adensosine A2A receptor antagonist.
4338. The device of item 4336 wherein the agent is an AKT inhibitor. 4339. The device of item 4336 wherein the agent is an alpha 2 integrin antagonist, wherein the alpha 2 integrin antagonist is Pharmaprojects No. 5754 (Merck KgaA).
4340. The device of item 4336 wherein the agent an alpha 4 integrin antagonist.
4341. The device of item 4336 wherein the agent is an alpha 7 nicotinic receptor agonist.
4342. The device of item 4336 wherein the agent is an angiogenesis inhibitor selected from the group consisting of AG-12,958 (Pfizer), ATN-161 (Attention LLC), neovastat, an angiogenesis inhibitor from Jerina AG (Germany), NM-3 (Mercian), VGA-1155 (Taisho), FCE-26644 (Pfizer), FCE-26950 (Pfizer), FPMA (Meiji Daries), FR-111142 (Fujisawa), GGTI-298, GM-1306 (Ligand), GPA-1-734 (Novartis), NNC-47-0011 (Novo Nordisk), herbamycin (Nippon Kayaku), lenalidomide (Celegene), IP-10 (NIH), ABT-828 (Abbott), KIN-841 (Tokushima University, Japan), SF-1126 (Semafore Pharmaceuticals), laminin technology (NIH), CHIR-258 (Chiron), NVP-AEW541 (Novartis), NVP-AEW541 (Novartis), Vt16907 (Alchemia), OXI-8007 (Oxigene), EG-3306 (Ark Therapeutics), Maspin (Arriva), ABT- 567 (Abbott), PPI-2458 (Praecis Pharmaceuticals), CC-5079, CC-4089 (Celgene), HIF-1 alpha inhibitors (Xenova), S-247 (Pfizer), AP-23573 (Ariad), AZD-9935 (Astra Zeneca), mebendazole (Introgen Therapeutics), MetAP-2 inhibitors (GlaxoSmithKline), AG-615 (Angiogene Pharmaceuticals), Tie-2 antagonists (Hybrigenics), NC-381, CYC-381, NC-169, NC-219, NC-383, NC-384, NC-407 (Lorus Therapeutics), ATN-224 (Atlenuon), ON-01370 (Onconova), Vitronectin antagonists (Amgen), SDX-103 (Salmedix), Vitronectin antagonists (Shire), CHP (Riemser), TEK (Amgen), Anecortave acetate (Alcon), T46.2 (Matrix Therapeutics), HG-2 (Heptagen), TEM antagonists (Genzyme), Oxi-4500 (Oxigene), ATN-161 (Attenuon), WX-293 (Wilex), M-2025 (Metris Therapeutics), Alphastatin (BioActa), YH-16 (Yantai Rongchang), BIBF-1120 (Boehringer Ingelheim), BAY-57-9352 (Bayer), AS- 1404 (Cancer Research Technology), SC-77964 (Pfizer), glycomimetics (BioTie Therapies), TIE-2 Inhibitors (Ontogen), DIMI, Octamer (Octamer), ABR-215050 (Active Biotech), ABT-518 (Abbott), KDR inhibitors (Abbott), BSF-466895 (Abbott), SCH-221153 (Schering-Plough), DAC.antiangiogenic (ConjuChem), TFPI (EntreMed), AZD-2171 (Astra-Zenaca), CDC-394 (Celgene), LY290293 (EIi Lilly), IDN-5390 (Indena), Kdr Kinase Inhibitors (Merck), CT-113020, CT-116433, CT-116563, CT-31890, CT-32228) (Cell Therapeutics), A-299620 (Abbott), TWEAK Inhibitor (Amgen), VEGF modulators (Johnson and Johnson), Tum-N53, tumstatin (Genzyme), Thios- 1 , Thios-2 (Thios Pharmaceuticals), MV-6401 (Miravant Medical Technologies), Spisulosine (PharmaMar), CEP-7055 (Cephalon), AUV-201 (Auvation), LM-609 (EIi Lilly), SKF-106615 (AnorMED), Oglufanide disodium (Cytran), BW-114 (Pharminox), Calreticulin (NIH), WX-678 (Wilex), SD-7784 (Pfizer), WX-UK1_<Wilex),-SH-268 (Schering AG), 2-Me-PGA (Celgene), S- 137 (Pfizer), ZD-6126 (Angiogene Pharmaceuticals), SG-292 (SignalGen), Benefin (Lane Labs), A6, A36 (Angstrom), SB-2723005 (GlaxoSmithKline), SC-7 (Cell Therapeutics), ZEN-014 (AEterna Zentaris), 2-methoxyestradiol (EntreMed), NK-130119 (Nippon Kayaku), CC-10004 (Celgene), AVE- 8062A (Ajinomoto), Tacedinaline (Pfizer), Actinonin (Tokyo Metropolitan Institute of Medical Science), Lenalidomide (Celgene), VGA-1155, BTO-956 (SRI International), ER-68203-00 (Eisai), CT-6685 (UCB), JKC-362 (Phoenix Pharmaceuticals), DMI-3798 (DMI Biosciences, Angiomate (Ipsen), ZD-6474 (AstraZeneca), CEP-5214 (Cephalon), Canstatin (Genzyme), NM-3 (Mercian), Oridigm (MediQuest Therapeutics), Exherin (Adherex), BLS-0597 (Boston Life Sciences), PTC-299 (PTC Therapeutics), NPI-2358 (Nereus Pharmaceuticals), CGP-79787 (Novartis), AEE-788 (Novartis), CKD-732 (Chong Kun Dang), CP-564959 (OSI Pharmaceuticals), CM-101 (CarboMed), CT-2584, CT3501 (Cell Therapeutics), combretastatin and analogues and derivatives thereof (Oxigene), Rebimastat (Bristol-Meyers Squibb), Dextrin 2-sulfate (ML Laboratories), Cilengitide (Merk KGaA), NSC-706704 (Pharminox), KRN- 951 (Kirin Brewery), Ukrain, NSC-631570 (Nowicky Pharma), Tecogalan sodium (Daiichi Pharmaceutical), Tz-93 (Tsumura), TBC-1635 (Encysive Pharmaceuticals), TAN-1120 (Takeda), Semaxanib (Pfizer), BDI-7800 (Biopharmacopae), SD-186, SD-983 (Bristol-Meyers Squibb), SB-223245 (GlaxoSmithKiine), SC-236 (Pfizer), RWJ-590973 (Johnson and Johnson), ILX-1850 (Genzyme), SC-68488, S-836 (Pfizer), CG-55069-11 (CuraGen), Ki-23057 (Kirin Brewery), CCX-700 (Chemoentryx), Pegaptanib octasodium (Giled Sciences), ANGIOCOL (available from Biostratum Inc.), or an analogue or derivative thereof.
4343. The device of item 4336 wherein the agent is an apoptosis antagonist.
4344. The device of item 4336 wherein the agent is an apoptosis activator.
4345. The device of item 4336 wherein the agent is a beta 1 integrin antagonist.
4346. The device of item 4336 wherein the agent is a beta tubulin inhibitor.
4347. The device of item 4336 wherein the agent is a blocker of enzyme production in Hepatitis C.
4348. The device of item 4336 wherein the agent is a Bruton's tyrosine kinase inhibitor. 4349. The device of item 4336 wherein the agent is a calcineurin inhibitor.
4350. The device of item 4336 wherein the agent is a caspase 3 inhibitor.
4351. The device of item 4336 wherein the agent is a CC chemokine receptor antagonist.
4352. The device of item 4336 wherein the agent is a cell cycle inhibitor selected from the group consisting of SNS-595 (Sunesis), synthadotin, KRX-0403, and an analogue or derivative thereof.
4353. The device of item 4336 wherein the agent is a cathepsin B inhibitor.
4354. The device of item 4336 wherein the agent is a cathepsin K inhibitor, wherein the cathepsin K inhibitor is 462795 (GlaxoSmithKline), INPL-022-D6 (Amura Therapeutics), an analogue or derivative thereof).
4355. The device of item 4336 wherein the agent is a cathepsin L inhibitor.
4356. The device of item 4336 wherein the agent is a CD40 antagonist.
4357. The device of item 4336 wherein the agent is a chemokine receptor agonist. 4358. The device of item 4336 wherein the agent is a chymase inhibitor.
4359. The device of item 4336 wherein the agent is a collagenase antagonist.
4360. The device of item 4336 wherein the agent is a CXCR antagonist.
4361. The device of item 4336 wherein the agent is a cyclin dependent kinase inhibitor selected from the group consisting of a CDK-1 inhibitor, a CDK-2 inhibitor, a CDK- 4 inhibitor, a CDK-6 inhibitor, a CAK1 inhibitor from GPC Biotech and Bristol-Myers Squibb, RGB-286199 (GPC Biotech), an anticancer agent from Astex Technology, a CAK1 inhibitor from GPC Biotech, a CDK inhibitor from Sanofi-Aventis, a CDK1/CDK2 inhibitor from Amgen, a CDK2 inhibitor from SUGEN-2 (Pfizer), a hearing loss therapy agent (Sound Pharmaceuticals), PD-0332991 (Pfizer), RGB-286199 (GPC Biotech), Ro-0505124 (Hoffmann-La Roche), a Ser/Thr kinase inhibitor from Lilly (EIi Lilly), CVT-2584 (CAS No. 199986-75-9) (CV Therapeutics), and an analogue or derivative thereof.
4362. The device of item 4336 wherein the agent is a cyclooxygenase 1 inhibitor.
4363. The device of item 4336 wherein the agent is a DHFR inhibitor.
4364. The device of item 4336 wherein the agent is a dual integrin inhibitor. 4365. The device of item 4336 wherein the agent is an elastase inhibitor.
4366. The device of item 4336 wherein the agent is an elongation factor-1 alpha inhibitor.
4367. The device of item 4336 wherein the agent is an endothelial growth factor antagonist.
4368. The device of item 4336 wherein the agent is an endothelial growth factor receptor kinase inhibitor selected from the group consisting of sorafenib tosylate (Bayer), AAL-993 (Novartis), ABP-309 (Novartis), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), E- 7080 (Eisai), EG-3306 (Ark Therapeutics), EXEL-2880 (Exelixis), GW- 654652 (GlaxoSmithKline), a KDR inhibitor from LG Life Sciences, CT-6685 and CT-6729 (UCB), KRN-633 and KRN-951 (Kirin Brewery), OSI-930 (OSI Pharmaceuticals), SP-5.2 (Supratek Pharma), SU-11657 (Pfizer), a Tie-2 antagonist (Hybrigenics), a VEGFR-2 kinase inhibitor (Bristol-Myers Squibb), XL-647 (Exelixis), a KDR inhibitor from Abbott Laboratories, sorafenib tosylate, and an analogue or derivative thereof.
4369. The device of item 4336 wherein the agent is an endotoxin antagonist.
4370. The device of item 4336 wherein the agent is an epothilone and tubulin binder.
4371. The device of item 4336 wherein the agent is an estrogen receptor antagonist. 4372. The device of item 4336 wherein the agent is an FGF inhibitor.
4373. The device of item 4336 wherein the agent is a farnexyl transferase inhibitor.
4374. The device of item 4336 wherein the agent is famesyttransferase inhibitor selected from the group of A-197574 (Abbott), a farnesyltransferase inhibitor from Servier, FPTIII (Strathclyde Institute for Drug R), LB-42908 (LG Life Sciences), Pharmaprojects No. 5063 (Genzyme), Pharmaprojects No. 5597 (Ipsen), Yissum Project No. B-1055 (Yissum), and an analogue or derivative thereof.
4375. The device of item 4336 wherein the agent is an FLT-3 kinase inhibitor.
4376. The device of item 4336 wherein the agent is an FGF receptor kinase inhibitor.
4377. The device of item 4336 wherein the agent is a fibrinogen antagonist selected from the group consisting of AUV-201 (Auvation), MG-13926 (Sanofi-Aventis), plasminogen activator (CAS No. 105913-11-9) (from Sanofi-Aventis or UCB), plasminogen activator-2 (tPA- 2) (Sanofi-Aventis), pro-urokinase (CAS No. 82657-92-9) (Sanofi-Aventis), and an analogue or derivative thereof.
4378. The device of item 4336 wherein the agent is a heat shock protein 90 antagonist selected from the group consisting of SRN-005 (Sirenade), geldanamycin, NSC-33050 (17-allylaminogeldanamycin; 17- AAG), 17-dimethylaminoethylamino-17-demethoxy-geldanamycin (17- DMAG), rifabutin (rifamycin XIV, r,4~didehydro-1-deoxy-1,4-dihydro-5'-(2- methyipropyi)-1-oxo-), and an analogue or derivative thereof.
4379. The device of item 4336 wherein the agent is a histone deacetylase inhibitor.
4380. The device of item 4336 wherein the agent is an HMGCoA reductase inhibitor selected from the group consisting of an atherosclerosis therapeutic from Lipid Sciences, ATI-16000 (ARYx Therapeutics), KS-01-019 (Kos Pharmaceuticals), Pharmaprojects No. 2197 (Sanofi-Aventi), RP 61969 (Sanofi-Aventis), and an analogue or derivative thereof.
4381. The device of item 4336 wherein the agent is an ICAM inhibitor.
4382. The device of item 4336 wherein the agent is an IL, ICE and IRAK antagonist, wherein the antagonist is a CJ-14877, CP-424174 (Pfizer), NF-61 (Negma-Lerads), and an analogue or derivative thereof.
4383. The device of item 4336 wherein the agent is an IL-2 inhibitor.
4384. The device of item 4336 wherein the agent is an immunosuppressant selected from the group consisting of teriflunomide (Sanofi Aventis), chlorsulfaquinoxalone (NSC-339004), chlorsulfaquinoxalone sulfate, CS-712 (Sankyo), ismomultin alfa (CAS No. 457913-93-8) (Akzo Nobel), antiallergics from GenPat77, antiinflammatories or AT-005 (Androclus Therapeutics), autoimmune disease therapeutics from EpiVax, BN-007 (Bone), budesonide (CAS No. 51333-22- 3) (MAP Pharmaceuticals), CO-14 (Genzyme), edratide (CAS No. 433922- 67-9) (Teva), EP-314 (Enanta), eprovafen (CAS No. 101335-99-3) (Sanofi- Aventis), HWA-131 (CAS No. 118788-41-3) (Sanofi-Aventis), immunomodulators from MerLion Pharmaceuticals, immunosuppressives from Alchemia, IPL-12 (Inflazyme), MDL-9563 (CAS No. 27086-86-8) (Sanofi-Aventis), Pharmaprojects No. 2330 (Sanofi-Aventis), Pharmaprojects No. 6426 (Abgenix), PXS-25 (Pharmaxis), rosmarinic acid (CAS No. 20283-92-5) (Sanofi-Aventis), RP 42927 or RP 54745 (CAS No. 135330-08-4) (Sanofi-Aventis), SGN-35 (Seattle Genetics), ST-1959 (Sigma-Tau), type I diabetes therapy from SYNX Pharma, UNIL-88 (Debiopharm), VP-025 (Vasogen), VR-694 (Vectura), PRTX-001 (Protalex), and an analogue or derivative thereof.
4385. The device of item 4336 wherein the agent is an IMPDH (inosine monophosphate).
4386. The device of item 4336 wherein the agent is an integrin antagonist.
4387. The device of item 4336 wherein the agent is an interleukin antagonist.
4388. The device of item 4336 wherein the agent is an inhibitor of type ill receptor tyrosine kinase.
4389. The device of item 4336 wherein the agent is an irreversible inhibitor of enzyme methionine aminopeptidase type 2.
4390. The device of item 4336 wherein the agent is an isozyme selective delta protein kinase C inhibitor. 4391. The device of item 4336 wherein the agent a JAK3 enzyme inhibitor.
4392. The device of item 4336 wherein the agent is a JNK inhibitor.
4393. The device of item 4336 wherein the agent is a kinase inhibitor.
4394. The device of item 4336 wherein the agent is kinesin antagonist.
4395. The device of item 4336 wherein the agent is a kinesin antagonist.
4396. The device of item 4336_wherein the agent is a leukotriene inhibitor and antagonist selected from the group consisting of ambicromii (CAS No. 58805-38-2) (Sanofi-Aventis), amelubant (CAS No. 346735-24-8) (Boehringer Ingelheim), DW-1141 (Dong Wha), ebselen (Daiichi Pharmaceutical), ibudilast (Kyorin), leucotriene inhibitors from Sanofi-Aventis, lymphotoxin-beta receptor (LT-D) from Biogen Idee, Pharmaprojects No. 1535 and 2728 (CAS No. 119340-33-9) (Sanofi- Aventis), R-112 (Rigel), Rev-5367 (CAS No. 92532-05-3) (Sanofi-Aventis), RG-14893 (CAS No. 141835-49-6) (Sanofi-Aventis), RG-5901-A (CAS No. 101910-24-1), 92532-23-5, RP 66153 (CAS No. 142422-79-5), RP 66364 (CAS No.186916-92-5), RP 69698 (CAS No. 141748-00-7) (Sanofi-Aventis), SC-411930 (Pfizer), SC-41930 (CAS No. 120072-59-5) (Pfizer), SC-50605 (CAS No. 138828-39-4) (Pfizer), SC-51146 (CAS No. 141059-52-1), SC- 53228 (CAS No. 153633-01-3) (Pfizer), spaglumic acid (ZY-15106) (CAS No.3106-85-2), 80619-64-3 (Novartis), tipredane (CAS No. 85197-77-9) (Bristol-Myers Squibb), U-75302 (CAS No. 119477-85-9) (Pfizer), and analogue or derivative thereof.
4397. The device of item 4336 wherein the agent is an MAP kinase inhibitor.
4398. The device of item 4336 wherein the agent is a matrix metalloproteinase inhibitor.
4399. The device of item 4336 wherein the agent is an MCP- CCR2 inhibitor.
4400. The device of item 4336 wherein the agent is an mTOR inhibitor.
4401. The device of item 4336 wherein the agent is an mTOR kinase inhibitor.
4402. The device of item 4336 wherein the agent is a microtubule inhibitor selected from the group consisting of antibody- maytansinoid conjugates from Biogen Idee, colchicines (MantiCore Pharmaceuticals), anticancer immunoconjugates from Johnson & Johnson, DIME from Octamer, gni-1f (GNI), huC242-DM4, huMy9-6-DM1 (ImmunoGen), IDN-5404 (Indena), IMO-098, IMOderm (Imotep), mebendazole (Introgen Therapeutics), microtubule poisons from Cambridge Enterprise, paclitaxel such as LOTAX from Aphios (CAS No. 33069-62-4), Genexol-PM from Samyang, Pharmaprojects No. 6383 (Tapestry Pharmaceuticals), RPR-112378 (Sanofi-Aventis), SGN-75 (Seattle Genetics), SPL-7435 (Starpharma), SSR-250411 (Sanofi-Aventis), trastuzumab-DM1 (Genentech), vinorelbine, and an analogue or derivative thereof. 4403. The device of item 4336 wherein the agent is an MIF inhibitor.
4404. The device of item 4336 wherein the agent is an MMP inhibitor.
4405. The device of item 4336 wherein the agent is a neurokinin (NK) antagonist selected from the group consisting of anthrotainin (CAS No. 148084-40-6) (Sanofi-Aventis), an IBS thereapeutic from ArQuIe, MDL-105212A (CAS No. 167261-60-1) (Ssanofi-Aventis), Pharmaprojects No. 2744, 3258 (CAS No. 139167-47-8) 4006, 4201, or 5986 (Sanofi-Aventis), RP 67580 (CAS No. 135911-02-3), SR-144190 (CAS No. 201152-86-5), SSR-240600, SSR-241586 (Sanofi-Aventis), TKA-457 (Novartis), vestipitant mesylate (CAS No. 334476-64-1) (GlaxoSmithKline), Win-64821 (Sanofi-Aventis), PRX-96026 (Predix Pharmaceuticals), and an analogue or derivative thereof.
4406. The device of item 4336 wherein the agent is an NF kappa B inhibitor selected from the group consisting of AVE-0545 or AVE- 0547 (Sanofi-Aventis), bortezomib (CAS No. 179324-69-7) (Millennium Pharmaceuticals), dexanabinol (CAS No. 112924-45-5) (Pharmos), dexlipotam (Viatris), Pharmaprojects No. 6283 (INDRA) (OXiGENE), IPL- 576092 (CAS No. 137571-30-3) (Inflazyme), NFKB decoy (Corgentech), NFKB decoy oligo (AnGes MG), NFKB's from Ariad, osteoporosis treatments or S5 (F005) from Fulcrum Pharmaceuticals, P61 (Phytopharm), R-flurbiprofen (CAS No. 5104-49-4) (Encore Pharmaceuticals), and an analogue or derivative thereof.
4407. The device of item 4336 wherein the agent is a nitric oxide agonist. 4408. The device of item 4336 wherein the agent is an ornithine decarboxylase inhibitor.
4409. The device of item 4336 wherein the agent is a p38 MAP kinase inhibitor selected from the group consisting of AZD-6703 (AstraZeneca), JX-401 (Jexys Pharmaceuticals), BMS-2 (Bristol-Myers Squibb), a p38 MAP kinase inhibitor from Novartis, a p38-alpha MAP kinase inhibitor from Amphora, Pharmaprojects No. 5704 (Pharmacopeia), RPR- 200765A (Sanofi-Aventis), SD-282 (Johnson & Johnson), TAK-715 (Takeda), and an analogue or derivative thereof.
4410. The device of item 4336 wherein the agent is a palmitoyl-protein thioesterase inhibitor.
4411. The device of item 4336 wherein the agent is a PDGF receptor kinase inhibitor selected from the group consisting of AAL-993, AMN-107, or ABP-309 (Novartis), AMG-706 (Amgen), BAY-57-9352 (Bayer), CDP-860 (UCB), E-7080 (Eisai), imatinib (CAS No. 152459-95-5) (Novartis), OSI-930 (OSI Pharmaceuticals), RPR-127963E (Sanofi-Aventis), RWJ-540973 (Johnson & Johnson), sorafenib tosylate (Bayer), SU-11657 (Pfizer), tandutinib (CAS No. 387867-13-2) (Millennium Pharmaceuticals), vatalanib (Novartis), ZK-CDK (Schering AG), and an analogue or derivative thereof.
4412. The device of item 4336 wherein the agent is (-)- halofenate (Metabolex), AMG-131 (Amgen), antidiabetics from Japan Tobacco, AZD-4619, AZD-8450, AZD-8677 (AstraZeneca), DRF-10945, balaglitazone (Dr Reddy's), CS-00088, CS-00098 (Chipscreen Biosciences), E-3030 (Eisai), etalocib (CAS No. 161172-51-6) (EIi Lilly), GSK-641597 (Ligand), GSK-677954 (GlaxoSmithKline), GW-409544 (Ligand), GW- 590735 (GlaxoSmithKline), K-111 (Hoffmann-La Roche), LY-518674 (EIi Lilly), LY-674 (Ligand), LY-929 (Ligand), MC-3001 , MC-3002 (MaxoCore Pharmaceuticals), metformin HCI + pioglitazone (CAS No. 1115-70-4 and 112529-15-4), ACTOPLUS MET from Andrx), muraglitazar (CAS No. 331741-94-7) (Bristol-Myers Squibb), naveglitazar (Ligand), oleoylethanolamide (Kadmus Pharmaceuticals), ONO-5129, pioglitazone hydrochloride (CAS No. 111025-46-8 and 112529-15-4) (Takeda), PLX-204 (Piexxikon), PPAR agonists from Genfit, PPAR delta agonists from EIi Lilly, PPAR-alpha agonists from CrystalGenomics, PPAR-gamma modulators and PPAR-β modulators from CareX, rosiglitazone maleate (CAS No. 122324- 73-4 or 155141-29-0) (GlaxoSmithKline), rosiglitazone maleate/glimepir (CAS No. 155141-29-0 and 93479-97-1), AVANDARYL, rosiglitazone maleate/metformin extend (CAS No. 155141-29-0 and 657-24-9), AVANDAMET, rosiglitazone maleate+metformin, AVANDAMET (GlaxoSmithKline), tesaglitazar (AstraZeneca), and an analogue or derivative thereof.
4413. The device of item 4336 wherein the agent is a phosphatase inhibitor.
4414. The device of item 4336 wherein the agent is a phosphodiesterase (PDE) inhibitor selected from the group consisting of avanafil (Tanabe Seiyaku), dasantafil (CAS No. 569351-91-3) (Schering- Plough), A-906119 (CAS No. 134072-58-5), DL-850 (Sanofi-Aventis), GRC- 3015, GRC-3566, GRC-3886 (Glenmark), HWA-153 (CAS No. 56395-66-5) (Sanofi-Aventis), hydroxypumafentrine (Altana), IBFB-130011, IBFB-14-016, IBFB-140301, IBFB-150007, IBFB-211913 (IBFB Pharma), L-826141 (Merck & Co), medorinone (CAS No. 88296-61-1) (Sanofi-Aventis), MEM- 1917 (Memory Pharmaceuticals), ND-1251 (Neuro3d), PDE inhibitors from ICOS, PDE IV inhibitors from Memory Pharmaceuticals and CrystalGenomics, Pharmaprojects No. 2742 and 6141 (Sanofi-Aventis), QAD-171 (Novartis), RHC-2963 (CAS No. 76993-12-9 and 76993-14-1), RPR-117658, RPR-122818 derivatives, SR-24870 , and RPR-132294 (Sanofi-Aventis), SK-350 (ln2Gen), stroke therapy agents from deCODE Genetics, TAS-203 (Taiho), tofimilast (CAS No. 185954-27-2) (Pfizer), UK- 371800 (Pfizer), WIN-65579 (CAS No. 158020-82-7) (Sanofi-Aventis), IBFB- 130020 (IBFB Pharma), OPC-6535 (CAS No. 145739-56-6) (Otsuka), a phosphodiesterase III inhibitor, enoximone, a phosphodiesterase IV inhibitor, fosfosal, Atopik (Barrier Therapeutics), triflusal, a phosphodiesterase V inhibitor, and an analogue or derivative thereof.
4415. The device of item 4336 wherein the agent is a PKC inhibitor.
4416. The device of item 4336 wherein the agent is a platelet activating factor antagonist.
4417. The device of item 4336 wherein the agent is a platelet- derived growth factor receptor kinase inhibitor.
4418. The device of item 4336 wherein the agent is a prolyl hydroxylase inhibitor.
4419. The device of item 4336 wherein the agent is a polymorphonuclear neutrophil inhibitor.
4420. The device of item 4336 wherein the agent is a protein kinase B inhibitor.
4421. The device of item 4336 wherein the agent is a protein kinase C stimulant. 4422. The device of item 4336 wherein the agent is a purine nucleoside analogue.
4423. The device of item 4336 wherein the agent is a purinoreceptor P2X antagonist.
4424. The device of item 4336 wherein the agent is a Raf kinase inhibitor.
4425. The device of item 4336 wherein the agent is a reversible inhibitor of ErbB1 and ErbB2.
4426. The device of item 4336 wherein the agent is a ribonucleoside triphosphate reductase inhibitor.
4427. The device, of item,4336 wherein the agent is an SDF-1 antagonist.
4428. The device of item 4336 wherein the agent is a sheddase inhibitor.
4429. The device of item 4336 wherein the agent is an SRC inhibitor.
4430. The device of item 4336 wherein the agent is a stromelysin inhibitor.
4431. The device of item 4336 wherein the agent is an Syk kinase inhibitor. 4432. The device of item 4336 wherein the agent is a telomerase inhibitor.
4433. The device of item 4336 wherein the agent is a TGF beta inhibitor selected from the group consisting of pirfenidone (CAS No. 53179-13-8) (MARNAC), tranilast (CAS No. 53902-12-8) (Kissei), IN-1130 (ln2Gen), mannose-6-phosphate (BTG), TGF-β antagonists from Inflazyme (Pharmaprojects No. 6075), TGF-β antagonists from Sydney, non-industrial source), TGF-β I receptor kinase inhibitors from EIi Lilly, TGF-β receptor inhibitors from Johnson & Johnson, and an analogue or derivative thereof.
4434. The device of item 4336 wherein the agent is a TNFα antagonist or TACE inhibitor selected from the group consisting of adalimumab (CAS No. 331731-18-1) (Cambridge Antibody Technology), AGIX-4207 (AtheroGenics), AGT-1 (Advanced Biotherapy), an antiinflammatory from Borean Pharma, Cellzome, or Paradigm Therapeutics, anti-inflammatory vaccine (TNF-alpha kinoid) from Neovacs, humanized anti-TNF antibody or an anti-TNF MAb (CB0006) Celltech (UCB), apratastat (CAS No. 287445-51-0) (Wyeth), BMS-561392 (Bristol-Myers Squibb), BN- 006 (Bone), certolizumab pegol (CAS No. 428863-50-7 or CH-138 (UCB), cilomilast (CAS No. 153259-65-5) (GlaxoSmithKline), CR-1 (Nuada Pharmaceuticals), CRx-119 (CombinatoRx), D-5410 (UCB), dacopafant (CAS No. 125372-33-0) (Sanofi-Aventis), dersalazine (CAS No. 188913-57- 7/188913-58-8) (Uriach), etanercept (CAS No. 185243-69-0) (Amgen), ethyl pyruvate (Critical (Critical Therapeutics), golimumab (CAS No. 476181-74-5) (Johnson & Johnson), hormono-immunotherapy from Ipsen, CDP571 (e.g., Humicade from UCB), IC-485 (ICOS), infliximab (CAS No. 170277-31-3) (Johnson & Johnson), IP-751 (Manhattan Pharmaceuticals), ISIS-104838 (CAS No. 250755-32-9) (ISIS Pharmaceuticals), lenalidomide (CAS No. 191732-72-6) (Celgene), lentinan (CAS No. 37339-90-5) (Ajinomoto), MDL- 201112 (CAS No. 142130-73-2) (Sanofi-Aventis), medroxyprogesterone (CAS No. 520-85-4) (InKine Pharmaceutical), N-acetylcysteine (CAS No. 616-91-1) (Zambon), NBE-P2 (DIREVO Biotech), nerelimomab (CAS No. 162774-06-3) (Chiron), OM-294DP (OM PHARMA), onercept (CAS No. 199685-57-9) (Yeda), PASSTNF-alpha (Verigen), pentoxifylline or oxypentifylline (Sanofi-Aventis), Pharmaprojects No. 4091 , 4241, 4295, or 4488 (Sanofi-Aventis), Pharmaprojects No. 5480 (Amgen), Pharmaprojects No. 6749 (Cengent), pirfenidone (CAS No. 53179-13-8) (MARNAC), RPR- 132294 (Sanofi-Aventis), S5 (F002) (Fulcrum Pharmaceuticals), simvastatin (CAS No. 79902-63-9) (Merck & Co), STA-6292 (Synta Pharmaceuticals), tacrolimus (CAS No. 104987-11-3) (Fujisawa LifeCycle Pharma), talactoferrin alfa (CAS No. 308240-58-6) (Agennix), thalidomide (CAS No. 50-35-1) (Celgene), TNF antagonists form ProStrakan, and Synergen, TNF inhibitors (Amgen), TNF-alpha antagonists from Dynavax Technologies and Jerina AG (Germany), TNF-alpha inhibitors from IBFB Pharma and Xencor (Xencor), torbafylline (CAS No. 105102-21-4) (Sanofi-Aventis), UR-1505 (Uriach), VT-346 (Viron Therapeutics), YSIL6 (Y-1S Therapeutics), YSTH2 (Y's Therapeutics), NPI-1302a-3 (Nereus Pharmaceuticals, a TNF antagonist from Jerina AG (Germany), dersalazine, and an analogue or derivative thereof.
4435. The device of item 4336 wherein the agent is a Toll receptor inhibitor.
4436. The device of item 4336 wherein the agent is a tubulin antagonist.
4437. The device of item 4336 wherein the agent is a tyrosine kinase inhibitor selected from the group consisting of SU-011248, SUTENT from Pfizer Inc. (New York, NY), BMS-354825, PN-355 (Paracelsian Pharmaceuticals), AGN-199659 (Allergan), AAL-993 or ABP-309 (Novartis), adaphostin (NIH), AEE-788 (Novartis), AG-013736 (OSI Pharmaceuticals), AG-13736 (Pfizer), ALT-110 (Alteris Therapeutics), AMG-706 (Amgen), anticancer MAbs from Xencor, anti-EGFrvlll MAbs from Abgenix, anti-HER2 MAb from Abiogen, AZD-2171 or AZD-9935 (AstraZeneca), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), CEP-5214 (Cephalon), CEP- 7055 (Cephalon), cetuximab (ImCIone Systems), CHIR-200131 and CHIR- 258 (Chiron), CP-547632 (OSI Pharmaceuticals), CP-724714 (Pfizer), CT- 301 (Creabilis Therapeutics), D-69491 (Baxter International), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EGFR/ErbB2 inhibitors from Array BioPharma, erlotinib (CAS No. 183319-69-9) (OSI Pharmaceuticals), EXEL-2880 (Exelixis), FK-778 (Sanofi-Aventis) , gefitinib (CAS No. 184475-35-2) (AstraZeneca), GW-2286 or GW-654652 (GlaxoSmithKline), her2/neu antigen from AlphaVax, HER-2/neu inhibitor from Generex, Herzyme (Medipad) (Sirna Therapeutics), HKI-272 (Wyeth), HuMax-EGFr (Genmab), idronoxil (CAS No. 81267-65-4) (Novogen), IGF-1 inhibitors from Ontogen, IMC-11 F8 (ImCIone Systems), kahalalide F (CAS No. 149204-42-2) (PharmaMar), KDR inhibitor from LG Life Sciences, KDR inhibitors from Abbott Laboratories, KDR kinase inhibitors (UCB), Kdr kinase inhibitors from Merck & Co, KRN-633 and KRN-951 (Kirin Brewery), KSB-102 (Xenova), lapatinib ditosylate (CAS No. 388082-78-8) (GlaxoSmithKline), matuzumab (Merck KGaA), MDX-214 (Medarex), ME-103 (Pharmexa), MED-A300 (Gerolymatos), MNAC-13 (Lay Line Genomics), nimotuzumab (Center of Molecular Immunology), NSC-330507 or NSC-707545 (NIH), NV-50 (Novogen), OSI-930 (OSI Pharmaceuticals), panitumumab (Abgenix), pelitinib (CAS No. 287933-82-7) (Wyeth), pertuzumab (CAS No. 380610-27- 5) (Genentech), Pharmaprojects No. 3985 (Sanofi-Aventis), prostate cancer therapeutics from Sequenom (SQPC35, SQPC36, SQPC90), removab and remoxab (Trion Pharma), RG-13022 (CAS No. 136831-48-6), RG-13291 (CAS No. 138989-50-1), or RG-14620 (CAS No. 136831-49-7) (Sanofi- Aventis), RM-6427 (Romark), RNAi breast cancer therapy from Benitec, RP 53801 (CAS No. 125882-88-4) (Sanofi-Aventis), sorafenib tosylate (Bayer), SU-11657 (Pfizer), Tie-2 antagonists from Semaia (Hybrigenics), Tie-2 inhibitors from Ontogen, trastuzumab (CAS No. 180288-69-1) (Genentech), tyrosine kinase inhibitors from Sanofi-Aventis, U3-1287, U3-1565, U3-1784, U3-1800 (U3 Pharma), vatalanib (Novartis), VEGFR-2 kinase inhibitor from Bristol-Myers Squibb, XL-647 (Exelixis), ZD-6474 (AstraZeneca), ZK-CDK (Schering AG), an EGFR tyrosine kinase inhibitor, EKB-569 (Wyeth), and an analogue or derivative thereof.
4438. The device of item 4336 wherein the agent is a VEGF inhibitor.
4439. The device of item 4336 wherein the agent is a vitamin D receptor agonist.
4440. The device of item 4336 wherein the agent is ZD-6474 (an angiogenesis inhibitor).
4441. The device of item 4336 wherein the agent is AP-23573 (an mTOR inhibitor).
4442. The device of item 4336 wherein the agent is synthadotin (a tubulin antagonist).
4443. The device of item 4336 wherein the agent is S-0885 (a collagenase inhibitor).
4444. The device of item 4336 wherein the agent is aplidine (an elongation factor-1 alpha inhibitor).
4445. The device of item 4336 wherein the agent is ixabepilone (an epithilone). 4446. The device of item 4336 wherein the agent is IDN-5390 (an angiogenesis inhibitor).
4447. The device of item 4336 wherein the agent is SB- 2723005 (an angiogenesis inhibitor).
4448. The device of item 4336 wherein the agent is ABT-518 (an angiogenesis inhibitor).
4449. The device of item 4336 wherein the agent is combretastatin (an angiogenesis inhibitor).
4450. The device of item 4336 wherein the agent is anecortave acetate (an angiogenesis inhibitor).
4451. The device of item 4336 wherein the agent is SB- 715992 (a kinesin antagonist).
4452. The device of item 4336 wherein the agent is temsirolimus (an mTOR inhibitor).
4453. The device of item 4336 wherein the agent is adalimumab (a TNFα antagonist).
4454. The device of item 4336, further comprising a polymer.
4455. The device of item 4336 wherein the anti-scarring agent inhibits adhesion between the device and a host into which the device is implanted. 4456. The device of item 4336 wherein the device delivers the anti-scarring agent locally to tissue proximate to the device.
4457. The device of item 4336, further comprising a coating, wherein the coating comprises the anti-scarring agent.
4458. The device of item 4336, further comprising a coating, wherein the coating is disposed on a surface of the device.
4459. The device of item 4336, further comprising a coating, wherein the coating directly contacts the device.
4460. The device of item 4336, further comprising a coating, wherein the coating indirectly contacts the device.
4461. Jhe device of item 4336, further comprising a coating, wherein the coating partially covers the device.
4462. The device of item 4336, further comprising a coating, wherein the coating completely covers the device.
4463. The device of item 4336, further comprising a coating, wherein the coating is a uniform coating.
4464. The device of item 4336, further comprising a coating, wherein the coating is a non-uniform coating.
4465. The device of item 4336, further comprising a coating, wherein the coating is a discontinuous coating. 4466. The device of item 4336, further comprising a coating, wherein the coating is a patterned coating.
4467. The device of item 4336, further comprising a coating, wherein the coating has a thickness of 100 Dm or less.
4468. The device of item 4336, further comprising a coating, wherein the coating has a thickness of 10 Dm or less.
4469. The device of item 4336, further comprising a coating, wherein the coating adheres to the surface of the device upon deployment of the device.
4470. The device of item 4336, further comprising a coating, wherein the coating is stable at room temperature for a period of 1 year.
4471. The device of item 4336, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 0.0001% to about 1% by weight.
4472. The device of item 4336, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 1 % to about 10% by weight.
4473. The device of item 4336, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 10% to about 25% by weight.
4474. The device of item 4336, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 25% to about 70% by weight. 4475. The device of item 4336, further comprising a coating, wherein the coating further comprises a polymer.
4476. The device of item 4336, further comprising a first coating having a first composition and the second coating having a second composition.
4477. The device of item 4336, further comprising a first coating having a first composition and the second coating having a second composition, wherein the first composition and the second composition are different.
4478. The device of item 4336, further comprising a polymeric carrier, wherein the polymeric carrier comprises a copolymer.
4479. The device of item 4336, further comprising a polymeric carrier, wherein the polymeric carrier comprises a block copolymer.
4480. The device of item 4336, further comprising a polymeric carrier, wherein the polymeric carrier comprises a random copolymer.
4481. The device of item 4336, further comprising a polymeric carrier, wherein the polymeric carrier comprises a biodegradable polymer.
4482. The device of item 4336, further comprising a polymeric carrier, wherein the polymeric carrier comprises a non-biodegradable polymer.
4483. The device of item 4336, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrophilic polymer. 4484. The device of item 4336, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrophobic polymer.
4485. The device of item 4336, further comprising a polymeric carrier, wherein the polymeric carrier comprises a polymer having hydrophilic domains.
4486. The device of item 4336, further comprising a polymeric carrier, wherein the polymeric carrier comprises a polymer having hydrophobic domains.
4487. The device of item 4336, further comprising a polymeric carrier, wherein the polymeric carrier comprises a non-conductive polymer.
4488. The device of item 4336, further comprising a polymeric carrier, wherein the polymeric carrier comprises an elastomer.
4489. The device of item 4336, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrogel.
4490. The device of item 4336, further comprising a polymeric carrier, wherein the polymeric carrier comprises a silicone polymer.
4491. The device of item 4336, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrocarbon polymer.
4492. The device of item 4336, further comprising a polymeric carrier, wherein the polymeric carrier comprises a styrene-derived polymer.
4493. The device of item 4336, further comprising a polymeric carrier, wherein the polymeric carrier comprises a butadiene polymer. 4494. The device of item 4336, further comprising a polymeric carrier, wherein the polymeric carrier comprises a macromer.
4495. The device of item 4336, further comprising a polymeric carrier, wherein the polymeric carrier comprises a poly(ethylene glycol) polymer.
4496. The device of item 4336, further comprising a polymeric carrier, wherein the polymeric carrier comprises an amorphous polymer.
4497. The device of item 4336, further comprising a lubricious coating.
4498. The device of item 4336 wherein the anti-scarring agent is located within pores or holes of the device.
4499. The device of item 4336 wherein the anti-scarring agent is located within a channel, lumen, or divet of the device.
4500. The device of item 4336, further comprising a second pharmaceutically active agent.
4501. The device of item 4336, further comprising an antiinflammatory agent.
4502. The device of item 4336, further comprising an agent that inhibits infection.
4503. The device of item 4336, further comprising an agent that inhibits infection, wherein the agent is an anthracycline. 4504. The device of item 4336, further comprising an agent that inhibits infection, wherein the agent is doxorubicin.
4505. The device of item 4336, further comprising an agent that inhibits infection, wherein the agent is mitoxantrone.
4506. The device of item 4336, further comprising an agent that inhibits infection, wherein the agent is a fluoropyrimidine.
4507. The device of item 4336, further comprising an agent that inhibits infection, wherein the agent is 5-fluorouracil (5-FU).
4508. The device of item 4336, further comprising an agent that inhibits infection, wherein the agent is a folic acid antagonist.
4509. The device of item 4336, further comprising an agent that inhibits infection, wherein the agent is methotrexate.
4510. The device of item 4336, further comprising an agent that inhibits infection, wherein the agent is a podophylotoxin.
4511. The device of item 4336, further comprising an agent that inhibits infection, wherein the agent is etoposide.
4512. The device of item 4336, further comprising an agent that inhibits infection, wherein the agent is a camptothecin.
4513. The device of item 4336, further comprising an agent that inhibits infection, wherein the agent is a hydroxyurea. 4514. The device of item 4336, further comprising an agent that inhibits infection, wherein the agent is a platinum complex.
4515. The device of item 4336, further comprising an agent that inhibits infection, wherein the agent is cisplatin.
4516. The device of item 4336, further comprising an antithrombotic agent.
4517. The device of item 4336, further comprising a visualization agent.
4518. The device of item 4336, further comprising a visualization agent, wherein the visualization agent is a radiopaque material, wherein the radiopaque material comprises a metal, a halogenated compound, or a barium containing compound.
4519. The device of item 4336, further comprising a visualization agent, wherein the visualization agent is a radiopaque material, wherein the radiopaque material comprises barium, tantalum, or technetium.
4520. The device of item 4336, further comprising a visualization agent, wherein the visualization agent is a MRI responsive material.
4521. The device of item 4336, further comprising a visualization agent, wherein the visualization agent comprises a gadolinium chelate. 4522. The device of item 4336, further comprising a visualization agent, wherein the visualization agent comprises iron, magnesium, manganese, copper, or chromium.
4523. The device of item 4336, further comprising a visualization agent, wherein the visualization agent comprises an iron oxide compound.
4524. The device of item 4336, further comprising a visualization agent, wherein the visualization agent comprises a dye, pigment, or colorant.
4525. The device of item 4336, further comprising an echogenic material.
4526. The deyice.Of item 4336, further comprising an echogenic material, wherein the echogenic material is in the form of a coating.
4527. The device of item 4336 wherein the device is sterile.
4528. The device of item 4336 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device.
4529. The device of item 4336 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is connective tissue. 4530. The device of item 4336 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is muscle tissue.
4531. The device of item 4336 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is nerve tissue.
4532. The device of item 4336 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is epithelium tissue.
4533. The device of item 4336 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from the time of deployment of the device to about 1 year.
4534. The device of item 4336 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from about 1 month to 6 months.
4535. The device of item 4336 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from about 1 - 90 days.
4536. The device of item 4336 wherein the anti-scarring agent is released in effective concentrations from the device at a constant rate.
4537. The device of item 4336 wherein the anti-scarring agent is released in effective concentrations from the device at an increasing rate. 4538. The device of item 4336 wherein the anti-scarring agent is released in effective concentrations from the device at a decreasing rate.
4539. The device of item 4336 wherein the anti-scarring agent is released in effective concentrations from the composition comprising the anti-scarring agent by diffusion over a period ranging from the time of deployment of the device to about 90 days.
4540. The device of item 4336 wherein the anti-scarring agent is released in effective concentrations from the composition comprising the anti-scarring agent by erosion of the composition over a period ranging from the time of deployment of the device to about 90 days.
4541. The device of item 4336 wherein the device comprises about 0.01 Dg to about 10 Dg of the anti-scarring agent.
4542. The device of item 4336 wherein the device comprises about 10 Dg to about 10 mg of the anti-scarring agent.
4543. The device of item 4336 wherein the device comprises about 10 mg to about 250 mg of the anti-scarring agent.
4544. The device of item 4336 wherein the device comprises about 250 mg to about 1000 mg of the anti-scarring agent.
4545. The device of item 4336 wherein the device comprises about 1000 mg to about 2500 mg of the anti-scarring agent.
4546. The device of item 4336 wherein a surface of the device comprises less than 0.01 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied. 4547. The device of item 4336 wherein a surface of the device comprises about 0.01 Dg to about 1 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
4548. The device of item 4336 wherein a surface of the device comprises about 1 Dg to about 10 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
4549. The device of item 4336 wherein a surface of the device comprises about 10 Dg to about 250 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
4550. The device of item 4336 wherein a surface of the device comprises about 250 Dg to about 1000 Dg of the anti-scarring agent of anti- scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
4551. The device of item 4336 wherein a surface of the device comprises about 1000 Dg to about 2500 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
4552. A method as in any one of items 4336-4551, wherein the device is a drainage tube.
4553. A method as in any one of items 4336-4551, wherein the device is a feeding tube.
4554. A method as in any one of items 4336-4551, wherein the device is a portosystemic shunt.
4555. A method as in any one of items 4336-4551, wherein the device is a shunt for ascite. 4556. A method as in any one of items 4336-4551, wherein the device is a nasogastric or nasoenteral tube.
4557. A method as in any one of items 4336-4551, wherein the device is a gastrostomy or percutaneous feeding tube.
4558. A method as in any one of items 4336-4551, wherein the device is a jejunostomy endoscopic tube.
4559. A method as in any one of items 4336-4551 , wherein the device is a colostomy device.
4560. A method as in any one of items 4336-4551 , wherein the device is a biliary T-tube.
4561. A method as in any one of items 4336-4551 , wherein the device is a biopsy forceps.
4562. A method as in any one of items 4336-4551, wherein the device is a biliary stone removal device.
4563. A method as in any one of items 4336-4551 , wherein the device is an endoscopic retrograde cholangiopancretography device.
4564. A method as in any one of items 4336-4551, wherein the device is a dilation balloon.
4565. A method as in any one of items 4336-4551, wherein the device is an enteral feeding device.
4566. A method as in any one of items 4336-4551 , wherein the device is a stent.
4567. A method as in any one of items 4336-4551, wherein the device is a low profile device. 4568. A method as in any one of items 4336-4551 , wherein the device is a virtual colonoscopy device.
4569. A method as in any one of items 4336-4551 , wherein the device is a capsule endoscope.
4570. A method as in any one of items 4336-4551, wherein the device is a retrieval device.
4571. A method as in any one of items 4336-4551, wherein the device is a gastrointestinal device adapted for examining the interior of the gastrointestinal tract.
4572. A method as in any one of items 4336-4551 , wherein the device is a gastrointestinal device adapted for irrigation or aspiration of the gastrointestinal tract.
4573. A method as in any one of items 4336-4551 , wherein the device is a colostomy device.
4574. A method as in any one of items 4336-4551 , wherein the device is a mechanical hemostatic device adapted for control of gastrointestinal bleeding.
4575. A method as in any one of items 4336-4551, wherein the device is a gastrointestinal device adapted for cleaning blocked gastrointestinal tract.
4576. A method as in any one of items 4336-4551 , wherein the device is a gastrointestinal device for providing communication between two bodily systems.
4577. A method as in any one of items 4336-4551, wherein the device is a portosystemic shunt. 4578. A method as in any one of items 4336-4551 , wherein the device is a dilation catheter.
4579. A device, comprising a central venous catheter (e.g., an implant) and an anti-scarring agent or a composition comprising an anti- scarring agent, wherein the agent inhibits scarring between the device and a host into which the device is implanted.
4580. The device of item 4579 wherein the agent is an adensosine A2A receptor antagonist.
4581. The device of item 4579 wherein the agent is an AKT inhibitor.
4582. The device of item 4579 wherein the agent is an alpha 2 integrin antagonist, wherein the alpha 2 integrin antagonist is Pharmaprojects No. 5754 (Merck KgaA).
4583. The device of item 4579 wherein the agent an alpha 4 integrin antagonist.
4584. The device of item 4579 wherein the agent is an alpha 7 nicotinic receptor agonist.
4585. The device of item 4579 wherein the agent is an angiogenesis inhibitor selected from the group consisting of AG-12,958 (Pfizer), ATN-161 (Attenuon LLC), neovastat, an angiogenesis inhibitor from Jerina AG (Germany), NM-3 (Mercian), VGA-1155 (Taisho), FCE-26644 (Pfizer), FCE-26950 (Pfizer), FPMA (Meiji Daries), FR-111142 (Fujisawa), GGTI-298, GM-1306 (Ligand), GPA-1734 (Novartis), NNC-47-0011 (Novo Nordisk), herbamycin (Nippon Kayaku), lenalidomide (Celegene), IP-10 (NIH), ABT-828 (Abbott), KIN-841 (Tokushima University, Japan), SF-1126 (Semafore Pharmaceuticals), laminin technology (NIH), CHIR-258 (Chiron), NVP-AEW541 (Novartis), NVP-AEW541 (Novartis), Vt16907 (Alchemia), OXI-8007 (Oxigene), EG-3306 (Ark Therapeutics), Maspin (Arriva), ABT- 567 (Abbott), PPI-2458 (Praecis Pharmaceuticals), CC-5079, CC-4089 (Celgene), HIF-1 alpha inhibitors (Xenova), S-247 (Pfizer), AP-23573 (Ariad), AZD-9935 (Astra Zeneca), mebendazole (Introgen Therapeutics), MetAP-2 inhibitors (GlaxoSmithKline), AG-615 (Angiogene Pharmaceuticals), Tie-2 antagonists (Hybrigenics), NC-381 , CYC-381, NC-169, NC-219, NC-383, NC-384, NC-407 (Lorus Therapeutics), ATN-224 (Attenuon), ON-01370 (Onconova), Vitronectin antagonists (Amgen), SDX-103 (Salmedix), Vitronectin antagonists (Shire), CHP (Riemser), TEK (Amgen), Anecortave acetate (Alcon), T46.2 (Matrix Therapeutics), HG-2 (Heptagen), TEM antagonists (Genzyme), Oxi-4500 (Oxigene), ATN-161 (Attenuon), WX-293 (Wilex), M-2025 (Metris Therapeutics), Alphastatin (BioActa), YH-16 (Yantai Rongchang), BIBF-1120 (Boehringer Ingelheim), BAY-57-9352 (Bayer), AS- 1404 (Cancer Research Technology), SC-77964 (Pfizer), glycomimetics (BioTie Therapies), TIE-2 Inhibitors (Ontogen), DIMI, Octamer (Octamer), ABR-215050 (Active Biotech), ABT-518 (Abbott), KDR inhibitors (Abbott), BSF-466895 (Abbott), SCH-221153 (Schering-Plough), DAC:antiangiogenic (ConjuChem), TFPI (EntreMed), AZD-2171 (Astra-Zenaca), CDC-394 (Celgene), LY290293 (EIi Lilly), IDN-5390 (Indena), Kdr Kinase Inhibitors (Merck), CT-113020, CT-116433, CT-116563, CT-31890, CT-32228) (Cell Therapeutics), A-299620 (Abbott), TWEAK Inhibitor (Amgen), VEGF modulators (Johnson and Johnson), Tum-N53, tumstatin (Genzyme), Thios- 1 , Thios-2 (Thios Pharmaceuticals), MV-6401 (Miravant Medical Technologies), Spisulosine (PharmaMar), CEP-7055 (Cephalon), AUV-201 (Auvation), LM-609 (EIi Lilly), SKF-106615 (AnorMED), Oglufanide disodium (Cytran), BW-114 (Pharminox), Calreticulin (NIH), WX-678 (Wilex), SD-7784 (Pfizer), WX-UK1 (Wilex), SH-268 (Schering AG), 2-Me-PGA (Celgene), S- 137 (Pfizer), ZD-6126 (Angiogene Pharmaceuticals), SG-292 (SignalGen), Benefin (Lane Labs), A6, A36 (Angstrom), SB-2723005 (GlaxoSmithKline), SC-7 (Cell Therapeutics), ZEN-014 (AEterna Zentaris), 2-methoxyestradiol (EntreMed), NK-130119 (Nippon Kayaku), CC-10004 (Ceigene), AVE- 8062A (Ajinomoto), Tacedinaline (Pfizer), Actinonin (Tokyo Metropolitan Institute of Medical Science), Lenalidomide (Ceigene), VGA-1155, BTO-956 (SRI International), ER-68203-00 (Eisai), CT-6685 (UCB), JKC-362 (Phoenix Pharmaceuticals), DMI-3798 (DMI Biosciences, Angiomate (Ipsen), ZD-6474 (AstraZeneca), CEP-5214 (Cephalon), Canstatin (Genzyme), NM-3 (Mercian), Oridigm (MediQuest Therapeutics), Exherin (Adherex), BLS-0597 (Boston Life Sciences), PTC-299 (PTC Therapeutics), NPI-2358 (Nereus Pharmaceuticals), CGP-79787 (Novartis), AEE-788 (Novartis), CKD-732 (Chong Kun Dang), CP-564959 (OSI Pharmaceuticals), CM-101 (CarboMed), CT-2584, CT3501 (Cell Therapeutics), combretastatin and analogues and derivatives thereof (Oxigene), Rebimastat (Bristol-Meyers Squibb), Dextrin 2-sulfate (ML Laboratories), Cilengitide (Merk KGaA), NSC-706704 (Pharminox), KRN- 951 (Kirin Brewery), Ukrain,_NSC-631570 (Nowicky Pharma), Tecogalan sodium (Daiichi Pharmaceutical), Tz-93 (Tsumura), TBC-1635 (Encysive Pharmaceuticals), TAN-1120 (Takeda), Semaxanib (Pfizer), BDI-7800 (Biopharmacopae), SD-186, SD-983 (Bristol-Meyers Squibb), SB-223245 (GlaxoSmithKline), SC-236 (Pfizer), RWJ-590973 (Johnson and Johnson), ILX-1850 (Genzyme), SC-68488, S-836 (Pfizer), CG-55069-11 (CuraGen), Ki-23057 (Kirin Brewery), CCX-700 (Chemoentryx), Pegaptanib octasodium (Giled Sciences), ANGIOCOL (available from Biostratum Inc.), or an analogue or derivative thereof.
4586. The device of item 4579 wherein the agent is an apoptosis antagonist.
4587. The device of item 4579 wherein the agent is an apoptosis activator. 4588. The device of item 4579 wherein the agent is a beta 1 integrin antagonist.
4589. The device of item 4579 wherein the agent is a beta tubulin inhibitor.
4590. The device of item 4579 wherein the agent is a blocker of enzyme production in Hepatitis C.
4591. The device of item 4579 wherein the agent is a Bruton's tyrosine kinase inhibitor.
4592. The device of item 4579 wherein the agent is a calcineurin inhibitor.
4593. The. device of.item 4579 wherein the agent is a caspase 3 inhibitor.
4594. The device of item 4579 wherein the agent is a CC chemokine receptor antagonist.
4595. The device of item 4579 wherein the agent is a cell cycle inhibitor selected from the group consisting of SNS-595 (Sunesis), synthadotin, KRX-0403, and an analogue or derivative thereof.
4596. The device of item 4579 wherein the agent is a cathepsin B inhibitor.
4597. The device of item 4579 wherein the agent is a cathepsin K inhibitor, wherein the cathepsin K inhibitor is 462795 B Λ_a O 2006/135479
(GlaxoSmithKline), INPL-022-D6 (Amura Therapeutics), an analogue or derivative thereof).
4598. The device of item 4579 wherein the agent is a cathepsin L inhibitor.
4599. The device of item 4579 wherein the agent is a CD40 antagonist.
4600. The device of item 4579 wherein the agent is a chemokine receptor agonist.
4601. The device of item 4579 wherein the agent is a chymase inhibitor.
4602,_ The device of item 4579 wherein the agent is a coilagenase antagonist.
4603. The device of item 4579 wherein the agent is a CXCR antagonist.
4604. The device of item 4579 wherein the agent is a cyclin dependent kinase inhibitor selected from the group consisting of a CDK-1 inhibitor, a CDK-2 inhibitor, a CDK- 4 inhibitor, a CDK-6 inhibitor, a CAK1 inhibitor from GPC Biotech and Bristol-Myers Squibb, RGB-286199 (GPC Biotech), an anticancer agent from Astex Technology, a CAK1 inhibitor from GPC Biotech, a CDK inhibitor from Sanofi-Aventis, a CDK1/CDK2 inhibitor from Amgen, a CDK2 inhibitor from SUGEN-2 (Pfizer), a hearing loss therapy agent (Sound Pharmaceuticals), PD-0332991 (Pfizer), RGB-286199 (GPC Biotech), Ro-0505124 (Hoffmann-La Roche), a Ser/Thr kinase inhibitor from Lilly (EIi Lilly), CVT-2584 (CAS No. 199986-75-9) (CV Therapeutics), and an analogue or derivative thereof.
4605. The device of item 4579 wherein the agent is a cyclooxygenase 1 inhibitor.
4606. The device of item 4579 wherein the agent is a DHFR inhibitor.
4607. The device of item 4579 wherein the agent is a dual integrin inhibitor.
4608. The device of item 4579 wherein the agent is an elastase inhibitor.
4609. The device of item 4579 wherein the agent is an elongation factor-1 alpha inhibitor.
4610. The device of item 4579 wherein the agent is an endothelial growth factor antagonist.
4611. The device of item 4579 wherein the agent is an endothelial growth factor receptor kinase inhibitor selected from the group consisting of sorafenib tosylate (Bayer), AAL-993 (Novartis), ABP-309 (Novartis), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), E- 7080 (Eisai), EG-3306 (Ark Therapeutics), EXEL-2880 (Exelixis), GW- 654652 (GlaxoSmithKline), a KDR inhibitor from LG Life Sciences, CT-6685 and CT-6729 (UCB), KRN-633 and KRN-951 (Kirin Brewery), OSI-930 (OSI Pharmaceuticals), SP-5.2 (Supratek Pharma), SU-11657 (Pfizer), a Tie-2 antagonist (Hybrigenics), a VEGFR-2 kinase inhibitor (Bristol-Myers Squibb), XL-647 (Exelixis), a KDR inhibitor from Abbott Laboratories, sorafenib tosylate, and an analogue or derivative thereof.
4612. The device of item 4579 wherein the agent is an endotoxin antagonist.
4613. The device of item 4579 wherein the agent is an epothilone and tubulin binder.
4614. The device of item 4579 wherein the agent is an estrogen receptor antagonist.
4615. The device of item 4579 wherein the agent is an FGF inhibitor.
4616. The device of item 4579 wherein the agent is a farnexyl transferase inhibitor.
4617. The device of item 4579 wherein the agent is famesyltransferase inhibitor selected from the group of A-197574 (Abbott), a farnesyltransferase inhibitor from Servier, FPTIII (Strathclyde Institute for Drug R), LB-42908 (LG Life Sciences), Pharmaprojects No. 5063 (Genzyme), Pharmaprojects No. 5597 (Ipsen), Yissum Project No. B-1055 (Yissum), and an analogue or derivative thereof.
4618. The device of item 4579 wherein the agent is an FLT-3 kinase inhibitor.
4619. The device of item 4579 wherein the agent is an FGF receptor kinase inhibitor. 4620. The device of item 4579 wherein the agent is a fibrinogen antagonist selected from the group consisting of AUV-201 (Auvation), MG-13926 (Sanofi-Aventis), plasminogen activator (CAS No. 105913-11-9) (from Sanofi-Aventis or UCB), plasminogen activator-2 (tPA- 2) (Sanofi-Aventis), pro-urokinase (CAS No. 82657-92-9) (Sanofi-Aventis), and an analogue or derivative thereof.
4621. The device of item 4579 wherein the agent is a heat shock protein 90 antagonist selected from the group consisting of SRN-005 (Sirenade), geldanamycin, NSC-33050 (17-allylaminogeldanamycin; 17- AAG), 17-dimethylaminoethylamino-17-demethoxy-geldanamycin (17- DMAG), rifabutin (rifamycin XIV, I'^-didehydro-i-deoxy-i^-dihydro-δ'-^- methylpropyl)-1-oxo-), and an analogue or derivative thereof.
4622. The device of item 4579 wherein the agent is a histone deacetylase inhibitor. . _ . . _ -
4623. The device of item 4579 wherein the agent is an HMGCoA reductase inhibitor selected from the group consisting of an atherosclerosis therapeutic from Lipid Sciences, ATI-16000 (ARYx Therapeutics), KS-01-019 (Kos Pharmaceuticals), Pharmaprojects No. 2197 (Sanofi-Aventi), RP 61969 (Sanofi-Aventis), and an analogue or derivative thereof.
4624. The device of item 4579 wherein the agent is an ICAM inhibitor.
4625. The device of item 4579 wherein the agent is an IL, ICE and IRAK antagonist, wherein the antagonist is a CJ-14877, CP-424174 (Pfizer), NF-61 (Negma-Lerads), and an analogue or derivative thereof. 4626. The device of item 4579 wherein the agent is an IL-2 inhibitor.
4627. The device of item 4579 wherein the agent is an immunosuppressant selected from the group consisting of teriflunomide (Sanofi Aventis), chlorsulfaquinoxalone (NSC-339004), chlorsulfaquinoxalone sulfate, CS-712 (Sankyo), ismomultin alfa (CAS No. 457913-93-8) (Akzo Nobel), antiallergics from GenPat77, antiinflammatories or AT-005 (Androclus Therapeutics), autoimmune disease therapeutics from EpiVax, BN-007 (Bone), budesonide (CAS No. 51333-22- 3) (MAP Pharmaceuticals), CO-14 (Genzyme), edratide (CAS No. 433922- 67-9) (Teva), EP-314 (Enanta), eprovafen (CAS No. 101335-99-3) (Sanofi- Aventis), HWA-131 (CAS No. 118788-41-3) (Sanofi-Aventis), immunomodulators from MerLion Pharmaceuticals, immunosuppressives from Alchemia, IPL-12 (Inflazyme), MDL-9563 (CAS No. 27086-86-8) (SanofirAventia), Pharmaprojects No. 2330 (Sanofi-Aventis), - - - - Pharmaprojects No. 6426 (Abgenix), PXS-25 (Pharmaxis), rosmarinic acid (CAS No. 20283-92-5) (Sanofi-Aventis), RP 42927 or RP 54745 (CAS No. 135330-08-4) (Sanofi-Aventis), SGN-35 (Seattle Genetics), ST-1959 (Sigma-Tau), type I diabetes therapy from SYNX Pharma, UNIL-88 (Debiopharm), VP-025 (Vasogen), VR-694 (Vectura), PRTX-001 (Protalex), and an analogue or derivative thereof.
4628. The device of item 4579 wherein the agent is an IMPDH (inosine monophosphate).
4629. The device of item 4579 wherein the agent is an integrin antagonist.
4630. The device of item 4579 wherein the agent is an interleukin antagonist. 4631. The device of item 4579 wherein the agent is an inhibitor of type III receptor tyrosine kinase.
4632. The device of item 4579 wherein the agent is an irreversible inhibitor of enzyme methionine aminopeptidase type 2.
4633. The device of item 4579 wherein the agent is an isozyme selective delta protein kinase C inhibitor.
4634. The device of item 4579 wherein the agent a JAK3 enzyme inhibitor.
4635. The device of item 4579 wherein the agent is a JNK inhibitor.
4636. The device of item 4579 wherein the agent is a kinase inhibitor.
4637. The device of item 4579 wherein the agent is kinesin antagonist.
4638. The device of item 4579 wherein the agent is a kinesin antagonist.
4639. The device of item 4579 wherein the agent is a leukotriene inhibitor and antagonist selected from the group consisting of ambicromil (CAS No. 58805-38-2) (Sanofi-Aventis), amelubant (CAS No. 346735-24-8) (Boehringer Ingelheim), DW-1141 (Dong Wha), ebselen (Daiichi Pharmaceutical), ibudilast (Kyorin), leucotriene inhibitors from Sanofi-Aventis, lymphotoxin-beta receptor (LT-D) from Biogen Idee, Pharmaprojects No. 1535 and 2728 (CAS No. 119340-33-9) (Sanofi- Aventis), R-112 (Rigel), Rev-5367 (CAS No. 92532-05-3) (Sanofi-Aventis), RG-14893 (CAS No. 141835-49-6) (Sanofi-Aventis), RG-5901-A (CAS No. 101910-24-1), 92532-23-5, RP 66153 (CAS No. 142422-79-5), RP 66364 (CAS No.186916-92-5), RP 69698 (CAS No. 141748-00-7) (Sanofi-Aventis), SC-411930 (Pfizer), SC-41930 (CAS No. 120072-59-5) (Pfizer), SC-50605 (CAS No. 138828-39-4) (Pfizer), SC-51146 (CAS No. 141059-52-1), SC- 53228 (CAS No. 153633-01-3) (Pfizer), spaglumic acid (ZY-15106) (CAS No.3106-85-2), 80619-64-3 (Novartis), tipredane (CAS No. 85197-77-9) (Bristol-Myers Squibb), U-75302 (CAS No. 119477-85-9) (Pfizer), and analogue or derivative thereof.
4640. The device of item 4579 wherein the agent is an MAP kinase inhibitor.
4641. The device of item 4579 wherein the agent is a matrix metalloproteinase inhibitor.,
4642. The device of item 4579 wherein the agent is an MCP- CCR2 inhibitor.
4643. The device of item 4579 wherein the agent is an mTOR inhibitor.
4644. The device of item 4579 wherein the agent is an mTOR kinase inhibitor.
4645. The device of item 4579 wherein the agent is a microtubule inhibitor selected from the group consisting of antibody- maytansinoid conjugates from Biogen Idee, colchicines (MantiCore Pharmaceuticals), anticancer immunoconjugates from Johnson & Johnson, DIME from Octamer, gni-1f (GNI), huC242-DM4, huMy9-6-DM1 (ImmunoGen), IDN-5404 (Indena), IMO-098, IMOderm (Imotep), mebendazole (Introgen Therapeutics), microtubule poisons from Cambridge Enterprise, paclitaxel such as LOTAX from Aphios (CAS No. 33069-62-4), Genexol-PM from Samyang, Pharmaprojects No. 6383 (Tapestry Pharmaceuticals), RPR-112378 (Sanofi-Aventis), SGN-75 (Seattle Genetics), SPL-7435 (Starpharma), SSR-250411 (Sanofi-Aventis), trastuzumab-DM1 (Genentech), vinorelbine, and an analogue or derivative thereof.
4646. The device of item 4579 wherein the agent is an MIF inhibitor.
4647. The device of item 4579 wherein the agent is an MMP inhibitor.
4648. The device of item 4579 wherein the agent is a neurokinin (NK) antagonist selected from the group consisting of anthrotainin (CAS No. 148084-40-6) (Sanofi-Aventis), an IBS thereapeutic from ArQuIe, MDL-105212A (CAS No. 167261-60-1) (Ssanofi-Aventis), Pharmaprojects No. 2744, 3258 (CAS No. 139167-47-8) 4006, 4201, or 5986 (Sanofi-Aventis), RP 67580 (CAS No. 135911-02-3), SR-144190 (CAS No. 201152-86-5), SSR-240600, SSR-241586 (Sanofi-Aventis), TKA-457 (Novartis), vestipitant mesylate (CAS No. 334476-64-1) (GlaxoSmithKline), Win-64821 (Sanofi-Aventis), PRX-96026 (Predix Pharmaceuticals), and an analogue or derivative thereof.
4649. The device of item 4579 wherein the agent is an NF kappa B inhibitor selected from the group consisting of AVE-0545 or AVE- 0547 (Sanofi-Aventis), bortezomib (CAS No. 179324-69-7) (Millennium Pharmaceuticals), dexanabinol (CAS No. 112924-45-5) (Pharmos), dexlipotam (Viatris), Pharmaprojects No. 6283 (INDRA) (OXiGENE), IPL- 576092 (CAS No. 137571-30-3) (Inflazyme), NFKB decoy (Corgentech), NFKB decoy oligo (AnGes MG), NFKB's from Ariad, osteoporosis treatments or S5 (F005) from Fulcrum Pharmaceuticals, P61 (Phytopharm), R-flurbiprofen (CAS No. 5104-49-4) (Encore Pharmaceuticals), and an analogue or derivative thereof.
4650. The device of item 4579 wherein the agent is a nitric oxide agonist.
4651. The device of item 4579 wherein the agent is an ornithine decarboxylase inhibitor.
4652. The device of item 4579 wherein the agent is a p38 MAP kinase inhibitor selected from the group consisting of AZD-6703 (AstraZeneca), JX-401 (Jexys Pharmaceuticals), BMS-2 (Bristol-Myers Squibb), a p38 MAP kinase inhibitorfrom Novartis, a p38-alpha MAP kinase inhibitor from Amphora, Pharmaprojects No. 5704 (Pharmacopeia), RPR- 200765A (Sanofi-Aventis), SD-282 (Johnson & Johnson), TAK-715 (Takeda), and an analogue or derivative thereof.
4653. The device of item 4579 wherein the agent is a palmitoyl-protein thioesterase inhibitor.
4654. The device of item 4579 wherein the agent is a PDGF receptor kinase inhibitor selected from the group consisting of AAL-993, AMN-107, or ABP-309 (Novartis), AMG-706 (Amgen), BAY-57-9352 (Bayer), CDP-860 (UCB), E-7080 (Eisai), imatinib (CAS No. 152459-95-5) (Novartis), OSI-930 (OSI Pharmaceuticals), RPR-127963E (Sanofi-Aventis), RWJ-540973 (Johnson & Johnson), sorafenib tosylate (Bayer), SU-11657 (Pfizer), tandutinib (CAS No. 387867-13-2) (Millennium Pharmaceuticals), vatatanib (Novartis), ZK-CDK (Schering AG), and an analogue or derivative thereof.
4655. The device of item 4579 wherein the agent is (-)- halofenate (Metabolex), AMG-131 (Amgen), antidiabetics from Japan Tobacco, AZD-4619, AZD-8450, AZD-8677 (AstraZeneca), DRF-10945, balaglitazone (Dr Reddy's), CS-00088, CS-00098 (Chipscreen Biosciences), E-3030 (Eisai), etalocib (CAS No. 161172-51-6) (EIi Lilly), GSK-641597 (Ligand), GSK-677954 (GlaxoSmithKline), GW-409544 (Ligand), GW- 590735 (GlaxoSmithKline), K-111 (Hoffmann-La Roche), LY-518674 (EIi Lilly), LY-674 (Ligand), LY-929 (Ligand), MC-3001 , MC-3002 (MaxoCore Pharmaceuticals), metformin HCI + pioglitazone (CAS No. 1115-70-4 and
112529-15-4), ACTOPLUS MET from Andrx), muraglitazar (CAS No. 331741-94-7) (Bristol-Myers Squibb), naveglitazar (Ligand), oleoylethanolamide (Kadmus Pharmaceuticals), ONO-5129, pioglitazone hydrochloride (CAS No. 111025-46-8 and 112529-15-4) (Takeda), PLX-204 (Plexxikon), PPAR agonists from Genfit, PPAR delta agonists from EIi Lilly, PPAR-alpha agonists from CrystalGenomics, PPAR-gamma modulators and PPAR-β modulators from CareX, rosiglitazone maleate (CAS No. 122324- 73-4 or 155141-29-0) (GlaxoSmithKline), rosiglitazone maleate/glimepir (CAS No. 155141-29-0 and 93479-97-1), AVANDARYL, rosiglitazone maleate/metformin extend (CAS No. 155141-29-0 and 657-24-9), AVANDAMET1 rosiglitazone maleate+metformin, AVANDAMET (GlaxoSmithKline), tesaglitazar (AstraZeneca), and an analogue or derivative thereof.
4656. The device of item 4579 wherein the agent is a phosphatase inhibitor.
4657. The device of item 4579 wherein the agent is a phosphodiesterase (PDE) inhibitor selected from the group consisting of avanafil (Tanabe Seiyaku), dasantafil (CAS No. 569351-91-3) (Schering- Plough), A-906119 (CAS No. 134072-58-5), DL-850 (Sanofi-Aventis), GRC- 3015, GRC-3566, GRC-3886 (Glenmark), HWA-153 (CAS No. 56395-66-5) (Sanofi-Aventis), hydroxypumafentrine (Altana), IBFB-130011 , IBFB-14-016, IBFB-140301, IBFB-150007, IBFB-211913 (IBFB Pharma), L-826141 (Merck & Co), medorinone (CAS No. 88296-61-1) (Sanofi-Aventis), MEM- 1917 (Memory Pharmaceuticals), ND-1251 (Neuro3d), PDE inhibitors from ICOS, PDE IV inhibitors from Memory Pharmaceuticals and CrystalGenomics, Pharmaprojects No. 2742 and 6141 (Sanofi-Aventis), QAD-171 (Novartis), RHC-2963 (CAS No. 76993-12-9 and 76993-14-1), RPR-117658, RPR-122818 derivatives, SR-24870 , and RPR-132294 (Sanofi-Aventis), SK-350 (ln2Gen), stroke therapy agents from deCODE Genetics, TAS-203 (Taiho), tofimilast (CAS No. 185954-27-2) (Pfizer), UK- 371800 (Pfizer), WIN-65579 (CAS No. 158020-82-7) (Sanofi-Aventis), IBFB- 130020 (IBFB Pharma), OPC-6535 (CAS No. 145739-56-6) (Otsuka), a phosphodiesterasejll inhibitor, enoximone, a phosphodiesterase IV inhibitor, fosfosal, Atopik (Barrier Therapeutics), triflusal, a phosphodiesterase V inhibitor, and an analogue or derivative thereof.
4658. The device of item 4579 wherein the agent is a PKC inhibitor.
4659. The device of item 4579 wherein the agent is a platelet activating factor antagonist.
4660. The device of item 4579 wherein the agent is a platelet- derived growth factor receptor kinase inhibitor.
4661. The device of item 4579 wherein the agent is a prolyl hydroxylase inhibitor. 4662. The device of item 4579 wherein the agent is a polymorphonuclear neutrophil inhibitor.
4663. The device of item 4579 wherein the agent is a protein kinase B inhibitor.
4664. The device of item 4579 wherein the agent is a protein kinase C stimulant.
4665. The device of item 4579 wherein the agent is a purine nucleoside analogue.
4666. The device of item 4579 wherein the agent is a purinoreceptor P2X antagonist.
4667- The device^of item 4579 wherein the agent is a Raf kinase inhibitor.
4668. The device of item 4579 wherein the agent is a reversible inhibitor of ErbB1 and ErbB2.
4669. The device of item 4579 wherein the agent is a ribonucleoside triphosphate reductase inhibitor.
4670. The device of item 4579 wherein the agent is an SDF-1 antagonist.
4671. The device of item 4579 wherein the agent is a sheddase inhibitor. 4672. The device of item 4579 wherein the agent is an SRC inhibitor.
4673. The device of item 4579 wherein the agent is a stromelysin inhibitor.
4674. The device of item 4579 wherein the agent is an Syk kinase inhibitor.
4675. The device of item 4579 wherein the agent is a telomerase inhibitor.
4676. The device of item 4579 wherein the agent is a TGF beta inhibitor selected from the group consisting of pirfenidone (CAS No. 53179-13-8) (MARNAC), tranilast (CAS No. 53902-12-8) (Kissei), IN-1130 (ln2Gen), mannose-6-phosphate (BTG), TGF-β antagonists from inflazyme (Pharmaprojects No. 6075), TGF-β antagonists from Sydney, non-industrial source), TGF-β I receptor kinase inhibitors from EIi Lilly, TGF-β receptor inhibitors from Johnson & Johnson, and an analogue or derivative thereof.
4677. The device of item 4579 wherein the agent is a TNFα antagonist or TACE inhibitor selected from the group consisting of adalimumab (CAS No. 331731-18-1) (Cambridge Antibody Technology), AGIX-4207 (AtheroGenics), AGT-1 (Advanced Biotherapy), an antiinflammatory from Borean Pharma, Cellzome, or Paradigm Therapeutics, anti-inflammatory vaccine (TNF-alpha kinoid) from Neovacs, humanized anti-TNF antibody or an anti-TNF MAb (CB0006) Celltech (UCB), apratastat (CAS No. 287445-51-0) (Wyeth), BMS-561392 (Bristol-Myers Squibb), BN- 006 (Bone), certolizumab pegol (CAS No. 428863-50-7 or CH-138 (UCB), cilomilast (CAS No. 153259-65-5) (GlaxoSmithKline), CR-1 (Nuada Pharmaceuticals), CRx-119 (CombinatoRx), D-5410 (UCB), dacopafant (CAS No. 125372-33-0) (Sanofi-Aventis), dersalazine (CAS No. 188913-57- 7/188913-58-8) (Uriach), etanercept (CAS No. 185243-69-0) (Amgen), ethyl pyruvate (Critical (Critical Therapeutics), golimumab (CAS No. 476181-74-5) (Johnson & Johnson), hormono-immunotherapy from Ipsen, CDP571 (e.g., Humicade from UCB), IC-485 (ICOS), infliximab (CAS No. 170277-31-3) (Johnson & Johnson), IP-751 (Manhattan Pharmaceuticals), ISIS-104838 (CAS No. 250755-32-9) (ISIS Pharmaceuticals), lenalidomide (CAS No. 191732-72-6) (Celgene), lentinan (CAS No. 37339-90-5) (Ajinomoto), MDL- 201112 (CAS No. 142130-73-2) (Sanofi-Aventis), medroxyprogesterone (CAS No. 520-85-4) (InKine Pharmaceutical), N-acetylcysteine (CAS No. 616-91-1) (Zambon), NBE-P2 (DIREVO Biotech), nerelimomab (CAS No. 162774-06-3) (Chiron), OM-294DP (OM PHARMA), onercept (CAS No. 199685-57-9) (Yeda), PASSTNF-alpha (Verigen), pentoxifylline or oxypentifylline (Sanofi-Aventis), Pharmaprojects No. 4091 , 4241 , 4295, or 4488 (Sanofi-Aventis), Pharmaprojects No. 5480 (Amgen), Pharmaprojects Noτ-6749 (Cengent), pirfenidone (CAS No. 53179-13-8) (MARNAC), RPR- 132294 (Sanofi-Aventis), S5 (F002) (Fulcrum Pharmaceuticals), simvastatin (CAS No. 79902-63-9) (Merck & Co), STA-6292 (Synta Pharmaceuticals), tacrolimus (CAS No. 104987-11-3) (Fujisawa LifeCycle Pharma), talactoferrin alfa (CAS No. 308240-58-6) (Agennix), thalidomide (CAS No. 50-35-1) (Celgene), TNF antagonists form ProStrakan, and Synergen, TNF inhibitors (Amgen), TNF-alpha antagonists from Dynavax Technologies and Jerina AG (Germany), TNF-alpha inhibitors from IBFB Pharma and Xencor (Xencor), torbafylline (CAS No. 105102-21-4) (Sanofi-Aventis), UR-1505 (Uriach), VT-346 (Viron Therapeutics), YSIL6 (Y's Therapeutics), YSTH2 (Y's Therapeutics), NPI-1302a-3 (Nereus Pharmaceuticals, a TNF antagonist from Jerina AG (Germany), dersalazine, and an analogue or derivative thereof.
4678. The device of item 4579 wherein the agent is a Toll receptor inhibitor. 4679. The device of item 4579 wherein the agent is a tubulin antagonist.
4680. The device of item 4579 wherein the agent is a tyrosine kinase inhibitor selected from the group consisting of SU-011248, SUTENT from Pfizer Inc. (New York, NY), BMS-354825, PN-355 (Paracelsian Pharmaceuticals), AGN-199659 (Allergan), AAL-993 or ABP-309 (Novartis), adaphostin (NIH), AEE-788 (Novartis), AG-013736 (OSI Pharmaceuticals), AG-13736 (Pfizer), ALT-110 (Alteris Therapeutics), AMG-706 (Amgen), anticancer MAbs from Xencor, anti-EGFrvlll MAbs from Abgenix, anti-HER2 MAb from Abiogen, AZD-2171 or AZD-9935 (AstraZeneca), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), CEP-5214 (Cephalon), CEP- 7055 (Cephalon), cetuximab (ImClone Systems), CHIR-200131 and CHIR- 258 (Chiron), CP-547632 (OSI Pharmaceuticals), CP-724714 (Pfizer), CT- 301 (Creabilis Therapeutics), D-69491 (Baxter International), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EGFR/ErbB2 inhibitors from Array BioPharma, erlotinib (CAS No. 183319-69-9) (OSI Pharmaceuticals), EXEL-2880 (Exelixis), FK-778 (Sanofi-Aventis) , gefitinib (CAS No. 184475-35-2) (AstraZeneca), GW-2286 or GW-654652 (GlaxoSmithKline), her2/neu antigen from AlphaVax, HER-2/neu inhibitor from Generex, Herzyme (Medipad) (Sirna Therapeutics), HKI-272 (Wyeth), HuMax-EGFr (Genmab), idronoxil (CAS No. 81267-65-4) (Novogen), IGF-1 inhibitors from Ontogen, IMC-11 F8 (ImClone Systems), kahalalide F (CAS No. 149204-42-2) (PharmaMar), KDR inhibitor from LG Life Sciences, KDR inhibitors from Abbott Laboratories, KDR kinase inhibitors (UCB), Kdr kinase inhibitors from Merck & Co, KRN-633 and KRN-951 (Kirin Brewery), KSB-102 (Xenova), lapatinib ditosylate (CAS No. 388082-78-8) (GlaxoSmithKline), matuzumab (Merck KGaA), MDX-214 (Medarex), ME-103 (Pharmexa), MED-A300 (Gerolymatos), MNAC-13 (Lay Line Genomics), nimotuzumab (Center of Molecular Immunology), NSC-330507 or NSC-707545 (NIH), NV-50 (Novogen), OSI-930 (OSI Pharmaceuticals), panitumumab (Abgenix), pelitinib (CAS No. 287933-82-7) (Wyeth), pertuzumab (CAS No. 380610-27- 5) (Genentech), Pharmaprojects No. 3985 (Sanofi-Aventis), prostate cancer therapeutics from Sequenom (SQPC35, SQPC36, SQPC90), removab and remoxab (Trion Pharma), RG-13022 (CAS No. 136831-48-6), RG-13291 (CAS No. 138989-50-1), or RG-14620 (CAS No. 136831-49-7) (Sanofi- Aventis), RM-6427 (Romark), RNAi breast cancer therapy from Benitec, RP 53801 (CAS No. 125882-88-4) (Sanofi-Aventis), sorafenib tosylate (Bayer), SU-11657 (Pfizer), Tie-2 antagonists from Semaia (Hybrigenics), Tie-2 inhibitors from Ontogen, trastuzumab (CAS No. 180288-69-1) (Genentech), tyrosine kinase inhibitors from Sanofi-Aventis, U3-1287, U3-1565, U3-1784, U3-1800 (U3 Pharma), vatalanib (Novartis), VEGFR-2 kinase inhibitor from Bristol-Myers Squibb, XL-647 (Exelixis), ZD-6474 (AstraZeneca), ZK-CDK (Schering AG)1 an EGFR tyrosine kinase inhibitor, EKB-569 (Wyeth), and an analogue or derivative thereof.
4681. The device of item 4579-wherein the agent is a VEGF inhibitor.
4682. The device of item 4579 wherein the agent is a vitamin D receptor agonist.
4683. The device of item 4579 wherein the agent is ZD-6474 (an angiogenesis inhibitor).
4684. The device of item 4579 wherein the agent is AP-23573 (an mTOR inhibitor).
4685. The device of item 4579 wherein the agent is synthadotin (a tubulin antagonist). 4686. The device of item 4579 wherein the agent is S-0885 (a collagenase inhibitor).
4687. The device of item 4579 wherein the agent is aplidine (an elongation factor-1 alpha inhibitor).
4688. The device of item 4579 wherein the agent is ixabepilone (an epithilone).
4689. The device of item 4579 wherein the agent is IDN-5390 (an angiogenesis inhibitor).
4690. The device of item 4579 wherein the agent is SB- 2723005 (an angiogenesis inhibitor).
4691. The device of item 4579 wherein the agent is ABT-518 (an angiogenesis inhibitor).
4692. The device of item 4579 wherein the agent is combretastatin (an angiogenesis inhibitor).
4693. The device of item 4579 wherein the agent is anecortave acetate (an angiogenesis inhibitor).
4694. The device of item 4579 wherein the agent is SB- 715992 (a kinesin antagonist).
4695. The device of item 4579 wherein the agent is temsirolimus (an mTOR inhibitor). 4696. The device of item 4579 wherein the agent is adalimumab (a TNFα antagonist).
4697. The device of item 4579, further comprising a polymer.
4698. The device of item 4579 wherein the anti-scarring agent inhibits adhesion between the device and a host into which the device is implanted.
4699. The device of item 4579 wherein the device delivers the anti-scarring agent locally to tissue proximate to the device.
4700. The device of item 4579, further comprising a coating, wherein the coating comprises the anti-scarring agent.
.4701. The device of item 4579, further comprising a coating, wherein the coating is disposed on a surface of the device.
4702. The device of item 4579, further comprising a coating, wherein the coating directly contacts the device.
4703. The device of item 4579, further comprising a coating, wherein the coating indirectly contacts the device.
4704. The device of item 4579, further comprising a coating, wherein the coating partially covers the device.
4705. The device of item 4579, further comprising a coating, wherein the coating completely covers the device. 4706. The device of item 4579, further comprising a coating, wherein the coating is a uniform coating.
4707. The device of item 4579, further comprising a coating, wherein the coating is a non-uniform coating.
4708. The device of item 4579, further comprising a coating, wherein the coating is a discontinuous coating.
4709. The device of item 4579, further comprising a coating, wherein the coating is a patterned coating.
4710. The device of item 4579, further comprising a coating, wherein the coating has a thickness of 100 Dm or less.
- - . 4711. The device of item 4579, further comprising a coating, wherein the coating has a thickness of 10 Dm or less.
4712. The device of item 4579, further comprising a coating, wherein the coating adheres to the surface of the device upon deployment of the device.
4713. The device of item 4579, further comprising a coating, wherein the coating is stable at room temperature for a period of 1 year.
4714. The device of item 4579, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 0.0001% to about 1% by weight. B Λ_a O 2006/135479
4715. The device of item 4579, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 1% to about 10% by weight.
4716. The device of item 4579, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 10% to about 25% by weight.
4717. The device of item 4579, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 25% to about 70% by weight.
4718. The device of item 4579, further comprising a coating, wherein the coating further comprises a polymer.
4719. The .device of item 4579, further comprising a first coating having a first composition and the second coating having a second composition.
4720. The device of item 4579, further comprising a first coating having a first composition and the second coating having a second composition, wherein the first composition and the second composition are different.
4721. The device of item 4579, further comprising a polymeric carrier, wherein the polymeric carrier comprises a copolymer.
4722. The device of item 4579, further comprising a polymeric carrier, wherein the polymeric carrier comprises a block copolymer. 4723. The device of item 4579, further comprising a polymeric carrier, wherein the polymeric carrier comprises a random copolymer.
4724. The device of item 4579, further comprising a polymeric carrier, wherein the polymeric carrier comprises a biodegradable polymer.
4725. The device of item 4579, further comprising a polymeric carrier, wherein the polymeric carrier comprises a non-biodegradable polymer.
4726. The device of item 4579, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrophilic polymer.
4727. The device of item 4579, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrophobic polymer.
4728. The device of item 4579, further comprising a polymeric carrier, wherein the polymeric carrier comprises a polymer having hydrophilic domains.
4729. The device of item 4579, further comprising a polymeric carrier, wherein the polymeric carrier comprises a polymer having hydrophobic domains.
4730. The device of item 4579, further comprising a polymeric carrier, wherein the polymeric carrier comprises a non-conductive polymer.
4731. The device of item 4579, further comprising a polymeric carrier, wherein the polymeric carrier comprises an elastomer. 4732. The device of item 4579, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrogel.
4733. The device of item 4579, further comprising a polymeric carrier, wherein the polymeric carrier comprises a silicone polymer.
4734. The device of item 4579, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrocarbon polymer.
4735. The device of item 4579, further comprising a polymeric carrier, wherein the polymeric carrier comprises a styrene-derived polymer.
4736. The device of item 4579, further comprising a polymeric carrier, wherein the polymeric carrier comprises a butadiene polymer.
4737. The device of item 4579, further comprising a polymeric carrier, wherein the polymeric carrier comprises a macromer.
4738. The device of item 4579, further comprising a polymeric carrier, wherein the polymeric carrier comprises a poly(ethylene glycol) polymer.
4739. The device of item 4579, further comprising a polymeric carrier, wherein the polymeric carrier comprises an amorphous polymer.
4740. The device of item 4579, further comprising a lubricious coating.
4741. The device of item 4579 wherein the anti-scarring agent is located within pores or holes of the device. 4742. The device of item 4579 wherein the anti-scarring agent is located within a channel, lumen, or divet of the device.
4743. The device of item 4579, further comprising a second pharmaceutically active agent.
4744. The device of item 4579, further comprising an antiinflammatory agent.
4745. The device of item 4579, further comprising an agent that inhibits infection.
4746. The device of item 4579, further comprising an agent that inhibits infection, wherein the agent is an anthracycline.
4747. The device of item 4579, further comprising an agent that inhibits infection, wherein the agent is doxorubicin.
4748. The device of item 4579, further comprising an agent that inhibits infection, wherein the agent is mitoxantrone.
4749. The device of item 4579, further comprising an agent that inhibits infection, wherein the agent is a fluoropyrimidine.
4750. The device of item 4579, further comprising an agent that inhibits infection, wherein the agent is 5-fluorouracil (5-FU).
4751. The device of item 4579, further comprising an agent that inhibits infection, wherein the agent is a folic acid antagonist. 4752. The device of item 4579, further comprising an agent that inhibits infection, wherein the agent is methotrexate.
4753. The device of item 4579, further comprising an agent that inhibits infection, wherein the agent is a podophyiotoxin.
4754. The device of item 4579, further comprising an agent that inhibits infection, wherein the agent is etoposide.
4755. The device of item 4579, further comprising an agent that inhibits infection, wherein the agent is a camptothecin.
4756. The device of item 4579, further comprising an agent that inhibits infection, wherein the agent is a hydroxyurea.
4757. The device of item 4579, further comprising an agent that inhibits infection, wherein the agent is a platinum complex.
4758. The device of item 4579, further comprising an agent that inhibits infection, wherein the agent is cisplatin.
4759. The device of item 4579, further comprising an antithrombotic agent.
4760. The device of item 4579, further comprising a visualization agent.
4761. The device of item 4579, further comprising a visualization agent, wherein the visualization agent is a radiopaque material, wherein the radiopaque material comprises a metal, a halogenated compound, or a barium containing compound. 4762. The device of item 4579, further comprising a visualization agent, wherein the visualization agent is a radiopaque material, wherein the radiopaque material comprises barium, tantalum, or technetium.
4763. The device of item 4579, further comprising a visualization agent, wherein the visualization agent is a MRI responsive material.
4764. The device of item 4579, further comprising a visualization agent, wherein the visualization agent comprises a gadolinium chelate.
4765. The device of item 4579, further comprising a visualization agent, wherein the visualization agent comprises iron, magnesium, manganese, copper, or chromium.
4766. The device of item 4579, further comprising a visualization agent, wherein the visualization agent comprises an iron oxide compound.
4767. The device of item 4579, further comprising a visualization agent, wherein the visualization agent comprises a dye, pigment, or colorant.
4768. The device of item 4579, further comprising an echogenic material.
4769. The device of item 4579, further comprising an echogenic material, wherein the echogenic material is in the form of a coating. 4770. The device of item 4579 wherein the device is sterile.
4771. The device of item 4579 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device.
4772. The device of item 4579 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is connective tissue.
4773. The device of item 4579 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is muscle tissue.
4774. The device of item 4579 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is nerve tissue.
4775. The device of item 4579 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is epithelium tissue.
4776. The device of item 4579 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from the time of deployment of the device to about 1 year.
4777. The device of item 4579 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from about 1 month to 6 months. 4778. The device of item 4579 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from about 1 - 90 days.
4779. The device of item 4579 wherein the anti-scarring agent is released in effective concentrations from the device at a constant rate.
4780. The device of item 4579 wherein the anti-scarring agent is released in effective concentrations from the device at an increasing rate.
4781. The device of item 4579 wherein the anti-scarring agent is released in effective concentrations from the device at a decreasing rate.
4782. The device of item 4579 wherein the anti-scarring agent is released in effective concentrations from the composition comprising the anti-scarring agent by diffusion over a period ranging from the time of deployment of the device to about 90 days.
4783. The device of item 4579 wherein the anti-scarring agent is released in effective concentrations from the composition comprising the anti-scarring agent by erosion of the composition over a period ranging from the time of deployment of the device to about 90 days.
4784. The device of item 4579 wherein the device comprises about 0.01 Dg to about 10 Dg of the anti-scarring agent.
4785. The device of item 4579 wherein the device comprises about 10 Dg to about 10 mg of the anti-scarring agent.
4786. The device of item 4579 wherein the device comprises about 10 mg to about 250 mg of the anti-scarring agent. 4787. The device of item 4579 wherein the device comprises about 250 mg to about 1000 mg of the anti-scarring agent.
4788. The device of item 4579 wherein the device comprises about 1000 mg to about 2500 mg of the anti-scarring agent.
4789. The device of item 4579 wherein a surface of the device comprises less than 0.01 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
4790. The device of item 4579 wherein a surface of the device comprises about 0.01 Dg to about 1 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
4791. The device of item 4579 wherein a surface of the device comprises about 1_Dgio about 10 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
4792. The device of item 4579 wherein a surface of the device comprises about 10 Dg to about 250 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
4793. The device of item 4579 wherein a surface of the device comprises about 250 Dg to about 1000 Dg of the anti-scarring agent of anti- scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
4794. The device of item 4579 wherein a surface of the device comprises about 1000 Dg to about 2500 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied. 4795. A method as in any one of items 4579-4794, wherein the device is a central venous catheter with a cuff.
4796. A method as in any one of items 4579-4794, wherein the device is a central venous catheter without a cuff.
4797. A method as in any one of items 4579-4794, wherein the device is a central venous catheter with a flange.
4798. A method as in any one of items 4579-4794, wherein the device is a central venous catheter without a flange.
4799. A method as in any one of items 4579-4794, wherein the device is a central venous catheter adapted for providing access to the circulatory system.
4800. A method as in any one of items 4579-4794, wherein the device is a central venous catheter adapted for providing multiple conduits for accessing the circulatory system.
4801. A method as in any one of items 4579-4794, wherein the device is a central venous catheter comprising a means for preventing infection as a result of long term use.
4802. A method as in any one of items 4579-4794, wherein the device is a central venous catheter adaptable for being used with an apparatus that provides a means for controlling the injection or withdrawl of bodily fluids through the central venous catheter.
4803. A method as in any one of items 4579-4794, wherein the device is a parenteral nutrition catheter.
4804. A method as in any one of items 4579-4794, wherein the device is a peripherally inserted central venous catheter. 4805. A method as in any one of items 4579-4794, wherein the device is a flow directed balloon tipped pulmonary artery catheter.
4806. A method as in any one of items 4579-4794, wherein the device is a long term central venous access catheter.
4807. A device, comprising a ventricular assist device (e.g., an implant) and an anti-scarring agent or a composition comprising an anti- scarring agent, wherein the agent inhibits scarring between the device and a host into which the device is implanted.
4808. The device of item 4807 wherein the agent is an adensosine A2A receptor antagonist.
4809. The device of item 4807 wherein the agent is an AKT inhibitor.
4810. The device of item 4807 wherein the agent is an alpha 2 integrin antagonist, wherein the alpha 2 integrin antagonist is Pharmaprojects No. 5754 (Merck KgaA).
4811. The device of item 4807 wherein the agent an alpha 4 integrin antagonist.
4812. The device of item 4807 wherein the agent is an alpha 7 nicotinic receptor agonist.
4813. The device of item 4807 wherein the agent is an angiogenesis inhibitor selected from the group consisting of AG-12,958 (Pfizer), ATN-161 (Attenuon LLC), neovastat, an angiogenesis inhibitor from Jerina AG (Germany), NM-3 (Mercian), VGA-1155 (Taisho), FCE-26644 (Pfizer), FCE-26950 (Pfizer), FPMA (Meiji Daries), FR-111142 (Fujisawa), GGTI-298, GM-1306 (Ligand), GPA-1734 (Novartis), NNC-47-0011 (Novo Nordisk), herbamycin (Nippon Kayaku), lenalidomide (Celegene), IP-10 (NIH), ABT-828 (Abbott), KIN-841 (Tokushima University, Japan), SF-1126 (Semafore Pharmaceuticals), laminin technology (NIH), CHIR-258 (Chiron), NVP-AEW541 (Novartis), NVP-AEW541 (Novartis), Vt16907 (Alchemia), OXI-8007 (Oxigene), EG-3306 (Ark Therapeutics), Maspin (Arriva), ABT- 567 (Abbott), PPI-2458 (Praecis Pharmaceuticals), CC-5079, CC-4089 (Celgene), HIF-1 alpha inhibitors (Xenova), S-247 (Pfizer), AP-23573 (Ariad), AZD-9935 (Astra Zeneca), mebendazole (Introgen Therapeutics), MetAP-2 inhibitors (GlaxoSmithKline), AG-615 (Angiogene Pharmaceuticals), Tie-2 antagonists (Hybrigenics), NC-381 , CYC-381, NC-169, NC-219, NC-383, NC-384, NC-407 (Lorus Therapeutics), ATN-224 (Attenuon), ON-01370 (Onconova), Vitronectin antagonists (Amgen), SDX-103 (Salmedix), Vitronectin antagonists (Shire), CHP (Riemser), TEK (Amgen), Anecortave acetate (Alcon), T46.2 (Matrix Therapeutics), HG-2 (Heptagen), TEM antagonists (Genzyme), Oxi-4500 (Oxigene), ATN-161 (Attenuon), WX-293 (Wilex), M-2025 (Metris Therapeutics), Alphastatin (BioActa), YH-16 (Yantai Rongchang), BIBF-1120 (Boehringer Ingelheim), BAY-57-9352 (Bayer), AS- 1404 (Cancer Research Technology), SC-77964 (Pfizer), glycomimetics (BioTie Therapies), TIE-2 Inhibitors (Ontogen), DIMI, Octamer (Octamer), ABR-215050 (Active Biotech), ABT-518 (Abbott), KDR inhibitors (Abbott), BSF-466895 (Abbott), SCH-221153 (Schering-Plough), DAC:antiangiogenic (ConjuChem), TFPI (EntreMed), AZD-2171 (Astra-Zenaca), CDC-394 (Celgene), LY290293 (EIi Lilly), IDN-5390 (Indena), Kdr Kinase Inhibitors (Merck), CT-113020, CT-116433, CT-116563, CT-31890, CT-32228) (Cell Therapeutics), A-299620 (Abbott), TWEAK Inhibitor (Amgen), VEGF modulators (Johnson and Johnson), Tum-N53, tumstatin (Genzyme), Thios- 1, Thios-2 (Thios Pharmaceuticals), MV-6401 (Miravant Medical Technologies), Spisulosine (PharmaMar), CEP-7055 (Cephalon), AUV-201 (Auvation), LM-609 (EIi Lilly), SKF-106615 (AnorMED), Oglufanide disodium (Cytran), BW-114 (Pharminox), Calreticulin (NIH), WX-678 (Wilex), SD-7784 B Λ_a O 2006/135479
(Pfizer), WX-UK1 (Wilex), SH-268 (Schering AG), 2-Me-PGA (Celgene), S- 137 (Pfizer), ZD-6126 (Angiogene Pharmaceuticals), SG-292 (SignalGen), Benefin (Lane Labs), A6, A36 (Angstrom), SB-2723005 (GlaxoSmithKline), SC-7 (Cell Therapeutics), ZEN-014 (AEterna Zentaris), 2-methoxyestradiol (EntreMed), NK-130119 (Nippon Kayaku), CC-10004 (Celgene), AVE- 8062A (Ajinomoto), Tacedinaline (Pfizer), Actinonin (Tokyo Metropolitan Institute of Medical Science), Lenalidomide (Celgene), VGA-1155, BTO-956 (SRI International), ER-68203-00 (Eisai), CT-6685 (UCB), JKC-362 (Phoenix Pharmaceuticals), DMI-3798 (DMI Biosciences, Angiomate (Ipsen), ZD-6474 (AstraZeneca), CEP-5214 (Cephalon), Canstatin (Genzyme), NM-3 (Mercian), Oridigm (MediQuest Therapeutics), Exherin (Adherex), BLS-0597 (Boston Life Sciences), PTC-299 (PTC Therapeutics), NPI-2358 (Nereus Pharmaceuticals), CGP-79787 (Novartis), AEE-788 (Novartis), CKD-732 (Chong Kun Dang), CP-564959 (OSI Pharmaceuticals), CM-101 (CarboMed), CT-2584, CT3501 (Cell Therapeutics), combretastatiα and analogues and derivatives thereof (Oxigene), Rebimastat (Bristol-Meyers Squibb), Dextrin 2-sulfate (ML Laboratories), Cilengitide (Merk KGaA), NSC-706704 (Pharminox), KRN- 951 (Kirin Brewery), Ukrain, NSC-631570 (Nowicky Pharma), Tecogalan sodium (Daiichi Pharmaceutical), Tz-93 (Tsumura), TBC-1635 (Encysive Pharmaceuticals), TAN-1120 (Takeda), Semaxanib (Pfizer), BDI-7800 (Biopharmacopae), SD-186, SD-983 (Bristol-Meyers Squibb), SB-223245 (GlaxoSmithKline), SC-236 (Pfizer), RWJ-590973 (Johnson and Johnson), ILX-1850 (Genzyme), SC-68488, S-836 (Pfizer), CG-55069-11 (CuraGen), Ki-23057 (Kirin Brewery), CCX-700 (Chemoentryx), Pegaptanib octasodium (Giled Sciences), ANGIOCOL (available from Biostratum Inc.), or an analogue or derivative thereof.
4814. The device of item 4807 wherein the agent is an apoptosis antagonist. 4815. The device of item 4807 wherein the agent is an apoptosis activator.
4816. The device of item 4807 wherein the agent is a beta 1 integrin antagonist.
4817. The device of item 4807 wherein the agent is a beta tubulin inhibitor.
4818. The device of item 4807 wherein the agent is a blocker of enzyme production in Hepatitis C.
4819. The device of item 4807 wherein the agent is a Bruton's tyrosine kinase inhibitor.
4820. The device of item 4807 wherein the agent is a calcineurin inhibitor.
4821. The device of item 4807 wherein the agent is a caspase 3 inhibitor.
4822. The device of item 4807 wherein the agent is a CC chemokine receptor antagonist.
4823. The device of item 4807 wherein the agent is a cell cycle inhibitor selected from the group consisting of SNS-595 (Sunesis), synthadotin, KRX-0403, and an analogue or derivative thereof.
4824. The device of item 4807 wherein the agent is a cathepsin B inhibitor. 4825. The device of item 4807 wherein the agent is a cathepsin K inhibitor, wherein the cathepsin K inhibitor is 462795 (GlaxoSmithKline), INPL-022-D6 (Amura Therapeutics), an analogue or derivative thereof).
4826. The device of item 4807 wherein the agent is a cathepsin L inhibitor.
4827. The device of item 4807 wherein the agent is a CD40 antagonist.
4828. The device of item 4807 wherein the agent is a chemokine receptor agonist.
4829. The device of item 4807 wherein the agent is a chymase inhibitor.^
4830. The device of item 4807 wherein the agent is a collagenase antagonist.
4831. The device of item 4807 wherein the agent is a CXCR antagonist.
4832. The device of item 4807 wherein the agent is a cyclin dependent kinase inhibitor selected from the group consisting of a CDK-1 inhibitor, a CDK-2 inhibitor, a CDK- 4 inhibitor, a CDK-6 inhibitor, a CAK1 inhibitor from GPC Biotech and Bristol-Myers Squibb, RGB-286199 (GPC Biotech), an anticancer agent from Astex Technology, a CAK1 inhibitor from GPC Biotech, a CDK inhibitor from Sanofi-Aventis, a CDK1/CDK2 inhibitor from Amgen, a CDK2 inhibitor from SUGEN-2 (Pfizer), a hearing loss therapy agent (Sound Pharmaceuticals), PD-0332991 (Pfizer), RGB-286199 (GPC Biotech), Ro-0505124 (Hoffmann-La Roche), a Ser/Thr kinase inhibitor from Lilly (EIi Lilly), CVT-2584 (CAS No. 199986-75-9) (CV Therapeutics), and an analogue or derivative thereof.
4833. The device of item 4807 wherein the agent is a cyclooxygenase 1 inhibitor.
4834. The device of item 4807 wherein the agent is a DHFR inhibitor.
4835. The device of item 4807 wherein the agent is a dual integrin inhibitor.
4836. The device of item 4807 wherein the agent is an elastase inhibitor.
4837. The device of item 4807 wherein the agent is an elongation factor- 1 alpha inhibitor.
4838. The device of item 4807 wherein the agent is an endothelial growth factor antagonist.
4839. The device of item 4807 wherein the agent is an endothelial growth factor receptor kinase inhibitor selected from the group consisting of sorafenib tosylate (Bayer), AAL-993 (Novartis), ABP-309 (Novartis), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), E- 7080 (Eisai), EG-3306 (Ark Therapeutics), EXEL-2880 (Exelixis), GW- 654652 (GlaxoSmithKline), a KDR inhibitor from LG Life Sciences, CT-6685 and CT-6729 (UCB), KRN-633 and KRN-951 (Kirin Brewery), OSI-930 (OSI Pharmaceuticals), SP-5.2 (Supratek Pharma), SU-11657 (Pfizer), a Tie-2 antagonist (Hybrigenics), a VEGFR-2 kinase inhibitor (Bristol-Myers Squibb), XL-647 (Exelixis), a KDR inhibitor from Abbott Laboratories, sorafenib tosylate, and an analogue or derivative thereof.
4840. The device of item 4807 wherein the agent is an endotoxin antagonist.
4841. The device of item 4807 wherein the agent is an epothilone and tubulin binder.
4842. The device of item 4807 wherein the agent is an estrogen receptor antagonist.
4843. The device of item 4807 wherein the agent is an FGF inhibitor.
4844. The device of item 4807 wherein the agent is a farnexyl transferase inhibitor.
4845. The device of item 4807 wherein the agent is farnesyltransferase inhibitor selected from the group of A-197574 (Abbott), a famesyltransferase inhibitor from Servier, FPTIII (Strathclyde Institute for Drug R), LB-42908 (LG Life Sciences), Pharmaprojects No. 5063 (Genzyme), Pharmaprojects No. 5597 (Ipsen), Yissum Project No. B-1055 (Yissum), and an analogue or derivative thereof.
4846. The device of item 4807 wherein the agent is an FLT-3 kinase inhibitor.
4847. The device of item 4807 wherein the agent is an FGF receptor kinase inhibitor. 4848. The device of item 4807 wherein the agent is a fibrinogen antagonist selected from the group consisting of AUV-201 (Auvation), MG-13926 (Sanofi-Aventis), plasminogen activator (CAS No. 105913-11-9) (from Sanofi-Aventis or UCB), plasminogen activator-2 (tPA- 2) (Sanofi-Aventis), pro-urokinase (CAS No. 82657-92-9) (Sanofi-Aventis), and an analogue or derivative thereof.
4849. The device of item 4807 wherein the agent is a heat shock protein 90 antagonist selected from the group consisting of SRN-005 (Sirenade), geldanamycin, NSC-33050 (17-allylaminogeldanamycin; 17- AAG), 17-dimethylaminoethylamino-17-demethoxy-geldanamycin (17- DMAG), rifabutin (rifamycin XIV, 1',4-didehydro-1-deoxy-1,4-dihydro-5'-(2- methylpropyl)-1-oxo-), and an analogue or derivative thereof.
4850. The device of item 4807 wherein the agent is a histone deacetylase inhibitor. ._ . . . - -
4851. The device of item 4807 wherein the agent is an HMGCoA reductase inhibitor selected from the group consisting of an atherosclerosis therapeutic from Lipid Sciences, ATI-16000 (ARYx Therapeutics), KS-01-019 (Kos Pharmaceuticals), Pharmaprojects No. 2197 (Sanofi-Aventi), RP 61969 (Sanofi-Aventis), and an analogue or derivative thereof.
4852. The device of item 4807 wherein the agent is an ICAM inhibitor.
4853. The device of item 4807 wherein the agent is an IL, ICE and IRAK antagonist, wherein the antagonist is a CJ-14877, CP-424174 (Pfizer), NF-61 (Negma-Lerads), and an analogue or derivative thereof. 4854. The device of item 4807 wherein the agent is an IL-2 inhibitor.
4855. The device of item 4807 wherein the agent is an immunosuppressant selected from the group consisting of teriflunomide (Sanofi Aventis), chlorsulfaquinoxalone (NSC-339004), chlorsulfaquinoxalone sulfate, CS-712 (Sankyo), ismomultin alfa (CAS No. 457913-93-8) (Akzo Nobel), antiallergics from GenPat77, antiinflammatories or AT-005 (Androclus Therapeutics), autoimmune disease therapeutics from EpiVax, BN-007 (Bone), budesonide (CAS No. 51333-22- 3) (MAP Pharmaceuticals), CO-14 (Genzyme), edratide (CAS No. 433922- 67-9) (Teva), EP-314 (Enanta), eprovafen (CAS No. 101335-99-3) (Sanofi- Aventis), HWA-131 (CAS No. 118788-41-3) (Sanofi-Aventis), immunomodulators from MerLion Pharmaceuticals, immunosuppressives from Alchemia, IPL-12 (Inflazyme), MDL-9563 (CAS No. 27086-86-8) (Sanofi-Aventis), Pharmaprojects No..2330 (Sanofi-Aventis), Pharmaprojects No. 6426 (Abgenix), PXS-25 (Pharmaxis), rosmarinic acid (CAS No. 20283-92-5) (Sanofi-Aventis), RP 42927 or RP 54745 (CAS No. 135330-08-4) (Sanofi-Aventis), SGN-35 (Seattle Genetics), ST-1959 (Sigma-Tau), type I diabetes therapy from SYNX Pharma, UNIL-88 (Debiopharm), VP-025 (Vasogen), VR-694 (Vectura), PRTX-001 (Protalex), and an analogue or derivative thereof.
4856. The device of item 4807 wherein the agent is an IMPDH (inosine monophosphate).
4857. The device of item 4807 wherein the agent is an integrin antagonist.
4858. The device of item 4807 wherein the agent is an interleukin antagonist. 4859. The device of item 4807 wherein the agent is an inhibitor of type III receptor tyrosine kinase.
4860. The device of item 4807 wherein the agent is an irreversible inhibitor of enzyme methionine aminopeptidase type 2.
4861. The device of item 4807 wherein the agent is an isozyme selective delta protein kinase C inhibitor.
4862. The device of item 4807 wherein the agent a JAK3 enzyme inhibitor.
4863. The device of item 4807 wherein the agent is a JNK inhibitor.
4864. The device of item 4807 wherein the agent is a kinase inhibitor.
4865. The device of item 4807 wherein the agent is kinesin antagonist.
4866. The device of item 4807 wherein the agent is a kinesin antagonist.
4867. The device of item 4807 wherein the agent is a leukotriene inhibitor and antagonist selected from the group consisting of ambicromil (CAS No. 58805-38-2) (Sanofi-Aventis), amelubant (CAS No. 346735-24-8) (Boehringer Ingelheim), DW-1141 (Dong Wha), ebselen (Daiichi Pharmaceutical), ibudilast (Kyorin), leucotriene inhibitors from Sanofi-Aventis, lymphotoxin-beta receptor (LT-D) from Biogen Idee, Pharmaprojects No. 1535 and 2728 (CAS No. 119340-33-9) (Sanofi- Aventis), R-112 (Rigel), Rev-5367 (CAS No. 92532-05-3) (Sanofi-Aventis), RG-14893 (CAS No. 141835-49-6) (Sanofi-Aventis), RG-5901-A (CAS No. 101910-24-1), 92532-23-5, RP 66153 (CAS No. 142422-79-5), RP 66364 (CAS No.186916-92-5), RP 69698 (CAS No. 141748-00-7) (Sanofi-Aventis), SC-411930 (Pfizer), SC-41930 (CAS No. 120072-59-5) (Pfizer), SC-50605 (CAS No. 138828-39-4) (Pfizer), SC-51146 (CAS No. 141059-52-1), SC- 53228 (CAS No. 153633-01-3) (Pfizer), spaglumic acid (ZY-15106) (CAS No.3106-85-2), 80619-64-3 (Novartis), tipredane (CAS No. 85197-77-9) (Bristol-Myers Squibb), U-75302 (CAS No. 119477-85-9) (Pfizer), and analogue or derivative thereof.
4868. The device of item 4807 wherein the agent is an MAP kinase inhibitor.
4869. The device of item 4807 wherein the agent is a matrix metalloproteinase inhibitor. -
4870. The device of item 4807 wherein the agent is an MCP- CCR2 inhibitor.
4871. The device of item 4807 wherein the agent is an mTOR inhibitor.
4872. The device of item 4807 wherein the agent is an mTOR kinase inhibitor.
4873. The device of item 4807 wherein the agent is a microtubule inhibitor selected from the group consisting of antibody- maytansinoid conjugates from Biogen Idee, colchicines (MantiCore Pharmaceuticals), anticancer immunoconjugates from Johnson & Johnson, DIME from Octamer, gnklf (GNI), huC242-DM4, huMy9-6-DM1 (ImmunoGen), IDN-5404 (Indena), IMO-098, IMOderm (Imotep), mebendazole (Introgen Therapeutics), microtubule poisons from Cambridge Enterprise, paclitaxel such as LOTAX from Aphios (CAS No. 33069-62-4), Genexol-PM from Samyang, Pharmaprojects No. 6383 (Tapestry Pharmaceuticals), RPR-112378 (Sanofi-Aventis), SGN-75 (Seattle Genetics), SPL-7435 (Starpharma), SSR-250411 (Sanofi-Aventis), trastuzumab-DM1 (Genentech), vinorelbine, and an analogue or derivative thereof.
4874. The device of item 4807 wherein the agent is an MIF inhibitor.
4875. The device of item 4807 wherein the agent is an MMP inhibitor.
4876. The device of item 4807 wherein the agent is a neurokinin (NK) antagonist selected from the group consisting of anthrotainin (CAS No. 148084-40-6) (Sanofi-Aventis), an IBS thereapeutic from ArQuIe, MDL-105212A (CAS No. 167261-60-1) (Ssanofi-Aventis), Pharmaprojects No. 2744, 3258 (CAS No. 139167-47-8) 4006, 4201, or 5986 (Sanofi-Aventis), RP 67580 (CAS No. 135911-02-3), SR-144190 (CAS No. 201152-86-5), SSR-240600, SSR-241586 (Sanofi-Aventis), TKA-457 (Novartis), vestipitant mesylate (CAS No. 334476-64-1) (GlaxoSmithKline), Win-64821 (Sanofi-Aventis), PRX-96026 (Predix Pharmaceuticals), and an analogue or derivative thereof.
4877. The device of item 4807 wherein the agent is an NF kappa B inhibitor selected from the group consisting of AVE-0545 or AVE- 0547 (Sanofi-Aventis), bortezomib (CAS No. 179324-69-7) (Millennium Pharmaceuticals), dexanabinol (CAS No. 112924-45-5) (Pharmos), dexlipotam (Viatris), Pharmaprojects No. 6283 (INDRA) (OXiGENE), IPL- 576092 (CAS No. 137571-30-3) (Inflazyme), NFKB decoy (Corgentech), NFKB decoy oligo (AnGes MG), NFKB's from Ariad, osteoporosis treatments or S5 (F005) from Fulcrum Pharmaceuticals, P61 (Phytopharm), R-flurbiprofen (CAS No. 5104-49-4) (Encore Pharmaceuticals), and an analogue or derivative thereof.
4878. The device of item 4807 wherein the agent is a nitric oxide agonist.
4879. The device of item 4807 wherein the agent is an ornithine decarboxylase inhibitor.
4880. The device of item 4807 wherein the agent is a p38 MAP kinase inhibitor selected from the group consisting of AZD-6703 (AstraZeneca), JX-401 (Jexys Pharmaceuticals), BMS-2 (Bristol-Myers Squibb), a p38 MAP kinase inhibitor from Novartis, a p38-alpha MAP kinase inhibitor from Amphora, Pharmaprojects No. 5704 (Pharmacopeia), RPR- 200765A (Sanofi-Aventis), SD-282 (Johnson & Johnson), TAK-715 (Takeda), and an analogue or derivative thereof.
4881. The device of item 4807 wherein the agent is a palmitoyl-protein thioesterase inhibitor.
4882. The device of item 4807 wherein the agent is a PDGF receptor kinase inhibitor selected from the group consisting of AAL-993, AMN-107, or ABP-309 (Novartis), AMG-706 (Amgen), BAY-57-9352 (Bayer), CDP-860 (UCB), E-7080 (Eisai), imatinib (CAS No. 152459-95-5) (Novartis), OSI-930 (OSI Pharmaceuticals), RPR-127963E (Sanofi-Aventis), RWJ-540973 (Johnson & Johnson), sorafenib tosylate (Bayer), SU-11657 (Pfizer), tandutinib (CAS No. 387867-13-2) (Millennium Pharmaceuticals), vatalanib (Novartis), ZK-CDK (Schering AG), and an analogue or derivative thereof.
4883. The device of item 4807 wherein the agent is (-)- halofenate (Metabolex), AMG-131 (Amgen), antidiabetics from Japan Tobacco, AZD-4619, AZD-8450, AZD-8677 (AstraZeneca), DRF-10945, balaglitazone (Dr Reddy's), CS-00088, CS-00098 (Chipscreen Biosciences), E-3030 (Eisai), etalocib (CAS No. 161172-51-6) (EIi Lilly), GSK-641597 (Ligand), GSK-677954 (GlaxoSmithKline), GW-409544 (Ligand), GW- 590735 (GlaxoSmithKline), K-111 (Hoffmann-La Roche), LY-518674 (EIi Lilly), LY-674 (Ligand), LY-929 (Ligand), MC-3001 , MC-3002 (MaxoCore Pharmaceuticals), metformin HCI + pioglitazone (CAS No. 1115-70-4 and
112529-15-4), ACTOPLUS MET from Andrx), muraglitazar (CAS No. 331741-94-7) (Bristol-Myers Squibb), naveglitazar (Ligand), oleoylethanolamide (Kadmus Pharmaceuticals), ONO-5129, pioglitazone hydrochloride (CAS No. 111025-46-8 and 112529-15-4) (Takeda), PLX-204 (Plexxikon), PPAR agonists from Genfit, PPAR delta agonists from EIi Lilly, PPAR-alpha agonists from CrystalGenomics, PPAR-gamma modulators and PPAR-β modulators from CareX, rosiglitazone maleate (CAS No. 122324- 73-4 or 155141-29-0) (GlaxoSmithKline), rosiglitazone maleate/glimepir (CAS No. 155141-29-0 and 93479-97-1), AVANDARYL, rosiglitazone maleate/metformin extend (CAS No. 155141-29-0 and 657-24-9), AVANDAMET, rosiglitazone maleate+metformin, AVANDAMET (GlaxoSmithKline), tesaglitazar (AstraZeneca), and an analogue or derivative thereof.
4884. The device of item 4807 wherein the agent is a phosphatase inhibitor.
4885. The device of item 4807 wherein the agent is a phosphodiesterase (PDE) inhibitor selected from the group consisting of avanafil (Tanabe Seiyaku), dasantafil (CAS No. 569351-91-3) (Schering- Plough), A-906119 (CAS No. 134072-58-5), DL-850 (Sanofi-Aventis), GRC- 3015, GRC-3566, GRC-3886 (Glenmark), HWA-153 (CAS No. 56395-66-5) (Sanofi-Aventis), hydroxypumafentrine (Altana), IBFB-130011 , IBFB-14-016, IBFB-140301 , IBFB-150007, IBFB-211913 (IBFB Pharma), L-826141 (Merck & Co), medorinone (CAS No. 88296-61-1) (Sanofi-Aventis), MEM- 1917 (Memory Pharmaceuticals), ND-1251 (Neuro3d), PDE inhibitors from ICOS, PDE IV inhibitors from Memory Pharmaceuticals and CrystalGenomics, Pharmaprojects No. 2742 and 6141 (Sanofi-Aventis), QAD-171 (Novartis), RHC-2963 (CAS No.76993-12-9 and 76993-14-1), RPFM 17658, RPR-122818 derivatives, SR-24870 , and RPR-132294 (Sanofi-Aventis), SK-350 (ln2Gen), stroke therapy agents from deCODE Genetics, TAS-203 (Taiho), tofimilast (CAS No. 185954-27-2) (Pfizer), UK- 371800 (Pfizer), WIN-65579 (CAS No. 158020-82-7) (Sanofi-Aventis), IBFB- 130020 (IBFB Pharma), OPC-6535 (CAS No. 145739-56-6) (Otsuka), a phosphodiesterase III inhibitor, enoximone, a phosphodiesterase IV inhibitor, fosfosal, Atopik (Barrier Therapeutics), triflusal, a phosphodiesterase V inhibitor, and an analogue or derivative thereof.
4886. The device of item 4807 wherein the agent is a PKC inhibitor.
4887. The device of item 4807 wherein the agent is a platelet activating factor antagonist.
4888. The device of item 4807 wherein the agent is a platelet- derived growth factor receptor kinase inhibitor.
4889. The device of item 4807 wherein the agent is a prolyl hydroxylase inhibitor. 4890. The device of item 4807 wherein the agent is a polymorphonuclear neutrophil inhibitor.
4891. The device of item 4807 wherein the agent is a protein kinase B inhibitor.
4892. The device of item 4807 wherein the agent is a protein kinase C stimulant.
4893. The device of item 4807 wherein the agent is a purine nucleoside analogue.
4894. The device of item 4807 wherein the agent is a purinoreceptor P2X antagonist.
4895. The device of item 4807 wherein the agent is a Raf kinase inhibitor.
4896. The device of item 4807 wherein the agent is a reversible inhibitor of ErbB1 and ErbB2.
4897. The device of item 4807 wherein the agent is a ribonucleoside triphosphate reductase inhibitor.
4898. The device of item 4807 wherein the agent is an SDF-1 antagonist.
4899. The device of item 4807 wherein the agent is a sheddase inhibitor. 4900. The device of item 4807 wherein the agent is an SRC inhibitor.
4901. The device of item 4807 wherein the agent is a stromelysin inhibitor.
4902. The device of item 4807 wherein the agent is an Syk kinase inhibitor.
4903. The device of item 4807 wherein the agent is a telomerase inhibitor.
4904. The device of item 4807 wherein the agent is a TGF beta inhibitor selected from the group consisting of pirfenidone (CAS No. 53179-13-8) (MARNAC), tranilast (CAS No. 53902-12-8) (Kissei), IN-1130 (ln2Gen), mannose-6-phosphate (BTG), TGF-β antagonists from Infiazyme (Pharmaprojects No. 6075), TGF-β antagonists from Sydney, non-industrial source), TGF-β I receptor kinase inhibitors from EIi Lilly, TGF-β receptor inhibitors from Johnson & Johnson, and an analogue or derivative thereof.
4905. The device of item 4807 wherein the agent is a TNFα antagonist or TACE inhibitor selected from the group consisting of adalimumab (CAS No. 331731-18-1) (Cambridge Antibody Technology), AGIX-4207 (AtheroGenics), AGT-1 (Advanced Biotherapy), an antiinflammatory from Borean Pharma, Cellzome, or Paradigm Therapeutics, anti-inflammatory vaccine (TNF-alpha kinoid) from Neovacs, humanized anti-TNF antibody or an anti-TNF MAb (CB0006) Celltech (UCB), apratastat (CAS No. 287445-51-0) (Wyeth), BMS-561392 (Bristol-Myers Squibb), BN- 006 (Bone), certolizumab pegol (CAS No. 428863-50-7 or CH-138 (UCB), cilomilast (CAS No. 153259-65-5) (GlaxoSmithKline), CR-1 (Nuada Pharmaceuticals), CRx-119 (CombinatoRx), D-5410 (UCB), dacopafant (CAS No. 125372-33-0) (Sanofi-Aventis), dersalazine (CAS No. 188913-57- 7/188913-58-8) (Uriach), etanercept (CAS No. 185243-69-0) (Amgen), ethyl pyruvate (Critical (Critical Therapeutics), golimumab (CAS No. 476181-74-5) (Johnson & Johnson), hormono-immunotherapy from Ipsen, CDP571 (e.g., Humicade from UCB), IC-485 (ICOS), infliximab (CAS No. 170277-31-3) (Johnson & Johnson), IP-751 (Manhattan Pharmaceuticals), ISIS-104838 (CAS No. 250755-32-9) (ISIS Pharmaceuticals), lenalidomide (CAS No. 191732-72-6) (Celgene), lentinan (CAS No. 37339-90-5) (Ajinomoto), MDL- 201112 (CAS No. 142130-73-2) (Sanofi-Aventis), medroxyprogesterone (CAS No. 520-85-4) (InKine Pharmaceutical), N-acetylcysteine (CAS No. 616-91-1) (Zambon), NBE-P2 (DIREVO Biotech), nerelimomab (CAS No. 162774-06-3) (Chiron), OM-294DP (OM PHARMA), onercept (CAS No. 199685-57-9) (Yeda), PASSTNF-alpha (Verigen), pentoxifylline or oxypentifylline (Sanofi-Aventis), Pharmaprojects No. 4091, 4241, 4295, or 4488 (Sanofi-Aventis), Pharmaprojects No. 5480 (Amgen), Pharmaprojects No. 6749 (Cengent), pirfenidone (CAS No. 53179--13-8) (MARNAC), RPR- 132294 (Sanofi-Aventis), S5 (F002) (Fulcrum Pharmaceuticals), simvastatin (CAS No. 79902-63-9) (Merck & Co), STA-6292 (Synta Pharmaceuticals), tacrolimus (CAS No. 104987-11-3) (Fujisawa LifeCycle Pharma), talactoferrin alfa (CAS No. 308240-58-6) (Agennix), thalidomide (CAS No. 50-35-1) (Celgene), TNF antagonists form ProStrakan, and Synergen, TNF inhibitors (Amgen), TNF-alpha antagonists from Dynavax Technologies and Jerina AG (Germany), TNF-alpha inhibitors from IBFB Pharma and Xencor (Xencor), torbafylline (CAS No. 105102-21-4) (Sanofi-Aventis), UR-1505 (Uriach), VT-346 (Viron Therapeutics), YSIL6 (Y's Therapeutics), YSTH2 (Y's Therapeutics), NPI-1302a-3 (Nereus Pharmaceuticals, a TNF antagonist from Jerina AG (Germany), dersalazine, and an analogue or derivative thereof.
4906. The device of item 4807 wherein the agent is a Toll receptor inhibitor. 4907. The device of item 4807 wherein the agent is a tubulin antagonist.
4908. The device of item 4807 wherein the agent is a tyrosine kinase inhibitor selected from the group consisting of SU-011248, SUTENT from Pfizer Inc. (New York, NY), BMS-354825, PN-355 (Paracelsian Pharmaceuticals), AGN-199659 (Allergan), AAL-993 or ABP-309 (Novartis), adaphostin (NIH), AEE-788 (Novartis), AG-013736 (OSI Pharmaceuticals), AG-13736 (Pfizer), ALT-110 (Alteris Therapeutics), AMG-706 (Amgen), anticancer MAbs from Xencor, anti-EGFrvlll MAbs from Abgenix, anti-HER2 MAb from Abiogen, AZD-2171 or AZD-9935 (AstraZeneca), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), CEP-5214 (Cephalon), CEP- 7055 (Cephalon), cetuximab (ImClone Systems), CHIR-200131 and CHIR- 258 (Chiron), CP-547632 (OSI Pharmaceuticals), CP-724714 (Pfizer), CT- 301 (Creabilis Therapeutics), D-69491 (Baxter International), E-7080 (Eisai), EG-3306 (Ark Therapeutics),. EGFR/ErbB2 inhibitors from Array BioPharma, erlotinib (CAS No. 183319-69-9) (OSI Pharmaceuticals), EXEL-2880 (Exelixis), FK-778 (Sanofi-Aventis) , gefitinib (CAS No. 184475-35-2) (AstraZeneca), GW-2286 or GW-654652 (GlaxoSmithKline), her2/neu antigen from AlphaVax, HER-2/neu inhibitor from Generex, Herzyme (Medipad) (Sirna Therapeutics), HKI-272 (Wyeth), HuMax-EGFr (Genmab), idronoxil (CAS No. 81267-65-4) (Novogen), IGF-1 inhibitors from Ontogen, IMC-11F8 (ImClone Systems), kahalalide F (CAS No. 149204-42-2) (PharmaMar), KDR inhibitor from LG Life Sciences, KDR inhibitors from Abbott Laboratories, KDR kinase inhibitors (UCB), Kdr kinase inhibitors from Merck & Co, KRN-633 and KRN-951 (Kirin Brewery), KSB-102 (Xenova), lapatinib ditosylate (CAS No. 388082-78-8) (GlaxoSmithKline), matuzumab (Merck KGaA), MDX-214 (Medarex), ME-103 (Pharmexa), MED-A300 (Gerolymatos), MNAC-13 (Lay Line Genomics), nimotuzumab (Center of Molecular Immunology), NSC-330507 or NSC-707545 (NIH), NV-50 (Novogen), OSI-930 (OSI Pharmaceuticals), panitumumab (Abgenix), pelitinib (CAS No. 287933-82-7) (Wyeth), pertuzumab (CAS No. 380610-27- 5) (Genentech), Pharmaprojects No. 3985 (Sanofi-Aventis), prostate cancer therapeutics from Sequenom (SQPC35, SQPC36, SQPC90), removab and remoxab (Trion Pharma), RG-13022 (CAS No. 136831-48-6), RG-13291 (CAS No. 138989-50-1), or RG-14620 (CAS No. 136831-49-7) (Sanofi- Aventis), RM-6427 (Romark), RNAi breast cancer therapy from Benitec, RP 53801 (CAS No. 125882-88-4) (Sanofi-Aventis), sorafenib tosyiate (Bayer), SU-11657 (Pfizer), Tie-2 antagonists from Semaia (Hybrigenics), Tie-2 inhibitors from Ontogen, trastuzumab (CAS No. 180288-69-1) (Genentech), tyrosine kinase inhibitors from Sanofi-Aventis, U3-1287, U3-1565, U3-1784, U3-1800 (U3 Pharma), vatalanib (Novartis), VEGFR-2 kinase inhibitor from Bristol-Myers Squibb, XL-647 (Exelixis), ZD-6474 (AstraZeneca), ZK-CDK (Schering AG), an EGFR tyrosine kinase inhibitor, EKB-569 (Wyeth), and an analogue or derivative thereof.
4909. The device of item_4807 wherein the agent is a VEGF inhibitor.
4910. The device of item 4807 wherein the agent is a vitamin D receptor agonist.
4911. The device of item 4807 wherein the agent is ZD-6474 (an angiogenesis inhibitor).
4912. The device of item 4807 wherein the agent is AP-23573 (an mTOR inhibitor).
4913. The device of item 4807 wherein the agent is synthadotin (a tubulin antagonist). 4914. The device of item 4807 wherein the agent is S-0885 (a collagenase inhibitor).
4915. The device of item 4807 wherein the agent is apiidine (an elongation factor-1 alpha inhibitor).
4916. The device of item 4807 wherein the agent is ixabepilone (an epithilone).
4917. The device of item 4807 wherein the agent is IDN-5390 (an angiogenesis inhibitor).
4918. The device of item 4807 wherein the agent is SB- 2723005 (an angiogenesis inhibitor).
4919. The device of item 4807 wherein the agent is ABT-518 (an angiogenesis inhibitor).
4920. The device of item 4807 wherein the agent is combretastatin (an angiogenesis inhibitor).
4921. The device of item 4807 wherein the agent is anecortave acetate (an angiogenesis inhibitor).
4922. The device of item 4807 wherein the agent is SB- 715992 (a kinesin antagonist).
4923. The device of item 4807 wherein the agent is temsirolimus (an mTOR inhibitor). B Λ_a O 2006/135479
4924. The device of item 4807 wherein the agent is adalimumab (a TNFα antagonist).
4925. The device of item 4807, further comprising a polymer.
4926. The device of item 4807 wherein the anti-scarring agent inhibits adhesion between the device and a host into which the device is implanted.
4927. The device of item 4807 wherein the device delivers the anti-scarring agent locally to tissue proximate to the device.
4928. The device of item 4807, further comprising a coating, wherein the coating comprises the anti-scarring agent.
4929. The device of item 4807, further comprising a coating, wherein the coating is disposed on a surface of the device.
4930. The device of item 4807, further comprising a coating, wherein the coating directly contacts the device.
4931. The device of item 4807, further comprising a coating, wherein the coating indirectly contacts the device.
4932. The device of item 4807, further comprising a coating, wherein the coating partially covers the device.
4933. The device of item 4807, further comprising a coating, wherein the coating completely covers the device. 4934. The device of item 4807, further comprising a coating, wherein the coating is a uniform coating.
4935. The device of item 4807, further comprising a coating, wherein the coating is a non-uniform coating.
4936. The device of item 4807, further comprising a coating, wherein the coating is a discontinuous coating.
4937. The device of item 4807, further comprising a coating, wherein the coating is a patterned coating.
4938. The device of item 4807, further comprising a coating, wherein the coating has a thickness of 100 Dm or less.
4939. The device of item 4807, further comprising a coating, wherein the coating has a thickness of 10 Dm or less.
4940. The device of item 4807, further comprising a coating, wherein the coating adheres to the surface of the device upon deployment of the device.
4941. The device of item 4807, further comprising a coating, wherein the coating is stable at room temperature for a period of 1 year.
4942. The device of item 4807, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 0.0001% to about 1% by weight. 4943. The device of item 4807, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 1% to about 10% by weight.
4944. The device of item 4807, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 10% to about 25% by weight.
4945. The device of item 4807, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 25% to about 70% by weight.
4946. The device of item 4807, further comprising a coating, wherein the coating further comprises a polymer.
4947. The device of item 4807,-further comprising a first coating having a first composition and the second coating having a second composition.
4948. The device of item 4807, further comprising a first coating having a first composition and the second coating having a second composition, wherein the first composition and the second composition are different.
4949. The device of item 4807, further comprising a polymeric carrier, wherein the polymeric carrier comprises a copolymer.
4950. The device of item 4807, further comprising a polymeric carrier, wherein the polymeric carrier comprises a block copolymer. 4951. The device of item 4807, further comprising a polymeric carrier, wherein the polymeric carrier comprises a random copolymer.
4952. The device of item 4807, further comprising a polymeric carrier, wherein the polymeric carrier comprises a biodegradable polymer.
4953. The device of item 4807, further comprising a polymeric carrier, wherein the polymeric carrier comprises a non-biodegradable polymer.
4954. The device of item 4807, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrophilic polymer.
4955. The device of item 4807, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrophobic polymer.
4956. The device of item 4807, further comprising a polymeric carrier, wherein the polymeric carrier comprises a polymer having hydrophilic domains.
4957. The device of item 4807, further comprising a polymeric carrier, wherein the polymeric carrier comprises a polymer having hydrophobic domains.
4958. The device of item 4807, further comprising a polymeric carrier, wherein the polymeric carrier comprises a non-conductive polymer.
4959. The device of item 4807, further comprising a polymeric carrier, wherein the polymeric carrier comprises an elastomer. 4960. The device of item 4807, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrogel.
4961. The device of item 4807, further comprising a polymeric carrier, wherein the polymeric carrier comprises a silicone polymer.
4962. The device of item 4807, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrocarbon polymer.
4963. The device of item 4807, further comprising a polymeric carrier, wherein the polymeric carrier comprises a styrene-derived polymer.
4964. The device of item 4807, further comprising a polymeric carrier, wherein the polymeric carrier comprises a butadiene polymer.
4965. The device of item 4807, further comprising a polymeric carrier, wherein the polymeric carrier comprises a macromer.
4966. The device of item 4807, further comprising a polymeric carrier, wherein the polymeric carrier comprises a poly(ethylene glycol) polymer.
4967. The device of item 4807, further comprising a polymeric carrier, wherein the polymeric carrier comprises an amorphous polymer.
4968. The device of item 4807, further comprising a lubricious coating.
4969. The device of item 4807 wherein the anti-scarring agent is located within pores or holes of the device. 4970. The device of item 4807 wherein the anti-scarring agent is located within a channel, lumen, or divet of the device.
4971. The device of item 4807, further comprising a second pharmaceutically active agent.
4972. The device of item 4807, further comprising an antiinflammatory agent.
4973. The device of item 4807, further comprising an agent that inhibits infection.
4974. The device of item 4807, further comprising an agent that inhibits infection, wherein the agent is an anthracycline.
4975. The device of item 4807, further comprising an agent that inhibits infection, wherein the agent is doxorubicin.
4976. The device of item 4807, further comprising an agent that inhibits infection, wherein the agent is mitoxantrone.
4977. The device of item 4807, further comprising an agent that inhibits infection, wherein the agent is a fluoropyrimidine.
4978. The device of item 4807, further comprising an agent that inhibits infection, wherein the agent is 5-fluorouracil (5-FU).
4979. The device of item 4807, further comprising an agent that inhibits infection, wherein the agent is a folic acid antagonist. B Λ_a 2006/135479
4980. The device of item 4807, further comprising an agent that inhibits infection, wherein the agent is methotrexate.
4981. The device of item 4807, further comprising an agent that inhibits infection, wherein the agent is a podophylotoxin.
4982. The device of item 4807, further comprising an agent that inhibits infection, wherein the agent is etoposide.
4983. The device of item 4807, further comprising an agent that inhibits infection, wherein the agent is a camptothecin.
4984. The device of item 4807, further comprising an agent that inhibits infection, wherein the agent is a hydroxyurea.
4985. The device of item 4807,. further comprising an agent that inhibits infection, wherein the agent is a platinum complex.
4986. The device of item 4807, further comprising an agent that inhibits infection, wherein the agent is cisplatin.
4987. The device of item 4807, further comprising an antithrombotic agent.
4988. The device of item 4807, further comprising a visualization agent.
4989. The device of item 4807, further comprising a visualization agent, wherein the visualization agent is a radiopaque material, wherein the radiopaque material comprises a metal, a halogenated compound, or a barium containing compound. 4990. The device of item 4807, further comprising a visualization agent, wherein the visualization agent is a radiopaque material, wherein the radiopaque material comprises barium, tantalum, or technetium.
4991. The device of item 4807, further comprising a visualization agent, wherein the visualization agent is a MRI responsive material.
4992. The device of item 4807, further comprising a visualization agent, wherein the visualization agent comprises a gadolinium chelate.
4993. The device of item 4807, further comprising a visualization agent, wherein the visualization agent comprises iron, magnesium, manganese, copper, or chromium.
4994. The device of item 4807, further comprising a visualization agent, wherein the visualization agent comprises an iron oxide compound.
4995. The device of item 4807, further comprising a visualization agent, wherein the visualization agent comprises a dye, pigment, or colorant.
4996. The device of item 4807, further comprising an echogenic material.
4997. The device of item 4807, further comprising an echogenic material, wherein the echogenic material is in the form of a coating. 4998. The device of item 4807 wherein the device is sterile.
4999. The device of item 4807 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device.
5000. The device of item 4807 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is connective tissue.
5001. The device of item 4807 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is muscle tissue.
5002. The device of item 4807 wherein the anti-scarring agent is released into tissue in the vicinity .of the device after deployment of the device, wherein the tissue is nerve tissue.
5003. The device of item 4807 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is epithelium tissue.
5004. The device of item 4807 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from the time of deployment of the device to about 1 year.
5005. The device of item 4807 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from about 1 month to 6 months. 5006. The device of item 4807 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from about 1 - 90 days.
5007. The device of item 4807 wherein the anti-scarring agent is released in effective concentrations from the device at a constant rate.
5008. The device of item 4807 wherein the anti-scarring agent is released in effective concentrations from the device at an increasing rate.
5009. The device of item 4807 wherein the anti-scarring agent is released in effective concentrations from the device at a decreasing rate.
5010. The device of item 4807 wherein the anti-scarring agent is released in effective concentrations from the composition comprising the anti-scarring agent by diffusion over a period ranging from the time of deployment of the device to about 90 days.
5011. The device of item 4807 wherein the anti-scarring agent is released in effective concentrations from the composition comprising the anti-scarring agent by erosion of the composition over a period ranging from the time of deployment of the device to about 90 days.
5012. The device of item 4807 wherein the device comprises about 0.01 Dg to about 10 Dg of the anti-scarring agent.
5013. The device of item 4807 wherein the device comprises about 10 Dg to about 10 mg of the anti-scarring agent.
5014. The device of item 4807 wherein the device comprises about 10 mg to about 250 mg of the anti-scarring agent. 5015. The device of item 4807 wherein the device comprises about 250 mg to about 1000 mg of the anti-scarring agent.
5016. The device of item 4807 wherein the device comprises about 1000 mg to about 2500 mg of the anti-scarring agent.
5017. The device of item 4807 wherein a surface of the device comprises less than 0.01 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
5018. The device of item 4807 wherein a surface of the device comprises about 0.01 Dg to about 1 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
5019. The device of item 4807 wherein a surface of the device comprises about 1 Dg to about 10 Dg of the anti-scaning agent per mm2 of device surface to which the anti-scarring agent is applied.
5020. The device of item 4807 wherein a surface of the device comprises about 10 Dg to about 250 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
5021. The device of item 4807 wherein a surface of the device comprises about 250 Dg to about 1000 Dg of the anti-scarring agent of anti- scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
5022. The device of item 4807 wherein a surface of the device comprises about 1000 Dg to about 2500 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied. 5023. A method as in any one of items 4807-5022, wherein the device is a left ventricular assist device.
5024. A method as in any one of items 4807-5022, wherein the device is a right ventricular assist device.
5025. A method as in any one of items 4807-5022, wherein the device is a biventricular assist device.
5026. A method as in any one of items 4807-5022, wherein the device is a cardiac assist device.
5027. A method as in any one of items 4807-5022, wherein the device is a mechanical assist device.
5028. A method as in any one of items 4807-5022, wherein the device is an artificial cardiac assist device.
5029. A method as in any one of items 4807-5022, wherein the device is an implantable heart assist system.
5030. A method as in any one of items 4807-5022, wherein the device is a heart assist pump.
5031. A method as in any one of items 4807-5022, wherein the device is an intra-ventricular cardiac assist device.
5032. A device, comprising a spinal implant and an anti- scarring agent or a composition comprising an anti-scarring agent, wherein the agent inhibits scarring between the device and a host into which the device is implanted.
5033. The device of item 5032 wherein the agent is an adensosine A2A receptor antagonist. 13030
5034. The device of item 5032 wherein the agent is an AKT inhibitor.
5035. The device of item 5032 wherein the agent is an alpha 2 integrin antagonist, wherein the alpha 2 integrin antagonist is Pharmaprojects No. 5754 (Merck KgaA).
5036. The device of item 5032 wherein the agent an alpha 4 integrin antagonist.
5037. The device of item 5032 wherein the agent is an alpha 7 nicotinic receptor agonist.
5038. The device of item 5032 wherein the agent is an angiogenesis inhibitor selected from the group consisting of AG-12,958 (Pfizer), ATN-16-1 (Attention LLC), neovastat, an angiogenesis inhibitor from Jerina AG (Germany), NM-3 (Mercian), VGA-1155 (Taisho), FCE-26644 (Pfizer), FCE-26950 (Pfizer), FPMA (Meiji Daries), FR-111142 (Fujisawa), GGTI-298, GM-1306 (Ligand), GPA-1734 (Novartis), NNC-47-0011 (Novo Nordisk), herbamycin (Nippon Kayaku), lenalidomide (Celegene), IP-10 (NIH), ABT-828 (Abbott), KIN-841 (Tokushima University, Japan), SF-1126 (Semafore Pharmaceuticals), laminin technology (NIH), CHlR-258 (Chiron), NVP-AEW541 (Novartis), NVP-AEW541 (Novartis), Vt16907 (Alchemia), OXI-8007 (Oxigene), EG-3306 (Ark Therapeutics), Maspin (Arriva), ABT- 567 (Abbott), PPI-2458 (Praecis Pharmaceuticals), CC-5079, CC-4089 (Celgene), HIF-1 alpha inhibitors (Xenova), S-247 (Pfizer), AP-23573 (Ariad), AZD-9935 (Astra Zeneca), mebendazole (Introgen Therapeutics), MetAP-2 inhibitors (GlaxoSmithKline), AG-615 (Angiogene Pharmaceuticals), Tie-2 antagonists (Hybrigenics), NC-381 , CYC-381, NC-169, NC-219, NC-383, NC-384, NC-407 (Lorus Therapeutics), ATN-224 (Attenuon), ON-01370 (Onconova), Vitronectin antagonists (Amgen), SDX-103 (Salmedix), Vitronectin antagonists (Shire), CHP (Riemser), TEK (Amgen), Anecortave acetate (Alcon), T46.2 (Matrix Therapeutics), HG-2 (Heptagen), TEM antagonists (Genzyme), Oxi-4500 (Oxigene), ATN-161 (Attenuon), WX-293 (Wilex), M-2025 (Metris Therapeutics), Alphastatin (BioActa), YH-16 (Yantai Rongchang), BIBF-1120 (Boehringer Ingelheim), BAY-57-9352 (Bayer), AS- 1404 (Cancer Research Technology), SC-77964 (Pfizer), glycomimetics (BioTie Therapies), TIE-2 Inhibitors (Ontogen), DIMI, Octamer (Octamer), ABR-215050 (Active Biotech), ABT-518 (Abbott), KDR inhibitors (Abbott), BSF-466895 (Abbott), SCH-221153 (Schering-Plough), DAC:antiangiogenic (ConjuChem), TFPI (EntreMed), AZD-2171 (Astra-Zenaca), CDC-394 (Celgene), LY290293 (EIi Lilly), IDN-5390 (Indena), Kdr Kinase Inhibitors (Merck), CT-113020, CT-116433, CT-116563, CT-31890, CT-32228) (Cell Therapeutics), A-299620 (Abbott), TWEAK Inhibitor (Amgen), VEGF modulators (Johnson and Johnson), Tum-N53, tumstatin (Genzyme), Thios- 1 , Thios-2 (Thios Pharmaceuticals), MV-6401 (Miravant Medical Technologies), Spisulosine (PharmaMar), CEP-7055 (Cephalon), AUV-201 (Auvation), LM-609 (EIi Lilly), SKF-106615 (AnorMED), Oglufanide disodium (Cytran), BW-114 (Pharminox), Calreticulin (NIH), WX-678 (Wilex), SD-7784 (Pfizer), WX-UK1 (Wilex), SH-268 (Schering AG), 2-Me-PGA (Celgene), S- 137 (Pfizer), ZD-6126 (Angiogene Pharmaceuticals), SG-292 (SignalGen), Benefin (Lane Labs), A6, A36 (Angstrom), SB-2723005 (GlaxoSmithKline), SC-7 (Cell Therapeutics), ZEN-014 (AEterna Zentaris), 2-methoxyestradiol (EntreMed), NK-130119 (Nippon Kayaku), CC-10004 (Celgene), AVE- 8062A (Ajinomoto), Tacedinaline (Pfizer), Actinonin (Tokyo Metropolitan Institute of Medical Science), Lenalidomide (Celgene), VGA-1155, BTO-956 (SRI International), ER-68203-00 (Eisai), CT-6685 (UCB), JKC-362 (Phoenix Pharmaceuticals), DMI-3798 (DMI Biosciences, Angiomate (Ipsen), ZD-6474 (AstraZeneca), CEP-5214 (Cephalon), Canstatin (Genzyme), NM-3 (Mercian), Oridigm (Med iQuest Therapeutics), Exherin (Adherex), BLS-0597 (Boston Life Sciences), PTC-299 (PTC Therapeutics), NPI-2358 (Nereus Pharmaceuticals), CGP-79787 (Novartis), AEE-788 (Novartis), CKD-732 (Chong Kun Dang), CP-564959 (OSI Pharmaceuticals), CM-101 (CarboMed), CT-2584, CT3501 (Cell Therapeutics), combretastatin and analogues and derivatives thereof (Oxigene), Rebimastat (Bristol-Meyers Squibb), Dextrin 2-sulfate (ML Laboratories), Cilengitide (Merk KGaA), NSC-706704 (Pharminox), KRN- 951 (Kirin Brewery), Ukrain, NSC-631570 (Nowicky Pharma), Tecogalan sodium (Daiichi Pharmaceutical), Tz-93 (Tsumura), TBC-1635 (Encysive Pharmaceuticals), TAN-1120 (Takeda), Semaxanib (Pfizer), BDI-7800 (Biopharmacopae), SD-186, SD-983 (Bristol-Meyers Squibb), SB-223245 (GlaxoSmithKline), SC-236 (Pfizer), RWJ-590973 (Johnson and Johnson), ILX-1850 (Genzyme), SC-68488, S-836 (Pfizer), CG-55069-11 (CuraGen), Ki-23057 (Kirin Brewery), CCX-700 (Chemoentryx), Pegaptanib octasodium (Giled Sciences), ANGIOCOL (available from Biostratum Inc.), or an analogue or derivative thereof.
5039. The device of item 5032 wherein the agentis an apoptosis antagonist.
5040. The device of item 5032 wherein the agent is an apoptosis activator.
5041. The device of item 5032 wherein the agent is a beta 1 integrin antagonist.
5042. The device of item 5032 wherein the agent is a beta tubulin inhibitor.
5043. The device of item 5032 wherein the agent is a blocker of enzyme production in Hepatitis C. 5044. The device of item 5032 wherein the agent is a Bruton's tyrosine kinase inhibitor.
5045. The device of item 5032 wherein the agent is a calcineurin inhibitor.
5046. The device of item 5032 wherein the agent is a caspase 3 inhibitor.
5047. The device of item 5032 wherein the agent is a CC chemokine receptor antagonist.
5048. The device of item 5032 wherein the agent is a cell cycle inhibitor selected from the group consisting of SNS-595 (Sunesis), synthadotin, KRX-0403, and an analogue or derivative thereof.
5049. The device of item 5032 wherein the agent is a cathepsin B inhibitor.
5050. The device of item 5032 wherein the agent is a cathepsin K inhibitor, wherein the cathepsin K inhibitor is 462795 (GlaxoSmithKline), INPL-022-D6 (Amura Therapeutics), an analogue or derivative thereof).
5051. The device of item 5032 wherein the agent is a cathepsin L inhibitor.
5052. The device of item 5032 wherein the agent is a CD40 antagonist. 5053. The device of item 5032 wherein the agent is a chemokine receptor agonist.
5054. The device of item 5032 wherein the agent is a chymase inhibitor.
5055. The device of item 5032 wherein the agent is a collagenase antagonist.
5056. The device of item 5032 wherein the agent is a CXCR antagonist.
5057. The device of item 5032 wherein the agent is a cyclin dependent kinase inhibitor selected from the group consisting of a CDK-1 inhibitor, a CDK-2 inhibitor, a CDK- 4 inhibitor, a CDK-6 inhibitor, a CAK1 inhibitor from GPC Biotech and Bristol-Myers Squibb, RGB-286199 (GPC Biotech), an anticancer agent from Astex Technology, a CAK1 inhibitor from GPC Biotech, a CDK inhibitor from Sanofi-Aventis, a CDK1/CDK2 inhibitor from Amgen, a CDK2 inhibitor from SUGEN-2 (Pfizer), a hearing loss therapy agent (Sound Pharmaceuticals), PD-0332991 (Pfizer), RGB-286199 (GPC Biotech), Ro-0505124 (Hoffmann-La Roche), a Ser/Thr kinase inhibitor from Lilly (EIi Lilly), CVT-2584 (CAS No. 199986-75-9) (CV Therapeutics), and an analogue or derivative thereof.
5058. The device of item 5032 wherein the agent is a cyclooxygenase 1 inhibitor.
5059. The device of item 5032 wherein the agent is a DHFR inhibitor. 5060. The device of item 5032 wherein the agent is a dual integrin inhibitor.
5061. The device of item 5032 wherein the agent is an elastase inhibitor.
5062. The device of item 5032 wherein the agent is an elongation factor-1 alpha inhibitor.
5063. The device of item 5032 wherein the agent is an endothelial growth factor antagonist.
5064. The device of item 5032 wherein the agent is an endothelial growth factor receptor kinase inhibitor selected from the group consisting of sorafenib tosylate (Bayer), AAL-993 (Novartis), ABP-309 (Novartis), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), E- 7080 (Eisai), EG-3306 (Ark Therapeutics), EXEL-2880 (Exelixis), GW- 654652 (GlaxoSmithK)ine), a KDR inhibitor from LG Life Sciences, CT-6685 and CT-6729 (UCB), KRN-633 and KRN-951 (Kirin Brewery), OSI-930 (OSI Pharmaceuticals), SP-5.2 (Supratek Pharma), SU-11657 (Pfizer), a Tie-2 antagonist (Hybrigenics), a VEGFR-2 kinase inhibitor (Bristol-Myers Squibb), XL-647 (Exelixis), a KDR inhibitor from Abbott Laboratories, sorafenib tosylate, and an analogue or derivative thereof.
5065. The device of item 5032 wherein the agent is an endotoxin antagonist.
5066. The device of item 5032 wherein the agent is an epothilone and tubulin binder. 5067. The device of item 5032 wherein the agent is an estrogen receptor antagonist.
5068. The device of item 5032 wherein the agent is an FGF inhibitor.
5069. The device of item 5032 wherein the agent is a farnexyl transferase inhibitor.
5070. The device of item 5032 wherein the agent is farnesyltransferase inhibitor selected from the group of A-197574 (Abbott), a famesyltransferase inhibitor from Servier, FPTIII (Strathclyde Institute for Drug R), LB-42908 (LG Life Sciences), Pharmaprojects No. 5063 (Genzyme), Pharmaprojects No. 5597 (Ipsen), Yissum Project No. B-1055 (Yissum), and an analogue or derivative thereof.
5071. The device of item 5032 wherein the agent is an FLT-3 kinase inhibitor.
5072. The device of item 5032 wherein the agent is an FGF receptor kinase inhibitor.
5073. The device of item 5032 wherein the agent is a fibrinogen antagonist selected from the group consisting of AUV-201 (Auvation), MG-13926 (Sanofi-Aventis), plasminogen activator (CAS No. 105913-11-9) (from Sanofi-Aventis or UCB), plasminogen activator-2 (tPA- 2) (Sanofi-Aventis), pro-urokinase (CAS No. 82657-92-9) (Sanofi-Aventis), and an analogue or derivative thereof.
5074. The device of item 5032 wherein the agent is a heat shock protein 90 antagonist selected from the group consisting of SRN-005 (Sirenade), geldanamycin, NSC-33050 (17-allylaminogeldanamycin; 17- AAG), ^-dimethylaminoethylamino-^-demethoxy-geldanamycin (17- DMAG), rifabutin (rifamycin XIV, 1',4-didehydro-1-deoxy-1 ,4-dihydro-5'-(2- methylpropyl)-1-oxo-), and an analogue or derivative thereof.
5075. The device of item 5032 wherein the agent is a histone deacetylase inhibitor.
5076. The device of item 5032 wherein the agent is an HMGCoA reductase inhibitor selected from the group consisting of an atherosclerosis therapeutic from Lipid Sciences, ATI-16000 (ARYx Therapeutics), KS-01-019 (Kos Pharmaceuticals), Pharmaprojects No. 2197 (Sanofi-Aventi), RP 61969 (Sanofi-Aventis), and an analogue or derivative thereof.
5077. The device of item 5032 wherein the agent is an ICAM inhibitor.
5078. The device of item 5032 wherein the agent is an IL, ICE and IRAK antagonist, wherein the antagonist is a CJ-14877, CP-424174 (Pfizer), NF-61 (Negma-Lerads), and an analogue or derivative thereof.
5079. The device of item 5032 wherein the agent is an IL-2 inhibitor.
5080. The device of item 5032 wherein the agent is an immunosuppressant selected from the group consisting of teriflunomide (Sanofi Aventis), chlorsulfaquinoxalone (NSC-339004), chlorsulfaquinoxalone sulfate, CS-712 (Sankyo), ismomultin alfa (CAS No. 457913-93-8) (Akzo Nobel), antiallergics from GenPat77, antiinflammatories or AT-005 (Androclus Therapeutics), autoimmune disease therapeutics from EpiVax, BN-007 (Bone), budesonide (CAS No. 51333-22- 3) (MAP Pharmaceuticals), CO-14 (Genzyme), edratide (CAS No. 433922- 67-9) (Teva), EP-314 (Enanta), eprovafen (CAS No. 101335-99-3) (Sanofi- Aventis), HWA-131 (CAS No. 118788-41-3) (Sanofi-Aventis), immunomodulators from MerLion Pharmaceuticals, immunosuppressives from Alchemia, IPL-12 (Inflazyme), MDL-9563 (CAS No. 27086-86-8) (Sanofi-Aventis), Pharmaprojects No. 2330 (Sanofi-Aventis), Pharmaprojects No. 6426 (Abgenix), PXS-25 (Pharmaxis), rosmarinic acid (CAS No. 20283-92-5) (Sanofi-Aventis), RP 42927 or RP 54745 (CAS No. 135330-08-4) (Sanofi-Aventis), SGN-35 (Seattle Genetics), ST-1959 (Sigma-Tau), type I diabetes therapy from SYNX Pharma, UNIL-88 (Debiopharm), VP-025 (Vasogen), VR-694 (Vectura), PRTX-001 (Protalex), and an analogue or derivative thereof.
5081. The device of item 5032 wherein the agent is an IMPDH (inosine monophosphate). _ -
5082. The device of item 5032 wherein the agent is an integrin antagonist.
5083. The device of item 5032 wherein the agent is an interleukin antagonist.
5084. The device of item 5032 wherein the agent is an inhibitor of type III receptor tyrosine kinase.
5085. The device of item 5032 wherein the agent is an irreversible inhibitor of enzyme methionine aminopeptidase type 2.
5086. The device of item 5032 wherein the agent is an isozyme selective delta protein kinase C inhibitor. 5087. The device of item 5032 wherein the agent a JAK3 enzyme inhibitor.
5088. The device of item 5032 wherein the agent is a JNK inhibitor.
5089. The device of item 5032 wherein the agent is a kinase inhibitor.
5090. The device of item 5032 wherein the agent is kinesin antagonist.
5091. The device of item 5032 wherein the agent is a kinesin antagonist.
5092. The device of item 5032 wherein the agent is a leukotriene inhibitor and antagonist selected from the group consisting of ambicromil (CAS No. 58805-38-2) (Sanofi-Aventis), amelubant (CAS No. 346735-24-8) (Boehringer Ingelheim), DW-1141 (Dong Wha), ebselen (Daiichi Pharmaceutical), ibudilast (Kyorin), leucotriene inhibitors from Sanofi-Aventis, lymphotoxin-beta receptor (LT-D) from Biogen Idee, Pharmaprojects No. 1535 and 2728 (CAS No. 119340-33-9) (Sanofi- Aventis), R-112 (Rigel), Rev-5367 (CAS No. 92532-05-3) (Sanofi-Aventis), RG-14893 (CAS No. 141835-49-6) (Sanofi-Aventis), RG-5901-A (CAS No. 101910-24-1), 92532-23-5, RP 66153 (CAS No. 142422-79-5), RP 66364 (CAS No.186916-92-5), RP 69698 (CAS No. 141748-00-7) (Sanofi-Aventis), SC-411930 (Pfizer), SC-41930 (CAS No. 120072-59-5) (Pfizer), SC-50605 (CAS No. 138828-39-4) (Pfizer), SC-51146 (CAS No. 141059-52-1), SC- 53228 (CAS No. 153633-01-3) (Pfizer), spaglumic acid (ZY-15106) (CAS No.3106-85-2), 80619-64-3 (Novartis), tipredane (CAS No. 85197-77-9) (Bristol-Myers Squibb), U-75302 (CAS No. 119477-85-9) (Pfizer), and analogue or derivative thereof.
5093. The device of item 5032 wherein the agent is an MAP kinase inhibitor.
5094. The device of item 5032 wherein the agent is a matrix metalloproteinase inhibitor.
5095. The device of item 5032 wherein the agent is an MCP- CCR2 inhibitor.
5096. The device of item 5032 wherein the agent is an mTOR inhibitor.
5097. The device of item 5032 wherein the agent is an mTOR kinase inhibitor.
5098. The device of item 5032 wherein the agent is a microtubule inhibitor selected from the group consisting of antibody- maytansinoid conjugates from Biogen Idee, colchicines (MantiCore Pharmaceuticals), anticancer immunoconjugates from Johnson & Johnson, DIME from Octamer, gni-1f (GNI), huC242-DM4, huMy9-6-DM1 (ImmunoGen), IDN-5404 (Indena), IMO-098, IMOderm (Imotep), mebendazole (Introgen Therapeutics), microtubule poisons from Cambridge Enterprise, paciitaxel such as LOTAX from Aphios (CAS No. 33069-62-4), Genexol-PM from Samyang, Pharmaprojects No. 6383 (Tapestry Pharmaceuticals), RPR-112378 (Sanofi-Aventis), SGN-75 (Seattle Genetics), SPL-7435 (Starpharma), SSR-250411 (Sanofi-Aventis), trastuzumab-DM1 (Genentech), vinorelbine, and an analogue or derivative thereof. 5099. The device of item 5032 wherein the agent is an MIF inhibitor.
5100. The device of item 5032 wherein the agent is an MMP inhibitor.
5101. The device of item 5032 wherein the agent is a neurokinin (NK) antagonist selected from the group consisting of anthrotainin (CAS No. 148084-40-6) (Sanofi-Aventis), an IBS thereapeutic from ArQuIe, MDL-105212A (CAS No. 167261-60-1) (Ssanofi-Aventis), Pharmaprojects No. 2744, 3258 (CAS No. 139167-47-8) 4006, 4201, or 5986 (Sanofi-Aventis), RP 67580 (CAS No. 135911-02-3), SR-144190 (CAS No. 201152-86-5), SSR-240600, SSR-241586 (Sanofi-Aventis), TKA-457 (Novartis), vestipitant mesylate (CAS No. 334476-64-1) (GlaxoSmithKline), Win-64821 (Sanofi-Aventis), PRX-96026 (Predix Pharmaceuticals), and an analogue or.derivative thereof . . . . ... . . .
5102. The device of item 5032 wherein the agent is an NF kappa B inhibitor selected from the group consisting of AVE-0545 or AVE- 0547 (Sanofi-Aventis), bortezomib (CAS No. 179324-69-7) (Millennium Pharmaceuticals), dexanabinol (CAS No. 112924-45-5) (Pharmos), dexlipotam (Viatris), Pharmaprojects No. 6283 (INDRA) (OXiGENE), IPL- 576092 (CAS No. 137571-30-3) (Jnflazyme), NFKB decoy (Corgentech), NFKB decoy oligo (AnGes MG), NFKB's from Ariad, osteoporosis treatments or S5 (F005) from Fulcrum Pharmaceuticals, P61 (Phytopharm), R-flurbiprofen (CAS No. 5104-49-4) (Encore Pharmaceuticals), and an analogue or derivative thereof.
5103. The device of item 5032 wherein the agent is a nitric oxide agonist. 5104. The device of item 5032 wherein the agent is an ornithine decarboxylase inhibitor.
5105. The device of item 5032 wherein the agent is a p38 MAP kinase inhibitor selected from the group consisting of AZD-6703 (AstraZeneca), JX-401 (Jexys Pharmaceuticals), BMS-2 (Bristol-Myers Squibb), a p38 MAP kinase inhibitor from Novartis, a p38-alpha MAP kinase inhibitor from Amphora, Pharmaprojects No. 5704 (Pharmacopeia), RPR- 200765A (Sanofi-Aventis), SD-282 (Johnson & Johnson), TAK-715 (Takeda), and an analogue or derivative thereof.
5106. The device of item 5032 wherein the agent is a palmitoyl-protein thioesterase inhibitor.
5107. The device of item 5032 wherein the agent is a PDGF receptor kinase inhibitor selected from the group consisting of AAL-993, AMN-107, or ABP-309 (Novartis), AMG-706 (Amgen), BAY-57-9352 (Bayer), CDP-860 (UCB), E-7080 (Eisai), imatinib (CAS No. 152459-95-5) (Novartis), OSI-930 (OSI Pharmaceuticals), RPR-127963E (Sanofi-Aventis), RWJ-540973 (Johnson & Johnson), sorafenib tosylate (Bayer), SU-11657 (Pfizer), tandutinib (CAS No. 387867-13-2) (Millennium Pharmaceuticals), vatalanib (Novartis), ZK-CDK (Schering AG), and an analogue or derivative thereof.
5108. The device of item 5032 wherein the agent is (-)- halofenate (Metabolex), AMG-131 (Amgen), antidiabetics from Japan Tobacco, AZD-4619, AZD-8450, AZD-8677 (AstraZeneca), DRF-10945, balaglitazone (Dr Reddy's), CS-00088, CS-00098 (Chipscreen Biosciences), E-3030 (Eisai), etalocib (CAS No. 161172-51-6) (EIi Lilly), GSK-641597 (Ligand), GSK-677954 (GlaxoSmithKline), GW-409544 (Ligand), GW- 590735 (GlaxoSmithKline), K-111 (Hoffmann-La Roche), LY-518674 (EIi Lilly), LY-674 (Ligand), LY-929 (Ligand), MC-3001, MC-3002 (MaxoCore Pharmaceuticals), metformin HCI + pioglitazone (CAS No. 1115-70-4 and 112529-15-4), ACTOPLUS MET from Andrx), muraglitazar (CAS No. 331741-94-7) (Bristol-Myers Squibb), naveglitazar (Ligand), oleoylethanolamide (Kadmus Pharmaceuticals), ONO-5129, pioglitazone hydrochloride (CAS No. 111025-46-8 and 112529-15-4) (Takeda), PLX-204 (Plexxikon), PPAR agonists from Genfit, PPAR delta agonists from EIi Lilly, PPAR-alpha agonists from CrystalGenomics, PPAR-gamma modulators and PPAR-β modulators from CareX, rosiglitazone maleate (CAS No. 122324- 73-4 or 155141-29-0) (GlaxoSmithK)ine), rosiglitazone maleate/glimepir (CAS No. 155141-29-0 and 93479-97-1), AVANDARYL, rosiglitazone maleate/metformin extend (CAS No. 155141-29-0 and 657-24-9), AVANDAMET, rosiglitazone maleate+metformin, AVANDAMET (GlaxoSmithKline), tesaglitazar (AstraZeneca), and an analogue or derivative thereof.
5109. The device of item 5032 wherein the agent is a phosphatase inhibitor.
5110. The device of item 5032 wherein the agent is a phosphodiesterase (PDE) inhibitor selected from the group consisting of avanafil (Tanabe Seiyaku), dasantafil (CAS No. 569351-91-3) (Schering- Plough), A-906119 (CAS No. 134072-58-5), DL-850 (Sanofi-Aventis), GRC- 3015, GRC-3566, GRC-3886 (Glenmark), HWA-153 (CAS No. 56395-66-5) (Sanofi-Aventis), hydroxypumafentrine (Altana), IBFB-130011, IBFB-14-016, IBFB-140301 , IBFB-150007, IBFB-211913 (IBFB Pharma), L-826141 (Merck & Co), medorinone (CAS No. 88296-61-1) (Sanofi-Aventis), MEM- 1917 (Memory Pharmaceuticals), ND-1251 (Neuro3d), PDE inhibitors from ICOS, PDE IV inhibitors from Memory Pharmaceuticals and CrystalGenomics, Pharmaprojects No. 2742 and 6141 (Sanofi-Aventis), QAD-171 (Novartis), RHC-2963 (CAS No. 76993-12-9 and 76993-14-1), RPR-117658, RPR-122818 derivatives, SR-24870 , and RPR-132294 (Sanofi-Aventis), SK-350 (ln2Gen), stroke therapy agents from deCODE Genetics, TAS-203 (Taiho), tofimilast (CAS No. 185954-27-2) (Pfizer), UK- 371800 (Pfizer), WIN-65579 (CAS No. 158020-82-7) (Sanofi-Aventis), IBFB- 130020 (IBFB Pharma), OPC-6535 (CAS No. 145739-56-6) (Otsuka), a phosphodiesterase III inhibitor, enoximone, a phosphodiesterase IV inhibitor, fosfosal, Atopik (Barrier Therapeutics), triflusal, a phosphodiesterase V inhibitor, and an analogue or derivative thereof.
5111. The device of item 5032 wherein the agent is a PKC inhibitor.
5112. The device of item 5032 wherein the agent is a platelet activating factor antagonist.
5113. The device of item 5032 wherein the agent is a platelet- derived growth factor receptor kinase inhibitor.
5114. The device of item 5032 wherein the agent is a prolyl hydroxylase inhibitor.
5115. The device of item 5032 wherein the agent is a polymorphonuclear neutrophil inhibitor.
5116. The device of item 5032 wherein the agent is a protein kinase B inhibitor.
5117. The device of item 5032 wherein the agent is a protein kinase C stimulant. 5118. The device of item 5032 wherein the agent is a purine nucleoside analogue.
5119. The device of item 5032 wherein the agent is a purinoreceptor P2X antagonist.
5120. The device of item 5032 wherein the agent is a Raf kinase inhibitor.
5121. The device of item 5032 wherein the agent is a reversible inhibitor of ErbB1 and ErbB2.
5122. The device of item 5032 wherein the agent is a ribonucleoside triphosphate reductase inhibitor.
5123. The device of jteni 5032 wherein the agent is an SDF-1 antagonist.
5124. The device of item 5032 wherein the agent is a sheddase inhibitor.
5125. The device of item 5032 wherein the agent is an SRC inhibitor.
5126. The device of item 5032 wherein the agent is a stromelysin inhibitor.
5127. The device of item 5032 wherein the agent is an Syk kinase inhibitor. 5128. The device of item 5032 wherein the agent is a telomerase inhibitor.
5129. The device of item 5032 wherein the agent is a TGF beta inhibitor selected from the group consisting of pirfenidone (CAS No. 53179-13-8) (MARNAC)1 tranilast (CAS No. 53902-12-8) (Kissei), IN-1130 (ln2Gen), mannose-6-phosphate (BTG), TGF-β antagonists from Inflazyme (Pharmaprojects No. 6075), TGF-β antagonists from Sydney, non-industrial source), TGF-β I receptor kinase inhibitors from EIi Lilly, TGF-β receptor inhibitors from Johnson & Johnson, and an analogue or derivative thereof.
5130. The device of item 5032 wherein the agent is a TNFα antagonist or TACE inhibitor selected from the group consisting of adalimumab (CAS No. 331731-18-1) (Cambridge Antibody Technology), AGIX-4207 (AtheroGenics), AGT-1 (Advanced Biotherapy), an antiinflammatory from Borean Pharma, Cellzome, or Paradigm Therapeutics, anti-inflammatory vaccine (TNF-alpha kinoid) from Neovacs, humanized anti-TNF antibody or an anti-TNF MAb (CB0006) Celltech (UCB), apratastat (CAS No. 287445-51-0) (Wyeth), BMS-561392 (Bristol-Myers Squibb), BN- 006 (Bone), certolizumab pegol (CAS No. 428863-50-7 or CH-138 (UCB), cilomilast (CAS No. 153259-65-5) (GlaxoSmithKline), CR-1 (Nuada Pharmaceuticals), CRx-119 (CombinatoRx), D-5410 (UCB), dacopafant (CAS No. 125372-33-0) (Sanofi-Aventis), dersalazine (CAS No. 188913-57- 7/188913-58-8) (Uriach), etanercept (CAS No. 185243-69-0) (Amgen), ethyl pyruvate (Critical (Critical Therapeutics), golimumab (CAS No. 476181-74-5) (Johnson & Johnson), hormono-immunotherapy from Ipsen, CDP571 (e.g., Humicade from UCB), IC-485 (ICOS), infliximab (CAS No. 170277-31-3) (Johnson & Johnson), IP-751 (Manhattan Pharmaceuticals), ISIS-104838 (CAS No. 250755-32-9) (ISIS Pharmaceuticals), lenalidomide (CAS No. 191732-72-6) (Celgene), lentinan (CAS No. 37339-90-5) (Ajinomoto), MDL- 201112 (CAS No. 142130-73-2) (Sanofi-Aventis), medroxyprogesterone (CAS No. 520-85-4) (InKine Pharmaceutical), N-acetylcysteine (CAS No. 616-91-1) (Zambon), NBE-P2 (DIREVO Biotech), nerelimomab (CAS No. 162774-06-3) (Chiron), OM-294DP (OM PHARMA), onercept (CAS No. 199685-57-9) (Yeda), PASSTNF-alpha (Verigen), pentoxifylline or oxypentifylline (Sanofi-Aventis), Pharmaprojects No. 4091 , 4241, 4295, or 4488 (Sanofi-Aventis), Pharmaprojects No. 5480 (Amgen), Pharmaprojects No. 6749 (Cengent), pirfenidone (CAS No. 53179-13-8) (MARNAC), RPR- 132294 (Sanofi-Aventis), S5 (F002) (Fulcrum Pharmaceuticals), simvastatin (CAS No. 79902-63-9) (Merck & Co), STA-6292 (Synta Pharmaceuticals), tacrolimus (CAS No. 104987-11-3) (Fujisawa LifeCycle Pharma), talactoferrin alfa (CAS No. 308240-58-6) (Agennix), thalidomide (CAS No. 50-35-1) (Celgene), TNF antagonists form ProStrakan, and Synergen, TNF inhibitors (Amgen), TNF-alpha antagonists from Dynavax Technologies and Jerina AG (Germany), TNF-alpha inhibitors from IBFB Pharma and Xencor (Xencor), torbafylline (CAS No. 105102-21-4) (Sanofi-Aventis), UR-1505 (Uriach), VT-346 (Viron Therapeutics), YSIL6 (Y1S Therapeutics), YSTH2 (Y's Therapeutics), NPI-1302a-3 (Nereus Pharmaceuticals, a TNF antagonist from Jerina AG (Germany), dersalazine, and an analogue or derivative thereof.
5131. The device of item 5032 wherein the agent is a Toll receptor inhibitor.
5132. The device of item 5032 wherein the agent is a tubulin antagonist.
5133. The device of item 5032 wherein the agent is a tyrosine kinase inhibitor selected from the group consisting of SU-011248, SUTENT from Pfizer Inc. (New York, NY), BMS-354825, PN-355 (Paracelsian Pharmaceuticals), AGN-199659 (Allergan), AAL-993 or ABP-309 (Novartis), adaphostin (NIH), AEE-788 (Novartis), AG-013736 (OSI Pharmaceuticals), AG-13736 (Pfizer), ALT-110 (Alteris Therapeutics), AMG-706 (Amgen), anticancer MAbs from Xencor, anti-EGFrvlll MAbs from Abgenix, anti-HER2 MAb from Abiogen, AZD-2171 or AZD-9935 (AstraZeneca), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), CEP-5214 (Cephalon), CEP- 7055 (Cephalon), cetuximab (ImCIone Systems), CHIR-200131 and CHIR- 258 (Chiron), CP-547632 (OSI Pharmaceuticals), CP-724714 (Pfizer), CT- 301 (Creabilis Therapeutics), D-69491 (Baxter International), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EGFR/ErbB2 inhibitors from Array BioPharma, erlotinib (CAS No. 183319-69-9) (OSI Pharmaceuticals), EXEL-2880 (Exelixis), FK-778 (Sanofi-Aventis) , gefitinib (CAS No. 184475-35-2) (AstraZeneca), GW-2286 or GW-654652 (GlaxoSmithKline), her2/neu antigen from AlphaVax, HER-2/neu inhibitor from Generex, Herzyme (Medipad) (Sima Therapeutics), HKI-272 (Wyeth), HuMax-EGFr (Genmab), idronoxil (CAS No. 81267-65-4) (Novogen), IGF-1 inhibitors from Ontogen, 1MC-11F8 (ImCIone Systems), kahalalide F (CAS No. 149204-42-2) (PharmaMar), KDR inhibitor from LG Life Sciences, KDR inhibitors from Abbott Laboratories, KDR kinase inhibitors (UCB), Kdr kinase inhibitors from Merck & Co, KRN-633 and KRN-951 (Kirin Brewery), KSB-102 (Xenova), lapatinib ditosylate (CAS No. 388082-78-8) (GlaxoSmithKline), matuzumab (Merck KGaA), MDX-214 (Medarex), ME-103 (Pharmexa), MED-A300 (Gerolymatos), MNAC-13 (Lay Line Genomics), nimotuzumab (Center of Molecular Immunology), NSC-330507 or NSC-707545 (NIH), NV-50 (Novogen), OSI-930 (OSl Pharmaceuticals), panitumumab (Abgenix), pelitinib (CAS No. 287933-82-7) (Wyeth), pertuzumab (CAS No. 380610-27- 5) (Genentech), Pharmaprojects No. 3985 (Sanofi-Aventis), prostate cancer therapeutics from Sequenom (SQPC35, SQPC36, SQPC90), removab and remoxab (Trion Pharma), RG-13022 (CAS No. 136831-48-6), RG-13291 (CAS No. 138989-50-1), or RG-14620 (CAS No. 136831-49-7) (Sanofi- Aventis), RM-6427 (Romark), RNAi breast cancer therapy from Benitec, RP 53801 (CAS No. 125882-88-4) (Sanofi-Aventis), sorafenib tosylate (Bayer), SU-11657 (Pfizer), Tie-2 antagonists from Semaia (Hybrigenics), Tie-2 inhibitors from Ontogen, trastuzumab (CAS No. 180288-69-1) (Genentech), tyrosine kinase inhibitors from Sanofi-Aventis, U3-1287, U3-1565, U3-1784, U3-1800 (U3 Pharrna), vatalanib (Novartis), VEGFR-2 kinase inhibitor from Bristol-Myers Squibb, XL-647 (Exelixis), ZD-6474 (AstraZeneca), ZK-CDK (Schering AG)1 an EGFR tyrosine kinase inhibitor, EKB-569 (Wyeth), and an analogue or derivative thereof.
5134. The device of item 5032 wherein the agent is a VEGF inhibitor.
5135. The device of item 5032 wherein the agent is a vitamin D receptor agonist.
5136. The device of item 5032 wherein the agent is ZD-6474 (an angiogenesis inhibitor).
5137. The device of item 5032 wherein the agent is AP-23573 (an mTOR inhibitor).
5138. The device of item 5032 wherein the agent is synthadotin (a tubulin antagonist).
5139. The device of item 5032 wherein the agent is S-0885 (a collagenase inhibitor).
5140. The device of item 5032 wherein the agent is aplidine (an elongation factor-1 alpha inhibitor).
5141. The device of item 5032 wherein the agent is ixabepilone (an epithilone). 5142. The device of item 5032 wherein the agent is IDN-5390 (an angiogenesis inhibitor).
5143. The device of item 5032 wherein the agent is SB- 2723005 (an angiogenesis inhibitor).
5144. The device of item 5032 wherein the agent is ABT-518 (an angiogenesis inhibitor).
5145. The device of item 5032 wherein the agent is combretastatin (an angiogenesis inhibitor).
5146. The device of item 5032 wherein the agent is anecortave acetate (an angiogenesis inhibitor).
5147. The device of item 5032 wherein ib& agent is SB- 715992 (a kinesin antagonist).
5148. The device of item 5032 wherein the agent is temsirolimus (an mTOR inhibitor).
5149. The device of item 5032 wherein the agent is adalimumab (a TNFα antagonist).
5150. The device of item 5032, further comprising a polymer.
5151. The device of item 5032 wherein the anti-scarring agent inhibits adhesion between the device and a host into which the device is implanted. 5152. The device of item 5032 wherein the device delivers the anti-scarring agent locally to tissue proximate to the device.
5153. The device of item 5032, further comprising a coating, wherein the coating comprises the anti-scarring agent.
5154. The device of item 5032, further comprising a coating, wherein the coating is disposed on a surface of the device.
5155. The device of item 5032, further comprising a coating, wherein the coating directly contacts the device.
5156. The device of item 5032, further comprising a coating, wherein the coating indirectly contacts the device.
5157. The device of item 5032, /further comprising a coating, wherein the coating partially covers the device.
5158. The device of item 5032, further comprising a coating, wherein the coating completely covers the device.
5159. The device of item 5032, further comprising a coating, wherein the coating is a uniform coating.
5160. The device of item 5032, further comprising a coating, wherein the coating is a non-uniform coating.
5161. The device of item 5032, further comprising a coating, wherein the coating is a discontinuous coating. 5162. The device of item 5032, further comprising a coating, wherein the coating is a patterned coating.
5163. The device of item 5032, further comprising a coating, wherein the coating has a thickness of 100 Dm or less.
5164. The device of item 5032, further comprising a coating, wherein the coating has a thickness of 10 Dm or less.
5165. The device of item 5032, further comprising a coating, wherein the coating adheres to the surface of the device upon deployment of the device.
5166. The device of item 5032, further comprising a coating, wherein the coating is stable at room temperature for a period of 1 year.
5167. The device of item 5032, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 0.0001% to about 1% by weight.
5168. The device of item 5032, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 1% to about 10% by weight.
5169. The device of item 5032, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 10% to about 25% by weight.
5170. The device of item 5032, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 25% to about 70% by weight. US2006/013030
5171. The device of item 5032, further comprising a coating, wherein the coating further comprises a polymer.
5172. The device of item 5032, further comprising a first coating having a first composition and the second coating having a second composition.
5173. The device of item 5032, further comprising a first coating having a first composition and the second coating having a second composition, wherein the first composition and the second composition are different.
5174. The device of item 5032, further comprising a polymeric carrier, wherein the polymeric carrier comprises a copolymer.
5175. The device of item 5032, further- comprising a polymeric . carrier, wherein the polymeric carrier comprises a block copolymer.
5176. The device of item 5032, further comprising a polymeric carrier, wherein the polymeric carrier comprises a random copolymer.
5177. The device of item 5032, further comprising a polymeric carrier, wherein the polymeric carrier comprises a biodegradable polymer.
5178. The device of item 5032, further comprising a polymeric carrier, wherein the polymeric carrier comprises a non-biodegradable polymer.
5179. The device of item 5032, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrophilic polymer. 5180. The device of item 5032, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrophobic polymer.
5181. The device of item 5032, further comprising a polymeric carrier, wherein the polymeric carrier comprises a polymer having hydrophilic domains.
5182. The device of item 5032, further comprising a polymeric carrier, wherein the polymeric carrier comprises a polymer having hydrophobic domains.
5183. The device of item 5032, further comprising a polymeric carrier, wherein the polymeric carrier comprises a non-conductive polymer.
5184. The device of item 5032, further comprising a polymeric carrier, wherein the polymeric carrier comprises an elastomer.
5185. The device of item 5032, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrogel.
5186. The device of item 5032, further comprising a polymeric carrier, wherein the polymeric carrier comprises a silicone polymer.
5187. The device of item 5032, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrocarbon polymer.
5188. The device of item 5032, further comprising a polymeric carrier, wherein the polymeric carrier comprises a styrene-derived polymer.
5189. The device of item 5032, further comprising a polymeric carrier, wherein the polymeric carrier comprises a butadiene polymer. US2006/013030
5190. The device of item 5032, further comprising a polymeric carrier, wherein the polymeric carrier comprises a macromer.
5191. The device of item 5032, further comprising a polymeric carrier, wherein the polymeric carrier comprises a poly(ethylene glycol) polymer.
5192. The device of item 5032, further comprising a polymeric carrier, wherein the polymeric carrier comprises an amorphous polymer.
5193. The device of item 5032, further comprising a lυbricioυs coating.
5194. The device of item 5032 wherein the anti-scarring agent is located within pores or holes of the device.
5195. The device of item 5032 wherein the anti-scarring agent is located within a channel, lumen, or divet of the device.
5196. The device of item 5032, further comprising a second pharmaceutically active agent.
5197. The device of item 5032, further comprising an antiinflammatory agent.
5198. The device of item 5032, further comprising an agent that inhibits infection.
5199. The device of item 5032, further comprising an agent that inhibits infection, wherein the agent is an anthracycline. 5200. The device of item 5032, further comprising an agent that inhibits infection, wherein the agent is doxorubicin.
5201. The device of item 5032, further comprising an agent that inhibits infection, wherein the agent is mitoxantrone.
5202. The device of item 5032, further comprising an agent that inhibits infection, wherein the agent is a fluoropyrimidine.
5203. The device of item 5032, further comprising an agent that inhibits infection, wherein the agent is 5-fluorouracil (5-FU).
5204. The device of item 5032, further comprising an agent that inhibits infection, wherein the agent is a folic acid antagonist.
5205. The device of item 5032, further comprising an agent that inhibits infection, wherein the agent is methotrexate.
5206. The device of item 5032, further comprising an agent that inhibits infection, wherein the agent is a podophylotoxin.
5207. The device of item 5032, further comprising an agent that inhibits infection, wherein the agent is etoposide.
5208. The device of item 5032, further comprising an agent that inhibits infection, wherein the agent is a camptothecin.
5209. The device of item 5032, further comprising an agent that inhibits infection, wherein the agent is a hydroxyurea. 5210. The device of item 5032, further comprising an agent that inhibits infection, wherein the agent is a platinum complex.
5211. The device of item 5032, further comprising an agent that inhibits infection, wherein the agent is cisplatin.
5212. The device of item 5032, further comprising an antithrombotic agent.
5213. The device of item 5032, further comprising a visualization agent.
5214. The device of item 5032, further comprising a visualization agent, wherein the visualization agent is a radiopaque material, wherein the radiopaque material comprises a metal, a halogenated compound, or a barium containing compound. - - . . ._ _. . .
5215. The device of item 5032, further comprising a visualization agent, wherein the visualization agent is a radiopaque material, wherein the radiopaque material comprises barium, tantalum, or technetium.
5216. The device of item 5032, further comprising a visualization agent, wherein the visualization agent is a MRI responsive material.
5217. The device of item 5032, further comprising a visualization agent, wherein the visualization agent comprises a gadolinium chelate. 5218. The device of item 5032, further comprising a visualization agent, wherein the visualization agent comprises iron, magnesium, manganese, copper, or chromium.
5219. The device of item 5032, further comprising a visualization agent, wherein the visualization agent comprises an iron oxide compound.
5220. The device of item 5032, further comprising a visualization agent, wherein the visualization agent comprises a dye, pigment, or colorant.
5221. The device of item 5032, further comprising an echogenic material.
5222. The device of item 5032, further comprising an echogenic material, wherein the echogenic material is in the form of a coating.
5223. The device of item 5032 wherein the device is sterile.
5224. The device of item 5032 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device.
5225. The device of item 5032 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is connective tissue. 5226. The device of item 5032 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is muscle tissue.
5227. The device of item 5032 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is nerve tissue.
5228. The device of item 5032 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is epithelium tissue.
5229. The device of item 5032 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from the time of deployment of the device to about 1 year.
5230. The device of item 5032 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from about 1 month to 6 months.
5231. The device of item 5032 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from about 1 - 90 days.
5232. The device of item 5032 wherein the anti-scarring agent is released in effective concentrations from the device at a constant rate.
5233. The device of item 5032 wherein the anti-scarring agent is released in effective concentrations from the device at an increasing rate. 5234. The device of item 5032 wherein the anti-scarring agent is released in effective concentrations from the device at a decreasing rate.
5235. The device of item 5032 wherein the anti-scarring agent is released in effective concentrations from the composition comprising the anti-scarring agent by diffusion over a period ranging from the time of deployment of the device to about 90 days.
5236. The device of item 5032 wherein the anti-scarring agent is released in effective concentrations from the composition comprising the anti-scarring agent by erosion of the composition over a period ranging from the time of deployment of the device to about 90 days.
5237. The device of item 5032 wherein the device comprises about 0.01 Dg to about 10 Gg of the anti-scarring agent.
5238. The device of item 5032 wherein the device comprises about 10 Dg to about 10 mg of the anti-scarring agent.
5239. The device of item 5032 wherein the device comprises about 10 mg to about 250 mg of the anti-scarring agent.
5240. The device of item 5032 wherein the device comprises about 250 mg to about 1000 mg of the anti-scarring agent.
5241. The device of item 5032 wherein the device comprises about 1000 mg to about 2500 mg of the anti-scarring agent.
5242. The device of item 5032 wherein a surface of the device comprises less than 0.01 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied. 5243. The device of item 5032 wherein a surface of the device comprises about 0.01 Dg to about 1 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
5244. The device of item 5032 wherein a surface of the device comprises about 1 Dg to about 10 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
5245. The device of item 5032 wherein a surface of the device comprises about 10 Dg to about 250 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
5246. The device of item 5032 wherein a surface of the device comprises about 250 Dg to about 1000 Dg of the anti-scarring agent of anti- scarring agent per mm2 of device surface to which the anti-scarring agent is applied.. . . . . _ . .. _.
5247. The device of item 5032 wherein a surface of the device comprises about 1000 Dg to about 2500 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
5248. A method as in any one of items 5032-5247, wherein the device is a spinal disc.
5249. A method as in any one of items 5032-5247, wherein the device is a vertebral implant.
5250. A method as in any one of items 5032-5247, wherein the device is a vertebral disc prosthesis.
5251. A method as in any one of items 5032-5247, wherein the device is a lumbar disc implant. 5252. A method as in any one of items 5032-5247, wherein the device is a cervical disc implant.
5253. A method as in any one of items 5032-5247, wherein the device is a intervertebral disc.
5254. A method as in any one of items 5032-5247, wherein the device is a spinl prosthesis.
5255. A method as in any one of items 5032-5247, wherein the device is a artificial disc.
5256. A method as in any one of items 5032-5247, wherein the device is a spinal disc endoprosthesis.
5257. A method as in any one of items 5032-5247, wherein the device is an intervertebral implant.
5258. A method as in any one of items 5032-5247, wherein the device is a spinal prosthesis.
5259. A method as in any one of items 5032-5247, wherein the device is an artificial disc.
5260. A method as in any one of items 5032-5247, wherein the device is a spinal disc endoprosthesis.
5261. A method as in any one of items 5032-5247, wherein the device is an intervertebral implant.
5262. A method as in any one of items 5032-5247, wherein the device is an implantable spinal graft.
5263. A method as in any one of items 5032-5247, wherein the device is an implantable bone graft. 30
5264. A method as in any one of items 5032-5247, wherein the device is an artificial lumbar disc.
5265. A method as in any one of items 5032-5247, wherein the device is a spinal nucleus implant.
5266. A method as in any one of items 5032-5247, wherein the device is an intervertebral disc spacer.
5267. A method as in any one of items 5032-5247, wherein the device is a fusion cage.
5268. A method as in any one of items 5032-5247, wherein the device is a fusion basket.
5269. A method as in any one of items 5032-5247, wherein the device is a fusion cage apparatus.
5270. A method as in any one of items 5032-5247, Wherein the device is an interbody cage.
5271. A method as in any one of items 5032-5247, wherein the device is an interbody implant.
5272. A method as in any one of items 5032-5247, wherein the device is a fusion cage anchoring device.
5273. A method as in any one of items 5032-5247, wherein the device is a bone fixation apparatus.
5274. A method as in any one of items 5032-5247, wherein the device is a fusion stabilization chamber.
5275. A method as in any one of items 5032-5247, wherein the device is an anchoring bone plate. 5276. A method as in any one of items 5032-5247, wherein the device is a bone screw.
5277. A device, comprising an implant that provides a surgical adhesion barrier and an anti-scarring agent or a composition comprising an anti-scarring agent, wherein the agent inhibits scarring between the device and a host into which the device is implanted.
5278. The device of item 5277 wherein the agent is an adensosine A2A receptor antagonist.
5279. The device of item 5277 wherein the agent is an AKT inhibitor.
5280. The device of item 5277 wherein the agent is an alpha 2 integrin antagonist, wherein the alpha 2 integrin antagonist is Pharmaprojects No. 5754 (Merck KgaA).
5281. The device of item 5277 wherein the agent an alpha 4 integrin antagonist.
5282. The device of item 5277 wherein the agent is an alpha 7 nicotinic receptor agonist.
5283. The device of item 5277 wherein the agent is an angiogenesis inhibitor selected from the group consisting of AG-12,958 (Pfizer), ATN-161 (Attenuon LLC), neovastat, an angiogenesis inhibitor from Jerina AG (Germany), NM-3 (Mercian), VGA-1155 (Taisho), FCE-26644 (Pfizer), FCE-26950 (Pfizer), FPMA (Meiji Daries), FR-111142 (Fujisawa), GGTI-298, GM-1306 (Ligand), GPA-1734 (Novartis), NNC-47-0011 (Novo Nordisk), herbamycin (Nippon Kayaku), lenalidomide (Celegene), IP-10 (NIH), ABT-828 (Abbott), KIN-841 (Tokushima University, Japan), SF-1126 (Semafore Pharmaceuticals), laminin technology (NIH), CHIR-258 (Chiron), NVP-AEW541 (Novartis), NVP-AEW541 (Novartis), Vt16907 (Alchemia), OXI-8007 (Oxigene), EG-3306 (Ark Therapeutics), Maspin (Arriva), ABT- 567 (Abbott), PPI-2458 (Praecis Pharmaceuticals), CC-5079, CC-4089 (Celgene), HIF-1 alpha inhibitors (Xenova), S-247 (Pfizer), AP-23573 (Ariad), AZD-9935 (Astra Zeneca), mebendazole (Introgen Therapeutics), MetAP-2 inhibitors (GlaxoSm/thKline), AG-615 (Angiogene Pharmaceuticals), Tie-2 antagonists (Hybrigenics), NC-381 , CYC-381, NC-169, NC-219, NC-383, NC-384, NC-407 (Lorus Therapeutics), ATN-224 (Attenuon), ON-01370 (Onconova), Vitronectin antagonists (Amgen), SDX-103 (Salmedix), Vitronectin antagonists (Shire), CHP (Riemser), TEK (Amgen), Anecortave acetate (Alcon), T46.2 (Matrix Therapeutics), HG-2 (Heptagen), TEM antagonists (Genzyme), Oxi-4500 (Oxigene), ATN-161 (Attenuon), WX-293 (Wilex), M-2025 (Metris Therapeutics), Alphastatin (BioActa), YH-16 (Yantai Rongchang), BIBF-1120 (Boehringer Ingelheim), BAY-57-9352 (Bayer), AS- 1404 (Cancer Research Technology), SC-77964 (Pfizer), glycomimetics (BioTie Therapies), TIE-2 Inhibitors (Ontogen), DlMl, Octamer (Octamer), ABR-215050 (Active Biotech), ABT-518 (Abbott), KDR inhibitors (Abbott), BSF-466895 (Abbott), SCH-221153 (Schering-Plough), DAC.antiangiogenic (ConjuChem), TFPI (EntreMed), AZD-2171 (Astra-Zenaca), CDC-394 (Celgene), LY290293 (EIi Lilly), IDN-5390 (Indena), Kdr Kinase Inhibitors (Merck), CT- 113020, CT~116433, CT-116563, CT-31890, CT-32228) (Cell Therapeutics), A-299620 (Abbott), TWEAK Inhibitor (Amgen), VEGF modulators (Johnson and Johnson), Tum-N53, tumstatin (Genzyme), Thios- 1, Thios-2 (Thios Pharmaceuticals), MV-6401 (Miravant Medical Technologies), Spisulosine (PharmaMar), CEP-7055 (Cephalon), AUV-201 (Auvation), LM-609 (EIi Lilly), SKF-106615 (AnorMED), Oglufanide disodium (Cytran), BW-114 (Pharminox), Calreticulin (NIH)1 WX-678 (Wilex), SD-7784 (Pfizer), WX-UK1 (Wilex), SH-268 (Schering AG), 2-Me-PGA (Celgene), S- 137 (Pfizer), ZD-6126 (Angiogene Pharmaceuticals), SG-292 (SignalGen), Benefin (Lane Labs), A6, A36 (Angstrom), SB-2723005 (GlaxoSmithKline), SC-7 (Ceil Therapeutics), ZEN-014 (AEterna Zentaris), 2-methoxyestradiol (EntreMed), NK-130119 (Nippon Kayaku), CC-10004 (Celgene), AVE- 8062A (Ajinomoto), Tacedinaline (Pfizer), Actinonin (Tokyo Metropolitan Institute of Medical Science), Lenalidomide (Celgene), VGA-1155, BTO-956 (SRI International), ER-68203-00 (Eisai), CT-6685 (UCB), JKC-362 (Phoenix Pharmaceuticals), DMI-3798 (DMI Biosciences, Angiomate (Ipsen), ZD-6474 (AstraZeneca), CEP-5214 (Cephalon), Canstatin (Genzyme), NM-3 (Mercian), Oridigm (MediQuest Therapeutics), Exherin (Adherex), BLS-0597 (Boston Life Sciences), PTC-299 (PTC Therapeutics), NPI-2358 (Nereus Pharmaceuticals), CGP-79787 (Novartis), AEE-788 (Novartis), CKD-732 (Chong Kun Dang), CP-564959 (OSI Pharmaceuticals), CM-101 (CarboMed), CT-2584, CT3501 (Cell Therapeutics), combretastatin and analogues and derivatives thereof (Oxigene), Rebimastat (Bristol-Meyers Squibb), Dextrin 2-sulfate (ML Laboratories), Cilengitide (Merk KGaA), NSC-706704 (Pharminox), KRN- 951 (Kirin Brewery), Ukrajn, NSC-631570 (Nowjcky Pharma), Tecogalan sodium (Daiichi Pharmaceutical), Tz-93 (Tsumura), TBC-1635 (Encysive Pharmaceuticals), TAN-1120 (Takeda), Semaxanib (Pfizer), BDI-7800 (Biopharmacopae), SD-186, SD-983 (Bristol-Meyers Squibb), SB-223245 (GlaxdSmithKline), SC-236 (Pfizer), RWJ-590973 (Johnson and Johnson), ILX-1850 (Genzyme), SC-68488, S-836 (Pfizer), CG-55069-11 (CuraGen), Ki-23057 (Kirin Brewery), CCX-700 (Chemoentryx), Pegaptanib octasodium (Giled Sciences), ANGlOCOL (available from Biostratum Inc.), or an analogue or derivative thereof.
5284. The device of item 5277 wherein the agent is an apoptosis antagonist.
5285. The device of item 5277 wherein the agent is an apoptosis activator. 5286. The device of item 5277 wherein the agent is a beta 1 integrin antagonist.
5287. The device of item 5277 wherein the agent is a beta tubulin inhibitor.
5288. The device of item 5277 wherein the agent is a blocker of enzyme production in Hepatitis C.
5289. The device of item 5277 wherein the agent is a Bruton's tyrosine kinase inhibitor.
5290. The device of item 5277 wherein the agent is a calcineurin inhibitor.
5291.. The device of item 5277 wherein the agent is a caspase 3 inhibitor.
5292. The device of item 5277 wherein the agent is a CC chemokine receptor antagonist.
5293. The device of item 5277 wherein the agent is a eel! cycle inhibitor selected from the group consisting of SNS-595 (Sunesis), synthadotin, KRX-0403, and an analogue or derivative thereof.
5294. The device of item 5277 wherein the agent is a cathepsin B inhibitor.
5295. The device of item 5277 wherein the agent is a cathepsin K inhibitor, wherein the cathepsin K inhibitor is 462795 (GlaxoSmithKline), INPL-022-D6 (Amura Therapeutics), an analogue or derivative thereof).
5296. The device of item 5277 wherein the agent is a cathepsin L inhibitor.
5297. The device of item 5277 wherein the agent is a CD40 antagonist.
5298. The device of item 5277 wherein the agent is a chemokine receptor agonist.
5299. The device of item 5277 wherein the agent is a chymase inhibitor.
5300. The device of item 5277- wherein the agent is a collagenase antagonist.
5301. The device of item 5277 wherein the agent is a CXCR antagonist.
5302. The device of item 5277 wherein the agent is a cyclin dependent kinase inhibitor selected from the group consisting of a CDK-1 inhibitor, a CDK-2 inhibitor, a CDK- 4 inhibitor, a CDK-6 inhibitor, a CAK1 inhibitor from GPC Biotech and Bristol-Myers Squibb, RGB-286199 (GPC Biotech), an anticancer agent from Astex Technology, a CAK1 inhibitor from GPC Biotech, a CDK inhibitor from Sanofi-Aventis, a CDK1/CDK2 inhibitor from Amgen, a CDK2 inhibitor from SUGEN-2 (Pfizer), a hearing loss therapy agent (Sound Pharmaceuticals), PD-0332991 (Pfizer), RGB-286199 (GPC Biotech), Ro-0505124 (Hoffmann-La Roche), a Ser/Thr kinase inhibitor from Lilly (EIi Lilly), CVT-2584 (CAS No. 199986-75-9) (CV Therapeutics), and an analogue or derivative thereof.
5303. The device of item 5277 wherein the agent is a cyclooxygenase 1 inhibitor.
5304. The device of item 5277 wherein the agent is a DHFR inhibitor.
5305. The device of item 5277 wherein the agent is a dual integrin inhibitor.
5306. The device of item 5277 wherein the agent is an elastase inhibitor.
- - - - 5307. The device of item 5277 wherein the agent is an elongation factor-1 alpha inhibitor.
5308. The device of item 5277 wherein the agent is an endothelial growth factor antagonist.
5309. The device of item 5277 wherein the agent is an endothelial growth factor receptor kinase inhibitor selected from the group consisting of sorafenib tosylate (Bayer), AAL-993 (Novartis), ABP-309 (Novartis), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), E- 7080 (Eisai), EG-3306 (Ark Therapeutics), EXEL-2880 (Exelixis), GW- 654652 (GlaxoSmithKline), a KDR inhibitor from LG Life Sciences, CT-6685 and CT-6729 (UCB), KRN-633 and KRN-951 (Kirin Brewery), OSI-930 (OSI Pharmaceuticals), SP-5.2 (Supratek Pharma), SU-11657 (Pfizer), a Tie-2 antagonist (Hybrigenics), a VEGFR-2 kinase inhibitor (Bristol-Myers Squibb), XL-647 (Exelixis), a KDR inhibitor from Abbott Laboratories, sorafenib tosylate, and an analogue or derivative thereof.
5310. The device of item 5277 wherein the agent is an endotoxin antagonist.
5311. The device of item 5277 wherein the agent is an epothilone and tubulin binder.
5312. The device of item 5277 wherein the agent is an estrogen receptor antagonist.
5313. The device of item 5277 wherein the agent is an FGF inhibitor.
5314. The device of item 5277 wherein the agent is a famexyl transferase inhibitor.
5315. The device of item 5277 wherein the agent is famesyltransferase inhibitor selected from the group of A-197574 (Abbott), a farnesyltransferase inhibitor from Servier, FPTIII (Strathclyde Institute for Drug R), LB-42908 (LG Life Sciences), Pharmaprojects No. 5063 (Genzyme), Pharmaprojects No. 5597 (Ipsen), Yissum Project No. B-1055 (Yissum), and an analogue or derivative thereof.
5316. The device of item 5277 wherein the agent is an FLT-3 kinase inhibitor.
5317. The device of item 5277 wherein the agent is an FGF receptor kinase inhibitor. 5318. The device of item 5277 wherein the agent is a fibrinogen antagonist selected from the group consisting of AUV-201 (Auvation), MG-13926 (Sanofi-Aventis), plasminogen activator (CAS No. 105913-11-9) (from Sanofi-Aventis or UCB), plasminogen activator-2 (tPA- 2) (Sanofi-Aventis), pro-urokinase (CAS No. 82657-92-9) (Sanofi-Aventis), and an analogue or derivative thereof.
5319. The device of item 5277 wherein the agent is a heat shock protein 90 antagonist selected from the group consisting of SRN-005 (Sirenade), geldanamycin, NSC-33050 (17-allylaminogeldanamycin; 17- AAG), 17-dimethylaminoethylamino-17-demethoxy-geldanamycin (17- DMAG), rifabutin (rifamycin XIV, r,4-didehydro-1-deoxy-1 ,4-dihydro-5'-(2- methylpropyl)-1~oxo-), and an analogue or derivative thereof.
5320. The device of item 5277 wherein the agent is a histone deacetylase -inhibitor.-- - . . -
5321. The device of item 5277 wherein the agent is an HMGCoA reductase inhibitor selected from the group consisting of an atherosclerosis therapeutic from Lipid Sciences, ATI-16000 (ARYx Therapeutics), KS-01-019 (Kos Pharmaceuticals), Pharmaprojects No. 2197 (Sanofi-Aventi), RP 61969 (Sanofi-Aventis), and an analogue or derivative thereof.
5322. The device of item 5277 wherein the agent is an ICAM inhibitor.
5323. The device of item 5277 wherein the agent is an IL, ICE and IRAK antagonist, wherein the antagonist is a CJ-14877, CP-424174 (Pfizer), NF-61 (Negma-Lerads), and an analogue or derivative thereof. 5324. The device of item 5277 wherein the agent is an IL-2 inhibitor.
5325. The device of item 5277 wherein the agent is an immunosuppressant selected from the group consisting of teriflunomide (Sanofi Aventis), chlorsulfaquinoxalone (NSC-339004), chlorsulfaquinoxalone sulfate, CS-712 (Sankyo), ismomultin alfa (CAS No. 457913-93-8) (Akzo Nobel), antiallergics from GenPat77, antiinflammatories or AT-005 (Androclus Therapeutics), autoimmune disease therapeutics from EpiVax, BN-007 (Bone), budesonide (CAS No. 51333-22- 3) (MAP Pharmaceuticals), CO-14 (Genzyme), edratide (CAS No. 433922- 67-9) (Teva), EP-314 (Enanta), eprovafen (CAS No. 101335-99-3) (Sanofi- Aventis), HWA- 131 (CAS No. 118788-41-3) (Sanofi-Aventis), immunomodulators from MerLion Pharmaceuticals, immunosuppressives from Alchemia, IPL-12 (Inflazyme), MDL-9563 (CAS No. 27086-86-8) (Sanofi-Aventis), Pharmaprojects No. 2330 (Sanofi-Aventis), Pharmaprojects No. 6426 (Abgenix), PXS-25 (Pharmaxis), rosmarinic acid (CAS No. 20283-92-5) (Sanofi-Aventis), RP 42927 or RP 54745 (CAS No. 135330-08-4) (Sanofi-Aventis), SGN-35 (Seattle Genetics), ST-1959 (Sigma-Tau), type I diabetes therapy from SYNX Pharma, UNIL-88 (Debiopharm), VP-025 (Vasogen), VR-694 (Vectura), PRTX-001 (Protalex), and an analogue or derivative thereof.
5326. The device of item 5277 wherein the agent is an IMPDH (inosine monophosphate).
5327. The device of item 5277 wherein the agent is an integrin antagonist.
5328. The device of item 5277 wherein the agent is an interleukin antagonist. 5329. The device of item 5277 wherein the agent is an inhibitor of type III receptor tyrosine kinase.
5330. The device of item 5277 wherein the agent is an irreversible inhibitor of enzyme methionine aminopeptidase type 2.
5331. The device of item 5277 wherein the agent is an isozyme selective delta protein kinase C inhibitor.
5332. The device of item 5277 wherein the agent a JAK3 enzyme inhibitor.
5333. The device of item 5277 wherein the agent is a JNK inhibitor.
5334. The device of item 5277 wherein the agent is a kinase inhibitor.
5335. The device of item 5277 wherein the agent is kinesin antagonist.
5336. The device of item 5277 wherein the agent is a kinesin antagonist.
5337. The device of item 5277 wherein the agent is a leukotriene inhibitor and antagonist selected from the group consisting of ambicromil (CAS No. 58805-38-2) (Sanofi-Aventis), amelubant (CAS No. 346735-24-8) (Boehringer Ingelheim), DW-1141 (Dong Wha), ebselen (Daiichi Pharmaceutical), ibudilast (Kyorin), leucotriene inhibitors from Sanofi-Aventis, lymphotoxin-beta receptor (LT-D) from Biogen Idee, Pharmaprojects No. 1535 and 2728 (CAS No. 119340-33-9) (Sanofi- Aventis), R-112 (Rigel), Rev-5367 (CAS No. 92532-05-3) (Sanofi-Aventis), RG-14893 (CAS No. 141835-49-6) (Sanofi-Aventis), RG-5901-A (CAS No. 101910-24-1), 92532-23-5, RP 66153 (CAS No. 142422-79-5), RP 66364 (CAS No.186916-92-5), RP 69698 (CAS No. 141748-00-7) (Sanofi-Aventis), SC-411930 (Pfizer), SC-41930 (CAS No. 120072-59-5) (Pfizer), SC-50605 (CAS No. 138828-39-4) (Pfizer), SC-51146 (CAS No. 141059-52-1), SC- 53228 (CAS No. 153633-01-3) (Pfizer), spaglumic acid (ZY-15106) (CAS No.3106-85-2), 80619-64-3 (Novartis), tipredane (CAS No. 85197-77-9) (Bristol-Myers Squibb), U-75302 (CAS No. 119477-85-9) (Pfizer), and analogue or derivative thereof.
5338. The device of item 5277 wherein the agent is an MAP kinase inhibitor.
5339. The device of item 5277 wherein the agent is a matrix metalloproteinase inhibitor.
5340. The device of item 5277 wherein the agent is an MCP- CCR2 inhibitor.
5341. The device of item 5277 wherein the agent is an mTOR inhibitor.
5342. The device of item 5277 wherein the agent is an mTOR kinase inhibitor.
5343. The device of item 5277 wherein the agent is a microtubule inhibitor selected from the group consisting of antibody- maytansinoid conjugates from Biogen Idee, colchicines (MantiCore Pharmaceuticals), anticancer immunoconjugates from Johnson & Johnson, DIME from Octamer, gni-1f (GNI), huC242-DM4, huMy9-6-DM1 (ImmunoGen), IDN-5404 (Indena), IMO-098, IMOderm (Imotep), mebendazole (Introgen Therapeutics), microtubule poisons from Cambridge Enterprise, paclitaxel such as LOTAX from Aphios (CAS No. 33069-62-4), Genexol-PM from Samyang, Pharmaprojects No. 6383 (Tapestry Pharmaceuticals), RPR-112378 (Sanofi-Aventis), SGN-75 (Seattle Genetics), SPL-7435 (Starpharma), SSR-250411 (Sanofi-Aventis), trastuzumab-DM1 (Genentech), vinorelbine, and an analogue or derivative thereof.
5344. The device of item 5277 wherein the agent is an MIF inhibitor.
5345. The device of item 5277 wherein the agent is an MMP inhibitor.
. 5346. The device of item 5277 wherein the agent is a „ neurokinin (NK) antagonist selected from the group consisting of anthrotainin (CAS No. 148084-40-6) (Sanofi-Aventis), an IBS thereapeutic from ArQuIe, MDL-105212A (CAS No. 167261-60-1) (Ssanofi-Aventis), Pharmaprojects No. 2744, 3258 (CAS No. 139167-47-8) 4006, 4201, or 5986 (Sanofi-Aventis), RP 67580 (CAS No. 135911-02-3), SR-144190 (CAS No. 201152-86-5), SSR-240600, SSR-241586 (Sanofi-Aventis), TKA-457 (Novartis), vestipitant mesylate (CAS No. 334476-64-1) (GlaxoSmithKline), Win-64821 (Sanofi-Aventis), PRX-96026 (Predix Pharmaceuticals), and an analogue or derivative thereof.
5347. The device of item 5277 wherein the agent is an NF kappa B inhibitor selected from the group consisting of AVE-0545 or AVE- 0547 (Sanofi-Aventis), bortezomib (CAS No. 179324-69-7) (Millennium Pharmaceuticals), dexanabinol (CAS No. 112924-45-5) (Pharmos), dexlipotam (Viatris), Pharmaprojects No. 6283 (INDRA) (OXiGENE), IPL- 576092 (CAS No. 137571-30-3) (Inflazyme), NFKB decoy (Corgentech), NFKB decoy oligo (AnGes MG), NFKB's from Ariad, osteoporosis treatments or S5 (F005) from Fulcrum Pharmaceuticals, P61 (Phytopharm), R-flurbiprofen (CAS No. 5104-49-4) (Encore Pharmaceuticals), and an analogue or derivative thereof.
5348. The device of item 5277 wherein the agent is a nitric oxide agonist.
5349. The device of item 5277 wherein the agent is an ornithine decarboxylase inhibitor.
5350. The device of item 5277 wherein the agent is a p38 MAP kinase inhibitor selected from the group consisting of AZD-6703 (AstraZeneca), JX-401 (Jexys Pharmaceuticals), BMS-2 (Bristol-Myers Squibb), a p38 MAP kinase inhibitor from Novartis, a p38=alpha MAP kinase inhibitor from Amphora, Pharmaprojects No. 5704 (Pharmacopeia), RPR- 200765A (Sanofi-Aventis), SD-282 (Johnson & Johnson), TAK-715 (Takeda), and an analogue or derivative thereof.
5351. The device of item 5277 wherein the agent is a palmitoyl-protein thioesterase inhibitor.
5352. The device of item 5277 wherein the agent is a PDGF receptor kinase inhibitor selected from the group consisting of AAL-993, AMN-107, or ABP-309 (Novartis), AMG-706 (Amgen), BAY-57-9352 (Bayer), CDP-860 (UCB), E-7080 (Eisai), imatinib (CAS No. 152459-95-5) (Novartis), OSI-930 (OSI Pharmaceuticals), RPR-127963E (Sanofi-Aventis), RWJ-540973 (Johnson & Johnson), sorafenib tosylate (Bayer), SU-11657 (Pfizer), tandutinib (CAS No. 387867-13-2) (Millennium Pharmaceuticals), vatalanib (Novartis), ZK-CDK (Schering AG)1 and an analogue or derivative thereof.
5353. The device of item 5277 wherein the agent is (-)- halofenate (Metabolex), AMG-131 (Amgen), antidiabetics from Japan Tobacco, AZD-4619, AZD-8450, AZD-8677 (AstraZeneca), DRF-10945, balaglitazone (Dr Reddy's), CS-00088, CS-00098 (Chipscreen Biosciences), E-3030 (Eisai), etalocib (CAS No. 161172-51-6) (EIi Lilly), GSK-641597 (Ligand), GSK-677954 (GlaxoSmithKline), GW-409544 (Ligand), GW- 590735 (GlaxoSmithKline), K-111 (Hoffmann-La Roche), LY-518674 (EIi Lilly), LY-674 (Ligand), LY-929 (Ligand), MC-3001 , MC-3002 (MaxoCore Pharmaceuticals), metformin HCI + pioglitazone (CAS No. 1115-70-4 and
112529-15-4), ACTOPLUS MET from Andrx), muraglitazar (CAS No. 331741-94-7) (Bristol-Myers Squibb), naveglitazar (Ligand), oleoylethanolamide (Kadmus Pharmaceuticals), ONO-5129, pioglitazone hydrochloride (CAS No. 111025-46-8 and 112529-15-4) (Takeda), PLX-204 (Plexxikon), PPAR agonists from Genfit, PPAR delta agonists from EIi Lilly, PPAR-alpha agonists from CrystalGenomics, PPAR-gamma modulators and PPAR-β modulators from CareX, rosiglitazone maleate (CAS No. 122324- 73-4 or 155141-29-0) (GlaxoSmithKline), rosiglitazone maleate/glimepir (CAS No. 155141-29-0 and 93479-97-1), AVANDARYL, rosiglitazone maleate/metformin extend (CAS No. 155141-29-0 and 657-24-9), AVANDAMET, rosiglitazone maleate+metformin, AVANDAMET (GlaxoSmithKline), tesaglitazar (AstraZeneca), and an analogue or derivative thereof.
5354. The device of item 5277 wherein the agent is a phosphatase inhibitor.
5355. The device of item 5277 wherein the agent is a phosphodiesterase (PDE) inhibitor selected from the group consisting of avanafil (Tanabe Seiyaku), dasantafil (CAS No. 569351-91-3) (Schering- Plough), A-906119 (CAS No. 134072-58-5), DL-850 (Sanofi-Aventis), GRC- 3015, GRC-3566, GRC-3886 (Glenmark), HWA-153 (CAS No. 56395-66-5) (Sanofi-Aventis), hydroxypumafentrine (Altana), IBFB-130011 , IBFB-14-016, IBFB-140301 , IBFB-150007, IBFB-211913 (IBFB Pharma), L-826141 (Merck & Co), medorinone (CAS No. 88296-61-1) (Sanofi-Aventis), MEM- 1917 (Memory Pharmaceuticals), ND-1251 (Neuro3d), PDE inhibitors from ICOS, PDE IV inhibitors from Memory Pharmaceuticals and CrystalGenomics, Pharmaprojects No. 2742 and 6141 (Sanofi-Aventis), QAD-171 (Novartis), RHC-2963 (CAS No. 76993-12-9 and 76993-14-1), RPR-117658, RPR-122818 derivatives, SR-24870 , and RPR-132294 (Sanofi-Aventis), SK-350 (ln2Gen), stroke therapy agents from deCODE Genetics, TAS-203 (Taiho), tofimilast (CAS No. 185954-27-2) (Pfizer), UK- 371800 (Pfizer), WIN-65579 (CAS No. 158020-82-7) (Sanofi-Aventis), IBFB- 130020 (IBFB Pharma), OPC-6535 (CAS No. 145739-56-6) (Otsuka), a phosphodiesterase III inhibitor, enoximone, a phosphodiesterase IV inhibitor, fosfosal, Atopik (Barrier Therapeutics), triflusal, a phosphodiesterase V inhibitor, and an analogue or derivative thereof.
5356. The device of item 5277 wherein the agent is a PKC inhibitor.
5357. The device of item 5277 wherein the agent is a platelet activating factor antagonist.
5358. The device of item 5277 wherein the agent is a platelet- derived growth factor receptor kinase inhibitor.
5359. The device of item 5277 wherein the agent is a prolyl hydroxylase inhibitor. 5360. The device of item 5277 wherein the agent is a polymorphonuclear neutrophil inhibitor.
5361. The device of item 5277 wherein the agent is a protein kinase B inhibitor.
5362. The device of item 5277 wherein the agent is a protein kinase C stimulant.
5363. The device of item 5277 wherein the agent is a purine nucleoside analogue.
5364. The device of item 5277 wherein the agent is a purinoreceptor P2X antagonist.
5365. The device of item 5277 wherein the agent is a Raf kinase inhibitor.
5366. The device of item 5277 wherein the agent is a reversible inhibitor of ErbB1 and ErbB2.
5367. The device of item 5277 wherein the agent is a ribonucleoside triphosphate reductase inhibitor.
5368. The device of item 5277 wherein the agent is an SDF-1 antagonist.
5369. The device of item 5277 wherein the agent is a sheddase inhibitor. 5370. The device of item 5277 wherein the agent is an SRC inhibitor.
5371. The device of item 5277 wherein the agent is a stromelysin inhibitor.
5372. The device of item 5277 wherein the agent is an Syk kinase inhibitor.
5373. The device of item 5277 wherein the agent is a telomerase inhibitor.
5374. The device of item 5277 wherein the agent is a TGF beta inhibitor selected from the group consisting of pirfenidone (CAS No. 53179-13-8) (MARNAC), tranilast (CAS No. 53902-12-8) (Kissei), IN-1130 (ln2Gen), mannose-6-phosphate (BTG), TGF-β antagonists from Inflazyme (Pharmaprojects No. 6075), TGF-β antagonists from Sydney, non-industrial source), TGF-β I receptor kinase inhibitors from EIi Lilly, TGF-β receptor inhibitors from Johnson & Johnson, and an analogue or derivative thereof.
5375. The device of item 5277 wherein the agent is a TNFα antagonist or TACE inhibitor selected from the group consisting of adalimumab (CAS No. 331731-18-1) (Cambridge Antibody Technology), AGIX-4207 (AtheroGenics), AGT-1 (Advanced Biotherapy), an antiinflammatory from Borean Pharma, Cellzome, or Paradigm Therapeutics, anti-inflammatory vaccine (TNF-alpha kinoid) from Neovacs, humanized anti-TNF antibody or an anti-TNF MAb (CB0006) Celltech (UCB), apratastat (CAS No. 287445-51-0) (Wyeth), BMS-561392 (Bristol-Myers Squibb), BN- 006 (Bone), certolizumab pegol (CAS No. 428863-50-7 or CH-138 (UCB), cilomilast (CAS No. 153259-65-5) (GlaxoSmithKline), CR-1 (Nuada Pharmaceuticals), CRx-119 (CombinatoRx), D-5410 (UCB), dacopafant (CAS No. 125372-33-0) (Sanofi-Aventis), dersalazine (CAS No. 188913-57- 7/188913-58-8) (Uriach), etanercept (CAS No. 185243-69-0) (Amgen), ethyl pyruvate (Critical (Critical Therapeutics), golimumab (CAS No. 476181-74-5) (Johnson & Johnson), hormono-immunotherapy from Ipsen, CDP571 (e.g., Humicade from UCB), IC-485 (ICOS), infliximab (CAS No. 170277-31-3) (Johnson & Johnson), IP-751 (Manhattan Pharmaceuticals), ISIS-104838 (CAS No. 250755-32-9) (ISIS Pharmaceuticals), lenaiidomide (CAS No. 191732-72-6) (Celgene), lentinan (CAS No. 37339-90-5) (Ajinomoto), MDL- 201112 (CAS No. 142130-73-2) (Sanofi-Aventis), medroxyprogesterone (CAS No. 520-85-4) (InKine Pharmaceutical), N-acetylcysteine (CAS No. 616-91-1) (Zambon), NBE-P2 (DIREVO Biotech), nerelimomab (CAS No. 162774-06-3) (Chiron), OM-294DP (OM PHARMA), onercept (CAS No. 199685-57-9) (Yeda), PASSTNF-alpha (Verigen), pentoxifylline or oxypentifylline (Sanofi-Aventis), Pharmaprojects No. 4091 , 4241 , 4295, or 4488 (Sanofi-Aventis), Pharmaprojects No. 5480 (Amgen), Pharmaprojects No. 6749 (Cengent), pirfenidone (CAS No. 53179-13r8)_(MARNAC), RPR- 132294 (Sanofi-Aventis), S5 (F002) (Fulcrum Pharmaceuticals), simvastatin (CAS No. 79902-63-9) (Merck & Co), STA-6292 (Synta Pharmaceuticals), tacrolimus (CAS No. 104987-11-3) (Fujisawa LifeCycle Pharma), talactoferrin alfa (CAS No. 308240-58-6) (Agennix), thalidomide (CAS No. 50-35-1) (Celgene), TNF antagonists form ProStrakan, and Synergen, TNF inhibitors (Amgen), TNF-alpha antagonists from Dynavax Technologies and Jerina AG (Germany), TNF-alpha inhibitors from IBFB Pharma and Xencor (Xencor), torbafylline (CAS No. 105102-21-4) (Sanofi-Aventis), UR-1505 (Uriach), VT-346 (Viron Therapeutics), YSIL6 (Y's Therapeutics), YSTH2 (Y's Therapeutics), NPI-1302a-3 (Nereus Pharmaceuticals, a TNF antagonist from Jerina AG (Germany), dersalazine, and an analogue or derivative thereof.
5376. The device of item 5277 wherein the agent is a Toll receptor inhibitor. 5377. The device of item 5277 wherein the agent is a tubulin antagonist.
5378. The device of item 5277 wherein the agent is a tyrosine kinase inhibitor selected from the group consisting of SU-011248, SUTENT from Pfizer Inc. (New York, NY), BMS-354825, PN-355 (Paracelsian Pharmaceuticals), AGN-199659 (Allergan), AAL-993 or ABP-309 (Novartis), adaphostin (NIH), AEE-788 (Novartis), AG-013736 (OSI Pharmaceuticals), AG-13736 (Pfizer), ALT-110 (Alteris Therapeutics), AMG-706 (Amgen), anticancer MAbs from Xencor, anti-EGFrvlll MAbs from Abgenix, anti-HER2 MAb from Abiogen, AZD-2171 or AZD-9935 (AstraZeneca), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), CEP-5214 (Cephalon), CEP- 7055 (Cephalon), cetuximab (ImClone Systems), CHIR-200131 and CHIR- 258 (Chiron), CP-547632 (OSI Pharmaceuticals), CP-724714 (Pfizer), CT- 301 (Creabilis Therapeutics), D-69491 (Baxter International), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EGFR/ErbB2 inhibitors from Array BioPharma, erlotinib (CAS No. 183319-69-9) (OSI Pharmaceuticals), EXEL-2880 (Exelixis), FK-778 (Sanofi-Aventis) , gefitinib (CAS No. 184475-35-2) (AstraZeneca), GW-2286 or GW-654652 (GlaxoSmithKline), her2/neu antigen from AlphaVax, HER-2/neu inhibitor from Generex, Herzyme (Medipad) (Sirna Therapeutics), HKI-272 (Wyeth), HuMax-EGFr (Genmab), idronoxil (CAS No. 81267-65-4) (Novogen), 1GF-1 inhibitors from Ontogen, IMC-11F8 (ImClone Systems), kahalalide F (CAS No. 149204-42-2) (PharmaMar), KDR inhibitor from LG Life Sciences, KDR inhibitors from Abbott Laboratories, KDR kinase inhibitors (UCB), Kdr kinase inhibitors from Merck & Co, KRN-633 and KRN-951 (Kirin Brewery), KSB-102 (Xenova), lapatinib ditosylate (CAS No. 388082-78-8) (GlaxoSmithKline), matuzumab (Merck KGaA), MDX-214 (Medarex), ME-103 (Pharmexa), MED-A300 (Gerolymatos), MNAC-13 (Lay Line Genomics), nimotuzumab (Center of Molecular Immunology), NSC-330507 or NSC-707545 (NIH), NV-50 (Novogen), OSI-930 (OSI Pharmaceuticals), panitumumab (Abgenix), pelitinib (CAS No. 287933-82-7) (Wyeth), pertuzumab (CAS No. 380610-27- 5) (Genentech), Pharmaprojects No. 3985 (Sanofi-Aventis), prostate cancer therapeutics from Sequenom (SQPC35, SQPC36, SQPC90), removab and remoxab (Trion Pharma), RG-13022 (CAS No. 136831-48-6), RG-13291 (CAS No. 138989-50-1), or RG-14620 (CAS No. 136831-49-7) (Sanofi- Aventis), RM-6427 (Romark), RNAi breast cancer therapy from Benitec, RP 53801 (CAS No. 125882-88-4) (Sanofi-Aventis), sorafenib tosylate (Bayer), SU-11657 (Pfizer), Tie-2 antagonists from Semaia (Hybrigenics), Tie-2 inhibitors from Ontogen, trastuzumab (CAS No. 180288-69-1) (Genentech), tyrosine kinase inhibitors from Sanofi-Aventis, U3-1287, U3-1565, U3-1784, U3-1800 (U3 Pharma), vatalanib (Novartis), VEGFR-2 kinase inhibitor from Bristol-Myers Squibb, XL-647 (Exelixis), ZD-6474 (AstraZeneca), ZK-CDK (Schering AG), an EGFR tyrosine kinase inhibitor, EKB-569 (Wyeth), and an analogue or derivative thereof.
5379. The device of item 5277 wherein the agent is a VEGF inhibitor.
5380. The device of item 5277 wherein the agent is a vitamin D receptor agonist.
5381. The device of item 5277 wherein the agent is ZD-6474 (an angiogenesis inhibitor).
5382. The device of item 5277 wherein the agent is AP-23573 (an mTOR inhibitor).
5383. The device of item 5277 wherein the agent is synthadotin (a tubulin antagonist). 5384. The device of item 5277 wherein the agent is S-0885 (a collagenase inhibitor).
5385. The device of item 5277 wherein the agent is aplidine (an elongation factor-1 alpha inhibitor).
5386. The device of item 5277 wherein the agent is ixabepilone (an epithilone).
5387. The device of item 5277 wherein the agent is IDN-5390 (an angiogenesis inhibitor).
5388. The device of item 5277 wherein the agent is SB- 2723005 (an angiogenesis inhibitor).
- 5389. The device of item 5277 wherein the agent js ABT-518 (an angiogenesis inhibitor).
5390. The device of item 5277 wherein the agent is combretastatin (an angiogenesis inhibitor).
5391. The device of item 5277 wherein the agent is anecortave acetate (an angiogenesis inhibitor).
5392. The device of item 5277 wherein the agent is SB- 715992 (a kinesin antagonist).
5393. The device of item 5277 wherein the agent is temsirolimus (an mTOR inhibitor). 5394. The device of item 5277 wherein the agent is adalimumab (a TNFα antagonist).
5395. The device of item 5277, further comprising a polymer.
5396. The device of item 5277 wherein the anti-scarring agent inhibits adhesion between the device and a host into which the device is implanted.
5397. The device of item 5277 wherein the device delivers the anti-scarring agent locally to tissue proximate to the device.
5398. The device of item 5277, further comprising a coating, wherein the coating comprises the anti-scarring agent.
5399.-The device-of item 5277, further comprising a coating, wherein the coating is disposed on a surface of the device.
5400. The device of item 5277, further comprising a coating, wherein the coating directly contacts the device.
5401. The device of item 5277, further comprising a coating, wherein the coating indirectly contacts the device.
5402. The device of item 5277, further comprising a coating, wherein the coating partially covers the device.
5403. The device of item 5277, further comprising a coating, wherein the coating completely covers the device. 5404. The device of item 5277, further comprising a coating, wherein the coating is a uniform coating.
5405. The device of item 5277, further comprising a coating, wherein the coating is a non-uniform coating.
5406. The device of item 5277, further comprising a coating, wherein the coating is a discontinuous coating.
5407. The device of item 5277, further comprising a coating, wherein the coating is a patterned coating.
5408. The device of item 5277, further comprising a coating, wherein the coating has a thickness of 100 Dm or less.
5409. The device of item -5277, further comprising a coating, wherein the coating has a thickness of 10 Dm or less.
5410. The device of item 5277, further comprising a coating, wherein the coating adheres to the surface of the device upon deployment of the device.
5411. The device of item 5277, further comprising a coating, wherein the coating is stable at room temperature for a period of 1 year.
5412. The device of item 5277, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 0.0001% to about 1% by weight. 5413. The device of item 5277, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 1% to about 10% by weight.
5414. The device of item 5277, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 10% to about 25% by weight.
5415. The device of item 5277, further comprising a coating, wherein the anti-scarring agent is present in the coating in an amount ranging between about 25% to about 70% by weight.
5416. The device of item 5277, further comprising a coating, wherein the coating further comprises a polymer.
5417. The device oiitem 5277, further comprising a first coating having a first composition and the second coating having a second composition.
5418. The device of item 5277, further comprising a first coating having a first composition and the second coating having a second composition, wherein the first composition and the second composition are different.
5419. The device of item 5277, further comprising a polymeric carrier, wherein the polymeric carrier comprises a copolymer.
5420. The device of item 5277, further comprising a polymeric carrier, wherein the polymeric carrier comprises a block copolymer. 5421. The device of item 5277, further comprising a polymeric carrier, wherein the polymeric carrier comprises a random copolymer.
5422. The device of item 5277, further comprising a polymeric carrier, wherein the polymeric carrier comprises a biodegradable polymer.
5423. The device of item 5277, further comprising a polymeric carrier, wherein the polymeric carrier comprises a non-biodegradable polymer.
5424. The device of item 5277, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrophilic polymer.
5425. The device of item 5277, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrophobic polymer.
5426. The device of item 5277, further comprising a polymeric carrier, wherein the polymeric carrier comprises a polymer having hydrophilic domains.
5427. The device of item 5277, further comprising a polymeric carrier, wherein the polymeric carrier comprises a polymer having hydrophobic domains.
5428. The device of item 5277, further comprising a polymeric carrier, wherein the polymeric carrier comprises a non-conductive polymer.
5429. The device of item 5277, further comprising a polymeric carrier, wherein the polymeric carrier comprises an elastomer. 5430. The device of item 5277, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrogel.
5431. The device of item 5277, further comprising a polymeric carrier, wherein the polymeric carrier comprises a silicone polymer.
5432. The device of item 5277, further comprising a polymeric carrier, wherein the polymeric carrier comprises a hydrocarbon polymer.
5433. The device of item 5277, further comprising a polymeric carrier, wherein the polymeric carrier comprises a styrene-derived polymer.
5434. The device of item 5277, further comprising a polymeric carrier, wherein the polymeric carrier comprises a butadiene polymer.
5435. The device of item 5277, further comprising a polymeric carrier, wherein the polymeric carrier comprises a macromer.
5436. The device of item 5277, further comprising a polymeric carrier, wherein the polymeric carrier comprises a poly(ethylene glycol) polymer.
5437. The device of item 5277, further comprising a polymeric carrier, wherein the polymeric carrier comprises an amorphous polymer.
5438. The device of item 5277, further comprising a lubricious coating.
5439. The device of item 5277 wherein the anti-scarring agent is located within pores or holes of the device. 5440. The device of item 5277 wherein the anti-scarring agent is located within a channel, lumen, or divet of the device.
5441. The device of item 5277, further comprising a second pharmaceutically active agent.
5442. The device of item 5277, further comprising an antiinflammatory agent.
5443. The device of item 5277, further comprising an agent that inhibits infection.
5444. The device of item 5277, further comprising an agent that inhibits infection, wherein the agent is an anthracycline.
_. . . . 5445. The device of item 5277, further comprising an agent, that inhibits infection, wherein the agent is doxorubicin.
5446. The device of item 5277, further comprising an agent that inhibits infection, wherein the agent is mitoxantrone.
5447. The device of item 5277, further comprising an agent that inhibits infection, wherein the agent is a fluoropyrimidine.
5448. The device of item 5277, further comprising an agent that inhibits infection, wherein the agent is 5-fluorouracil (5-FU).
5449. The device of item 5277, further comprising an agent that inhibits infection, wherein the agent is a folic acid antagonist. 5450. The device of item 5277, further comprising an agent that inhibits infection, wherein the agent is methotrexate.
5451. The device of item 5277, further comprising an agent that inhibits infection, wherein the agent is a podophyiotoxin.
5452. The device of item 5277, further comprising an agent that inhibits infection, wherein the agent is etoposide.
5453. The device of item 5277, further comprising an agent that inhibits infection, wherein the agent is a camptothecin.
5454. The device of item 5277, further comprising an agent that inhibits infection, wherein the agent is a hydroxyurea.
5455. The device-of item 5277, further comprising an agent that inhibits infection, wherein the agent is a platinum complex.
5456. The device of item 5277, further comprising an agent that inhibits infection, wherein the agent is cisplatin.
5457. The device of item 5277, further comprising an antithrombotic agent.
5458. The device of item 5277, further comprising a visualization agent.
5459. The device of item 5277, further comprising a visualization agent, wherein the visualization agent is a radiopaque material, wherein the radiopaque material comprises a metal, a halogenated compound, or a barium containing compound. 5460. The device of item 5277, further comprising a visualization agent, wherein the visualization agent is a radiopaque material, wherein the radiopaque material comprises barium, tantalum, or technetium.
5461. The device of item 5277, further comprising a visualization agent, wherein the visualization agent is a MRI responsive material.
5462. The device of item 5277, further comprising a visualization agent, wherein the visualization agent comprises a gadolinium chelate.
5463. The device of item 5277, further comprising a visualization agent, wherein the visualization agent comprises iron, magnesium, manganese, copper, or chromium.
5464. The device of item 5277, further comprising a visualization agent, wherein the visualization agent comprises an iron oxide compound.
5465. The device of item 5277, further comprising a visualization agent, wherein the visualization agent comprises a dye, pigment, or colorant.
5466. The device of item 5277, further comprising an echogenic material.
5467. The device of item 5277, further comprising an echogenic material, wherein the echogenic material is in the form of a coating. 5468. The device of item 5277 wherein the device is sterile.
5469. The device of item 5277 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device.
5470. The device of item 5277 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is connective tissue.
5471. The device of item 5277 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is muscle tissue.
5472. The device of item 5277 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is nerve tissue.
5473. The device of item 5277 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device, wherein the tissue is epithelium tissue.
5474. The device of item 5277 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from the time of deployment of the device to about 1 year.
5475. The device of item 5277 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from about 1 month to 6 months. 5476. The device of item 5277 wherein the anti-scarring agent is released in effective concentrations from the device over a period ranging from about 1 - 90 days.
5477. The device of item 5277 wherein the anti-scarring agent is released in effective concentrations from the device at a constant rate.
5478. The device of item 5277 wherein the anti-scarring agent is released in effective concentrations from the device at an increasing rate.
5479. The device of item 5277 wherein the anti-scarring agent is released in effective concentrations from the device at a decreasing rate.
5480. The device of item 5277 wherein the anti-scarring agent is released in effective concentrations from the composition comprising the anti-scarring agent by diffusion over a period ranging from the time of deployment of the device to about 90 days.
5481. The device of item 5277 wherein the anti-scarring agent is released in effective concentrations from the composition comprising the anti-scarring agent by erosion of the composition over a period ranging from the time of deployment of the device to about 90 days.
5482. The device of item 5277 wherein the device comprises about 0.01 Dg to about 10 Dg of the anti-scarring agent.
5483. The device of item 5277 wherein the device comprises about 10 Dg to about 10 mg of the anti-scarring agent.
5484. The device of item 5277 wherein the device comprises about 10 mg to about 250 mg of the anti-scarring agent. 5485. The device of item 5277 wherein the device comprises about 250 mg to about 1000 mg of the anti-scarring agent.
5486. The device of item 5277 wherein the device comprises about 1000 mg to about 2500 mg of the anti-scarring agent.
5487. The device of item 5277 wherein a surface of the device comprises less than 0.01 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring .agent is applied.
5488. The device of item 5277 wherein a surface of the device comprises about 0.01 Dg to about 1 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
5489. The device of item 5277 wherein a surface of the device comprises about 1 Dg to about 10 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
5490. The device of item 5277 wherein a surface of the device comprises about 10 Dg to about 250 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
5491. The device of item 5277 wherein a surface of the device comprises about 250 Dg to about 1000 Dg of the anti-scarring agent of anti- scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
5492. The device of item 5277 wherein a surface of the device comprises about 1000 Dg to about 2500 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied. 5493. A method for inhibiting scarring comprising placing a device that comprises a medical device (e.g., an implant) and an anti- scarring agent or a composition comprising an anti-scarring agent into an animal host, wherein the agent inhibits scarring.
5494. The method of item 5493 wherein the agent is an adensosine A2A receptor antagonist.
5495. The method of item 5493 wherein the agent is an AKT inhibitor.
5496. The method of item 5493 wherein the agent is an alpha 2 integrin antagonist, wherein the alpha 2 integrin antagonist is Pharmaprojects No. 5754 (Merck KgaA).
5497. The method of item 5493 wherein the agent is an alpha 4 integrin antagonist.
5498. The method of item 5493 wherein the agent is an alpha 7 nicotinic receptor agonist.
5499. The method of item 5493 wherein the agent is an angiogenesis inhibitor selected from the group consisting of AG-12,958 (Pfizer), ATN-161 (Attention LLC), neovastat, an angiogenesis inhibitor from Jerina AG (Germany), NM-3 (Mercian), VGA-1155 (Taisho), FCE-26644 (Pfizer), FCE-26950 (Pfizer), FPMA (Meiji Daries), FR-111142 (Fujisawa), GGTI-298, GM-1306 (Ligand), GPA-1734 (Novartis), NNC-47-0011 (Novo Nordisk), herbamycin (Nippon Kayaku), lenalidomide (Celegene), IP-10 (NIH), ABT-828 (Abbott), KIN-841 (Tokushima University, Japan), SF-1126 (Semafore Pharmaceuticals), laminin technology (NIH), CHIR-258 (Chiron), NVP-AEW541 (Novartis), NVP-AEW541 (Novartis), Vt16907 (Alchemia), OXI-8007 (Oxigene), EG-3306 (Ark Therapeutics), Maspin (Arriva), ABT- 567 (Abbott), PPI-2458 (Praecis Pharmaceuticals), CC-5079, CC-4089 (Celgene), HIF-1 alpha inhibitors (Xenova), S-247 (Pfizer), AP-23573 (Ariad), AZD-9935 (Astra Zeneca), mebendazole (Introgen Therapeutics), MetAP-2 inhibitors (GlaxoSmithKline), AG-615 (Angiogene Pharmaceuticals), Tie-2 antagonists (Hybrigenics), NC-381 , CYC-381, NC-169, NC-219, NC-383, NC-384, NC-407 (Lorus Therapeutics), ATN-224 (Attenuon), ON-01370 (Onconova), Vitronectin antagonists (Amgen), SDX-103 (Salmedix), Vitronectin antagonists (Shire), CHP (Riemser), TEK (Amgen), Anecortave acetate (Alcon), T46.2 (Matrix Therapeutics), HG-2 (Heptagen), TEM antagonists (Genzyme), Oxi-4500 (Oxigene), ATN-161 (Attenuon), WX-293 (Wilex), M-2025 (Metris Therapeutics), Alphastatin (BioActa), YH-16 (Yantai Rongchang), BIBF-1120 (Boehringer Ingelheim), BAY-57-9352 (Bayer), AS- 1404 (Cancer Research Technology), SC-77964 (Pfizer), glycomimetics (BioTie Therapies), TIE-2 Inhibitors (Ontogen), DIMI, Octamer (Octamer), ABR-215050 (Active Biotech), ABT=518 (Abbott), KDR inhibitors (Abbott), BSF-466895 (Abbott), SCH-221153 (Schering-Plough), DAC:antiangiogenic (ConjuChem), TFPI (EntreMed), AZD-2171 (Astra-Zenaca), CDC-394 (Celgene), LY290293 (EIi Lilly), IDN-5390 (Indena), Kdr Kinase Inhibitors (Merck), CT-113020, CT-116433, CT-116563, CT-31890, CT-32228) (Cell Therapeutics), A-299620 (Abbott), TWEAK Inhibitor (Amgen), VEGF modulators (Johnson and Johnson), Tum-N53, tumstatin (Genzyme), Thios- 1, Thios-2 (Thios Pharmaceuticals), MV-6401 (Miravant Medical Technologies), Spisulosine (PharmaMar), CEP-7055 (Cephalon), AUV-201 (Auvation), LM-609 (EIi Lilly), SKF-106615 (AnorMED), Oglufanide disodium (Cytran), BW-114 (Pharminox), Calreticulin (NIH), WX-678 (Wilex), SD-7784 (Pfizer), WX-UK1 (Wilex), SH-268 (Schering AG), 2-Me-PGA (Celgene), S- 137 (Pfizer), ZD-6126 (Angiogene Pharmaceuticals), SG-292 (SignalGen), Benefin (Lane Labs), A6, A36 (Angstrom), SB-2723005 (GlaxoSmithKline), SC-7 (Cell Therapeutics), ZEN-014 (AEterna Zentaris), 2-methoxyestradiol (EntreMed), NK-130119 (Nippon Kayaku), CC-10004 (Celgene), AVE- 8062A (Ajinomoto), Tacedinaline (Pfizer), Actinonin (Tokyo Metropolitan Institute of Medical Science), Lenalidomide (Celgene), VGA-1155, BTO-956 (SRI International), ER-68203-00 (Eisai), CT-6685 (UCB), JKC-362 (Phoenix Pharmaceuticals), DMI-3798 (DMI Biosciences, Angiomate (Ipsen), ZD-6474 (AstraZeneca), CEP-5214 (Cephalon), Canstatin (Genzyme), NM-3 (Mercian), Oridigm (MediQuest Therapeutics), Exherin (Adherex), BLS-0597 (Boston Life Sciences), PTC-299 (PTC Therapeutics), NPI-2358 (Nereus Pharmaceuticals), CGP-79787 (Novartis), AEE-788 (Novartis), CKD-732 (Chong Kun Dang), CP-564959 (OSI Pharmaceuticals), CM-101 (CarboMed), CT-2584, CT3501 (Cell Therapeutics), combretastatin and analogues and derivatives thereof (Oxigene), Rebimastat (Bristol-Meyers Squibb), Dextrin 2-sulfate (ML Laboratories), Cilengitide (Merk KGaA), NSC-706704 (Pharminox), KRN- 951 (Kirin Brewery), Ukrain, NSC-631570 (Nowicky Pharma), Tecogalan sodium (Daiichi Pharmaceutical), Tz-93 (Tsumura), TBC-1635 (Encysive Pharmaceuticals), TAN-1120 (Takeda),_Semaxanib (Pfizer), BDI-7800 (Biopharmacopae), SD-186, SD-983 (Bristol-Meyers Squibb), SB-223245 (GlaxoSmithKline), SC-236 (Pfizer), RWJ-590973 (Johnson and Johnson), ILX-1850 (Genzyme), SC-68488, S-836 (Pfizer), CG-55069-11 (CuraGen), Ki-23057 (Kirin Brewery), CCX-700 (Chemoentryx), Pegaptanib octasodium (Giled Sciences), ANGIOCOL (available from Biostratum Inc.), or an analogue or derivative thereof.
5500. The method of item 5493 wherein the agent is an apoptosis antagonist.
5501. The method of item 5493 wherein the agent is an apoptosis activator.
5502. The method of item 5493 wherein the agent is a beta 1 integrin antagonist. 5503. The method of item 5493 wherein the agent is a beta tubulin inhibitor.
5504. The method of item 5493 wherein the agent is a blocker of enzyme production in Hepatitis C.
5505. The method of item 5493 wherein the agent is a Bruton's tyrosine kinase inhibitor.
5506. The method of item 5493 wherein the agent is a calcineurin inhibitor.
5507. The method of item 5493 wherein the agent is a caspase 3 inhibitor.
5508. The method of item 5493 wherein the agent is a CC chemokine receptor antagonist.
5509. The method of item 5493 wherein the agent is a cell cycle inhibitor selected from the group consisting of SNS-595 (Sunesis), synthadotin, KRX-0403, and an analogue or derivative thereof.
5510. The method of item 5493 wherein the agent is a cathepsin B inhibitor.
5511. The method of item 5493 wherein the agent is a cathepsin K inhibitor, wherein the cathepsin K inhibitor is 462795 (GlaxoSmithKline), INPL-022-D6 (Amura Therapeutics), or an analogue or derivative thereof. 5512. The method of item 5493 wherein the agent is a cathepsin L inhibitor.
5513. The method of item 5493 wherein the agent is a CD40 antagonist.
5514. The method of item 5493 wherein the agent is a chemokine receptor agonist.
5515. The method of item 5493 wherein the agent is a chymase inhibitor.
5516. The method of item 5493 wherein the agent is a collagenase antagonist.
5517. The method of item 5493 wherein the agent is a CXCR antagonist.
5518. The method of item 5493 wherein the agent is a cyclin dependent kinase inhibitor selected from the group consisting of a CDK-1 inhibitor, a CDK-2 inhibitor, a CDK- 4 inhibitor, a CDK-6 inhibitor, a CAK1 inhibitor from GPC Biotech and Bristol-Myers Squibb, RGB-286199 (GPC Biotech), an anticancer agent from Astex Technology, a CAK1 inhibitor from GPC Biotech, a CDK inhibitor from Sanofi-Aventis, a CDK1/CDK2 inhibitor from Amgen, a CDK2 inhibitor from SUGEN-2 (Pfizer), a hearing loss therapy agent (Sound Pharmaceuticals), PD-0332991 (Pfizer), RGB-286199 (GPC Biotech), Ro-0505124 (Hoffmann-La Roche), a Ser/Thr kinase inhibitor from Lilly (EIi Lilly), CVT-2584 (CAS No. 199986-75-9) (CV Therapeutics), and an analogue or derivative thereof. 5519. The method of item 5493 wherein the agent is a cyclooxygenase 1 inhibitor.
5520. The method of item 5493 wherein the agent is a DHFR inhibitor.
5521. The method of item 5493 wherein the agent is a dual integrin inhibitor.
5522. The method of item 5493 wherein the agent is an elastase inhibitor.
5523. The method of item 5493 wherein the agent is an elongation factor-1 alpha inhibitor.
5524. The method of item 5493 wherein the agent is an endothelial growth factor antagonist.
5525. The method of item 5493 wherein the agent is an endothelial growth factor receptor kinase inhibitor selected from the group consisting of sorafenib tosylate (Bayer), AAL-993 (Novartis), ABP-309 (Novartis), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), E- 7080 (Eisai), EG-3306 (Ark Therapeutics), EXEL-2880 (Exelixis), GW- 654652 (GlaxoSmithKline), a KDR inhibitor from LG Life Sciences, CT-6685 and CT-6729 (UCB), KRN-633 and KRN-951 (Kirin Brewery), OSI-930 (OSI Pharmaceuticals), SP-5.2 (Supratek Pharma), SU-11657 (Pfizer), a Tie-2 antagonist (Hybrigenics), a VEGFR-2 kinase inhibitor (Bristol-Myers Squibb), XL-647 (Exelixis), a KDR inhibitor from Abbott Laboratories, sorafenib tosylate, and an analogue or derivative thereof. 5526. The method of item 5493 wherein the agent is an endotoxin antagonist.
5527. The method of item 5493 wherein the agent is an epothilone and tubulin binder.
5528. The method of item 5493 wherein the agent is an estrogen receptor antagonist.
5529. The method of item 5493 wherein the agent is an FGF inhibitor.
5530. The method of item 5493 wherein the agent is a farnexyl transferase inhibitor.
5531. The method of jtem 5493 wherein the agent is^ farnesyltransferase inhibitor selected from the group of A-197574 (Abbott), a farnesyltransferase inhibitor from Servier, FPTIII (Strathclyde Institute for Drug R), LB-42908 (LG Life Sciences), Pharmaprojects No. 5063 (Genzyme), Pharmaprojects No. 5597 (Ipsen), Yissum Project No. B-1055 (Yϊssum), and an analogue or derivative thereof.
5532. The method of item 5493 wherein the agent is an FLT-3 kinase inhibitor.
5533. The method of item 5493 wherein the agent is an FGF receptor kinase inhibitor.
5534. The method of item 5493 wherein the agent is a fibrinogen antagonist selected from the group consisting of AUV-201 (Auvation), MG-13926 (Sanofi-Aventis), plasminogen activator (CAS No. 105913-11-9) (from Sanofi-Aventis or UCB), plasminogen activator-2 (tPA- 2) (Sanofi-Aventis), pro-urokinase (CAS No. 82657-92-9) (Sanofi-Aventis), and an analogue or derivative thereof.
5535. The method of item 5493 wherein the agent is a heat shock protein 90 antagonist selected from the group consisting of SRN-005 (Sirenade), geldanamycin, NSC-33050 (17-allylaminogeldanamycin; 17- AAG), 17-dimethylaminoethylamino-17-demethoxy-geldanamycin (17- DMAG), rifabutin (rifamycin XIV, r,4-didehydro-1-deoxy-1 ,4-dihydro-5'-(2- methylpropyl)-1-oxo-), and an analogue or derivative thereof.
5536. The method of item 5493 wherein the agent is a histone deacetylase inhibitor.
5537. The method of item 5493 wherein the agent is an HMGCoA reductase inhibitor selected from the group consisting of an atherosclerosis therapeutic from Lipid Sciences, ATI-16000 (ARYx Therapeutics), KS-01-019 (Kos Pharmaceuticals), Pharmaprojects No. 2197 (Sanofi-Aventi), RP 61969 (Sanofi-Aventis), and an analogue or derivative thereof.
5538. The method of item 5493 wherein the agent is an ICAM inhibitor.
5539. The method of item 5493 wherein the agent is an IL, ICE and IRAK antagonist, wherein the antagonist is a CJ-14877, CP-424174 (Pfizer), NF-61 (Negma-Lerads), and an analogue or derivative thereof.
5540. The method of item 5493 wherein the agent is an IL-2 inhibitor. 5541. The method of item 5493 wherein the agent is an immunosuppressant selected from the group consisting of teriflunomide (Sanofi Aventis), chlorsulfaquinoxalone (NSC-339004), chlorsulfaquinoxalone sulfate, CS-712 (Sankyo), ismomultin alfa (CAS No. 457913-93-8) (Akzo Nobel), antiallergics from GenPatTT, antiinflammatories or AT-005 (Androclus Therapeutics), autoimmune disease therapeutics from EpiVax, BN-007 (Bone), budesonide (CAS No. 51333-22- 3) (MAP Pharmaceuticals), CO-14 (Genzyme), edratide (CAS No. 433922- 67-9) (Teva), EP-314 (Enanta), eprovafen (CAS No. 101335-99-3) (Sanofi- Aventis), HWA-131 (CAS No. 118788-41-3) (Sanofi-Aventis), immunomodulators from MerLion Pharmaceuticals, immunosuppressives from Alchemia, IPL-12 (Inflazyme), MDL-9563 (CAS No. 27086-86-8) (Sanofi-Aventis), Pharmaprojects No. 2330 (Sanofi-Aventis), Pharmaprojects No. 6426 (Abgenix), PXS-25 (Pharmaxis), rosmarinic acid (CAS No. 20283-92-5) (Sanofi-Aventis), RP 42927 or RP 54745 (CAS No. 135330-08-4) (Sanofi-Aventis), SGN-35 (Seattle Genetics), ST-1959 (Sigma-Tau), type I diabetes therapy from SYNX Pharma, UNIL-88 (Debiopharm), VP-025 (Vasogen), VR-694 (Vectura), PRTX-001 (Protalex), and an analogue or derivative thereof.
5542. The method of item 5493 wherein the agent is an IMPDH (inosine monophosphate).
5543. The method of item 5493 wherein the agent is an integrin antagonist.
5544. The method of item 5493 wherein the agent is an interleukin antagonist.
5545. The method of item 5493 wherein the agent is an inhibitor of type III receptor tyrosine kinase. 5546. The method of item 5493 wherein the agent is an irreversible inhibitor of enzyme methionine aminopeptidase type 2.
5547. The method of item 5493 wherein the agent is an isozyme selective delta protein kinase C inhibitor.
5548. The method of item 5493 wherein the agent a JAK3 enzyme inhibitor.
5549. The method of item 5493 wherein the agent is a JNK inhibitor.
5550. The method of item 5493 wherein the agent is a kinase inhibitor.
5551. The method of item 5493 wherein the agent is kinesin antagonist.
5552. The method of item 5493 wherein the agent is a kinesin antagonist.
5553. The method of item 5493 wherein the agent is a leukotriene inhibitor and antagonist selected from the group consisting of ambicromil (CAS No. 58805-38-2) (Sanofi-Aventis), amelubant (CAS No. 346735-24-8) (Boehringer Ingelheim), DW-1141 (Dong Wha), ebselen (Daiichi Pharmaceutical), ibudilast (Kyorin), leucotriene inhibitors from Sanofi-Aventis, lymphotoxin-beta receptor (LT-D) from Biogen Idee, Pharmaprojects No. 1535 and 2728 (CAS No. 119340-33-9) (Sanofi- Aventis), R-112 (Rigel), Rev-5367 (CAS No. 92532-05-3) (Sanofi-Aventis), RG-14893 (CAS No. 141835-49-6) (Sanofi-Aventis), RG-5901-A (CAS No. 101910-24-1), 92532-23-5, RP 66153 (CAS No. 142422-79-5), RP 66364 (CAS No.186916-92-5), RP 69698 (CAS No. 141748-00-7) (Sanofi-Aventis), SC-411930 (Pfizer), SC-41930 (CAS No. 120072-59-5) (Pfizer), SC-50605 (CAS No. 138828-39-4) (Pfizer), SC-51146 (CAS No. 141059-52-1), SC- 53228 (CAS No. 153633-01-3) (Pfizer), spaglumic acid (ZY-15106) (CAS No.3106-85-2), 80619-64-3 (Novartis), tipredane (CAS No. 85197-77-9) (Bristol-Myers Squibb), U-75302 (CAS No. 119477-85-9) (Pfizer), and analogue or derivative thereof.
5554. The method of item 5493 wherein the agent is an MAP kinase inhibitor.
5555. The method of item 5493 wherein the agent is a matrix metalloproteinase inhibitor.
5556. The method of item 5493 wherein the agent is an MCP- CCR2 inhibitor.
5557. The method of item 5493 wherein the agent is an mTOR inhibitor.
5558. The method of item 5493 wherein the agent is an mTOR kinase inhibitor.
5559. The method of item 5493 wherein the agent is a microtubule inhibitor selected from the group consisting of antibody- maytansinoid conjugates from Biogen Idee, colchicines (MantiCore Pharmaceuticals), anticancer immunoconjugates from Johnson & Johnson, DIME from Octamer, gni-1f (GNI), huC242-DM4, huMy9-6-DM1 (ImmunoGen), IDN-5404 (Indena), IMO-098, IMOderm (Imotep), mebendazole (Introgen Therapeutics), microtubule poisons from Cambridge Enterprise, paclitaxel such as LOTAX from Aphios (CAS No. 33069-62-4), Genexol-PM from Samyang, Pharmaprojects No. 6383 (Tapestry Pharmaceuticals), RPR-112378 (Sanofi-Aventis), SGN-75 (Seattle Genetics), SPL-7435 (Starpharma), SSR-250411 (Sanofi-Aventis), trastuzumab-DM1 (Genentech), vinorelbine, and an analogue or derivative thereof.
5560. The method of item 5493 wherein the agent is an MIF inhibitor.
5561. The method of item 5493 wherein the agent is an MMP inhibitor.
5562. The method of item 5493 wherein the agent is a neurokinin (NK) antagonist selected from the group consisting of anthrotainin (CAS No. 148084-40-6) (Sanofi-Aventis), an IBS thereapeutic from ArQuIe, MDL-105212A (CAS No. 167261-60-1) (Ssanofi-Aventis), Pharmaprojects No. 2744, 3258 (CAS No. 139167-47-8) 4006, 4201, or 5986 (Sanofi-Aventis), RP 67580 (CAS No. 135911-02-3), SR-144190 (CAS No. 201152-86-5), SSR-240600, SSR-241586 (Sanofi-Aventis), TKA-457 (Novartis), vestipitant mesylate (CAS No. 334476-64-1) (GlaxoSmithKline), Win-64821 (Sanofi-Aventis), PRX-96026 (Predix Pharmaceuticals), and an analogue or derivative thereof.
5563. The method of item 5493 wherein the agent is an NF kappa B inhibitor selected from the group consisting of AVE-0545 or AVE- 0547 (Sanofi-Aventis), bortezomib (CAS No. 179324-69-7) (Millennium Pharmaceuticals), dexanabinol (CAS No. 112924-45-5) (Pharmos), dexlipotam (Viatris), Pharmaprojects No. 6283 (INDRA) (OXiGENE), IPL- 576092 (CAS No. 137571-30-3) (Inflazyme), NFKB decoy (Corgentech), NFKB decoy oligo (AnGes MG), NFKB's from Ariad, osteoporosis treatments or S5 (F005) from Fulcrum Pharmaceuticals, P61 (Phytopharm), R-flurbiprofen (CAS No. 5104-49-4) (Encore Pharmaceuticals), and an analogue or derivative thereof.
5564. The method of item 5493 wherein the agent is a nitric oxide agonist.
5565. The method of item 5493 wherein the agent is an ornithine decarboxylase inhibitor.
5566. The method of item 5493 wherein the agent is a p38 MAP kinase inhibitor selected from the group consisting of AZD-6703 (AstraZeneca), JX-401 (Jexys Pharmaceuticals), BMS-2 (Bristol-Myers Squibb), a p38 MAP kinase inhibitor from Novartis, a p38-alpha MAP kinase inhibitor from Amphora, Pharmaprojects No. 5704 (Pharmacopeia), RPR- 200765A (Sanofi-Aventis), SD-282 (Johnson & Johnson), TAK-715 (Takeda), and an analogue or derivative thereof.
5567. The method of item 5493 wherein the agent is a palmitoyl-protein thioesterase inhibitor.
5568. The method of item 5493 wherein the agent is a PDGF receptor kinase inhibitor selected from the group consisting of AAL-993, AMN-107, or ABP-309 (Novartis), AMG-706 (Amgen), BAY-57-9352 (Bayer), CDP-860 (UCB), E-7080 (Eisai), imatinib (CAS No. 152459-95-5) (Novartis), OSI-930 (OSI Pharmaceuticals), RPR-127963E (Sanofi-Aventis), RWJ-540973 (Johnson & Johnson), sorafenib tosylate (Bayer), SlM 1657 (Pfizer), tandutinib (CAS No. 387867-13-2) (Millennium Pharmaceuticals), vatalanib (Novartis), ZK-CDK (Schering AG), and an analogue or derivative thereof. 5569. The method of item 5493 wherein the agent is (-)- halofenate (Metabolex), AMG-131 (Amgen), antidiabetics from Japan Tobacco, AZD-4619, AZD-8450, AZD-8677 (AstraZeneca), DRF-10945, balaglitazone (Dr Reddy's), CS-00088, CS-00098 (Chipscreen Biosciences), E-3030 (Eisai), etalocib (CAS No. 161172-51-6) (EIi Lilly), GSK-641597 (Ligand), GSK-677954 (GlaxoSmithKline), GW-409544 (Ligand), GW- 590735 (GlaxoSmithKline), K-111 (Hoffmann-La Roche), LY-518674 (EIi Lilly), LY-674 (Ligand), LY-929 (Ligand), MC-3001 , MC-3002 (MaxoCore Pharmaceuticals), metformin HCI + pioglitazone (CAS No. 1115-70-4 and
112529-15-4), ACTOPLUS MET from Andrx), muraglitazar (CAS No. 331741-94-7) (Bristol-Myers Squibb), naveglitazar (Ligand), oleoylethanolamide (Kadmus Pharmaceuticals), ONO-5129, pioglitazone hydrochloride (CAS No. 111025-46-8 and 112529-15-4) (Takeda), PLX-204 (Plexxikon), PPAR agonists from Genfit, PPAR delta agonists from EIi Lilly, PPAR-alpha agonists from CrystalGenomics, PPAR-gamma modulators and PPAR-β modulators from CareX, rosiglitazone maleate (CAS No. 122324- 73-4 or 155141-29-0) (GlaxoSmithKline), rosiglitazone maleate/glimepir (CAS No. 155141-29-0 and 93479-97-1), AVANDARYL, rosiglitazone maleate/metformin extend (CAS No. 155141-29-0 and 657-24-9), AVANDAMET, rosiglitazone maleate+metformin, AVANDAMET (GlaxoSmithKline), tesaglitazar (AstraZeneca), and an analogue or derivative thereof.
5570. The method of item 5493 wherein the agent is a phosphatase inhibitor.
5571. The method of item 5493 wherein the agent is a phosphodiesterase (PDE) inhibitor selected from the group consisting of avanafil (Tanabe Seiyaku), dasantafil (CAS No. 569351-91-3) (Schering- Plough), A-906119 (CAS No. 134072-58-5), DL-850 (Sanofi-Aventis), GRC- 3015, GRC-3566, GRC-3886 (Glenmark), HWA-153 (CAS No. 56395-66-5) (Sanofi-Aventis), hydroxypumafentrine (Altana), IBFB-130011 , IBFB-14-016, IBFB-140301 , IBFB-150007, IBFB-211913 (IBFB Pharma), L-826141 (Merck & Co), medorinone (CAS No. 88296-61-1) (Sanofi-Aventis), MEM- 1917 (Memory Pharmaceuticals), ND-1251 (Neuro3d), PDE inhibitors from ICOS, PDE IV inhibitors from Memory Pharmaceuticals and CrystalGenomics, Pharmaprojects No. 2742 and 6141 (Sanofi-Aventis), QAD-171 (Novartis), RHC-2963 (CAS No. 76993-12-9 and 76993-14-1), RPR-117658, RPR-122818 derivatives, SR-24870 , and RPR-132294 (Sanofi-Aventis), SK-350 (ln2Gen), stroke therapy agents from deCODE Genetics, TAS-203 (Taiho), tofimilast (CAS No. 185954-27-2) (Pfizer), UK- 371800 (Pfizer), WIN-65579 (CAS No. 158020-82-7) (Sanofi-Aventis), IBFB- 130020 (IBFB Pharma), OPC-6535 (CAS No. 145739-56-6) (Otsuka), a phosphodiesterase III inhibitor, enoximone, a phosphodiesterase IV inhibitor, fosfosal, Atopik (Barrier Therapeutics), triflusal, a phosphodiesterase V inhibitor, and an analogue or derivative thereof.
5572. The method of item 5493 wherein the agent is a PKC inhibitor.
5573. The method of item 5493 wherein the agent is a platelet activating factor antagonist.
5574. The method of item 5493 wherein the agent is a platelet-derived growth factor receptor kinase inhibitor.
5575. The method of item 5493 wherein the agent is a prolyl hydroxylase inhibitor.
5576. The method of item 5493 wherein the agent is a polymorphonuclear neutrophil inhibitor. 5577. The method of item 5493 wherein the agent is a protein kinase B inhibitor.
5578. The method of item 5493 wherein the agent is a protein kinase C stimulant.
5579. The method of item 5493 wherein the agent is a purine nucleoside analogue.
5580. The method of item 5493 wherein the agent is a purinoreceptor P2X antagonist.
5581. The method of item 5493 wherein the agent is a Raf kinase inhibitor.
5582. The method of item 5493 wherein the agent is a reversible inhibitor of ErbB1 and ErbB2.
5583. The method of item 5493 wherein the agent is a ribonucleoside triphosphate reductase inhibitor.
5584. The method of item 5493 wherein the agent is an SDF- 1 antagonist.
5585. The method of item 5493 wherein the agent is a sheddase inhibitor.
5586. The method of item 5493 wherein the agent is an SRC inhibitor. 5587. The method of item 5493 wherein the agent is a stromelysin inhibitor.
5588. The method of item 5493 wherein the agent is an Syk kinase inhibitor.
5589. The method of item 5493 wherein the agent is a telomerase inhibitor.
5590. The method of item 5493 wherein the agent is a TGF beta inhibitor selected from the group consisting of pirfenidone (CAS No. 53179-13-8) (MARNAC), tranilast (CAS No. 53902-12-8) (Kissei), IN-1130 (ln2Gen), mannose-6-phosphate (BTG), TGF-β antagonists from Inflazyme (Pharmaprojects No. 6075), TGF-β antagonists from Sydney, non-industrial source), TGF-β I receptor kinase inhibitors from EIi Lilly, TGF-β receptor inhibitors from Johnson & Johnson, and an analogue or derivative thereof.
5591. The method of item 5493 wherein the agent is a TN Fa antagonist or TACE inhibitor selected from the group consisting of adalimumab (CAS No. 331731-18-1) (Cambridge Antibody Technology), AGIX-4207 (AtheroGenics), AGT-1 (Advanced Biotherapy), an antiinflammatory from Borean Pharma, Cellzome, or Paradigm Therapeutics, anti-inflammatory vaccine (TNF-alpha kinoid) from Neovacs, humanized anti-TNF antibody or an anti-TNF MAb (CB0006) Celltech (UCB), apratastat (CAS No. 287445-51-0) (Wyeth), BMS-561392 (Bristol-Myers Squibb), BN- 006 (Bone), certolizumab pegol (CAS No. 428863-50-7 or CH-138 (UCB), cilomilast (CAS No. 153259-65-5) (GlaxoSmithKline), CR-1 (Nuada Pharmaceuticals), CRx-119 (CombinatoRx), D-5410 (UCB), dacopafant (CAS No. 125372-33-0) (Sanofi-Aventis), dersalazine (CAS No. 188913-57- 7/188913-58-8) (Uriach), etanercept (CAS No. 185243-69-0) (Amgen), ethyl pyruvate (Critical (Critical Therapeutics), golimumab (CAS No. 476181-74-5) (Johnson & Johnson), hormono-immunotherapy from Ipsen, CDP571 {e.g., Humicade from UCB), IC-485 (ICOS), infliximab (CAS No. 170277-31-3) (Johnson & Johnson), IP-751 (Manhattan Pharmaceuticals), ISIS-104838 (CAS No. 250755-32-9) (ISIS Pharmaceuticals), lenalidomide (CAS No. 191732-72-6) (Celgene), lentinan (CAS No. 37339-90-5) (Ajinomoto), MDL- 201112 (CAS No. 142130-73-2) (Sanofi-Aventis), medroxyprogesterone (CAS No. 520-85-4) (InKine Pharmaceutical), N-acetylcysteine (CAS No. 616-91-1) (Zambon), NBE-P2 (DIREVO Biotech), nereiimomab (CAS No. 162774-06-3) (Chiron), OM-294DP (OM PHARMA), onercept (CAS No. 199685-57-9) (Yeda), PASSTNF-alpha (Verigen), pentoxifylline or oxypentifylline (Sanofi-Aventis), Pharmaprojects No. 4091, 4241 , 4295, or 4488 (Sanofi-Aventis), Pharmaprojects No. 5480 (Amgen), Pharmaprojects No. 6749 (Cengent), pirfenidone (CAS No. 53179-13-8) (MARNAC), RPR- 132294 (Sanofi-Aventis), S5 (F002) (Fulcrum Pharmaceuticals), simvastatin (CAS No. 79902-63-9) (Merck & Co), STA-6292 (Synta Pharmaceuticals), tacrolimus (CAS No. 104987-11 -3) (Fujisawa LifeCycle Pharma), talactoferrin alfa (CAS No. 308240-58-6) (Agennix), thalidomide (CAS No. 50-35-1) (Celgene), TNF antagonists form ProStrakan, and Synergen, TNF inhibitors (Amgen), TNF-alpha antagonists from Dynavax Technologies and Jerina AG (Germany), TNF-alpha inhibitors from IBFB Pharma and Xencor (Xencor), torbafylline (CAS No. 105102-21-4) (Sanofi-Aventis), UR-1505 (Uriach), VT-346 (Viron Therapeutics), YSIL6 (Y's Therapeutics), YSTH2 (Y's Therapeutics), NPI-1302a-3 (Nereus Pharmaceuticals, a TNF antagonist from Jerina AG (Germany), dersalazine, and an analogue or derivative thereof.
5592. The method of item 5493 wherein the agent is a To)I receptor inhibitor.
5593. The method of item 5493 wherein the agent is a tubulin antagonist. 5594. The method of item 5493 wherein the agent is a tyrosine kinase inhibitor selected from the group consisting of SU-011248, SUTENT from Pfizer Inc. (New York, NY)1 BMS-354825, PN-355 (Paracelsian Pharmaceuticals), AGN-199659 (Allergan), AAL-993 or ABP- 309 (Novartis), adaphostin (NIH), AEE-788 (Novartis), AG-013736 (OSI Pharmaceuticals), AG-13736 (Pfizer), ALT-110 (Alteris Therapeutics), AMG- 706 (Amgen), anticancer MAbs from Xencor, anti-EGFrvlll MAbs from Abgenix, anti-HER2 MAb from Abiogen, AZD-2171 or AZD-9935 (AstraZeneca), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), CEP-5214 (Cephalon), CEP-7055 (Cephalon), cetuximab (ImClone Systems), CHIR-200131 and CHIR-258 (Chiron), CP-547632 (OSI Pharmaceuticals), CP-724714 (Pfizer), CT-301 (Creabilis Therapeutics), D- 69491 (Baxter International), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EGFR/ErbB2 inhibitors from Array BioPharma, erlotinib (CAS No. 183319- 69-9) (OSI Pharmaceuticals), EXEL-2880 (Exelixis), FK-778 (Sanofi- Aventis) , gefitinib (CAS No. 184475-35-2) (AstraZeneca), GW-2286 or GW- 654652 (GlaxoSmithKline), her2/neu antigen from AlphaVax, HER-2/neu inhibitor from Generex, Herzyme (Medipad) (Sirna Therapeutics), HKl-272 (Wyeth), HuMax-EGFr (Genmab), idronoxil (CAS No. 81267-65-4) (Novogen), IGF-1 inhibitors from Ontogen, IMC-11 F8 (ImClone Systems), kahalalide F (CAS No. 149204-42-2) (PharmaMar), KDR inhibitor from LG Life Sciences, KDR inhibitors from Abbott Laboratories, KDR kinase inhibitors (UCB), Kdr kinase inhibitors from Merck & Co, KRN-633 and KRN- 951 (Kirin Brewery), KSB-102 (Xenova), lapatinib ditosylate (CAS No. 388082-78-8) (GlaxoSmithKline), matuzumab (Merck KGaA), MDX-214 (Medarex), ME-103 (Pharmexa), MED-A300 (Gerolymatos), MNAC-13 (Lay Line Genomics), nimotuzumab (Center of Molecular Immunology), NSC- 330507 or NSC-707545 (NlH), NV-50 (Novogen), OSI-930 (OSI Pharmaceuticals), panitumumab (Abgenix), pelitinib (CAS No. 287933-82-7) (Wyeth), pertuzumab (CAS No. 380610-27-5) (Genentech), Pharmaprojects No. 3985 (Sanofi-Aventis), prostate cancer therapeutics from Sequenom (SQPC35, SQPC36, SQPC90), removab and remoxab (Trion Pharma), RG- 13022 (CAS No. 136831-48-6), RG-13291 (CAS No. 138989-50-1), or RG- 14620 (CAS No. 136831-49-7) (Sanofi-Aventis), RM-6427 (Romark), RNAi breast cancer therapy from Benitec, RP 53801 (CAS No. 125882-88-4) (Sanofi-Aventis), sorafenib tosylate (Bayer), SU-11657 (Pfizer), Tie-2 antagonists from Semaia (Hybrigenics), Tie-2 inhibitors from Ontogen, trastuzumab (CAS No. 180288-69-1) (Genentech), tyrosine kinase inhibitors from Sanofi-Aventis, U3-1287, U3-1565, U3-1784, U3-1800 (U3 Pharma), vatalanib (Novartis), VEGFR-2 kinase inhibitor from Bristol-Myers Squibb, XL-647 (Exelixis), ZD-6474 (AstraZeneca), ZK-CDK (Schering AG), an EGFR tyrosine kinase inhibitor, EKB-569 (Wyeth), and an analogue or derivative thereof.
5595. The method of item 5493 wherein the agent is a VEGF inhibitor.
5596. The method of item 5493 wherein the agent is a vitamin D receptor agonist.
5597. The method of item 5493 wherein the agent is ZD-6474 (an angiogenesis inhibitor).
5598. The method of item 5493 wherein the agent is AP- 23573 (an mTOR inhibitor).
5599. The method of item 5493 wherein the agent is synthadotin (a tubulin antagonist).
5600. The method of item 5493 wherein the agent is S-0885 (a collagenase inhibitor). 5601. The method of item 5493 wherein the agent is aplidine (an elongation factor-1 alpha inhibitor).
5602. The method of item 5493 wherein the agent is ixabepilone (an epithilone).
5603. The method of item 5493 wherein the agent is IDN- 5390 (an angiogenesis inhibitor).
5604. The method of item 5493 wherein the agent is SB- 2723005 (an angiogenesis inhibitor).
5605. The method of item 5493 wherein the agent is ABT-518 (an angiogenesis inhibitor).
,. -_. .. 5606. The methαcLαf item 5493 wherein the agent is combretastatin (an angiogenesis inhibitor).
5607. The method of item 5493 wherein the agent is anecortave acetate (an angiogenesis inhibitor).
5608. The method of item 5493 wherein the agent is SB- 715992 (a kinesin antagonist).
5609. The method of item 5493 wherein the agent is temsirolimus (an mTOR inhibitor).
5610. The method of item 5493 wherein the agent is adalimumab (a TNFα antagonist). 5611. The method of item 5493, wherein the composition comprises a polymer.
5612. The method of item 5493, wherein the composition comprises a polymer, and the" polymer is, or comprises, a copolymer.
5613. The method of item 5493, wherein the composition comprises a polymer, and the polymer is, or comprises, a block copolymer.
5614. The method of item 5493, wherein the composition comprises a polymer, and the polymer is, or comprises, a random copolymer.
5615. The method of item 5493, wherein the composition comprises a polymer, and the polymer is, or comprises, a biodegradable polymer. _ .
5616. The method of item 5493, wherein the composition comprises a polymer, and the polymer is, or comprises, a nonbiodegradable polymer.
5617. The method of item 5493, wherein the composition comprises a polymer, and the polymer is, or comprises, a hydrophilic polymer.
5618. The method of item 5493, wherein the composition comprises a polymer, and the polymer is, or comprises, a hydrophobic polymer. 5619. The method of item 5493, wherein the composition comprises a polymer, and the polymer is, or comprises, a polymer having hydrophilic domains.
5620. The method of item 5493, wherein the composition comprises a polymer, and the polymer is, or comprises, a polymer having hydrophobic domains.
5621. The method of item 5493, wherein the composition comprises a polymer, and the polymer is, or comprises, a non-conductive polymer.
5622. The method of item 5493, wherein the composition comprises a polymer, and the polymer is, or comprises, an elastomer.
5623. The method of item 5493, wherein the composition comprises a polymer, and the polymer is, or comprises, a hydrogel.
5624. The method of item 5493, wherein the composition comprises a polymer, and the polymer is, or comprises, a silicone polymer.
5625. The method of item 5493, wherein the composition comprises a polymer, and the polymer is, or comprises, a hydrocarbon polymer.
5626. The method of item 5493, wherein the composition comprises a polymer, and the polymer is, or comprises, a styrene-derived polymer. T/US2006/013030
5627. The method of item 5493, wherein the composition comprises a polymer, and the polymer is, or comprises, a butadiene-derived polymer.
5628. The method of item 5493, wherein the composition comprises a polymer, and the polymer is, or comprises, a macromer.
5629. The method of item 5493, wherein the composition comprises a polymer, and the polymer is, or comprises, a poly(ethylene glycol) polymer.
5630. The method of item 5493, wherein the composition comprises a polymer, and the polymer is, or comprises, an amorphous polymer.
5631. The method of item 5493, wherein the composition further comprises a second pharmaceutically active agent.
5632. The method of item 5493, wherein the composition further comprises an anti-inflammatory agent.
5633. The method of item 5493, wherein the composition further comprises an agent that inhibits infection.
5634. The method of item 5493, wherein the composition further comprises an anthracycline.
5635. The method of item 5493, wherein the composition further comprises doxorubicin. 5636. The method of item 5493 wherein the composition further comprises mitoxantrone.
5637. The method of item 5493 wherein the composition further comprises a fluoropyrimidine.
5638. The method of item 5493, wherein the composition further comprises 5-fluorouracil (5-FU).
5639. The method of item 5493, wherein the composition further comprises a folic acid antagonist.
5640. The method of item 5493, wherein the composition further comprises methotrexate.
5641. The method of item 5493, wherein the composition further comprises a podophylotoxin.
5642. The method of item 5493, wherein the composition further comprises etoposide.
5643. The method of item 5493, wherein the composition further comprises camptothecin.
5644. The method of item 5493, wherein the composition further comprises a hydroxyurea.
5645. The method of item 5493, wherein the composition further comprises a platinum complex. 5646. The method of item 5493, wherein the composition further comprises cisplatin.
5647. The method of item 5493 wherein the composition further comprises an anti-thrombotic agent.
5648. The method of item 5493, wherein the composition further comprises a visualization agent.
5649. The method of item 5493, wherein the composition further comprises a visualization agent, and the visualization agent is a radiopaque material, wherein the radiopaque material comprises a metal, a halogenated compound, or a barium containing compound.
5650. The method of item 5493, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, barium, tantalum, or technetium.
5651. The method of item 5493, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, an MRI responsive material.
5652. The method of item 5493, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, a gadolinium chelate.
5653. The method of item 5493, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, iron, magnesium, manganese, copper, or chromium. 5654. The method of item 5493, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, iron oxide compound.
5655. The method of item 5493, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, a dye, pigment, or colorant.
5656. The method of item 5493 wherein the agent is released in effective concentrations from the composition comprising the agent by diffusion over a period ranging from the time of administration to about 90 days.
5657. The method of item 5493 wherein the agent is released in effective concentrations from the composition comprising the agent by erosion of the composition over a period ranging from the time of administration to about 90 days.
5658. The method of item 5493 wherein the composition further comprises an inflammatory cytokine.
5659. The method of item 5493 wherein the composition further comprises an agent that stimulates cell proliferation.
5660. The method of item 5493 wherein the composition further comprises a polymeric carrier.
5661. The method of item 5493 wherein the composition is in the form of a gel, paste, or spray. 5662. The method of item 5493 wherein the implant is partially constructed with the agent or the composition.
5663. The method of item 5493 wherein the implant is fully constructed with the agent or the composition.
5664. The method of item 5493 wherein the implant is impregnated with the agent or the composition.
5665. The method of item 5493, wherein the agent or the composition forms a coating, and the coating directly contacts the implant.
5666. The method of item 5493, wherein the agent or the composition forms a coating, and the coating indirectly contacts the implant.
5667. Tha method of item 5493 wherein the agent or the composition forms a coating, and the coating partially covers the implant.
5668. The method of item 5493, wherein the agent or the composition forms a coating, and the coating completely covers the implant.
5669. The method of item 5493 wherein the agent or the composition is located within pores or holes of the implant.
5670. The method of item 5493 wherein the agent or the composition is located within a channel, lumen, or divet of the implant.
5671. The method of item 5493 wherein the implant further comprising an echogenic material. 5672. The method of item 5493 wherein the implant further comprises an echogenic material, wherein the echogenic material is in the form of a coating.
5673. The method of item 5493 wherein the implant is sterile.
5674. The method of item 5493 wherein the agent is delivered from the implant, wherein the agent is released into tissue in the vicinity of the implant after deployment of the implant.
5675. The method of item 5493 wherein the agent is delivered from the implant, wherein the agent is released into tissue in the vicinity of the implant after deployment of the implant, wherein the tissue is connective tissue.
5676.- The method of item 5493 wherein the_ agent is delivered from the implant, wherein the agent is released into tissue in the vicinity of the implant after deployment of the implant, wherein the tissue is muscle tissue.
5677. The method of item 5493 wherein the agent is delivered from the implant, wherein the agent is released into tissue in the vicinity of the implant after deployment of the implant, wherein the tissue is nerve tissue.
5678. The method of item 5493 wherein the agent is delivered from the implant, wherein the agent is released into tissue in the vicinity of the implant after deployment of the implant, wherein the tissue is epithelium tissue. 5679. The method of item 5493 wherein the agent is delivered from the implant, wherein the agent is released in effective concentrations from the implant over a period ranging from the time of deployment of the implant to about 1 year.
5680. The method of item 5493 wherein the agent is delivered from the implant, wherein the agent is released in effective concentrations from the implant over a period ranging from about 1 month to 6 months.
5681. The method of item 5493 wherein the agent is delivered from the implant, wherein the agent is released in effective concentrations from the implant over a period ranging from about 1 - 90 days.
5682. The method of item 5493 wherein the agent is delivered from the implant, wherein the agent is released in effective concentrations from the implant at a constant rate.
5683. The method of item 5493 wherein the agent is delivered from the implant, wherein the agent is released in effective concentrations from the implant at an increasing rate.
5684. The method of item 5493 wherein the agent is delivered from the implant, wherein the agent is released in effective concentrations from the implant at a decreasing rate.
5685. The method of item 5493 wherein the agent is delivered from the implant, wherein the implant comprises about 0.01 Dg to about 10 Dg of the agent. 5686. The method of item 5493 wherein the agent is delivered from the implant, wherein the implant comprises about 10 Dg to about 10 mg of the agent.
5687. The method of item 5493 wherein the agent is delivered from the implant, wherein the implant comprises about 10 mg to about 250 mg of the agent.
5688. The method of item 5493 wherein the agent is delivered from the implant, wherein the implant comprises about 250 mg to about
1000 mg of the agent.
5689. The method of item 5493 wherein the agent is delivered from the implant, wherein the implant comprises about 1000 mg to about 2500 mg of the agent.
5690. The method of item 5493 wherein the agent is delivered from the implant, wherein a surface of the implant comprises less than 0.01 Dg of the agent per mm2 of implant surface to which the agent is applied.
5691. The method of item 5493 wherein the agent is delivered from the implant, wherein a surface of the implant comprises about 0.01 Dg to about 1 Dg of the agent per mm2 of implant surface to which the agent is applied.
5692. The method of item 5493 wherein the agent is delivered from the implant, wherein a surface of the implant comprises about 1 Dg to about 10 Dg of the agent per mm2 of implant surface to which the agent is applied. 5693. The method of item 5493 wherein the agent is delivered from the implant, wherein a surface of the implant comprises about 10 Dg to about 250 Dg of the agent per mm2 of implant surface to which the agent is applied.
5694. The method of item 5493 wherein the agent is delivered from the implant, wherein a surface of the implant comprises about 250 Dg to about 1000 Dg of the agent per mm2 of implant surface to which the agent is applied.
5695. The method of item 5493 wherein the agent is delivered from the implant, wherein a surface of the implant comprises about 1000 Dg to about 2500 Dg of the agent per mm2 of implant surface to which the agent is applied.
5696. The method of item 5493, wherein the implant further comprises a coating, and the coating is a uniform coating.
5697. The method of item 5493, wherein the implant further comprises a coating, and the coating is a non-uniform coating.
5698. The method of item 5493, wherein the implant further comprises a coating, and the coating is a discontinuous coating.
5699. The method of item 5493, wherein the implant further comprises a coating, and the coating is a patterned coating.
5700. The method of item 5493, wherein the implant further comprises a coating, and the coating has a thickness of 100 Dm or less. 5701. The method of item 5493, wherein the implant further comprises a coating, and the coating has a thickness of 10 Dm or less.
5702. The method of item 5493, wherein the implant further comprises a coating, and the coating adheres to the surface of the implant upon deployment of the implant.
5703. The method of item 5493, wherein the implant further comprises a coating, and the coating is stable at room temperature for a period of at least 1 year.
5704. The method of item 5493, wherein the implant further comprises a coating, and the agent is present in the coating in an amount ranging between about 0.0001% to about 1% by weight.
5705. The method of item 5493, wherein the implant further comprises a coating, and the agent is present in the coating in an amount ranging between about 1% to about 10% by weight.
5706. The method of item 5493, wherein the implant further comprises a coating, and the agent is present in the coating in an amount ranging between about 10% to about 25% by weight.
5707. The method of item 5493, wherein the implant further comprises a coating, and the agent is present in the coating in an amount ranging between about 25% to about 70% by weight.
5708. The method of item 5493, wherein the implant further comprises a coating, and the coating comprises a polymer. 5709. The method of item 5493, wherein the implant comprises a first coating having a first composition and a second coating having a second composition.
5710. The method of item 5493, wherein the implant comprises a first coating having a first composition and a second coating having a second composition, wherein the first composition and the second composition are different.
5711. A method as in any one of items 5493-5710, wherein the device is an intravascular device.
5712. A method as in any one of items 5493-5710, wherein the device is a gastrointestinal stent.
5713. A method as in any one of items 5493-5710, wherein the device is a tracheal and bronchial stent.
5714. A method as in any one of items 5493-5710, wherein the device is a genital urinary stent.
5715. A method as in any one of items 5493-5710, wherein the device is an ear and nose stent.
5716. A method as in any one of items 5493-5710, wherein the device is an ear ventilation.
5717. A method as in any one of items 5493-5710, wherein the device is an intraocular implant.
5718. A method as in any one of items 5493-5710, wherein the device is a vascular graft.
5719. A method as in any one of items 5493-5710, wherein the device comprises a film or a mesh. 5720. A method as in any one of items 5493-5710, wherein the device is a glaucoma drainage device.
5721. A method as in any one of items 5493-5710, wherein the device is a prosthetic heart valve or a component thereof.
5722. A method as in any one of items 5493-5710, wherein the device is a penile implant.
5723. A method as in any one of items 5493-5710, wherein the device is an endotracheal or tracheostomy tube.
5724. A method as in any one of items 5493-5710, wherein the device is a peritoneal dialysis catheter.
5725. A method as in any one of items 5493-5710, wherein the device is a central nervous system shunt or a pressure monitoring device.
5726. A method as in any one of items 5493-5710, wherein the device is an inferior vena cava filter.
5727. A method as in any one of items 5493-5710, wherein the device is a gastrointestinal device.
5728. A method as in any one of items 5493-5710, wherein the device is a central venous catheter.
5729. A method as in any one of items 5493-5710, wherein the device is a ventricular assist device.
5730. A method as in any one of items 5493-5710, wherein the device is a spinal implant.
5731. A method as in any one of items 5493-5710, wherein the device is an implantable electrical device. 5732. A method as in any one of items 5493-5710, wherein the device is an implantable sensor.
5733. A method as in any one of items 5493-5710, wherein the device is an implantable pump
5734. A method as in any one of items 5493-5710, wherein the device is a soft tissue implant.
5735. A method for inhibiting scarring comprising placing a device that comprises an intravascular implant and an anti-scarring agent or a composition comprising an anti-scarring agent into an animal host, wherein the agent inhibits scarring.
5736. The method of item 5735 wherein the agent is an adensosine A2A receptor antagonist.
5737. The method of item 5735 wherein the agent is an AKT inhibitor.
5738. The method of item 5735 wherein the agent is an alpha 2 integrin antagonist, wherein the alpha 2 integrin antagonist is Pharmaprojects No. 5754 (Merck KgaA).
5739. The method of item 5735 wherein the agent is an alpha 4 integrin antagonist.
5740. The method of item 5735 wherein the agent is an alpha 7 nicotinic receptor agonist.
5741. The method of item 5735 wherein the agent is an angiogenesis inhibitor selected from the group consisting of AG-12,958 (Pfizer), ATN-161 (Attention LLC), neovastat, an angiogenesis inhibitor from Jerina AG (Germany), NM-3 (Mercian), VGA-1155 (Taisho), FCE-26644 (Pfizer), FCE-26950 (Pfizer), FPMA (Meiji Daries), FR-111142 (Fujisawa), GGTI-298, GM-1306 (Ligand), GPA-1734 (Novartis), NNC-47-0011 (Novo Nordisk), herbamycin (Nippon Kayaku), lenalidomide (Celegene), IP-10 (NIH), ABT-828 (Abbott), KIN-841 (Tokushima University, Japan), SF-1126 (Semafore Pharmaceuticals), laminin technology (NIH), CHIR-258 (Chiron), NVP-AEW541 (Novartis), NVP-AEW541 (Novartis), Vt16907 (Alchemia), OXI-8007 (Oxigene), EG-3306 (Ark Therapeutics), Maspin (Arriva), ABT- 567 (Abbott), PPI-2458 (Praecis Pharmaceuticals), CC-5079, CC-4089 (Celgene), HIF-1 alpha inhibitors (Xenova), S-247 (Pfizer), AP-23573 (Ariad), AZD-9935 (Astra Zeneca), mebendazole (Introgen Therapeutics), MetAP-2 inhibitors (GlaxoSmithKline), AG-615 (Angiogene Pharmaceuticals), Tie-2 antagonists (Hybrigenics), NC-381 , CYC-381, NC-169, NC-219, NC-383, NC-384, NC-407 (Lorus Therapeutics), ATN-224 (Attenuon), ON-01370 (Onconova), Vitronectin antagonists (Amgen), SDX-103 (Salmedix), Vitronectin antagonists (Shire), CHP (Riemser), TEK (Amgen), Anecortave acetate (Alcon), T46.2 (Matrix Therapeutics), HG-2 (Heptagen), TEM antagonists (Genzyme), Oxi-4500 (Oxigene), ATN-161 (Attenuon), WX-293 (Wilex), M-2025 (Metris Therapeutics), Alphastatin (BioActa), YH-16 (Yantai Rongchang), BIBF-1120 (Boehringer Ingelheim), BAY-57-9352 (Bayer), AS- 1404 (Cancer Research Technology), SC-77964 (Pfizer), glycomimetics (BioTie Therapies), TIE-2 Inhibitors (Ontogen), DIMI, Octamer (Octamer), ABR-215050 (Active Biotech), ABT-518 (Abbott), KDR inhibitors (Abbott), BSF-466895 (Abbott), SCH-221153 (Schering-Plough), DAC:antiangiogenic (ConjuChem), TFPI (EntreMed), AZD-2171 (Astra-Zenaca), CDC-394 (Celgene), LY290293 (EIi Lilly), IDN-5390 (Indena), Kdr Kinase Inhibitors (Merck), CT-113020, CT-116433, CT-116563, CT-31890, CT-32228) (Cell Therapeutics), A-299620 (Abbott), TWEAK Inhibitor (Amgen), VEGF modulators (Johnson and Johnson), Tum-N53, tumstatin (Genzyme), Thios- 1 , Thios-2 (Thios Pharmaceuticals), MV-6401 (Miravant Medical Technologies), Spisulosine (PharmaMar), CEP-7055 (Cephalon), AUV-201 (Auvation), LM-609 (EIi Lilly), SKF-106615 (AnorMED), Oglufanide disodium (Cytran), BW-114 (Pharminox), Calreticulin (NIH), WX-678 (Wilex), SD-7784 (Pfizer), WX-UK1 (Wilex), SH-268 (Schering AG), 2-Me-PGA (Celgene), S- 137 (Pfizer), ZD-6126 (Angiogene Pharmaceuticals), SG-292 (SignalGen), Benefin (Lane Labs), A6, A36 (Angstrom), SB-2723005 (GlaxoSmithKline), SC-7 (Cell Therapeutics), ZEN-014 (AEterna Zentaris), 2-methoxyestradiol (EntreMed), NK-130119 (Nippon Kayaku), CC-10004 (Celgene), AVE- 8062A (Ajinomoto), Tacedinaline (Pfizer), Actinonin (Tokyo Metropolitan Institute of Medical Science), Lenalidomide (Celgene), VGA-1155, BTO-956 (SRI International), ER-68203-00 (Eisai), CT-6685 (UCB), JKC-362 (Phoenix Pharmaceuticals), DMI-3798 (DMI Biosciences, Angiomate (Ipsen), ZD-6474 (AstraZeneca), CEP-5214 (Cephalon), Canstatin (Genzyme), NM-3 (Mercian), Oridigm (MediQuest Therapeutics), Exherin (Adherex), BLS-0597 (Boston Life Sciences), PTC-299 (PTC Therapeutics), NPI-2358 (Nereus Pharmaceuticals), CGP-79787 (Novartis), AEE-788 (Novartis), CKD-732 (Chong Kun Dang), CP-564959 (OSI Pharmaceuticals), CM-101 (CarboMed), CT-2584, CT3501 (Cell Therapeutics), combretastatin and analogues and derivatives thereof (Oxigene), Rebimastat (Bristol-Meyers Squibb), Dextrin 2-sulfate (ML Laboratories), Cilengitide (Merk KGaA), NSC-706704 (Pharminox), KRN- 951 (Kirin Brewery), Ukrain, NSC-631570 (Nowicky Pharma), Tecogalan sodium (Daiichi Pharmaceutical), Tz-93 (Tsumura), TBC-1635 (Encysive Pharmaceuticals), TAN-1120 (Takeda), Semaxanib (Pfizer), BDI-7800 (Biopharmacopae), SD-186, SD-983 (Bristol-Meyers Squibb), SB-223245 (GlaxoSmithKline), SC-236 (Pfizer), RWJ-590973 (Johnson and Johnson), ILX-1850 (Genzyme), SC-68488, S-836 (Pfizer), CG-55069-11 (CuraGen), Ki-23057 (Kirin Brewery), CCX-700 (Chemoentryx), Pegaptanib octasodium (Giled Sciences), ANGIOCOL (available from Biostratum Inc.), or an analogue or derivative thereof. 5742. The method of item 5735 wherein the agent is an apoptosis antagonist.
5743. The method of item 5735 wherein the agent is an apoptosis activator.
5744. The method of item 5735 wherein the agent is a beta 1 integrin antagonist.
5745. The method of item 5735 wherein the agent is a beta tubulin inhibitor.
5746. The method of item 5735 wherein the agent is a blocker of enzyme production in Hepatitis C.
5747. The method of item 5735 wherein the agent is a Bruton's tyrosine kinase inhibitor.
5748. The method of item 5735 wherein the agent is a calcineurin inhibitor.
5749. The method of item 5735 wherein the agent is a caspase 3 inhibitor.
5750. The method of item 5735 wherein the agent is a CC chemokine receptor antagonist.
5751. The method of item 5735 wherein the agent is a cell cycle inhibitor selected from the group consisting of SNS-595 (Sunesis), synthadotin, KRX-0403, and an analogue or derivative thereof. 5752. The method of item 5735 wherein the agent is a cathepsin B inhibitor.
5753. The method of item 5735 wherein the agent is a cathepsin K inhibitor, wherein the cathepsin K inhibitor is 462795 (GlaxoSmithKline), INPL-022-D6 (Amura Therapeutics), or an analogue or derivative thereof.
5754. The method of item 5735 wherein the agent is a cathepsin L inhibitor.
5755. The method of item 5735 wherein the agent is a CD40 antagonist.
5756. The method of item 5735 wherein the agent is a chemokine receptor agonist.
5757. The method of item 5735 wherein the agent is a chymase inhibitor.
5758. The method of item 5735 wherein the agent is a collagenase antagonist.
5759. The method of item 5735 wherein the agent is a CXCR antagonist.
5760. The method of item 5735 wherein the agent is a cyclin dependent kinase inhibitor selected from the group consisting of a CDK-1 inhibitor, a CDK-2 inhibitor, a CDK- 4 inhibitor, a CDK-6 inhibitor, a CAK1 inhibitor from GPC Biotech and Bristol-Myers Squibb, RGB-286199 (GPC Biotech), an anticancer agent from Astex Technology, a CAK1 inhibitor from GPC Biotech, a CDK inhibitor from Sanofi-Aventis, a CDK1/CDK2 inhibitor from Amgen, a CDK2 inhibitor from SUGEN-2 (Pfizer), a hearing loss therapy agent (Sound Pharmaceuticals), PD-0332991 (Pfizer), RGB-286199 (GPC Biotech), Ro-0505124 (Hoffmann-La Roche), a Ser/Thr kinase inhibitor from Lilly (EIi Lilly), CVT-2584 (CAS No. 199986-75-9) (CV Therapeutics), and an analogue or derivative thereof.
5761. The method of item 5735 wherein the agent is a cyclooxygenase 1 inhibitor.
5762. The method of item 5735 wherein the agent is a DHFR inhibitor.
5763. The method of item 5735 wherein the agent is a dual integrin inhibitor.
5764. The method of item 5735 wherein the agent is an elastase inhibitor.
5765. The method of item 5735 wherein the agent is an elongation factor-1 alpha inhibitor.
5766. The method of item 5735 wherein the agent is an endothelial growth factor antagonist.
5767. The method of item 5735 wherein the agent is an endothelial growth factor receptor kinase inhibitor selected from the group consisting of sorafenib tosylate (Bayer), AAL-993 (Novartis), ABP-309 (Novartis), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), E- 7080 (Eisai), EG-3306 (Ark Therapeutics), EXEL-2880 (Exelixis), GW- 654652 (GlaxoSmithKline), a KDR inhibitor from LG Life Sciences, CT-6685 and CT-6729 (UCB), KRN-633 and KRN-951 (Kirin Brewery), OSI-930 (OSI Pharmaceuticals), SP-5.2 (Supratek Pharma), SU-11657 (Pfizer), a Tie-2 antagonist (Hybrigenics), a VEGFR-2 kinase inhibitor (Bristol-Myers Squibb), XL-647 (Exelixis), a KDR inhibitor from Abbott Laboratories, sorafenib tosylate, and an analogue or derivative thereof.
5768. The method of item 5735 wherein the agent is an endotoxin antagonist.
5769. The method of item 5735 wherein the agent is an epothilone and tubulin binder.
5770. The method of item 5735 wherein the agent is an estrogen receptor antagonist.
5771. The method of item 5735 wherein the agent is an FGF inhibitor.
5772. The method of item 5735 wherein the agent is a farnexyl transferase inhibitor.
5773. The method of item 5735 wherein the agent is famesyltransferase inhibitor selected from the group of A-197574 (Abbott), a famesyltransferase inhibitor from Servier, FPTIII (Strathclyde Institute for Drug R), LB-42908 (LG Life Sciences), Pharmaprojects No. 5063 (Genzyme), Pharmaprojects No. 5597 (Ipsen), Yissum Project No. B-1055 (Yissum), and an analogue or derivative thereof.
5774. The method of item 5735 wherein the agent is an FLT-3 kinase inhibitor. 5775. The method of item 5735 wherein the agent is an FGF receptor kinase inhibitor.
5776. The method of item 5735 wherein the agent is a fibrinogen antagonist selected from the group consisting of AUV-201 (Auvation), MG-13926 (Sanofi-Aventis), plasminogen activator (CAS No. 105913-11-9) (from Sanofi-Aventis or UCB), plasminogen activator-2 (tPA- 2) (Sanofi-Aventis), pro-urokinase (CAS No. 82657-92-9) (Sanofi-Aventis), and an analogue or derivative thereof.
5777. The method of item 5735 wherein the agent is a heat shock protein 90 antagonist selected from the group consisting of SRN-005 (Sirenade), geldanamycin, NSC-33050 (17-allylaminogeldanamycin; 17- AAG), 17-dimethylaminoethylamino-17-demethoxy-geldanamycin (17- DMAG), rifabutin (rifamycin XIV, 1',4-didehydro-1-deoxy-1 ,4-dihydro-5'-(2- methylpropyl)-1-oxo-), and an analogue or derivative thereof.
5778. The method of item 5735 wherein the agent is a histone deacetylase inhibitor.
5779. The method of item 5735 wherein the agent is an HMGCoA reductase inhibitor selected from the group consisting of an atherosclerosis therapeutic from Lipid Sciences, ATI-16000 (ARYx Therapeutics), KS-01-019 (Kos Pharmaceuticals), Pharmaprojects No. 2197 (Sanofi-Aventi), RP 61969 (Sanofi-Aventis), and an analogue or derivative thereof.
5780. The method of item 5735 wherein the agent is an ICAM inhibitor. 5781. The method of item 5735 wherein the agent is an IL, ICE and IRAK antagonist, wherein the antagonist is a CJ-14877, CP-424174 (Pfizer), NF-61 (Negma-Lerads), and an analogue or derivative thereof.
5782. The method of item 5735 wherein the agent is an IL-2 inhibitor.
5783. The method of item 5735 wherein the agent is an immunosuppressant selected from the group consisting of teriflunomide (Sanofi Aventis), chlorsulfaquinoxalone (NSC-339004), chlorsulfaquinoxalone sulfate, CS-712 (Sankyo), ismomultin alfa (CAS No. 457913-93-8) (Akzo Nobel), antiallergics from GenPat77, antiinflammatories or AT-005 (Androclus Therapeutics), autoimmune disease therapeutics from EpiVax, BN-007 (Bone), budesonide (CAS No. 51333-22- 3) (MAP Pharmaceuticals), CO-14 (Genzyme), edratide (CAS No. 433922- 67-9) (Teva), EP-314 (Enanta), eprovafen (CAS No. 101335-99-3) (Sanofi- Aventis), HWA-131 (CAS No. 118788-41-3) (Sanofi-Aventis), immunomodulators from MerLion Pharmaceuticals, immunosuppressives from Alchemia, IPL-12 (Inflazyme), MDL-9563 (CAS No. 27086-86-8) (Sanofi-Aventis), Pharmaprojects No. 2330 (Sanofi-Aventis), Pharmaprojects No. 6426 (Abgenix), PXS-25 (Pharmaxis), rosmarinic acid (CAS No. 20283-92-5) (Sanofi-Aventis), RP 42927 or RP 54745 (CAS No. 135330-08-4) (Sanofi-Aventis), SGN-35 (Seattle Genetics), ST-1959 (Sigma-Tau), type I diabetes therapy from SYNX Pharma, UNIL-88 (Debiopharm), VP-025 (Vasogen), VR-694 (Vectura), PRTX-001 (Protalex), and an analogue or derivative thereof.
5784. The method of item 5735 wherein the agent is an IMPDH (inosine monophosphate). 5785. The method of item 5735 wherein the agent is an integrin antagonist.
5786. The method of item 5735 wherein the agent is an interleukin antagonist.
5787. The method of item 5735 wherein the agent is an inhibitor of type III receptor tyrosine kinase.
5788. The method of item 5735 wherein the agent is an irreversible inhibitor of enzyme methionine aminopeptidase type 2.
5789. The method of item 5735 wherein the agent is an isozyme selective delta protein kinase C inhibitor.
5790. The method of item 5735 wherein the agent a JAK3 enzyme inhibitor.
5791. The method of item 5735 wherein the agent is a JNK inhibitor.
5792. The method of item 5735 wherein the agent is a kinase inhibitor.
5793. The method of item 5735 wherein the agent is kinesin antagonist.
5794. The method of item 5735 wherein the agent is a kinesin antagonist. 5795. The method of item 5735 wherein the agent is a leukotriene inhibitor and antagonist selected from the group consisting of ambicromil (CAS No. 58805-38-2) (Sanofi-Aventis), amelubant (CAS No. 346735-24-8) (Boehringer Ingelheim), DW-1141 (Dong Wha), ebselen (Daiichi Pharmaceutical), ibudilast (Kyorin), leucotriene inhibitors from Sanofi-Aventis, lymphotoxin-beta receptor (LT-D) from Biogen Idee, Pharmaprojects No. 1535 and 2728 (CAS No. 119340-33-9) (Sanofi- Aventis), R-112 (Rigel), Rev-5367 (CAS No. 92532-05-3) (Sanofi-Aventis), RG-14893 (CAS No. 141835-49-6) (Sanofi-Aventis), RG-5901-A (CAS No. 101910-24-1), 92532-23-5, RP 66153 (CAS No. 142422-79-5), RP 66364 (CAS No.186916-92-5), RP 69698 (CAS No. 141748-00-7) (Sanofi-Aventis), SC-411930 (Pfizer), SC-41930 (CAS No. 120072-59-5) (Pfizer), SC-50605 (CAS No. 138828-39-4) (Pfizer), SC-51146 (CAS No. 141059-52-1), SC- 53228 (CAS No. 153633-01-3) (Pfizer), spaglumic acid (ZY-15106) (CAS No.3106-85-2), 80619-64-3 (Novartis), tipredane (CAS No. 85197-77-9) (Bristol-Myers Squibb), U-75302 (CAS No. 119477-85-9) (Pfizer), and analogue or derivative thereof.
5796. The method of item 5735 wherein the agent is an MAP kinase inhibitor.
5797. The method of item 5735 wherein the agent is a matrix metalloproteinase inhibitor.
5798. The method of item 5735 wherein the agent is an MCP- CCR2 inhibitor.
5799. The method of item 5735 wherein the agent is an mTOR inhibitor. 5800. The method of item 5735 wherein the agent is an mTOR kinase inhibitor.
5801. The method of item 5735 wherein the agent is a microtubule inhibitor selected from the group consisting of antibody- maytansinoid conjugates from Biogen Idee, colchicines (MantiCore Pharmaceuticals), anticancer immunoconjugates from Johnson & Johnson, DIME from Octamer, gni-1f (GNI), huC242-DM4, huMy9-6-DM1 (ImmunoGen), IDN-5404 (lndena), IMO-098, IMOderm (Imotep), mebendazole (Introgen Therapeutics), microtubule poisons from Cambridge Enterprise, paclitaxel such as LOTAX from Aphios (CAS No. 33069-62-4), Genexol-PM from Samyang, Pharmaprojects No. 6383 (Tapestry Pharmaceuticals), RPR-112378 (Sanofi-Aventis), SGN-75 (Seattle Genetics), SPL-7435 (Starpharma), SSR-250411 (Sanofi-Aventis), trastuzumab-DM1 (Genentech), vinorelbine, and an analogue or derivative thereof.
5802. The method of item 5735 wherein the agent is an MIF inhibitor.
5803. The method of item 5735 wherein the agent is an MMP inhibitor.
5804. The method of item 5735 wherein the agent is a neurokinin (NK) antagonist selected from the group consisting of anthrotainin (CAS No. 148084-40-6) (Sanofi-Aventis), an IBS thereapeutic from ArQuIe, MDL-105212A (CAS No. 167261-60-1) (Ssanofi-Aventis), Pharmaprojects No. 2744, 3258 (CAS No. 139167-47-8) 4006, 4201, or 5986 (Sanofi-Aventis), RP 67580 (CAS No. 135911-02-3), SR-144190 (CAS No. 201152-86-5), SSR-240600, SSR-241586 (Sanofi-Aventis), TKA-457 (Novartis), vestipitant mesylate (CAS No. 334476-64-1) (GlaxoSmithKline), Win-64821 (Sanofi-Aventis), PRX-96026 (Predix Pharmaceuticals), and an analogue or derivative thereof.
5805. The method of item 5735 wherein the agent is an NF kappa B inhibitor selected from the group consisting of AVE-0545 or AVE- 0547 (Sanofi-Aventis), bortezomib (CAS No. 179324-69-7) (Millennium Pharmaceuticals), dexanabinol (CAS No. 112924-45-5) (Pharmos), dexlipotam (Viatris), Pharmaprojects No. 6283 (INDRA) (OXiGENE), IPL- 576092 (CAS No. 137571-30-3) (Inflazyme), NFKB decoy (Corgentech), NFKB decoy oligo (AnGes MG), NFKB's from Ariad, osteoporosis treatments or S5 (F005) from Fulcrum Pharmaceuticals, P61 (Phytopharm), R-flurbiprofen (CAS No. 5104-49-4) (Encore Pharmaceuticals), and an analogue or derivative thereof.
5806. The method of item 5735 wherein the agent is a nitric oxide agonist.
5807. The method of item 5735 wherein the agent is an ornithine decarboxylase inhibitor.
5808. The method of item 5735 wherein the agent is a p38 MAP kinase inhibitor selected from the group consisting of AZD-6703 (AstraZeneca), JX-401 (Jexys Pharmaceuticals), BMS-2 (Bristol-Myers Squibb), a p38 MAP kinase inhibitor from Novartis, a p38-alpha MAP kinase inhibitor from Amphora, Pharmaprojects No. 5704 (Pharmacopeia), RPR- 200765A (Sanofi-Aventis), SD-282 (Johnson & Johnson), TAK-715 (Takeda), and an analogue or derivative thereof.
5809. The method of item 5735 wherein the agent is a palmitoyl-protein thioesterase inhibitor. 5810. The method of item 5735 wherein the agent is a PDGF receptor kinase inhibitor selected from the group consisting of AAL-993, AMN-107, or ABP-309 (Novartis), AMG-706 (Amgen), BAY-57-9352 (Bayer), CDP-860 (UCB)1 E-7080 (Eisai), imatinib (CAS No. 152459-95-5) (Novartis), OSI-930 (OSI Pharmaceuticals), RPR-127963E (Sanofi-Aventis), RWJ-540973 (Johnson & Johnson), sorafenib tosylate (Bayer), SU-11657 (Pfizer), tandutinib (CAS No. 387867-13-2) (Millennium Pharmaceuticals), vatalanib (Novartis), ZK-CDK (Schering AG), and an analogue or derivative thereof.
5811. The method of item 5735 wherein the agent is (-)- halofenate (Metabolex), AMG-131 (Amgen), antidiabetics from Japan Tobacco, AZD-4619, AZD-8450, AZD-8677 (AstraZeneca), DRF-10945, balaglitazone (Dr Reddy's), CS-00088, CS-00098 (Chipscreen Biosciences), E-3030 (Eisai), etalocib (CAS No. 161172-51-6) (EIi Lilly), GSK-641597 (Ligand), GSK-677954 (GlaxoSmithKline), GW-409544 (Ligand), GW- 590735 (GlaxoSmithKline), K-111 (Hoffmann-La Roche), LY-518674 (EIi Lilly), LY-674 (Ligand), LY-929 (Ligand), MC-3001 , MC-3002 (MaxoCore Pharmaceuticals), metformin HCI + pioglitazone (CAS No. 1115-70-4 and 112529-15-4), ACTOPLUS MET from Andrx), muraglitazar (CAS No. 331741-94-7) (Bristol-Myers Squibb), naveglitazar (Ligand), oleoylethanolamide (Kadmus Pharmaceuticals), ONO-5129, pioglitazone hydrochloride (CAS No. 111025-46-8 and 112529-15-4) (Takeda), PLX-204 (Plexxikon), PPAR agonists from Genfit, PPAR delta agonists from EIi Lilly, PPAR-alpha agonists from CrystalGenomics, PPAR-gamma modulators and PPAR-β modulators from CareX, rosiglitazone maleate (CAS No. 122324- 73-4 or 155141-29-0) (GlaxoSmithKline), rosiglitazone maleate/glimepir (CAS No. 155141-29-0 and 93479-97-1), AVANDARYL, rosiglitazone maleate/metformin extend (CAS No. 155141-29-0 and 657-24-9), AVANDAMET, rosiglitazone maleate+metformin, AVANDAMET (GlaxoSmithKline), tesaglitazar (AstraZeneca), and an analogue or derivative thereof.
5812. The method of item 5735 wherein the agent is a phosphatase inhibitor.
5813. The method of item 5735 wherein the agent is a phosphodiesterase (PDE) inhibitor selected from the group consisting of avanafil (Tanabe Seiyaku), dasantafil (CAS No. 569351-91-3) (Schering- Plough), A-906119 (CAS No. 134072-58-5), DL-850 (Sanofi-Aventis), GRC- 3015, GRC-3566, GRC-3886 (Glenmark), HWA-153 (CAS No. 56395-66-5) (Sanofi-Aventis), hydroxypumafentrine (Altana), IBFB-130011 , IBFB-14-016, IBFB-140301 , IBFB-150007, IBFB-211913 (IBFB Pharma), L-826141 (Merck & Co), medorinone (CAS No. 88296-61-1) (Sanofi-Aventis), MEM- 1917 (Memory Pharmaceuticals), ND-1251 (Neuro3d), PDE inhibitors from ICOS, PDE IV inhibitors, from Memory Pharmaceuticals and CrystalGenomics, Pharmaprojects No. 2742 and 6141 (Sanofi-Aventis), QAD-171 (Novartis), RHC-2963 (CAS No. 76993-12-9 and 76993-14-1), RPR-117658, RPR-122818 derivatives, SR-24870 , and RPR-132294 (Sanofi-Aventis), SK-350 (ln2Gen), stroke therapy agents from deCODE Genetics, TAS-203 (Taiho), tofimilast (CAS No. 185954-27-2) (Pfizer), UK- 371800 (Pfizer), WIN-65579 (CAS No. 158020-82-7) (Sanofi-Aventis), IBFB- 130020 (IBFB Pharma), OPC-6535 (CAS No. 145739-56-6) (Otsuka), a phosphodiesterase III inhibitor, enoximone, a phosphodiesterase IV inhibitor, fosfosal, Atopik (Barrier Therapeutics), triflusal, a phosphodiesterase V inhibitor, and an analogue or derivative thereof.
5814. The method of item 5735 wherein the agent is a PKC inhibitor. 5815. The method of item 5735 wherein the agent is a platelet activating factor antagonist.
5816. The method of item 5735 wherein the agent is a platelet-derived growth factor receptor kinase inhibitor.
5817. The method of item 5735 wherein the agent is a prolyl hydroxylase inhibitor.
5818. The method of item 5735 wherein the agent is a polymorphonuclear neutrophil inhibitor.
5819. The method of item 5735 wherein the agent is a protein kinase B inhibitor.
5820. The method of item 5735 wherein the agent is a protein kinase C stimulant.
5821. The method of item 5735 wherein the agent is a purine nucleoside analogue.
5822. The method of item 5735 wherein the agent is a purinoreceptor P2X antagonist.
5823. The method of item 5735 wherein the agent is a Raf kinase inhibitor.
5824. The method of item 5735 wherein the agent is a reversible inhibitor of ErbB1 and ErbB2. 5825. The method of item 5735 wherein the agent is a ribonucleoside triphosphate reductase inhibitor.
5826. The method of item 5735 wherein the agent is an SDF- 1 antagonist.
5827. The method of item 5735 wherein the agent is a sheddase inhibitor.
5828. The method of item 5735 wherein the agent is an SRC inhibitor.
5829. The method of item 5735 wherein the agent is a stromelysin inhibitor.
— . 5830. The method. of item.5735 whejeinjhe agent is an Syk kinase inhibitor.
5831. The method of item 5735 wherein the agent is a telomerase inhibitor.
5832. The method of item 5735 wherein the agent is a TGF beta inhibitor selected from the group consisting of pirfenidone (CAS No. 53179-13-8) (MARNAC), tranilast (CAS No. 53902-12-8) (Kissei), IN-1130 (ln2Gen), mannose-6-phosphate (BTG), TGF-β antagonists from Inflazyme (Pharmaprojects No. 6075), TGF-β antagonists from Sydney, non-industrial source), TGF-β I receptor kinase inhibitors from EIi Lilly, TGF-β receptor inhibitors from Johnson & Johnson, and an analogue or derivative thereof.
5833. The method of item 5735 wherein the agent is a TN Fa antagonist or TACE inhibitor selected from the group consisting of adalimumab (CAS No. 331731-18-1) (Cambridge Antibody Technology), AGIX-4207 (AtheroGenics), AGT-1 (Advanced Biotherapy), an antiinflammatory from Borean Pharma, Cellzome, or Paradigm Therapeutics, anti-inflammatory vaccine (TNF-alpha kinoid) from Neovacs, humanized anti-TNF antibody or an anti-TNF MAb (CB0006) Celltech (UCB), apratastat (CAS No. 287445-51-0) (Wyeth), BMS-561392 (Bristol-Myers Squibb), BN- 006 (Bone), certolizumab pegol (CAS No. 428863-50-7 or CH-138 (UCB), cilomilast (CAS No. 153259-65-5) (GlaxoSmithKline), CR-1 (Nuada Pharmaceuticals), CRx-119 (CombinatoRx), D-5410 (UCB), dacopafant (CAS No. 125372-33-0) (Sanofi-Aventis), dersalazine (CAS No. 188913-57- 7/188913-58-8) (Uriach), etanercept (CAS No. 185243-69-0) (Amgen), ethyl pyruvate (Critical (Critical Therapeutics), golimumab (CAS No. 476181-74-5) (Johnson & Johnson), hormono-immunotherapy from Ipsen, CDP571 (e.g., Humicade from UCB), IC-485 (ICOS), infliximab (CAS No. 170277-31-3) (Johnson & Johnson), IP-751 (Manhattan Pharmaceuticals), ISIS-104838 (CAS NIo..250755-32-9) (LSJS Pharmaceuticals), lenalidqmjde (CAS Np. 191732-72-6) (Celgene), lentinan (CAS No. 37339-90-5) (Ajinomoto), MDL- 201112 (CAS No. 142130-73-2) (Sanofi-Aventis), medroxyprogesterone (CAS No. 520-85-4) (InKine Pharmaceutical), N-acetylcysteine (CAS No. 616-91-1) (Zambon), NBE-P2 (DIREVO Biotech), nerelimomab (CAS No. 162774-06-3) (Chiron), OM-294DP (OM PHARMA), onercept (CAS No. 199685-57-9) (Yeda), PASSTNF-alpha (Verigen), pentoxifylline or oxypentifylline (Sanofi-Aventis), Pharmaprojects No. 4091 , 4241, 4295, or 4488 (Sanofi-Aventis), Pharmaprojects No. 5480 (Amgen), Pharmaprojects No. 6749 (Cengent), pirfenidone (CAS No. 53179-13-8) (MARNAC), RPR- 132294 (Sanofi-Aventis), S5 (F002) (Fulcrum Pharmaceuticals), simvastatin (CAS No. 79902-63-9) (Merck & Co), STA-6292 (Synta Pharmaceuticals), tacrolimus (CAS No. 104987-11-3) (Fujisawa LifeCycle Pharma), talactoferrin alfa (CAS No. 308240-58-6) (Agennix), thalidomide (CAS No. 50-35-1) (Celgene), TNF antagonists form ProStrakan, and Synergen, TNF inhibitors (Amgen), TNF-alpha antagonists from Dynavax Technologies and Jerina AG (Germany), TNF-alpha inhibitors from IBFB Pharma and Xencor (Xencor), torbafylline (CAS No. 105102-21-4) (Sanofi-Aventis), UR-1505 (Uriach), VT-346 (Viron Therapeutics), YSIL6 (Y's Therapeutics), YSTH2 (Y's Therapeutics), NPI-1302a-3 (Nereus Pharmaceuticals, a TNF antagonist from Jerina AG (Germany), dersalazine, and an analogue or derivative thereof.
5834. The method of item 5735 wherein the agent is a Toll receptor inhibitor.
5835. The method of item 5735 wherein the agent is a tubulin antagonist.
5836. The method of item 5735 wherein the agent is a tyrosine kinase inhibitor selected from the group consisting of SU-011248, SUTENT from Pfizer Inc. (New York, NY), BMS-354825, PN-355 (Paracelsian Pharmaceuticals), AGN-199659 (Allergan), AAL-993 or ABP- 309 (Novartis), adaphostin (NIH), AEE-788 (Novartis), AG-013736 (OSI Pharmaceuticals), AG-13736 (Pfizer), ALT-110 (Alteris Therapeutics), AMG- 706 (Amgen), anticancer MAbs from Xencor, anti-EGFrvlll MAbs from Abgenix, anti-HER2 MAb from Abiogen, AZD-2171 or AZD-9935 (AstraZeneca), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), CEP-5214 (Cephalon), CEP-7055 (Cephalon), cetuximab (ImClone Systems), CHIR-200131 and CHIR-258 (Chiron), CP-547632 (OSI Pharmaceuticals), CP-724714 (Pfizer), CT-301 (Creabilis Therapeutics), D- 69491 (Baxter International), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EGFR/ErbB2 inhibitors from Array BioPharma, erlotinib (CAS No. 183319- 69-9) (OSI Pharmaceuticals), EXEL-2880 (Exelixis), FK-778 (Sanofi- Aventis) , gefitinib (CAS No. 184475-35-2) (AstraZeneca), GW-2286 or GW- 654652 (GlaxoSmithKline), her2/neu antigen from AlphaVax, HER-2/neu inhibitor from Generex, Herzyme (Medipad) (Slrna Therapeutics), HKI-272 (Wyeth), HuMax-EGFr (Genmab), idronoxil (CAS No. 81267-65-4) (Novogen), IGF-1 inhibitors from Ontogen, IMC-11 F8 (ImCIone Systems), kahalalide F (CAS No. 149204-42-2) (PharmaMar), KDR inhibitor from LG Life Sciences, KDR inhibitors from Abbott Laboratories, KDR kinase inhibitors (UCB), Kdr kinase inhibitors from Merck & Co, KRN-633 and KRN- 951 (Kirin Brewery), KSB-102 (Xenova), lapatinib ditosylate (CAS No. 388082-78-8) (GlaxoSmithKline), matuzumab (Merck KGaA), MDX-214 (Medarex), ME-103 (Pharmexa), MED-A300 (Gerolymatos), MNAC-13 (Lay Line Genomics), nimotuzumab (Center of Molecular Immunology), NSC- 330507 or NSC-707545 (NIH), NV-50 (Novogen), OSI-930 (OSI Pharmaceuticals), panitumumab (Abgenix), pelitinib (CAS No. 287933-82-7) (Wyeth), pertuzumab (CAS No. 380610-27-5) (Genentech), Pharmaprojects No. 3985 (Sanofi-Aventis), prostate cancer therapeutics from Sequenom (SQPC35, SQPC36, SQPC90), removab and remoxab (Trion Pharma), RG- 13022 (CAS No. 136831-48-6), RG-13291 (CAS No. 138989-50-1), or RG- 14620 (CAS No. 136831-49-7) (Sanofi-Aventis), RM-6427 (Remark), RNAi breast cancer therapy from Benitec, RP 53801 (CAS No. 125882-88-4) (Sanofi-Aventis), sorafenib tosylate (Bayer), SU-11657 (Pfizer), Tie-2 antagonists from Semaia (Hybrigenics), Tie-2 inhibitors from Ontogen, trastuzumab (CAS No. 180288-69-1) (Genentech), tyrosine kinase inhibitors from Sanofi-Aventis, U3-1287, U3-1565, U3-1784, U3-1800 (U3 Pharma), vatalanib (Novartis), VEGFR-2 kinase inhibitor from Bristol-Myers Squibb, XL-647 (Exelixis), ZD-6474 (AstraZeneca), ZK-CDK (Schering AG), an EGFR tyrosine kinase inhibitor, EKB-569 (Wyeth), and an analogue or derivative thereof.
5837. The method of item 5735 wherein the agent is a VEGF inhibitor.
5838. The method of item 5735 wherein the agent is a vitamin D receptor agonist. 5839. The method of item 5735 wherein the agent is ZD-6474 (an angiogenesis inhibitor).
5840. The method of item 5735 wherein the agent is AP- 23573 (an mTOR inhibitor).
5841. The method of item 5735 wherein the agent is synthadotin (a tubulin antagonist).
5842. The method of item 5735 wherein the agent is S-0885 (a collagenase inhibitor).
5843. The method of item 5735 wherein the agent is aplidine (an elongation factor-1 alpha inhibitor).
5844. The method of item 5735 wherein the agent is ixabepilone (an epithilone).
5845. The method of item 5735 wherein the agent is IDN- 5390 (an angiogenesis inhibitor).
5846. The method of item 5735 wherein the agent is SB- 2723005 (an angiogenesis inhibitor).
5847. The method of item 5735 wherein the agent is ABT-518 (an angiogenesis inhibitor).
5848. The method of item 5735 wherein the agent is combretastatin (an angiogenesis inhibitor). 5849. The method of item 5735 wherein the agent is anecortave acetate (an angiogenesis inhibitor).
5850. The method of item 5735 wherein the agent is SB- 715992 (a kinesin antagonist).
5851. The method of item 5735 wherein the agent is temsirolimus (an mTOR inhibitor).
5852. The method of item 5735 wherein the agent is adalimumab (a TNFα antagonist).
5853. The method of item 5735, wherein the composition comprises a polymer.
5854. The method of item 5735, wherein the composition comprises a polymer, and the polymer is, or comprises, a copolymer.
5855. The method of item 5735, wherein the composition comprises a polymer, and the polymer is, or comprises, a block copolymer.
5856. The method of item 5735, wherein the composition comprises a polymer, and the polymer is, or comprises, a random copolymer.
5857. The method of item 5735, wherein the composition comprises a polymer, and the polymer is, or comprises, a biodegradable polymer. 5858. The method of item 5735, wherein the composition comprises a polymer, and the polymer is, or comprises, a nonbiodegradable polymer.
5859. The method of item 5735, wherein the composition comprises a polymer, and the polymer is, or comprises, a hydrophilic polymer.
5860. The method of item 5735, wherein the composition comprises a polymer, and the polymer is, or comprises, a hydrophobic polymer.
5861. The method of item 5735, wherein the composition comprises a polymer, and the polymer is, or comprises, a polymer having hydrophilic domains.
5862. The method of item 5735, wherein the composition comprises a polymer, and the polymer is, or comprises, a polymer having hydrophobic domains.
5863. The method of item 5735, wherein the composition comprises a polymer, and the polymer is, or comprises, a non-conductive polymer.
5864. The method of item 5735, wherein the composition comprises a polymer, and the polymer is, or comprises, an elastomer.
5865. The method of item 5735, wherein the composition comprises a polymer, and the polymer is, or comprises, a hydrogel. 5866. The method of item 5735, wherein the composition comprises a polymer, and the polymer is, or comprises, a silicone polymer.
5867. The method of item 5735, wherein the composition comprises a polymer, and the polymer is, or comprises, a hydrocarbon polymer.
5868. The method of item 5735, wherein the composition comprises a polymer, and the polymer is, or comprises, a styrene-derived polymer.
5869. The method of item 5735, wherein the composition comprises a polymer, and the polymer is, or comprises, a butadiene-derived polymer.
5870. The method of item 5735, wherein the composition comprises a polymer, and the polymer is, or comprises, a macromer.
5871. The method of item 5735, wherein the composition comprises a polymer, and the polymer is, or comprises, a poly(ethylene glycol) polymer.
5872. The method of item 5735, wherein the composition comprises a polymer, and the polymer is, or comprises, an amorphous polymer.
5873. The method of item 5735, wherein the composition further comprises a second pharmaceutically active agent.
5874. The method of item 5735, wherein the composition further comprises an anti-inflammatory agent. 5875. The method of item 5735, wherein the composition further comprises an agent that inhibits infection.
5876. The method of item 5735, wherein the composition further comprises an anthracycline.
5877. The method of item 5735, wherein the composition further comprises doxorubicin.
5878. The method of item 5735 wherein the composition further comprises mitoxantrone.
5879. The method of item 5735 wherein the composition further comprises a fluoropyrimidine.
5880. The method of item 5735, wherein the composition further comprises 5-fluorouracil (5-FU).
5881. The method of item 5735, wherein the composition further comprises a folic acid antagonist.
5882. The method of item 5735, wherein the composition further comprises methotrexate.
5883. The method of item 5735, wherein the composition further comprises a podophylotoxin.
5884. The method of item 5735, wherein the composition further comprises etoposide. 5885. The method of item 5735, wherein the composition further comprises camptothecin.
5886. The method of item 5735, wherein the composition further comprises a hydroxyurea.
5887. The method of item 5735, wherein the composition further comprises a platinum complex.
5888. The method of item 5735, wherein the composition further comprises cisplatin.
5889. The method of item 5735 wherein the composition further comprises an anti-thrombotic agent.
5890. The method of item 5735, wherein the composition further comprises a visualization agent.
5891. The method of item 5735, wherein the composition further comprises a visualization agent, and the visualization agent is a radiopaque material, wherein the radiopaque material comprises a metal, a halogenated compound, or a barium containing compound.
5892. The method of item 5735, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, barium, tantalum, or technetium.
5893. The method of item 5735, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, an MRI responsive material. 5894. The method of item 5735, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, a gadolinium chelate.
5895. The method of item 5735, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, iron, magnesium, manganese, copper, or chromium.
5896. The method of item 5735, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, iron oxide compound.
5897. The method of item 5735, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, a dye, pigment, or colorant.
5898. The method of item 5735 wherein the agent is released in effective concentrations from the composition comprising the agent by diffusion over a period ranging from the time of administration to about 90 days.
5899. The method of item 5735 wherein the agent is released in effective concentrations from the composition comprising the agent by erosion of the composition over a period ranging from the time of administration to about 90 days.
5900. The method of item 5735 wherein the composition further comprises an inflammatory cytokine.
5901. The method of item 5735 wherein the composition further comprises an agent that stimulates cell proliferation. 5902. The method of item 5735 wherein the composition further comprises a polymeric carrier.
5903. The method of item 5735 wherein the composition is in the form of a gel, paste, or spray.
5904. The method of item 5735 wherein the implant is partially constructed with the agent or the composition.
5905. The method of item 5735 wherein the implant is fully constructed with the agent or the composition.
5906. The method of item 5735 wherein the implant is impregnated with the agent or the composition.
. . . „ . 5907. The method of item 5735, wherein the agent or the composition forms a coating, and the coating directly contacts the implant.
5908. The method of item 5735, wherein the agent or the composition forms a coating, and the coating indirectly contacts the implant.
5909. The method of item 5735 wherein the agent or the composition forms a coating, and the coating partially covers the implant.
5910. The method of item 5735, wherein the agent or the composition forms a coating, and the coating completely covers the implant.
5911. The method of item 5735 wherein the agent or the composition is located within pores or holes of the implant. 5912. The method of item 5735 wherein the agent or the composition is located within a channel, lumen, or divet of the implant.
5913. The method of item 5735 wherein the implant further comprising an echogenic material.
5914. The method of item 5735 wherein the implant further comprises an echogenic material, wherein the echogenic material is in the form of a coating.
5915. The method of item 5735 wherein the implant is sterile.
5916. The method of item 5735 wherein the agent is delivered from the implant, wherein the agent is released into tissue in the vicinity of the implant after deployment of the implant.
5917. The method of item 5735 wherein the agent is delivered from the implant, wherein the agent is released into tissue in the vicinity of the implant after deployment of the implant, wherein the tissue is connective tissue.
5918. The method of item 5735 wherein the agent is delivered from the implant, wherein the agent is released into tissue in the vicinity of the implant after deployment of the implant, wherein the tissue is muscle tissue.
5919. The method of item 5735 wherein the agent is delivered from the implant, wherein the agent is released into tissue in the vicinity of the implant after deployment of the implant, wherein the tissue is nerve tissue. 5920. The method of item 5735 wherein the agent is delivered from the implant, wherein the agent is released into tissue in the vicinity of the implant after deployment of the implant, wherein the tissue is epithelium tissue.
5921. The method of item 5735 wherein the agent is delivered from the implant, wherein the agent is released in effective concentrations from the implant over a period ranging from the time of deployment of the implant to about 1 year.
5922. The method of item 5735 wherein the agent is delivered from the implant, wherein the agent is released in effective concentrations from the implant over a period ranging from about 1 month to 6 months.
5923. The method of item 5735 wherein the agent is delivered from the implant, wherein the agent is released in effective concentrations from the implant over a period ranging from about 1 - 90 days.
5924. The method of item 5735 wherein the agent is delivered from the implant, wherein the agent is released in effective concentrations from the implant at a constant rate.
5925. The method of item 5735 wherein the agent is delivered from the implant, wherein the agent is released in effective concentrations from the implant at an increasing rate.
5926. The method of item 5735 wherein the agent is delivered from the implant, wherein the agent is released in effective concentrations from the implant at a decreasing rate. 5927. The method of item 5735 wherein the agent is delivered from the implant, wherein the implant comprises about 0.01 Dg to about 10 Dg of the agent.
5928. The method of item 5735 wherein the agent is delivered from the implant, wherein the implant comprises about 10 Dg to about 10 mg of the agent.
5929. The method of item 5735 wherein the agent is delivered from the implant, wherein the implant comprises about 10 mg to about 250 mg of the agent.
5930. The method of item 5735 wherein the agent is delivered from the implant, wherein the implant comprises about 250 mg to about 1000 mg of the agent.
5931. The method of item 5735 wherein the agent is delivered from the implant, wherein the implant comprises about 1000 mg to about 2500 mg of the agent.
5932. The method of item 5735 wherein the agent is delivered from the implant, wherein a surface of the implant comprises less than 0.01 Dg of the agent per mm2 of implant surface to which the agent is applied.
5933. The method of item 5735 wherein the agent is delivered from the implant, wherein a surface of the implant comprises about 0.01 Dg to about 1 Dg of the agent per mm2 of implant surface to which the agent is applied.
5934. The method of item 5735 wherein the agent is delivered from the implant, wherein a surface of the implant comprises about 1 Dg to about 10 Dg of the agent per mm2 of implant surface to which the agent is applied.
5935. The method of item 5735 wherein the agent is delivered from the implant, wherein a surface of the implant comprises about 10 Dg to about 250 Dg of the agent per mm2 of implant surface to which the agent is applied.
5936. The method of item 5735 wherein the agent is delivered from the implant, wherein a surface of the implant comprises about 250 Dg to about 1000 Dg of the agent per mm2 of implant surface to which the agent is applied.
5937. The method of item 5735 wherein the agent is delivered from the implant, wherein a surface of the implant comprises about 1000 Dg to about 2500 Dg of the agent per mm2 of implant surface to which the agent is applied.
5938. The method of item 5735, wherein the implant further comprises a coating, and the coating is a uniform coating.
5939. The method of item 5735, wherein the implant further comprises a coating, and the coating is a non-uniform coating.
5940. The method of item 5735, wherein the implant further comprises a coating, and the coating is a discontinuous coating.
5941. The method of item 5735, wherein the implant further comprises a coating, and the coating is a patterned coating. 5942. The method of item 5735, wherein the implant further comprises a coating, and the coating has a thickness of 100 Dm or less.
5943. The method of item 5735, wherein the implant further comprises a coating, and the coating has a thickness of 10 Dm or less.
5944. The method of item 5735, wherein the implant further comprises a coating, and the coating adheres to the surface of the implant upon deployment of the implant.
5945. The method of item 5735, wherein the implant further comprises a coating, and the coating is stable at room temperature for a period of at least 1 year.
5946. The method of item 5735, wherein the implant further comprises a coating, and the agent is present in the coating in an amount ranging between about 0.0001% to about 1% by weight.
5947. The method of item 5735, wherein the implant further comprises a coating, and the agent is present in the coating in an amount ranging between about 1% to about 10% by weight.
5948. The method of item 5735, wherein the implant further comprises a coating, and the agent is present in the coating in an amount ranging between about 10% to about 25% by weight.
5949. The method of item 5735, wherein the implant further comprises a coating, and the agent is present in the coating in an amount ranging between about 25% to about 70% by weight. 5950. The method of item 5735, wherein the implant further comprises a coating, and the coating comprises a polymer.
5951. The method of item 5735, wherein the implant comprises a first coating having a first composition and a second coating having a second composition.
5952. The method of item 5735, wherein the implant comprises a first coating having a first composition and a second coating having a second composition, wherein the first composition and the second composition are different.
5953. A method as in any one of items 5735-5952, wherein the device is a catheter.
5954. A method as in any one of items 5735-5952, wherein the device is a balloon catheter. • - — - .
5955. A method as in any one of items 5735-5952, wherein the device is a balloon.
5956. A method as in any one of items 5735-5952, wherein the device is a stent graft.
5957. A method as in any one of items 5735-5952, wherein the device is a guidewire.
5958. A method as in any one of items 5735-5952, wherein the device is a stent.
5959. A method as in any one of items 5735-5952, wherein the device is an intravascular stent.
5960. A method as in any one of items 5735-5952, wherein the device is a metallic stent. 5961. A method as in any one of items 5735-5952, wherein the device is a polymeric stent.
5962. A method as in any one of items 5735-5952, wherein the device is a biodegradable stent.
5963. A method as in any one of items 5735-5952, wherein the device is a non-biodegradable stent.
5964. A method as in any one of items 5735-5952, wherein the device is a self-expandable stent.
5965. A method as in any one of items 5735-5952, wherein the device is a balloon expandable stent.
5966. A method as in any one of items 5735-5952, wherein the device is a covered stent.
5967. A method as in any one of items 5735-5952, wherein the device is a drug eluting stent.
5968. A method as in any one of items 5735-5952, wherein the device is a stent that comprises a radio-opaque material.
5969. A method as in any one of items 5735-5952, wherein the device is a stent that comprises an echogenic material.
5970. A method as in any one of items 5735-5952, wherein the device is a stent that comprises an MRI responsive material.
5971. A method as in any one of items 5735-5952, wherein the device is an anastomotic connector device.
5972. A method as in any one of items 5735-5952, wherein the device is an artery to artery anastomotic connector device. 5973. A method as in any one of items 5735-5952, wherein the device is a vein to artery anastomotic connector device.
5974. A method as in any one of items 5735-5952, wherein the device is an artery to vein anastomotic connector device.
5975. A method as in any one of items 5735-5952, wherein the device is an artery to synthetic graft anastomotic connector device.
5976. A method as in any one of items 5735-5952, wherein the device is a synthetic graft to artery anastomotic connector device.
5977. A method as in any one of items 5735-5952, wherein the device is a vein to synthetic graft anastomotic connector device.
5978. A method as in any one of items 5735-5952, wherein the device is a synthetic graft to vein anastomotic connector device.
59797 A" method as in any one of items 5735-5952; wherein the device is a vascular clip.
5980. A method as in any one of items 5735-5952, wherein the device is a vascular suture.
5981. A method as in any one of items 5735-5952, wherein the device is a vascular clamp.
5982. A method as in any one of items 5735-5952, wherein the device is a suturing device.
5983. A method as in any one of items 5735-5952, wherein the device is an anastomotic coupler.
5984. A method as in any one of items 5735-5952, wherein the device is an automated or modified suture device. 5985. A method as in any one of items 5735-5952, wherein the device is a micromechanical anastomotic connector device.
5986. A method as in any one of items 5735-5952, wherein the device is an anastomotic coupling device that facilitates automated attachment of a graft or vessel to an aperature or orifice in a target vessel without the use of sutures or staples.
5987. A method as in any one of items 5735-5952, wherein the device is an anastomotic coupling device that comprises a tubular graft conduit and may be placed in a side wall of a target vessel so that the tubular graft conduit may be extended from the target vessel.
5988. A method as in any one of items 5735-5952, wherein the device is an anastomotic coupler in the form of a frame.
5989. A method as in any one of items 5735-5952, wherein the device is an anastomotic coupler in a ring-like form.
5990. A method as in any one of items 5735-5952, wherein the device is a resorbable anastomotic coupler.
5991. A method as in any one of items 5735-5952, wherein the device is an anastomotic coupler that comprises a bioabsorbable and elastomeric material.
5992. A method as in any one of items 5735-5952, wherein the device is an anastomotic coupler adapted to connect a first blood vessel with a second blood vessel with a graft vessel.
5993. A method as in any one of items 5735-5952, wherein the device is an anastomotic coupler adapted to connect a first blood vessel with a second blood vessel without a graft vessel. 5994. A method as in any one of items 5735-5952, wherein the device is an anastomotic coupler that is incorporated in the design of a vascular graft.
5995. A method as in any one of items 5735-5952, wherein the device is an anastomotic coupler that comprises a graft that incorporates a fixation mechanism.
5996. A method as in any one of items 5735-5952, wherein the device is an anastomotic coupler that comprises a compressible, expandable fitting for securing the ends of a bypass graft to two vessels.
5997. A method as in any one of items 5735-5952, wherein the device is an anastomotic coupler that comprises a pair of coupling disc members for joining two vessels in an end to end or end to side fashion.
5998. A method as in any one of items 5735-5952, wherein the device is a proximal aortic connector.
5999. A method as in any one of items 5735-5952, wherein the device is a distal coronary connector.
6000. A method as in any one of items 5735-5952, wherein the device is a bypass device made of a biocompatible material.
6001. A method as in any one of items 5735-5952, wherein the device is a bypass device made of at least partially a metal or metal alloy.
6002. A method as in any one of items 5735-5952, wherein the device is a bypass device made of at least partially a synthetic polymer.
6003. A method as in any one of items 5735-5952, wherein the device is a bypass device made of at least partially naturally derived polymer. 6004. A method as in any one of items 5735-5952, wherein the device is a tubular anastomotic coupler that comprises a tubular structure that may be attached directly to a proximal blood vessel.
6005. A method as in any one of items 5735-5952, wherein the device is a tubular anastomotic coupler that comprises a tubular structure that may be attached directly to a distal blood vessel.
6006. A method as in any one of items 5735-5952, wherein the device is a tubular anastomotic coupler that has a proximal end attachable to a proximal vessel and a distal end attachable to a bypass graft.
6007. A method as in any one of items 5735-5952, wherein the device is a tubular anatsomotic coupler that has a proximal end attachable to a graft vessel that is secured to a proximal blood vessel and a distal end attachable to a distal blood vessel.
6008. A method as in any one of items 5735-5952, wherein the device is an anastomotic connector device adapted for end to end anastomosis procedures.
6009. A method as in any one of items 5735-5952, wherein the device is an anastomotic stent.
6010. A method as in any one of items 5735-5952, wherein the device is an anastomotic sleeve.
6011. A method as in any one of items 5735-5952, wherein the device is an anastomotic connector device adapted for end to side anastomosis procedures.
6012. A method as in any one of items 5735-5952, wherein the device is a single lumen bypass device. 6013. A method as in any one of items 5735-5952, wherein the device is a multi-lumen bypass device.
6014. A method as in any one of items 5735-5952, wherein the device is an anastomotic coupling device that comprises a single tubular portion that may be used as a shunt to divert blood from a source vessel to a graft vessel.
6015. A method as in any one of items 5735-5952, wherein the device is an anastomotic coupling device that comprises more than one tubular portion, and wherein at least one tubular portion my be used as a shunt for diverting blood between a source vessel and a target vessel.
6016. A method as in any one of items 5735-5952, wherein the device is an anastomotic connector device that comprises a tubular portion, and wherein one or more ends of the tubular portion may be inserted into the end or into the side of one or more blood vessels.
6017. A method as in any one of items 5735-5952, wherein the device is a multi-lumen anastomotic connector device that at least one arm of the device may be attached to a graft vessel.
6018. A method as in any one of items 5735-5952, wherein the device is an anastomotic connector device that includes three or more tubular arms that extend form a junction site.
6019. A method as in any one of items 5735-5952, wherein the device is a multi-lumen anastomotic connector device that is generally T-shaped.
6020. A method as in any one of items 5735-5952, wherein the device is a multi-lumen anastomotic connector device that is generally Y-shaped. 6021. A method as in any one of items 5735-5952, wherein the device is an anastomotic connector device that comprises a tube for bypassing blood flow directly from a portion of the heart to a coronary artery.
6022. A method as in any one of items 5735-5952, wherein the device is an anastomotic connector device that comprises a network of interconnected tubular conduits.
6023. A method as in any one of items 5735-5952, wherein the device is an anastomotic connector device that is configured with two or more termini that provide a vessel interface without the need for sutures and a fluid communication through an intersecting lumen.
6024. A method for inhibiting scarring comprising placing a device that comprises a gastrointestinal stent and an anti-scarring agent or a composition comprising an anti-scarring agent into an animal host, wherein the agent inhibits scarring.
6025. The method of item 6024 wherein the agent is an adensosine A2A receptor antagonist.
6026. The method of item 6024 wherein the agent is an AKT inhibitor.
6027. The method of item 6024 wherein the agent is an alpha 2 integrin antagonist, wherein the alpha 2 integrin antagonist is Pharmaprojects No. 5754 (Merck KgaA).
6028. The method of item 6024 wherein the agent is an alpha 4 integrin antagonist. 6029. The method of item 6024 wherein the agent is an alpha 7 nicotinic receptor agonist.
6030. The method of item 6024 wherein the agent is an angiogenesis inhibitor selected from the group consisting of AG-12,958 (Pfizer), ATN-161 (Attention LLC), neovastat, an angiogenesis inhibitor from Jerina AG (Germany), NM-3 (Mercian), VGA-1155 (Taisho), FCE-26644 (Pfizer), FCE-26950 (Pfizer), FPMA (Meiji Daries), FR-111142 (Fujisawa), GGTI-298, GM-1306 (Ligand), GPA-1734 (Novartis), NNC-47-0011 (Novo Nordisk), herbamycin (Nippon Kayaku), lenalidomide (Celegene), IP-10 (NIH), ABT-828 (Abbott), KIN-841 (Tokushima University, Japan), SF-1126 (Semafore Pharmaceuticals), laminin technology (NIH), CHIR-258 (Chiron), NVP-AEW541 (Novartis), NVP-AEW541 (Novartis), Vt16907 (Alchemia), OXI-8007 (Oxigene), EG-3306 (Ark Therapeutics), Maspin (Arriva), ABT- 567 (Abbott), PPI-2458 (Praecis Pharmaceuticals), CC-5079, CC-4089 (Celgene), HIF-1 alpha inhibitors (Xenova), S-247 (Pfizer), AP-23573 (Ariad), AZD-9935 (Astra Zeneca), mebendazole (Introgen Therapeutics), MetAP-2 inhibitors (GlaxoSmithKline), AG-615 (Angiogene Pharmaceuticals), Tie-2 antagonists (Hybrigenics), NC-381 , CYC-381, NC-169, NC-219, NC-383, NC-384, NC-407 (Lorus Therapeutics), ATN-224 (Attenuon), ON-01370 (Onconova), Vitronectin antagonists (Amgen), SDX-103 (Salmedix), Vitronectin antagonists (Shire), CHP (Riemser), TEK (Amgen), Anecortave acetate (Alcon), T46.2 (Matrix Therapeutics), HG-2 (Heptagen), TEM antagonists (Genzyme), Oxi-4500 (Oxigene), ATN-161 (Attenuon), WX-293 (Wilex), M-2025 (Metris Therapeutics), Alphastatin (BioActa), YH-16 (Yantai Rongchang), BIBF-1120 (Boehringer Ingelheim), BAY-57-9352 (Bayer), AS- 1404 (Cancer Research Technology), SC-77964 (Pfizer), glycomimetics (BioTie Therapies), TIE-2 Inhibitors (Ontogen), DIMI, Octamer (Octamer), ABR-215050 (Active Biotech), ABT-518 (Abbott), KDR inhibitors (Abbott), BSF-466895 (Abbott), SCH-221153 (Schering-Plough), DAC:antiangiogenic (ConjuChem), TFPI (EntreMed), AZD-2171 (Astra-Zenaca), CDC-394 (Celgene), LY290293 (EIi Lilly), IDN-5390 (Indena), Kdr Kinase Inhibitors (Merck), CT-113020, CT-116433, CT-116563, CT-31890, CT-32228) (Cell Therapeutics), A-299620 (Abbott), TWEAK Inhibitor (Amgen), VEGF modulators (Johnson and Johnson), Tum-N53, tumstatin (Genzyme), Thios- 1 , Thios-2 (Thios Pharmaceuticals), MV-6401 (Miravant Medical Technologies), Spisulosine (PharmaMar), CEP-7055 (Cephalon), AUV-201 (Auvation), LM-609 (EIi Lilly), SKF-106615 (AnorMED), Oglufanide disodium (Cytran), BW-114 (Pharminox), Calreticulin (NIH), WX-678 (Wilex), SD-7784 (Pfizer), WX-UK1 (Wilex), SH-268 (Schering AG), 2-Me-PGA (Celgene), S- 137 (Pfizer), ZD-6126 (Angiogene Pharmaceuticals), SG-292 (SignalGen), Benefin (Lane Labs), A6, A36 (Angstrom), SB-2723005 (GlaxoSmithKline), SC-7 (Cell Therapeutics), ZEN-014 (AEterna Zentaris), 2-methoxyestradiol (EntreMed), NK-130119 (Nippon Kayaku), CC-10004 (Celgene), AVE- 8062A (Ajinomoto), Tacedinaline (Pfizer), Actinonin (Tokyo Metropolitan Institute of Medical Science), Lenalidomide (Celgene), VGA-1155, BTO-956 (SRI International), ER-68203r00.(Eisai), CT=6685 (UCB), JKC-362. (Phoenix Pharmaceuticals), DMI-3798 (DMl Biosciences, Angiomate (Ipsen), ZD-6474 (AstraZeneca), CEP-5214 (Cephalon), Canstatin (Genzyme), NM-3 (Mercian), Oridigm (MediQuest Therapeutics), Exherin (Adherex), BLS-0597 (Boston Life Sciences), PTC-299 (PTC Therapeutics), NPI-2358 (Nereus Pharmaceuticals), CGP-79787 (Novartis), AEE-788 (Novartis), CKD-732 (Chong Kun Dang), CP-564959 (OSI Pharmaceuticals), CM-101 (CarboMed), CT-2584, CT3501 (Cell Therapeutics), combretastatin and analogues and derivatives thereof (Oxigene), Rebimastat (Bristol-Meyers Squibb), Dextrin 2-sulfate (ML Laboratories), Cilengitide (Merk KGaA), NSC-706704 (Pharminox), KRN- 951 (Kirin Brewery), Ukrain, NSC-631570 (Nowicky Pharma), Tecogalan sodium (Daiichi Pharmaceutical), Tz-93 (Tsumura), TBC-1635 (Encysive Pharmaceuticals), TAN-1120 (Takeda), Semaxanib (Pfizer), BDI-7800 (Biopharmacopae), SD-186, SD-983 (Bristol-Meyers Squibb), SB-223245 (GlaxoSmithKline), SC-236 (Pfizer), RWJ-590973 (Johnson and Johnson), ILX-1850 (Genzyme), SC-68488, S-836 (Pfizer), CG-55069-11 (CuraGen), Ki-23057 (Kirin Brewery), CCX-700 (Chemoentryx), Pegaptanib octasodium (Giled Sciences), ANGIOCOL (available from Biostratum Inc.), or an analogue or derivative thereof.
6031. The method of item 6024 wherein the agent is an apoptosis antagonist.
6032. The method of item 6024 wherein the agent is an apoptosis activator.
6033. The method of item 6024 wherein the agent is a beta 1 integrin antagonist.
6034. The method of item 6024 wherein the agent is a beta tubulin inhibitor.
6035. The method of item 6024 wherein the agent is a blocker of enzyme production in Hepatitis C.
6036. The method of item 6024 wherein the agent is a Bruton's tyrosine kinase inhibitor.
6037. The method of item 6024 wherein the agent is a calcineurin inhibitor.
6038. The method of item 6024 wherein the agent is a caspase 3 inhibitor.
6039. The method of item 6024 wherein the agent is a CC chemokine receptor antagonist. 6040. The method of item 6024 wherein the agent is a cell cycle inhibitor selected from the group consisting of SNS-595 (Sunesis), synthadotin, KRX-0403, and an analogue or derivative thereof.
6041. The method of item 6024 wherein the agent is a cathepsin B inhibitor.
6042. The method of item 6024 wherein the agent is a cathepsin K inhibitor, wherein the cathepsin K inhibitor is 462795 (GlaxoSmithKline), INPL-022-D6 (Amura Therapeutics), or an analogue or derivative thereof.
6043. The method of item 6024 wherein the agent is a cathepsin L inhibitor.
6044. The method of item 6024 wherein the agent is a CD40 antagonist.
6045. The method of item 6024 wherein the agent is a chemokine receptor agonist.
6046. The method of item 6024 wherein the agent is a chymase inhibitor.
6047. The method of item 6024 wherein the agent is a collagenase antagonist.
6048. The method of item 6024 wherein the agent is a CXCR antagonist. 6049. The method of item 6024 wherein the agent is a cyclin dependent kinase inhibitor selected from the group consisting of a CDK-1 inhibitor, a CDK-2 inhibitor, a CDK- 4 inhibitor, a CDK-6 inhibitor, a CAK1 inhibitor from GPC Biotech and Bristol-Myers Squibb, RGB-286199 (GPC Biotech), an anticancer agent from Astex Technology, a CAK1 inhibitor from GPC Biotech, a CDK inhibitor from Sanofi-Aventis, a CDK1/CDK2 inhibitor from Amgen, a CDK2 inhibitor from SUGEN-2 (Pfizer), a hearing loss therapy agent (Sound Pharmaceuticals), PD-0332991 (Pfizer), RGB-286199 (GPC Biotech), Ro-0505124 (Hoffmann-La Roche), a Ser/Thr kinase inhibitor from Lilly (EIi Lilly), CVT-2584 (CAS No. 199986-75-9) (CV Therapeutics), and an analogue or derivative thereof.
6050. The method of item 6024 wherein the agent is a cyclooxygenase 1 inhibitor.
6051._The method of item 6024. wherein the agent is a DHFR inhibitor.
6052. The method of item 6024 wherein the agent is a dual integrin inhibitor.
6053. The method of item 6024 wherein the agent is an elastase inhibitor.
6054. The method of item 6024 wherein the agent is an elongation factor-1 alpha inhibitor.
6055. The method of item 6024 wherein the agent is an endothelial growth factor antagonist. 6056. The method of item 6024 wherein the agent is an endothelial growth factor receptor kinase inhibitor selected from the group consisting of sorafenib tosylate (Bayer), AAL-993 (Novartis), ABP-309 (Novartis), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), E- 7080 (Eisai), EG-3306 (Ark Therapeutics), EXEL-2880 (Exelixis), GW- 654652 (GlaxoSmithKline), a KDR inhibitor from LG Life Sciences, CT-6685 and CT-6729 (UCB), KRN-633 and KRN-951 (Kirin Brewery), OSI-930 (OSI Pharmaceuticals), SP-5.2 (Supratek Pharma), SU-11657 (Pfizer), a Tie-2 antagonist (Hybrigenics), a VEGFR-2 kinase inhibitor (Bristol-Myers Squibb), XL-647 (Exelixis), a KDR inhibitor from Abbott Laboratories, sorafenib tosylate, and an analogue or derivative thereof.
6057. The method of item 6024 wherein the agent is an endotoxin antagonist.
6058. The method of item 6024 wherein the_agent is an epothilone and tubulin binder.
6059. The method of item 6024 wherein the agent is an estrogen receptor antagonist.
6060. The method of item 6024 wherein the agent is an FGF inhibitor.
6061. The method of item 6024 wherein the agent is a famexyl transferase inhibitor.
6062. The method of item 6024 wherein the agent is farnesyltransferase inhibitor selected from the group of A-197574 (Abbott), a farnesyltransferase inhibitor from Servier, FPTIII (Strathclyde Institute for Drug R), LB-42908 (LG Life Sciences), Pharmaprojects No. 5063 (Genzyme), Pharmaprojects No. 5597 (Ipsen), Yissum Project No. B-1055 (Yissum), and an analogue or derivative thereof.
6063. The method of item 6024 wherein the agent is an FLT-3 kinase inhibitor.
6064. The method of item 6024 wherein the agent is an FGF receptor kinase inhibitor.
6065. The method of item 6024 wherein the agent is a fibrinogen antagonist selected from the group consisting of AUV-201 (Auvation), MG-13926 (Sanofi-Aventis), plasminogen activator (CAS No. 105913-11-9) (from Sanofi-Aventis or UCB), plasminogen activator-2 (tPA- 2) (Sanofi-Aventis), pro-urokinase (CAS No. 82657-92-9) (Sanofi-Aventis), and an analogue or derivative thereof.
6066. The method of item 6024 wherein the agent is a heat shock protein 90 antagonist selected from the group consisting of SRN-005 (Sirenade), geldanamycin, NSC-33050 (17-allylaminogeldanamycin; 17- AAG), 17-dimethylaminoethylamino-17-demethoxy-geldanamycin (17- DMAG), rifabutin (rifamycin XIV, 1',4-didehydro-1-deoxy-1 ,4-dihydro-5'-(2- methylpropyl)-1-oxo-)τ and an analogue or derivative thereof.
6067. The method of item 6024 wherein the agent is a histone deacetylase inhibitor.
6068. The method of item 6024 wherein the agent is an HMGCoA reductase inhibitor selected from the group consisting of an atherosclerosis therapeutic from Lipid Sciences, ATI-16000 (ARYx Therapeutics), KS-01-019 (Kos Pharmaceuticals), Pharmaprojects No. 2197 (Sanofi-Aventi), RP 61969 (Sanofi-Aventϊs), and an analogue or derivative thereof.
6069. The method of item 6024 wherein the agent is an ICAM inhibitor.
6070. The method of item 6024 wherein the agent is an IL, ICE and IRAK antagonist, wherein the antagonist is a CJ-14877, CP-424174 (Pfizer), NF-61 (Negma-Lerads), and an analogue or derivative thereof.
6071. The method of item 6024 wherein the agent is an IL-2 inhibitor.
6072. The method of item 6024 wherein the agent is an immunosuppressant selected from the group consisting of teriflunomide (Sanofi Aventis), chlorsulfaqumoxalone (NSC-339004), chlorsulfaquinoxalone sulfate, CS-712 (Sankyo), ismomultin alfa (CAS No. 457913-93-8) (Akzo Nobel), antiallergics from GenPat77, antiinflammatories or AT-005 (Androclus Therapeutics), autoimmune disease therapeutics from EpiVax, BN-007 (Bone), budesonide (CAS No. 51333-22- 3) (MAP Pharmaceuticals), CO-14 (Genzyme), edratide (CAS No. 433922- 67-9) (Teva), EP-314 (Enanta), eprovafen (CAS No. 101335-99-3) (Sanofi- Aventis), HWA-131 (CAS No. 118788-41-3) (Sanofi-Aventis), immunomodulators from MerLion Pharmaceuticals, immunosuppressives from Alchemia, IPL-12 (Inflazyme), MDL-9563 (CAS No. 27086-86-8) (Sanofi-Aventis), Pharmaprojects No. 2330 (Sanofi-Aventis), Pharmaprojects No. 6426 (Abgenix), PXS-25 (Pharmaxis), rosmarinic acid (CAS No. 20283-92-5) (Sanofi-Aventis), RP 42927 or RP 54745 (CAS No. 135330-08-4) (Sanofi-Aventis), SGN-35 (Seattle Genetics), ST-1959 (Sigma-Tau), type I diabetes therapy from SYNX Pharma, UNIL-88 (Debiopharm), VP-025 (Vasogen), VR-694 (Vectura), PRTX-001 (Protalex), and an analogue or derivative thereof.
6073. The method of item 6024 wherein the agent is an IMPDH (inosine monophosphate).
6074. The method of item 6024 wherein the agent is an integrin antagonist.
6075. The method of item 6024 wherein the agent is an interleukin antagonist.
6076. The method of item 6024 wherein the agent is an inhibitor of type III receptor tyrosine kinase.
6077. The method of item 6024 wherein the agent is an irreversible inhibitor of enzyme methionine aminopeptidase type 2.
6078. The method of item 6024 wherein the agent is an isozyme selective delta protein kinase C inhibitor.
6079. The method of item 6024 wherein the agent a JAK3 enzyme inhibitor.
6080. The method of item 6024 wherein the agent is a JNK inhibitor.
6081. The method of item 6024 wherein the agent is a kinase inhibitor. 6082. The method of item 6024 wherein the agent is kinesin antagonist.
6083. The method of item 6024 wherein the agent is a kinesin antagonist.
6084. The method of item 6024 wherein the agent is a leukotriene inhibitor and antagonist selected from the group consisting of ambicromil (CAS No. 58805-38-2) (Sanofi-Aventis), amelubant (CAS No. 346735-24-8) (Boehringer Ingelheim), DW-1141 (Dong Wha), ebselen (Daiichi Pharmaceutical), ibudilast (Kyorin), leucotriene inhibitors from Sanofi-Aventis, lymphotoxin-beta receptor (LT-D) from Biogen Idee, Pharmaprojects No. 1535 and 2728 (CAS No. 119340-33-9) (Sanofi- Aventis), R-112 (Rigel), Rev-5367 (CAS No. 92532-05-3) (Sanofi-Aventis), RG-14893 (CAS No. 141835-49-6) (Sanofi-Aventis), RG-5901-A (CAS No. 101910-24-1), 92532-23-5, RP 66153 (CAS No. 142422r79-5), RP 66364 (CAS No.186916-92-5), RP 69698 (CAS No. 141748-00-7) (Sanofi-Aventis), SC-411930 (Pfizer), SC-41930 (CAS No. 120072-59-5) (Pfizer), SC-50605 (CAS No. 138828-39-4) (Pfizer), SC-51146 (CAS No. 141059-52-1), SC- 53228 (CAS No. 153633-01-3) (Pfizer), spaglumic acid (ZY-15106) (CAS No.3106-85-2), 80619-64-3 (Novartis), tipredane (CAS No. 85197-77-9) (Bristol-Myers Squibb), U-75302 (CAS No. 119477-85-9) (Pfizer), and analogue or derivative thereof.
6085. The method of item 6024 wherein the agent is an MAP kinase inhibitor.
6086. The method of item 6024 wherein the agent is a matrix metalloproteinase inhibitor. 6087. The method of item 6024 wherein the agent is an MCP- CCR2 inhibitor.
6088. The method of item 6024 wherein the agent is an mTOR inhibitor.
6089. The method of item 6024 wherein the agent is an mTOR kinase inhibitor.
6090. The method of item 6024 wherein the agent is a microtubule inhibitor selected from the group consisting of antibody- maytansinoid conjugates from Biogen Idee, colchicines (MantiCore Pharmaceuticals), anticancer immunoconjugates from Johnson & Johnson, DIME from Octamer, gni-1f (GNI), huC242-DM4, huMy9-6-DM1 (ImmunoGen), IDN-5404 (Indena), IMO-098, IMOderm (Imotep), mebendazole (Introgen Therapeutics), microtubule poisons from Cambridge Enterprise, paclitaxel such as LOTAX from Aphios (CAS No. 33069-62-4), Genexol-PM from Samyang, Pharmaprojects No. 6383 (Tapestry Pharmaceuticals), RPR-112378 (Sanofi-Aventis), SGN-75 (Seattle Genetics), SPL-7435 (Starpharma), SSR-250411 (Sanofi-Aventis), trastuzumab-DM1 (Genentech), vinorelbine, and an analogue or derivative thereof.
6091. The method of item 6024 wherein the agent is an MIF inhibitor.
6092. The method of item 6024 wherein the agent is an MMP inhibitor.
6093. The method of item 6024 wherein the agent is a neurokinin (NK) antagonist selected from the group consisting of anthrotainin (CAS No. 148084-40-6) (Sanofi-Aventis), an IBS thereapeutic from ArQuIe, MDL-105212A (CAS No. 167261-60-1) (Ssanofi-Aventis), Pharmaprojects No. 2744, 3258 (CAS No. 139167-47-8) 4006, 4201, or 5986 (Sanofi-Aventis), RP 67580 (CAS No. 135911-02-3), SR-144190 (CAS No. 201152-86-5), SSR-240600, SSR-241586 (Sanofi-Aventis), TKA-457 (Novartis), vestipitant mesylate (CAS No. 334476-64-1) (GlaxoSmithKline), Win-64821 (Sanofi-Aventis), PRX-96026 (Predix Pharmaceuticals), and an analogue or derivative thereof.
6094. The method of item 6024 wherein the agent is an NF kappa B inhibitor selected from the group consisting of AVE-0545 or AVE- 0547 (Sanofi-Aventis), bortezomib (CAS No. 179324-69-7) (Millennium Pharmaceuticals), dexanabinol (CAS No. 112924-45-5) (Pharmos), dexlipotam (Viatris), Pharmaprojects No. 6283 (INDRA) (OXiGENE), IPL- 576092 (CAS No. 137571-30-3) (Inflazyme), NFKB decoy (Corgentech), NFKB decoy oligo (AnGes MG), NFKB's from Ariad, osteoporosjs treatments or S5 (F005) from Fulcrum Pharmaceuticals, P61 (Phytopharm), R-flurbiprofen (CAS No. 5104-49-4) (Encore Pharmaceuticals), and an analogue or derivative thereof.
6095. The method of item 6024 wherein the agent is a nitric oxide agonist.
6096. The method of item 6024 wherein the agent is an ornithine decarboxylase inhibitor.
6097. The method of item 6024 wherein the agent is a p38 MAP kinase inhibitor selected from the group consisting of AZD-6703 (AstraZeneca), JX-401 (Jexys Pharmaceuticals), BMS-2 (Bristol-Myers Squibb), a p38 MAP kinase inhibitor from Novartis, a p38-alpha MAP kinase inhibitor from Amphora, Pharmaprojects No. 5704 (Pharmacopeia), RPR- 200765A (Sanofi-Aventis), SD-282 (Johnson & Johnson), TAK-715 (Takeda), and an analogue or derivative thereof.
6098. The method of item 6024 wherein the agent is a palmitoyl-protein thioesterase inhibitor.
6099. The method of item 6024 wherein the agent is a PDGF receptor kinase inhibitor selected from the group consisting of AAL-993, AMN-107, or ABP-309 (Novartis), AMG-706 (Amgen), BAY-57-9352 (Bayer), CDP-860 (UCB), E-7080 (Eisai), imatinib (CAS No. 152459-95-5) (Novartis), OSI-930 (OSI Pharmaceuticals), RPR-127963E (Sanofi-Aventis), RWJ-540973 (Johnson & Johnson), sorafenib tosylate (Bayer), SU-11657 (Pfizer), tandutinib (CAS No. 387867-13-2) (Millennium Pharmaceuticals), vatalanib (Novartis), ZK-CDK (Schering AG), and an analogue or derivative thereof.
6100. The method of item 6024 wherein the agent is (-)- halofenate (Metabolex), AMG-131 (Amgen), antidiabetics from Japan Tobacco, AZD-4619, AZD-8450, AZD-8677 (AstraZeneca), DRF-10945, balaglitazone (Dr Reddy's), CS-00088, CS-00098 (Chipscreen Biosciences), E-3030 (Eisai), etalocib (CAS No. 161172-51-6) (EIi Lilly), GSK-641597 (Ligand), GSK-677954 (GlaxoSmithKline), GW-409544 (Ligand), GW- 590735 (GlaxoSmithKline), K-111 (Hoffmann-La Roche), LY-518674 (EIi Lilly), LY-674 (Ligand), LY-929 (Ligand), MC-3001, MC-3002 (MaxoCore Pharmaceuticals), metformin HCI + pioglitazone (CAS No. 1115-70-4 and
112529-15-4), ACTOPLUS MET from Andrx), muraglitazar (CAS No. 331741-94-7) (Bristol-Myers Squibb), naveglitazar (Ligand), oleoylethanolamide (Kadmus Pharmaceuticals), ONO-5129, pioglitazone hydrochloride (CAS No. 111025-46-8 and 112529-15-4) (Takeda), PLX-204 (Plexxikon), PPAR agonists from Genfit, PPAR delta agonists from EIi Lilly, PPAR-alpha agonists from CrystalGenomics, PPAR-gamma modulators and PPAR-β modulators from CareX, rosiglitazone maleate (CAS No. 122324- 73-4 or 155141-29-0) (GlaxoSmithKline), rosiglitazone maleate/glimepir (CAS No. 155141-29-0 and 93479-97-1), AVANDARYL, rosiglitazone maleate/metformin extend (CAS No. 155141-29-0 and 657-24-9), AVANDAMET, rosiglitazone maleate+metformin, AVANDAMET (GlaxoSmithKline), tesaglitazar (AstraZeneca), and an analogue or derivative thereof.
6101. The method of item 6024 wherein the agent is a phosphatase inhibitor.
6102. The method of item 6024 wherein the agent is a phosphodiesterase (PDE) inhibitor selected from the group consisting of avanafil (Tanabe Seiyaku), dasantafil (CAS No. 569351-91-3) (Schering- Plough), A-906119 (CAS No. 134072-58-5), DL-850 (Sanofi-Aventis), GRC- 3015,-GRO3566, GRC-3886 (Glenmark), HWA-153 (CAS No. 56395-66-5) (Sanofi-Aventis), hydroxypumafentrine (Altana), IBFB-130011, IBFB-14-016, IBFB-140301 , IBFB-150007, IBFB-211913 (IBFB Pharma), L-826141 (Merck & Co), medorinone (CAS No. 88296-61-1) (Sanofi-Aventis), MEM- 1917 (Memory Pharmaceuticals), ND-1251 (Neuro3d), PDE inhibitors from ICOS, PDE IV inhibitors from Memory Pharmaceuticals and CrystalGenomics, Pharmaprojects No. 2742 and 6141 (Sanofi-Aventis), QAD-171 (Novartis), RHC-2963 (CAS No. 76993-12-9 and 76993-14-1), RPR-117658, RPR-122818 derivatives, SR-24870 , and RPR-132294 (Sanofi-Aventis), SK-350 (ln2Gen), stroke therapy agents from deCODE Genetics, TAS-203 (Taiho), tofimilast (CAS No. 185954-27-2) (Pfizer), UK- 371800 (Pfizer), WIN-65579 (CAS No. 158020-82-7) (Sanofi-Aventis), IBFB- 130020 (IBFB Pharma), OPC-6535 (CAS No. 145739-56-6) (Otsuka), a phosphodiesterase III inhibitor, enoximone, a phosphodiesterase IV inhibitor, fosfosal, Atopik (Barrier Therapeutics), triflusal, a phosphodiesterase V inhibitor, and an analogue or derivative thereof. 6103. The method of item 6024 wherein the agent is a PKC inhibitor.
6104. The method of item 6024 wherein the agent is a platelet activating factor antagonist.
6105. The method of item 6024 wherein the agent is a platelet-derived growth factor receptor kinase inhibitor.
6106. The method of item 6024 wherein the agent is a prolyl hydroxylase inhibitor.
6107. The method of item 6024 wherein the agent is a polymorphonuclear neutrophil inhibitor.
6108. The method of item 6024 wherein the agent is a protein kinase B inhibitor.
6109. The method of item 6024 wherein the agent is a protein kinase C stimulant.
6110. The method of item 6024 wherein the agent is a purine nucleoside analogue.
6111. The method of item 6024 wherein the agent is a purinoreceptor P2X antagonist.
6112. The method of item 6024 wherein the agent is a Raf kinase inhibitor. 6113. The method of item 6024 wherein the agent is a reversible inhibitor of ErbB1 and ErbB2.
6114. The method of item 6024 wherein the agent is a ribonucleoside triphosphate reductase inhibitor.
6115. The method of item 6024 wherein the agent is an SDF- 1 antagonist.
6116. The method of item 6024 wherein the agent is a sheddase inhibitor.
6117. The method of item 6024 wherein the agent is an SRC inhibitor.
j3118. The method of item 6024 wherein the agent is a stromelysin inhibitor.
6119. The method of item 6024 wherein the agent is an Syk kinase inhibitor.
6120. The method of item 6024 wherein the agent is a teiomerase inhibitor.
6121. The method of item 6024 wherein the agent is a TGF beta inhibitor selected from the group consisting of pirfenidone (CAS No. 53179-13-8) (MARNAC), tranilast (CAS No. 53902-12-8) (Kissei), IN-1130 (ln2Gen), mannose-6-phosphate (BTG), TGF-β antagonists from Inflazyme (Pharmaprojects No. 6075), TGF-β antagonists from Sydney, non-industrial source), TGF-β I receptor kinase inhibitors from EIi Lilly, TGF-β receptor inhibitors from Johnson & Johnson, and an analogue or derivative thereof. 6122. The method of item 6024 wherein the agent is a TNFα antagonist or TACE inhibitor selected from the group consisting of adalimumab (CAS No. 331731-18-1) (Cambridge Antibody Technology), AGIX-4207 (AtheroGenics), AGT-1 (Advanced Biotherapy), an antiinflammatory from Borean Pharma, Cellzome, or Paradigm Therapeutics, anti-inflammatory vaccine (TNF-alpha kinoid) from Neovacs, humanized anti-TNF antibody or an anti-TNF MAb (CB0006) Celltech (UCB), apratastat (CAS No. 287445-51-0) (Wyeth), BMS-561392 (Bristol-Myers Squibb), BN- 006 (Bone), certolizumab pegol (CAS No. 428863-50-7 or CH-138 (UCB), cilomilast (CAS No. 153259-65-5) (GlaxoSmithKline), CR-1 (Nuada Pharmaceuticals), CRx-119 (CombinatoRx), D-5410 (UCB), dacopafant (CAS No. 125372-33-0) (Sanofi-Aventis), dersalazine (CAS No. 188913-57- 7/188913-58-8) (Uriach), etanercept (CAS No. 185243-69-0) (Amgen), ethyl pyruvate (Critical (Critical Therapeutics), golimumab (CAS No. 476181-74-5) (Johnson & Johnson), hormono-immunotherapy from Ipsen, CDP571 (e.g., Hurnicade-from UCB)r IC-485 (ICOS), infliximabiCAS No. 170277-31-3) (Johnson & Johnson), IP-751 (Manhattan Pharmaceuticals), ISIS-104838 (CAS No. 250755-32-9) (ISIS Pharmaceuticals), lenalidomide (CAS No. 191732-72-6) (Celgene), lentinan (CAS No. 37339-90-5) (Ajinomoto), MDL- 201112 (CAS No. 142130-73-2) (Sanofi-Aventis), medroxyprogesterone (CAS No. 520-85-4) (InKine Pharmaceutical), N-acetylcysteϊne (CAS No. 616-91-1) (Zambon), NBE-P2 (DIREVO Biotech), nerelimomab (CAS No. 162774-06-3) (Chiron), OM-294DP (OM PHARMA), onercept (CAS No. 199685-57-9) (Yeda), PASSTNF-alpha (Verigen), pentoxifylline or oxypentifylline (Sanofi-Aventis), Pharmaprojects No. 4091 , 4241 , 4295, or 4488 (Sanofi-Aventis), Pharmaprojects No. 5480 (Amgen), Pharmaprojects No. 6749 (Cengent), pirfenidone (CAS No. 53179-13-8) (MARNAC), RPR- 132294 (Sanofi-Aventis), S5 (F002) (Fulcrum Pharmaceuticals), simvastatin (CAS No. 79902-63-9) (Merck & Co), STA-6292 (Synta Pharmaceuticals), tacrolimus (CAS No. 104987-11-3) (Fujisawa LifeCycle Pharma), talactoferrin alfa (CAS No. 308240-58-6) (Agennix), thalidomide (CAS No. 50-35-1) (Celgene), TNF antagonists form ProStrakan, and Synergen, TNF inhibitors (Amgen), TNF-alpha antagonists from Dynavax Technologies and Jerina AG (Germany), TNF-alpha inhibitors from IBFB Pharma and Xencor (Xencor), torbafylline (CAS No. 105102-21-4) (Sanofi-Aventis), UR-1505 (Uriach), VT-346 (Viron Therapeutics), YSIL6 (Y's Therapeutics), YSTH2 (Y's Therapeutics), NPI-1302a-3 (Nereus Pharmaceuticals, a TNF antagonist from Jerina AG (Germany), dersalazine, and an analogue or derivative thereof.
6123. The method of item 6024 wherein the agent is a Toll receptor inhibitor.
6124. The method of item 6024 wherein the agent is a tubulin antagonist.
6125. The method of item.6024_whereiη the agent is a tyrosine kinase inhibitor selected from the group consisting of SU-011248, SUTENT from Pfizer Inc. (New York, NY), BMS-354825, PN-355 (Paracelsian Pharmaceuticals), AGN-199659 (Allergan), AAL-993 or ABP- 309 (Novartis), adaphostin (NIH), AEE-788 (Novartis), AG-013736 (OSI Pharmaceuticals), AG-13736 (Pfizer), ALT-110 (Alteris Therapeutics), AMG- 706 (Amgen), anticancer MAbs from Xencor, anti-EGFrvlll MAbs from Abgenix, anti-HER2 MAb from Abiogen, AZD-2171 or AZD-9935 (AstraZeneca), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), CEP-5214 (Cephalon), CEP-7055 (Cephalon), cetuximab (ImClone Systems), CHIR-200131 and CHIR-258 (Chiron), CP-547632 (OSI Pharmaceuticals), CP-724714 (Pfizer), CT-301 (Creabilis Therapeutics), D- 69491 (Baxter International), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EGFR/ErbB2 inhibitors from Array BioPharma, erlotinib (CAS No. 183319- 69-9) (OSI Pharmaceuticals), EXEL-2880 (Exelixis), FK-778 (Sanofi- Aventis) , gefitinib (CAS No. 184475-35-2) (AstraZeneca), GW-2286 or GW- US2006/013030
654652 (GlaxoSmithKline), her2/neu antigen from AlphaVax, HER-2/neu inhibitor from Generex, Herzyme (Medipad) (Sima Therapeutics), HKI-272 (Wyeth), HuMax-EGFr (Genmab), idronoxil (CAS No. 81267-65-4) (Novogen), IGF-1 inhibitors from Ontogen, IMC-11F8 (ImCIone Systems), kahalalide F (CAS No. 149204-42-2) (PharmaMar), KDR inhibitor from LG Life Sciences, KDR inhibitors from Abbott Laboratories, KDR kinase inhibitors (UCB), Kdr kinase inhibitors from Merck & Co, KRN-633 and KRN- 951 (Kirin Brewery), KSB-102 (Xenova), lapatinib ditosylate (CAS No. 388082-78-8) (GlaxoSmithKline), matuzumab (Merck KGaA), MDX-214 (Medarex), ME-103 (Pharmexa), MED-A300 (Gerolymatos), MNAC-13 (Lay Line Genomics), nimotuzumab (Center of Molecular Immunology), NSC- 330507 or NSC-707545 (NIH), NV-50 (Novogen), OSI-930 (OSI Pharmaceuticals), panitumumab (Abgenix), pelitinib (CAS No. 287933-82-7) (Wyeth), pertuzumab (CAS No. 380610-27-5) (Genentech), Pharmaprojects No. 3985 (Sanofi-Aventis), prostate cancer therapeutics from Sequenom (SQPC35, SQPC36, SQPC90), removab and remoxab (Trion Pharma), RG- 13022 (CAS No. 136831-48-6), RG-13291 (CAS No. 138989-50-1), or RG- 14620 (CAS No. 136831-49-7) (Sanofi-Aventis), RM-6427 (Romark), RNAi breast cancer therapy from Benitec, RP 53801 (CAS No. 125882-88-4) (Sanofi-Aventis), sorafenib tosylate (Bayer), SU-11657 (Pfizer), Tie-2 antagonists from Semaia (Hybrigenics), Tie-2 inhibitors from Ontogen, trastuzumab (CAS No. 180288-69-1) (Genentech), tyrosine kinase inhibitors from Sanofi-Aventis, U3-1287, U3-1565, U3-1784, U3-1800 (U3 Pharma), vatalanib (Novartis), VEGFR-2 kinase inhibitor from Bristol-Myers Squibb, XL-647 (Exelixis), ZD-6474 (AstraZeneca), ZK-CDK (Schering AG), an EGFR tyrosine kinase inhibitor, EKB-569 (Wyeth), and an analogue or derivative thereof.
6126. The method of item 6024 wherein the agent is a VEGF inhibitor. 6127. The method of item 6024 wherein the agent is a vitamin D receptor agonist.
6128. The method of item 6024 wherein the agent is ZD-6474 (an angiogenesis inhibitor).
6129. The method of item 6024 wherein the agent is AP- 23573 (an mTOR inhibitor).
6130. The method of item 6024 wherein the agent is synthadotin (a tubulin antagonist).
6131. The method of item 6024 wherein the agent is S-0885 (a collagenase inhibitor).
-6132. The method of item 6024_wherein the agent is aplidine (an elongation factor-1 alpha inhibitor).
6133. The method of item 6024 wherein the agent is ixabepilone (an epithilone).
6134. The method of item 6024 wherein the agent is IDN- 5390 (an angiogenesis inhibitor).
6135. The method of item 6024 wherein the agent is SB- 2723005 (an angiogenesis inhibitor).
6136. The method of item 6024 wherein the agent is ABT-518 (an angiogenesis inhibitor). 6137. The method of item 6024 wherein the agent is combretastatin (an angiogenesis inhibitor).
6138. The method of item 6024 wherein the agent is anecortave acetate (an angiogenesis inhibitor).
6139. The method of item 6024 wherein the agent is SB- 715992 (a kinesin antagonist).
6140. The method of item 6024 wherein the agent is temsirolimus (an mTOR inhibitor).
6141. The method of item 6024 wherein the agent is adalimumab (a TNFα antagonist).
6142. The method of item 6024, wherein the composition comprises a polymer.
6143. The method of item 6024, wherein the composition comprises a polymer, and the polymer is, or comprises, a copolymer.
6144. The method of item 6024, wherein the composition comprises a polymer, and the polymer is, or comprises, a block copolymer.
6145. The method of item 6024, wherein the composition comprises a polymer, and the polymer is, or comprises, a random copolymer.
6146. The method of item 6024, wherein the composition comprises a polymer, and the polymer is, or comprises, a biodegradable polymer. 6147. The method of item 6024, wherein the composition comprises a polymer, and the polymer is, or comprises, a nonbiodegradable polymer.
6148. The method of item 6024, wherein the composition comprises a polymer, and the polymer is, or comprises, a hydrophilic polymer.
6149. The method of item 6024, wherein the composition comprises a polymer, and the polymer is, or comprises, a hydrophobic polymer.
6150. The method of item 6024, wherein the composition comprises a polymer, and the polymer is, or comprises, a polymer having hydrophilic domains.
6151. The method of item 6024, wherein the composition comprises a polymer, and the polymer is, or comprises, a polymer having hydrophobic domains.
6152. The method of item 6024, wherein the composition comprises a polymer, and the polymer is, or comprises, a non-conductive polymer.
6153. The method of item 6024, wherein the composition comprises a polymer, and the polymer is, or comprises, an elastomer.
6154. The method of item 6024, wherein the composition comprises a polymer, and the polymer is, or comprises, a hydrogel. 6155. The method of item 6024, wherein the composition comprises a polymer, and the polymer is, or comprises, a silicone polymer.
6156. The method of item 6024, wherein the composition comprises a polymer, and the polymer is, or comprises, a hydrocarbon polymer.
6157. The method of item 6024, wherein the composition comprises a polymer, and the polymer is, or comprises, a styrene-derived polymer.
6158. The method of item 6024, wherein the composition comprises a polymer, and the polymer is, or comprises, a butadiene-derived polymer.
6159. The method of item 6024, wherein the composition comprises a polymer, and the polymer is, or comprises, a macromer.
6160. The method of item 6024, wherein the composition comprises a polymer, and the polymer is, or comprises, a poly(ethylene glycol) polymer.
6161. The method of item 6024, wherein the composition comprises a polymer, and the polymer is, or comprises, an amorphous polymer.
6162. The method of item 6024, wherein the composition further comprises a second pharmaceutically active agent.
6163. The method of item 6024, wherein the composition further comprises an anti-inflammatory agent. 6164. The method of item 6024, wherein the composition further comprises an agent that inhibits infection.
6165. The method of item 6024, wherein the composition further comprises an anthracycline.
6166. The method of item 6024, wherein the composition further comprises doxorubicin.
6167. The method of item 6024 wherein the composition further comprises mitoxantrone.
6168. The method of item 6024 wherein the composition further comprises a fluoropyrimidine.
6169. The method of item 6024, wherein the composition further comprises 5-fluorouracil (5-FU).
6170. The method of item 6024, wherein the composition further comprises a folic acid antagonist.
6171. The method of item 6024, wherein the composition further comprises methotrexate.
6172. The method of item 6024, wherein the composition further comprises a podophyiotoxin.
6173. The method of item 6024, wherein the composition further comprises etoposide. 6174. The method of item 6024, wherein the composition further comprises camptothecin.
6175. The method of item 6024, wherein the composition further comprises a hydroxyurea.
6176. The method of item 6024, wherein the composition further comprises a platinum complex.
6177. The method of item 6024, wherein the composition further comprises cisplatin.
6178. The method of item 6024 wherein the composition further comprises an anti-thrombotic agent.
6179: The method of item 6024, wherein the composition further comprises a visualization agent.
6180. The method of item 6024, wherein the composition further comprises a visualization agent, and the visualization agent is a radiopaque material, wherein the radiopaque material comprises a metal, a halogenated compound, or a barium containing compound.
6181. The method of item 6024, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, barium, tantalum, or technetium.
6182. The method of item 6024, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, an MRI responsive material. 6183. The method of item 6024, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, a gadolinium chelate.
6184. The method of item 6024, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, iron, magnesium, manganese, copper, or chromium.
6185. The method of item 6024, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, iron oxide compound.
6186. The method of item 6024, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, a dye, pigment, or colorant.
6187. The method of item 6024 wherein the agent is released in effective concentrations from the composition comprising the agent by diffusion over a period ranging from the time of administration to about 90 days.
6188. The method of item 6024 wherein the agent is released in effective concentrations from the composition comprising the agent by erosion of the composition over a period ranging from the time of administration to about 90 days.
6189. The method of item 6024 wherein the composition further comprises an inflammatory cytokine.
6190. The method of item 6024 wherein the composition further comprises an agent that stimulates cell proliferation. 6191. The method of item 6024 wherein the composition further comprises a polymeric carrier.
6192. The method of item 6024 wherein the composition is in the form of a gel, paste, or spray.
6193. The method of item 6024 wherein the implant is partially constructed with the agent or the composition.
6194. The method of item 6024 wherein the implant is fully constructed with the agent or the composition.
6195. The method of item 6024 wherein the implant is impregnated with the agent or the composition.
6196. The method of item 6024, wherein the agent or the composition forms a coating, and the coating directly contacts the implant.
6197. The method of item 6024, wherein the agent or the composition forms a coating, and the coating indirectly contacts the implant.
6198. The method of item 6024 wherein the agent or the composition forms a coating, and the coating partially covers the implant.
6199. The method of item 6024, wherein the agent or the composition forms a coating, and the coating completely covers the implant.
6200. The method of item 6024 wherein the agent or the composition is located within pores or holes of the implant. 6201. The method of item 6024 wherein the agent or the composition is located within a channel, lumen, or divet of the implant.
6202. The method of item 6024 wherein the implant further comprising an echogenic material.
6203. The method of item 6024 wherein the implant further comprises an echogenic material, wherein the echogenic material is in the form of a coating.
6204. The method of item 6024 wherein the implant is sterile.
6205. The method of item 6024 wherein the agent is delivered from the implant, wherein the agent is released into tissue in the vicinity of the implant after deployment of the implant.
6206. The method of item 6024 wherein the agent is delivered from the implant, wherein the agent is released into tissue in the vicinity of the implant after deployment of the implant, wherein the tissue is connective tissue.
6207. The method of item 6024 wherein the agent is delivered from the implant, wherein the agent is released into tissue in the vicinity of the implant after deployment of the implant, wherein the tissue is muscle tissue.
6208. The method of item 6024 wherein the agent is delivered from the implant, wherein the agent is released into tissue in the vicinity of the implant after deployment of the implant, wherein the tissue is nerve tissue. 6209. The method of item 6024 wherein the agent is delivered from the implant, wherein the agent is released into tissue in the vicinity of the implant after deployment of the implant, wherein the tissue is epithelium tissue.
6210. The method of item 6024 wherein the agent is delivered from the implant, wherein the agent is released in effective concentrations from the implant over a period ranging from the time of deployment of the implant to about 1 year.
6211. The method of item 6024 wherein the agent is delivered from the implant, wherein the agent is released in effective concentrations from the implant over a period ranging from about 1 month to 6 months.
6212. The method of item 6024 wherein the agent is delivered from the implant, wherein the agent is released in effective concentrations from the implant over a period ranging from about 1 - 90 days.
6213. The method of item 6024 wherein the agent is delivered from the implant, wherein the agent is released in effective concentrations from the implant at a constant rate.
6214. The method of item 6024 wherein the agent is delivered from the implant, wherein the agent is released in effective concentrations from the implant at an increasing rate.
6215. The method of item 6024 wherein the agent is delivered from the implant, wherein the agent is released in effective concentrations from the implant at a decreasing rate. 6216. The method of item 6024 wherein the agent is delivered from the implant, wherein the implant comprises about 0.01 Dg to about 10 Dg of the agent.
6217. The method of item 6024 wherein the agent is delivered from the implant, wherein the implant comprises about 10 Dg to about 10 mg of the agent.
6218. The method of item 6024 wherein the agent is delivered from the implant, wherein the implant comprises about 10 mg to about 250 mg of the agent.
6219. The method of item 6024 wherein the agent is delivered from the implant, wherein the implant comprises about 250 mg to about
1000 mg of the agent.
6220. The method of item 6024 wherein the agent is delivered from the implant, wherein the implant comprises about 1000 mg to about 2500 mg of the agent.
6221. The method of item 6024 wherein the agent is delivered from the implant, wherein a surface of the implant comprises less than 0.01 Dg of the agent per mm2 of implant surface to which the agent is applied.
6222. The method of item 6024 wherein the agent is delivered from the implant, wherein a surface of the implant comprises about 0.01 Dg to about 1 Dg of the agent per mm2 of implant surface to which the agent is applied.
6223. The method of item 6024 wherein the agent is delivered from the implant, wherein a surface of the implant comprises about 1 Dg to about 10 Dg of the agent per mm2 of implant surface to which the agent is applied.
6224. The method of item 6024 wherein the agent is delivered from the implant, wherein a surface of the implant comprises about 10 Dg to about 250 Dg of the agent per mm2 of implant surface to which the agent is applied.
6225. The method of item 6024 wherein the agent is delivered from the implant, wherein a surface of the implant comprises about 250 Dg to about 1000 Dg of the agent per mm2 of implant surface to which the agent is applied.
6226. The method of item 6024 wherein the agent is delivered from the implant, wherein a surface of the implant comprises about 1000 Dg io about 2500 Dg of the agent per mm2 of implant surface to which the agent is applied.
6227. The method of item 6024, wherein the implant further comprises a coating, and the coating is a uniform coating.
6228. The method of item 6024, wherein the implant further comprises a coating, and the coating is a non-uniform coating.
6229. The method of item 6024, wherein the implant further comprises a coating, and the coating is a discontinuous coating.
6230. The method of item 6024, wherein the implant further comprises a coating, and the coating is a patterned coating. 6231. The method of item 6024, wherein the implant further comprises a coating, and the coating has a thickness of 100 Dm or less.
6232. The method of item 6024, wherein the implant further comprises a coating, and the coating has a thickness of 10 Dm or less.
6233. The method of item 6024, wherein the implant further comprises a coating, and the coating adheres to the surface of the implant upon deployment of the implant.
6234. The method of item 6024, wherein the implant further comprises a coating, and the coating is stable at room temperature for a period of at least 1 year.
6235. The method of item 6024, wherein the implant further comprises a coating, and the agent is present in the coating in an amount ranging between about 0.0001% to about 1% by weight.
6236. The method of item 6024, wherein the implant further comprises a coating, and the agent is present in the coating in an amount ranging between about 1% to about 10% by weight.
6237. The method of item 6024, wherein the implant further comprises a coating, and the agent is present in the coating in an amount ranging between about 10% to about 25% by weight.
6238. The method of item 6024, wherein the implant further comprises a coating, and the agent is present in the coating in an amount ranging between about 25% to about 70% by weight. 6239. The method of item 6024, wherein the implant further comprises a coating, and the coating comprises a polymer.
6240. The method of item 6024, wherein the implant comprises a first coating having a first composition and a second coating having a second composition.
6241. The method of item 6024, wherein the implant comprises a first coating having a first composition and a second coating having a second composition, wherein the first composition and the second composition are different.
6242. A method as in any one of items 6024-6241 , wherein the device is an esophageal stent.
6243. A method as in any one of items 6024-6241 , wherein the device is a biliary stent. . . . .._ . ..
6244. A method as in any one of items 6024-6241 , wherein the device is a colonic stent.
6245. A method as in any one of items 6024-6241, wherein the device is a pancreatic stent.
6246. A method for inhibiting scarring comprising placing a device that comprises a tracheal and bronchial stent (e.g., an implant) and an anti-scarring agent or a composition comprising an anti-scarring agent into an animal host, wherein the agent inhibits scarring.
6247. The method of item 6246 wherein the agent is an adensosine A2A receptor antagonist. 6248. The method of item 6246 wherein the agent is an AKT inhibitor.
6249. The method of item 6246 wherein the agent is an alpha 2 integrin antagonist, wherein the alpha 2 integrin antagonist is Pharmaprojects No. 5754 (Merck KgaA).
6250. The method of item 6246 wherein the agent is an alpha 4 integrin antagonist.
6251. The method of item 6246 wherein the agent is an alpha 7 nicotinic receptor agonist.
6252. The method of item 6246 wherein the agent is an angiogenesis inhibitor selected from the group consisting of AG-12,958 (Pfizer), ATN-161 (Attenuon LLC), neovastat, an angiogenesis inhibitor from Jerina AG (Germany), NM-3 (Mercian), VGA-1155 (Taisho), FCE-26644 (Pfizer), FCE-26950 (Pfizer), FPMA (Meiji Daries), FR-111142 (Fujisawa), GGTI-298, GM-1306 (Ligand), GPA-1734 (Novartis), NNC-47-0011 (Novo Nordisk), herbamycin (Nippon Kayaku), lenalidomide (Celegene), IP-10 (NIH), ABT-828 (Abbott), KIN-841 (Tokushima University, Japan), SF-1126 (Semafore Pharmaceuticals), laminin technology (NIH), CHIR-258 (Chiron), NVP-AEW541 (Novartis), NVP-AEW541 (Novartis), Vt16907 (Alchemia), OXI-8007 (Oxigene), EG-3306 (Ark Therapeutics), Maspin (Arriva), ABT- 567 (Abbott), PPI-2458 (Praecis Pharmaceuticals), CC-5079, CC-4089 (Celgene), HIF-1 alpha inhibitors (Xenova), S-247 (Pfizer), AP-23573 (Ariad), AZD-9935 (Astra Zeneca), mebendazole (Introgen Therapeutics), MetAP-2 inhibitors (GlaxoSmithKline), AG-615 (Angiogene Pharmaceuticals), Tie-2 antagonists (Hybrigenics), NC-381 , CYC-381, NC-169, NC-219, NC-383, NC-384, NC-407 (Lorus Therapeutics), ATN-224 (Attenuon), ON-01370 (Onconova), Vitronectin antagonists (Amgen), SDX-103 (Salmedix), Vitronectin antagonists (Shire), CHP (Riemser), TEK (Amgen), Anecortave acetate (Alcon), T46.2 (Matrix Therapeutics), HG-2 (Heptagen), TEM antagonists (Genzyme), Oxi-4500 (Oxigene), ATN-161 (Attenuon), WX-293 (Wilex), M-2025 (Metris Therapeutics), Alphastatin (BioActa), YH-16 (Yantai Rongchang), BIBF-1120 (Boehringer Ingelheim), BAY-57-9352 (Bayer), AS- 1404 (Cancer Research Technology), SC-77964 (Pfizer), glycomimetics (BioTie Therapies), TIE-2 Inhibitors (Ontogen), DIMI, Octamer (Octamer), ABR-215050 (Active Biotech), ABT-518 (Abbott), KDR inhibitors (Abbott), BSF-466895 (Abbott), SCH-221153 (Schering-Plough), DAC:antiangiogenic (ConjuChem), TFPI (EntreMed), AZD-2171 (Astra-Zenaca), CDC-394 (Celgene), LY290293 (EIi Lilly), IDN-5390 (Indena), Kdr Kinase Inhibitors (Merck), CT-113020, CT-116433, CT-116563, CT-31890, CT-32228) (Cell Therapeutics), A-299620 (Abbott), TWEAK Inhibitor (Amgen), VEGF modulators (Johnson and Johnson), Tum-N53, tumstatin (Genzyme), Thios- 1 , Thios-2 (Thios Pharmaceuticals), MV-6401 (Miravant Medical Technologies^ Spisulosine (PharmaMar), CEP-7055 (Cephalon), AUV-201 (Auvation), LM-609 (EIi Lilly), SKF-106615 (AnorMED), Oglufanide disodium (Cytran), BW-114 (Pharminox), Calreticulin (NIH), WX-678 (Wilex), SD-7784 (Pfizer), WX-UK1 (Wilex), SH-268 (Schering AG), 2-Me-PGA (Celgene), S- 137 (Pfizer), ZD-6126 (Angiogene Pharmaceuticals), SG-292 (SignalGen), Benefin (Lane Labs), A6, A36 (Angstrom), SB-2723005 (GlaxoSmithKline), SC-7 (Cell Therapeutics), ZEN-014 (AEterna Zentaris), 2-methoxyestradiol (EntreMed), NK-130119 (Nippon Kayaku), CC-10004 (Celgene), AVE- 8062A (Ajinomoto), Tacedinaline (Pfizer), Actinonin (Tokyo Metropolitan Institute of Medical Science), Lenalidomide (Celgene), VGA-1155, BTO-956 (SRI International), ER-68203-00 (Eisai), CT-6685 (UCB), JKC-362 (Phoenix Pharmaceuticals), DMI-3798 (DMI Biosciences, Angiomate (Ipsen), ZD-6474 (AstraZeneca), CEP-5214 (Cephalon), Canstatin (Genzyme), NM-3 (Mercian), Oridigm (MediQuest Therapeutics), Exherin (Adherex), BLS-0597 (Boston Life Sciences), PTC-299 (PTC Therapeutics), NPI-2358 (Nereus Pharmaceuticals), CGP-79787 (Novartis), AEE-788 (Novartis), CKD-732 (Chong Kun Dang), CP-564959 (OSI Pharmaceuticals), CM-101 (CarboMed), CT-2584, CT3501 (Cell Therapeutics), combretastatin and analogues and derivatives thereof (Oxigene), Rebimastat (Bristol-Meyers Squibb), Dextrin 2-sulfate (ML Laboratories), Cilengitide (Merk KGaA), NSC-706704 (Pharminox), KRN- 951 (Kirin Brewery), Ukrain, NSC-631570 (Nowicky Pharma), Tecogalan sodium (Daiichi Pharmaceutical), Tz-93 (Tsumura), TBC-1635 (Encysive Pharmaceuticals), TAN-1120 (Takeda), Semaxanib (Pfizer), BDI-7800 (Biopharmacopae), SD-186, SD-983 (Bristol-Meyers Squibb), SB-223245 (GlaxoSmithKline), SC-236 (Pfizer), RWJ-590973 (Johnson and Johnson), ILX-1850 (Genzyme), SC-68488, S-836 (Pfizer), CG-55069-11 (CuraGen), Ki-23057 (Kirin Brewery), CCX-700 (Chemoentryx), Pegaptanib octasodium (Giled Sciences), ANGIOCOL (available from Biostratum Inc.), or an analogue or derivative thereof.
6253. The method of item 6246 wherein the agent is an apoptosis antagonist.
6254. The method of item 6246 wherein the agent is an apoptosis activator.
6255. The method of item 6246 wherein the agent is a beta 1 integrin antagonist.
6256. The method of item 6246 wherein the agent is a beta tubulin inhibitor.
6257. The method of item 6246 wherein the agent is a blocker of enzyme production in Hepatitis C. 6258. The method of item 6246 wherein the agent is a Bruton's tyrosine kinase inhibitor.
6259. The method of item 6246 wherein the agent is a calcineurin inhibitor.
6260. The method of item 6246 wherein the agent is a caspase 3 inhibitor.
6261. The method of item 6246 wherein the agent is a CC chemokine receptor antagonist.
6262. The method of item 6246 wherein the agent is a cell cycle inhibitor selected from the group consisting of SNS-595 (Sunesis), synthadotin, KRX-0403, and an analogue or derivative thereof.
6263. The method of item 6246 wherein the agent is a cathepsin B inhibitor.
6264. The method of item 6246 wherein the agent is a cathepsin K inhibitor, wherein the cathepsin K inhibitor is 462795 (GlaxoSmithKline), INPL-022-D6 (Amura Therapeutics), or an analogue or derivative thereof.
6265. The method of item 6246 wherein the agent is a cathepsin L inhibitor.
6266. The method of item 6246 wherein the agent is a CD40 antagonist. 6267. The method of item 6246 wherein the agent is a chemokine receptor agonist.
6268. The method of item 6246 wherein the agent is a chymase inhibitor.
6269. The method of item 6246 wherein the agent is a collagenase antagonist.
6270. The method of item 6246 wherein the agent is a CXCR antagonist.
6271. The method of item 6246 wherein the agent is a cyclin dependent kinase inhibitor selected from the group consisting of a CDK-1 inhibitor, a CDK-2 inhibitor, a CDK- 4 inhibitor, a CDK-6 inhibitor, a CAK1 inhibitor from GPC Biotech and Bristol-Myers Squibb, RGB-286199 (GPC Biotech), an anticancer agent from Astex Technology, a CAK1 inhibitor from GPC Biotech, a CDK inhibitor from Sanofi-Aventis, a CDK1/CDK2 inhibitor from Amgen, a CDK2 inhibitor from SUGEN-2 (Pfizer), a hearing loss therapy agent (Sound Pharmaceuticals), PD-0332991 (Pfizer), RGB-286199 (GPC Biotech), Ro-0505124 (Hoffmann-La Roche), a Ser/Thr kinase inhibitor from Lilly (EIi Lilly), CVT-2584 (CAS No. 199986-75-9) (CV Therapeutics), and an analogue or derivative thereof.
6272. The method of item 6246 wherein the agent is a cyclooxygenase 1 inhibitor.
6273. The method of item 6246 wherein the agent is a DHFR inhibitor. 6274. The method of item 6246 wherein the agent is a dual integrin inhibitor.
6275. The method of item 6246 wherein the agent is an elastase inhibitor.
6276. The method of item 6246 wherein the agent is an elongation factor-1 alpha inhibitor.
6277. The method of item 6246 wherein the agent is an endothelial growth factor antagonist.
6278. The method of item 6246 wherein the agent is an endothelial growth factor receptor kinase inhibitor selected from the group consisting of sorafenib tosylate (Bayer), AAL-993 (Novartis), ABP-309 (Novartis), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), E- 7080 (Eisai), EG-3306 (Ark Therapeutics), EXEL-2880 (Exelixis), GW- 654652 (GlaxoSmithKline), a KDR inhibitor from LG Life Sciences, CT-6685 and CT-6729 (UCB), KRN-633 and KRN-951 (Kirin Brewery), OSI-930 (OSI Pharmaceuticals), SP-5.2 (Supratek Pharma), SU-11657 (Pfizer), a Tie-2 antagonist (Hybrigenics), a VEGFR-2 kinase inhibitor (Bristol-Myers Squibb), XL-647 (Exelixis), a KDR inhibitor from Abbott Laboratories, sorafenib tosylate, and an analogue or derivative thereof.
6279. The method of item 6246 wherein the agent is an endotoxin antagonist.
6280. The method of item 6246 wherein the agent is an epothilone and tubulin binder. 6281. The method of item 6246 wherein the agent is an estrogen receptor antagonist.
6282. The method of item 6246 wherein the agent is an FGF inhibitor.
6283. The method of item 6246 wherein the agent is a farnexyl transferase inhibitor.
6284. The method of item 6246 wherein the agent is farnesyltransferase inhibitor selected from the group of A-197574 (Abbott), a famesyltransferase inhibitor from Servier, FPTIII (Strathclyde Institute for Drug R), LB-42908 (LG Life Sciences), Pharmaprojects No. 5063 (Genzyme), Pharmaprojects No. 5597 (Ipsen), Yissum Project No. B-1055 (Yissum), and an analogue or derivative thereof.
6285. The method of item 6246 wherein the agent is an FLT-3 kinase inhibitor.
6286. The method of item 6246 wherein the agent is an FGF receptor kinase inhibitor.
6287. The method of item 6246 wherein the agent is a fibrinogen antagonist selected from the group consisting of AUV-201 (Auvation), MG-13926 (Sanofi-Aventis), plasminogen activator (CAS No. 105913-11-9) (from Sanofi-Aventis or UCB), plasminogen activator-2 (tPA- 2) (Sanofi-Aventis), pro-urokinase (CAS No. 82657-92-9) (Sanofi-Aventis), and an analogue or derivative thereof.
6288. The method of item 6246 wherein the agent is a heat shock protein 90 antagonist selected from the group consisting of SRN-005 (Sirenade), geldanamycin, NSC-33050 (17-allylaminogeldanamycin; 17- AAG), IT-dimethylaminoethylamino-IT-demethoxy-geldanamycin (17- DMAG), rifabutin (rifamycin XIV, 1',4-didehydro-1-deoxy-1 ,4-dihydro-5l-(2- methylpropyl)-1-oxo-), and an analogue or derivative thereof.
6289. The method of item 6246 wherein the agent is a histone deacetylase inhibitor.
6290. The method of item 6246 wherein the agent is an HMGCoA reductase inhibitor selected from the group consisting of an atherosclerosis therapeutic from Lipid Sciences, ATI-16000 (ARYx Therapeutics), KS-01-019 (Kos Pharmaceuticals), Pharmaprojects No. 2197 (Sanofi-Aventi), RP 61969 (Sanofi-Aventis), and an analogue or derivative thereof.
6291. The method of item 6246 wherein the agent is an ICAM inhibitor.
6292. The method of item 6246 wherein the agent is an IL, ICE and IRAK antagonist, wherein the antagonist is a CJ-14877, CP-424174 (Pfizer), NF-61 (Negma-Lerads), and an analogue or derivative thereof.
6293. The method of item 6246 wherein the agent is an IL-2 inhibitor.
6294. The method of item 6246 wherein the agent is an immunosuppressant selected from the group consisting of teriflunomide (Sanofi Aventis), chlorsulfaquinoxalone (NSC-339004), chlorsulfaquinoxalone sulfate, CS-712 (Sankyo), ismomultin alfa (CAS No. 457913-93-8) (Akzo Nobel), antiallergics from GenPat77, antiinflammatories or AT-005 (Androclus Therapeutics), autoimmune disease therapeutics from EpiVax, BN-007 (Bone), budesonide (CAS No. 51333-22- 3) (MAP Pharmaceuticals), CO-14 (Genzyme), edratide (CAS No. 433922- 67-9) (Teva), EP-314 (Enanta), eprovafen (CAS No. 101335-99-3) (Sanofi- Aventis), HWA-131 (CAS No. 118788-41-3) (Sanofi-Aventis), immunomodulators from MerLion Pharmaceuticals, immunosuppressives from Alchemia, IPL-12 (Inflazyme), MDL-9563 (CAS No. 27086-86-8) (Sanofi-Aventis), Pharmaprojects No. 2330 (Sanofi-Aventis), Pharmaprojects No. 6426 (Abgenix), PXS-25 (Pharmaxis), rosmarinic acid (CAS No. 20283-92-5) (Sanofi-Aventis), RP 42927 or RP 54745 (CAS No. 135330-08-4) (Sanofi-Aventis), SGN-35 (Seattle Genetics), ST-1959 (Sigma-Tau), type I diabetes therapy from SYNX Pharma, UNIL-88 (Debiopharm), VP-025 (Vasogen), VR-694 (Vectura), PRTX-001 (Protalex), and an analogue or derivative thereof.
6295. The method of item 6246 wherein the agent is an IMPDH (inosine monophosphate).
6296. The method of item 6246 wherein the agent is an integrin antagonist.
6297. The method of item 6246 wherein the agent is an interleukin antagonist.
6298. The method of item 6246 wherein the agent is an inhibitor of type III receptor tyrosine kinase.
6299. The method of item 6246 wherein the agent is an irreversible inhibitor of enzyme methionine aminopeptidase type 2.
6300. The method of item 6246 wherein the agent is an isozyme selective delta protein kinase C inhibitor. 6301. The method of item 6246 wherein the agent a JAK3 enzyme inhibitor.
6302. The method of item 6246 wherein the agent is a JNK inhibitor.
6303. The method of item 6246 wherein the agent is a kinase inhibitor.
6304. The method of item 6246 wherein the agent is kinesin antagonist.
6305. The method of item 6246 wherein the agent is a kinesin antagonist.
6306. The method of item 6246 wherein the agent is a leukotriene inhibitor and antagonist selected from the group consisting of ambicromil (CAS No. 58805-38-2) (Sanofi-Aventis), amelubant (CAS No. 346735-24-8) (Boehringer Ingelheim), DW-1141 (Dong Wha), ebselen (Daiichi Pharmaceutical), ibudilast (Kyorin), leucotriene inhibitors from Sanofi-Aventis, lymphotoxin-beta receptor (LT-D) from Biogen Idee, Pharmaprojects No. 1535 and 2728 (CAS No. 119340-33-9) (Sanofi- Aventis), R-112 (Rigel), Rev-5367 (CAS No. 92532-05-3) (Sanofi-Aventis), RG-14893 (CAS No. 141835-49-6) (Sanofi-Aventis), RG-5901-A (CAS No. 101910-24-1), 92532-23-5, RP 66153 (CAS No. 142422-79-5), RP 66364 (CAS No.186916-92-5), RP 69698 (CAS No. 141748-00-7) (Sanofi-Aventis), SC-411930 (Pfizer), SC-41930 (CAS No. 120072-59-5) (Pfizer), SC-50605 (CAS No. 138828-39-4) (Pfizer), SC-51146 (CAS No. 141059-52-1), SC- 53228 (CAS No. 153633-01-3) (Pfizer), spaglumic acid (ZY-15106) (CAS No.3106-85-2), 80619-64-3 (Novartis), tipredane (CAS No. 85197-77-9) (Bristol-Myers Squibb), U-75302 (CAS No. 119477-85-9) (Pfizer), and analogue or derivative thereof.
6307. The method of item 6246 wherein the agent is an MAP kinase inhibitor.
6308. The method of item 6246 wherein the agent is a matrix metalloproteinase inhibitor.
6309. The method of item 6246 wherein the agent is an MCP- CCR2 inhibitor.
6310. The method of item 6246 wherein the agent is an mTOR inhibitor.
6311. The method of item 6246 wherein the agent is an mTOR kinase inhibitor.
6312. The method of item 6246 wherein the agent is a microtubule inhibitor selected from the group consisting of antibody- maytansinoid conjugates from Biogen Idee, colchicines (MantiCore Pharmaceuticals), anticancer immunoconjugates from Johnson & Johnson, DIME from Octamer, gni-1f (GNI), huC242-DM4, huMy9-6-DM1 (ImmunoGen), IDN-5404 (Indena), IMO-098, IMOderm (Imotep), mebendazole (Introgen Therapeutics), microtubule poisons from Cambridge Enterprise, paclitaxel such as LOTAX from Aphios (CAS No. 33069-62-4), Genexol-PM from Samyang, Pharmaprojects No. 6383 (Tapestry Pharmaceuticals), RPR-112378 (Sanofi-Aventis), SGN-75 (Seattle Genetics), SPL-7435 (Starpharma), SSR-250411 (Sanofi-Aventis), trastuzumab-DM1 (Genentech), vinorelbine, and an analogue or derivative thereof. 6313. The method of item 6246 wherein the agent is an MIF inhibitor.
6314. The method of item 6246 wherein the agent is an MMP inhibitor.
6315. The method of item 6246 wherein the agent is a neurokinin (NK) antagonist selected from the group consisting of anthrotainin (CAS No. 148084-40-6) (Sanofi-Aventis), an IBS thereapeutic from ArQuIe, MDL-105212A (CAS No. 167261-60-1) (Ssanofi-Aventis), Pharmaprojects No. 2744, 3258 (CAS No. 139167-47-8) 4006, 4201, or 5986 (Sanofi-Aventis), RP 67580 (CAS No. 135911-02-3), SR-144190 (CAS No. 201152-86-5), SSR-240600, SSR-241586 (Sanofi-Aventis), TKA-457 (Novartis), vestipitant mesylate (CAS No. 334476-64-1) (GlaxoSmithKline), Win-64821 (Sanofi-Aventis), PRX-96026 (Predix Pharmaceuticals), and an analogue or derivative thereof.
6316. The method of item 6246 wherein the agent is an NF kappa B inhibitor selected from the group consisting of AVE-0545 or AVE- 0547 (Sanofi-Aventis), bortezomib (CAS No. 179324-69-7) (Millennium Pharmaceuticals), dexanabinol (CAS No. 112924-45-5) (Pharmos), dexlipotam (Viatris), Pharmaprojects No. 6283 (INDRA) (OXiGENE), IPL- 576092 (CAS No. 137571-30-3) (Inflazyme), NFKB decoy (Corgentech), NFKB decoy oligo (AnGes MG), NFKB's from Ariad, osteoporosis treatments or S5 (F005) from Fulcrum Pharmaceuticals, P61 (Phytopharm), R-flurbiprofen (CAS No. 5104-49-4) (Encore Pharmaceuticals), and an analogue or derivative thereof.
6317. The method of item 6246 wherein the agent is a nitric oxide agonist. 6318. The method of item 6246 wherein the agent is an ornithine decarboxylase inhibitor.
6319. The method of item 6246 wherein the agent is a p38 MAP kinase inhibitor selected from the group consisting of AZD-6703 (AstraZeneca), JX-401 (Jexys Pharmaceuticals), BMS-2 (Bristol-Myers Squibb), a p38 MAP kinase inhibitor from Novartis, a p38-alpha MAP kinase inhibitor from Amphora, Pharmaprojects No. 5704 (Pharmacopeia), RPR- 200765A (Sanofi-Aventis), SD-282 (Johnson & Johnson), TAK-715 (Takeda), and an analogue or derivative thereof.
6320. The method of item 6246 wherein the agent is a palmitoyl-protein thioesterase inhibitor.
6321. The method of item 6246 wherein the agent is a PDGF receptor kinase inhibitor selected from the group consisting of AAL-993, AMN-107, or ABP-309 (Novartis), AMG-706 (Amgen), BAY-57-9352 (Bayer), CDP-860 (UCB), E-7080 (Eisai), imatinib (CAS No. 152459-95-5) (Novartis), OSI-930 (OSI Pharmaceuticals), RPR-127963E (Sanofi-Aventis), RWJ-540973 (Johnson & Johnson), sorafenib tosylate (Bayer), SU-11657 (Pfizer), tandutinib (CAS No. 387867-13-2) (Millennium Pharmaceuticals), vatalanib (Novartis), ZK-CDK (Schering AG), and an analogue or derivative thereof.
6322. The method of item 6246 wherein the agent is (-)- halofenate (Metabolex), AMG-131 (Amgen), antidiabetics from Japan Tobacco, AZD-4619, AZD-8450, AZD-8677 (AstraZeneca), DRF-10945, balaglitazone (Dr Reddy's), CS-00088, CS-00098 (Chipscreen Biosciences), E-3030 (Eisai), etalocib (CAS No. 161172-51-6) (EIi Lilly), GSK-641597 (Ligand), GSK-677954 (GlaxoSmithKline), GW-409544 (Ligand), GW- 590735 (GlaxoSmithKline), K-111 (Hoffmann-La Roche), LY-518674 (EIi Lilly), LY-674 (Ligand), LY-929 (Ligand), MC-3001 , MC-3002 (MaxoCore Pharmaceuticals), metformin HCI + pioglitazone (CAS No. 1115-70-4 and 112529-15-4), ACTOPLUS MET from Andrx), muraglitazar (CAS No. 331741-94-7) (Bristol-Myers Squibb), naveglitazar (Ligand), oleoylethanolamide (Kadmus Pharmaceuticals), ONO-5129, pioglitazone hydrochloride (CAS No. 111025-46-8 and 112529-15-4) (Takeda), PLX-204 (Plexxikon), PPAR agonists from Genfit, PPAR delta agonists from EIi Lilly, PPAR-alpha agonists from CrystalGenomics, PPAR-gamma modulators and PPAR-β modulators from CareX, rosiglitazone maleate (CAS No. 122324- 73-4 or 155141-29-0) (GlaxoSmithKline), rosiglitazone maleate/glimepir (CAS No. 155141-29-0 and 93479-97-1), AVANDARYL1 rosiglitazone maleate/metformin extend (CAS No. 155141-29-0 and 657-24-9), AVANDAMET, rosiglitazone maleate+metformin, AVANDAMET (GlaxoSmithKline), tesaglitazar (AstraZeneca), and an analogue or derivative thereof.
6323. The method of item 6246 wherein the agent is a phosphatase inhibitor.
6324. The method, of item 6246 wherein the agent is a phosphodiesterase (PDE) inhibitor selected from the group consisting of avanafil (Tanabe Seiyaku), dasantafil (CAS No. 569351-91-3) (Schering- Plough), A-906119 (CAS No. 134072-58-5), DL-850 (Sanofi-Aventis), GRC- 3015, GRC-3566, GRC-3886 (Glenmark), HWA-153 (CAS No. 56395-66-5) (Sanofi-Aventis), hydroxypumafentrine (Altana), IBFB-130011 , 1BFB-14-016, IBFB-140301 , IBFB-150007, IBFB-211913 (IBFB Pharma), L-826141 (Merck & Co), medorinone (CAS No. 88296-61-1) (Sanofi-Aventis), MEM- 1917 (Memory Pharmaceuticals), ND-1251 (Neuro3d), PDE inhibitors from ICOS, PDE IV inhibitors from Memory Pharmaceuticals and CrystalGenomics, Pharmaprojects No. 2742 and 6141 (Sanofi-Aventis), QAD-171 (Novartis), RHC-2963 (CAS No. 76993-12-9 and 76993-14-1), RPR-117658, RPR-122818 derivatives, SR-24870 , and RPR-132294 (Sanofi-Aventis), SK-350 (ln2Gen), stroke therapy agents from deCODE Genetics, TAS-203 (Taiho), tofimilast (CAS No. 185954-27-2) (Pfizer), UK- 371800 (Pfizer), WIN-65579 (CAS No. 158020-82-7) (Sanofi-Aventis), IBFB- 130020 (IBFB Pharma), OPC-6535 (CAS No. 145739-56-6) (Otsuka), a phosphodiesterase III inhibitor, enoximone, a phosphodiesterase IV inhibitor, fosfosal, Atopik (Barrier Therapeutics), triflusal, a phosphodiesterase V inhibitor, and an analogue or derivative thereof.
6325. The method of item 6246 wherein the agent is a PKC inhibitor.
6326. The method of item 6246 wherein the agent is a platelet activating factor antagonist.
6327. The method of item 6246 wherein the agent is a platelet-derived growth factor receptor kinase inhibitor.
6328. The method of item 6246 wherein the agent is a prolyl hydroxylase inhibitor.
6329. The method of item 6246 wherein the agent is a polymorphonuclear neutrophil inhibitor.
6330. The method of item 6246 wherein the agent is a protein kinase B inhibitor.
6331. The method of item 6246 wherein the agent is a protein kinase C stimulant. 6332. The method of item 6246 wherein the agent is a purine nucleoside analogue.
6333. The method of item 6246 wherein the agent is a purinoreceptor P2X antagonist.
6334. The method of item 6246 wherein the agent is a Raf kinase inhibitor.
6335. The method of item 6246 wherein the agent is a reversible inhibitor of ErbB1 and ErbB2.
6336. The method of item 6246 wherein the agent is a ribonucleoside triphosphate reductase inhibitor.
6337. The method of item 6246 wherein the agent is an SDF- 1 antagonist.
6338. The method of item 6246 wherein the agent is a sheddase inhibitor.
6339. The method of item 6246 wherein the agent is an SRC inhibitor.
6340. The method of item 6246 wherein the agent is a stromelysin inhibitor.
6341. The method of item 6246 wherein the agent is an Syk kinase inhibitor. 6342. The method of item 6246 wherein the agent is a telomerase inhibitor.
6343. The method of item 6246 wherein the agent is a TGF beta inhibitor selected from the group consisting of pirfenidone (CAS No. 53179-13-8) (MARNAC)1 tranilast (CAS No. 53902-12-8) (Kissei), IN-1130 (ln2Gen), mannose-6-phosphate (BTG), TGF-β antagonists from Inflazyme (Pharmaprojects No. 6075), TGF-β antagonists from Sydney, non-industrial source), TGF-β I receptor kinase inhibitors from EIi Lilly, TGF-β receptor inhibitors from Johnson & Johnson, and an analogue or derivative thereof.
6344. The method of item 6246 wherein the agent is a TN Fa antagonist or TACE inhibitor selected from the group consisting of adalimumab (CAS No. 331731-18-1) (Cambridge Antibody Technology), AGIX-4207 (AtheroGenics), AGT-1 (Advanced Biotherapy), an antiinflammatory from Borean Pharma, Cellzome, or Paradigm Therapeutics, anti-inflammatory vaccine (TNF-alpha kinoid) from Neovacs, humanized anti-TNF antibody or an anti-TNF MAb (CB0006) Celltech (UCB), apratastat (CAS No. 287445-51-0) (Wyeth), BMS-561392 (Bristol-Myers Squibb), BN- 006 (Bone), certolizumab pegol (CAS No. 428863-50-7 or CH-138 (UCB), cilomilast (CAS No. 153259-65-5) (GlaxoSmithKline), CR-1 (Nuada Pharmaceuticals), CRx-119 (CombinatoRx), D-5410 (UCB), dacopafant (CAS No. 125372-33-0) (Sanofi-Aventis), dersalazine (CAS No. 188913-57- 7/188913-58-8) (Uriach), etanercept (CAS No. 185243-69-0) (Amgen), ethyl pyruvate (Critical (Critical Therapeutics), golimumab (CAS No. 476181-74-5) (Johnson & Johnson), hormono-immunotherapy from Ipsen, CDP571 (e.g., Humicade from UCB), IC-485 (ICOS), infliximab (CAS No. 170277-31-3) (Johnson & Johnson), IP-751 (Manhattan Pharmaceuticals), ISIS-104838 (CAS No. 250755-32-9) (ISIS Pharmaceuticals), lenalidomide (CAS No. 191732-72-6) (Celgene), lentinan (CAS No. 37339-90-5) (Ajinomoto), MDL- 201112 (CAS No. 142130-73-2) (Sanofi-Aventis), medroxyprogesterone (CAS No. 520-85-4) (InKine Pharmaceutical), N-acetylcysteine (CAS No. 616-91-1) (Zambon), NBE-P2 (DIREVO Biotech), nerelimomab (CAS No. 162774-06-3) (Chiron), OM-294DP (OM PHARMA)1 onercept (CAS No. 199685-57-9) (Yeda), PASSTNF-alpha (Verigen), pentoxifylline or oxypentifylline (Sanofi-Aventis), Pharmaprojects No. 4091 , 4241 , 4295, or 4488 (Sanofi-Aventis), Pharmaprojects No. 5480 (Amgen), Pharmaprojects No. 6749 (Cengent), pirfenidone (CAS No. 53179-13-8) (MARNAC), RPR- 132294 (Sanofi-Aventis), S5 (F002) (Fulcrum Pharmaceuticals), simvastatin (CAS No. 79902-63-9) (Merck & Co), STA-6292 (Synta Pharmaceuticals), tacrolimus (CAS No. 104987-11-3) (Fujisawa LifeCycle Pharma), talactoferrin alfa (CAS No. 308240-58-6) (Agennix), thalidomide (CAS No. 50-35-1) (Celgene), TNF antagonists form ProStrakan, and Synergen, TNF inhibitors (Amgen), TNF-alpha antagonists from Dynavax Technologies and Jerina AG (Germany), TNF-alpha inhibitors from IBFB Pharma and Xencor (Xencor), torbafylline (CAS No. 105102-21-4) (Sanofi-Aventis), UR-1505 (Uriach), VT-346 (Won Therapeutics), YSIL6 (Y's Therapeutics), YSTH2 (Y's Therapeutics), NPI-1302a-3 (Nereus Pharmaceuticals, a TNF antagonist from Jerina AG (Germany), dersalazine, and an analogue or derivative thereof.
6345. The method of item 6246 wherein the agent is a Toll receptor inhibitor.
6346. The method of item 6246 wherein the agent is a tubulin antagonist.
6347. The method of item 6246 wherein the agent is a tyrosine kinase inhibitor selected from the group consisting of SU-011248, SUTENT from Pfizer Inc. (New York, NY), BMS-354825; PN-355 (Paracelsian Pharmaceuticals), AGN-199659 (Allergan), AAL-993 or ABP- 309 (Novartis), adaphostin (NIH), AEE-788 (Novartis), AG-013736 (OSI Pharmaceuticals), AG-13736 (Pfizer), ALT-110 (Alteris Therapeutics), AMG- 706 (Amgen), anticancer MAbs from Xencor, anti-EGFrvlll MAbs from Abgenix, anti-HER2 MAb from Abiogen, AZD-2171 or AZD-9935 (AstraZeneca), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), CEP-5214 (Cephalon), CEP-7055 (Cephalon), cetuximab (ImCIone Systems), CHIR-200131 and CHIR-258 (Chiron), CP-547632 (OSI Pharmaceuticals), CP-724714 (Pfizer), CT-301 (Creabilis Therapeutics), D- 69491 (Baxter International), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EGFR/ErbB2 inhibitors from Array BioPharma, erlotinib (CAS No. 183319- 69-9) (OSI Pharmaceuticals), EXEL-2880 (Exelixis), FK-778 (Sanofi- Aventis) , gefitinib (CAS No. 184475-35-2) (AstraZeneca), GW-2286 or GW- 654652 (GlaxoSmithKline), her2/neu antigen from AlphaVax, HER-2/neu inhibitor from Generex, Herzyme (Medipad) (Sirna Therapeutics), HKI-272 (Wyeth), HuMax-EGFr (Genmab), idronoxil (CAS No. 81267-65-4) (Novogen), IGF-1 inhibitors from Ontogen, IMC-11F8 (ImCIone Systems), kahalalide F (CAS No. 149204-42-2) (PharmaMar), KDR inhibitor from LG Life Sciences, KDR inhibitors from Abbott Laboratories, KDR kinase inhibitors (UCB), Kdr kinase inhibitors from Merck & Co, KRN-633 and KRN- 951 (Kirin Brewery), KSB-102 (Xenova), lapatinib ditosylate (CAS No. 388082-78-8) (GlaxoSmithKline), matuzumab (Merck KGaA), MDX-214 (Medarex), ME-103 (Pharmexa), MED-A300 (Gerolymatos), MNAC-13 (Lay Line Genomics), nimotuzumab (Center of Molecular Immunology), NSC- 330507 or NSC-707545 (NIH), NV-50 (Novogen), OSI-930 (OSI Pharmaceuticals), panitumumab (Abgenix), pelitinib (CAS No. 287933-82-7) (Wyeth), pertuzumab (CAS No. 380610-27-5) (Genentech), Pharmaprojects No. 3985 (Sanofi-Aventis), prostate cancer therapeutics from Sequenom (SQPC35, SQPC36, SQPC90), removab and remoxab (Trion Pharma), RG- 13022 (CAS No. 136831-48-6), RG-13291 (CAS No. 138989-50-1), or RG- 14620 (CAS No. 136831-49-7) (Sanofi-Aventis), RM-6427 (Romark), RNAi breast cancer therapy from Benitec, RP 53801 (CAS No. 125882-88-4) (Sanofi-Aventis), sorafenib tosylate (Bayer), SU-11657 (Pfizer), Tie-2 antagonists from Semaia (Hybrigenics), Tie-2 inhibitors from Ontogen, trastuzumab (CAS No. 180288-69-1) (Genentech), tyrosine kinase inhibitors from Sanofi-Aventis, U3-1287, U3-1565, U3-1784, U3-1800 (U3 Pharma), vatalanib (Novartis), VEGFR-2 kinase inhibitor from Bristol-Myers Squibb, XL-647 (Exelixis), ZD-6474 (AstraZeneca), ZK-CDK (Schering AG), an EGFR tyrosine kinase inhibitor, EKB-569 (Wyeth), and an analogue or derivative thereof.
6348. The method of item 6246 wherein the agent is a VEGF inhibitor.
6349. The method of item 6246 wherein the agent is a vitamin D receptor agonist.
6350. The method of item 6246 wherein the agent is ZD-6474 (an angiogenesis inhibitor).
6351. The method of item 6246 wherein the agent is AP- 23573 (an mTOR inhibitor).
6352. The method of item 6246 wherein the agent is synthadotin (a tubulin antagonist).
6353. The method of item 6246 wherein the agent is S-0885 (a collagenase inhibitor).
6354. The method of item 6246 wherein the agent is aplidine (an elongation factor-1 alpha inhibitor).
6355. The method of item 6246 wherein the agent is ixabepilone (an epithilone). 6356. The method of item 6246 wherein the agent is IDN- 5390 (an angiogenesis inhibitor).
6357. The method of item 6246 wherein the agent is SB- 2723005 (an angiogenesis inhibitor).
6358. The method of item 6246 wherein the agent is ABT-518 (an angiogenesis inhibitor).
6359. The method of item 6246 wherein the agent is combretastatin (an angiogenesis inhibitor).
6360. The method of item 6246 wherein the agent is anecortave acetate (an angiogenesis inhibitor).
6361. The method of itemj3246 wherein the agent is SB- 715992 (a kinesin antagonist).
6362. The method of item 6246 wherein the agent is temsirolimus (an mTOR inhibitor).
6363. The method of item 6246 wherein the agent is adalimumab (a TNFα antagonist).
6364. The method of item 6246, wherein the composition comprises a polymer.
6365. The method of item 6246, wherein the composition comprises a polymer, and the polymer is, or comprises, a copolymer. 6366. The method of item 6246, wherein the composition comprises a polymer, and the polymer is, or comprises, a block copolymer.
6367. The method of item 6246, wherein the composition comprises a polymer, and the polymer is, or comprises, a random copolymer.
6368. The method of item 6246, wherein the composition comprises a polymer, and the polymer is, or comprises, a biodegradable polymer.
6369. The method of item 6246, wherein the composition comprises a polymer, and the polymer is, or comprises, a nonbiodegradable polymer.
6370. The method of item 6246, wherein the composition comprises a polymer, and the polymer is, or comprises, a hydrophilic polymer.
6371. The method of item 6246, wherein the composition comprises a polymer, and the polymer is, or comprises, a hydrophobic polymer.
6372. The method of item 6246, wherein the composition comprises a polymer, and the polymer is, or comprises, a polymer having hydrophilic domains.
6373. The method of item 6246, wherein the composition comprises a polymer, and the polymer is, or comprises, a polymer having hydrophobic domains. 6374. The method of item 6246, wherein the composition comprises a polymer, and the polymer is, or comprises, a non-conductive polymer.
6375. The method of item 6246, wherein the composition comprises a polymer, and the polymer is, or comprises, an elastomer.
6376. The method of item 6246, wherein the composition comprises a polymer, and the polymer is, or comprises, a hydrogel.
6377. The method of item 6246, wherein the composition comprises a polymer, and the polymer is, or comprises, a silicone polymer.
6378. The method of item 6246, wherein the composition comprises a polymer, and the polymer is, or comprises, a hydrocarbon polymer.
6379. The method of item 6246, wherein the composition comprises a polymer, and the polymer is, or comprises, a styrene-derived polymer.
6380. The method of item 6246, wherein the composition comprises a polymer, and the polymer is, or comprises, a butadiene-derived polymer.
6381. The method of item 6246, wherein the composition comprises a polymer, and the polymer is, or comprises, a macromer.
6382. The method of item 6246, wherein the composition comprises a polymer, and the polymer is, or comprises, a poly(ethylene glycol) polymer. 6383. The method of item 6246, wherein the composition comprises a polymer, and the polymer is, or comprises, an amorphous polymer.
6384. The method of item 6246, wherein the composition further comprises a second pharmaceutically active agent.
6385. The method of item 6246, wherein the composition further comprises an anti-inflammatory agent.
6386. The method of item 6246, wherein the composition further comprises an agent that inhibits infection.
6387. The method of item 6246, wherein the composition further comprises an anthracycline.
6388. The method of item 6246, wherein the composition further comprises doxorubicin.
6389. The method of item 6246 wherein the composition further comprises mitoxantrone.
6390. The method of item 6246 wherein the composition further comprises a fluoropyrimidine.
6391. The method of item 6246, wherein the composition further comprises 5-fluorouracil (5-FU).
6392. The method of item 6246, wherein the composition further comprises a folic acid antagonist. 6393. The method of item 6246, wherein the composition further comprises methotrexate.
6394. The method of item 6246, wherein the composition further comprises a podophylotoxin.
6395. The method of item 6246, wherein the composition further comprises etoposide.
6396. The method of item 6246, wherein the composition further comprises camptothecin.
6397. The method of item 6246, wherein the composition further comprises a hydroxyurea.
6398. The method of item 6246,_wherein the composition further comprises a platinum complex.
6399. The method of item 6246, wherein the composition further comprises cisplatin.
6400. The method of item 6246 wherein the composition further comprises an antithrombotic agent.
6401. The method of item 6246, wherein the composition further comprises a visualization agent.
6402. The method of item 6246, wherein the composition further comprises a visualization agent, and the visualization agent is a radiopaque material, wherein the radiopaque material comprises a metal, a halogenated compound, or a barium containing compound. 6403. The method of item 6246, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, barium, tantalum, or technetium.
6404. The method of item 6246, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, an MRI responsive material.
6405. The method of item 6246, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, a gadolinium chelate.
6406. The method of item 6246, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, iron, magnesium, manganese, copper, or chromium.
6407. The method of item 6246, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, iron oxide compound.
6408. The method of item 6246, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, a dye, pigment, or colorant.
6409. The method of item 6246 wherein the agent is released in effective concentrations from the composition comprising the agent by diffusion over a period ranging from the time of administration to about 90 days.
6410. The method of item 6246 wherein the agent is released in effective concentrations from the composition comprising the agent by erosion of the composition over a period ranging from the time of administration to about 90 days.
6411. The method of item 6246 wherein the composition further comprises an inflammatory cytokine.
6412. The method of item 6246 wherein the composition further comprises an agent that stimulates cell proliferation.
6413. The method of item 6246 wherein the composition further comprises a polymeric carrier.
6414. The method of item 6246 wherein the composition is in the form of a gel, paste, or spray.
6415. The method of item 6246 wherein the implant is partially constructed with the agent or the composition.
6416. The method of item 6246 wherein the implant is fully constructed with the agent or the composition.
6417. The method of item 6246 wherein the implant is impregnated with the agent or the composition.
6418. The method of item 6246, wherein the agent or the composition forms a coating, and the coating directly contacts the implant.
6419. The method of item 6246, wherein the agent or the composition forms a coating, and the coating indirectly contacts the implant. 6420. The method of item 6246 wherein the agent or the composition forms a coating, and the coating partially covers the implant.
6421. The method of item 6246, wherein the agent or the composition forms a coating, and the coating completely covers the implant.
6422. The method of item 6246 wherein the agent or the composition is located within pores or holes of the implant.
6423. The method of item 6246 wherein the agent or the composition is located within a channel, lumen, or divet of the implant.
6424. The method of item 6246 wherein the implant further comprising an echogenic material.
- - — 6425. _ The method of item 6246 wherein the implant further comprises an echogenic material, wherein the echogenic material is in the form of a coating.
6426. The method of item 6246 wherein the implant is sterile.
6427. The method of item 6246 wherein the agent is delivered from the implant, wherein the agent is released into tissue in the vicinity of the implant after deployment of the implant.
6428. The method of item 6246 wherein the agent is delivered from the implant, wherein the agent is released into tissue in the vicinity of the implant after deployment of the implant, wherein the tissue is connective tissue. 6429. The method of item 6246 wherein the agent is delivered from the implant, wherein the agent is released into tissue in the vicinity of the implant after deployment of the implant, wherein the tissue is muscle tissue.
6430. The method of item 6246 wherein the agent is delivered from the implant, wherein the agent is released into tissue in the vicinity of the implant after deployment of the implant, wherein the tissue is nerve tissue.
6431. The method of item 6246 wherein the agent is delivered from the implant, wherein the agent is released into tissue in the vicinity of the implant after deployment of the implant, wherein the tissue is epithelium tissue.
6432. The method of item 6246 wherein the agent is delivered from the implant, wherein the agent is released in effective concentrations from the implant over a period ranging from the time of deployment of the implant to about 1 year.
6433. The method of item 6246 wherein the agent is delivered from the implant, wherein the agent is released in effective concentrations from the implant over a period ranging from about 1 month to 6 months.
6434. The method of item 6246 wherein the agent is delivered from the implant, wherein the agent is released in effective concentrations from the implant over a period ranging from about 1 - 90 days.
6435. The method of item 6246 wherein the agent is delivered from the implant, wherein the agent is released in effective concentrations from the implant at a constant rate. 6436. The method of item 6246 wherein the agent is delivered from the implant, wherein the agent is released in effective concentrations from the implant at an increasing rate.
6437. The method of item 6246 wherein the agent is delivered from the implant, wherein the agent is released in effective concentrations from the implant at a decreasing rate.
6438. The method of item 6246 wherein the agent is delivered from the implant, wherein the implant comprises about 0.01 Dg to about 10 Dg of the agent.
6439. The method of item 6246 wherein the agent is delivered from the implant, wherein the implant comprises about 10 Dg to about 10 mg of the agent.
6440. The method of item 6246 wherein the agent is delivered from the implant, wherein the implant comprises about 10 mg to about 250 mg of the agent.
6441. The method of item 6246 wherein the agent is delivered from the implant, wherein the implant comprises about 250 mg to about 1000 mg of the agent.
6442. The method of item 6246 wherein the agent is delivered from the implant, wherein the implant comprises about 1000 mg to about 2500 mg of the agent.
6443. The method of item 6246 wherein the agent is delivered from the implant, wherein a surface of the implant comprises less than 0.01 Dg of the agent per mm2 of implant surface to which the agent is applied. 6444. The method of item 6246 wherein the agent is delivered from the implant, wherein a surface of the implant comprises about 0.01 Dg to about 1 Dg of the agent per mm2 of implant surface to which the agent is applied.
6445. The method of item 6246 wherein the agent is delivered from the implant, wherein a surface of the implant comprises about 1 Dg to about 10 Dg of the agent per mm2 of implant surface to which the agent is applied.
6446. The method of item 6246 wherein the agent is delivered from the implant, wherein a surface of the implant comprises about 10 Dg to about 250 Dg of the agent per mm2 of implant surface to which the agent is applied.
6447. The method of item 6246 wherein the agent is delivered from the implant, wherein a surface of the implant comprises about 250 Dg to about 1000 Dg of the agent per mm2 of implant surface to which the agent is applied.
6448. The method of item 6246 wherein the agent is delivered from the implant, wherein a surface of the implant comprises about 1000 Dg to about 2500 Dg of the agent per mm2 of implant surface to which the agent is applied.
6449. The method of item 6246, wherein the implant further comprises a coating, and the coating is a uniform coating.
6450. The method of item 6246, wherein the implant further comprises a coating, and the coating is a non-uniform coating. 6451. The method of item 6246, wherein the implant further comprises a coating, and the coating is a discontinuous coating.
6452. The method of item 6246, wherein the implant further comprises a coating, and the coating is a patterned coating.
6453. The method of item 6246, wherein the implant further comprises a coating, and the coating has a thickness of 100 Dm or less.
6454. The method of item 6246, wherein the implant further comprises a coating, and the coating has a thickness of 10 Dm or less.
6455. The method of item 6246, wherein the implant further comprises a coating, and the coating adheres to the surface of the implant upon deployment of the implant.
6456. The method of item 6246, wherein the implant further comprises a coating, and the coating is stable at room temperature for a period of at least 1 year.
6457. The method of item 6246, wherein the implant further comprises a coating, and the agent is present in the coating in an amount ranging between about 0.0001% to about 1% by weight.
6458. The method of item 6246, wherein the implant further comprises a coating, and the agent is present in the coating in an amount ranging between about 1 % to about 10% by weight.
6459. The method of item 6246, wherein the implant further comprises a coating, and the agent is present in the coating in an amount ranging between about 10% to about 25% by weight. 6460. The method of item 6246, wherein the implant further comprises a coating, and the agent is present in the coating in an amount ranging between about 25% to about 70% by weight.
6461. The method of item 6246, wherein the implant further comprises a coating, and the coating comprises a polymer.
6462. The method of item 6246, wherein the implant comprises a first coating having a first composition and a second coating having a second composition.
6463. The method of item 6246, wherein the implant comprises a first coating having a first composition and a second coating having a second composition, wherein the first composition and the second composition are different.
6464. A method as in any one of items 6246-6463, wherein the device is a tracheal stent.
6465. A method as in any one of items 6246-6463, wherein the device is a bronchial stent.
6466. A method as in any one of items 6246-6463, wherein the device is a metallic tracheal stent.
6467. A method as in any one of items 6246-6463, wherein the device is a metallic bronchial stent.
6468. A method as in any one of items 6246-6463, wherein the device is a polymeric tracheal stent.
6469. A method as in any one of items 6246-6463, wherein the device is a polymeric bronchial stent. 6470. A method for inhibiting scarring comprising placing a device that comprises a genital urinary stent (e.g., an implant) and an anti- scarring agent or a composition comprising an anti-scarring agent into an animal host, wherein the agent inhibits scarring.
6471. The method of item 6470 wherein the agent is an adensosine A2A receptor antagonist.
6472. The method of item 6470 wherein the agent is an AKT inhibitor.
6473. The method of item 6470 wherein the agent is an alpha 2 integrin antagonist, wherein the alpha 2 integrin antagonist is Pharmaprojects No. 5754 (Merck KgaA).
6474. The method of item 6470 wherein the agent is an alpha 4 integrin antagonist.
6475. The method of item 6470 wherein the agent is an alpha 7 nicotinic receptor agonist.
6476. The method of item 6470 wherein the agent is an angiogenesis inhibitor selected from the group consisting of AG-12,958 (Pfizer), ATN-161 (Attenuon LLC), neovastat, an angiogenesis inhibitor from Jerina AG (Germany), NM-3 (Mercian), VGA-1155 (Taisho), FCE-26644 (Pfizer), FCE-26950 (Pfizer), FPMA (Meiji Daries), FFM 11142 (Fujisawa), GGTI-298, GM-1306 (Ligand), GPA-1734 (Novartis), NNC-47-0011 (Novo Nordisk), herbamycin (Nippon Kayaku), lenalidomide (Celegene), IP-10 (NIH), ABT-828 (Abbott), KIN-841 (Tokushima University, Japan), SF-1126 (Semafore Pharmaceuticals), laminin technology (NIH), CHIR-258 (Chiron), NVP-AEW541 (Novartis), NVP-AEW541 (Novartis), Vt16907 (Alchemia), OXI-8007 (Oxigene), EG-3306 (Ark Therapeutics), Maspin (Arriva), ABT- 567 (Abbott), PPI-2458 (Praecis Pharmaceuticals), CC-5079, CC-4089 (Celgene), HIF-1 alpha inhibitors (Xenova), S-247 (Pfizer), AP-23573 (Ariad), AZD-9935 (Astra Zeneca), mebendazole (Introgen Therapeutics), MetAP-2 inhibitors (GlaxoSmithKline), AG-615 (Angiogene Pharmaceuticals), Tie-2 antagonists (Hybrigenics), NC-381 , CYC-381 , NC-169, NC-219, NC-383, NC-384, NC-407 (Lorus Therapeutics), ATN-224 (Attenuon), ON-01370 (Onconova), Vitronectin antagonists (Amgen), SDX-103 (Salmedix), Vitronectin antagonists (Shire), CHP (Riemser), TEK (Amgen), Anecortave acetate (Alcon), T46.2 (Matrix Therapeutics), HG-2 (Heptagen), TEM antagonists (Genzyme), Oxi-4500 (Oxigene), ATN-161 (Attenuon), WX-293 (Wilex), M-2025 (Metris Therapeutics), Alphastatin (BioActa), YH-16 (Yantai Rongchang), BIBF-1120 (Boehringer Ingelheim), BAY-57-9352 (Bayer), AS- 1404 (Cancer Research Technology), SC-77964 (Pfizer), glycomimetics (BioTie Therapies), TIE-2 Inhibitors (Ontogen), DIMI, Octamer (Octamer), ABR-215050 (Active Biotech), ABT-518 (Abbott), KDR inhibitors (Abbott), BSF-466895 (Abbott), SCH-221153 (Schering-Plough), DAC:antiangiogenic (ConjuChem), TFPI (EntreMed), AZD-2171 (Astra-Zenaca), CDC-394 (Celgene), LY290293 (EIi Lilly), IDN-5390 (Indena), Kdr Kinase Inhibitors (Merck), CT-113020, CT-116433, CT-116563, CT-31890, CT-32228) (Cell Therapeutics), A-299620 (Abbott), TWEAK Inhibitor (Amgen), VEGF modulators (Johnson and Johnson), Tum-N53, tumstatin (Genzyme), Thios- 1 , Thios-2 (Thios Pharmaceuticals), MV-6401 (Miravant Medical Technologies), Spisulosine (PharmaMar), CEP-7055 (Cephalon), AUV-201 (Auvation), LM-609 (EIi Lilly), SKF-106615 (AnorMED), Oglufanide disodium (Cylran), BW-114 (Pharminox), Calreticulin (NIH), WX-678 (Wilex), SD-7784 (Pfizer), WX-UK1 (Wilex), SH-268 (Schering AG), 2-Me-PGA (Celgene), S- 137 (Pfizer), ZD-6126 (Angiogene Pharmaceuticals), SG-292 (SignalGen), Benefin (Lane Labs), A6, A36 (Angstrom), SB-2723005 (GlaxoSmithKline), SC-7 (Cell Therapeutics), ZEN-014 (AEterna Zentaris), 2-methoxyestradiol (EntreMed), NK-130119 (Nippon Kayaku), CC-10004 (Celgene), AVE- 8062A (Ajinomoto), Tacedinaline (Pfizer), Actinonin (Tokyo Metropolitan Institute of Medical Science), Lenalidomide (Celgene), VGA-1155, BTO-956 (SRI International), ER-68203-00 (Eisai), CT-6685 (UCB), JKC-362 (Phoenix Pharmaceuticals), DMI-3798 (DMI Biosciences, Angiomate (Ipsen), ZD-6474 (AstraZeneca), CEP-5214 (Cephalon), Canstatin (Genzyme), NM-3 (Mercian), Oridigm (MediQuest Therapeutics), Exherin (Adherex), BLS-0597 (Boston Life Sciences), PTC-299 (PTC Therapeutics), NPI-2358 (Nereus Pharmaceuticals), CGP-79787 (Novartis), AEE-788 (Novartis), CKD-732 (Chong Kun Dang), CP-564959 (OSI Pharmaceuticals), CM-101 (CarboMed), CT-2584, CT3501 (Cell Therapeutics), combretastatin and analogues and derivatives thereof (Oxigene), Rebimastat (Bristol-Meyers Squibb), Dextrin 2-sulfate (ML Laboratories), Cilengitide (Merk KGaA), NSC-706704 (Pharminox), KRN- 951 (Kirin Brewery), Ukrain, NSC-631570 (Nowicky Pharma), Tecogalan sodium (Daiichi Pharmaceutical), Tz-93 (Tsumura), TBC-1635 (Encysive Pharmaceuticals), TAN-1120 (Takeda), Semaxanib (Pfizer), BDI-7800 (Biopharmacopae), SD-186, SD-983 (Bristol-Meyers Squibb), SB-223245 (GlaxoSmithKline), SC-236 (Pfizer), RWJ-590973 (Johnson and Johnson), ILX-1850 (Genzyme), SC-68488, S-836 (Pfizer), CG-55069-11 (CuraGen), Ki-23057 (Kirin Brewery), CCX-700 (Chemoentryx), Pegaptanib octasodium (Giled Sciences), ANGlOCOL (available from Biostratum Inc.), or an analogue or derivative thereof.
6477. The method of item 6470 wherein the agent is an apoptosis antagonist.
6478. The method of item 6470 wherein the agent is an apoptosis activator.
6479. The method of item 6470 wherein the agent is a beta 1 integrin antagonist. 6480. The method of item 6470 wherein the agent is a beta tubulin inhibitor.
6481. The method of item 6470 wherein the agent is a blocker of enzyme production in Hepatitis C.
6482. The method of item 6470 wherein the agent is a Bruton's tyrosine kinase inhibitor.
6483. The method of item 6470 wherein the agent is a calcineurin inhibitor.
6484. The method of item 6470 wherein the agent is a caspase 3 inhibitor.
6485. The method of item 6470 wherein the agent is a CC chemokine receptor antagonist.
6486. The method of item 6470 wherein the agent is a cell cycle inhibitor selected from the group consisting of SNS-595 (Sunesis), synthadotin, KRX-0403, and an analogue or derivative thereof.
6487. The method of item 6470 wherein the agent is a cathepsin B inhibitor.
6488. The method of item 6470 wherein the agent is a cathepsin K inhibitor, wherein the cathepsin K inhibitor is 462795 (GlaxoSmithKline), INPL-022-D6 (Amura Therapeutics), or an analogue or derivative thereof. 6489. The method of item 6470 wherein the agent is a cathepsin L inhibitor.
6490. The method of item 6470 wherein the agent is a CD40 antagonist.
6491. The method of item 6470 wherein the agent is a chemokine receptor agonist.
6492. The method of item 6470 wherein the agent is a chymase inhibitor.
6493. The method of item 6470 wherein the agent is a collagenase antagonist.
6494. The method of item 6470 wherein the agent is a CXCR antagonist.
6495. The method of item 6470 wherein the agent is a cyclin dependent kinase inhibitor selected from the group consisting of a CDK-1 inhibitor, a CDK-2 inhibitor, a CDK- 4 inhibitor, a CDK-6 inhibitor, a CAK1 inhibitor from GPC Biotech and Bristol-Myers Squibb, RGB-286199 (GPC Biotech), an anticancer agent from Astex Technology, a CAK1 inhibitor from GPC Biotech, a CDK inhibitor from Sanofi-Aventis, a CDK1/CDK2 inhibitor from Amgen, a CDK2 inhibitor from SUGEN-2 (Pfizer), a hearing loss therapy agent (Sound Pharmaceuticals), PD-0332991 (Pfizer), RGB-286199 (GPC Biotech), Ro-0505124 (Hoffmann-La Roche), a Ser/Thr kinase inhibitor from Lilly (EIi Lilly), CVT-2584 (CAS No. 199986-75-9) (CV Therapeutics), and an analogue or derivative thereof. 6496. The method of item 6470 wherein the agent is a cyclooxygenase 1 inhibitor.
6497. The method of item 6470 wherein the agent is a DHFR inhibitor.
6498. The method of item 6470 wherein the agent is a dual integrin inhibitor.
6499. The method of item 6470 wherein the agent is an elastase inhibitor.
6500. The method of item 6470 wherein the agent is an elongation factor-1 alpha inhibitor.
6501. The method of item 6470 wherein the agent is an endothelial growth factor antagonist.
6502. The method of item 6470 wherein the agent is an endothelial growth factor receptor kinase inhibitor selected from the group consisting of sorafenib tosylate (Bayer), AAL-993 (Novartis), ABP-309 (Novartis), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), E- 7080 (Eisai), EG-3306 (Ark Therapeutics), EXEL-2880 (Exelixis), GW- 654652 (GlaxoSmithKline), a KDR inhibitor from LG Life Sciences, CT-6685 and CT-6729 (UCB), KRN-633 and KRN-951 (Kirin Brewery), OSI-930 (OSI Pharmaceuticals), SP-5.2 (Supratek Pharma), SU-11657 (Pfizer), a Tie-2 antagonist (Hybrigenics), a VEGFR-2 kinase inhibitor (Bristol-Myers Squibb), XL-647 (Exelixis), a KDR inhibitor from Abbott Laboratories, sorafenib tosylate, and an analogue or derivative thereof. 6503. The method of item 6470 wherein the agent is an endotoxin antagonist.
6504. The method of item 6470 wherein the agent is an epothilone and tubulin binder.
6505. The method of item 6470 wherein the agent is an estrogen receptor antagonist.
6506. The method of item 6470 wherein the agent is an FGF inhibitor.
6507. The method of item 6470 wherein the agent is a farnexyl transferase inhibitor.
6508. The method of item 6470 wherein the agent is farnesyltransferase inhibitor selected from the group of A-197574 (Abbott), a famesyltransferase inhibitor from Servier, FPTIII (Strathclyde Institute for Drug R), LB-42908 (LG Life Sciences), Pharmaprojects No. 5063 (Genzyme), Pharmaprojects No. 5597 (Ipsen), Yissum Project No. B-1055 (Yissum), and an analogue or derivative thereof.
6509. The method of item 6470 wherein the agent is an FLT-3 kinase inhibitor.
6510. The method of item 6470 wherein the agent is an FGF receptor kinase inhibitor.
6511. The method of item 6470 wherein the agent is a fibrinogen antagonist selected from the group consisting of AUV-201 (Auvation), MG-13926 (Sanofi-Aventis), plasminogen activator (CAS No. 105913-11-9) (from Sanofi-Aventis or UCB), plasminogen activator-2 (tPA- 2) (Sanofi-Aventis), pro-urokinase (CAS No. 82657-92-9) (Sanofi-Aventis), and an analogue or derivative thereof.
6512. The method of item 6470 wherein the agent is a heat shock protein 90 antagonist selected from the group consisting of SRN-005 (Sirenade), geldanamycin, NSC-33050 (17-allylaminogeldanamycin; 17- AAG), 17-dimethylaminoethylamino-17-demethoxy-geldanamycin (17- DMAG), rifabutin (rifamycin XIV, 1\4-didehydro-1-deoxy-1 ,4-dihydro-5'-(2- methylpropyl)-1-oxo-), and an analogue or derivative thereof.
6513. The method of item 6470 wherein the agent is a histone deacetylase inhibitor.
6514. The method of item 6470 wherein the agent is an HMGCoA reductase inhibitor selected from the group consisting of an atherosclerosis therapeutic from Lipid Sciences, ATI-16000 (ARYx Therapeutics), KS-01-019 (Kos Pharmaceuticals), Pharmaprojects No. 2197 (Sanofi-Aventi), RP 61969 (Sanofi-Aventis), and an analogue or derivative thereof.
6515. The method of item 6470 wherein the agent is an ICAM inhibitor.
6516. The method of item 6470 wherein the agent is an IL, ICE and IRAK antagonist, wherein the antagonist is a CJ-14877, CP-424174 (Pfizer), NF-61 (Negma-Lerads), and an analogue or derivative thereof.
6517. The method of item 6470 wherein the agent is an IL-2 inhibitor. 6518. The method of item 6470 wherein the agent is an immunosuppressant selected from the group consisting of teriflunomide (Sanofi Aventis), chlorsulfaquinoxalone (NSC-339004), chlorsulfaquinoxalone sulfate, CS-712 (Sankyo), ismomultin alfa (CAS No. 457913-93-8) (Akzo Nobel), antiallergics from GenPat77, antiinflammatories or AT-005 (Androclus Therapeutics), autoimmune disease therapeutics from EpiVax, BN-007 (Bone), budesonide (CAS No. 51333-22- 3) (MAP Pharmaceuticals), CO-14 (Genzyme), edratide (CAS No. 433922- 67-9) (Teva), EP-314 (Enanta), eprovafen (CAS No. 101335-99-3) (Sanofi- Aventis), HWA-131 (CAS No. 118788-41-3) (Sanofi-Aventis), immunomodulators from MerLion Pharmaceuticals, immunosuppressives from Alchemia, IPL-12 (Inflazyme), MDL-9563 (CAS No. 27086-86-8) (Sanofi-Aventis), Pharmaprojects No. 2330 (Sanofi-Aventis), Pharmaprojects No. 6426 (Abgenix), PXS-25 (Pharmaxis), rosmarinic acid (CAS No. 20283-92-5) (Sanofi-Aventis), RP 42927 or RP 54745 (CAS No. 135330-08-4) (Sanofi-Aventis), SGN-35 (Seattle Genetics), ST-1959 (Sigma-Tau), type I diabetes therapy from SYNX Pharma, UNIL-88 (Debiopharm), VP-025 (Vasogen), VR-694 (Vectura), PRTX-001 (Protalex), and an analogue or derivative thereof.
6519. The method of item 6470 wherein the agent is an IMPDH (inosine monophosphate).
6520. The method of item 6470 wherein the agent is an integrin antagonist.
6521. The method of item 6470 wherein the agent is an interleukin antagonist.
6522. The method of item 6470 wherein the agent is an inhibitor of type ill receptor tyrosine kinase. 6523. The method of item 6470 wherein the agent is an irreversible inhibitor of enzyme methionine aminopeptidase type 2.
6524. The method of item 6470 wherein the agent is an isozyme selective delta protein kinase C inhibitor.
6525. The method of item 6470 wherein the agent a JAK3 enzyme inhibitor.
6526. The method of item 6470 wherein the agent is a JNK inhibitor.
6527. The method of item 6470 wherein the agent is a kinase inhibitor.
6528. The method of item 6470 wherein the agent is kinesin antagonist.
6529. The method of item 6470 wherein the agent is a kinesin antagonist.
6530. The method of item 6470 wherein the agent is a leukotriene inhibitor and antagonist selected from the group consisting of ambicromil (CAS No. 58805-38-2) (Sanofi-Aventis), amelubant (CAS No. 346735-24-8) (Boehringer Ingelheim), DW-1141 (Dong Wha), ebselen (Daiichi Pharmaceutical), ibudilast (Kyorin), leucotriene inhibitors from Sanofi-Aventis, lymphotoxin-beta receptor (LT-D) from Biogen Idee, Pharmaprojects No. 1535 and 2728 (CAS No. 119340-33-9) (Sanofi- Aventis), R-112 (Rigel), Rev-5367 (CAS No. 92532-05-3) (Sanofi-Aventis), RG-14893 (CAS No. 141835-49-6) (Sanofi-Aventis), RG-5901-A (CAS No. 101910-24-1), 92532-23-5, RP 66153 (CAS No. 142422-79-5), RP 66364 (CAS No.186916-92-5), RP 69698 (CAS No. 141748-00-7) (Sanofi-Aventis), SC-411930 (Pfizer), SC-41930 (CAS No. 120072-59-5) (Pfizer), SC-50605 (CAS No. 138828-39-4) (Pfizer), SC-51146 (CAS No. 141059-52-1), SC- 53228 (CAS No. 153633-01-3) (Pfizer), spaglumic acid (ZY-15106) (CAS No.3106-85-2), 80619-64-3 (Novartis), tipredane (CAS No. 85197-77-9) (Bristol-Myers Squibb), U-75302 (CAS No. 119477-85-9) (Pfizer), and analogue or derivative thereof.
6531. The method of item 6470 wherein the agent is an MAP kinase inhibitor.
6532. The method of item 6470 wherein the agent is a matrix metalloproteinase inhibitor.
6533. The method of item 6470 wherein the agent is an MCP- CCR2 inhibitor.
6534. The method of item 6470 wherein the agent is an mTOR inhibitor.
6535. The method of item 6470 wherein the agent is an mTOR kinase inhibitor.
6536. The method of item 6470 wherein the agent is a microtubule inhibitor selected from the group consisting of antibody- maytansinoid conjugates from Biogen Idee, colchicines (MantiCore Pharmaceuticals), anticancer immunoconjugates from Johnson & Johnson, DIME from Octamer, gni-1f (GNI), huC242-DM4, huMy9-6-DM1 (ImmunoGen), IDN-5404 (Indena), IMO-098, IMOderm (Imotep), mebendazole (Introgen Therapeutics), microtubule poisons from Cambridge Enterprise, paclitaxel such as LOTAX from Aphios (CAS No. 33069-62-4), Genexoi-PM from Samyang, Pharmaprojects No. 6383 (Tapestry Pharmaceuticals), RPR-112378 (Sanofi-Aventis), SGN-75 (Seattle Genetics), SPL-7435 (Starpharma), SSR-250411 (Sanofi-Aventis), trastuzumab-DM1 (Genentech), vinorelbine, and an analogue or derivative thereof.
6537. The method of item 6470 wherein the agent is an MIF inhibitor.
6538. The method of item 6470 wherein the agent is an MMP inhibitor.
6539. The method of item 6470 wherein the agent is a neurokinin (NK) antagonist selected from the group consisting of anthrotainin (CAS No. 148084-40-6) (Sanofi-Aventis), an IBS thereapeutic from ArQuIe, MDL-105212A (CAS No. 167261-60-1) (Ssanofi-Aventis), Pharmaprojects No. 2744, 3258 (CAS No. 139167-47-8) 4006, 4201, or 5986 (Sanofi-Aventis), RP 67580 (CAS No. 135911-02-3), SR-144190 (CAS No. 201152-86-5), SSR-240600, SSR-241586 (Sanofi-Aventis), TKA-457 (Novartis), vestipitant mesylate (CAS No. 334476-64-1) (GlaxoSmithKline), Win-64821 (Sanofi-Aventis), PRX-96026 (Predix Pharmaceuticals), and an analogue or derivative thereof.
6540. The method of item 6470 wherein the agent is an NF kappa B inhibitor selected from the group consisting of AVE-0545 or AVE- 0547 (Sanofi-Aventis), bortezomib (CAS No. 179324-69-7) (Millennium Pharmaceuticals), dexanabinol (CAS No. 112924-45-5) (Pharmos), dexlipotam (Viatris), Pharmaprojects No. 6283 (INDRA) (OXiGENE), IPL- 576092 (CAS No. 137571-30-3) (Inflazyme), NFKB decoy (Corgentech), NFKB decoy oligo (AnGes MG), NFKB's from Ariad, osteoporosis treatments or S5 (F005) from Fulcrum Pharmaceuticals, P61 (Phytopharm), R-flurbiprofen (CAS No. 5104-49-4) (Encore Pharmaceuticals), and an analogue or derivative thereof.
6541. The method of item 6470 wherein the agent is a nitric oxide agonist.
6542. The method of item 6470 wherein the agent is an ornithine decarboxylase inhibitor.
6543. The method of item 6470 wherein the agent is a p38 MAP kinase inhibitor selected from the group consisting of AZD-6703 (AstraZeneca), JX-401 (Jexys Pharmaceuticals), BMS-2 (Bristol-Myers Squibb), a p38 MAP kinase inhibitor from Novartis, a p38-alpha MAP kinase inhibitor from Amphora, Pharmaprojects No. 5704 (Pharmacopeia), RPR- 200765A (Sanofi-Aventis), SD-282 (Johnson & Johnson), TAK-715 (Takeda), and an analogue or derivative thereof.
6544. The method of item 6470 wherein the agent is a palmitoyl-protein thioesterase inhibitor.
6545. The method of item 6470 wherein the agent is a PDGF receptor kinase inhibitor selected from the group consisting of AAL-993, AMN-107, or ABP-309 (Novartis), AMG-706 (Amgen), BAY-57-9352 (Bayer), CDP-860 (UCB), E-7080 (Eisai), imatinib (CAS No. 152459-95-5) (Novartis), OSI-930 (OSI Pharmaceuticals), RPR-127963E (Sanofi-Aventis), RWJ-540973 (Johnson & Johnson), sorafenib tosylate (Bayer), SU-11657 (Pfizer), tandutinib (CAS No. 387867-13-2) (Millennium Pharmaceuticals), vatalanib (Novartis), ZK-CDK (Schering AG), and an analogue or derivative thereof. 6546. The method of item 6470 wherein the agent is (-)- haiofenate (Metabolex), AMG-131 (Amgen), antidiabetics from Japan Tobacco, AZD-4619, AZD-8450, AZD-8677 (AstraZeneca), DRF-10945, balaglitazone (Dr Reddy's), CS-00088, CS-00098 (Chipscreen Biosciences), E-3030 (Eisai), etalocib (CAS No. 161172-51-6) (EIi Lilly), GSK-641597 (Ligand), GSK-677954 (GlaxoSmithKline), GW-409544 (Ligand), GW- 590735 (GlaxoSmithKline), K-111 (Hoffmann-La Roche), LY-518674 (EIi Lilly), LY-674 (Ligand), LY-929 (Ligand), MC-3001 , MC-3002 (MaxoCore Pharmaceuticals), metformin HCI + pioglitazone (CAS No. 1115-70-4 and
112529-15-4), ACTOPLUS MET from Andrx), muraglitazar (CAS No. 331741-94-7) (Bristol-Myers Squibb), naveglitazar (Ligand), oleoylethanolamide (Kadmus Pharmaceuticals), ONO-5129, pioglitazone hydrochloride (CAS No. 111025-46-8 and 112529-15-4) (Takeda), PLX-204 (Plexxikon), PPAR agonists from Genfit, PPAR delta agonists from EIi Lilly, PPAR-alpha agonists from CrystalGenomics, PPAR-gamma modulators and PPAR-β modulators from CareX, rosiglitazone maleate (CAS No. 122324- 73-4 or 155141-29-0) (GlaxoSmithKline), rosiglitazone maleate/glimepir (CAS No. 155141-29-0 and 93479-97-1), AVANDARYL, rosiglitazone maleate/metformin extend (CAS No. 155141-29-0 and 657-24-9), AVANDAMET, rosiglitazone maleate+metformin, AVANDAMET (GlaxoSmithKline), tesaglitazar (AstraZeneca), and an analogue or derivative thereof.
6547. The method of item 6470 wherein the agent is a phosphatase inhibitor.
6548. The method of item 6470 wherein the agent is a phosphodiesterase (PDE) inhibitor selected from the group consisting of avanafil (Tanabe Seiyaku), dasantafil (CAS No. 569351-91-3) (Schering- Plough), A-906119 (CAS No. 134072-58-5), DL-850 (Sanofi-Aventis), GRC- 3015, GRC-3566, GRC-3886 (Glenmark), HWA-153 (CAS No. 56395-66-5) (Sanofi-Aventis), hydroxypumafentrine (Altana), IBFB-130011 , IBFB-14-016, IBFB-140301 , IBFB-150007, IBFB-211913 (IBFB Pharma), L-826141 (Merck & Co), medorinone (CAS No. 88296-61-1) (Sanofi-Aventis), MEM- 1917 (Memory Pharmaceuticals), ND-1251 (Neuro3d), PDE inhibitors from ICOS, PDE IV inhibitors from Memory Pharmaceuticals and CrystalGenomics, Pharmaprojects No. 2742 and 6141 (Sanofi-Aventis), QAD-171 (Novartis), RHC-2963 (CAS No. 76993-12-9 and 76993-14-1), RPFM 17658, RPR-122818 derivatives, SR-24870 , and RPR-132294 (Sanofi-Aventis), SK-350 (ln2Gen), stroke therapy agents from deCODE Genetics, TAS-203 (Taiho), tofimilast (CAS No. 185954-27-2) (Pfizer), UK- 371800 (Pfizer), WIN-65579 (CAS No. 158020-82-7) (Sanofi-Aventis), IBFB- 130020 (IBFB Pharma), OPC-6535 (CAS No. 145739-56-6) (Otsuka), a phosphodiesterase III inhibitor, enoximone, a phosphodiesterase IV inhibitor, fosfosal, Atopik (Barrier Therapeutics), triflusal, a phosphodiesterase V inhibitor, and an analogue or derivative thereof.
6549. The method of item 6470 wherein the agent is a PKC inhibitor.
6550. The method of item 6470 wherein the agent is a platelet activating factor antagonist.
6551. The method of item 6470 wherein the agent is a platelet-derived growth factor receptor kinase inhibitor.
6552. The method of item 6470 wherein the agent is a prolyl hydroxylase inhibitor.
6553. The method of item 6470 wherein the agent is a polymorphonuclear neutrophil inhibitor. 6554. The method of item 6470 wherein the agent is a protein kinase B inhibitor.
6555. The method of item 6470 wherein the agent is a protein kinase C stimulant.
6556. The method of item 6470 wherein the agent is a purine nucleoside analogue.
6557. The method of item 6470 wherein the agent is a purinoreceptor P2X antagonist.
6558. The method of item 6470 wherein the agent is a Raf kinase inhibitor.
6559. The method of item 6470 wherein the agent is a reversible inhibitor of ErbB1 and ErbB2.
6560. The method of item 6470 wherein the agent is a ribonucleoside triphosphate reductase inhibitor.
6561. The method of item 6470 wherein the agent is an SDF- 1 antagonist.
6562. The method of item 6470 wherein the agent is a sheddase inhibitor.
6563. The method of item 6470 wherein the agent is an SRC inhibitor. 6564. The method of item 6470 wherein the agent is a stromelysin inhibitor.
6565. The method of item 6470 wherein the agent is an Syk kinase inhibitor.
6566. The method of item 6470 wherein the agent is a telomerase inhibitor.
6567. The method of item 6470 wherein the agent is a TGF beta inhibitor selected from the group consisting of pirfenidone (CAS No. 53179-13-8) (MARNAC), tranilast (CAS No. 53902-12-8) (Kissei), IN-1130 (ln2Gen), mannose-6-phosphate (BTG), TGF-β antagonists from Inflazyme (Pharmaprojects No. 6075), TGF-β antagonists from Sydney, non-industrial source), TGF-β I receptor kinase inhibitors from EIi Lilly, TGF-β receptor inhibitors from Johnson & Johnson, and an analogue or derivative thereof.
6568. The method of item 6470 wherein the agent is a TNFα antagonist or TACE inhibitor selected from the group consisting of adalimumab (CAS No. 331731-18-1) (Cambridge Antibody Technology), AGIX-4207 (AtheroGenics), AGT-1 (Advanced Biotherapy), an antiinflammatory from Borean Pharma, Cellzome, or Paradigm Therapeutics, anti-inflammatory vaccine (TNF-alpha kinoid) from Neovacs, humanized anti-TNF antibody or an anti-TNF MAb (CB0006) Celltech (UCB), apratastat (CAS No. 287445-51-0) (Wyeth), BMS-561392 (Bristol-Myers Squibb), BN- 006 (Bone), certolizumab pegol (CAS No. 428863-50-7 or CH-138 (UCB), cilomilast (CAS No. 153259-65-5) (GlaxoSmithKline), CR-1 (Nuada Pharmaceuticals), CRx-119 (CombinatoRx), D-5410 (UCB), dacopafant (CAS No. 125372-33-0) (Sanofi-Aventis), dersalazine (CAS No. 188913-57- 7/188913-58-8) (Uriach), etanercept (CAS No. 185243-69-0) (Amgen), ethyl pyruvate (Critical (Critical Therapeutics), golimumab (CAS No. 476181-74-5) (Johnson & Johnson), hormono-immunotherapy from Ipsen, CDP571 (e.g., Humicade from UCB), IC-485 (ICOS), infliximab (CAS No. 170277-31-3) (Johnson & Johnson), IP-751 (Manhattan Pharmaceuticals), ISIS-104838 (CAS No. 250755-32-9) (ISIS Pharmaceuticals), lenalidomide (CAS No. 191732-72-6) (Celgene), lentinan (CAS No. 37339-90-5) (Ajinomoto), MDL- 201112 (CAS No. 142130-73-2) (Sanofi-Aventis), medroxyprogesterone (CAS No. 520-85-4) (InKine Pharmaceutical), N-acetylcysteine (CAS No. 616-91-1) (Zambon), NBE-P2 (DIREVO Biotech), nerelimomab (CAS No. 162774-06-3) (Chiron), OM-294DP (OM PHARMA), onercept (CAS No. 199685-57-9) (Yeda), PASSTNF-alpha (Verigen), pentoxifylline or oxypentifylline (Sanofi-Aventis), Pharmaprojects No. 4091 , 4241 , 4295, or 4488 (Sanofi-Aventis), Pharmaprojects No. 5480 (Amgen), Pharmaprojects No. 6749 (Cengent), pirfenidone (CAS No. 53179-13-8) (MARNAC), RPR- 132294 (Sanofi-Aventis), S5 (F002) (Fulcrum Pharmaceuticals), simvastatin (CAS No. 79902-63-9) (Merck & Co), STA-6292 (Synta Pharmaceuticals), tacrolimus (CAS No. 104987-11-3) (Fujisawa LifeCycle Pharma), talactoferrin alfa (CAS No. 308240-58-6) (Agennix), thalidomide (CAS No. 50-35-1) (Celgene), TNF antagonists form ProStrakan, and Synergen, TNF inhibitors (Amgen), TNF-alpha antagonists from Dynavax Technologies and Jerina AG (Germany), TNF-alpha inhibitors from IBFB Pharma and Xencor (Xencor), torbafylline (CAS No. 105102-21-4) (Sanofi-Aventis), UR-1505 (Uriach), VT-346 (Viron Therapeutics), YSIL6 (Y's Therapeutics), YSTH2 (Y's Therapeutics), NPI-1302a-3 (Nereus Pharmaceuticals, a TNF antagonist from Jerina AG (Germany), dersalazine, and an analogue or derivative thereof.
6569. The method of item 6470 wherein the agent is a Toll receptor inhibitor.
6570. The method of item 6470 wherein the agent is a tubulin antagonist. 6571. The method of item 6470 wherein the agent is a tyrosine kinase inhibitor selected from the group consisting of SU-011248, SUTENT from Pfizer Inc. (New York, NY), BMS-354825, PN-355 (Paracelsian Pharmaceuticals), AGN-199659 (Allergan), AAL-993 or ABP- 309 (Novartis), adaphostin (NIH), AEE-788 (Novartis), AG-013736 (OSI Pharmaceuticals), AG-13736 (Pfizer), ALT-110 (Alteris Therapeutics), AMG- 706 (Amgen), anticancer MAbs from Xencor, anti-EGFrvlll MAbs from Abgenix, anti-HER2 MAb from Abiogen, AZD-2171 or AZD-9935 (AstraZeneca), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), CEP-5214 (Cephalon), CEP-7055 (Cephalon), cetuximab (ImClone Systems), CHIR-200131 and CHIR-258 (Chiron), CP-547632 (OSI Pharmaceuticals), CP-724714 (Pfizer), CT-301 (Creabilis Therapeutics), D- 69491 (Baxter International), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EGFR/ErbB2 inhibitors from Array BioPharma, erlotinib (CAS No. 183319- 69-9) (OSI Pharmaceuticals), EXEL-2880 (Exelixis), FK-778 (Sanofi- Aventis) , gefitinib (CAS No. 184475-35-2) (AstraZeneca), GW-2286 or GW- 654652 (GlaxoSmithKline), her2/neu antigen from AlphaVax, HER-2/neu inhibitor from Generex, Herzyme (Medipad) (Sirna Therapeutics), HKI-272 (Wyeth), HuMax-EGFr (Genmab), idronoxil (CAS No. 81267-65-4) (Novogen), IGF-1 inhibitors from Ontogen, IMC-11 F8 (ImClone Systems), kahalalide F (CAS No. 149204-42-2) (PharmaMar), KDR inhibitor from LG Life Sciences, KDR inhibitors from Abbott Laboratories, KDR kinase inhibitors (UCB), Kdr kinase inhibitors from Merck & Co, KRN-633 and KRN- 951 (Kirin Brewery), KSB-102 (Xenova), lapatinib ditosylate (CAS No. 388082-78-8) (GlaxoSmithKline), matuzumab (Merck KGaA), MDX-214 (Medarex), ME-103 (Pharmexa), MED-A300 (Gerolymatos), MNAC-13 (Lay Line Genomics), nimotuzumab (Center of Molecular Immunology), NSC- 330507 or NSC-707545 (NIH), NV-50 (Novogen), OSI-930 (OSI Pharmaceuticals), panitumumab (Abgenix), pelitinib (CAS No. 287933-82-7) (Wyeth), pertuzumab (CAS No. 380610-27-5) (Genentech), Pharmaprojects No. 3985 (Sanofi-Aventis), prostate cancer therapeutics from Sequenom (SQPC35, SQPC36, SQPC90), removab and remoxab (Trion Pharma), RG- 13022 (CAS No. 136831-48-6), RG-13291 (CAS No. 138989-50-1), or RG- 14620 (CAS No. 136831-49-7) (Sanofi-Aventis), RM-6427 (Romark), RNAi breast cancer therapy from Benitec, RP 53801 (CAS No. 125882-88-4) (Sanofi-Aventis), sorafenib tosylate (Bayer), SU-11657 (Pfizer), Tie-2 antagonists from Semaia (Hybrigenics), Tie-2 inhibitors from Ontogen, trastuzumab (CAS No. 180288-69-1) (Genentech), tyrosine kinase inhibitors from Sanofi-Aventis, U3-1287, U3-1565, U3-1784, U3-1800 (U3 Pharma), vatalanib (Novartis), VEGFR-2 kinase inhibitor from Bristol-Myers Squibb, XL-647 (Exelixis), ZD-6474 (AstraZeneca), ZK-CDK (Schering AG), an EGFR tyrosine kinase inhibitor, EKB-569 (Wyeth), and an analogue or derivative thereof.
6572. The method of item 6470 wherein the agent is a VEGF inhibitor.
6573. The method of item 6470 wherein the agent is a vitamin D receptor agonist.
6574. The method of item 6470 wherein the agent is ZD-6474 (an angiogenesis inhibitor).
6575. The method of item 6470 wherein the agent is AP- 23573 (an mTOR inhibitor).
6576. The method of item 6470 wherein the agent is synthadotin (a tubulin antagonist).
6577. The method of item 6470 wherein the agent is S-0885 (a collagenase inhibitor). 6578. The method of item 6470 wherein the agent is aplidine (an elongation factor-1 alpha inhibitor).
6579. The method of item 6470 wherein the agent is ixabepilone (an epithilone).
6580. The method of item 6470 wherein the agent is IDN- 5390 (an angiogenesis inhibitor).
6581. The method of item 6470 wherein the agent is SB- 2723005 (an angiogenesis inhibitor).
6582. The method of item 6470 wherein the agent is ABT-518 (an angiogenesis inhibitor).
6583. The method of item 6470 wherein the agent is combretastatin (an angiogenesis inhibitor).
6584. The method of item 6470 wherein the agent is anecortave acetate (an angiogenesis inhibitor).
6585. The method of item 6470 wherein the agent is SB- 715992 (a kinesin antagonist).
6586. The method of item 6470 wherein the agent is temsirolimus (an mTOR inhibitor).
6587. The method of item 6470 wherein the agent is adalimumab (a TNFα antagonist). 6588. The method of item 6470, wherein the composition comprises a polymer.
6589. The method of item 6470, wherein the composition comprises a polymer, and the polymer is, or comprises, a copolymer.
6590. The method of item 6470, wherein the composition comprises a polymer, and the polymer is, or comprises, a block copolymer.
6591. The method of item 6470, wherein the composition comprises a polymer, and the polymer is, or comprises, a random copolymer.
6592. The method of item 6470, wherein the composition comprises a polymer, and the polymer is, or comprises, a biodegradable polymer.
6593. The method of item 6470, wherein the composition comprises a polymer, and the polymer is, or comprises, a nonbiodegradable polymer.
6594. The method of item 6470, wherein the composition comprises a polymer, and the polymer is, or comprises, a hydrophilic polymer.
6595. The method of item 6470, wherein the composition comprises a polymer, and the polymer is, or comprises, a hydrophobic polymer. 6596. The method of item 6470, wherein the composition comprises a polymer, and the polymer is, or comprises, a polymer having hydrophilic domains.
6597. The method of item 6470, wherein the composition comprises a polymer, and the polymer is, or comprises, a polymer having hydrophobic domains.
6598. The method of item 6470, wherein the composition comprises a polymer, and the polymer is, or comprises, a non-conductive polymer.
6599. The method of item 6470, wherein the composition comprises a polymer, and the polymer is, or comprises, an elastomer.
6600. The method of item 6470, wherein the composition comprises a polymer, and the polymer is, or comprises, a hydrogel.
6601. The method of item 6470, wherein the composition comprises a polymer, and the polymer is, or comprises, a silicone polymer.
6602. The method of item 6470, wherein the composition comprises a polymer, and the polymer is, or comprises, a hydrocarbon polymer.
6603. The method of item 6470, wherein the composition comprises a polymer, and the polymer is, or comprises, a styrene-derived polymer. 6604. The method of item 6470, wherein the composition comprises a polymer, and the polymer is, or comprises, a butadiene-derived polymer.
6605. The method of item 6470, wherein the composition comprises a polymer, and the polymer is, or comprises, a macromer.
6606. The method of item 6470, wherein the composition comprises a polymer, and the polymer is, or comprises, a poly(ethylene glycol) polymer.
6607. The method of item 6470, wherein the composition comprises a polymer, and the polymer is, or comprises, an amorphous polymer.
6608. The method of item 6470, wherein the composition further comprises a second pharmaceutically active agent.
6609. The method of item 6470, wherein the composition further comprises an anti-inflammatory agent.
6610. The method of item 6470, wherein the composition further comprises an agent that inhibits infection.
6611. The method of item 6470, wherein the composition further comprises an anthracycline.
6612. The method of item 6470, wherein the composition further comprises doxorubicin. 6613. The method of item 6470 wherein the composition further comprises mitoxantrone.
6614. The method of item 6470 wherein the composition further comprises a fluoropyrimidine.
6615. The method of item 6470, wherein the composition further comprises 5-fluorouracil (5-FU).
6616. The method of item 6470, wherein the composition further comprises a folic acid antagonist.
6617. The method of item 6470, wherein the composition further comprises methotrexate.
6618. The method of item 6470, wherein the composition further comprises a podophylotoxin.
6619. The method of item 6470, wherein the composition further comprises etoposide.
6620. The method of item 6470, wherein the composition further comprises camptothecin.
6621. The method of item 6470, wherein the composition further comprises a hydroxyurea.
6622. The method of item 6470, wherein the composition further comprises a platinum complex. 6623. The method of item 6470, wherein the composition further comprises cisplatin.
6624. The method of item 6470 wherein the composition further comprises an anti-thrombotic agent.
6625. The method of item 6470, wherein the composition further comprises a visualization agent.
6626. The method of item 6470, wherein the composition further comprises a visualization agent, and the visualization agent is a radiopaque material, wherein the radiopaque material comprises a metal, a halogenated compound, or a barium containing compound.
6627. The method of item 6470, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, barium, tantalum, or technetium.
6628. The method of item 6470, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, an MRI responsive material.
6629. The method of item 6470, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, a gadolinium chelate.
6630. The method of item 6470, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, iron, magnesium, manganese, copper, or chromium. 6631. The method of item 6470, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, iron oxide compound.
6632. The method of item 6470, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, a dye, pigment, or colorant.
6633. The method of item 6470 wherein the agent is released in effective concentrations from the composition comprising the agent by diffusion over a period ranging from the time of administration to about 90 days.
6634. The method of item 6470 wherein the agent is released in effective concentrations from the composition comprising the agent by erosion of the composition over a period ranging from the time of administration to about 90 days.
6635. The method of item 6470 wherein the composition further comprises an inflammatory cytokine.
6636. The method of item 6470 wherein the composition further comprises an agent that stimulates cell proliferation.
6637. The method of item 6470 wherein the composition further comprises a polymeric carrier.
6638. The method of item 6470 wherein the composition is in the form of a gel, paste, or spray. 6639. The method of item 6470 wherein the implant is partially constructed with the agent or the composition.
6640. The method of item 6470 wherein the implant is fully constructed with the agent or the composition.
6641. The method of item 6470 wherein the implant is impregnated with the agent or the composition.
6642. The method of item 6470, wherein the agent or the composition forms a coating, and the coating directly contacts the implant.
6643. The method of item 6470, wherein the agent or the composition forms a coating, and the coating indirectly contacts the implant.
6644. The method of item 6470 wherein the agent or the composition forms a coating, and the coating partially covers the implant.
6645. The method of item 6470, wherein the agent or the composition forms a coating, and the coating completely covers the implant.
6646. The method of item 6470 wherein the agent or the composition is located within pores or holes of the implant.
6647. The method of item 6470 wherein the agent or the composition is located within a channel, lumen, or divet of the implant.
6648. The method of item 6470 wherein the implant further comprising an echogenic material. 6649. The method of item 6470 wherein the implant further comprises an echogenic material, wherein the echogenic material is in the form of a coating.
6650. The method of item 6470 wherein the implant is sterile.
6651. The method of item 6470 wherein the agent is delivered from the implant, wherein the agent is released into tissue in the vicinity of the implant after deployment of the implant.
6652. The method of item 6470 wherein the agent is delivered from the implant, wherein the agent is released into tissue in the vicinity of the implant after deployment of the implant, wherein the tissue is connective tissue.
6653. The method of item 6470 wherein the agent is delivered from the implant, wherein the agent is released into tissue in the vicinity of the implant after deployment of the implant, wherein the tissue is muscle tissue.
6654. The method of item 6470 wherein the agent is delivered from the implant, wherein the agent is released into tissue in the vicinity of the implant after deployment of the implant, wherein the tissue is nerve tissue.
6655. The method of item 6470 wherein the agent is delivered from the implant, wherein the agent is released into tissue in the vicinity of the implant after deployment of the implant, wherein the tissue is epithelium tissue. 6656. The method of item 6470 wherein the agent is delivered from the implant, wherein the agent is released in effective concentrations from the implant over a period ranging from the time of deployment of the implant to about 1 year.
6657. The method of item 6470 wherein the agent is delivered from the implant, wherein the agent is released in effective concentrations from the implant over a period ranging from about 1 month to 6 months.
6658. The method of item 6470 wherein the agent is delivered from the implant, wherein the agent is released in effective concentrations from the implant over a period ranging from about 1 - 90 days.
6659. The method of item 6470 wherein the agent is delivered from the implant, wherein the agent is released in effective concentrations from the implant at a constant rate.
6660. The method of item 6470 wherein the agent is delivered from the implant, wherein the agent is released in effective concentrations from the implant at an increasing rate.
6661. The method of item 6470 wherein the agent is delivered from the implant, wherein the agent is released in effective concentrations from the implant at a decreasing rate.
6662. The method of item 6470 wherein the agent is delivered from the implant, wherein the implant comprises about 0.01 Dg to about 10 Dg of the agent. 6663. The method of item 6470 wherein the agent is delivered from the implant, wherein the implant comprises about 10 Dg to about 10 mg of the agent.
6664. The method of item 6470 wherein the agent is delivered from the implant, wherein the implant comprises about 10 mg to about 250 mg of the agent.
6665. The method of item 6470 wherein the agent is delivered from the implant, wherein the implant comprises about 250 mg to about 1000 mg of the agent.
6666. The method of item 6470 wherein the agent is delivered from the implant, wherein the implant comprises about 1000 mg to about 2500 mg of the agent.
6667. The method of item 6470 wherein the agent is delivered from the implant, wherein a surface of the implant comprises less than 0.01 Dg of the agent per mm2 of implant surface to which the agent is applied.
6668. The method of item 6470 wherein the agent is delivered from the implant, wherein a surface of the implant comprises about 0.01 Dg to about 1 Dg of the agent per mm2 of implant surface to which the agent is applied.
6669. The method of item 6470 wherein the agent is delivered from the implant, wherein a surface of the implant comprises about 1 Dg to about 10 Dg of the agent per mm2 of implant surface to which the agent is applied. 6670. The method of item 6470 wherein the agent is delivered from the implant, wherein a surface of the implant comprises about 10 Dg to about 250 Dg of the agent per mm2 of implant surface to which the agent is applied.
6671. The method of item 6470 wherein the agent is delivered from the implant, wherein a surface of the implant comprises about 250 Dg to about 1000 Dg of the agent per mm2 of implant surface to which the agent is applied.
6672. The method of item 6470 wherein the agent is delivered from the implant, wherein a surface of the implant comprises about 1000 Dg to about 2500 Dg of the agent per mm2 of implant surface to which the agent is applied.
6673. The method of item 6470, wherein the implant further comprises a coating, and the coating is a uniform coating.
6674. The method of item 6470, wherein the implant further comprises a coating, and the coating is a non-uniform coating.
6675. The method of item 6470, wherein the implant further comprises a coating, and the coating is a discontinuous coating.
6676. The method of item 6470, wherein the implant further comprises a coating, and the coating is a patterned coating.
6677. The method of item 6470, wherein the implant further comprises a coating, and the coating has a thickness of 100 Dm or less. 6678. The method of item 6470, wherein the implant further comprises a coating, and the coating has a thickness of 10 Dm or less.
6679. The method of item 6470, wherein the implant further comprises a coating, and the coating adheres to the surface of the implant upon deployment of the implant.
6680. The method of item 6470, wherein the implant further comprises a coating, and the coating is stable at room temperature for a period of at least 1 year.
6681. The method of item 6470, wherein the implant further comprises a coating, and the agent is present in the coating in an amount ranging between about 0.0001% to about 1% by weight.
6682. The method of item 6470, wherein the implant further comprises a coating, and the agent is present in the coating in an amount ranging between about 1% to about 10% by weight.
6683. The method of item 6470, wherein the implant further comprises a coating, and the agent is present in the coating in an amount ranging between about 10% to about 25% by weight.
6684. The method of item 6470, wherein the implant further comprises a coating, and the agent is present in the coating in an amount ranging between about 25% to about 70% by weight.
6685. The method of item 6470, wherein the implant further comprises a coating, and the coating comprises a polymer. 6686. The method of item 6470, wherein the implant comprises a first coating having a first composition and a second coating having a second composition.
6687. The method of item 6470, wherein the implant comprises a first coating having a first composition and a second coating having a second composition, wherein the first composition and the second composition are different.
6688. A method as in any one of items 6470-6687, wherein the device is a ureteric stent.
6689. A method as in any one of items 6470-6687, wherein the device is a urethral stent.
6690. A method as in any one of items 6470-6687, wherein the device is a fallopian tube stent.
6691. A method as in any one of items 6470-6687, wherein the device is a prostate stent.
6692. A method as in any one of items 6470-6687, wherein the device is a metallic genital urinary stent.
6693. A method as in any one of items 6470-6687, wherein the device is a polymeric genital urinary stent.
6694. A method for inhibiting scarring comprising placing a device that comprises an ear and nose stent (e.g., an implant) and an anti- scarring agent or a composition comprising an anti-scarring agent into an animal host, wherein the agent inhibits scarring. 6695. The method of item 6694 wherein the agent is an adensosine A2A receptor antagonist.
6696. The method of item 6694 wherein the agent is an AKT inhibitor.
6697. The method of item 6694 wherein the agent is an alpha 2 integrin antagonist, wherein the alpha 2 integrin antagonist is Pharmaprojects No. 5754 (Merck KgaA).
6698. The method of item 6694 wherein the agent is an alpha 4 integrin antagonist.
6699. The method of item 6694 wherein the agent is an alpha 7 nicotinic receptor agonist.
6700. The method of item 6694 wherein the agent is an angiogenesis inhibitor selected from the group consisting of AG-12,958 (Pfizer), ATN-161 (Attenuon LLC), neovastat, an angiogenesis inhibitor from Jerina AG (Germany), NM-3 (Mercian), VGA-1155 (Taisho), FCE-26644 (Pfizer), FCE-26950 (Pfizer), FPMA (Meiji Daries), FR-111142 (Fujisawa), GGTI-298, GM-1306 (Ligand), GPA-1734 (Novartis), NNC-47-0011 (Novo Nordisk), herbamycin (Nippon Kayaku), lenalidomide (Celegene), IP-10 (NIH), ABT-828 (Abbott), KIN-841 (Tokushima University, Japan), SF-1126 (Semafore Pharmaceuticals), laminin technology (NIH), CHIR-258 (Chiron), NVP-AEW541 (Novartis), NVP-AEW541 (Novartis), Vt16907 (Alchemia), OXI-8007 (Oxigene), EG-3306 (Ark Therapeutics), Maspin (Arriva), ABT- 567 (Abbott), PPI-2458 (Praecis Pharmaceuticals), CC-5079, CC-4089 (Celgene), HIF-1 alpha inhibitors (Xenova), S-247 (Pfizer), AP-23573 (Ariad), AZD-9935 (Astra Zeneca), mebendazole (Introgen Therapeutics), MetAP-2 inhibitors (GlaxoSmithKline), AG-615 (Angiogene Pharmaceuticals), Tie-2 antagonists (Hybrigenics), NC-381 , CYC-381, NC-169, NC-219, NC-383, NC-384, NC-407 (Lorus Therapeutics), ATN-224 (Attenuon), ON-01370 (Onconova), Vitronectin antagonists (Amgen), SDX-103 (Salmedix), Vitronectin antagonists (Shire), CHP (Riemser), TEK (Amgen), Anecortave acetate (Alcon), T46.2 (Matrix Therapeutics), HG-2 (Heptagen), TEM antagonists (Genzyme), Oxi-4500 (Oxigene), ATN-161 (Attenuon), WX-293 (Wilex), M-2025 (Metris Therapeutics), Alphastatin (BioActa), YH-16 (Yantai Rongchang), BIBF-1120 (Boehringer Ingelheim), BAY-57-9352 (Bayer), AS- 1404 (Cancer Research Technology), SC-77964 (Pfizer), glycomimetics (BioTie Therapies), TIE-2 Inhibitors (Ontogen), DIMI1 Octamer (Octamer), ABR-215050 (Active Biotech), ABT-518 (Abbott), KDR inhibitors (Abbott), BSF-466895 (Abbott), SCH-221153 (Schering-Plough), DAC:antiangiogenic (ConjuChem), TFPI (EntreMed), AZD-2171 (Astra-Zenaca), CDC-394 (Celgene), LY290293 (EIi Lilly), IDN-5390 (Indena), Kdr Kinase Inhibitors (Merck), CT-113020, CT-116433, CT-116563, CT-31890, CT-32228) (Cell Therapeutics), A-299620 (Abbott), TWEAK Inhibitor (Amgen), VEGF modulators (Johnson and Johnson), Tum-N53, tumstatin (Genzyme), Thios- 1 , Thios-2 (Thios Pharmaceuticals), MV-6401 (Miravant Medical Technologies), Spisulosine (PharmaMar), CEP-7055 (Cephalon), AUV-201 (Auvation), LM-609 (EIi Lilly), SKF-106615 (AnorMED), Oglufanide disodium (Cytran), BW-114 (Pharminox), Calreticulin (NIH), WX-678 (Wilex), SD-7784 (Pfizer), WX-UK1 (Wilex), SH-268 (Schering AG), 2-Me-PGA (Celgene), S- 137 (Pfizer), ZD-6126 (Angiogene Pharmaceuticals), SG-292 (SignalGen), Benefin (Lane Labs), A6, A36 (Angstrom), SB-2723005 (GlaxoSmithKline), SC-7 (Cell Therapeutics), ZEN-014 (AEtema Zentaris), 2-methoxyestradiol (EntreMed), NK-130119 (Nippon Kayaku), CC-10004 (Celgene), AVE- 8062A (Ajinomoto), Tacedinaline (Pfizer), Actinonin (Tokyo Metropolitan Institute of Medical Science), Lenalidomide (Celgene), VGA-1155, BTO-956 (SRI International), ER-68203-00 (Eisai), CT-6685 (UCB), JKC-362 (Phoenix Pharmaceuticals), DMI-3798 (DMI Biosciences, Angiomate (Ipsen), ZD-6474 (AstraZeneca), CEP-5214 (Cephalon), Canstatin (Genzyme), NM-3 (Mercian), Oridigm (MediQuest Therapeutics), Exherin (Adherex), BLS-0597 (Boston Life Sciences), PTC-299 (PTC Therapeutics), NPI-2358 (Nereus Pharmaceuticals), CGP-79787 (Novartis), AEE-788 (Novartis), CKD-732 (Chong Kun Dang), CP-564959 (OSI Pharmaceuticals), CM-101 (CarboMed), CT-2584, CT3501 (Cell Therapeutics), combretastatin and analogues and derivatives thereof (Oxigene), Rebimastat (Bristol-Meyers Squibb), Dextrin 2-sulfate (ML Laboratories), Cilengitide (Merk KGaA), NSC-706704 (Pharminox), KRN- 951 (Kirin Brewery), Ukrain, NSC-631570 (Nowicky Pharma), Tecogalan sodium (Daiichi Pharmaceutical), Tz-93 (Tsumura), TBC-1635 (Encysive Pharmaceuticals), TAN-1120 (Takeda), Semaxanib (Pfizer), BDI-7800 (Biopharmacopae), SD-186, SD-983 (Bristol-Meyers Squibb), SB-223245 (GlaxoSmithKline), SC-236 (Pfizer), RWJ-590973 (Johnson and Johnson), ILX-1850 (Genzyme), SC-68488, S-836 (Pfizer), CG-55069-11 (CuraGen), Ki-23057 (Kirin Brewery), CCX-700 (Chemoentryx), Pegaptanib octasodium (Giled Sciences), ANGIOCOL (available from Biostratum Inc.), or an analogue or derivative thereof.
6701. The method of item 6694 wherein the agent is an apoptosis antagonist.
6702. The method of item 6694 wherein the agent is an apoptosis activator.
6703. The method of item 6694 wherein the agent is a beta 1 integrin antagonist.
6704. The method of item 6694 wherein the agent is a beta tubulin inhibitor. 6705. The method of item 6694 wherein the agent is a blocker of enzyme production in Hepatitis C.
6706. The method of item 6694 wherein the agent is a Bruton's tyrosine kinase inhibitor.
6707. The method of item 6694 wherein the agent is a calcineurin inhibitor.
6708. The method of item 6694 wherein the agent is a caspase 3 inhibitor.
6709. The method of item 6694 wherein the agent is a CC chemokine receptor antagonist.
6710. The method of item 6694 wherein the agent is a cell cycle inhibitor selected from the group consisting of SNS-595 (Sunesis), synthadotin, KRX-0403, and an analogue or derivative thereof.
6711. The method of item 6694 wherein the agent is a cathepsin B inhibitor.
6712. The method of item 6694 wherein the agent is a cathepsin K inhibitor, wherein the cathepsin K inhibitor is 462795 (GlaxoSmithKline), INPL-022-D6 (Amura Therapeutics), or an analogue or derivative thereof.
6713. The method of item 6694 wherein the agent is a cathepsin L inhibitor. 6714. The method of item 6694 wherein the agent is a CD40 antagonist.
6715. The method of item 6694 wherein the agent is a chemokine receptor agonist.
6716. The method of item 6694 wherein the agent is a chymase inhibitor.
6717. The method of item 6694 wherein the agent is a collagenase antagonist.
6718. The method of item 6694 wherein the agent is a CXCR antagonist.
6719. The method of item 6694 wherein the agent is a cyclin dependent kinase inhibitor selected from the group consisting of a CDK-1 inhibitor, a CDK-2 inhibitor, a CDK- 4 inhibitor, a CDK-6 inhibitor, a CAK1 inhibitor from GPC Biotech and Bristol-Myers Squibb, RGB-286199 (GPC Biotech), an anticancer agent from Astex Technology, a CAK1 inhibitor from GPC Biotech, a CDK inhibitor from Sanofi-Aventis, a CDK1/CDK2 inhibitor from Amgen, a CDK2 inhibitor from SUGEN-2 (Pfizer), a hearing loss therapy agent (Sound Pharmaceuticals), PD-0332991 (Pfizer), RGB-286199 (GPC Biotech), Ro-0505124 (Hoffmann-La Roche), a Ser/Thr kinase inhibitor from Lilly (EIi Lilly), CVT-2584 (CAS No. 199986-75-9) (CV Therapeutics), and an analogue or derivative thereof.
6720. The method of item 6694 wherein the agent is a cyclooxygenase 1 inhibitor. 6721. The method of item 6694 wherein the agent is a DHFR inhibitor.
6722. The method of item 6694 wherein the agent is a dual integrin inhibitor.
6723. The method of item 6694 wherein the agent is an elastase inhibitor.
6724. The method of item 6694 wherein the agent is an elongation factor-1 alpha inhibitor.
6725. The method of item 6694 wherein the agent is an endothelial growth factor antagonist.
6726. The method of item 6694 wherein the agent is an endothelial growth factor receptor kinase inhibitor selected from the group consisting of sorafenib tosylate (Bayer), AAL-993 (Novartis), ABP-309 (Novartis), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), E- 7080 (Eisai), EG-3306 (Ark Therapeutics), EXEL-2880 (Exelixis), GW- 654652 (GlaxoSmithKline), a KDR inhibitor from LG Life Sciences, CT-6685 and CT-6729 (UCB), KRN-633 and KRN-951 (Kirin Brewery), OSI-930 (OSI Pharmaceuticals), SP-5.2 (Supratek Pharma), SU-11657 (Pfizer), a Tie-2 antagonist (Hybrigenics), a VEGFR-2 kinase inhibitor (Bristol-Myers Squibb), XL-647 (Exelixis), a KDR inhibitor from Abbott Laboratories, sorafenib tosylate, and an analogue or derivative thereof.
6727. The method of item 6694 wherein the agent is an endotoxin antagonist. 6728. The method of item 6694 wherein the agent is an epothilone and tubulin binder.
6729. The method of item 6694 wherein the agent is an estrogen receptor antagonist.
6730. The method of item 6694 wherein the agent is an FGF inhibitor.
6731. The method of item 6694 wherein the agent is a famexyl transferase inhibitor.
6732. The method of item 6694 wherein the agent is farnesyltransferase inhibitor selected from the group of A-197574 (Abbott), a farnesyltransferase inhibitor from Servier, FPTIII (Strathclyde Institute for Drug R), LB-42908 (LG Life Sciences), Pharmaprojects No. 5063 (Genzyme), Pharmaprojects No. 5597 (Ipsen), Yissum Project No. B-1055 (Yissum), and an analogue or derivative thereof.
6733. The method of item 6694 wherein the agent is an FLT-3 kinase inhibitor.
6734. The method of item 6694 wherein the agent is an FGF receptor kinase inhibitor.
6735. The method of item 6694 wherein the agent is a fibrinogen antagonist selected from the group consisting of AUV-201 (Auvation), MG-13926 (Sanofi-Aventis), plasminogen activator (CAS No. 105913-11-9) (from Sanofi-Aventis or UCB), plasminogen activator-2 (tPA- 2) (Sanofi-Aventis), pro-urokinase (CAS No. 82657-92-9) (Sanofi-Aventis), and an analogue or derivative thereof. 6736. The method of item 6694 wherein the agent is a heat shock protein 90 antagonist selected from the group consisting of SRN-005 (Sirenade), geldanamycin, NSC-33050 (17-allylaminogeldanamycin; 17- AAG), ^-dimethylaminoethylamino-^-demethoxy-geldanamycin (17- DMAG)1 rifabutin (rifamycin XIV, f,4-didehydro-1-deoxy-1 ,4-dihydro-5'-(2- methylpropyl)-1-oxo-), and an analogue or derivative thereof.
6737. The method of item 6694 wherein the agent is a histone deacetylase inhibitor.
6738. The method of item 6694 wherein the agent is an HMGCoA reductase inhibitor selected from the group consisting of an atherosclerosis therapeutic from Lipid Sciences, ATI-16000 (ARYx Therapeutics), KS-01-019 (Kos Pharmaceuticals), Pharmaprojects No. 2197 (Sanofi-Aventi), RP 61969 (Sanofi-Aventis), and an analogue or derivative thereof.
6739. The method of item 6694 wherein the agent is an ICAM inhibitor.
6740. The method of item 6694 wherein the agent is an IL, ICE and IRAK antagonist, wherein the antagonist is a CJ-14877, CP-424174 (Pfizer), NF-61 (Negma-Lerads), and an analogue or derivative thereof.
6741. The method of item 6694 wherein the agent is an IL-2 inhibitor.
6742. The method of item 6694 wherein the agent is an immunosuppressant selected from the group consisting of teriflunomide (Sanofi Aventis), chlorsulfaquinoxalone (NSC-339004), chlorsulfaquinoxalone sulfate, CS-712 (Sankyo), ismomultin alfa (CAS No. 457913-93-8) (Akzo Nobel), antiallergics from GenPat77, antiinflammatories or AT-005 (Androclus Therapeutics), autoimmune disease therapeutics from EpiVax, BN-007 (Bone), budesonide (CAS No. 51333-22- 3) (MAP Pharmaceuticals), CO-14 (Genzyme), edratide (CAS No. 433922- 67-9) (Teva), EP-314 (Enanta), eprovafen (CAS No. 101335-99-3) (Sanofi- Aventis), HWA-131 (CAS No. 118788-41-3) (Sanofi-Aventis), immunomodulators from MerLion Pharmaceuticals, immunosuppressives from Alchemia, IPL-12 (Inflazyme), MDL-9563 (CAS No. 27086-86-8) (Sanofi-Aventis), Pharmaprojects No. 2330 (Sanofi-Aventis), Pharmaprojects No. 6426 (Abgenix), PXS-25 (Pharmaxis), rosmarinic acid (CAS No. 20283-92-5) (Sanofi-Aventis), RP 42927 or RP 54745 (CAS No. 135330-08-4) (Sanofi-Aventis), SGN-35 (Seattle Genetics), ST-1959 (Sigma-Tau), type I diabetes therapy from SYNX Pharma, UNIL-88 (Debiopharm), VP-025 (Vasogen), VR-694 (Vectura), PRTX-001 (Protalex), and an analogue or derivative thereof.
6743. The method of item 6694 wherein the agent is an IMPDH (inosine monophosphate).
6744. The method of item 6694 wherein the agent is an integrin antagonist.
6745. The method of item 6694 wherein the agent is an interleukin antagonist.
6746. The method of item 6694 wherein the agent is an inhibitor of type III receptor tyrosine kinase.
6747. The method of item 6694 wherein the agent is an irreversible inhibitor of enzyme methionine aminopeptidase type 2. 6748. The method of item 6694 wherein the agent is an isozyme selective delta protein kinase C inhibitor.
6749. The method of item 6694 wherein the agent a JAK3 enzyme inhibitor.
6750. The method of item 6694 wherein the agent is a JNK inhibitor.
6751. The method of item 6694 wherein the agent is a kinase inhibitor.
6752. The method of item 6694 wherein the agent is kinesin antagonist.
6753. The method of item 6694 wherein the agent is a kinesin antagonist.
6754. The method of item 6694 wherein the agent is a leυkotriene inhibitor and antagonist selected from the group consisting of ambicromil (CAS No. 58805-38-2) (Sanofi-Aventis), amelubant (CAS No. 346735-24-8) (Boehringer Ingelheim), DW-1141 (Dong Wha), ebselen (Daiichi Pharmaceutical), ibudilast (Kyorin), leucotriene inhibitors from Sanofi-Aventis, lymphotoxin-beta receptor (LT-D) from Biogen Idee, Pharmaprojects No. 1535 and 2728 (CAS No. 119340-33-9) (Sanofi- Aventis), R-112 (Rigel), Rev-5367 (CAS No. 92532-05-3) (Sanofi-Aventis), RG-14893 (CAS No. 141835-49-6) (Sanofi-Aventis), RG-5901-A (CAS No. 101910-24-1), 92532-23-5, RP 66153 (CAS No. 142422-79-5), RP 66364 (CAS No.186916-92-5), RP 69698 (CAS No. 141748-00-7) (Sanofi-Aventis), SC-411930 (Pfizer), SC-41930 (CAS No. 120072-59-5) (Pfizer), SC-50605 (CAS No. 138828-39-4) (Pfizer), SC-51146 (CAS No. 141059-52-1), SC- 53228 (CAS No. 153633-01-3) (Pfizer), spaglumic acid (ZY-15106) (CAS No.3106-85-2), 80619-64-3 (Novartis), tipredane (CAS No. 85197-77-9) (Bristol-Myers Squibb), U-75302 (CAS No. 119477-85-9) (Pfizer), and analogue or derivative thereof.
6755. The method of item 6694 wherein the agent is an MAP kinase inhibitor.
6756. The method of item 6694 wherein the agent is a matrix metalloproteinase inhibitor.
6757. The method of item 6694 wherein the agent is an MCP- CCR2 inhibitor.
6758. The method of item 6694 wherein the agent is an mTOR inhibitor.
6759. The method of item 6694 wherein the agent is an mTOR kinase inhibitor.
6760. The method of item 6694 wherein the agent is a microtubule inhibitor selected from the group consisting of antibody- maytansinoid conjugates from Biogen Idee, colchicines (MantiCore Pharmaceuticals), anticancer immunoconjugates from Johnson & Johnson, DIME from Octamer, gni-1f (GNI), huC242-DM4, huMy9-6-DM1 (ImmunoGen), IDN-5404 (Indena), IMO-098, IMOderm (Imotep), mebendazole (Introgen Therapeutics), microtubule poisons from Cambridge Enterprise, paclitaxel such as LOTAX from Aphios (CAS No. 33069-62-4), Genexol-PM from Samyang, Pharmaprojects No. 6383 (Tapestry Pharmaceuticals), RPR-112378 (Sanofi-Aventis), SGN-75 (Seattle Genetics), SPL-7435 (Starpharma), SSR-250411 (Sanofi-Aventis), trastuzumab-DM1 (Genentech), vinorelbine, and an analogue or derivative thereof.
6761. The method of item 6694 wherein the agent is an MIF inhibitor.
6762. The method of item 6694 wherein the agent is an MMP inhibitor.
6763. The method of item 6694 wherein the agent is a neurokinin (NK) antagonist selected from the group consisting of anthrotainin (CAS No. 148084-40-6) (Sanofi-Aventis), an IBS thereapeutic from ArQuIe, MDL-105212A (CAS No. 167261-60-1) (Ssanofi-Aventis), Pharmaprojects No. 2744, 3258 (CAS No. 139167-47-8) 4006, 4201 , or 5986 (Sanofi-Aventis), RP 67580 (CAS No. 135911-02-3), SR-144190 (CAS No. 201152-86-5), SSR-240600, SSR-241586 (Sanofi-Aventis), TKA-457 (Novartis), vestipitant mesylate (CAS No. 334476-64-1) (GlaxoSmithKline), Win-64821 (Sanofi-Aventis), PRX-96026 (Predix Pharmaceuticals), and an analogue or derivative thereof.
6764. The method of item 6694 wherein the agent is an NF kappa B inhibitor selected from the group consisting of AVE-0545 or AVE- 0547 (Sanofi-Aventis), bortezomib (CAS No. 179324-69-7) (Millennium Pharmaceuticals), dexanabinol (CAS No. 112924-45-5) (Pharmos), dexlipotam (Viatris), Pharmaprojects No. 6283 (INDRA) (OXiGENE), IPL- 576092 (CAS No. 137571-30-3) (Inflazyme), NFKB decoy (Corgentech), NFKB decoy oligo (AnGes MG), NFKB's from Ariad, osteoporosis treatments or S5 (F005) from Fulcrum Pharmaceuticals, P61 (Phytopharm), R-flurbiprofen (CAS No. 5104-49-4) (Encore Pharmaceuticals), and an analogue or derivative thereof. 6765. The method of item 6694 wherein the agent is a nitric oxide agonist.
6766. The method of item 6694 wherein the agent is an ornithine decarboxylase inhibitor.
6767. The method of item 6694 wherein the agent is a p38 MAP kinase inhibitor selected from the group consisting of AZD-6703 (AstraZeneca), JX-401 (Jexys Pharmaceuticals), BMS-2 (Bristol-Myers Squibb), a p38 MAP kinase inhibitor from Novartis, a p38-alpha MAP kinase inhibitor from Amphora, Pharmaprojects No. 5704 (Pharmacopeia), RPR- 200765A (Sanofi-Aventis), SD-282 (Johnson & Johnson), TAK-715 (Takeda), and an analogue or derivative thereof.
6768. The method of item 6694 wherein the agent is a palmitoyl-protein thioesterase inhibitor.
6769. The method of item 6694 wherein the agent is a PDGF receptor kinase inhibitor selected from the group consisting of AAL-993, AMN-107, or ABP-309 (Novartis), AMG-706 (Amgen), BAY-57-9352 (Bayer), CDP-860 (UCB), E-7080 (Eisai), imatinib (CAS No. 152459-95-5) (Novartis), OSI-930 (OSI Pharmaceuticals), RPR-127963E (Sanofi-Aventis), RWJ-540973 (Johnson & Johnson), sorafenib tosylate (Bayer), SU-11657 (Pfizer), tandutinib (CAS No. 387867-13-2) (Millennium Pharmaceuticals), vatalanib (Novartis), ZK-CDK (Schering AG), and an analogue or derivative thereof.
6770. The method of item 6694 wherein the agent is (-)- halofenate (Metabolex), AMG-131 (Amgen), antidiabetics from Japan Tobacco, AZD-4619, AZD-8450, AZD-8677 (AstraZeneca), DRF-10945, balaglitazone (Dr Reddy's), CS-00088, CS-00098 (Chipscreen Biosciences), E-3030 (Eisai), etalocib (CAS No. 161172-51-6) (EIi Lilly), GSK-641597 (Ligand), GSK-677954 (GlaxoSmithKline), GW-409544 (Ligand), GW- 590735 (GlaxoSmithKline), K-111 (Hoffmann-La Roche), LY-518674 (EIi Lilly), LY-674 (Ligand), LY-929 (Ligand), MC-3001 , MC-3002 (MaxoCore Pharmaceuticals), metformin HCI + pioglitazone (CAS No. 1115-70-4 and 112529-15-4), ACTOPLUS MET from Andrx), muraglitazar (CAS No. 331741-94-7) (Bristol-Myers Squibb), naveglitazar (Ligand), oleoylethanolamide (Kadmus Pharmaceuticals), ONO-5129, pioglitazone hydrochloride (CAS No. 111025-46-8 and 112529-15-4) (Takeda), PLX-204 (Plexxikon), PPAR agonists from Genfit, PPAR delta agonists from EIi Lilly, PPAR-alpha agonists from CrystalGenomics, PPAR-gamma modulators and PPAR-β modulators from CareX, rosiglitazone maleate (CAS No. 122324- 73-4 or 155141-29-0) (GlaxoSmithKline), rosiglitazone maleate/glimepir (CAS No. 155141-29-0 and 93479-97-1), AVANDARYL, rosiglitazone maleate/metformin extend (CAS No. 155141-29-0 and 657-24-9), AVANDAMET, rosiglitazone maleate+metformin, AVANDAMET (GlaxoSmithKline), tesaglitazar (AstraZeneca), and an analogue or derivative thereof.
6771. The method of item 6694 wherein the agent is a phosphatase inhibitor.
6772. The method of item 6694 wherein the agent is a phosphodiesterase (PDE) inhibitor selected from the group consisting of avanafil (Tanabe Seiyaku), dasantafil (CAS No. 569351-91-3) (Schering- Plough), A-906119 (CAS No. 134072-58-5), DL-850 (Sanofi-Aventis), GRC- 3015, GRC-3566, GRC-3886 (Glenmark), HWA-153 (CAS No. 56395-66-5) (Sanofi-Aventis), hydroxypumafentrine (Altana), IBFB-130011, IBFB-14-016, IBFB-140301 , IBFB-150007, IBFB-211913 (IBFB Pharma), L-826141 (Merck & Co), medorinone (CAS No. 88296-61-1) (Sanofi-Aventis), MEM- 1917 (Memory Pharmaceuticals), ND-1251 (Neuro3d), PDE inhibitors from ICOS1 PDE IV inhibitors from Memory Pharmaceuticals and CrystalGenomics, Pharmaprojects No. 2742 and 6141 (Sanofi-Aventis), QAD-171 (Novartis), RHC-2963 (CAS No. 76993-12-9 and 76993-14-1), RPR-117658, RPR-122818 derivatives, SR-24870 , and RPR-132294 (Sanofi-Aventis), SK-350 (ln2Gen), stroke therapy agents from deCODE Genetics, TAS-203 (Taiho), tofimilast (CAS No. 185954-27-2) (Pfizer), UK- 371800 (Pfizer), WIN-65579 (CAS No. 158020-82-7) (Sanofi-Aventis), IBFB- 130020 (IBFB Pharma), OPC-6535 (CAS No. 145739-56-6) (Otsuka), a phosphodiesterase III inhibitor, enoximone, a phosphodiesterase IV inhibitor, fosfosal, Atopik (Barrier Therapeutics), triflusal, a phosphodiesterase V inhibitor, and an analogue or derivative thereof.
6773. The method of item 6694 wherein the agent is a PKC inhibitor.
6774. The method of item 6694 wherein the agent is a platelet activating factor antagonist.
6775. The method of item 6694 wherein the agent is a platelet-derived growth factor receptor kinase inhibitor.
6776. The method of item 6694 wherein the agent is a prolyl hydroxylase inhibitor.
6777. The method of item 6694 wherein the agent is a polymorphonuclear neutrophil inhibitor.
6778. The method of item 6694 wherein the agent is a protein kinase B inhibitor. 6779. The method of item 6694 wherein the agent is a protein kinase C stimulant.
6780. The method of item 6694 wherein the agent is a purine nucleoside analogue.
6781. The method of item 6694 wherein the agent is a purinoreceptor P2X antagonist.
6782. The method of item 6694 wherein the agent is a Raf kinase inhibitor.
6783. The method of item 6694 wherein the agent is a reversible inhibitor of ErbB1 and ErbB2.
6784. The method of item 6694 wherein the agent is a ribonucleoside triphosphate reductase inhibitor.
6785. The method of item 6694 wherein the agent is an SDF- 1 antagonist.
6786. The method of item 6694 wherein the agent is a sheddase inhibitor.
6787. The method of item 6694 wherein the agent is an SRC inhibitor.
6788. The method of item 6694 wherein the agent is a stromelysin inhibitor. 6789. The method of item 6694 wherein the agent is an Syk kinase inhibitor.
6790. The method of item 6694 wherein the agent is a telomerase inhibitor.
6791. The method of item 6694 wherein the agent is a TGF beta inhibitor selected from the group consisting of pirfenidone (CAS No. 53179-13-8) (MARNAC)1 tranilast (CAS No. 53902-12-8) (Kissei), 1N-1130 (ln2Gen), mannose-6-phosphate (BTG)1 TGF-β antagonists from Inflazyme (Pharmaprojects No. 6075), TGF-β antagonists from Sydney, non-industrial source), TGF-β I receptor kinase inhibitors from EIi Lilly, TGF-β receptor inhibitors from Johnson & Johnson, and an analogue or derivative thereof.
6792. The method of item 6694 wherein the agent is a TNFα antagonist or TACE inhibitor selected from the group consisting of adalimumab (CAS No. 331731-18-1) (Cambridge Antibody Technology), AGIX-4207 (AtheroGenics), AGT-1 (Advanced Biotherapy), an antiinflammatory from Borean Pharma, Cellzome, or Paradigm Therapeutics, anti-inflammatory vaccine (TNF-alpha kinoid) from Neovacs, humanized anti-TNF antibody or an anti-TNF MAb (CB0006) Celltech (UCB), apratastat (CAS No. 287445-51-0) (Wyeth), BMS-561392 (Bristol-Myers Squibb), BN- 006 (Bone), certolizumab pegol (CAS No. 428863-50-7 or CH-138 (UCB), cilomilast (CAS No. 153259-65-5) (GlaxoSmithKline), CR-1 (Nuada Pharmaceuticals), CRx-119 (CombinatoRx), D-5410 (UCB), dacopafant (CAS No. 125372-33-0) (Sanofi-Aventis), dersalazine (CAS No. 188913-57- 7/188913-58-8) (Uriach), etanercept (CAS No. 185243-69-0) (Amgen), ethyl pyruvate (Critical (Critical Therapeutics), golimumab (CAS No. 476181-74-5) (Johnson & Johnson), hormono-immunotherapy from Ipsen, CDP571 (e.g., Humicade from UCB), IC-485 (ICOS), infliximab (CAS No. 170277-31-3) (Johnson & Johnson), IP-751 (Manhattan Pharmaceuticals), ISIS-104838 (CAS No. 250755-32-9) (ISIS Pharmaceuticals), lenalidomide (CAS No. 191732-72-6) (Celgene), lentinan (CAS No. 37339-90-5) (Ajinomoto), MDL- 201112 (CAS No. 142130-73-2) (Sanofi-Aventis), medroxyprogesterone (CAS No. 520-85-4) (InKine Pharmaceutical), N-acetylcysteine (CAS No. 616-91-1) (Zambon), NBE-P2 (DIREVO Biotech), nerelimomab (CAS No. 162774-06-3) (Chiron), OM-294DP (OM PHARMA), onercept (CAS No. 199685-57-9) (Yeda), PASSTNF-alpha (Verigen), pentoxifylline or oxypentifylline (Sanofi-Aventis), Pharmaprojects No. 4091 , 4241, 4295, or 4488 (Sanofi-Aventis), Pharmaprojects No. 5480 (Amgen), Pharmaprojects No. 6749 (Cengent), pirfenidone (CAS No. 53179-13-8) (MARNAC), RPR- 132294 (Sanofi-Aventis), S5 (F002) (Fulcrum Pharmaceuticals), simvastatin (CAS No. 79902-63-9) (Merck & Co), STA-6292 (Synta Pharmaceuticals), tacrolimus (CAS No. 104987-11-3) (Fujisawa LifeCycle Pharma), talactoferrin alfa (CAS No. 308240-58-6) (Agennix), thalidomide (CAS No. 50-35-1) (Celgene), TNF antagonists form ProStrakan, and Synergen, TNF inhibitors (Amgen), TNF-alpha antagonists from Dynavax Technologies and Jerina AG (Germany), TNF-alpha inhibitors from IBFB Pharma and Xencor (Xencor), torbafylline (CAS No. 105102-21-4) (Sanofi-Aventis), UR-1505 (Uriach), VT-346 (Viron Therapeutics), YSIL6 (Y's Therapeutics), YSTH2 (Y's Therapeutics), NPI-1302a-3 (Nereus Pharmaceuticals, a TNF antagonist from Jerina AG (Germany), dersalazine, and an analogue or derivative thereof.
6793. The method of item 6694 wherein the agent is a Toll receptor inhibitor.
6794. The method of item 6694 wherein the agent is a tubulin antagonist.
6795. The method of item 6694 wherein the agent is a tyrosine kinase inhibitor selected from the group consisting of SU-011248, SUTENT from Pfizer Inc. (New York, NY), BMS-354825, PN-355 (Paracelsian Pharmaceuticals), AGN-199659 (Allergan), AAL-993 or ABP- 309 (Novartis), adaphostin (NIH), AEE-788 (Novartis), AG-013736 (OSI Pharmaceuticals), AG-13736 (Pfizer), ALT-110 (Alteris Therapeutics), AMG- 706 (Amgen), anticancer MAbs from Xencor, anti-EGFrvlll MAbs from Abgenix, anti-HER2 MAb from Abiogen, AZD-2171 or AZD-9935 (AstraZeneca), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), CEP-5214 (Cephalon), CEP-7055 (Cephalon), cetuximab (ImClone Systems), CHIR-200131 and CHIR-258 (Chiron), CP-547632 (OSI Pharmaceuticals), CP-724714 (Pfizer), CT-301 (Creabilis Therapeutics), D- 69491 (Baxter International), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EGFR/ErbB2 inhibitors from Array BioPharma, erlotinib (CAS No. 183319- 69-9) (OSI Pharmaceuticals), EXEL-2880 (Exelixis), FK-778 (Sanofi- Aventis) , gefitinib (CAS No. 184475-35-2) (AstraZeneca), GW-2286 or GW- 654652 (GlaxoSmithKline), her2/neu antigen from AlphaVax, HER-2/neu inhibitor from Generex, Herzyme (Medipad) (Sirna Therapeutics), HKI-272 (Wyeth), HuMax-EGFr (Genmab), idronoxil (CAS No. 81267-65-4) (Novogen), IGF-1 inhibitors from Ontogen, IMC-11F8 (ImClone Systems), kahalalide F (CAS No. 149204-42-2) (PharmaMar), KDR inhibitor from LG Life Sciences, KDR inhibitors from Abbott Laboratories, KDR kinase inhibitors (UCB), Kdr kinase inhibitors from Merck & Co, KRN-633 and KRN- 951 (Kirin Brewery), KSB-102 (Xenova), lapatinib ditosylate (CAS No. 388082-78-8) (GlaxoSmithKline), matuzumab (Merck KGaA), MDX-214 (Medarex), ME-103 (Pharmexa), MED-A300 (Gerolymatos), MNAC-13 (Lay Line Genomics), nimotuzumab (Center of Molecular Immunology), NSC- 330507 or NSC-707545 (NIH), NV-50 (Novogen), OSI-930 (OSI Pharmaceuticals), panitumumab (Abgenix), pelitinib (CAS No. 287933-82-7) (Wyeth), pertuzumab (CAS No. 380610-27-5) (Genentech), Pharmaprojects No. 3985 (Sanofi-Aventis), prostate cancer therapeutics from Sequenom (SQPC35, SQPC36, SQPC90), removab and remoxab (Trion Pharma), RG- 13022 (CAS No. 136831-48-6), RG-13291 (CAS No. 138989-50-1), or RG- 14620 (CAS No. 136831-49-7) (Sanofi-Aventis), RM-6427 (Romark), RNAi breast cancer therapy from Benitec, RP 53801 (CAS No. 125882-88-4) (Sanofi-Aventis), sorafenib tosylate (Bayer), SU-11657 (Pfizer), Tie-2 antagonists from Semaia (Hybrigenics), Tie-2 inhibitors from Ontogen, trastuzumab (CAS No. 180288-69-1) (Genentech), tyrosine kinase inhibitors from Sanofi-Aventis, U3-1287, U3-1565, U3-1784, U3-1800 (U3 Pharma), vatalanib (Novartis), VEGFR-2 kinase inhibitor from Bristol-Myers Squibb, XL-647 (Exelixis), ZD-6474 (AstraZeneca), ZK-CDK (Schering AG), an EGFR tyrosine kinase inhibitor, EKB-569 (Wyeth), and an analogue or derivative thereof.
6796. The method of item 6694 wherein the agent is a VEGF inhibitor.
6797. The method of item 6694 wherein the agent is a vitamin D receptor agonist.
6798. The method of item 6694 wherein the agent is ZD-6474 (an angiogenesis inhibitor).
6799. The method of item 6694 wherein the agent is AP- 23573 (an mTOR inhibitor).
6800. The method of item 6694 wherein the agent is synthadotin (a tubulin antagonist).
6801. The method of item 6694 wherein the agent is S-0885 (a collagenase inhibitor).
6802. The method of item 6694 wherein the agent is aplidine (an elongation factor-1 alpha inhibitor). 6803. The method of item 6694 wherein the agent is ixabepilone (an epithilone).
6804. The method of item 6694 wherein the agent is IDN- 5390 (an angiogenesis inhibitor).
6805. The method of item 6694 wherein the agent is SB- 2723005 (an angiogenesis inhibitor).
6806. The method of item 6694 wherein the agent is ABT-518 (an angiogenesis inhibitor).
6807. The method of item 6694 wherein the agent is combretastatin (an angiogenesis inhibitor).
6808. The method of item 6694 wherein the agent is anecortave acetate (an angiogenesis inhibitor).
6809. The method of item 6694 wherein the agent is SB- 715992 (a kinesin antagonist).
6810. The method of item 6694 wherein the agent is temsirolimus (an mTOR inhibitor).
6811. The method of item 6694 wherein the agent is adalimumab (a TNFα antagonist).
6812. The method of item 6694, wherein the composition comprises a polymer. 6813. The method of item 6694, wherein the composition comprises a polymer, and the polymer is, or comprises, a copolymer.
6814. The method of item 6694, wherein the composition comprises a polymer, and the polymer is, or comprises, a block copolymer.
6815. The method of item 6694, wherein the composition comprises a polymer, and the polymer is, or comprises, a random copolymer.
6816. The method of item 6694, wherein the composition comprises a polymer, and the polymer is, or comprises, a biodegradable polymer.
6817. The method of item 6694, wherein the composition comprises a polymer, and the polymer is, or comprises, a nonbiodegradable polymer.
6818. The method of item 6694, wherein the composition comprises a polymer, and the polymer is, or comprises, a hydrophilic polymer.
6819. The method of item 6694, wherein the composition comprises a polymer, and the polymer is, or comprises, a hydrophobic polymer.
6820. The method of item 6694, wherein the composition comprises a polymer, and the polymer is, or comprises, a polymer having hydrophilic domains. 6821. The method of item 6694, wherein the composition comprises a polymer, and the polymer is, or comprises, a polymer having hydrophobic domains.
6822. The method of item 6694, wherein the composition comprises a polymer, and the polymer is, or comprises, a non-conductive polymer.
6823. The method of item 6694, wherein the composition comprises a polymer, and the polymer is, or comprises, an elastomer.
6824. The method of item 6694, wherein the composition comprises a polymer, and the polymer is, or comprises, a hydrogel.
6825. The method of item 6694, wherein the composition comprises a polymer, and the polymer is, or comprises, a silicone polymer.
6826. The method of item 6694, wherein the composition comprises a polymer, and the polymer is, or comprises, a hydrocarbon polymer.
6827. The method of item 6694, wherein the composition comprises a polymer, and the polymer is, or comprises, a styrene-derived polymer.
6828. The method of item 6694, wherein the composition comprises a polymer, and the polymer is, or comprises, a butadiene-derived polymer.
6829. The method of item 6694, wherein the composition comprises a polymer, and the polymer is, or comprises, a macromer. 6830. The method of item 6694, wherein the composition comprises a polymer, and the polymer is, or comprises, a poly(ethylene glycol) polymer.
6831. The method of item 6694, wherein the composition comprises a polymer, and the polymer is, or comprises, an amorphous polymer.
6832. The method of item 6694, wherein the composition further comprises a second pharmaceutically active agent.
6833. The method of item 6694, wherein the composition further comprises an anti-inflammatory agent.
6834. The method of item 6694, wherein the composition further comprises an agent that inhibits infection.
6835. The method of item 6694, wherein the composition further comprises an anthracycline.
6836. The method of item 6694, wherein the composition further comprises doxorubicin.
6837. The method of item 6694 wherein the composition further comprises mitoxantrone.
6838. The method of item 6694 wherein the composition further comprises a fluoropyrimidine.
6839. The method of item 6694, wherein the composition further comprises 5-fluorouracil (5-FU). 6840. The method of item 6694, wherein the composition further comprises a folic acid antagonist.
6841. The method of item 6694, wherein the composition further comprises methotrexate.
6842. The method of item 6694, wherein the composition further comprises a podophylotoxin.
6843. The method of item 6694, wherein the composition further comprises etoposide.
6844. The method of item 6694, wherein the composition further comprises camptothecin.
6845. The method of item 6694, wherein the composition further comprises a hydroxyurea.
6846. The method of item 6694, wherein the composition further comprises a platinum complex.
6847. The method of item 6694, wherein the composition further comprises cisplatin.
6848. The method of item 6694 wherein the composition further comprises an anti-thrombotic agent.
6849. The method of item 6694, wherein the composition further comprises a visualization agent. 6850. The method of item 6694, wherein the composition further comprises a visualization agent, and the visualization agent is a radiopaque material, wherein the radiopaque material comprises a metal, a halogenated compound, or a barium containing compound.
6851. The method of item 6694, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, barium, tantalum, or technetium.
6852. The method of item 6694, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, an MRI responsive material.
6853. The method of item 6694, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, a gadolinium chelate.
6854. The method of item 6694, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, iron, magnesium, manganese, copper, or chromium.
6855. The method of item 6694, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, iron oxide compound.
6856. The method of item 6694, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, a dye, pigment, or colorant.
6857. The method of item 6694 wherein the agent is released in effective concentrations from the composition comprising the agent by diffusion over a period ranging from the time of administration to about 90 days.
6858. The method of item 6694 wherein the agent is released in effective concentrations from the composition comprising the agent by erosion of the composition over a period ranging from the time of administration to about 90 days.
6859. The method of item 6694 wherein the composition further comprises an inflammatory cytokine.
6860. The method of item 6694 wherein the composition further comprises an agent that stimulates cell proliferation.
6861. The method of item 6694 wherein the composition further comprises a polymeric carrier.
6862. The method of item 6694 wherein the composition is in the form of a gel, paste, or spray.
6863. The method of item 6694 wherein the implant is partially constructed with the agent or the composition.
6864. The method of item 6694 wherein the implant is fully constructed with the agent or the composition.
6865. The method of item 6694 wherein the implant is impregnated with the agent or the composition.
6866. The method of item 6694, wherein the agent or the composition forms a coating, and the coating directly contacts the implant. 6867. The method of item 6694, wherein the agent or the composition forms a coating, and the coating indirectly contacts the implant.
6868. The method of item 6694 wherein the agent or the composition forms a coating, and the coating partially covers the implant.
6869. The method of item 6694, wherein the agent or the composition forms a coating, and the coating completely covers the implant.
6870. The method of item 6694 wherein the agent or the composition is located within pores or holes of the implant.
6871. The method of item 6694 wherein the agent or the composition is located within a channel, lumen, or divet of the implant.
6872. The method of item 6694 wherein the implant further comprising an echogenic material.
6873. The method of item 6694 wherein the implant further comprises an echogenic material, wherein the echogenic material is in the form of a coating.
6874. The method of item 6694 wherein the implant is sterile.
6875. The method of item 6694 wherein the agent is delivered from the implant, wherein the agent is released into tissue in the vicinity of the implant after deployment of the implant.
6876. The method of item 6694 wherein the agent is delivered from the implant, wherein the agent is released into tissue in the vicinity of the implant after deployment of the implant, wherein the tissue is connective tissue.
6877. The method of item 6694 wherein the agent is delivered from the implant, wherein the agent is released into tissue in the vicinity of the implant after deployment of the implant, wherein the tissue is muscle tissue.
6878. The method of item 6694 wherein the agent is delivered from the implant, wherein the agent is released into tissue in the vicinity of the implant after deployment of the implant, wherein the tissue is nerve tissue.
6879. The method of item 6694 wherein the agent is delivered from the implant, wherein the agent is released into tissue in the vicinity of the implant after deployment of the implant, wherein the tissue is epithelium tissue.
6880. The method of item 6694 wherein the agent is delivered from the implant, wherein the agent is released in effective concentrations from the implant over a period ranging from the time of deployment of the implant to about 1 year.
6881. The method of item 6694 wherein the agent is delivered from the implant, wherein the agent is released in effective concentrations from the implant over a period ranging from about 1 month to 6 months.
6882. The method of item 6694 wherein the agent is delivered from the implant, wherein the agent is released in effective concentrations from the implant over a period ranging from about 1 - 90 days. 6883. The method of item 6694 wherein the agent is delivered from the implant, wherein the agent is released in effective concentrations from the implant at a constant rate.
6884. The method of item 6694 wherein the agent is delivered from the implant, wherein the agent is released in effective concentrations from the implant at an increasing rate.
6885. The method of item 6694 wherein the agent is delivered from the implant, wherein the agent is released in effective concentrations from the implant at a decreasing rate.
6886. The method of item 6694 wherein the agent is delivered from the implant, wherein the implant comprises about 0.01 Dg to about 10 Dg of the agent.
6887. The method of item 6694 wherein the agent is delivered from the implant, wherein the implant comprises about 10 Dg to about 10 mg of the agent.
6888. The method of item 6694 wherein the agent is delivered from the implant, wherein the implant comprises about 10 mg to about 250 mg of the agent.
6889. The method of item 6694 wherein the agent is delivered from the implant, wherein the implant comprises about 250 mg to about
1000 mg of the agent.
6890. The method of item 6694 wherein the agent is delivered from the implant, wherein the implant comprises about 1000 mg to about 2500 mg of the agent. 6891. The method of item 6694 wherein the agent is delivered from the implant, wherein a surface of the implant comprises less than 0.01 Dg of the agent per mm2 of implant surface to which the agent is applied.
6892. The method of item 6694 wherein the agent is delivered from the implant, wherein a surface of the implant comprises about 0.01 Dg to about 1 Dg of the agent per mm2 of implant surface to which the agent is applied.
6893. The method of item 6694 wherein the agent is delivered from the implant, wherein a surface of the implant comprises about 1 Dg to about 10 Dg of the agent per mm2 of implant surface to which the agent is applied.
6894. The method of item 6694 wherein the agent is delivered from the implant, wherein a surface of the implant comprises about 10 Dg to about 250 Dg of the agent per mm2 of implant surface to which the agent is applied.
6895. The method of item 6694 wherein the agent is delivered from the implant, wherein a surface of the implant comprises about 250 Dg to about 1000 Dg of the agent per mm2 of implant surface to which the agent is applied.
6896. The method of item 6694 wherein the agent is delivered from the implant, wherein a surface of the implant comprises about 1000 Dg to about 2500 Dg of the agent per mm2 of implant surface to which the agent is applied.
6897. The method of item 6694, wherein the implant further comprises a coating, and the coating is a uniform coating. 6898. The method of item 6694, wherein the implant further comprises a coating, and the coating is a non-uniform coating.
6899. The method of item 6694, wherein the implant further comprises a coating, and the coating is a discontinuous coating.
6900. The method of item 6694, wherein the implant further comprises a coating, and the coating is a patterned coating.
6901. The method of item 6694, wherein the implant further comprises a coating, and the coating has a thickness of 100 Dm or less.
6902. The method of item 6694, wherein the implant further comprises a coating, and the coating has a thickness of 10 Dm or less.
6903. The method of item 6694, wherein the implant further comprises a coating, and the coating adheres to the surface of the implant upon deployment of the implant.
6904. The method of item 6694, wherein the implant further comprises a coating, and the coating is stable at room temperature for a period of at least 1 year.
6905. The method of item 6694, wherein the implant further comprises a coating, and the agent is present in the coating in an amount ranging between about 0.0001% to about 1% by weight.
6906. The method of item 6694, wherein the implant further comprises a coating, and the agent is present in the coating in an amount ranging between about 1% to about 10% by weight. 6907. The method of item 6694, wherein the implant further comprises a coating, and the agent is present in the coating in an amount ranging between about 10% to about 25% by weight.
6908. The method of item 6694, wherein the implant further comprises a coating, and the agent is present in the coating in an amount ranging between about 25% to about 70% by weight.
6909. The method of item 6694, wherein the implant further comprises a coating, and the coating comprises a polymer.
6910. The method of item 6694, wherein the implant comprises a first coating having a first composition and a second coating having a second composition.
6911. The method of item 6694, wherein the implant comprises a first coating having a first composition and a second coating having a second composition, wherein the first composition and the second composition are different.
6912. A method as in any one of items 6694-6911 , wherein the device is a lacrimal duct stent.
6913. A method as in any one of items 6694-6911, wherein the device is an Eustachian tube stent.
6914. A method as in any one of items 6694-6911 , wherein the device is a nasal stent.
6915. A method as in any one of items 6694-6911, wherein the device is a sinus stent. 6916. A method for inhibiting scarring comprising placing a device that comprises an ear ventilation tube (e.g., an implant) and an anti- scarring agent or a composition comprising an anti-scarring agent into an animal host, wherein the agent inhibits scarring.
6917. The method of item 6916 wherein the agent is an adensosine A2A receptor antagonist.
6918. The method of item 6916 wherein the agent is an AKT inhibitor.
6919. The method of item 6916 wherein the agent is an alpha 2 integrin antagonist, wherein the alpha 2 integrin antagonist is Pharmaprojects No. 5754 (Merck KgaA).
6920. The method of item 6916 wherein the agent is an alpha 4 integrin antagonist.
6921. The method of item 6916 wherein the agent is an alpha 7 nicotinic receptor agonist.
6922. The method of item 6916 wherein the agent is an angiogenesis inhibitor selected from the group consisting of AG-12,958 (Pfizer), ATN-161 (Attenuon LLC), neovastat, an angiogenesis inhibitor from Jerina AG (Germany), NM-3 (Mercian), VGA-1155 (Taisho), FCE-26644 (Pfizer), FCE-26950 (Pfizer), FPMA (Meiji Daries), FFM 11142 (Fujisawa), GGTI-298, GM-1306 (Ligand), GPA-1734 (Novartis), NNC-47-0011 (Novo Nordisk), herbamycin (Nippon Kayaku), lenalidomide (Celegene), IP-10 (NIH), ABT-828 (Abbott), KIN-841 (Tokushima University, Japan), SF-1126 (Semafore Pharmaceuticals), laminin technology (NIH), CHIR-258 (Chiron), NVP-AEW541 (Novartis), NVP-AEW541 (Novartis), Vt16907 (Alchemia), OXI-8007 (Oxigene), EG-3306 (Ark Therapeutics), Maspin (Arriva), ABT- 567 (Abbott), PPI-2458 (Praecis Pharmaceuticals), CC-5079, CC-4089 (Celgene), HIF-1 alpha inhibitors (Xenova), S-247 (Pfizer), AP-23573 (Ariad), AZD-9935 (Astra Zeneca), mebendazole (Introgen Therapeutics), MetAP-2 inhibitors (GlaxoSmithKline), AG-615 (Angiogene Pharmaceuticals), Tie-2 antagonists (Hybrigenics), NC-381 , CYC-381, NC-169, NC-219, NC-383, NC-384, NC-407 (Lorus Therapeutics), ATN-224 (Attenuon), ON-01370 (Onconova), Vitronectin antagonists (Amgen), SDX-103 (Salmedix), Vitronectin antagonists (Shire), CHP (Riemser), TEK (Amgen), Anecortave acetate (Alcon), T46.2 (Matrix Therapeutics), HG-2 (Heptagen), TEM antagonists (Genzyme), Oxi-4500 (Oxigene), ATN-161 (Attenuon), WX-293 (Wilex), M-2025 (Metris Therapeutics), Alphastatin (BioActa), YH-16 (Yantai Rongchang), BIBF-1120 (Boehringer Ingelheim), BAY-57-9352 (Bayer), AS- 1404 (Cancer Research Technology), SC-77964 (Pfizer), glycomimetics (BioTie Therapies), TIE-2 Inhibitors (Ontogen), DIMI, Octamer (Octamer), ABR-215050 (Active Biotech), ABT-518 (Abbott), KDR inhibitors (Abbott), BSF-466895 (Abbott), SCH-221153 (Schering-Plough), DAC:antiangiogenic (ConjuChem), TFPI (EntreMed), AZD-2171 (Astra-Zenaca), CDC-394 (Celgene), LY290293 (EIi Lilly), IDN-5390 (Indena), Kdr Kinase Inhibitors (Merck), CT-113020, CT-116433, CT-116563, CT-31890, CT-32228) (Cell Therapeutics), A-299620 (Abbott), TWEAK Inhibitor (Amgen), VEGF modulators (Johnson and Johnson), Tum-N53, tumstatin (Genzyme), Thios- 1 , Thios-2 (Thios Pharmaceuticals), MV-6401 (Miravant Medical Technologies), Spisulosine (PharmaMar), CEP-7055 (Cephalon), AUV-201 (Auvation), LM-609 (EIi Lilly), SKF-106615 (AnorMED), Oglufanide disodium (Cytran), BW-114 (Pharminox), Calreticulin (NIH), WX-678 (Wilex), SD-7784 (Pfizer), WX-UK1 (Wiiex), SH-268 (Schering AG), 2-Me-PGA (Celgene), S- 137 (Pfizer), ZD-6126 (Angiogene Pharmaceuticals), SG-292 (SignalGen), Benefin (Lane Labs), A6, A36 (Angstrom), SB-2723005 (GlaxoSmithKline), SC-7 (Cell Therapeutics), ZEN-014 (AEterna Zentaris), 2-methoxyestradiol (EntreMed), NK-130119 (Nippon Kayaku), CC-10004 (Celgene), AVE- 8062A (Ajinomoto), Tacedinaline (Pfizer), Actinonin (Tokyo Metropolitan Institute of Medical Science), Lenalidomide (Celgene), VGA-1155, BTO-956 (SRI International), ER-68203-00 (Eisai), CT-6685 (UCB), JKC-362 (Phoenix Pharmaceuticals), DMI-3798 (DMI Biosciences, Angiomate (Ipsen), ZD-6474 (AstraZeneca), CEP-5214 (Cephalon), Canstatin (Genzyme), NM-3 (Mercian), Oridigm (MediQuest Therapeutics), Exherin (Adherex), BLS-0597 (Boston Life Sciences), PTC-299 (PTC Therapeutics), NPI-2358 (Nereus Pharmaceuticals), CGP-79787 (Novartis), AEE-788 (Novartis), CKD-732 (Chong Kun Dang), CP-564959 (OSI Pharmaceuticals), CM-101 (CarboMed), CT-2584, CT3501 (Cell Therapeutics), combretastatin and analogues and derivatives thereof (Oxigene), Rebimastat (Bristol-Meyers Squibb), Dextrin 2-sulfate (ML Laboratories), Cilengitide (Merk KGaA), NSC-706704 (Pharminox), KRN- 951 (Kirin Brewery), Ukrain, NSC-631570 (Nowicky Pharma), Tecogalan sodium (Daiichi Pharmaceutical), Tz-93 (Tsumura), TBC-1635 (Encysive Pharmaceuticals), TAN-1120 (Takeda), Semaxanib (Pfizer), BDI-7800 (Biopharmacopae), SD-186, SD-983 (Bristol-Meyers Squibb), SB-223245 (GlaxoSmithKline), SC-236 (Pfizer), RWJ-590973 (Johnson and Johnson), ILX-1850 (Genzyme), SC-68488, S-836 (Pfizer), CG-55069-11 (CuraGen), Ki-23057 (Kirin Brewery), CCX-700 (Chemoentryx), Pegaptanib octasodium (Giled Sciences), ANGIOCOL (available from Biostratum Inc.), or an analogue or derivative thereof.
6923. The method of item 6916 wherein the agent is an apoptosis antagonist.
6924. The method of item 6916 wherein the agent is an apoptosis activator.
6925. The method of item 6916 wherein the agent is a beta 1 integrin antagonist. 6926. The method of item 6916 wherein the agent is a beta tubulin inhibitor.
6927. The method of item 6916 wherein the agent is a blocker of enzyme production in Hepatitis C.
6928. The method of item 6916 wherein the agent is a Bruton's tyrosine kinase inhibitor.
6929. The method of item 6916 wherein the agent is a calcineurin inhibitor.
6930. The method of item 6916 wherein the agent is a caspase 3 inhibitor.
6931. The method of item 6916 wherein the agent is a CC chemokine receptor antagonist.
6932. The method of item 6916 wherein the agent is a cell cycle inhibitor selected from the group consisting of SNS-595 (Sunesis), synthadotin, KRX-0403, and an analogue or derivative thereof.
6933. The method of item 6916 wherein the agent is a cathepsin B inhibitor.
6934. The method of item 6916 wherein the agent is a cathepsin K inhibitor, wherein the cathepsin K inhibitor is 462795 (GlaxoSmithKline), INPL-022-D6 (Amura Therapeutics), or an analogue or derivative thereof. 6935. The method of item 6916 wherein the agent is a cathepsin L inhibitor.
6936. The method of item 6916 wherein the agent is a CD40 antagonist.
6937. The method of item 6916 wherein the agent is a chemokine receptor agonist.
6938. The method of item 6916 wherein the agent is a chymase inhibitor.
6939. The method of item 6916 wherein the agent is a collagenase antagonist.
6940. The method of item 6916 wherein the agent is a CXCR antagonist.
6941. The method of item 6916 wherein the agent is a cyclin dependent kinase inhibitor selected from the group consisting of a CDK-1 inhibitor, a CDK-2 inhibitor, a CDK- 4 inhibitor, a CDK-6 inhibitor, a CAK1 inhibitor from GPC Biotech and Bristol-Myers Squibb, RGB-286199 (GPC Biotech), an anticancer agent from Astex Technology, a CAK1 inhibitor from GPC Biotech, a CDK inhibitor from Sanofi-Aventis, a CDK1/CDK2 inhibitor from Amgen, a CDK2 inhibitor from SUGEN-2 (Pfizer), a hearing loss therapy agent (Sound Pharmaceuticals), PD-0332991 (Pfizer), RGB-286199 (GPC Biotech), Ro-0505124 (Hoffmann-La Roche), a Ser/Thr kinase inhibitor from Lilly (Eli Lilly), CVT-2584 (CAS No. 199986-75-9) (CV Therapeutics), and an analogue or derivative thereof. 6942. The method of item 6916 wherein the agent is a cyclooxygenase 1 inhibitor.
6943. The method of item 6916 wherein the agent is a DHFR inhibitor.
6944. The method of item 6916 wherein the agent is a dual integrin inhibitor.
6945. The method of item 6916 wherein the agent is an elastase inhibitor.
6946. The method of item 6916 wherein the agent is an elongation factor-1 alpha inhibitor.
6947. The method of item 6916 wherein the agent is an endothelial growth factor antagonist.
6948. The method of item 6916 wherein the agent is an endothelial growth factor receptor kinase inhibitor selected from the group consisting of sorafenib tosylate (Bayer), AAL-993 (Novartis), ABP-309 (Novartis), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), E- 7080 (Eisai), EG-3306 (Ark Therapeutics), EXEL-2880 (Exelixis), GW- 654652 (GlaxoSmithKline), a KDR inhibitor from LG Life Sciences, CT-6685 and CT-6729 (UCB), KRN-633 and KRN-951 (Kirin Brewery), OSI-930 (OSI Pharmaceuticals), SP-5.2 (Supratek Pharma), SLM 1657 (Pfizer), a Tie-2 antagonist (Hybrigenics), a VEGFR-2 kinase inhibitor (Bristol-Myers Squibb), XL-647 (Exelixis), a KDR inhibitor from Abbott Laboratories, sorafenib tosylate, and an analogue or derivative thereof. 6949. The method of item 6916 wherein the agent is an endotoxin antagonist.
6950. The method of item 6916 wherein the agent is an epothilone and tubulin binder.
6951. The method of item 6916 wherein the agent is an estrogen receptor antagonist.
6952. The method of item 6916 wherein the agent is an FGF inhibitor.
6953. The method of item 6916 wherein the agent is a farnexyl transferase inhibitor.
6954. The method of item 6916 wherein the agent is farnesyltransferase inhibitor selected from the group of A-197574 (Abbott), a farnesyltransferase inhibitor from Servier, FPTIII (Strathclyde Institute for Drug R), LB-42908 (LG Life Sciences), Pharmaprojects No. 5063 (Genzyme), Pharmaprojects No. 5597 (Ipsen), Yissum Project No. B-1055 (Yissum), and an analogue or derivative thereof.
6955. The method of item 6916 wherein the agent is an FLT-3 kinase inhibitor.
6956. The method of item 6916 wherein the agent is an FGF receptor kinase inhibitor.
6957. The method of item 6916 wherein the agent is a fibrinogen antagonist selected from the group consisting of AUV-201 (Auvation), MG-13926 (Sanofi-Aventis), plasminogen activator (CAS No. 105913-11-9) (from Sanofi-Aventis or UCB), plasminogen activator-2 (tPA- 2) (Sanofi-Aventis), pro-urokinase (CAS No. 82657-92-9) (Sanofi-Aventis), and an analogue or derivative thereof.
6958. The method of item 6916 wherein the agent is a heat shock protein 90 antagonist selected from the group consisting of SRN-005 (Sirenade), geldanamycin, NSC-33050 (17-allylaminogeldanamycin; 17- AAG), 17-dimethylaminoethylamino-17-demethoxy-geldanamycin (17- DMAG), rifabutin (rifamycin XIV, 1',4-didehydro-1-deoxy~1,4-dihydro-5'-(2- methylpropyl)-1 -OXO-), and an analogue or derivative thereof.
6959. The method of item 6916 wherein the agent is a histone deacetylase inhibitor.
6960. The method of item 6916 wherein the agent is an HMGCoA reductase inhibitor selected from the group consisting of an atherosclerosis therapeutic from Lipid Sciences, ATI-16000 (ARYx Therapeutics), KS-01-019 (Kos Pharmaceuticals), Pharmaprojects No. 2197 (Sanofi-Aventi), RP 61969 (Sanofi-Aventis), and an analogue or derivative thereof.
6961. The method of item 6916 wherein the agent is an ICAM inhibitor.
6962. The method of item 6916 wherein the agent is an IL1 ICE and IRAK antagonist, wherein the antagonist is a CJ-14877, CP-424174 (Pfizer), NF-61 (Negma-Lerads), and an analogue or derivative thereof.
6963. The method of item 6916 wherein the agent is an IL-2 inhibitor. 6964. The method of item 6916 wherein the agent is an immunosuppressant selected from the group consisting of teriflunomide (Sanofi Aventis), chlorsulfaquinoxalone (NSC-339004), chlorsulfaquinoxalone sulfate, CS-712 (Sankyo), ismomultin alfa (CAS No. 457913-93-8) (Akzo Nobel), antiallergics from GenPat77, antiinflammatories or AT-005 (Androclus Therapeutics), autoimmune disease therapeutics from EpiVax, BN-007 (Bone), budesonide (CAS No. 51333-22- 3) (MAP Pharmaceuticals), CO-14 (Genzyme), edratide (CAS No. 433922- 67-9) (Teva), EP-314 (Enanta), eprovafen (CAS No. 101335-99-3) (Sanofi- Aventis), HWA-131 (CAS No. 118788-41-3) (Sanofi-Aventis), immunomodulators from MerLion Pharmaceuticals, immunosuppressives from Alchemia, IPL-12 (Inflazyme), MDL-9563 (CAS No. 27086-86-8) (Sanofi-Aventis), Pharmaprojects No. 2330 (Sanofi-Aventis), Pharmaprojects No. 6426 (Abgenix), PXS-25 (Pharmaxis), rosmarinic acid (CAS No. 20283-92-5) (Sanofi-Aventis), RP 42927 or RP 54745 (CAS No. 135330-08-4) (Sanofi-Aventis), βGN-35 (Seattle Genetics), ST-1959 (Sigma-Tau), type I diabetes therapy from SYNX Pharma, UNIL-88 (Debiopharm), VP-025 (Vasogen), VR-694 (Vectura), PRTX-001 (Protalex), and an analogue or derivative thereof.
6965. The method of item 6916 wherein the agent is an IMPDH (inosine monophosphate).
6966. The method of item 6916 wherein the agent is an integrin antagonist.
6967. The method of item 6916 wherein the agent is an interleukin antagonist.
6968. The method of item 6916 wherein the agent is an inhibitor of type III receptor tyrosine kinase. 6969. The method of item 6916 wherein the agent is an irreversible inhibitor of enzyme methionine aminopeptidase type 2.
6970. The method of item 6916 wherein the agent is an isozyme selective delta protein kinase C inhibitor.
6971. The method of item 6916 wherein the agent a JAK3 enzyme inhibitor.
6972. The method of item 6916 wherein the agent is a JNK inhibitor.
6973. The method of item 6916 wherein the agent is a kinase inhibitor.
6974. The method of item 6916 wherein the agent is kinesin antagonist.
6975. The method of item 6916 wherein the agent is a kinesin antagonist.
6976. The method of item 6916 wherein the agent is a leukotriene inhibitor and antagonist selected from the group consisting of ambicromil (CAS No. 58805-38-2) (Sanofi-Aventis), amelubant (CAS No. 346735-24-8) (Boehringer Ingelheim), DW-1141 (Dong Wha), ebselen (Daiichi Pharmaceutical), ibudilast (Kyorin), leucotriene inhibitors from Sanofi-Aventis, lymphotoxin-beta receptor (LT-D) from Biogen Idee, Pharmaprojects No. 1535 and 2728 (CAS No. 119340-33-9) (Sanofi- Aventis), R-112 (Rigel), Rev-5367 (CAS No. 92532-05-3) (Sanofi-Aventis), RG-14893 (CAS No. 141835-49-6) (Sanofi-Aventis), RG-5901-A (CAS No. 101910-24-1), 92532-23-5, RP 66153 (CAS No. 142422-79-5), RP 66364 (CAS No.186916-92-5), RP 69698 (CAS No. 141748-00-7) (Sanofi-Aventis), SC-411930 (Pfizer), SC-41930 (CAS No. 120072-59-5) (Pfizer), SC-50605 (CAS No. 138828-39-4) (Pfizer), SC-51146 (CAS No. 141059-52-1), SC- 53228 (CAS No. 153633-01-3) (Pfizer), spaglumic acid (ZY-15106) (CAS No.3106-85-2), 80619-64-3 (Novartis), tipredane (CAS No. 85197-77-9) (Bristol-Myers Squibb), U-75302 (CAS No. 119477-85-9) (Pfizer), and analogue or derivative thereof.
6977. The method of item 6916 wherein the agent is an MAP kinase inhibitor.
6978. The method of item 6916 wherein the agent is a matrix metalloproteinase inhibitor.
6979. The method of item 6916 wherein the agent is an MCP- CCR2 inhibitor.
6980. The method of item 6916 wherein the agent is an mTOR inhibitor.
6981. The method of item 6916 wherein the agent is an mTOR kinase inhibitor.
6982. The method of item 6916 wherein the agent is a microtubule inhibitor selected from the group consisting of antibody- maytansinoid conjugates from Biogen Idee, colchicines (MantiCore Pharmaceuticals), anticancer immunoconjugates from Johnson & Johnson, DIME from Octamer, gni-1f (GNl)1 huC242-DM4, huMy9-6-DM1 (ImmunoGen), IDN-5404 (Indena), IMO-098, IMOderm (Imotep), mebendazole (Introgen Therapeutics), microtubule poisons from Cambridge Enterprise, paclitaxel such as LOTAX from Aphios (CAS No. 33069-62-4), Genexol-PM from Samyang, Pharmaprojects No. 6383 (Tapestry Pharmaceuticals), RPR-112378 (Sanofi-Aventis), SGN-75 (Seattle Genetics), SPL-7435 (Starpharma), SSR-250411 (Sanofi-Aventis), trastuzumab-DM1 (Genentech), vinorelbine, and an analogue or derivative thereof.
6983. The method of item 6916 wherein the agent is an MIF inhibitor.
6984. The method of item 6916 wherein the agent is an MMP inhibitor.
6985. The method of item 6916 wherein the agent is a neurokinin (NK) antagonist selected from the group consisting of anthrotainin (CAS No. 148084-40-6) (Sanofi-Aventis), an IBS thereapeutic from ArQuIe, MDL-105212A (CAS No. 167261-60-1) (Ssanofi-Aventis), Pharmaprojects No. 2744, 3258 (CAS No. 139167-47-8) 4006, 4201 , or 5986 (Sanofi-Aventis), RP 67580 (CAS No. 135911-02-3), SR-144190 (CAS No. 201152-86-5), SSR-240600, SSR-241586 (Sanofi-Aventis), TKA-457 (Novartis), vestipitant mesylate (CAS No. 334476-64-1) (GlaxoSmithKline), Win-64821 (Sanofi-Aventis), PRX-96026 (Predix Pharmaceuticals), and an analogue or derivative thereof.
6986. The method of item 6916 wherein the agent is an NF kappa B inhibitor selected from the group consisting of AVE-0545 or AVE- 0547 (Sanofi-Aventis), bortezomib (CAS No. 179324-69-7) (Millennium Pharmaceuticals), dexanabinol (CAS No. 112924-45-5) (Pharmos), dexlipotam (Viatris), Pharmaprojects No. 6283 (INDRA) (OXiGENE), IPL- 576092 (CAS No. 137571-30-3) (Inflazyme), NFKB decoy (Corgentech), NFKB decoy oligo (AnGes MG), NFKB's from Ariad, osteoporosis treatments or S5 (F005) from Fulcrum Pharmaceuticals, P61 (Phytopharm), R-flurbiprofen (CAS No. 5104-49-4) (Encore Pharmaceuticals), and an analogue or derivative thereof.
6987. The method of item 6916 wherein the agent is a nitric oxide agonist.
6988. The method of item 6916 wherein the agent is an ornithine decarboxylase inhibitor.
6989. The method of item 6916 wherein the agent is a p38 MAP kinase inhibitor selected from the group consisting of AZD-6703 (AstraZeneca), JX-401 (Jexys Pharmaceuticals), BMS-2 (Bristol-Myers Squibb), a p38 MAP kinase inhibitor from Novartis, a p38-alpha MAP kinase inhibitor from Amphora, Pharmaprojects No. 5704 (Pharmacopeia), RPR- 200765A (Sanofi-Aventis), SD-282 (Johnson & Johnson), TAK-715 (Takeda), and an analogue or derivative thereof.
6990. The method of item 6916 wherein the agent is a palmitoyl-protein thioesterase inhibitor.
6991. The method of item 6916 wherein the agent is a PDGF receptor kinase inhibitor selected from the group consisting of AAL-993, AMN-107, or ABP-309 (Novartis), AMG-706 (Amgen), BAY-57-9352 (Bayer), CDP-860 (UCB), E-7080 (Eisai), imatinib (CAS No. 152459-95-5) (Novartis), OSI-930 (OSI Pharmaceuticals), RPR-127963E (Sanofi-Aventis), RWJ-540973 (Johnson & Johnson), sorafenib tosylate (Bayer), SU-11657 (Pfizer), tandutinib (CAS No. 387867-13-2) (Millennium Pharmaceuticals), vatalanib (Novartis), ZK-CDK (Schering AG), and an analogue or derivative thereof. 6992. The method of item 6916 wherein the agent is (-)- halofenate (Metabolex), AMG-131 (Amgen), antidiabetics from Japan Tobacco, AZD-4619, AZD-8450, AZD-8677 (AstraZeneca), DRF-10945, balaglitazone (Dr Reddy's), CS-00088, CS-00098 (Chipscreen Biosciences), E-3030 (Eisai), etalocib (CAS No. 161172-51-6) (EIi Lilly), GSK-641597 (Ligand), GSK-677954 (GlaxoSmithKline), GW-409544 (Ligand), GW- 590735 (GlaxoSmithKline), K-111 (Hoffmann-La Roche), LY-518674 (EIi Lilly), LY-674 (Ligand), LY-929 (Ligand), MC-3001, MC-3002 (MaxoCore Pharmaceuticals), metformin HCI + pioglitazone (CAS No. 1115-70-4 and
112529-15-4), ACTOPLUS MET from Andrx), muraglitazar (CAS No. 331741-94-7) (Bristol-Myers Squibb), naveglitazar (Ligand), oleoylethanolamide (Kadmus Pharmaceuticals), ONO-5129, pioglitazone hydrochloride (CAS No. 111025-46-8 and 112529-15-4) (Takeda), PLX-204 (Plexxikon), PPAR agonists from Genfit, PPAR delta agonists from EIi Lilly, PPAR-alpha agonists from CrystalGenomics, PPAR-gamma modulators and PPAR-β modulators from CareX, rosiglitazone maleate (CAS No. 122324- 73-4 or 155141-29-0) (GlaxoSmithKline), rosiglitazone maleate/glimepir (CAS No. 155141-29-0 and 93479-97-1), AVANDARYL, rosiglitazone maleate/metformin extend (CAS No. 155141-29-0 and 657-24-9), AVANDAMET, rosiglitazone maleate+metformin, AVANDAMET (GlaxoSmithKline), tesaglitazar (AstraZeneca), and an analogue or derivative thereof.
6993. The method of item 6916 wherein the agent is a phosphatase inhibitor.
6994. The method of item 6916 wherein the agent is a phosphodiesterase (PDE) inhibitor selected from the group consisting of avanafil (Tanabe Seiyaku), dasantafil (CAS No. 569351-91-3) (Schering- Plough), A-906119 (CAS No. 134072-58-5), DL-850 (Sanofi-Aventis), GRC- 3015, GRC-3566, GRC-3886 (Glenmark), HWA-153 (CAS No. 56395-66-5) (Sanofi-Aventis), hydroxypumafentrine (Altana), IBFB-130011 , IBFB-14-016, IBFB-140301 , IBFB-150007, IBFB-211913 (IBFB Pharma), L-826141 (Merck & Co), medorinone (CAS No. 88296-61-1) (Sanofi-Aventis), MEM- 1917 (Memory Pharmaceuticals), ND-1251 (Neuro3d), PDE inhibitors from ICOS, PDE IV inhibitors from Memory Pharmaceuticals and CrystalGenomics, Pharmaprojects No. 2742 and 6141 (Sanofi-Aventis), QAD-171 (Novartis), RHC-2963 (CAS No. 76993-12-9 and 76993-14-1), RPR-117658, RPR-122818 derivatives, SR-24870 , and RPR-132294 (Sanofi-Aventis), SK-350 (ln2Gen), stroke therapy agents from deCODE Genetics, TAS-203 (Taiho), tofimilast (CAS No. 185954-27-2) (Pfizer), UK- 371800 (Pfizer), WIN-65579 (CAS No. 158020-82-7) (Sanofi-Aventis), IBFB- 130020 (IBFB Pharma), OPC-6535 (CAS No. 145739-56-6) (Otsuka), a phosphodiesterase III inhibitor, enoximone, a phosphodiesterase IV inhibitor, fosfosal, Atopik (Barrier Therapeutics), triflusal, a phosphodiesterase V inhibitor, and an analogue or derivative thereof.
6995. The method of item 6916 wherein the agent is a PKC inhibitor.
6996. The method of item 6916 wherein the agent is a platelet activating factor antagonist.
6997. The method of item 6916 wherein the agent is a platelet-derived growth factor receptor kinase inhibitor.
6998. The method of item 6916 wherein the agent is a prolyl hydroxylase inhibitor.
6999. The method of item 6916 wherein the agent is a polymorphonuclear neutrophil Inhibitor. 7000. The method of item 6916 wherein the agent is a protein kinase B inhibitor.
7001. The method of item 6916 wherein the agent is a protein kinase C stimulant.
7002. The method of item 6916 wherein the agent is a purine nucleoside analogue.
7003. The method of item 6916 wherein the agent is a purinoreceptor P2X antagonist.
7004. The method of item 6916 wherein the agent is a Raf kinase inhibitor.
- 7005. The method of item 6916 wherein the agent is a reversible inhibitor of ErbB1 and ErbB2.
7006. The method of item 6916 wherein the agent is a ribonucleoside triphosphate reductase inhibitor.
7007. The method of item 6916 wherein the agent is an SDF- 1 antagonist.
7008. The method of item 6916 wherein the agent is a sheddase inhibitor.
7009. The method of item 6916 wherein the agent is an SRC inhibitor. 7010. The method of item 6916 wherein the agent is a stromelysin inhibitor.
7011. The method of item 6916 wherein the agent is an Syk kinase inhibitor.
7012. The method of item 6916 wherein the agent is a telomerase inhibitor.
7013. The method of item 6916 wherein the agent is a TGF beta inhibitor selected from the group consisting of pirfenidone (CAS No. 53179-13-8) (MARNAC), tranilast (CAS No. 53902-12-8) (Kissei), IN-1130 (ln2Gen), mannose-6-phosphate (BTG), TGF-β antagonists from Inflazyme (Pharmaprojects No. 6075), TGF-β antagonists from Sydney, non-industrial source), TGF-β I receptor kinase inhibitors from EIi Lilly, TGF-β receptor inhibitors from Johnson & Johnson, and an analogue or. derivative thereof.
7014. The method of item 6916 wherein the agent is a TNFα antagonist or TACE inhibitor selected from the group consisting of adalimumab (CAS No. 331731-18-1) (Cambridge Antibody Technology), AGIX-4207 (AtheroGenics), AGT-1 (Advanced Biotherapy), an antiinflammatory from Borean Pharma, Cellzome, or Paradigm Therapeutics, anti-inflammatory vaccine (TNF-alpha kinoid) from Neovacs, humanized anti-TNF antibody or an anti-TNF MAb (CB0006) Celltech (UCB), apratastat (CAS No. 287445-51-0) (Wyeth), BMS-561392 (Bristol-Myers Squibb), BN- 006 (Bone), certolizumab pegol (CAS No. 428863-50-7 or CH-138 (UCB), cilomilast (CAS No. 153259-65-5) (GlaxoSmithKline), CR-1 (Nuada Pharmaceuticals), CRx-119 (CombinatoRx), D-5410 (UCB), dacopafant (CAS No. 125372-33-0) (Sanofi-Aventis), dersalazine (CAS No. 188913-57- 7/188913-58-8) (Uriach), etanercept (CAS No. 185243-69-0) (Amgen), ethyl pyruvate (Critical (Critical Therapeutics), golimumab (CAS No. 476181-74-5) (Johnson & Johnson), hormono-immunotherapy from Ipsen, CDP571 (e.g., Humicade from UCB), IC-485 (ICOS), infliximab (CAS No. 170277-31-3) (Johnson & Johnson), IP-751 (Manhattan Pharmaceuticals), ISIS-104838 (CAS No. 250755-32-9) (ISIS Pharmaceuticals), lenalidomide (CAS No. 191732-72-6) (Celgene), lentinan (CAS No. 37339-90-5) (Ajinomoto), MDL- 201112 (CAS No. 142130-73-2) (Sanofi-Aventis), medroxyprogesterone (CAS No. 520-85-4) (InKine Pharmaceutical), N-acetylcysteine (CAS No. 616-91-1) (Zambon), NBE-P2 (DIREVO Biotech), nerelimomab (CAS No. 162774-06-3) (Chiron), OM-294DP (OM PHARMA), onercept (CAS No. 199685-57-9) (Yeda), PASSTNF-alpha (Verigen), pentoxifylline or oxypentifylline (Sanofi-Aventis), Pharmaprojects No. 4091 , 4241 , 4295, or 4488 (Sanofi-Aventis), Pharmaprojects No. 5480 (Amgen), Pharmaprojects No. 6749 (Cengent), pirfenidone (CAS No. 53179-13-8) (MARNAC), RPR- 132294 (Sanofi-Aventis), S5 (F002) (Fulcrum Pharmaceuticals), simvastatin (CAS No. 79902-63-9) (Merck & Co), STA-6292 (Synta Pharmaceuticals), tacrolimus (CAS No. 104987-11-3)_(Fujisawa LjfβCycle Pharma), talactoferrin alfa (CAS No. 308240-58-6) (Agennix), thalidomide (CAS No. 50-35-1) (Celgene), TNF antagonists form ProStrakan, and Synergen, TNF inhibitors (Amgen), TNF-alpha antagonists from Dynavax Technologies and Jerina AG (Germany), TNF-alpha inhibitors from IBFB Pharma and Xencor (Xencor), torbafylline (CAS No. 105102-21-4) (Sanofi-Aventis), UR-1505 (Uriach), VT-346 (Viron Therapeutics), YSIL6 (Y's Therapeutics), YSTH2 (Y's Therapeutics), NPI-1302a-3 (Nereus Pharmaceuticals, a TNF antagonist from Jerina AG (Germany), dersalazine, and an analogue or derivative thereof.
7015. The method of item 6916 wherein the agent is a Toll receptor inhibitor.
7016. The method of item 6916 wherein the agent is a tubulin antagonist. 7017. The method of item 6916 wherein the agent is a tyrosine kinase inhibitor selected from the group consisting of SU-011248, SUTENT from Pfizer Inc. (New York, NY), BMS-354825, PN-355 (Paracelsian Pharmaceuticals), AGN-199659 (Allergan), AAL-993 or ABP- 309 (Novartis), adaphostin (NIH), AEE-788 (Novartis), AG-013736 (OSI Pharmaceuticals), AG-13736 (Pfizer), ALT-110 (Alteris Therapeutics), AMG- 706 (Amgen), anticancer MAbs from Xencor, anti-EGFrvlll MAbs from Abgenix, anti-HER2 MAb from Abiogen, AZD-2171 or AZD-9935 (AstraZeneca), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), CEP-5214 (Cephalon), CEP-7055 (Cephalon), cetuximab (ImClone Systems), CHIR-200131 and CHIR-258 (Chiron), CP-547632 (OSI Pharmaceuticals), CP-724714 (Pfizer), CT-301 (Creabilis Therapeutics), D- 69491 (Baxter International), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EGFR/ErbB2 inhibitors from Array BioPharma, erlotinib (CAS No. 183319- 69-9) (OSI Pharmaceuticals), EXEL-2880 (Exelixis), FK-778 (Sanofi- Aventis) , gefitinib (CAS No. 184475-35-2) (AstraZeneca), GW-2286 or GW- 654652 (GlaxoSmithKline), her2/neu antigen from AlphaVax, HER-2/neu inhibitor from Generex, Herzyme (Medipad) (Sirna Therapeutics), HKI-272 (Wyeth), HuMax-EGFr (Genmab), idronoxil (CAS No. 81267-65-4) (Novogen), IGF-1 inhibitors from Ontogen, IMC-11F8 (ImClone Systems), kahalalide F (CAS No. 149204-42-2) (PharmaMar), KDR inhibitor from LG Life Sciences, KDR inhibitors from Abbott Laboratories, KDR kinase inhibitors (UCB), Kdr kinase inhibitors from Merck & Co, KRN-633 and KRN- 951 (Kirin Brewery), KSB-102 (Xenova), lapatinib ditosylate (CAS No. 388082-78-8) (GlaxoSmithKline), matuzumab (Merck KGaA), MDX-214 (Medarex), ME-103 (Pharmexa), MED-A300 (Gerolymatos), MNAC-13 (Lay Line Genomics), nimotuzumab (Center of Molecular Immunology), NSC- 330507 or NSC-707545 (NIH), NV-50 (Novogen), OSI-930 (OSI Pharmaceuticals), panitumumab (Abgenix), pelitinib (CAS No. 287933-82-7) (Wyeth), pertuzumab (CAS No. 380610-27-5) (Genentech), Pharmaprojects No. 3985 (Sanofi-Aventis), prostate cancer therapeutics from Sequenom (SQPC35, SQPC36, SQPC90), removab and remoxab (Trion Pharma), RG- 13022 (CAS No. 136831-48-6), RG-13291 (CAS No. 138989-50-1), or RG- 14620 (CAS No. 136831-49-7) (Sanofi-Aventis), RM-6427 (Romark), RNAi breast cancer therapy from Benitec, RP 53801 (CAS No. 125882-88-4) (Sanofi-Aventis), sorafenib tosylate (Bayer), SU-11657 (Pfizer), Tie-2 antagonists from Semaia (Hybrigenics), Tie-2 inhibitors from Ontogen, trastuzumab (CAS No. 180288-69-1) (Genentech), tyrosine kinase inhibitors from Sanofi-Aventis, U3-1287, U3-1565, U3-1784, U3-1800 (U3 Pharma), vatalanib (Novartis), VEGFR-2 kinase inhibitor from Bristol-Myers Squibb, XL-647 (Exelixis), ZD-6474 (AstraZeneca), ZK-CDK (Schering AG), an EGFR tyrosine kinase inhibitor, EKB-569 (Wyeth), and an analogue or derivative thereof.
7018. The method of item 6916 wherein the agent is a VEGF inhibitor.
7019. The method of item 6916 wherein the agent is a vitamin D receptor agonist.
7020. The method of item 6916 wherein the agent is ZD-6474 (an angiogenesis inhibitor).
7021. The method of item 6916 wherein the agent is AP- 23573 (an mTOR inhibitor).
7022. The method of item 6916 wherein the agent is synthadotin (a tubulin antagonist).
7023. The method of item 6916 wherein the agent is S-0885 (a collagenase inhibitor). 7024. The method of item 6916 wherein the agent is aplidine (an elongation factor-1 alpha inhibitor).
7025. The method of item 6916 wherein the agent is ixabepilone (an epithilone).
7026. The method of item 6916 wherein the agent is IDN- 5390 (an angiogenesis inhibitor).
7027. The method of item 6916 wherein the agent is SB- 2723005 (an angiogenesis inhibitor).
7028. The method of item 6916 wherein the agent is ABT-518 (an angiogenesis inhibitor).
7029. The method of item 6916 wherein the agent is combretastatin (an angiogenesis inhibitor).
7030. The method of item 6916 wherein the agent is anecortave acetate (an angiogenesis inhibitor).
7031. The method of item 6916 wherein the agent is SB- 715992 (a kinesin antagonist).
7032. The method of item 6916 wherein the agent is temsirolimus (an mTOR inhibitor).
7033. The method of item 6916 wherein the agent is adalimumab (a TNFα antagonist). 7034. The method of item 6916, wherein the composition comprises a polymer.
7035. The method of item 6916, wherein the composition comprises a polymer, and the polymer is, or comprises, a copolymer.
7036. The method of item 6916, wherein the composition comprises a polymer, and the polymer is, or comprises, a block copolymer.
7037. The method of item 6916, wherein the composition comprises a polymer, and the polymer is, or comprises, a random copolymer.
7038. The method of item 6916, wherein the composition comprises a polymer, and the polymer is, or comprises, a biodegradable polymer. -
7039. The method of item 6916, wherein the composition comprises a polymer, and the polymer is, or comprises, a nonbiodegradable polymer.
7040. The method of item 6916, wherein the composition comprises a polymer, and the polymer is, or comprises, a hydrophilic polymer.
7041. The method of item 6916, wherein the composition comprises a polymer, and the polymer is, or comprises, a hydrophobic polymer. 7042. The method of item 6916, wherein the composition comprises a polymer, and the polymer is, or comprises, a polymer having hydrophilic domains.
7043. The method of item 6916, wherein the composition comprises a polymer, and the polymer is, or comprises, a polymer having hydrophobic domains.
7044. The method of item 6916, wherein the composition comprises a polymer, and the polymer is, or comprises, a non-conductive polymer.
7045. The method of item 6916, wherein the composition comprises a polymer, and the polymer is, or comprises, an elastomer.
7046. The method of item 6916, wherein the composition comprises a polymer, and the polymer is, or comprises, a hydrogel.
7047. The method of item 6916, wherein the composition comprises a polymer, and the polymer is, or comprises, a silicone polymer.
7048. The method of item 6916, wherein the composition comprises a polymer, and the polymer is, or comprises, a hydrocarbon polymer.
7049. The method of item 6916, wherein the composition comprises a polymer, and the polymer is, or comprises, a styrene-derived polymer. 7050. The method of item 6916, wherein the composition comprises a polymer, and the polymer is, or comprises, a butadiene-derived polymer.
7051. The method of item 6916, wherein the composition comprises a polymer, and the polymer is, or comprises, a macromer.
7052. The method of item 6916, wherein the composition comprises a polymer, and the polymer is, or comprises, a poly(ethylene glycol) polymer.
7053. The method of item 6916, wherein the composition comprises a polymer, and the polymer is, or comprises, an amorphous polymer.
7054. The method of item 691.6, wherein the composition further comprises a second pharmaceutically active agent.
7055. The method of item 6916, wherein the composition further comprises an anti-inflammatory agent.
7056. The method of item 6916, wherein the composition further comprises an agent that inhibits infection.
7057. The method of item 6916, wherein the composition further comprises an anthracycline.
7058. The method of item 6916, wherein the composition further comprises doxorubicin. 7059. The method of item 6916 wherein the composition further comprises mitoxantrone.
7060. The method of item 6916 wherein the composition further comprises a fluoropyrimidine.
7061. The method of item 6916, wherein the composition further comprises 5-fluorouracil (5-FU).
7062. The method of item 6916, wherein the composition further comprises a folic acid antagonist.
7063. The method of item 6916, wherein the composition further comprises methotrexate.
7064. The method of item 6916, wherein the composition further comprises a podophylotoxin.
7065. The method of item 6916, wherein the composition further comprises etoposide.
7066. The method of item 6916, wherein the composition further comprises camptothecin.
7067. The method of item 6916, wherein the composition further comprises a hydroxyurea.
7068. The method of item 6916, wherein the composition further comprises a platinum complex. 7069. The method of item 6916, wherein the composition further comprises cisplatin.
7070. The method of item 6916 wherein the composition further comprises an anti-thrombotic agent.
7071. The method of item 6916, wherein the composition further comprises a visualization agent.
7072. The method of item 6916, wherein the composition further comprises a visualization agent, and the visualization agent is a radiopaque material, wherein the radiopaque material comprises a metal, a halogenated compound, or a barium containing compound.
7073. The method of item 6916, wherein the composition furthencomprises a visualization agent, and the visualization agent is, or comprises, barium, tantalum, or technetium.
7074. The method of item 6916, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, an MRI responsive material.
7075. The method of item 6916, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, a gadolinium chelate.
7076. The method of item 6916, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, iron, magnesium, manganese, copper, or chromium. 7077. The method of item 6916, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, iron oxide compound.
7078. The method of item 6916, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, a dye, pigment, or colorant.
7079. The method of item 6916 wherein the agent is released in effective concentrations from the composition comprising the agent by diffusion over a period ranging from the time of administration to about 90 days.
7080. The method of item 6916 wherein the agent is released in effective concentrations from the composition comprising the agent by erosion of the composition over a period ranging from the time of administration to about 90 days.
7081. The method of item 6916 wherein the composition further comprises an inflammatory cytokine.
7082. The method of item 6916 wherein the composition further comprises an agent that stimulates cell proliferation.
7083. The method of item 6916 wherein the composition further comprises a polymeric carrier.
7084. The method of item 6916 wherein the composition is in the form of a gel, paste, or spray. 7085. The method of item 6916 wherein the implant is partially constructed with the agent or the composition.
7086. The method of item 6916 wherein the implant is fully constructed with the agent or the composition.
7087. The method of item 6916 wherein the implant is impregnated with the agent or the composition.
7088. The method of item 6916, wherein the agent or the composition forms a coating, and the coating directly contacts the implant.
7089. The method of item 6916, wherein the agent or the composition forms a coating, and the coating indirectly contacts the implant.
7090. The method of item 6916 wherein the agent or the composition forms a coating, and the coating partially covers the implant.
7091. The method of item 6916, wherein the agent or the composition forms a coating, and the coating completely covers the implant.
7092. The method of item 6916 wherein the agent or the composition is located within pores or holes of the implant.
7093. The method of item 6916 wherein the agent or the composition is located within a channel, lumen, or divet of the implant.
7094. The method of item 6916 wherein the implant further comprising an echogenic material. 7095. The method of item 6916 wherein the implant further comprises an echogenic material, wherein the echogenic material is in the form of a coating.
7096. The method of item 6916 wherein the implant is sterile.
7097. The method of item 6916 wherein the agent is delivered from the implant, wherein the agent is released into tissue in the vicinity of the implant after deployment of the implant.
7098. The method of item 6916 wherein the agent is delivered from the implant, wherein the agent is released into tissue in the vicinity of the implant after deployment of the implant, wherein the tissue is connective tissue.
7099. The method of item 6916 wherein the agent is delivered from the implant, wherein the agent is released into tissue in the vicinity of the implant after deployment of the implant, wherein the tissue is muscle tissue.
7100. The method of item 6916 wherein the agent is delivered from the implant, wherein the agent is released into tissue in the vicinity of the implant after deployment of the implant, wherein the tissue is nerve tissue.
7101. The method of item 6916 wherein the agent is delivered from the implant, wherein the agent is released into tissue in the vicinity of the implant after deployment of the implant, wherein the tissue is epithelium tissue. 7102. The method of item 6916 wherein the agent is delivered from the implant, wherein the agent is released in effective concentrations from the implant over a period ranging from the time of deployment of the implant to about 1 year.
7103. The method of item 6916 wherein the agent is delivered from the implant, wherein the agent is released in effective concentrations from the implant over a period ranging from about 1 month to 6 months.
7104. The method of item 6916 wherein the agent is delivered from the implant, wherein the agent is released in effective concentrations from the implant over a period ranging from about 1 - 90 days.
7105. The method of item 6916 wherein the agent is delivered from the implant, wherein the agent is released in effective concentrations from the implant at a constant rate.
7106. The method of item 6916 wherein the agent is delivered from the implant, wherein the agent is released in effective concentrations from the implant at an increasing rate.
7107. The method of item 6916 wherein the agent is delivered from the implant, wherein the agent is released in effective concentrations from the implant at a decreasing rate.
7108. The method of item 6916 wherein the agent is delivered from the implant, wherein the implant comprises about 0.01 Dg to about 10 Dg of the agent. 7109. The method of item 6916 wherein the agent is delivered from the implant, wherein the implant comprises about 10 Dg to about 10 mg of the agent.
7110. The method of item 6916 wherein the agent is delivered from the implant, wherein the implant comprises about 10 mg to about 250 mg of the agent.
7111. The method of item 6916 wherein the agent is delivered from the implant, wherein the implant comprises about 250 mg to about 1000 mg of the agent.
7112. The method of item 6916 wherein the agent is delivered from the implant, wherein the implant comprises about 1000 mg to about 2500 mg of the agent.
7113. The method of item 6916 wherein the agent is delivered from the implant, wherein a surface of the implant comprises less than 0.01 Dg of the agent per mm2 of implant surface to which the agent is applied.
7114. The method of item 6916 wherein the agent is delivered from the implant, wherein a surface of the implant comprises about 0.01 Dg to about 1 Dg of the agent per mm2 of implant surface to which the agent is applied.
7115. The method of item 6916 wherein the agent is delivered from the implant, wherein a surface of the implant comprises about 1 Dg to about 10 Dg of the agent per mm2 of implant surface to which the agent is applied. 7116. The method of item 6916 wherein the agent is delivered from the implant, wherein a surface of the implant comprises about 10 Dg to about 250 Dg of the agent per mm2 of implant surface to which the agent is applied.
7117. The method of item 6916 wherein the agent is delivered from the implant, wherein a surface of the implant comprises about 250 Dg to about 1000 Dg of the agent per mm2 of implant surface to which the agent is applied.
7118. The method of item 6916 wherein the agent is delivered from the implant, wherein a surface of the implant comprises about 1000 Dg to about 2500 Dg of the agent per mm2 of implant surface to which the agent is applied.
7119. The method of item 6916, wherein the implant further comprises a coating, and the coating is a uniform coating.
7120. The method of item 6916, wherein the implant further comprises a coating, and the coating is a non-uniform coating.
7121. The method of item 6916, wherein the implant further comprises a coating, and the coating is a discontinuous coating.
7122. The method of item 6916, wherein the implant further comprises a coating, and the coating is a patterned coating.
7123. The method of item 6916, wherein the implant further comprises a coating, and the coating has a thickness of 100 Dm or less. 7124. The method of item 6916, wherein the implant further comprises a coating, and the coating has a thickness of 10 Dm or less.
7125. The method of item 6916, wherein the implant further comprises a coating, and the coating adheres to the surface of the implant upon deployment of the implant.
7126. The method of item 6916, wherein the implant further comprises a coating, and the coating is stable at room temperature for a period of at least 1 year.
7127. The method of item 6916, wherein the implant further comprises a coating, and the agent is present in the coating in an amount ranging between about 0.0001 % to about 1% by weight.
7128. The method of item 6916, wherein the implant further comprises a coating, and the agent is present in the coating in an amount ranging between about 1% to about 10% by weight.
7129. The method of item 6916, wherein the implant further comprises a coating, and the agent is present in the coating in an amount ranging between about 10% to about 25% by weight.
7130. The method of item 6916, wherein the implant further comprises a coating, and the agent is present in the coating in an amount ranging between about 25% to about 70% by weight.
7131. The method of item 6916, wherein the implant further comprises a coating, and the coating comprises a polymer. 7132. The method of item 6916, wherein the implant comprises a first coating having a first composition and a second coating having a second composition.
7133. The method of item 6916, wherein the implant comprises a first coating having a first composition and a second coating having a second composition, wherein the first composition and the second composition are different.
7134. A method as in any one of items 6916-7133, wherein the device is a grommet shaped tube.
7135. A method as in any one of items 6916-7133, wherein the device is a T-tube.
7136. A method as in any one of items 6916-7133, wherein the device is a tympanostomy tube.
7137. A method as in any one of items 6916-7133, wherein the device is a drain tube.
7138. A method as in any one of items 6916-7133, wherein the device is a tympanic tube.
7139. A method as in any one of items 6916-7133, wherein the device is an otological tube.
7140. A method as in any one of items 6916-7133, wherein the device is a myringotomy tube. 7141. A method as in any one of items 6916-7133, wherein the device is an artificial Eustachian tube.
7142. A method as in any one of items 6916-7133, wherein the device is an Eustachian rube prosthesis.
7143. A method as in any one of items 6916-7133, wherein the device is an Eustachian stent.
7144. A method for inhibiting scarring comprising placing a device that comprises an intraocular implant (e.g., an implant) and an anti- scarring agent or a composition comprising an anti-scarring agent into an animal host, wherein the agent inhibits scarring.
7145. The method of item 7144 wherein the agent is an adensosine A2A receptor antagonist.
7146. The method of item 7144 wherein the agent is an AKT inhibitor.
7147. The method of item 7144 wherein the agent is an alpha 2 integrin antagonist, wherein the alpha 2 integrin antagonist is Pharmaprojects No. 5754 (Merck KgaA).
7148. The method of item 7144 wherein the agent is an alpha 4 integrin antagonist.
7149. The method of item 7144 wherein the agent is an alpha 7 nicotinic receptor agonist. 7150. The method of item 7144 wherein the agent is an angiogenesis inhibitor selected from the group consisting of AG-12,958 (Pfizer), ATN-161 (Attenuon LLC), neovastat, an angiogenesis inhibitor from Jerina AG (Germany), NM-3 (Mercian), VGA-1155 (Taisho), FCE-26644 (Pfizer), FCE-26950 (Pfizer), FPMA (Meiji Daries), FR-111142 (Fujisawa), GGTI-298, GM-1306 (Ligand), GPA-1734 (Novartis), NNC-47-0011 (Novo Nordisk), herbamycin (Nippon Kayaku), lenalidomide (Celegene), IP-10 (NIH), ABT-828 (Abbott), KIN-841 (Tokushima University, Japan), SF-1126 (Semafore Pharmaceuticals), laminin technology (NIH), CHIR-258 (Chiron), NVP-AEW541 (Novartis), NVP-AEW541 (Novartis), Vt16907 (Alchemia), OXI-8007 (Oxigene), EG-3306 (Ark Therapeutics), Maspin (Arriva), ABT- 567 (Abbott), PPI-2458 (Praecis Pharmaceuticals), CC-5079, CC-4089 (Celgene), HIF-1 alpha inhibitors (Xenova), S-247 (Pfizer), AP-23573 (Ariad), AZD-9935 (Astra Zeneca), mebendazole (Introgen Therapeutics), MetAP-2 inhibitors (GlaxoSmithKline), AG-615 (Angiogene Pharmaceuticals), Tie-2 antagonists (Hybrigenics), NC-381 , CYC-381, NC-169, NC-219, NC-383, NC-384, NC-407 (Lorus Therapeutics), ATN-224 (Attenuon), ON-01370 (Onconova), Vitronectin antagonists (Amgen), SDX-103 (Salmedix), Vitronectin antagonists (Shire), CHP (Riemser), TEK (Amgen), Anecortave acetate (Alcon), T46.2 (Matrix Therapeutics), HG-2 (Heptagen), TEM antagonists (Genzyme), Oxi-4500 (Oxigene), ATN-161 (Attenuon), WX-293 (Wilex), M-2025 (Metris Therapeutics), Alphastatin (BioActa), YH-16 (Yantai Rongchang), BIBF-1120 (Boehringer Ingelheim), BAY-57-9352 (Bayer), AS- 1404 (Cancer Research Technology), SC-77964 (Pfizer), glycomimetics (BioTie Therapies), TIE-2 Inhibitors (Ontogen), DIMI, Octamer (Octamer), ABR-215050 (Active Biotech), ABT-518 (Abbott), KDR inhibitors (Abbott), BSF-466895 (Abbott), SCH-221153 (Schering-Plough), DAC:antiangiogenic (ConjuChem), TFPI (EntreMed), AZD-2171 (Astra-Zenaca), CDC-394 (Celgene), LY290293 (EIi Lilly), IDN-5390 (Indena), Kdr Kinase Inhibitors (Merck), CT-113020, CT-116433, CT-116563, CT-31890, CT-32228) (Cell Therapeutics), A-299620 (Abbott), TWEAK Inhibitor (Amgen), VEGF modulators (Johnson and Johnson), Tum-N53, tumstatin (Genzyme), Thios- 1 , Thios-2 (Thios Pharmaceuticals), MV-6401 (Miravant Medical Technologies), Spisulosine (PharmaMar), CEP-7055 (Cephalon), AUV-201 (Auvation), LM-609 (EIi Lilly), SKF-106615 (AnorMED), Oglufanide disodium (Cytran), BW-114 (Pharminox), Calreticulin (NIH), WX-678 (Wilex), SD-7784 (Pfizer), WX-UK1 (Wilex), SH-268 (Schering AG), 2-Me-PGA (Celgene), S- 137 (Pfizer), ZD-6126 (Angiogene Pharmaceuticals), SG-292 (SignalGen), Benefin (Lane Labs), A6, A36 (Angstrom), SB-2723005 (GlaxoSmithKline), SC-7 (Cell Therapeutics), ZEN-014 (AEterna Zentaris), 2-methoxyestradiol (EntreMed), NK-130119 (Nippon Kayaku), CC-10004 (Celgene), AVE- 8062A (Ajinomoto), Tacedinaline (Pfizer), Actinonin (Tokyo Metropolitan Institute of Medical Science), Lenalidomide (Celgene), VGA-1155, BTO-956 (SRI International), ER-68203-00 (Eisai), CT-6685 (UCB), JKC-362 (Phoenix Pharmaceuticals), DMI-3798 (DMI Biosciences, Angiomate (Ipsen), ZD-6474 (AstraZeneca), CEP-5214 (Cephalon), Canstatin (Genzyme), NM-3 (Mercian), Oridigm (MediQuest Therapeutics), Exherin (Adherex), BLS-0597 (Boston Life Sciences), PTC-299 (PTC Therapeutics), NPI-2358 (Nereus Pharmaceuticals), CGP-79787 (Novartis), AEE-788 (Novartis), CKD-732 (Chong Kun Dang), CP-564959 (OSI Pharmaceuticals), CM-101 (CarboMed), CT-2584, CT3501 (Cell Therapeutics), combretastatin and analogues and derivatives thereof (Oxigene), Rebimastat (Bristol-Meyers Squibb), Dextrin 2-sulfate (ML Laboratories), Cilengitide (Merk KGaA), NSC-706704 (Pharminox), KRN- 951 (Kirin Brewery), Ukrain, NSC-631570 (Nowicky Pharma), Tecogalan sodium (Daiichi Pharmaceutical), Tz-93 (Tsumura), TBC-1635 (Encysive Pharmaceuticals), TAN-1120 (Takeda), Semaxanib (Pfizer), BDI-7800 (Biopharmacopae), SD-186, SD-983 (Bristol-Meyers Squibb), SB-223245 (GlaxoSmithKline), SC-236 (Pfizer), RWJ-590973 (Johnson and Johnson), ILX-1850 (Genzyme), SC-68488, S-836 (Pfizer), CG-55069-11 (CuraGen), Ki-23057 (Kirin Brewery), CCX-700 (Chemoentryx), Pegaptanib octasodium (Giled Sciences), ANGIOCOL (available from Biostratum Inc.), or an analogue or derivative thereof.
7151. The method of item 7144 wherein the agent is an apoptosis antagonist.
7152. The method of item 7144 wherein the agent is an apoptosis activator.
7153. The method of item 7144 wherein the agent is a beta 1 integrin antagonist.
7154. The method of item 7144 wherein the agent is a beta tubulin inhibitor.
7155. The method of item 7144 wherein the agent is a blocker of enzyme production in Hepatitis C.
7156. The method of item 7144 wherein the agent is a Bruton's tyrosine kinase inhibitor.
7157. The method of item 7144 wherein the agent is a calcineurin inhibitor.
7158. The method of item 7144 wherein the agent is a i caspase 3 inhibitor.
7159. The method of item 7144 wherein the agent is a CC chemokine receptor antagonist. 7160. The method of item 7144 wherein the agent is a cell cycle inhibitor selected from the group consisting of SNS-595 (Sunesis), synthadotin, KRX-0403, and an analogue or derivative thereof.
7161. The method of item 7144 wherein the agent is a cathepsin B inhibitor.
7162. The method of item 7144 wherein the agent is a cathepsin K inhibitor, wherein the cathepsin K inhibitor is 462795 (GlaxoSmithKline), INPL-022-D6 (Amura Therapeutics), or an analogue or derivative thereof.
7163. The method of item 7144 wherein the agent is a cathepsin L inhibitor.
7164. The method of item 7144 wherein the agent is a CD40 antagonist.
7165. The method of item 7144 wherein the agent is a chemokine receptor agonist.
7166. The method of item 7144 wherein the agent is a chymase inhibitor.
7167. The method of item 7144 wherein the agent is a collagenase antagonist.
7168. The method of item 7144 wherein the agent is a CXCR antagonist. 7169. The method of item 7144 wherein the agent is a cyclin dependent kinase inhibitor selected from the group consisting of a CDK-1 inhibitor, a CDK-2 inhibitor, a CDK- 4 inhibitor, a CDK-6 inhibitor, a CAK1 inhibitor from GPC Biotech and Bristol-Myers Squibb, RGB-286199 (GPC Biotech), an anticancer agent from Astex Technology, a CAK1 inhibitor from GPC Biotech, a CDK inhibitor from Sanofi-Aventis, a CDK1/CDK2 inhibitor from Amgen, a CDK2 inhibitor from SUGEN-2 (Pfizer), a hearing loss therapy agent (Sound Pharmaceuticals), PD-0332991 (Pfizer), RGB-286199 (GPC Biotech), Ro-0505124 (Hoffmann-La Roche), a Ser/Thr kinase inhibitor from Lilly (EIi Lilly), CVT-2584 (CAS No. 199986-75-9) (CV Therapeutics), and an analogue or derivative thereof.
7170. The method of item 7144 wherein the agent is a cyclooxygenase 1 inhibitor.
7171. The method of item_7144 wherein the agent is a DHFR inhibitor.
7172. The method of item 7144 wherein the agent is a dual integrin inhibitor.
7173. The method of item 7144 wherein the agent is an elastase inhibitor.
7174. The method of item 7144 wherein the agent is an elongation factor-1 alpha inhibitor.
7175. The method of item 7144 wherein the agent is an endothelial growth factor antagonist. 7176. The method of item 7144 wherein the agent is an endothelial growth factor receptor kinase inhibitor selected from the group consisting of sorafenib tosylate (Bayer), AAL-993 (Novartis), ABP-309 (Novartis), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), E- 7080 (Eisai), EG-3306 (Ark Therapeutics), EXEL-2880 (Exelixis), GW- 654652 (GlaxoSmithKline), a KDR inhibitor from LG Life Sciences, CT-6685 and CT-6729 (UCB), KRN-633 and KRN-951 (Kirin Brewery), OSI-930 (OSI Pharmaceuticals), SP-5.2 (Supratek Pharma), SU-11657 (Pfizer), a Tie-2 antagonist (Hybrigenics), a VEGFR-2 kinase inhibitor (Bristol-Myers Squibb), XL-647 (Exelixis), a KDR inhibitor from Abbott Laboratories, sorafenib tosylate, and an analogue or derivative thereof.
7177. The method of item 7144 wherein the agent is an endotoxin antagonist.
7178. The method of item 7144 wherein the agent is an epothilone and tubulin binder.
7179. The method of item 7144 wherein the agent is an estrogen receptor antagonist.
7180. The method of item 7144 wherein the agent is an FGF inhibitor.
7181. The method of item 7144 wherein the agent is a farnexyl transferase inhibitor.
7182. The method of item 7144 wherein the agent is farnesyltransferase inhibitor selected from the group of A-197574 (Abbott), a famesyltransferase inhibitor from Servier, FPTIII (Strathclyde Institute for Drug R), LB-42908 (LG Life Sciences), Pharmaprojects No. 5063 (Genzyme), Pharmaprojects No. 5597 (Ipsen), Yissum Project No. B-1055 (Yissum), and an analogue or derivative thereof.
7183. The method of item 7144 wherein the agent is an FLT-3 kinase inhibitor.
7184. The method of item 7144 wherein the agent is an FGF receptor kinase inhibitor.
7185. The method of item 7144 wherein the agent is a fibrinogen antagonist selected from the group consisting of AUV-201 (Auvation), MG-13926 (Sanofi-Aventis), plasminogen activator (CAS No. 105913-11-9) (from Sanofi-Aventis or UCB), plasminogen activator-2 (tPA- 2) (Sanofi-Aventis), pro-urokinase (CAS No. 82657-92-9) (Sanofi-Aventis), and an analogue or derivative thereof.
7186. The method of item 7144 wherein the agent is a heat shock protein 90 antagonist selected from the group consisting of SRN-005 (Sirenade), geldanamycin, NSC-33050 (17-allylaminogeldanamycin; 17- AAG), 17-dimethylaminoethylamino-17-demethoxy-geldanamycin (17- DMAG), rifabutin (rifamycin XIV, 1',4-didehydro-1-deoxy-1 ,4-dihydro-5'-(2- methylpropyl)-1-oxo-), and an analogue or derivative thereof.
7187. The method of item 7144 wherein the agent is a histone deacetylase inhibitor.
7188. The method of item 7144 wherein the agent is an HMGCoA reductase inhibitor selected from the group consisting of an atherosclerosis therapeutic from Lipid Sciences, ATI-16000 (ARYx Therapeutics), KS-01-019 (Kos Pharmaceuticals), Pharmaprojects No. 2197 (Sanofi-Aventi), RP 61969 (Sanofi-Aventis), and an analogue or derivative thereof.
7189. The method of item 7144 wherein the agent is an ICAM inhibitor.
7190. The method of item 7144 wherein the agent is an IL, ICE and IRAK antagonist, wherein the antagonist is a CJ-14877, CP-424174 (Pfizer), NF-61 (Negma-Lerads), and an analogue or derivative thereof.
7191. The method of item 7144 wherein the agent is an IL-2 inhibitor.
7192. The method of item 7144 wherein the agent is an immunosuppressant selected from the group consisting of teriflunomide (Sanofi Aventis), chlorsulfaquinoxaIone (NSC-339Q04), ._ chlorsulfaquinoxalone sulfate, CS-712 (Sankyo), ismomultin alfa (CAS No. 457913-93-8) (Akzo Nobel), antiallergics from GenPat77, antiinflammatories or AT-005 (And roclus Therapeutics), autoimmune disease therapeutics from EpiVax, BN-007 (Bone), budesonide (CAS No. 51333-22- 3) (MAP Pharmaceuticals), CO-14 (Genzyme), edratide (CAS No. 433922- 67-9) (Teva), EP-314 (Enanta), eprovafen (CAS No. 101335-99-3) (Sanofi- Aventis), HWA-131 (CAS No. 118788-41-3) (Sanofi-Aventis), immunomodulators from MerLion Pharmaceuticals, immunosuppressives from Alchemia, IPL-12 (Inflazyme), MDL-9563 (CAS No. 27086-86-8) (Sanofi-Aventis), Pharmaprojects No. 2330 (Sanofi-Aventis), Pharmaprojects No. 6426 (Abgenix), PXS-25 (Pharmaxis), rosmarinic acid (CAS No. 20283-92-5) (Sanofi-Aventis), RP 42927 or RP 54745 (CAS No. 135330-08-4) (Sanofi-Aventis), SGN-35 (Seattle Genetics), ST-1959 (Sigma-Tau), type I diabetes therapy from SYNX Pharma, UNIL-88 (Debiopharm), VP-025 (Vasogen), VR-694 (Vectura), PRTX-001 (Protalex), and an analogue or derivative thereof.
7193. The method of item 7144 wherein the agent is an IMPDH (inosine monophosphate).
7194. The method of item 7144 wherein the agent is an integrin antagonist.
7195. The method of item 7144 wherein the agent is an interleukin antagonist.
7196. The method of item 7144 wherein the agent is an inhibitor of type III receptor tyrosine kinase.
7197. The method of item 7144 wherein the agent is an irreversible inhibitor of enzyme methionine aminopeptidase type 2.
7198. The method of item 7144 wherein the agent is an isozyme selective delta protein kinase C inhibitor.
7199. The method of item 7144 wherein the agent a JAK3 enzyme inhibitor.
7200. The method of item 7144 wherein the agent is a JNK inhibitor.
7201. The method of item 7144 wherein the agent is a kinase inhibitor. 7202. The method of item 7144 wherein the agent is kinesin antagonist.
7203. The method of item 7144 wherein the agent is a kinesin antagonist.
7204. The method of item 7144 wherein the agent is a leukotriene inhibitor and antagonist selected from the group consisting of ambicromil (CAS No. 58805-38-2) (Sanofi-Aventis), amelubant (CAS No. 346735-24-8) (Boehringer Ingelheim), DW-1141 (Dong Wha), ebselen (Daiichi Pharmaceutical), ibudilast (Kyorin), leucotriene inhibitors from Sanofi-Aventis, lymphotoxin-beta receptor (LT-D) from Biogen Idee, Pharmaprojects No. 1535 and 2728 (CAS No. 119340-33-9) (Sanofi- Aventis), R-112 (Rigel), Rev-5367 (CAS No. 92532-05-3) (Sanofi-Aventis), RG-14893 (CAS No. 141835-49-6) (Sanofi-Aventis), RG-5901-A (CAS No. 101910-24-1), 92532-23-5, RP 66:153.(CAS No, 142422-79-5), RP 66364 (CAS No.186916-92-5), RP 69698 (CAS No. 141748-00-7) (Sanofi-Aventis), SC-411930 (Pfizer), SC-41930 (CAS No. 120072-59-5) (Pfizer), SC-50605 (CAS No. 138828-39-4) (Pfizer), SC-51146 (CAS No. 141059-52-1), SC- 53228 (CAS No. 153633-01-3) (Pfizer), spaglumic acid (ZY-15106) (CAS No.3106-85-2), 80619-64-3 (Novartis), tipredane (CAS No. 85197-77-9) (Bristol-Myers Squibb), U-75302 (CAS No. 119477-85-9) (Pfizer), and analogue or derivative thereof.
7205. The method of item 7144 wherein the agent is an MAP kinase inhibitor.
7206. The method of item 7144 wherein the agent is a matrix metalloproteinase inhibitor. 7207. The method of item 7144 wherein the agent is an MCP- CCR2 inhibitor.
7208. The method of item 7144 wherein the agent is an mTOR inhibitor.
7209. The method of item 7144 wherein the agent is an mTOR kinase inhibitor.
7210. The method of item 7144 wherein the agent is a microtubule inhibitor selected from the group consisting of antibody- maytansinoϊd conjugates from Biogen Idee, colchicines (MantiCore Pharmaceuticals), anticancer immunoconjugates from Johnson & Johnson, DIME from Octamer, gni-1f (GNI), huC242-DM4, huMy9-6-DM1 (ImmunoGen), IDN-5404 (Indena), IMO-098, IMOderm (Imotep), mebendazole. (I ntrogen Therapeutics), microtubule poisons from Cambridge Enterprise, paclitaxel such as LOTAX from Aphios (CAS No. 33069-62-4), Genexol-PM from Samyang, Pharmaprojects No. 6383 (Tapestry Pharmaceuticals), RPR-112378 (Sanofi-Aventis), SGN-75 (Seattle Genetics), SPL-7435 (Starpharma), SSR-250411 (Sanofi-Aventis), trastuzumab-DM1 (Genentech), vinorelbine, and an analogue or derivative thereof.
7211. The method of item 7144 wherein the agent is an MIF inhibitor.
7212. The method of item 7144 wherein the agent is an MMP inhibitor.
7213. The method of item 7144 wherein the agent is a neurokinin (NK) antagonist selected from the group consisting of anthrotainin (CAS No. 148084-40-6) (Sanofi-Aventis), an IBS thereapeutic from ArQuIe, MDL-105212A (CAS No. 167261-60-1) (Ssanofi-Aventis), Pharmaprojects No. 2744, 3258 (CAS No. 139167-47-8) 4006, 4201, or 5986 (Sanofi-Aventis), RP 67580 (CAS No. 135911-02-3), SR-144190 (CAS No. 201152-86-5), SSR-240600, SSR-241586 (Sanofi-Aventis), TKA-457 (Novartis), vestipitant mesylate (CAS No. 334476-64-1) (GlaxoSmithKline), Win-64821 (Sanofi-Aventis), PRX-96026 (Predix Pharmaceuticals), and an analogue or derivative thereof.
7214. The method of item 7144 wherein the agent is an NF kappa B inhibitor selected from the group consisting of AVE-0545 or AVE- 0547 (Sanofi-Aventis), bortezomib (CAS No. 179324-69-7) (Millennium Pharmaceuticals), dexanabinol (CAS No. 112924-45-5) (Pharmos), dexlipotam (Viatris), Pharmaprojects No. 6283 (INDRA) (OXiGENE), IPL- 576092 (CAS No. 137571-30-3) (Inflazyme), NFKB decoy (Corgentech), NFKB decoy oligo (AnGes MG), NFKB's from Ariad, osteoporosis treatments or S5 (F005) from Fulcrum Pharmaceuticals, P61 (Phytopharm), R-flurbiprofen (CAS No. 5104-49-4) (Encore Pharmaceuticals), and an analogue or derivative thereof.
7215. The method of item 7144 wherein the agent is a nitric oxide agonist.
7216. The method of item 7144 wherein the agent is an ornithine decarboxylase inhibitor.
7217. The method of item 7144 wherein the agent is a p38 MAP kinase inhibitor selected from the group consisting of AZD-6703 (AstraZeneca), JX-401 (Jexys Pharmaceuticals), BMS-2 (Bristol-Myers Squibb), a p38 MAP kinase inhibitor from Novartis, a p38-alpha MAP kinase inhibitor from Amphora, Pharmaprojects No. 5704 (Pharmacopeia), RPR- 200765A (Sanofi-Aventis), SD-282 (Johnson & Johnson), TAK-715 (Takeda), and an analogue or derivative thereof.
7218. The method of item 7144 wherein the agent is a palmitoyl-protein thioesterase inhibitor.
7219. The method of item 7144 wherein the agent is a PDGF receptor kinase inhibitor selected from the group consisting of AAL-993, AMN-107, or ABP-309 (Novartis), AMG-706 (Amgen), BAY-57-9352 (Bayer), CDP-860 (UCB), E-7080 (Eisai), imatinib (CAS No. 152459-95-5) (Novartis), OSI-930 (OSI Pharmaceuticals), RPR-127963E (Sanofi-Aventis), RWJ-540973 (Johnson & Johnson), sorafenib tosylate (Bayer), SlM 1657 (Pfizer), tandutinib (CAS No. 387867-13-2) (Millennium Pharmaceuticals), vatalanib (Novartis), ZK-CDK (Schering AG), and an analogue or derivative thereof.
7220. The method of item 7144 wherein the agent is (-)- halofenate (Metabolex), AMG-131 (Amgen), antidiabetics from Japan Tobacco, AZD-4619, AZD-8450, AZD-8677 (AstraZeneca), DRF-10945, balaglitazone (Dr Reddy's), CS-00088, CS-00098 (Chipscreen Biosciences), E-3030 (Eisai), etalocib (CAS No. 161172-51-6) (EIi Lilly), GSK-641597 (Ligand), GSK-677954 (GlaxoSmithKline), GW-409544 (Ligand), GW- 590735 (GlaxoSmithKline), K-111 (Hoffmann-La Roche), LY-518674 (EIi Lilly), LY-674 (Ligand), LY-929 (Ligand), MC-3001, MC-3002 (MaxoCore Pharmaceuticals), metformin HCI + pioglitazone (CAS No. 1115-70-4 and 112529-15-4), ACTOPLUS MET from Andrx), muraglitazar (CAS No. 331741-94-7) (Bristol-Myers Squibb), naveglitazar (Ligand), oieoylethanolamide (Kadmus Pharmaceuticals), ONO-5129, pioglitazone hydrochloride (CAS No. 111025-46-8 and 112529-15-4) (Takeda), PLX-204 (Plexxikon), PPAR agonists from Genfit, PPAR delta agonists from EIi Lilly, PPAR-alpha agonists from CrystalGenomics, PPAR-gamma modulators and PPAR-β modulators from CareX, rosiglitazone maleate (CAS No. 122324- 73-4 or 155141-29-0) (GlaxoSmithKline), rosiglitazone maleate/glimepir (CAS No. 155141-29-0 and 93479-97-1), AVANDARYL, rosiglitazone maleate/metformin extend (CAS No. 155141-29-0 and 657-24-9), AVANDAMET, rosiglitazone maleate+metformin, AVANDAMET (GlaxoSmithKline), tesaglitazar (AstraZeneca), and an analogue or derivative thereof.
7221. The method of item 7144 wherein the agent is a phosphatase inhibitor.
7222. The method of item 7144 wherein the agent is a phosphodiesterase (PDE) inhibitor selected from the group consisting of avanafil (Tanabe Seiyaku), dasantafil (CAS No. 569351-91-3) (Schering- Plough), A-906119 (CAS No. 134072-58-5), DL-850 (Sanofi-Aventis), GRC- 3015, GRC-3566, GRC-3886 (Glenmark), HWA-153 (CAS No, 56395-66-5) (Sanofi-Aventis), hydroxypumafentrine (Altana), IBFB-130011 , IBFB-14-016, IBFB-140301, IBFB-150007, IBFB-211913 (IBFB Pharma), L-826141 (Merck & Co), medorinone (CAS No. 88296-61-1) (Sanofi-Aventis), MEM- 1917 (Memory Pharmaceuticals), ND-1251 (Neuro3d), PDE inhibitors from ICOS, PDE IV inhibitors from Memory Pharmaceuticals and CrystalGenomics, Pharmaprojects No. 2742 and 6141 (Sanofi-Aventis), QAD-171 (Novartis), RHC-2963 (CAS No. 76993-12-9 and 76993-14-1), RPR-117658, RPR-122818 derivatives, SR-24870 , and RPR-132294 (Sanofi-Aventis), SK-350 (ln2Gen), stroke therapy agents from deCODE Genetics, TAS-203 (Taiho), tofimilast (CAS No. 185954-27-2) (Pfizer), UK- 371800 (Pfizer), WIN-65579 (CAS No. 158020-82-7) (Sanofi-Aventis), IBFB- 130020 (IBFB Pharma), OPC-6535 (CAS No. 145739-56-6) (Otsuka), a phosphodiesterase III inhibitor, enoximone, a phosphodiesterase IV inhibitor, fosfosal, Atopik (Barrier Therapeutics), triflusal, a phosphodiesterase V inhibitor, and an analogue or derivative thereof. 7223. The method of item 7144 wherein the agent is a PKC inhibitor.
7224. The method of item 7144 wherein the agent is a platelet activating factor antagonist.
7225. The method of item 7144 wherein the agent is a platelet-derived growth factor receptor kinase inhibitor.
7226. The method of item 7144 wherein the agent is a prolyl hydroxylase inhibitor.
7227. The method of item 7144 wherein the agent is a polymorphonuclear neutrophil inhibitor.
--7228. r The method of item 7144 wherein the agent is a protein, kinase B inhibitor.
7229. The method of item 7144 wherein the agent is a protein kinase C stimulant.
7230. The method of item 7144 wherein the agent is a purine nucleoside analogue.
7231. The method of item 7144 wherein the agent is a purinoreceptor P2X antagonist.
7232. The method of item 7144 wherein the agent is a Raf kinase inhibitor. 7233. The method of item 7144 wherein the agent is a reversible inhibitor of ErbB1 and ErbB2.
7234. The method of item 7144 wherein the agent is a ribonucleoside triphosphate reductase inhibitor.
7235. The method of item 7144 wherein the agent is an SDF- 1 antagonist.
7236. The method of item 7144 wherein the agent is a sheddase inhibitor.
7237. The method of item 7144 wherein the agent is an SRC inhibitor.
. .. . _ 7238. The method of item 71.44 wherein the agent is a stromelysin inhibitor.
7239. The method of item 7144 wherein the agent is an Syk kinase inhibitor.
7240. The method of item 7144 wherein the agent is a telomerase inhibitor.
7241. The method of item 7144 wherein the agent is a TGF beta inhibitor selected from the group consisting of pirfenidone (CAS No. 53179-13-8) (MARNAC), tranilast (CAS No. 53902-12-8) (Kissei), IN-1130 (ln2Gen), mannose-6-phosphate (BTG), TGF-β antagonists from Inflazyme (Pharmaprojects No. 6075), TGF-β antagonists from Sydney, non-industrial source), TGF-β I receptor kinase inhibitors from EIi Lilly, TGF-β receptor inhibitors from Johnson & Johnson, and an analogue or derivative thereof. 7242. The method of item 7144 wherein the agent is a TNFα antagonist or TACE inhibitor selected from the group consisting of adalimumab (CAS No. 331731-18-1) (Cambridge Antibody Technology), AGIX-4207 (AtheroGenics), AGT-1 (Advanced Biotherapy), an antiinflammatory from Borean Pharma, Cellzome, or Paradigm Therapeutics, anti-inflammatory vaccine (TNF-alpha kinoid) from Neovacs, humanized anti-TNF antibody or an anti-TNF MAb (CB0006) Celltech (UCB), apratastat (CAS No. 287445-51-0) (Wyeth), BMS-561392 (Bristol-Myers Squibb), BN- 006 (Bone), certolizumab pegol (CAS No. 428863-50-7 or CH-138 (UCB), cilomilast (CAS No. 153259-65-5) (GlaxoSmithKline), CR-1 (Nuada Pharmaceuticals), CRx-119 (CombinatoRx), D-5410 (UCB), dacopafant (CAS No. 125372-33-0) (Sanofi-Aventis), dersalazine (CAS No. 188913-57- 7/188913-58-8) (Uriach), etanercept (CAS No. 185243-69-0) (Amgen), ethyl pyruvate (Critical (Critical Therapeutics), golimumab (CAS No. 476181-74-5) (Johnson & Johnson), hormono-immunotherapy from Ipsen, CDP571 (e.g., Humicade from UCB), IC-485 (ICOS), infliximab (CAS No. 170277-31-3) (Johnson & Johnson), IP-751 (Manhattan Pharmaceuticals), ISIS-104838 (CAS No. 250755-32-9) (ISIS Pharmaceuticals), lenalidomide (CAS No. 191732-72-6) (Celgene), lentinan (CAS No. 37339-90-5) (Ajinomoto), MDL- 201112 (CAS No. 142130-73-2) (Sanofi-Aventis), medroxyprogesterone (CAS No. 520-85-4) (InKine Pharmaceutical), N-acetylcysteine (CAS No. 616-91-1) (Zambon), NBE-P2 (DIREVO Biotech), nerelimomab (CAS No. 162774-06-3) (Chiron), OM-294DP (OM PHARMA), onercept (CAS No. 199685-57-9) (Yeda), PASSTNF-alpha (Verigen), pentoxifylline or oxypentifylline (Sanofi-Aventis), Pharmaprojects No. 4091 , 4241 , 4295, or 4488 (Sanofi-Aventis), Pharmaprojects No. 5480 (Amgen), Pharmaprojects No. 6749 (Cengent), pirfenidone (CAS No. 53179-13-8) (MARNAC), RPR- 132294 (Sanofi-Aventis), S5 (F002) (Fulcrum Pharmaceuticals), simvastatin (CAS No. 79902-63-9) (Merck & Co), STA-6292 (Synta Pharmaceuticals), tacrolimus (CAS No. 104987-11-3) (Fujisawa LifeCycle Pharma), talactoferrin alfa (CAS No. 308240-58-6) (Agennix), thalidomide (CAS No. 50-35-1) (Celgene), TNF antagonists form ProStrakan, and Synergen, TNF inhibitors (Amgen), TNF-alpha antagonists from Dynavax Technologies and Jerina AG (Germany), TNF-alpha inhibitors from IBFB Pharma and Xencor (Xencor), torbafylline (CAS No. 105102-21-4) (Sanofi-Aventis), UR-1505 (Uriach), VT-346 (Viron Therapeutics), YSIL6 (Y's Therapeutics), YSTH2 (Y's Therapeutics), NPI-1302a-3 (Nereus Pharmaceuticals, a TNF antagonist from Jerina AG (Germany), dersalazine, and an analogue or derivative thereof.
7243. The method of item 7144 wherein the agent is a Toll receptor inhibitor.
7244. The method of item 7144 wherein the agent is a tubulin antagonist.
7245. The method of item 7144 wherein the agent is a - tyrosine kinase inhibitor selected from the group consisting of SU-011248, SUTENT from Pfizer Inc. (New York, NY), BMS-354825, PN-355 (Paracelsian Pharmaceuticals), AGN-199659 (Allergan), AAL-993 or ABP- 309 (Novartis), adaphostin (NIH), AEE-788 (Novartis), AG-013736 (OSI Pharmaceuticals), AG-13736 (Pfizer), ALT-110 (Alteris Therapeutics), AMG- 706 (Amgen), anticancer MAbs from Xencor, anti-EGFrvlll MAbs from Abgenix, anti-HER2 MAb from Abiogen, AZD-2171 or AZD-9935 (AstraZeneca), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), CEP-5214 (Cephalon), CEP-7055 (Cephalon), cetuximab (ImClone Systems), CHIR-200131 and CHIR-258 (Chiron), CP-547632 (OSI Pharmaceuticals), CP-724714 (Pfizer), CT-301 (Creabilis Therapeutics), D- 69491 (Baxter International), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EGFR/ErbB2 inhibitors from Array BioPharma, erlotinib (CAS No. 183319- 69-9) (OSI Pharmaceuticals), EXEL-2880 (Exelixis), FK-778 (Sanofi- Aventis) , gefitinib (CAS No. 184475-35-2) (AstraZeneca), GW-2286 or GW- 654652 (GlaxoSmithKline), her2/neu antigen from AlphaVax, HER-2/neu inhibitor from Generex, Herzyme (Medipad) (Sirna Therapeutics), HKI-272 (Wyeth), HuMax-EGFr (Genmab), idronoxil (CAS No. 81267-65-4) (Novogen), IGF-1 inhibitors from Ontogen, IMC-11 F8 (ImCIone Systems), kahalalide F (CAS No. 149204-42-2) (PharmaMar), KDR inhibitor from LG Life Sciences, KDR inhibitors from Abbott Laboratories, KDR kinase inhibitors (UCB), Kdr kinase inhibitors from Merck & Co, KRN-633 and KRN- 951 (Kirin Brewery), KSB-102 (Xenova), lapatinib ditosylate (CAS No. 388082-78-8) (GlaxoSmithKline), matuzumab (Merck KGaA), MDX-214 (Medarex), ME-103 (Pharmexa), MED-A300 (Gerolymatos), MNAC-13 (Lay Line Genomics), nimotuzumab (Center of Molecular Immunology), NSC- 330507 or NSC-707545 (NIH), NV-50 (Novogen), OSI-930 (OSI Pharmaceuticals), panitumumab (Abgenix), pelitinib (CAS No. 287933-82-7) (Wyeth), pertuzumab (CAS No. 380610-27-5) (Genentech), Pharmaprojects No. 3985 (Sanofi-Aventis), prostate cancer therapeutics from Sequenom (SQPC35, SQPC36, SQPC90), removab and remoxab (Trion Pharma), RG- 13022 (CAS No. 136831-48-6), RG-13291 (CAS No. 138989-50-1), or RG- 14620 (CAS No. 136831-49-7) (Sanofi-Aventis), RM-6427 (Romark), RNAi breast cancer therapy from Benitec, RP 53801 (CAS No. 125882-88-4) (Sanofi-Aventis), sorafenib tosylate (Bayer), SU-11657 (Pfizer), Tie-2 antagonists from Semaia (Hybrigenics), Tie-2 inhibitors from Ontogen, trastuzumab (CAS No. 180288-69-1) (Genentech), tyrosine kinase inhibitors from Sanofi-Aventis, U3-1287, U3-1565, U3-1784, U3-1800 (U3 Pharma), vatalanib (Novartis), VEGFR-2 kinase inhibitor from Bristol-Myers Squibb, XL-647 (Exelixis), ZD-6474 (AstraZeneca), ZK-CDK (Schering AG), an EGFR tyrosine kinase inhibitor, EKB-569 (Wyeth), and an analogue or derivative thereof.
7246. The method of item 7144 wherein the agent is a VEGF inhibitor. 7247. The method of item 7144 wherein the agent is a vitamin D receptor agonist.
7248. The method of item 7144 wherein the agent is ZD-6474 (an angiogenesis inhibitor).
7249. The method of item 7144 wherein the agent is AP- 23573 (an mTOR inhibitor).
7250. The method of item 7144 wherein the agent is synthadotin (a tubulin antagonist).
7251. The method of item 7144 wherein the agent is S-0885 (a collagenase inhibitor).
7252. The method of item 7144 wherein the agent is aplidine (an elongation factor-1 alpha inhibitor).
7253. The method of item 7144 wherein the agent is ixabepilone (an epithilone).
7254. The method of item 7144 wherein the agent is IDN- 5390 (an angiogenesis inhibitor).
7255. The method of item 7144 wherein the agent is SB- 2723005 (an angiogenesis inhibitor).
7256. The method of item 7144 wherein the agent is ABT-518 (an angiogenesis inhibitor). 7257. The method of item 7144 wherein the agent is combretastatin (an angiogenesis inhibitor).
7258. The method of item 7144 wherein the agent is anecortave acetate (an angiogenesis inhibitor).
7259. The method of item 7144 wherein the agent is SB- 715992 (a kinesin antagonist).
7260. The method of item 7144 wherein the agent is temsirolimus (an mTOR inhibitor).
7261. The method of item 7144 wherein the agent is adalimumab (a TNFα antagonist).
-7262. -The method of item 7144, wherein the composition comprises a polymer.
7263. The method of item 7144, wherein the composition comprises a polymer, and the polymer is, or comprises, a copolymer.
7264. The method of item 7144, wherein the composition comprises a polymer, and the polymer is, or comprises, a block copolymer.
7265. The method of item 7144, wherein the composition comprises a polymer, and the polymer is, or comprises, a random copolymer.
7266. The method of item 7144, wherein the composition comprises a polymer, and the polymer is, or comprises, a biodegradable polymer. 7267. The method of item 7144, wherein the composition comprises a polymer, and the polymer is, or comprises, a nonbiodegradable polymer.
7268. The method of item 7144, wherein the composition comprises a polymer, and the polymer is, or comprises, a hydrophilic polymer.
7269. The method of item 7144, wherein the composition comprises a polymer, and the polymer is, or comprises, a hydrophobic polymer.
7270. The method of item 7144, wherein the composition comprises a polymer, and the polymer is, or comprises, a polymer having hydrophilic domains.
7271. The method of item 7144, wherein the composition comprises a polymer, and the polymer is, or comprises, a polymer having hydrophobic domains.
7272. The method of item 7144, wherein the composition comprises a polymer, and the polymer is, or comprises, a non-conductive polymer.
7273. The method of item 7144, wherein the composition comprises a polymer, and the polymer is, or comprises, an elastomer.
7274. The method of item 7144, wherein the composition comprises a polymer, and the polymer is, or comprises, a hydrogel. 7275. The method of item 7144, wherein the composition comprises a polymer, and the polymer is, or comprises, a silicone polymer.
7276. The method of item 7144, wherein the composition comprises a polymer, and the polymer is, or comprises, a hydrocarbon polymer.
7277. The method of item 7144, wherein the composition comprises a polymer, and the polymer is, or comprises, a styrene-derived polymer.
7278. The method of item 7144, wherein the composition comprises a polymer, and the polymer is, or comprises, a butadiene-derived polymer.
7279. The method of item 7144, wherein the composition - comprises a polymer, and the polymer is, or comprises, a macromer.
7280. The method of item 7144, wherein the composition comprises a polymer, and the polymer is, or comprises, a poly(ethylene glycol) polymer.
7281. The method of item 7144, wherein the composition comprises a polymer, and the polymer is, or comprises, an amorphous polymer.
7282. The method of item 7144, wherein the composition further comprises a second pharmaceutically active agent.
7283. The method of item 7144, wherein the composition further comprises an anti-inflammatory agent. 7284. The method of item 7144, wherein the composition further comprises an agent that inhibits infection.
7285. The method of item 7144, wherein the composition further comprises an anthracycline.
7286. The method of item 7144, wherein the composition further comprises doxorubicin.
7287. The method of item 7144 wherein the composition further comprises mitoxantrone.
7288. The method of item 7144 wherein the composition further comprises a fluoropyrimidine.
7289. The method of item 7144, wherein the composition further comprises 5-fluorouracil (5-FU).
7290. The method of item 7144, wherein the composition further comprises a folic acid antagonist.
7291. The method of item 7144, wherein the composition further comprises methotrexate.
7292. The method of item 7144, wherein the composition further comprises a podophylotoxin.
7293. The method of item 7144, wherein the composition further comprises etoposide. 7294. The method of item 7144, wherein the composition further comprises camptothecin.
7295. The method of item 7144, wherein the composition further comprises a hydroxyurea.
7296. The method of item 7144, wherein the composition further comprises a platinum complex.
7297. The method of item 7144, wherein the composition further comprises cisplatin.
7298. The method of item 7144 wherein the composition further comprises an anti-thrombotic agent.
- - - - 7299. The method of item 7144, wherein the composition further comprises a visualization agent.
7300. The method of item 7144, wherein the composition further comprises a visualization agent, and the visualization agent is a radiopaque material, wherein the radiopaque material comprises a metal, a halogenated compound, or a barium containing compound.
7301. The method of item 7144, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, barium, tantalum, or technetium.
7302. The method of item 7144, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, an MRI responsive material. 7303. The method of item 7144, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, a gadolinium chelate.
7304. The method of item 7144, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, iron, magnesium, manganese, copper, or chromium.
7305. The method of item 7144, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, iron oxide compound.
7306. The method of item 7144, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, a dye, pigment, or colorant.
7307. The method of item 7144 wherein the agent is released in effective concentrations from the composition comprising the agent by diffusion over a period ranging from the time of administration to about 90 days.
7308. The method of item 7144 wherein the agent is released in effective concentrations from the composition comprising the agent by erosion of the composition over a period ranging from the time of administration to about 90 days.
7309. The method of item 7144 wherein the composition further comprises an inflammatory cytokine.
7310. The method of item 7144 wherein the composition further comprises an agent that stimulates cell proliferation. 7311. The method of item 7144 wherein the composition further comprises a polymeric carrier.
7312. The method of item 7144 wherein the composition is in the form of a gel, paste, or spray.
7313. The method of item 7144 wherein the implant is partially constructed with the agent or the composition.
7314. The method of item 7144 wherein the implant is fully constructed with the agent or the composition.
7315. The method of item 7144 wherein the implant is impregnated with the agent or the composition.
7316. The method of item 7144, wherein the agent or the composition forms a coating, and the coating directly contacts the implant.
7317. The method of item 7144, wherein the agent or the composition forms a coating, and the coating indirectly contacts the implant.
7318. The method of item 7144 wherein the agent or the composition forms a coating, and the coating partially covers the implant.
7319. The method of item 7144, wherein the agent or the composition forms a coating, and the coating completely covers the implant.
7320. The method of item 7144 wherein the agent or the composition is located within pores or holes of the implant. 7321. The method of item 7144 wherein the agent or the composition is located within a channel, lumen, or divet of the implant.
7322. The method of item 7144 wherein the implant further comprising an echogenic material.
7323. The method of item 7144 wherein the implant further comprises an echogenic material, wherein the echogenic material is in the form of a coating.
7324. The method of item 7144 wherein the implant is sterile.
7325. The method of item 7144 wherein the agent is delivered from the implant, wherein the agent is released into tissue in the vicinity of the implant after deployment of the implant.
7326. The method of item 7144 wherein the agent is delivered from the implant, wherein the agent is released into tissue in the vicinity of the implant after deployment of the implant, wherein the tissue is connective tissue.
7327. The method of item 7144 wherein the agent is delivered from the implant, wherein the agent is released into tissue in the vicinity of the implant after deployment of the implant, wherein the tissue is muscle tissue.
7328. The method of item 7144 wherein the agent is delivered from the implant, wherein the agent is released into tissue in the vicinity of the implant after deployment of the implant, wherein the tissue is nerve tissue. 7329. The method of item 7144 wherein the agent is delivered from the implant, wherein the agent is released into tissue in the vicinity of the implant after deployment of the implant, wherein the tissue is epithelium tissue.
7330. The method of item 7144 wherein the agent is delivered from the implant, wherein the agent is released in effective concentrations from the implant over a period ranging from the time of deployment of the implant to about 1 year.
7331. The method of item 7144 wherein the agent is delivered from the implant, wherein the agent is released in effective concentrations from the implant over a period ranging from about 1 month to 6 months.
7332. The method of item 7144 wherein the agent is delivered from the implant, wherein the agent-is released in effective concentrations from the implant over a period ranging from about 1 - 90 days.
7333. The method of item 7144 wherein the agent is delivered from the implant, wherein the agent is released in effective concentrations from the implant at a constant rate.
7334. The method of item 7144 wherein the agent is delivered from the implant, wherein the agent is released in effective concentrations from the implant at an increasing rate.
7335. The method of item 7144 wherein the agent is delivered from the implant, wherein the agent is released in effective concentrations from the implant at a decreasing rate. 7336. The method of item 7144 wherein the agent is delivered from the implant, wherein the implant comprises about 0.01 Dg to about 10 Dg of the agent.
7337. The method of item 7144 wherein the agent is delivered from the implant, wherein the implant comprises about 10 Dg to about 10 mg of the agent.
7338. The method of item 7144 wherein the agent is delivered from the implant, wherein the implant comprises about 10 mg to about 250 mg of the agent.
7339. The method of item 7144 wherein the agent is delivered from the implant, wherein the implant comprises about 250 mg to about 1000 mg of the agent.
7340. The method of item 7144 wherein the agent is delivered from the implant, wherein the implant comprises about 1000 mg to about 2500 mg of the agent.
7341. The method of item 7144 wherein the agent is delivered from the implant, wherein a surface of the implant comprises less than 0.01 Dg of the agent per mm2 of implant surface to which the agent is applied.
7342. The method of item 7144 wherein the agent is delivered from the implant, wherein a surface of the implant comprises about 0.01 Dg to about 1 Dg of the agent per mm2 of implant surface to which the agent is applied.
7343. The method of item 7144 wherein the agent is delivered from the implant, wherein a surface of the implant comprises about 1 Dg to about 10 Dg of the agent per mm2 of implant surface to which the agent is applied.
7344. The method of item 7144 wherein the agent is delivered from the implant, wherein a surface of the implant comprises about 10 Dg to about 250 Dg of the agent per mm2 of implant surface to which the agent is applied.
7345. The method of item 7144 wherein the agent is delivered from the implant, wherein a surface of the implant comprises about 250 Dg to about 1000 Dg of the agent per mm2 of implant surface to which the agent is applied.
7346. The method of item 7144 wherein the agent is delivered from the implant, wherein a surface of the implant comprises about 1000 Dg to about 2500 Dg of the agent per mm2 of implant surface to which the agent is applied.
7347. The method of item 7144, wherein the implant further comprises a coating, and the coating is a uniform coating.
7348. The method of item 7144, wherein the implant further comprises a coating, and the coating is a non-uniform coating.
7349. The method of item 7144, wherein the implant further comprises a coating, and the coating is a discontinuous coating.
7350. The method of item 7144, wherein the implant further comprises a coating, and the coating is a patterned coating. 7351. The method of item 7144, wherein the implant further comprises a coating, and the coating has a thickness of 100 Dm or less.
7352. The method of item 7144, wherein the implant further comprises a coating, and the coating has a thickness of 10 Dm or less.
7353. The method of item 7144, wherein the implant further comprises a coating, and the coating adheres to the surface of the implant upon deployment of the implant.
7354. The method of item 7144, wherein the implant further comprises a coating, and the coating is stable at room temperature for a period of at least 1 year.
7355. The method of item 7144, wherein the implant further comprises a coating, and the agent is present in the coating in an amount ranging between about 0.0001% to about 1% by weight.
7356. The method of item 7144, wherein the implant further comprises a coating, and the agent is present in the coating in an amount ranging between about 1% to about 10% by weight.
7357. The method of item 7144, wherein the implant further comprises a coating, and the agent is present in the coating in an amount ranging between about 10% to about 25% by weight.
7358. The method of item 7144, wherein the implant further comprises a coating, and the agent is present in the coating in an amount ranging between about 25% to about 70% by weight. 7359. The method of item 7144, wherein the implant further comprises a coating, and the coating comprises a polymer.
7360. The method of item 7144, wherein the implant comprises a first coating having a first composition and a second coating having a second composition.
7361. The method of item 7144, wherein the implant comprises a first coating having a first composition and a second coating having a second composition, wherein the first composition and the second composition are different.
7362. A method as in any one of items 7144-7361 , wherein the device is an intraocular lens device for preventing lens opacification.
7363. A method as in any one of items 7144-7361 , wherein the device is a polymethylmethacrylate intraocular lens.
7364. A method as in any one of items 7144-7361 , wherein the device is a silicone intraocular lens.
7365. A method as in any one of items 7144-7361 , wherein the device is an achromatic lens.
7366. A method as in any one of items 7144-7361, wherein the device is a pseudophako.
7367. A method as in any one of items 7144-7361, wherein the device is a phakic lens. 7368. A method as in any one of items 7144-7361 , wherein the device is an aphakic lens.
7369. A method as in any one of items 7144-7361, wherein the device is a multi-focal intraocular lens.
7370. A method as in any one of items 7144-7361, wherein the device is a hydrophilic and hydrophobic acrylic intraocular lens.
7371. A method as in any one of items 7144-7361 , wherein the device is an intraocular implant.
7372. A method as in any one of items 7144-7361 , wherein the device is an optic lens.
- 7373. A method as in any one of items-7144-7361 , wherein the device is a rigid gas permeable lens.
7374. A method as in any one of items 7144-7361, wherein the device is a foldable intraocular lens.
7375. A method as in any one of items 7144-7361 , wherein the device is a rigid intraocular lens.
7376. A method as in any one of items 7144-7361 , wherein the device is a corrective implant for vision impairment.
7377. A method as in any one of items 7144-7361 , wherein the device is an intraocular implant adapted for being used in conjunction with a transplant for the cornea. 7378. A method as in any one of items 7144-7361, wherein the device is an intraocular implant adapted for being used in conjunction with treatment of secondary cataract after extracapsular cataract extraction.
7379. A method for inhibiting scarring comprising placing a device that comprises a medical device for treating hypertropic scar or keloid (e.g., an implant) and an anti-scarring agent or a composition comprising an anti-scarring agent into an animal host, wherein the agent inhibits scarring.
7380. The method of item 7379 wherein the agent is an adensosine A2A receptor antagonist.
7381. The method of item 7379 wherein the agent is an AKT inhibitor.
7382. The method of item 7379 wherein the agent is an alpha 2 integrin antagonist, wherein the alpha 2 integrin antagonist is Pharmaprojects No. 5754 (Merck KgaA).
7383. The method of item 7379 wherein the agent is an alpha 4 integrin antagonist.
7384. The method of item 7379 wherein the agent is an alpha 7 nicotinic receptor agonist.
7385. The method of item 7379 wherein the agent is an angiogenesis inhibitor selected from the group consisting of AG-12,958 (Pfizer), ATN-161 (Attenuon LLC), neovastat, an angiogenesis inhibitor from Jerina AG (Germany), NM-3 (Mercian), VGA-1155 (Taisho), FCE-26644 (Pfizer), FCE-26950 (Pfizer), FPMA (Meiji Daries), FR-111142 (Fujisawa), GGTI-298, GM-1306 (Ligand), GPA-1734 (Novartis), NNC-47-0011 (Novo Nordisk), herbamycin (Nippon Kayaku), lenalidomide (Celegene), IP-10 (NIH), ABT-828 (Abbott), KIN-841 (Tokushima University, Japan), SF-1126 (Semafore Pharmaceuticals), laminin technology (NIH), CHIR-258 (Chiron), NVP-AEW541 (Novartis), NVP-AEW541 (Novartis), Vt16907 (Alchemia), OXI-8007 (Oxigene), EG-3306 (Ark Therapeutics), Maspin (Arriva), ABT- 567 (Abbott), PPI-2458 (Praecis Pharmaceuticals), CC-5079, CC-4089 (Celgene), HIF-1 alpha inhibitors (Xenova), S-247 (Pfizer), AP-23573 (Ariad), AZD-9935 (Astra Zeneca), mebendazole (Introgen Therapeutics), MetAP-2 inhibitors (GlaxoSmithKline), AG-615 (Angiogene Pharmaceuticals), Tie-2 antagonists (Hybrigenics), NC-381, CYC-381 , NC-169, NC-219, NC-383, NC-384, NC-407 (Lorus Therapeutics), ATN-224 (Attenuon), ON-01370 (Onconova), Vitronectin antagonists (Amgen), SDX-103 (Salmedix), Vitronectin antagonists (Shire), CHP (Riemser), TEK (Amgen), Anecortave acetate (Alcon), T46.2 (Matrix Therapeutics), HG-2 (Heptagen), TEM antagonists (Genzyme), Oxi-4500 (Oxigene), ATN-161 (Attenuon), WX-293 (Wilex), M-2025 (Metris Therapeutics), Alphastatin (BioActa), YH-16 (Yantai Rongchang), BIBF-1120 (Boehringer Ingelheim), BAY-57-9352 (Bayer), AS- 1404 (Cancer Research Technology), SC-77964 (Pfizer), glycomimetics (BioTie Therapies), TIE-2 Inhibitors (Ontogen), DIMI, Octamer (Octamer), ABR-215050 (Active Biotech), ABT-518 (Abbott), KDR inhibitors (Abbott), BSF-466895 (Abbott), SCH-221153 (Schering-Plough), DAC:antiangiogenic (ConjuChem), TFPI (EntreMed), AZD-2171 (Astra-Zenaca), CDC-394 (Celgene), LY290293 (EIi Lilly), IDN-5390 (Indena), Kdr Kinase Inhibitors (Merck), CT-113020, CT-116433, CT-116563, CT-31890, CT-32228) (Cell Therapeutics), A-299620 (Abbott), TWEAK Inhibitor (Amgen), VEGF modulators (Johnson and Johnson), Tum-N53, tumstatin (Genzyme), Thios- 1 , Thios-2 (Thios Pharmaceuticals), MV-6401 (Miravant Medical Technologies), Spisulosine (PharmaMar), CEP-7055 (Cephalon), AUV-201 (Auvation), LM-609 (EIi Lilly), SKF-106615 (AnorMED), Oglufanide disodium (Cytran), BW-114 (Pharminox), Calreticulin (NIH), WX-678 (Wilex), SD-7784 (Pfizer), WX-UK1 (Wilex), SH-268 (Schering AG), 2-Me-PGA (Celgene), S- 137 (Pfizer), ZD-6126 (Angiogene Pharmaceuticals), SG-292 (SignalGen), Benefin (Lane Labs), A6, A36 (Angstrom), SB-2723005 (GlaxoSmithKline), SC-7 (Cell Therapeutics), ZEN-014 (AEterna Zentaris), 2-methoxyestradiol (EntreMed), NK-130119 (Nippon Kayaku), CC-10004 (Celgene), AVE- 8062A (Ajinomoto), Tacedinaline (Pfizer), Actinonin (Tokyo Metropolitan Institute of Medical Science), Lenalidomide (Celgene), VGA-1155, BTO-956 (SRI International), ER-68203-00 (Eisai), CT-6685 (UCB), JKC-362 (Phoenix Pharmaceuticals), DMI-3798 (DMI Biosciences, Angiomate (Ipsen), ZD-6474 (AstraZeneca), CEP-5214 (Cephalon), Canstatin (Genzyme), NM-3 (Mercian), Oridigm (MediQuest Therapeutics), Exherin (Adherex), BLS-0597 (Boston Life Sciences), PTC-299 (PTC Therapeutics), NPI-2358 (Nereus Pharmaceuticals), CGP-79787 (Novartis), AEE-788 (Novartis), CKD-732 (Chong Kun Dang), CP-564959 (OSI Pharmaceuticals), CM-101 (CarboMed), CT-2584, CT3501 (Cell Therapeutics), combretastatin and analogues and derivatives-thereof (Oxigene), Rebimastat (Bristol-Meyers Squibb), Dextrin 2-sulfate (ML Laboratories), Cilengitide (Merk KGaA), NSC-706704 (Pharminox), KRN- 951 (Kirin Brewery), Ukrain, NSC-631570 (Nowicky Pharma), Tecogalan sodium (Daiichi Pharmaceutical), Tz-93 (Tsumura), TBC-1635 (Encysive Pharmaceuticals), TAN-1120 (Takeda), Semaxanib (Pfizer), BDI-7800 (Biopharmacopae), SD-186, SD-983 (Bristol-Meyers Squibb), SB-223245 (GlaxoSmithKline), SC-236 (Pfizer), RWJ-590973 (Johnson and Johnson), ILX-1850 (Genzyme), SC-68488, S-836 (Pfizer), CG-55069-11 (CuraGen), Ki-23057 (Kirin Brewery), CCX-700 (Chemoentryx), Pegaptanib octasodium (Giled Sciences), ANGIOCOL (available from Biostratum Inc.), or an analogue or derivative thereof.
7386. The method of item 7379 wherein the agent is an apoptosis antagonist. 7387. The method of item 7379 wherein the agent is an apoptosis activator.
7388. The method of item 7379 wherein the agent is a beta 1 integrin antagonist.
7389. The method of item 7379 wherein the agent is a beta tubulin inhibitor.
7390. The method of item 7379 wherein the agent is a blocker of enzyme production in Hepatitis C.
7391. The method of item 7379 wherein the agent is a Bruton's tyrosine kinase inhibitor.
- 7392. The method of item 7379 wherein the agent is a calcineurin inhibitor.
7393. The method of item 7379 wherein the agent is a caspase 3 inhibitor.
7394. The method of item 7379 wherein the agent is a CC chemokine receptor antagonist.
7395. The method of item 7379 wherein the agent is a cell cycle inhibitor selected from the group consisting of SNS-595 (Sunesis), synthadotin, KRX-0403, and an analogue or derivative thereof.
7396. The method of item 7379 wherein the agent is a cathepsin B inhibitor. 7397. The method of item 7379 wherein the agent is a cathepsin K inhibitor, wherein the cathepsin K inhibitor is 462795 (GlaxoSmithKline), INPL-022-D6 (Amura Therapeutics), or an analogue or derivative thereof.
7398. The method of item 7379 wherein the agent is a cathepsin L inhibitor.
7399. The method of item 7379 wherein the agent is a CD40 antagonist.
7400. The method of item 7379 wherein the agent is a chemokine receptor agonist.
7401. The method of item 7379 wherein the agent is a chymase inhibitor. . . . . - . .. _ _ . ..
7402. The method of item 7379 wherein the agent is a collagenase antagonist.
7403. The method of item 7379 wherein the agent is a CXCR antagonist.
7404. The method of item 7379 wherein the agent is a cyclin dependent kinase inhibitor selected from the group consisting of a CDK-1 inhibitor, a CDK-2 inhibitor, a CDK- 4 inhibitor, a CDK-6 inhibitor, a CAK1 inhibitor from GPC Biotech and Bristol-Myers Squibb, RGB-286199 (GPC Biotech), an anticancer agent from Astex Technology, a CAK1 inhibitor from GPC Biotech, a CDK inhibitor from Sanofi-Aventis, a CDK1/CDK2 inhibitor from Amgen, a CDK2 inhibitor from SUGEN-2 (Pfizer), a hearing loss therapy agent (Sound Pharmaceuticals), PD-0332991 (Pfizer), RGB-286199 (GPC Biotech), Ro-0505124 (Hoffmann-La Roche), a Ser/Thr kinase inhibitor from Lilly (EIi Lilly), CVT-2584 (CAS No. 199986-75-9) (CV Therapeutics), and an analogue or derivative thereof.
7405. The method of item 7379 wherein the agent is a cyclooxygenase 1 inhibitor.
7406. The method of item 7379 wherein the agent is a DHFR inhibitor.
7407. The method of item 7379 wherein the agent is a dual integrin inhibitor.
7408. The method of item 7379 wherein the agent is an elastase inhibitor.
7409. The method of item 7379 wherein the agent is an elongation factor-1 alpha inhibitor.
7410. The method of item 7379 wherein the agent is an endothelial growth factor antagonist.
7411. The method of item 7379 wherein the agent is an endothelial growth factor receptor kinase inhibitor selected from the group consisting of sorafenib tosylate (Bayer), AAL-993 (Novartis), ABP-309 (Novartis), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), E- 7080 (Eisai), EG-3306 (Ark Therapeutics), EXEL-2880 (Exelixis), GW- 654652 (GlaxoSmithKline), a KDR inhibitor from LG Life Sciences, CT-6685 and CT-6729 (UCB), KRN-633 and KRN-951 (Kirin Brewery), OSI-930 (OSI Pharmaceuticals), SP-5.2 (Supratek Pharma), SU-11657 (Pfizer), a Tie-2 antagonist (Hybrigenics), a VEGFR-2 kinase inhibitor (Bristol-Myers Squibb), XL-647 (Exelixis), a KDR inhibitor from Abbott Laboratories, sorafenib tosylate, and an analogue or derivative thereof.
7412. The method of item 7379 wherein the agent is an endotoxin antagonist.
7413. The method of item 7379 wherein the agent is an epothilone and tubulin binder.
7414. The method of item 7379 wherein the agent is an estrogen receptor antagonist.
7415. The method of item 7379 wherein the agent is an FGF inhibitor.
7416. The method of item 7379 wherein the agent is a farnexyl transferase inhibitor.
7417. The method of item 7379 wherein the agent is farnesyltransferase inhibitor selected from the group of A-197574 (Abbott), a farnesyltransferase inhibitor from Servier, FPTIII (Strathclyde Institute for Drug R), LB-42908 (LG Life Sciences), Pharmaprojects No. 5063 (Genzyme), Pharmaprojects No. 5597 (Ipsen), Yissum Project No. B-1055 (Yissum), and an analogue or derivative thereof.
7418. The method of item 7379 wherein the agent is an FLT-3 kinase inhibitor.
7419. The method of item 7379 wherein the agent is an FGF receptor kinase inhibitor. 7420. The method of item 7379 wherein the agent is a fibrinogen antagonist selected from the group consisting of AUV-201 (Auvation), MG-13926 (Sanofi-Aventis), plasminogen activator (CAS No. 105913-11-9) (from Sanofi-Aventis or UCB), plasminogen activator-2 (tPA- 2) (Sanofi-Aventis), pro-urokinase (CAS No. 82657-92-9) (Sanofi-Aventis), and an analogue or derivative thereof.
7421. The method of item 7379 wherein the agent is a heat shock protein 90 antagonist selected from the group consisting of SRN-005 (Sirenade), geldanamycin, NSC-33050 (17-allylaminogeldanamycin; 17- AAG), 17-dimethylaminoethylamino-17-demethoxy-geldanamycin (17- DMAG), rifabutin (rifamycin XIV, 1',4-didehydro-1-deoxy-1 ,4-dihydro-5'-(2- methylpropyl)-1-oxo-), and an analogue or derivative thereof.
7422. The method of item 7379 wherein the agent is a histone deacetylase inhibitor.-
7423. The method of item 7379 wherein the agent is an HMGCoA reductase inhibitor selected from the group consisting of an atherosclerosis therapeutic from Lipid Sciences, ATI-16000 (ARYx Therapeutics), KS-01-019 (Kos Pharmaceuticals), Pharmaprojects No. 2197 (Sanofi-Aventi), RP 61969 (Sanofi-Aventis), and an analogue or derivative thereof.
7424. The method of item 7379 wherein the agent is an ICAM inhibitor.
7425. The method of item 7379 wherein the agent is an IL, ICE and IRAK antagonist, wherein the antagonist is a CJ-14877, CP-424174 (Pfizer), NF-61 (Negma-Lerads), and an analogue or derivative thereof. 7426. The method of item 7379 wherein the agent is an IL-2 inhibitor.
7427. The method of item 7379 wherein the agent is an immunosuppressant selected from the group consisting of teriflunomide (Sanofi Aventis), chlorsulfaquinoxalone (NSC-339004), chlorsulfaquinoxalone sulfate, CS-712 (Sankyo), ismomultin alfa (CAS No. 457913-93-8) (Akzo Nobel), antiallergics from GenPat77, antiinflammatories or AT-005 (Androclus Therapeutics), autoimmune disease therapeutics from EpiVax, BN-007 (Bone), budesonide (CAS No. 51333-22- 3) (MAP Pharmaceuticals), CO-14 (Genzyme), edratide (CAS No. 433922- 67-9) (Teva), EP-314 (Enanta), eprovafen (CAS No. 101335-99-3) (Sanofi- Aventis), HWA-131 (CAS No. 118788-41-3) (Sanofi-Aventis), immunomodulators from MerLion Pharmaceuticals, immunosuppressives from Alchemia, IPL-12 (Inflazyme), MDL-9563 (CAS No. 27086-86-8) (Sanofi-Aventis), Pharmaprojects No. 2330 (Sanofi-Aventis), Pharmaprojects No. 6426 (Abgenix), PXS-25 (Pharmaxis), rosmarinic acid (CAS No. 20283-92-5) (Sanofi-Aventis), RP 42927 or RP 54745 (CAS No. 135330-08-4) (Sanofi-Aventis), SGN-35 (Seattle Genetics), ST-1959 (Sigma-Tau), type I diabetes therapy from SYNX Pharma, UNIL-88 (Debiopharm), VP-025 (Vasogen), VR-694 (Vectura), PRTX-001 (Protalex), and an analogue or derivative thereof.
7428. The method of item 7379 wherein the agent is an IMPDH (inosine monophosphate).
7429. The method of item 7379 wherein the agent is an integrin antagonist.
7430. The method of item 7379 wherein the agent is an interleukin antagonist. 7431. The method of item 7379 wherein the agent is an inhibitor of type III receptor tyrosine kinase.
7432. The method of item 7379 wherein the agent is an irreversible inhibitor of enzyme methionine aminopeptidase type 2.
7433. The method of item 7379 wherein the agent is an isozyme selective delta protein kinase C inhibitor.
7434. The method of item 7379 wherein the agent a JAK3 enzyme inhibitor.
7435. The method of item 7379 wherein the agent is a JNK inhibitor.
7436. The method of item 7379 wherein the agent is a kinase inhibitor.
7437. The method of item 7379 wherein the agent is kinesin antagonist.
7438. The method of item 7379 wherein the agent is a kinesin antagonist.
7439. The method of item 7379 wherein the agent is a leukotriene inhibitor and antagonist selected from the group consisting of ambicromil (CAS No. 58805-38-2) (Sanofi-Aventis), amelubant (CAS No. 346735-24-8) (Boehringer Ingelheim), DW-1141 (Dong Wha), ebselen (Daiichi Pharmaceutical), ibudilast (Kyorin), leucotriene inhibitors from Sanofi-Aventis, lymphotoxin-beta receptor (LT-D) from Biogen Idee, Pharmaprojects No. 1535 and 2728 (CAS No. 119340-33-9) (Sanofi- Aventis), R-112 (Rigel), Rev-5367 (CAS No. 92532-05-3) (Sanofi-Aventis), RG-14893 (CAS No. 141835-49-6) (Sanofi-Aventis), RG-5901-A (CAS No. 101910-24-1), 92532-23-5, RP 66153 (CAS No. 142422-79-5), RP 66364 (CAS No.186916-92-5), RP 69698 (CAS No. 141748-00-7) (Sanofi-Aventis), SC-411930 (Pfizer), SC-41930 (CAS No. 120072-59-5) (Pfizer), SC-50605 (CAS No. 138828-39-4) (Pfizer), SC-51146 (CAS No. 141059-52-1), SC- 53228 (CAS No. 153633-01-3) (Pfizer), spaglumic acid (ZY-15106) (CAS No.3106-85-2), 80619-64-3 (Novartis), tipredane (CAS No. 85197-77-9) (Bristol-Myers Squibb), U-75302 (CAS No. 119477-85-9) (Pfizer), and analogue or derivative thereof.
7440. The method of item 7379 wherein the agent is an MAP kinase inhibitor.
7441. The method of item 7379 wherein the agent is a matrix metalloproteinase inhibitor.
7442. The method of item 7379 wherein the agent is an MCP- CCR2 inhibitor.
7443. The method of item 7379 wherein the agent is an mTOR inhibitor.
7444. The method of item 7379 wherein the agent is an mTOR kinase inhibitor.
7445. The method of item 7379 wherein the agent is a microtubule inhibitor selected from the group consisting of antibody- maytansinoid conjugates from Biogen Idee, colchicines (MantiCore Pharmaceuticals), anticancer immunoconjugates from Johnson & Johnson, DIME from Octamer, gni-1f (GNI), huC242-DM4, huMy9-6-DM1 (ImmunoGen), IDN-5404 (Indena), IMO-098, IMOderm (Imotep), mebendazole (Introgen Therapeutics), microtubule poisons from Cambridge Enterprise, paclitaxel such as LOTAX from Aphios (CAS No. 33069-62-4), Genexol-PM from Samyang, Pharmaprojects No. 6383 (Tapestry Pharmaceuticals), RPR-112378 (Sanofi-Aventis), SGN-75 (Seattle Genetics), SPL-7435 (Starpharma), SSR-250411 (Sanofi-Aventis), trastuzumab-DM1 (Genentech), vinorelbine, and an analogue or derivative thereof.
7446. The method of item 7379 wherein the agent is an MIF inhibitor.
7447. The method of item 7379 wherein the agent is an MMP inhibitor.
7448. The method of item 7379 wherein the agent is a neurokinin (NK) antagonist selected from the group consisting of anthrotainin (CAS No. 148084-40-6) (Sanofi-Aventis), an IBS thereapeutic from ArQuIe, MDL-105212A (CAS No. 167261-60-1) (Ssanofi-Aventis), Pharmaprojects No. 2744, 3258 (CAS No. 139167-47-8) 4006, 4201 , or 5986 (Sanofi-Aventis), RP 67580 (CAS No. 135911-02-3), SR-144190 (CAS No. 201152-86-5), SSR-240600, SSR-241586 (Sanofi-Aventis), TKA-457 (Novartis), vestipitant mesylate (CAS No. 334476-64-1) (GlaxoSmithKline), Win-64821 (Sanofi-Aventis), PRX-96026 (Predix Pharmaceuticals), and an analogue or derivative thereof.
7449. The method of item 7379 wherein the agent is an NF kappa B inhibitor selected from the group consisting of AVE-0545 or AVE- 0547 (Sanofi-Aventis), bortezomib (CAS No. 179324-69-7) (Millennium Pharmaceuticals), dexanabinol (CAS No. 112924-45-5) (Pharmos), dexlipotam (Viatris), Pharmaprojects No. 6283 (INDRA) (OXiGENE), IPL- 576092 (CAS No. 137571-30-3) (Inflazyme), NFKB decoy (Corgentech), NFKB decoy oligo (AnGes MG), NFKB's from Ariad, osteoporosis treatments or S5 (F005) from Fulcrum Pharmaceuticals, P61 (Phytopharm), R-flurbiprofen (CAS No. 5104-49-4) (Encore Pharmaceuticals), and an analogue or derivative thereof.
7450. The method of item 7379 wherein the agent is a nitric oxide agonist.
7451. The method of item 7379 wherein the agent is an ornithine decarboxylase inhibitor.
7452. The method of item 7379 wherein the agent is a p38 MAP kinase inhibitor selected from the group consisting of AZD-6703 (AstraZeneca), JX-401 (Jexys Pharmaceuticals), BMS-2 (Bristol-Myers Squibb), a p38 MAP kinase inhibitor from Novartisτ a p38-alpha MAP kinase inhibitor from Amphora, Pharmaprojects No. 5704 (Pharmacopeia), RPR- 200765A (Sanofi-Aventis), SD-282 (Johnson & Johnson), TAK-715 (Takeda), and an analogue or derivative thereof.
7453. The method of item 7379 wherein the agent is a palmitoyl-protein thioesterase inhibitor.
7454. The method of item 7379 wherein the agent is a PDGF receptor kinase inhibitor selected from the group consisting of AAL-993, AMN-107, or ABP-309 (Novartis), AMG-706 (Amgen), BAY-57-9352 (Bayer), CDP-860 (UCB), E-7080 (Eisai), imatinib (CAS No. 152459-95-5) (Novartis), OSI-930 (OSI Pharmaceuticals), RPR-127963E (Sanofi-Aventis), RWJ-540973 (Johnson & Johnson), sorafenib tosylate (Bayer), SU-11657 (Pfizer), tandutinib (CAS No. 387867-13-2) (Millennium Pharmaceuticals), vatalanib (Novartis), ZK-CDK (Schering AG), and an analogue or derivative thereof.
7455. The method of item 7379 wherein the agent is (-)- halofenate (Metabolex), AMG-131 (Amgen), antidiabetics from Japan Tobacco, AZD-4619, AZD-8450, AZD-8677 (AstraZeneca), DRF-10945, balaglitazone (Dr Reddy's), CS-00088, CS-00098 (Chipscreen Biosciences), E-3030 (Eisai), etalocib (CAS No. 161172-51-6) (EIi Lilly), GSK-641597 (Ligand), GSK-677954 (GlaxoSmithKline), GW-409544 (Ligand), GW- 590735 (GlaxoSmithKline), K-111 (Hoffmann-La Roche), LY-518674 (EIi Lilly), LY-674 (Ligand), LY-929 (Ligand), MC-3001 , MC-3002 (MaxoCore Pharmaceuticals), metformin HCI + pioglitazone (CAS No. 1115-70-4 and
112529-15-4), ACTOPLUS MET from Andrx), muraglitazar (CAS No. 331741-94-7) (Bristol-Myers Squibb), naveglitazar (Ligand), oleoylethanolamide (Kadmus Pharmaceuticals), ONO-5129, pioglitazone hydrochloride (CAS No; 111025-46-8 and 112529-15-4) (Takeda), PLX-204 - (Plexxikon), PPAR agonists from Genfit, PPAR delta agonists from EIi Lilly, PPAR-alpha agonists from CrystalGenomics, PPAR-gamma modulators and PPAR-β modulators from CareX, rosiglitazone maleate (CAS No. 122324- 73-4 or 155141-29-0) (GlaxoSmithKline), rosiglitazone maleate/glimepir (CAS No. 155141-29-0 and 93479-97-1), AVANDARYL, rosiglitazone maleate/metformin extend (CAS No. 155141-29-0 and 657-24-9), AVANDAMET, rosiglitazone maleate+metformin, AVANDAMET (GlaxoSmithKline), tesaglitazar (AstraZeneca), and an analogue or derivative thereof.
7456. The method of item 7379 wherein the agent is a phosphatase inhibitor.
7457. The method of item 7379 wherein the agent is a phosphodiesterase (PDE) inhibitor selected from the group consisting of avanafil (Tanabe Seiyaku), dasantafil (CAS No. 569351-91-3) (Schering- Plough), A-906119 (CAS No. 134072-58-5), DL-850 (Sanofi-Aventis), GRC- 3015, GRC-3566, GRC-3886 (Glenmark), HWA-153 (CAS No. 56395-66-5) (Sanofi-Aventis), hydroxypumafentrine (Altana), IBFB-130011 , IBFB-14-016, IBFB-140301 , IBFB-150007, IBFB-211913 (IBFB Pharma), L-826141 (Merck & Co), medorinone (CAS No. 88296-61-1) (Sanofi-Aventis), MEM- 1917 (Memory Pharmaceuticals), ND-1251 (Neuro3d), PDE inhibitors from ICOS, PDE IV inhibitors from Memory Pharmaceuticals and CrystalGenomics, Pharmaprojects No. 2742 and 6141 (Sanofi-Aventis), QAD-171 (Novartis), RHC-2963 (CAS No. 76993-12-9 and 76993-14-1), RPR-117658, RPR-122818 derivatives, SR-24870 , and RPR-132294 (Sanofi-Aventis), SK-350 (ln2Gen), stroke therapy agents from deCODE Genetics, TAS-203 (Taiho), tofimilast (CAS No. 185954-27-2) (Pfizer), UK- 371800 (Pfizer), WIN-65579 (CAS No. 158020-82-7) (Sanofi-Aventis), IBFB- 130020 (IBFB Pharma), OPC-6535 (CAS No. 145739-56-6) (Otsuka), a phosphodiesterase III inhibitor, enoximone, a phosphodiesterase IV inhibitor, fosfosal, Atopik (Barrier Therapeutics), triflusal, a phosphodiesterase V inhibitor, and an analogue or derivative thereof.
7458. The method of item 7379 wherein the agent is a PKC inhibitor.
7459. The method of item 7379 wherein the agent is a platelet activating factor antagonist.
7460. The method of item 7379 wherein the agent is a platelet-derived growth factor receptor kinase inhibitor.
7461. The method of item 7379 wherein the agent is a prolyl hydroxylase inhibitor. 7462. The method of item 7379 wherein the agent is a polymorphonuclear neutrophil inhibitor.
7463. The method of item 7379 wherein the agent is a protein kinase B inhibitor.
7464. The method of item 7379 wherein the agent is a protein kinase C stimulant.
7465. The method of item 7379 wherein the agent is a purine nucleoside analogue.
7466. The method of item 7379 wherein the agent is a purinoreceptor P2X antagonist.
7467. The method of item 7379 wherein the agent is a Raf kinase inhibitor.
7468. The method of item 7379 wherein the agent is a reversible inhibitor of ErbB1 and ErbB2.
7469. The method of item 7379 wherein the agent is a ribonucleoside triphosphate reductase inhibitor.
7470. The method of item 7379 wherein the agent is an SDF- 1 antagonist.
7471. The method of item 7379 wherein the agent is a sheddase inhibitor. 7472. The method of item 7379 wherein the agent is an SRC inhibitor.
7473. The method of item 7379 wherein the agent is a stromelysin inhibitor.
7474. The method of item 7379 wherein the agent is an Syk kinase inhibitor.
7475. The method of item 7379 wherein the agent is a telomerase inhibitor.
7476. The method of item 7379 wherein the agent is a TGF beta inhibitor selected from the group consisting of pirfenidone (CAS No. 53179-13-8) (MARNAC)1 tranilast (CAS No. 53902-12-8) (Kissei), IN-1130 (ln2Gen), mannose-6-phosphate (BTG)J TGF-β antagonists from Inflazyme (Pharmaprojects No. 6075), TGF-β antagonists from Sydney, non-industrial source), TGF-β I receptor kinase inhibitors from EIi Lilly, TGF-β receptor inhibitors from Johnson & Johnson, and an analogue or derivative thereof.
7477. The method of item 7379 wherein the agent is a TNFα antagonist or TACE inhibitor selected from the group consisting of adalimumab (CAS No. 331731-18-1) (Cambridge Antibody Technology), AGIX-4207 (AtheroGenics), AGT-1 (Advanced Biotherapy), an antiinflammatory from Borean Pharma, Cellzome, or Paradigm Therapeutics, anti-inflammatory vaccine (TNF-alpha kinoid) from Neovacs, humanized anti-TNF antibody or an anti-TNF MAb (CB0006) Celltech (UCB), apratastat (CAS No. 287445-51-0) (Wyeth), BMS-561392 (Bristol-Myers Squibb), BN- 006 (Bone), certolizumab pegol (CAS No. 428863-50-7 or CH-138 (UCB), cilomilast (CAS No. 153259-65-5) (GlaxoSmithKline), CR-1 (Nuada Pharmaceuticals), CRx-119 (CombinatoRx), D-5410 (UCB), dacopafant (CAS No. 125372-33-0) (Sanofi-Aventis), dersalazine (CAS No. 188913-57- 7/188913-58-8) (Uriach), etanercept (CAS No. 185243-69-0) (Amgen), ethyl pyruvate (Critical (Critical Therapeutics), golimumab (CAS No. 476181-74-5) (Johnson & Johnson), hormono-immunotherapy from Ipsen, CDP571 (e.g., Humicade from UCB), IC-485 (ICOS), infliximab (CAS No. 170277-31-3) (Johnson & Johnson), IP-751 (Manhattan Pharmaceuticals), ISIS-104838 (CAS No. 250755-32-9) (ISIS Pharmaceuticals), lenalidomide (CAS No. 191732-72-6) (Celgene), lentinan (CAS No. 37339-90-5) (Ajinomoto), MDL- 201112 (CAS No. 142130-73-2) (Sanofi-Aventis), medroxyprogesterone (CAS No. 520-85-4) (InKine Pharmaceutical), N-acetylcysteine (CAS No. 616-91-1) (Zambon), NBE-P2 (DIREVO Biotech), nerelimomab (CAS No. 162774-06-3) (Chiron), OM-294DP (OM PHARMA), onercept (CAS No. 199685-57-9) (Yeda), PASSTNF-alpha (Verigen), pentoxifylline or oxypentifylline (Sanofi-Aventis), Pharmaprojects No. 4091 , 4241 , 4295, or 4488 (Sanofi-Aventis), Pharmaprojects No. 5480 (Amgen), Pharmaprojects No. 6749 (Gengent), pirfenidone (CAS No: 53179-13-8) (MARNAC), RPR- 132294 (Sanofi-Aventis), S5 (F002) (Fulcrum Pharmaceuticals), simvastatin (CAS No. 79902-63-9) (Merck & Co), STA-6292 (Synta Pharmaceuticals), tacrolimus (CAS No. 104987-11-3) (Fujisawa LifeCycle Pharma), talactoferrin alfa (CAS No. 308240-58-6) (Agennix), thalidomide (CAS No. 50-35-1) (Celgene), TNF antagonists form ProStrakan, and Synergen, TNF inhibitors (Amgen), TNF-alpha antagonists from Dynavax Technologies and Jerina AG (Germany), TNF-alpha inhibitors from IBFB Pharma and Xencor (Xencor), torbafylline (CAS No. 105102-21-4) (Sanofi-Aventis), UR-1505 (Uriach), VT-346 (Viron Therapeutics), YSIL6 (Y's Therapeutics), YSTH2 (Y's Therapeutics), NPI-1302a-3 (Nereus Pharmaceuticals, a TNF antagonist from Jerina AG (Germany), dersalazine, and an analogue or derivative thereof.
7478. The method of item 7379 wherein the agent is a Toll receptor inhibitor. 7479. The method of item 7379 wherein the agent is a tubulin antagonist.
7480. The method of item 7379 wherein the agent is a tyrosine kinase inhibitor selected from the group consisting of SU-011248, SUTENT from Pfizer Inc. (New York, NY), BMS-354825, PN-355 (Paracelsian Pharmaceuticals), AGN-199659 (Allergan), AAL-993 or ABP- 309 (Novartis), adaphostin (NIH), AEE-788 (Novartis), AG-013736 (OSI Pharmaceuticals), AG-13736 (Pfizer), ALT-110 (Alteris Therapeutics), AMG- 706 (Amgen), anticancer MAbs from Xencor, anti-EGFrvlll MAbs from Abgenix, anti-HER2 MAb from Abiogen, AZD-2171 or AZD-9935 (AstraZeneca), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), CEP-5214 (Cephalon), CEP-7055 (Cephalon), cetuximab (ImClone Systems), CHIR-200131 and CHIR-258 (Chiron), CP-547632 (OSI Pharmaceuticals), CP-724714 (Pfizer), CT-301 (Creabilis Therapeutics), D- 69491 (Baxter International), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EGFR/ErbB2 inhibitors from Array BioPharma, erlotinib (CAS No. 183319- 69-9) (OSI Pharmaceuticals), EXEL-2880 (Exelixis), FK-778 (Sanofi- Aventis) , gefitinib (CAS No. 184475-35-2) (AstraZeneca), GW-2286 or GW- 654652 (GlaxoSmithKline), her2/neu antigen from AlphaVax, HER-2/neu inhibitor from Generex, Herzyme (Medipad) (Sirna Therapeutics), HKI-272 (Wyeth), HuMax-EGFr (Genmab), idronoxil (CAS No. 81267-65-4) (Novogen), IGF-1 inhibitors from Ontogen, IMC-11 F8 (ImClone Systems), kahalalide F (CAS No. 149204-42-2) (PharmaMar), KDR inhibitor from LG Life Sciences, KDR inhibitors from Abbott Laboratories, KDR kinase inhibitors (UCB), Kdr kinase inhibitors from Merck & Co, KRN-633 and KRN- 951 (Kirin Brewery), KSB-102 (Xenova), lapatinib ditosylate (CAS No. 388082-78-8) (GlaxoSmithKline), matuzumab (Merck KGaA), MDX-214 (Medarex), ME-103 (Pharmexa), MED-A300 (Gerolymatos), MNAC-13 (Lay Line Genomics), nimotuzumab (Center of Molecular Immunology), NSC- 330507 or NSC-707545 (NIH), NV-50 (Novogen), OSI-930 (OSI Pharmaceuticals), panitumumab (Abgenix), pelitinib (CAS No. 287933-82-7) (Wyeth), pertuzumab (CAS No. 380610-27-5) (Genentech), Pharmaprojects No. 3985 (Sanofi-Aventis), prostate cancer therapeutics from Sequenom (SQPC35, SQPC36, SQPC90), removab and remoxab (Trion Pharma), RG- 13022 (CAS No. 136831-48-6), RG-13291 (CAS No. 138989-50-1), or RG- 14620 (CAS No. 136831-49-7) (Sanofi-Aventis), RM-6427 (Romark), RNAi breast cancer therapy from Benitec, RP 53801 (CAS No. 125882-88-4) (Sanofi-Aventis), sorafenib tosylate (Bayer), SU-11657 (Pfizer), Tie-2 antagonists from Semaia (Hybrigenics), Tie-2 inhibitors from Ontogen, trastuzumab (CAS No. 180288-69-1) (Genentech), tyrosine kinase inhibitors from Sanofi-Aventis, U3-1287, U3-1565, U3-1784, U3-1800 (U3 Pharma), vatalanib (Novartis), VEGFR-2 kinase inhibitor from Bristol-Myers Squibb, XL-647 (Exelixis), ZD-6474 (AstraZeneca), ZK-CDK (Schering AG), an EGFR tyrosine kinase inhibitor, EKB-569 (Wyeth), and an analogue or derivative thereof.
7481. The method of item 7379 wherein the agent is a VEGF inhibitor.
7482. The method of item 7379 wherein the agent is a vitamin D receptor agonist.
7483. The method of item 7379 wherein the agent is ZD-6474 (an angiogenesis inhibitor).
7484. The method of item 7379 wherein the agent is AP- 23573 (an mTOR inhibitor).
7485. The method of item 7379 wherein the agent is synthadotin (a tubulin antagonist). 7486. The method of item 7379 wherein the agent is S-0885 (a collagenase inhibitor).
7487. The method of item 7379 wherein the agent is aplidine (an elongation factor-1 alpha inhibitor).
7488. The method of item 7379 wherein the agent is ixabepilone (an epithilone).
7489. The method of item 7379 wherein the agent is IDN- 5390 (an angiogenesis inhibitor).
7490. The method of item 7379 wherein the agent is SB- 2723005 (an angiogenesis inhibitor).
-7491. The method of item 7379 wherein the agent is ABT-518 (an angiogenesis inhibitor).
7492. The method of item 7379 wherein the agent is combretastatin (an angiogenesis inhibitor).
7493. The method of item 7379 wherein the agent is anecortave acetate (an angiogenesis inhibitor).
7494. The method of item 7379 wherein the agent is SB- 715992 (a kinesin antagonist).
7495. The method of item 7379 wherein the agent is temsirolimus (an mTOR inhibitor). 7496. The method of item 7379 wherein the agent is adalimumab (a TNFα antagonist).
7497. The method of item 7379, wherein the composition comprises a polymer.
7498. The method of item 7379, wherein the composition comprises a polymer, and the polymer is, or comprises, a copolymer.
7499. The method of item 7379, wherein the composition comprises a polymer, and the polymer is, or comprises, a block copolymer.
7500. The method of item 7379, wherein the composition comprises a polymer, and the polymer is, or comprises, a random copolymer.
7501. The method of item 7379, wherein the composition comprises a polymer, and the polymer is, or comprises, a biodegradable polymer.
7502. The method of item 7379, wherein the composition comprises a polymer, and the polymer is, or comprises, a nonbiodegradable polymer.
7503. The method of item 7379, wherein the composition comprises a polymer, and the polymer is, or comprises, a hydrophilic polymer.
7504. The method of item 7379, wherein the composition comprises a polymer, and the polymer is, or comprises, a hydrophobic polymer. 7505. The method of item 7379, wherein the composition comprises a polymer, and the polymer is, or comprises, a polymer having hydrophilic domains.
7506. The method of item 7379, wherein the composition comprises a polymer, and the polymer is, or comprises, a polymer having hydrophobic domains.
7507. The method of item 7379, wherein the composition comprises a polymer, and the polymer is, or comprises, a non-conductive polymer.
7508. The method of item 7379, wherein the composition comprises a polymer, and the polymer is, or comprises, an elastomer.
7509. The method of item 7379, wherein the composition comprises a polymer, and the polymer is, or comprises, a hydrogel.
7510. The method of item 7379, wherein the composition comprises a polymer, and the polymer is, or comprises, a silicone polymer.
7511. The method of item 7379, wherein the composition comprises a polymer, and the polymer is, or comprises, a hydrocarbon polymer.
7512. The method of item 7379, wherein the composition comprises a polymer, and the polymer is, or comprises, a styrene-derived polymer. 7513. The method of item 7379, wherein the composition comprises a polymer, and the polymer is, or comprises, a butadiene-derived polymer.
7514. The method of item 7379, wherein the composition comprises a polymer, and the polymer is, or comprises, a macromer.
7515. The method of item 7379, wherein the composition comprises a polymer, and the polymer is, or comprises, a poly(ethylene glycol) polymer.
7516. The method of item 7379, wherein the composition comprises a polymer, and the polymer is, or comprises, an amorphous polymer.
7517. The method of item 7379,- wherein the composition - - further comprises a second pharmaceutically active agent.
7518. The method of item 7379, wherein the composition further comprises an anti-inflammatory agent.
7519. The method of item 7379, wherein the composition further comprises an agent that inhibits infection.
7520. The method of item 7379, wherein the composition further comprises an anthracycline.
7521. The method of item 7379, wherein the composition further comprises doxorubicin. 7522. The method of item 7379 wherein the composition further comprises mitoxantrone.
7523. The method of item 7379 wherein the composition further comprises a fluoropyrimidine.
7524. The method of item 7379, wherein the composition further comprises 5-fluorouracil (5-FU).
7525. The method of item 7379, wherein the composition further comprises a folic acid antagonist.
7526. The method of item 7379, wherein the composition further comprises methotrexate.
- - . - 7527. The method of item 7379, wherein the composition further comprises a podophylotoxin.
7528. The method of item 7379, wherein the composition further comprises etoposide.
7529. The method of item 7379, wherein the composition further comprises camptothecin.
7530. The method of item 7379, wherein the composition further comprises a hydroxyurea.
7531. The method of item 7379, wherein the composition further comprises a platinum complex. 7532. The method of item 7379, wherein the composition further comprises cisplatin.
7533. The method of item 7379 wherein the composition further comprises an anti-thrombotic agent.
7534. The method of item 7379, wherein the composition further comprises a visualization agent.
7535. The method of item 7379, wherein the composition further comprises a visualization agent, and the visualization agent is a radiopaque material, wherein the radiopaque material comprises a metal, a halogenated compound, or a barium containing compound.
7536. The method of item 7379, wherein the composition further comprises a visualization, agent, and the [.visualization agent is, or comprises, barium, tantalum, or technetium.
7537. The method of item 7379, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, an MRI responsive material.
7538. The method of item 7379, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, a gadolinium chelate.
7539. The method of item 7379, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, iron, magnesium, manganese, copper, or chromium. 7540. The method of item 7379, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, iron oxide compound.
7541. The method of item 7379, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, a dye, pigment, or colorant.
7542. The method of item 7379 wherein the agent is released in effective concentrations from the composition comprising the agent by diffusion over a period ranging from the time of administration to about 90 days.
7543. The method of item 7379 wherein the agent is released in effective concentrations from the composition comprising the agent by erosion of the composition over a period ranging from the time of- - administration to about 90 days.
7544. The method of item 7379 wherein the composition further comprises an inflammatory cytokine.
7545. The method of item 7379 wherein the composition further comprises an agent that stimulates cell proliferation.
7546. The method of item 7379 wherein the composition further comprises a polymeric carrier.
7547. The method of item 7379 wherein the composition is in the form of a gel, paste, or spray. 7548. The method of item 7379 wherein the implant is partially constructed with the agent or the composition.
7549. The method of item 7379 wherein the implant is fully constructed with the agent or the composition.
7550. The method of item 7379 wherein the implant is impregnated with the agent or the composition.
7551. The method of item 7379, wherein the agent or the composition forms a coating, and the coating directly contacts the implant.
7552. The method of item 7379, wherein the agent or the composition forms a coating, and the coating indirectly contacts the implant.
7553. The method of item 7379 wherein the agent or the composition forms a coating, and the coating partially covers the implant.
7554. The method of item 7379, wherein the agent or the composition forms a coating, and the coating completely covers the implant.
7555. The method of item 7379 wherein the agent or the composition is located within pores or holes of the implant.
7556. The method of item 7379 wherein the agent or the composition is located within a channel, lumen, or divet of the implant.
7557. The method of item 7379 wherein the implant further comprising an echogenic material. 7558. The method of item 7379 wherein the implant further comprises an echogenic material, wherein the echogenic material is in the form of a coating.
7559. The method of item 7379 wherein the implant is sterile.
7560. The method of item 7379 wherein the agent is delivered from the implant, wherein the agent is released into tissue in the vicinity of the implant after deployment of the implant.
7561. The method of item 7379 wherein the agent is delivered from the implant, wherein the agent is released into tissue in the vicinity of the implant after deployment of the implant, wherein the tissue is connective tissue.
7562. The method of item 7379 wherein the agent is delivered from the implant, wherein the agent is released into tissue in the vicinity of the implant after deployment of the implant, wherein the tissue is muscle tissue.
7563. The method of item 7379 wherein the agent is delivered from the implant, wherein the agent is released into tissue in the vicinity of the implant after deployment of the implant, wherein the tissue is nerve tissue.
7564. The method of item 7379 wherein the agent is delivered from the implant, wherein the agent is released into tissue in the vicinity of the implant after deployment of the implant, wherein the tissue is epithelium tissue. 7565. The method of item 7379 wherein the agent is delivered from the implant, wherein the agent is released in effective concentrations from the implant over a period ranging from the time of deployment of the implant to about 1 year.
7566. The method of item 7379 wherein the agent is delivered from the implant, wherein the agent is released in effective concentrations from the implant over a period ranging from about 1 month to 6 months.
7567. The method of item 7379 wherein the agent is delivered from the implant, wherein the agent is released in effective concentrations from the implant over a period ranging from about 1 - 90 days.
7568. The method of item 7379 wherein the agent is delivered from the implant, wherein the agent is released in effective concentrations from the implant at a constant rate.
7569. The method of item 7379 wherein the agent is delivered from the implant, wherein the agent is released in effective concentrations from the implant at an increasing rate.
7570. The method of item 7379 wherein the agent is delivered from the implant, wherein the agent is released in effective concentrations from the implant at a decreasing rate.
7571. The method of item 7379 wherein the agent is delivered from the implant, wherein the implant comprises about 0.01 Dg to about 10 Dg of the agent. 7572. The method of item 7379 wherein the agent is delivered from the implant, wherein the implant comprises about 10 Dg to about 10 mg of the agent.
7573. The method of item 7379 wherein the agent is delivered from the implant, wherein the implant comprises about 10 mg to about 250 mg of the agent.
7574. The method of item 7379 wherein the agent is delivered from the implant, wherein the implant comprises about 250 mg to about
1000 mg of the agent.
7575. The method of item 7379 wherein the agent is delivered from the implant, wherein the implant comprises about 1000 mg to about 2500 mg of the agent.
7576. The method of item 7379 wherein the agent is delivered from the implant, wherein a surface of the implant comprises less than 0.01 Dg of the agent per mm2 of implant surface to which the agent is applied.
7577. The method of item 7379 wherein the agent is delivered from the implant, wherein a surface of the implant comprises about 0.01 Dg to about 1 Dg of the agent per mm2 of implant surface to which the agent is applied.
7578. The method of item 7379 wherein the agent is delivered from the implant, wherein a surface of the implant comprises about 1 Dg to about 10 Dg of the agent per mm2 of implant surface to which the agent is applied. 7579. The method of item 7379 wherein the agent is delivered from the implant, wherein a surface of the implant comprises about 10 Dg to about 250 Dg of the agent per mm2 of implant surface to which the agent is applied.
7580. The method of item 7379 wherein the agent is delivered from the implant, wherein a surface of the implant comprises about 250 Dg to about 1000 Dg of the agent per mm2 of implant surface to which the agent is applied.
7581. The method of item 7379 wherein the agent is delivered from the implant, wherein a surface of the implant comprises about 1000 Dg to about 2500 Dg of the agent per mm2 of implant surface to which the agent is applied.
7582. The method of item 7379, wherein the implant further comprises a coating, and the coating is a uniform coating.
7583. The method of item 7379, wherein the implant further comprises a coating, and the coating is a non-uniform coating.
7584. The method of item 7379, wherein the implant further comprises a coating, and the coating is a discontinuous coating.
7585. The method of item 7379, wherein the implant further comprises a coating, and the coating is a patterned coating.
7586. The method of item 7379, wherein the implant further comprises a coating, and the coating has a thickness of 100 Dm or less. 7587. The method of item 7379, wherein the implant further comprises a coating, and the coating has a thickness of 10 Dm or less.
7588. The method of item 7379, wherein the implant further comprises a coating, and the coating adheres to the surface of the implant upon deployment of the implant.
7589. The method of item 7379, wherein the implant further comprises a coating, and the coating is stable at room temperature for a period of at least 1 year.
7590. The method of item 7379, wherein the implant further comprises a coating, and the agent is present in the coating in an amount ranging between about 0.0001% to about 1% by weight.
7591. The method of item 7379, wherein the implant further comprises a coating, and the agent is present in the coating in an amount ranging between about 1 % to about 10% by weight.
7592. The method of item 7379, wherein the implant further comprises a coating, and the agent is present in the coating in an amount ranging between about 10% to about 25% by weight.
7593. The method of item 7379, wherein the implant further comprises a coating, and the agent is present in the coating in an amount ranging between about 25% to about 70% by weight.
7594. The method of item 7379, wherein the implant further comprises a coating, and the coating comprises a polymer. 7595. The method of item 7379, wherein the implant comprises a first coating having a first composition and a second coating having a second composition.
7596. The method of item 7379, wherein the implant comprises a first coating having a first composition and a second coating having a second composition, wherein the first composition and the second composition are different.
7597. A method as in any one of items 7379-7596, wherein the device is a device for treating hypertropic scar or keloid that comprises an external tissue expansion device.
7598. A method as in any one of items 7379-7596, wherein the device is a device for treating hypertropic scar or keloid that comprises a masking element, and wherein the masking element may be pressed onto the scar tissue.
7599. A method as in any one of items 7379-7596, wherein the device is a device for treating hypertropic scar or keloid that comprises a locking element and a grasping structure so that the dermal and epidermal layers of a skin wound can be pushed together.
7600. A method for inhibiting scarring comprising placing a device that comprises a vascular graft (e.g., an implant) and an anti-scarring agent or a composition comprising an anti-scarring agent into an animal host, wherein the agent inhibits scarring.
7601. The method of item 7600 wherein the agent is an adensosine A2A receptor antagonist. 7602. The method of item 7600 wherein the agent is an AKT inhibitor.
7603. The method of item 7600 wherein the agent is an alpha 2 integrin antagonist, wherein the alpha 2 integrin antagonist is Pharmaprojects No. 5754 (Merck KgaA).
7604. The method of item 7600 wherein the agent is an alpha 4 integrin antagonist.
7605. The method of item 7600 wherein the agent is an alpha 7 nicotinic receptor agonist.
7606. The method of item 7600 wherein the agent is an angiogenesis inhibitor selected from the group consisting of AG-12,958 (Pfizer)^ ATN-161 (Attention LLC), neovastat, an angiogenesis inhibitor from Jerina AG (Germany), NM-3 (Mercian), VGA-1155 (Taisho), FCE-26644 (Pfizer), FCE-26950 (Pfizer), FPMA (Meiji Daries), FR-111142 (Fujisawa), GGTI-298, GM-1306 (Ligand), GPA-1734 (Novartis), NNC-47-0011 (Novo Nordisk), herbamycin (Nippon Kayaku), lenalidomide (Celegene), IP-10 (NIH), ABT-828 (Abbott), KIN-841 (Tokushima University, Japan), SF-1126 (Semafore Pharmaceuticals), laminin technology (NIH), CHIR-258 (Chiron), NVP-AEW541 (Novartis), NVP-AEW541 (Novartis), Vt16907 (Alchemia), OXI-8007 (Oxigene), EG-3306 (Ark Therapeutics), Maspin (Arriva), ABT- 567 (Abbott), PPI-2458 (Praecis Pharmaceuticals), CC-5079, CC-4089 (Celgene), HIF-1 alpha inhibitors (Xenova), S-247 (Pfizer), AP-23573 (Ariad), AZD-9935 (Astra Zeneca), mebendazole (Introgen Therapeutics), MetAP-2 inhibitors (GlaxoSmithKline), AG-615 (Angiogene Pharmaceuticals), Tie-2 antagonists (Hybrigenics), NC-381 , CYC-381 , NC-169, NC-219, NC-383, NC-384, NC-407 (Lorus Therapeutics), ATN-224 (Attenuon), ON-01370 (Onconova), Vitronectin antagonists (Amgen), SDX-103 (Salmedix), Vitronectin antagonists (Shire), CHP (Riemser), TEK (Amgen), Anecortave acetate (Alcon), T46.2 (Matrix Therapeutics), HG-2 (Heptagen), TEM antagonists (Genzyme), Oxi-4500 (Oxigene), ATN-161 (Attenuon), WX-293 (Wilex), M-2025 (Metris Therapeutics), Alphastatin (BioActa), YH-16 (Yantai Rongchang), BIBF-1120 (Boehringer Ingelheim), BAY-57-9352 (Bayer), AS- 1404 (Cancer Research Technology), SC-77964 (Pfizer), glycomimetics (BioTie Therapies), TIE-2 Inhibitors (Ontogen), DIMI, Octamer (Octamer), ABR-215050 (Active Biotech), ABT-518 (Abbott), KDR inhibitors (Abbott), BSF-466895 (Abbott), SCH-221153 (Schering-Plough), DAC:antiangiogenic (ConjuChem), TFPI (EntreMed), AZD-2171 (Astra-Zenaca), CDC-394 (Celgene), LY290293 (EIi Lilly), IDN-5390 (Indena), Kdr Kinase Inhibitors (Merck), CT-113020, CT-116433, CT-116563, CT-31890, CT-32228) (Cell Therapeutics), A-299620 (Abbott), TWEAK Inhibitor (Amgen), VEGF modulators (Johnson and Johnson), Tum-N53, tumstatin (Genzyme), Thios- 1 , Thios-2 (Thios Pharmaceuticals), MV-6401 (Miravant Medical Technologies), Spisulosine (PharmaMar), CEP-7055 (Cephalon), AUV-201 (Auvation), LM-609 (EIi Lilly), SKF-106615 (AnorMED), Oglufanide disodium (Cytran), BW-114 (Pharminox), Calreticulin (NIH), WX-678 (Wilex), SD-7784 (Pfizer), WX-UK1 (Wilex), SH-268 (Schering AG), 2-Me-PGA (Celgene), S- 137 (Pfizer), ZD-6126 (Angiogene Pharmaceuticals), SG-292 (SignalGen), Benefin (Lane Labs), A6, A36 (Angstrom), SB-2723005 (GlaxoSmithKline), SC-7 (Cell Therapeutics), ZEN-014 (AEterna Zentaris), 2-methoxyestradiol (EntreMed), NK-130119 (Nippon Kayaku), CC-10004 (Celgene), AVE- 8062A (Ajinomoto), Tacedinaline (Pfizer), Actinonin (Tokyo Metropolitan Institute of Medical Science), Lenalidomide (Celgene), VGA-1155, BTO-956 (SRI International), ER-68203-00 (Eisai), CT-6685 (UCB), JKC-362 (Phoenix Pharmaceuticals), DMI-3798 (DMI Biosciences, Angiomate (Ipsen), ZD-6474 (AstraZeneca), CEP-5214 (Cephalon), Canstatin (Genzyme), NM-3 (Mercian), Oridigm (MediQuest Therapeutics), Exherin (Adherex), BLS-0597 (Boston Life Sciences), PTC-299 (PTC Therapeutics), NPI-2358 (Nereus Pharmaceuticals), CGP-79787 (Novartis), AEE-788 (Novartis), CKD-732 (Chong Kun Dang), CP-564959 (OSI Pharmaceuticals), CM-101 (CarboMed), CT-2584, CT3501 (Cell Therapeutics), combretastatin and analogues and derivatives thereof (Oxigene), Rebimastat (Bristol-Meyers Squibb), Dextrin 2-sulfate (ML Laboratories), Cilengitide (Merk KGaA), NSC-706704 (Pharminox), KRN- 951 (Kirin Brewery), Ukrain, NSC-631570 (Nowicky Pharma), Tecogalan sodium (Daiichi Pharmaceutical), Tz-93 (Tsumura), TBC-1635 (Encysive Pharmaceuticals), TAN-1120 (Takeda), Semaxanib (Pfizer), BDI-7800 (Biopharmacopae), SD-186, SD-983 (Bristol-Meyers Squibb), SB-223245 (GlaxoSmithKline), SC-236 (Pfizer), RWJ-590973 (Johnson and Johnson), ILX-1850 (Genzyme), SC-68488, S-836 (Pfizer), CG-55069-11 (CuraGen), Ki-23057 (Kirin Brewery), CCX-700 (Chemoentryx), Pegaptanib octasodium (Giled Sciences), ANGIOCOL (available from Biostratum Inc.), or an analogue or derivative thereof.
7607. The method of item 7600 wherein the agent is an apoptosis antagonist.
7608. The method of item 7600 wherein the agent is an apoptosis activator.
7609. The method of item 7600 wherein the agent is a beta 1 integrin antagonist.
7610. The method of item 7600 wherein the agent is a beta tubulin inhibitor.
7611. The method of item 7600 wherein the agent is a blocker of enzyme production in Hepatitis C. 7612. The method of item 7600 wherein the agent is a Bruton's tyrosine kinase inhibitor.
7613. The method of item 7600 wherein the agent is a calcineurin inhibitor.
7614. The method of item 7600 wherein the agent is a caspase 3 inhibitor.
7615. The method of item 7600 wherein the agent is a CC chemokine receptor antagonist.
7616. The method of item 7600 wherein the agent is a cell cycle inhibitor selected from the group consisting of SNS-595 (Sunesis), synthadotin, KRX-0403, and an analogue or derivative thereof.
7617. The method of item 7600 wherein the agent is a cathepsin B inhibitor.
7618. The method of item 7600 wherein the agent is a cathepsin K inhibitor, wherein the cathepsin K inhibitor is 462795 (GlaxoSmithKline), INPL-022-D6 (Amura Therapeutics), or an analogue or derivative thereof.
7619. The method of item 7600 wherein the agent is a cathepsin L inhibitor.
7620. The method of item 7600 wherein the agent is a CD40 antagonist. 7621. The method of item 7600 wherein the agent is a chemokine receptor agonist.
7622. The method of item 7600 wherein the agent is a chymase inhibitor.
7623. The method of item 7600 wherein the agent is a collagenase antagonist.
7624. The method of item 7600 wherein the agent is a CXCR antagonist.
7625. The method of item 7600 wherein the agent is a cyclin dependent kinase inhibitor selected from the group consisting of a CDK-1 inhibitor, a CDK-2 inhibitor, a CDK- 4 inhibitor, a CDK-6 inhibitor, a CAK1 inhibitor from GPC Biotech and Bristol-Myers Squibb, RGB-286199 (GPC Biotech), an anticancer agent from Astex Technology, a CAK1 inhibitor from GPC Biotech, a CDK inhibitor from Sanofi-Aventis, a CDK1/CDK2 inhibitor from Amgen, a CDK2 inhibitor from SUGEN-2 (Pfizer), a hearing loss therapy agent (Sound Pharmaceuticals), PD-0332991 (Pfizer), RGB-286199 (GPC Biotech), Ro-0505124 (Hoffmann-La Roche), a Ser/Thr kinase inhibitor from Lilly (EIi Lilly), CVT-2584 (CAS No. 199986-75-9) (CV Therapeutics), and an analogue or derivative thereof.
7626. The method of item 7600 wherein the agent is a cyclooxygenase 1 inhibitor.
7627. The method of item 7600 wherein the agent is a DHFR inhibitor. 7628. The method of item 7600 wherein the agent is a dual integrin inhibitor.
7629. The method of item 7600 wherein the agent is an elastase inhibitor.
7630. The method of item 7600 wherein the agent is an elongation factor-1 alpha inhibitor.
7631. The method of item 7600 wherein the agent is an endothelial growth factor antagonist.
7632. The method of item 7600 wherein the agent is an endothelial growth factor receptor kinase inhibitor selected from the group consisting of sorafenib tosylate (Bayer), AAL-993 (Novartis), ABP-309 (Novartis), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), E- 7080 (Eisai), EG-3306 (Ark Therapeutics), EXEL-2880 (Exelixis), GW- 654652 (GlaxoSmithKline), a KDR inhibitor from LG Life Sciences, CT-6685 and CT-6729 (UCB), KRN-633 and KRN-951 (Kirin Brewery), OSI-930 (OSI Pharmaceuticals), SP-5.2 (Supratek Pharma), SU-11657 (Pfizer), a Tie-2 antagonist (Hybrigenics), a VEGFR-2 kinase inhibitor (Bristol-Myers Squibb), XL-647 (Exelixis), a KDR inhibitor from Abbott Laboratories, sorafenib tosylate, and an analogue or derivative thereof.
7633. The method of item 7600 wherein the agent is an endotoxin antagonist.
7634. The method of item 7600 wherein the agent is an epothilone and tubulin binder. 7635. The method of item 7600 wherein the agent is an estrogen receptor antagonist.
7636. The method of item 7600 wherein the agent is an FGF inhibitor.
7637. The method of item 7600 wherein the agent is a famexyl transferase inhibitor.
7638. The method of item 7600 wherein the agent is farnesyltransferase inhibitor selected from the group of A-197574 (Abbott), a farnesyltransferase inhibitor from Servier, FPTIII (Strathclyde Institute for Drug R), LB-42908 (LG Life Sciences), Pharmaprojects No. 5063 (Genzyme), Pharmaprojects No. 5597 (Ipsen), Yissum Project No. B-1055 (Yissum), and an analogue or derivative thereof.
7639. The method of item 7600 wherein the agent is an FLT-3 kinase inhibitor.
7640. The method of item 7600 wherein the agent is an FGF receptor kinase inhibitor.
7641. The method of item 7600 wherein the agent is a fibrinogen antagonist selected from the group consisting of AUV-201 (Auvation), MG-13926 (Sanofi-Aventis), plasminogen activator (CAS No. 105913-11-9) (from Sanofi-Aventis or UCB), plasminogen activator-2 (tPA- 2) (Sanofi-Aventis), pro-urokinase (CAS No. 82657-92-9) (Sanofi-Aventis), and an analogue or derivative thereof.
7642. The method of item 7600 wherein the agent is a heat shock protein 90 antagonist selected from the group consisting of SRN-005 (Sirenade), geldanamycin, NSC-33050 (17-allylaminogeldanamycin; 17- AAG), ^-dimethylaminoethylamino-^-demethoxy-geldanamycin (17- DMAG), rifabutin (rifamycin XIV, 1',4~didehydro-1-deoxy-1 ,4-dihydro-5'-(2- methylpropyl)-1-oxo-), and an analogue or derivative thereof.
7643. The method of item 7600 wherein the agent is a histone deacetylase inhibitor.
7644. The method of item 7600 wherein the agent is an HMGCoA reductase inhibitor selected from the group consisting of an atherosclerosis therapeutic from Lipid Sciences, ATI-16000 (ARYx Therapeutics), KS-01-019 (Kos Pharmaceuticals), Pharmaprojects No. 2197 (Sanofi-Aventi), RP 61969 (Sanofi-Aventis), and an analogue or derivative thereof.
7645. The method of item 7600 wherein the agent is an ICAM inhibitor.
7646. The method of item 7600 wherein the agent is an IL, ICE and IRAK antagonist, wherein the antagonist is a CJ-14877, CP-424174 (Pfizer), NF-61 (Negma-Lerads), and an analogue or derivative thereof.
7647. The method of item 7600 wherein the agent is an IL-2 inhibitor.
7648. The method of item 7600 wherein the agent is an immunosuppressant selected from the group consisting of teriflunomide (Sanofi Aventis), chlorsulfaquinoxalone (NSC-339004), chlorsulfaquinoxalone sulfate, CS-712 (Sankyo), ismomultin alfa (CAS No. 457913-93-8) (Akzo Nobel), antiallergics from GenPat77, antiinflammatories or AT-005 (Androclus Therapeutics), autoimmune disease therapeutics from EpiVax, BN-007 (Bone), budesonide (CAS No. 51333-22- 3) (MAP Pharmaceuticals), CO-14 (Genzyme), edratide (CAS No. 433922- 67-9) (Teva), EP-314 (Enanta), eprovafen (CAS No. 101335-99-3) (Sanofi- Aventis), HWA-131 (CAS No. 118788-41-3) (Sanofi-Aventis), immunomodulators from MerLion Pharmaceuticals, immunosuppressives from Alchemia, IPL-12 (Inflazyme), MDL-9563 (CAS No. 27086-86-8) (Sanofi-Aventis), Pharmaprojects No. 2330 (Sanofi-Aventis), Pharmaprojects No. 6426 (Abgenix), PXS-25 (Pharmaxis), rosmarinic acid (CAS No. 20283-92-5) (Sanofi-Aventis), RP 42927 or RP 54745 (CAS No. 135330-08-4) (Sanofi-Aventis), SGN-35 (Seattle Genetics), ST-1959 (Sigma-Tau), type I diabetes therapy from SYNX Pharma, UNIL-88 (Debiopharm), VP-025 (Vasogen), VR-694 (Vectura), PRTX-001 (Protalex), and an analogue or derivative thereof.
7649. The method of item 7600 wherein the agent is an IMPDH (inosine monophosphate).
7650. The method of item 7600 wherein the agent is an integrin antagonist.
7651. The method of item 7600 wherein the agent is an interleukin antagonist.
7652. The method of item 7600 wherein the agent is an inhibitor of type III receptor tyrosine kinase.
7653. The method of item 7600 wherein the agent is an irreversible inhibitor of enzyme methionine aminopeptidase type 2.
7654. The method of item 7600 wherein the agent is an isozyme selective delta protein kinase C inhibitor. 7655. The method of item 7600 wherein the agent a JAK3 enzyme inhibitor.
7656. The method of item 7600 wherein the agent is a JNK inhibitor.
7657. The method of item 7600 wherein the agent is a kinase inhibitor.
7658. The method of item 7600 wherein the agent is kinesin antagonist.
7659. The method of item 7600 wherein the agent is a kinesin antagonist.
7660. The method^ of item 7600 wherein the. agent is a leukotriene inhibitor and antagonist selected from the group consisting of ambicromil (CAS No. 58805-38-2) (Sanofi-Aventis), amelubant (CAS No. 346735-24-8) (Boehringer Ingelheim), DW-1141 (Dong Wha), ebselen (Daiichi Pharmaceutical), ibudilast (Kyorin), leucotriene inhibitors from Sanofi-Aventis, lymphotoxin-beta receptor (LT-D) from Biogen Idee, Pharmaprojects No. 1535 and 2728 (CAS No. 119340-33-9) (Sanofi- Aventis), R-112 (Rigel), Rev-5367 (CAS No. 92532-05-3) (Sanofi-Aventis), RG-14893 (CAS No. 141835-49-6) (Sanofi-Aventis), RG-5901-A (CAS No. 101910-24-1), 92532-23-5, RP 66153 (CAS No. 142422-79-5), RP 66364 (CAS No.186916-92-5), RP 69698 (CAS No. 141748-00-7) (Sanofi-Aventis), SC-411930 (Pfizer), SC-41930 (CAS No. 120072-59-5) (Pfizer), SC-50605 (CAS No. 138828-39-4) (Pfizer), SC-51146 (CAS No. 141059-52-1), SC- 53228 (CAS No. 153633-01-3) (Pfizer), spaglumic acid (ZY-15106) (CAS No.3106-85-2), 80619-64-3 (Novartis), tipredane (CAS No. 85197-77-9) (Bristol-Myers Squibb), U-75302 (CAS No. 119477-85-9) (Pfizer), and analogue or derivative thereof.
7661. The method of item 7600 wherein the agent is an MAP kinase inhibitor.
7662. The method of item 7600 wherein the agent is a matrix metalloproteinase inhibitor.
7663. The method of item 7600 wherein the agent is an MCP- CCR2 inhibitor.
7664. The method of item 7600 wherein the agent is an mTOR inhibitor.
7665. The method of item 7600 wherein the agent is an mTOR kinase inhibitor.
7666. The method of item 7600 wherein the agent is a microtubule inhibitor selected from the group consisting of antibody- maytansinoid conjugates from Biogen Idee, colchicines (MantiCore Pharmaceuticals), anticancer immunoconjugates from Johnson & Johnson, DIME from Octamer, gni-1f (GNI), huC242-DM4, huMy9-6-DM1 (ImmunoGen), IDN-5404 (Indena), IMO-098, IMOderm (Imotep), mebendazole (Introgen Therapeutics), microtubule poisons from Cambridge Enterprise, paclitaxel such as LOTAX from Aphios (CAS No. 33069-62-4), Genexol-PM from Samyang, Pharmaprojects No. 6383 (Tapestry Pharmaceuticals), RPR-112378 (Sanofi-Aventis), SGN-75 (Seattle Genetics), SPL-7435 (Starpharma), SSR-250411 (Sanofi-Aventis), trastuzumab-DM1 (Genentech), vinorelbine, and an analogue or derivative thereof. 7667. The method of item 7600 wherein the agent is an MIF inhibitor.
7668. The method of item 7600 wherein the agent is an MMP inhibitor.
7669. The method of item 7600 wherein the agent is a neurokinin (NK) antagonist selected from the group consisting of anthrotainin (CAS No. 148084-40-6) (Sanofi-Aventis), an IBS thereapeutic from ArQuIe, MDL-105212A (CAS No. 167261-60-1) (Ssanofi-Aventis), Pharmaprojects No. 2744, 3258 (CAS No. 139167-47-8) 4006, 4201 , or 5986 (Sanofi-Aventis), RP 67580 (CAS No. 135911-02-3), SR-144190 (CAS No. 201152-86-5), SSR-240600, SSR-241586 (Sanofi-Aventis), TKA-457 (Novartis), vestipitant mesylate (CAS No. 334476-64-1) (GlaxoSmithKline), Win-64821 (Sanofi-Aventis), PRX-96026 (Predix Pharmaceuticals), and an analogue or derivative thereof. „ _
7670. The method of item 7600 wherein the agent is an NF kappa B inhibitor selected from the group consisting of AVE-0545 or AVE- 0547 (Sanofi-Aventis), bortezomib (CAS No. 179324-69-7) (Millennium Pharmaceuticals), dexanabinol (CAS No. 112924-45-5) (Pharmos), dexlipotam (Viatris), Pharmaprojects No. 6283 (INDRA) (OXiGENE), IPL- 576092 (CAS No. 137571-30-3) (Inflazyme), NFKB decoy (Corgentech), NFKB decoy oligo (AnGes MG), NFKB's from Ariad, osteoporosis treatments or S5 (F005) from Fulcrum Pharmaceuticals, P61 (Phytopharm), R-flurbiprofen (CAS No. 5104-49-4) (Encore Pharmaceuticals), and an analogue or derivative thereof.
7671. The method of item 7600 wherein the agent is a nitric oxide agonist. 7672. The method of item 7600 wherein the agent is an ornithine decarboxylase inhibitor.
7673. The method of item 7600 wherein the agent is a p38 MAP kinase inhibitor selected from the group consisting of AZD-6703 (AstraZeneca), JX-401 (Jexys Pharmaceuticals), BMS-2 (Bristol-Myers Squibb), a p38 MAP kinase inhibitor from Novartis, a p38-alpha MAP kinase inhibitor from Amphora, Pharmaprojects No. 5704 (Pharmacopeia), RPR- 200765A (Sanofi-Aventis), SD-282 (Johnson & Johnson), TAK-715 (Takeda), and an analogue or derivative thereof.
7674. The method of item 7600 wherein the agent is a palmitoyl-protein thioesterase inhibitor.
7675. The method of item 7600 wherein the agent is a PDGF receptor kinase inhibitor selected from the group consisting of AAL-993, AMN-107, or ABP-309 (Novartis), AMG-706 (Amgen), BAY-57-9352 (Bayer), CDP-860 (UCB), E-7080 (Eisai), imatinib (CAS No. 152459-95-5) (Novartis), OSI-930 (OSI Pharmaceuticals), RPR-127963E (Sanofi-Aventis), RWJ-540973 (Johnson & Johnson), sorafenib tosylate (Bayer), SU-11657 (Pfizer), tandutinib (CAS No. 387867-13-2) (Millennium Pharmaceuticals), vatalanib (Novartis), ZK-CDK (Schering AG), and an analogue or derivative thereof.
7676. The method of item 7600 wherein the agent is (-)- halofenate (Metabolex), AMG-131 (Amgen), antidiabetics from Japan Tobacco, AZD-4619, AZD-8450, AZD-8677 (AstraZeneca), DRF-10945, balaglitazone (Dr Reddy's), CS-00088, CS-00098 (Chipscreen Biosciences), E-3030 (Eisai), etalocib (CAS No. 161172-51-6) (EIi Lilly), GSK-641597 (Ligand), GSK-677954 (GlaxoSmithKline), GW-409544 (Ligand), GW- 590735 (GlaxoSmithKline), K-111 (Hoffmann-La Roche), LY-518674 (EIi Lilly), LY-674 (Ligand), LY-929 (Ligand), MC-3001 , MC-3002 (MaxoCore Pharmaceuticals), metformin HCI + pioglitazone (CAS No. 1115-70-4 and 112529-15-4), ACTOPLUS MET from Andrx), muraglitazar (CAS No. 331741-94-7) (Bristol-Myers Squibb), naveglitazar (Ligand), oleoylethanolamide (Kadmus Pharmaceuticals), ONO-5129, pioglitazone hydrochloride (CAS No. 111025-46-8 and 112529-15-4) (Takeda), PLX-204 (Plexxikon), PPAR agonists from Genfit, PPAR delta agonists from EIi Lilly, PPAR-alpha agonists from CrystalGenomics, PPAR-gamma modulators and PPAR-β modulators from CareX, rosiglitazone maleate (CAS No. 122324- 73-4 or 155141-29-0) (GlaxoSmithKline), rosiglitazone maleate/glimepir (CAS No. 155141-29-0 and 93479-97-1), AVANDARYL, rosiglitazone maleate/metformin extend (CAS No. 155141-29-0 and 657-24-9), AVANDAMET, rosiglitazone maleate+metformin, AVANDAMET (GlaxoSmithKline), tesaglitazar (AstraZeneca), and an analogue or derivative thereof.
7677. The method of item 7600 wherein the agent is a phosphatase inhibitor.
7678. The method of item 7600 wherein the agent is a phosphodiesterase (PDE) inhibitor selected from the group consisting of avanafil (Tanabe Seiyaku), dasantafil (CAS No. 569351-91-3) (Schering- Plough), A-906119 (CAS No. 134072-58-5), DL-850 (Sanofi-Aventis), GRC- 3015, GRC-3566, GRC-3886 (Glenmark), HWA-153 (CAS No. 56395-66-5) (Sanofi-Aventis), hydroxypumafentrine (Altana), IBFB-130011, IBFB-14-016, IBFB-140301 , IBFB-150007, IBFB-211913 (IBFB Pharma), L-826141 (Merck & Co), medorinone (CAS No. 88296-61-1) (Sanofi-Aventis), MEM- 1917 (Memory Pharmaceuticals), ND-1251 (Neuro3d), PDE inhibitors from ICOS, PDE IV inhibitors from Memory Pharmaceuticals and CrystalGenomics, Pharmaprojects No. 2742 and 6141 (Sanofi-Aventis), QAD-171 (Novartis), RHC-2963 (CAS No. 76993-12-9 and 76993-14-1), RPR-117658, RPR-122818 derivatives, SR-24870 , and RPR-132294 (Sanofi-Aventis), SK-350 (ln2Gen), stroke therapy agents from deCODE Genetics, TAS-203 (Taiho), tofimilast (CAS No. 185954-27-2) (Pfizer), UK- 371800 (Pfizer), WIN-65579 (CAS No. 158020-82-7) (Sanofi-Aventis), IBFB- 130020 (IBFB Pharma), OPC-6535 (CAS No. 145739-56-6) (Otsuka), a phosphodiesterase III inhibitor, enoximone, a phosphodiesterase IV inhibitor, fosfosal, Atopik (Barrier Therapeutics), triflusal, a phosphodiesterase V inhibitor, and an analogue or derivative thereof.
7679. The method of item 7600 wherein the agent is a PKC inhibitor.
7680. The method of item 7600 wherein the agent is a platelet activating factor antagonist.
7681. The method of item 7600 wherein the agent is a platelet-derived growth factor receptor kinase inhibitor.
7682. The method of item 7600 wherein the agent is a prolyl hydroxylase inhibitor.
7683. The method of item 7600 wherein the agent is a polymorphonuclear neutrophil inhibitor.
7684. The method of item 7600 wherein the agent is a protein kinase B inhibitor.
7685. The method of item 7600 wherein the agent is a protein kinase C stimulant. 7686. The method of item 7600 wherein the agent is a purine nucleoside analogue.
7687. The method of item 7600 wherein the agent is a purinoreceptor P2X antagonist.
7688. The method of item 7600 wherein the agent is a Raf kinase inhibitor.
7689. The method of item 7600 wherein the agent is a reversible inhibitor of ErbB1 and ErbB2.
7690. The method of item 7600 wherein the agent is a ribonucleoside triphosphate reductase inhibitor.
7691. The method of item 7600 wherein the agent is an SDF- 1 antagonist.
7692. The method of item 7600 wherein the agent is a sheddase inhibitor.
7693. The method of item 7600 wherein the agent is an SRC inhibitor.
7694. The method of item 7600 wherein the agent is a stromelysin inhibitor.
7695. The method of item 7600 wherein the agent is an Syk kinase inhibitor. 7696. The method of item 7600 wherein the agent is a telomerase inhibitor.
7697. The method of item 7600 wherein the agent is a TGF beta inhibitor selected from the group consisting of pirfenidone (CAS No. 53179-13-8) (MARNAC)1 tranilast (CAS No. 53902-12-8) (Kissei), IN-1130 (ln2Gen), mannose-6-phosphate (BTG), TGF-β antagonists from Inflazyme (Pharmaprojects No. 6075), TGF-β antagonists from Sydney, non-industrial source), TGF-β I receptor kinase inhibitors from EIi Lilly, TGF-β receptor inhibitors from Johnson & Johnson, and an analogue or derivative thereof.
7698. The method of item 7600 wherein the agent is a TN Fa antagonist or TACE inhibitor selected from the group consisting of adalimumab (CAS No. 331731-18-1) (Cambridge Antibody Technology), AGIX-4207 (AtheroGenics), AGT-1 (Advanced Biotherapy), an antiinflammatory from Borean Pharma, Cellzome, or Paradigm Therapeutics, anti-inflammatory vaccine (TNF-alpha kinoid) from Neovacs, humanized anti-TNF antibody or an anti-TNF MAb (CB0006) Celltech (UCB), apratastat (CAS No. 287445-51-0) (Wyeth), BMS-561392 (Bristol-Myers Squibb), BN- 006 (Bone), certolizumab pegol (CAS No. 428863-50-7 or CH-138 (UCB), cilomilast (CAS No. 153259-65-5) (GlaxoSmithKline), CR-1 (Nuada Pharmaceuticals), CRx-119 (CombinatoRx), D-5410 (UCB), dacopafant (CAS No. 125372-33-0) (Sanofi-Aventis), dersalazine (CAS No. 188913-57- 7/188913-58-8) (Uriach), etanercept (CAS No. 185243-69-0) (Amgen), ethyl pyruvate (Critical (Critical Therapeutics), golimumab (CAS No. 476181-74-5) (Johnson & Johnson), hormono-immunotherapy from Ipsen, CDP571 (e.g., Humicade from UCB), IC-485 (ICOS), infliximab (CAS No. 170277-31-3) (Johnson & Johnson), IP-751 (Manhattan Pharmaceuticals), ISIS-104838 (CAS No. 250755-32-9) (ISIS Pharmaceuticals), lenalidomide (CAS No. 191732-72-6) (Celgene), lentinan (CAS No. 37339-90-5) (Ajinomoto), MDL- 201112 (CAS No. 142130-73-2) (Sanofi-Aventis), medroxyprogesterone (CAS No. 520-85-4) (InKine Pharmaceutical), N-acetylcysteine (CAS No. 616-91-1) (Zambon), NBE-P2 (DlREVO Biotech), nerelimomab (CAS No. 162774-06-3) (Chiron), OM-294DP (OM PHARMA), onercept (CAS No. 199685-57-9) (Yeda), PASSTNF-alpha (Verigen), pentoxifylline or oxypentifylline (Sanofi-Aventis), Pharmaprojects No. 4091 , 4241 , 4295, or 4488 (Sanofi-Aventis), Pharmaprojects No. 5480 (Amgen), Pharmaprojects No. 6749 (Cengent), pirfenidone (CAS No. 53179-13-8) (MARNAC), RPR- 132294 (Sanofi-Aventis), S5 (F002) (Fulcrum Pharmaceuticals), simvastatin (CAS No. 79902-63-9) (Merck & Co), STA-6292 (Synta Pharmaceuticals), tacrolimus (CAS No. 104987-11-3) (Fujisawa LifeCycle Pharma), talactoferrin alfa (CAS No. 308240-58-6) (Agennix), thalidomide (CAS No. 50-35-1) (Celgene), TNF antagonists form ProStrakan, and Synergen, TNF inhibitors (Amgen), TNF-alpha antagonists from Dynavax Technologies and Jerina AG (Germany), TNF-alpha inhibitors from IBFB Pharma and Xencor (Xencor), torbafylline (CAS No. 105102-21-4) (Sanofi-Aventis), UR-1505 (Uriach), VT-346 (Viron Therapeutics), YSI L6-(¥'s Therapeutics),- YSTH2 (Y's Therapeutics), NPI-1302a-3 (Nereus Pharmaceuticals, a TNF antagonist from Jerina AG (Germany), dersalazine, and an analogue or derivative thereof.
7699. The method of item 7600 wherein the agent is a Toll receptor inhibitor.
7700. The method of item 7600 wherein the agent is a tubulin antagonist.
7701. The method of item 7600 wherein the agent is a tyrosine kinase inhibitor selected from the group consisting of SU-011248, SUTENT from Pfizer Inc. (New York, NY), BMS-354825, PN-355 (Paracelsian Pharmaceuticals), AGN-199659 (Allergan), AAL-993 or ABP- 309 (Novartis), adaphostin (NIH), AEE-788 (Novartis), AG-013736 (OSI Pharmaceuticals), AG-13736 (Pfizer), ALT-110 (Alteris Therapeutics), AMG- 706 (Amgen), anticancer MAbs from Xencor, anti-EGFrvlll MAbs from Abgenix, anti-HER2 MAb from Abiogen, AZD-2171 or AZD-9935 (AstraZeneca), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), CEP-5214 (Cephalon), CEP-7055 (Cephalon), cetuximab (ImCIone Systems), CHIR-200131 and CHIR-258 (Chiron), CP-547632 (OSI Pharmaceuticals), CP-724714 (Pfizer), CT-301 (Creabilis Therapeutics), D- 69491 (Baxter International), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EGFR/ErbB2 inhibitors from Array BioPharma, erlotinib (CAS No. 183319- 69-9) (OSI Pharmaceuticals), EXEL-2880 (Exelixis), FK-778 (Sanofi- Aventis) , gefitinib (CAS No. 184475-35-2) (AstraZeneca), GW-2286 or GW- 654652 (GlaxoSmithKline), her2/neu antigen from AlphaVax, HER-2/neu inhibitor from Generex, Herzyme (Medipad) (Sima Therapeutics), HKI-272 (Wyeth), HuMax-EGFr (Genmab), idronoxil (CAS No. 81267-65-4) (Novogen), IGF-1 inhibitors from Ontogen, IMC-11 F8 (ImCIone Systems), kahalalide F (CAS No. 149204-42-2) (PharmaMar), KDR inhibitor from LG Life Sciences, KDR inhibitors from Abbott Laboratories, KDR kinase inhibitors (UCB), Kdr kinase inhibitors from Merck & Co, KRN-633 and KRN- 951 (Kirin Brewery), KSB-102 (Xenova), lapatinib ditosylate (CAS No. 388082-78-8) (GlaxoSmithKline), matuzumab (Merck KGaA), MDX-214 (Medarex), ME-103 (Pharmexa), MED-A300 (Gerolymatos), MNAC-13 (Lay Line Genomics), nimotuzumab (Center of Molecular Immunology), NSC- 330507 or NSC-707545 (NIH), NV-50 (Novogen), OSI-930 (OSI Pharmaceuticals), panitumumab (Abgenix), pelitinib (CAS No. 287933-82-7) (Wyeth), pertuzumab (CAS No. 380610-27-5) (Genentech), Pharmaprojects No. 3985 (Sanofi-Aventis), prostate cancer therapeutics from Sequenom (SQPC35, SQPC36, SQPC90), removab and remoxab (Trion Pharma), RG- 13022 (CAS No. 136831-48-6), RG-13291 (CAS No. 138989-50-1), or RG- 14620 (CAS No. 136831-49-7) (Sanofi-Aventis), RM-6427 (Romark), RNAi breast cancer therapy from Benitec, RP 53801 (CAS No. 125882-88-4) (Sanofi-Aventis), sorafenib tosylate (Bayer), SU-11657 (Pfizer), Tie-2 antagonists from Semaia (Hybrigenics), Tie-2 inhibitors from Ontogen, trastuzumab (CAS No. 180288-69-1) (Genentech), tyrosine kinase inhibitors from Sanofi-Aventis, U3-1287, U3-1565, U3-1784, U3-1800 (U3 Pharma), vatalanib (Novartis), VEGFR-2 kinase inhibitor from Bristol-Myers Squibb, XL-647 (Exelixis), ZD-6474 (AstraZeneca), ZK-CDK (Schering AG), an EGFR tyrosine kinase inhibitor, EKB-569 (Wyeth), and an analogue or derivative thereof.
7702. The method of item 7600 wherein the agent is a VEGF inhibitor.
7703. The method of item 7600 wherein the agent is a vitamin D receptor agonist.
7704. The method of item 7600 wherein the agent is ZD-6474 (an angiogenesis inhibitor).- - - --
7705. The method of item 7600 wherein the agent is AP- 23573 (an mTOR inhibitor).
7706. The method of item 7600 wherein the agent is synthadotin (a tubulin antagonist).
7707. The method of item 7600 wherein the agent is S-0885 (a collagenase inhibitor).
7708. The method of item 7600 wherein the agent is aplidine (an elongation factor-1 alpha inhibitor).
7709. The method of item 7600 wherein the agent is ixabepilone (an epithilone). 7710. The method of item 7600 wherein the agent is IDN- 5390 (an angiogenesis inhibitor).
7711. The method of item 7600 wherein the agent is SB- 2723005 (an angiogenesis inhibitor).
7712. The method of item 7600 wherein the agent is ABT-518 (an angiogenesis inhibitor).
7713. The method of item 7600 wherein the agent is combretastatin (an angiogenesis inhibitor).
7714. The method of item 7600 wherein the agent is anecortave acetate (an angiogenesis inhibitor).
7715. The method of item 7600 wherein the agent is SB- 715992 (a kinesin antagonist).
7716. The method of item 7600 wherein the agent is temsirolimus (an mTOR inhibitor).
7717. The method of item 7600 wherein the agent is adalimumab (a TNFα antagonist).
7718. The method of item 7600, wherein the composition comprises a polymer.
7719. The method of item 7600, wherein the composition comprises a polymer, and the polymer is, or comprises, a copolymer. 7720. The method of item 7600, wherein the composition comprises a polymer, and the polymer is, or comprises, a block copolymer.
7721. The method of item 7600, wherein the composition comprises a polymer, and the polymer is, or comprises, a random copolymer.
7722. The method of item 7600, wherein the composition comprises a polymer, and the polymer is, or comprises, a biodegradable polymer.
7723. The method of item 7600, wherein the composition comprises a polymer, and the polymer is, or comprises, a nonbiodegradable polymer.
7724. The method of item 7600, wherein the composition comprises a polymer, and the polymer is, or comprises, a hydrophilic polymer.
7725. The method of item 7600, wherein the composition comprises a polymer, and the polymer is, or comprises, a hydrophobic polymer.
7726. The method of item 7600, wherein the composition comprises a polymer, and the polymer is, or comprises, a polymer having hydrophilic domains.
7727. The method of item 7600, wherein the composition comprises a polymer, and the polymer is, or comprises, a polymer having hydrophobic domains. 7728. The method of item 7600, wherein the composition comprises a polymer, and the polymer is, or comprises, a non-conductive polymer.
7729. The method of item 7600, wherein the composition comprises a polymer, and the polymer is, or comprises, an elastomer.
7730. The method of item 7600, wherein the composition comprises a polymer, and the polymer is, or comprises, a hydrogel.
7731. The method of item 7600, wherein the composition comprises a polymer, and the polymer is, or comprises, a silicone polymer.
7732. The method of item 7600, wherein the composition comprises a polymer, and the polymer is, or comprises, a hydrocarbon polymer.
7733. The method of item 7600, wherein the composition comprises a polymer, and the polymer is, or comprises, a styrene-derived polymer.
7734. The method of item 7600, wherein the composition comprises a polymer, and the polymer is, or comprises, a butadiene-derived polymer.
7735. The method of item 7600, wherein the composition comprises a polymer, and the polymer is, or comprises, a macromer.
7736. The method of item 7600, wherein the composition comprises a polymer, and the polymer is, or comprises, a poly(ethylene glycol) polymer. 7737. The method of item 7600, wherein the composition comprises a polymer, and the polymer is, or comprises, an amorphous polymer.
7738. The method of item 7600, wherein the composition further comprises a second pharmaceutically active agent.
7739. The method of item 7600, wherein the composition further comprises an anti-inflammatory agent.
7740. The method of item 7600, wherein the composition further comprises an agent that inhibits infection.
7741. The method of item 7600, wherein the composition further comprises an anthracycline.
7742. The method of item 7600, wherein the composition further comprises doxorubicin.
7743. The method of item 7600 wherein the composition further comprises mitoxantrone.
7744. The method of item 7600 wherein the composition further comprises a fluoropyrimidine.
7745. The method of item 7600, wherein the composition further comprises 5-fluorouracil (5-FU).
7746. The method of item 7600, wherein the composition further comprises a folic acid antagonist. 7747. The method of item 7600, wherein the composition further comprises methotrexate.
7748. The method of item 7600, wherein the composition further comprises a podophylotoxin.
7749. The method of item 7600, wherein the composition further comprises etoposide.
7750. The method of item 7600, wherein the composition further comprises camptothecin.
7751. The method of item 7600, wherein the composition further comprises a hydroxyurea.
7752. The method of item 7600, wherein the composition further comprises a platinum complex.
7753. The method of item 7600, wherein the composition further comprises cisplatin.
7754. The method of item 7600 wherein the composition further comprises an anti-thrombotic agent.
7755. The method of item 7600, wherein the composition further comprises a visualization agent.
7756. The method of item 7600, wherein the composition further comprises a visualization agent, and the visualization agent is a radiopaque material, wherein the radiopaque material comprises a metal, a halogenated compound, or a barium containing compound. 7757. The method of item 7600, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, barium, tantalum, or technetium.
7758. The method of item 7600, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, an MRI responsive material.
7759. The method of item 7600, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, a gadolinium chelate.
7760. The method of item 7600, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, iron, magnesium, manganese, copper, or chromium.
7761. The method of item 7600, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, iron oxide compound.
7762. The method of item 7600, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, a dye, pigment, or colorant.
7763. The method of item 7600 wherein the agent is released in effective concentrations from the composition comprising the agent by diffusion over a period ranging from the time of administration to about 90 days.
7764. The method of item 7600 wherein the agent is released in effective concentrations from the composition comprising the agent by erosion of the composition over a period ranging from the time of administration to about 90 days.
7765. The method of item 7600 wherein the composition further comprises an inflammatory cytokine.
7766. The method of item 7600 wherein the composition further comprises an agent that stimulates cell proliferation.
7767. The method of item 7600 wherein the composition further comprises a polymeric carrier.
7768. The method of item 7600 wherein the composition is in the form of a gel, paste, or spray.
7769. The method of item 760frwherein the implant is partially constructed with the agent or the composition.
7770. The method of item 7600 wherein the implant is fully constructed with the agent or the composition.
7771. The method of item 7600 wherein the implant is impregnated with the agent or the composition.
7772. The method of item 7600, wherein the agent or the composition forms a coating, and the coating directly contacts the implant.
7773. The method of item 7600, wherein the agent or the composition forms a coating, and the coating indirectly contacts the implant. 7774. The method of item 7600 wherein the agent or the composition forms a coating, and the coating partially covers the implant.
7775. The method of item 7600, wherein the agent or the composition forms a coating, and the coating completely covers the implant.
7776. The method of item 7600 wherein the agent or the composition is located within pores or holes of the implant.
7777. The method of item 7600 wherein the agent or the composition is located within a channel, lumen, or divet of the implant.
7778. The method of item 7600 wherein the implant further comprising an echogenic material.
7779. The method of item 7600 wherein-the implant further comprises an echogenic material, wherein the echogenic material is in the form of a coating.
7780. The method of item 7600 wherein the implant is sterile.
7781. The method of item 7600 wherein the agent is delivered from the implant, wherein the agent is released into tissue in the vicinity of the implant after deployment of the implant.
7782. The method of item 7600 wherein the agent is delivered from the implant, wherein the agent is released into tissue in the vicinity of the implant after deployment of the implant, wherein the tissue is connective tissue. 7783. The method of item 7600 wherein the agent is delivered from the implant, wherein the agent is released into tissue in the vicinity of the implant after deployment of the implant, wherein the tissue is muscle tissue.
7784. The method of item 7600 wherein the agent is delivered from the implant, wherein the agent is released into tissue in the vicinity of the implant after deployment of the implant, wherein the tissue is nerve tissue.
7785. The method of item 7600 wherein the agent is delivered from the implant, wherein the agent is released into tissue in the vicinity of the implant after deployment of the implant, wherein the tissue is epithelium tissue.
7786. The method of item 7600 wherein the-agent is delivered from the implant, wherein the agent is released in effective concentrations from the implant over a period ranging from the time of deployment of the implant to about 1 year.
7787. The method of item 7600 wherein the agent is delivered from the implant, wherein the agent is released in effective concentrations from the implant over a period ranging from about 1 month to 6 months.
7788. The method of item 7600 wherein the agent is delivered from the implant, wherein the agent is released in effective concentrations from the implant over a period ranging from about 1 - 90 days.
7789. The method of item 7600 wherein the agent is delivered from the implant, wherein the agent is released in effective concentrations from the implant at a constant rate. 7790. The method of item 7600 wherein the agent is delivered from the implant, wherein the agent is released in effective concentrations from the implant at an increasing rate.
7791. The method of item 7600 wherein the agent is delivered from the implant, wherein the agent is released in effective concentrations from the implant at a decreasing rate.
7792. The method of item 7600 wherein the agent is delivered from the implant, wherein the implant comprises about 0.01 Dg to about 10 Dg of the agent.
7793. The method of item 7600 wherein the agent is delivered from the implant, wherein the implant comprises about 10 Dg to about 10 mg of the agent.
7794. The method of item 7600 wherein the agent is delivered from the implant, wherein the implant comprises about 10 mg to about 250 mg of the agent.
7795. The method of item 7600 wherein the agent is delivered from the implant, wherein the implant comprises about 250 mg to about 1000 mg of the agent.
7796. The method of item 7600 wherein the agent is delivered from the implant, wherein the implant comprises about 1000 mg to about 2500 mg of the agent.
7797. The method of item 7600 wherein the agent is delivered from the implant, wherein a surface of the implant comprises less than 0.01 Dg of the agent per mm2 of implant surface to which the agent is applied. 7798. The method of item 7600 wherein the agent is delivered from the implant, wherein a surface of the implant comprises about 0.01 Dg to about 1 Dg of the agent per mm2 of implant surface to which the agent is applied.
7799. The method of item 7600 wherein the agent is delivered from the implant, wherein a surface of the implant comprises about 1 Dg to about 10 Dg of the agent per mm2 of implant surface to which the agent is applied.
7800. The method of item 7600 wherein the agent is delivered from the implant, wherein a surface of the implant comprises about 10 Dg to about 250 Dg of the agent per mm2 of implant surface to which the agent is applied.
7801. The method of item 7600 wherein the agent is delivered from the implant, wherein a surface of the implant comprises about 250 Dg to about 1000 Dg of the agent per mm2 of implant surface to which the agent is applied.
7802. The method of item 7600 wherein the agent is delivered from the implant, wherein a surface of the implant comprises about 1000 Dg to about 2500 Dg of the agent per mm2 of implant surface to which the agent is applied.
7803. The method of item 7600, wherein the implant further comprises a coating, and the coating is a uniform coating.
7804. The method of item 7600, wherein the implant further comprises a coating, and the coating is a non-uniform coating. 7805. The method of item 7600, wherein the implant further comprises a coating, and the coating is a discontinuous coating.
7806. The method of item 7600, wherein the implant further comprises a coating, and the coating is a patterned coating.
7807. The method of item 7600, wherein the implant further comprises a coating, and the coating has a thickness of 100 Dm or less.
7808. The method of item 7600, wherein the implant further comprises a coating, and the coating has a thickness of 10 Dm or less.
7809. The method of item 7600, wherein the implant further comprises a coating, and the coating adheres to the surface of the implant upon deployment of the implant.
7810. The method of item 7600, wherein the implant further comprises a coating, and the coating is stable at room temperature for a period of at least 1 year.
7811. The method of item 7600, wherein the implant further comprises a coating, and the agent is present in the coating in an amount ranging between about 0.0001% to about 1% by weight.
7812. The method of item 7600, wherein the implant further comprises a coating, and the agent is present in the coating in an amount ranging between about 1% to about 10% by weight.
7813. The method of item 7600, wherein the implant further comprises a coating, and the agent is present in the coating in an amount ranging between about 10% to about 25% by weight. 7814. The method of item 7600, wherein the implant further comprises a coating, and the agent is present in the coating in an amount ranging between about 25% to about 70% by weight.
7815. The method of item 7600, wherein the implant further comprises a coating, and the coating comprises a polymer.
7816. The method of item 7600, wherein the implant comprises a first coating having a first composition and a second coating having a second composition.
7817. The method of item 7600, wherein the implant comprises a first coating having a first composition and a second coating having a second composition, wherein the first composition and the second composition are different.
7818. A method as in any one of items 7600-7817, wherein the device is an extravascular graft.
7819. A method as in any one of items 7600-7817, wherein the device is an intravascular graft.
7820. A method as in any one of items 7600-7817, wherein the device is a vascular graft adapted for replacing a blood vessel damaged by aneurysm.
7821. A method as in any one of items 7600-7817, wherein the device is a vascular graft adapted for replacing a blood vessel damaged by intimal hyperplasia. 7822. A method as in any one of items 7600-7817, wherein the device is a vascular graft adapted for replacing a blood vessel damaged by thrombosis.
7823. A method as in any one of items 7600-7817, wherein the device is a vascular graft adapted for providing access to blood vessel.
7824. A method as in any one of items 7600-7817, wherein the device is a vascular graft adapted for providing an alternative conduit for blood flow through a damaged or diseased area in a vein.
7825. A method as in any one of items 7600-7817, wherein the device is a vascular graft adapted for providing an alternative conduit for blood flow through a damaged or diseased area in an artery.
7826. A method as in any one of items 7600-7817, wherein the device is a synthetic bypass graft.
7827. A method as in any one of items 7600-7817, wherein the device is a femoreal-popliteal bypass graft.
7828. A method as in any one of items 7600-7817, wherein the device is a femoral-femoral bypass graft.
7829. A method as in any one of items 7600-7817, wherein the device is an axillary-femoral bypass graft.
7830. A method as in any one of items 7600-7817, wherein the device is a vein graft. 7831. A method as in any one of items 7600-7817, wherein the device is a peripheral vein graft.
7832. A method as in any one of items 7600-7817, wherein the device is a coronary vein graft.
7833. A method as in any one of items 7600-7817, wherein the device is an internal mammary graft.
7834. A method as in any one of items 7600-7817, wherein the device is a bifurcated vascular graft.
7835. A method as in any one of items 7600-7817, wherein the device is an intraluminal graft.
7836. A method as in any one of items 7600-7817, wherein the device is a prosthetic vascular graft.
7837. A method for inhibiting scarring comprising placing a device that comprises a hemodialysis access device (e.g., an implant) and an anti-scarring agent or a composition comprising an anti-scarring agent into an animal host, wherein the agent inhibits scarring.
7838. The method of item 7837 wherein the agent is an adensosine A2A receptor antagonist.
7839. The method of item 7837 wherein the agent is an AKT inhibitor. 7840. The method of item 7837 wherein the agent is an alpha 2 integrin antagonist, wherein the alpha 2 integrin antagonist is Pharmaprojects No. 5754 (Merck KgaA).
7841. The method of item 7837 wherein the agent is an alpha 4 integrin antagonist.
7842. The method of item 7837 wherein the agent is an alpha 7 nicotinic receptor agonist.
7843. The method of item 7837 wherein the agent is an angiogenesis inhibitor selected from the group consisting of AG-12,958 (Pfizer), ATN-161 (Attention LLC), neovastat, an angiogenesis inhibitor from Jerina AG (Germany), NM-3 (Mercian), VGA-1155 (Taisho), FCE-26644 (Pfizer), FCE-26950 (Pfizer), FPMA (Meiji Daries), FR-111142 (Fujisawa), GGTI-298, GM-1306 (Ligand), GPA-1734 (Novartis), NNC-47-0011 (Novo Nordisk), herbamycin (Nippon Kayaku), lenalidomide (Celegene), IP-10 (NIH), ABT-828 (Abbott), KIN-841 (Tokushima University, Japan), SF-1126 (Semafore Pharmaceuticals), laminin technology (NIH), CHIR-258 (Chiron), NVP-AEW541 (Novartis), NVP-AEW541 (Novartis), Vt 16907 (Alchemia), OXI-8007 (Oxigene), EG-3306 (Ark Therapeutics), Maspin (Arriva), ABT- 567 (Abbott), PPI-2458 (Praecis Pharmaceuticals), CC-5079, CC-4089 (Celgene), HIF-1 alpha inhibitors (Xenova), S-247 (Pfizer), AP-23573 (Ariad), AZD-9935 (Astra Zeneca), mebendazole (Introgen Therapeutics), MetAP-2 inhibitors (GlaxoSmithKline), AG-615 (Angiogene Pharmaceuticals), Tie-2 antagonists (Hybrigenics), NC-381 , CYC-381, NC-169, NC-219, NC-383, NC-384, NC-407 (Lorus Therapeutics), ATN-224 (Attenuon), ON-01370 (Onconova), Vitronectin antagonists (Amgen), SDX-103 (Salmedix), Vitronectin antagonists (Shire), CHP (Riemser), TEK (Amgen), Anecortave acetate (Alcon), T46.2 (Matrix Therapeutics), HG-2 (Heptagen), TEM antagonists (Genzyme), Oxi-4500 (Oxigene), ATN-161 (Attenuon), WX-293 (Wilex), M-2025 (Metris Therapeutics), Alphastatin (BioActa), YH-16 (Yantai Rongchang), BIBF-1120 (Boehringer Ingelheim), BAY-57-9352 (Bayer), AS- 1404 (Cancer Research Technology), SC-77964 (Pfizer), glycomimetics (BioTie Therapies), TIE-2 Inhibitors (Ontogen), DIMI, Octamer (Octamer), ABR-215050 (Active Biotech), ABT-518 (Abbott), KDR inhibitors (Abbott), BSF-466895 (Abbott), SCH-221153 (Schering-Plough), DAC:antiangiogenic (ConjuChem), TFPI (EntreMed), AZD-2171 (Astra-Zenaca), CDC-394 (Celgene), LY290293 (EIi Lilly), IDN-5390 (Indena), Kdr Kinase Inhibitors (Merck), CT-113020, CT-116433, CT-116563, CT-31890, CT-32228) (Cell Therapeutics), A-299620 (Abbott), TWEAK Inhibitor (Amgen), VEGF modulators (Johnson and Johnson), Tum-N53, tumstatin (Genzyme), Thios- 1 , Thios-2 (Thios Pharmaceuticals), MV-6401 (Miravant Medical Technologies), Spisulosine (PharmaMar), CEP-7055 (Cephalon), AUV-201 (Auvation), LM-609 (EIi Lilly), SKF-106615 (AnorMED), Oglufanide disodium (Cytran), BW-114 (Pharminox), Calreticulin (NIH), WX-678 (Wilex), SD-7784 (Pfizer), WX-UK1 (Wilex), SH-268 (Schering AG), 2-Me-PGA (Celgene), S- 137 (Pfizer), ZD-6126 (Angiogene Pharmaceuticals), SG-292 (SignalGen), Benefin (Lane Labs), A6, A36 (Angstrom), SB-2723005 (GlaxoSmithKline), SC-7 (Cell Therapeutics), ZEN-014 (AEterna Zentaris), 2-methoxyestradiol (EntreMed), NK-130119 (Nippon Kayaku), CC-10004 (Celgene), AVE- 8062A (Ajinomoto), Tacedinaline (Pfizer), Actinonin (Tokyo Metropolitan Institute of Medical Science), Lenalidomide (Celgene), VGA-1155, BTO-956 (SRI International), ER-68203-00 (Eisai), CT-6685 (UCB), JKC-362 (Phoenix Pharmaceuticals), DMI-3798 (DMI Biosciences, Angiomate (Ipsen), ZD-6474 (AstraZeneca), CEP-5214 (Cephalon), Canstatin (Genzyme), NM-3 (Mercian), Oridigm (MediQuest Therapeutics), Exherin (Adherex), BLS-0597 (Boston Life Sciences), PTC-299 (PTC Therapeutics), NPl-2358 (Nereus Pharmaceuticals), CGP-79787 (Novartis), AEE-788 (Novartis), CKD-732 (Chong Kun Dang), CP-564959 (OSI Pharmaceuticals), CM-101 (CarboMed), CT-2584, CT3501 (Cell Therapeutics), combretastatin and analogues and derivatives thereof (Oxigene), Rebimastat (Bristol-Meyers Squibb), Dextrin 2-sulfate (ML Laboratories), Cilengitide (Merk KGaA), NSC-706704 (Pharminox), KRN- 951 (Kirin Brewery), Ukrain, NSC-631570 (Nowicky Pharma), Tecogalan sodium (Daiichi Pharmaceutical), Tz-93 (Tsumura), TBC-1635 (Encysive Pharmaceuticals), TAN-1120 (Takeda), Semaxanib (Pfizer), BDI-7800 (Biopharmacopae), SD-186, SD-983 (Bristol-Meyers Squibb), SB-223245 (GlaxoSmithKline), SC-236 (Pfizer), RWJ-590973 (Johnson and Johnson), ILX-1850 (Genzyme), SC-68488, S-836 (Pfizer), CG-55069-11 (CuraGen),
Ki-23057 (Kirin Brewery), CCX-700 (Chemoentryx), Pegaptanib octasodium
/" (Giled Sciences), ANGIOCOL (available from Biostratum Inc.), or an , analogue or derivative thereof.
7844. The method of item 7837 wherein the agent is an apoptosis antagonist.
7845. The method of item 7837 wherein the agent is an apoptosis activator.
7846. The method of item 7837 wherein the agent is a beta 1 integrin antagonist.
7847. The method of item 7837 wherein the agent is a beta tubulin inhibitor.
7848. The method of item 7837 wherein the agent is a blocker of enzyme production in Hepatitis C.
7849. The method of item 7837 wherein the agent is a Bruton's tyrosine kinase inhibitor. 7850. The method of item 7837 wherein the agent is a calcineurin inhibitor.
7851. The method of item 7837 wherein the agent is a caspase 3 inhibitor.
7852. The method of item 7837 wherein the agent is a CC chemokine receptor antagonist.
7853. The method of item 7837 wherein the agent is a cell cycle inhibitor selected from the group consisting of SNS-595 (Sunesis), synthadotin, KRX-0403, and an analogue or derivative thereof.
7854. The method of item 7837 wherein the agent is a cathepsin B inhibitor.
7855. The method of item 7837 wherein the agent is a cathepsin K inhibitor, wherein the cathepsin K inhibitor is 462795 (GlaxoSmithKline), INPL-022-D6 (Amura Therapeutics), or an analogue or derivative thereof.
7856. The method of item 7837 wherein the agent is a cathepsin L inhibitor.
7857. The method of item 7837 wherein the agent is a CD40 antagonist.
7858. The method of item 7837 wherein the agent is a chemokine receptor agonist. 7859. The method of item 7837 wherein the agent is a chymase inhibitor.
7860. The method of item 7837 wherein the agent is a collagenase antagonist.
7861. The method of item 7837 wherein the agent is a CXCR antagonist.
7862. The method of item 7837 wherein the agent is a cyclin dependent kinase inhibitor selected from the group consisting of a CDK-1 inhibitor, a CDK-2 inhibitor, a CDK- 4 inhibitor, a CDK-6 inhibitor, a CAK1 inhibitor from GPC Biotech and Bristol-Myers Squibb, RGB-286199 (GPC Biotech), an anticancer agent from Astex Technology, a CAK1 inhibitor from GPC Biotech, a CDK inhibitor from Sanofi-Aventis, a CDK1/CDK2 inhibitor from Amgen, a CDK2 inhibitor from SUGEN-2 (Pfizer), a hearing loss therapy agent (Sound Pharmaceuticals), PD-0332991 (Pfizer), RGB-286199 (GPC Biotech), Ro-0505124 (Hoffmann-La Roche), a Ser/Thr kinase inhibitor from Lilly (EIi Lilly), CVT-2584 (CAS No. 199986-75-9) (CV Therapeutics), and an analogue or derivative thereof.
7863. The method of item 7837 wherein the agent is a cyclooxygenase 1 inhibitor.
7864. The method of item 7837 wherein the agent is a DHFR inhibitor.
7865. The method of item 7837 wherein the agent is a dual integrin inhibitor. 7866. The method of item 7837 wherein the agent is an elastase inhibitor.
7867. The method of item 7837 wherein the agent is an elongation factor-1 alpha inhibitor.
7868. The method of item 7837 wherein the agent is an endothelial growth factor antagonist.
7869. The method of item 7837 wherein the agent is an endothelial growth factor receptor kinase inhibitor selected from the group consisting of sorafenib tosylate (Bayer), AAL-993 (Novartis), ABP-309 (Novartis), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), E- 7080 (Eisai), EG-3306 (Ark Therapeutics), EXEL-2880 (Exelixis), GW- 654652 (GlaxoSmithKline), a KDR inhibitor from LG Life Sciences, CT-6685 and CT-6729 (UCB), KRN-633 and KRN-951 (Kirin Brewery), OSI-930 (OSI Pharmaceuticals), SP-5.2 (Supratek Pharma), SU-11657 (Pfizer), a Tie-2 antagonist (Hybrigenics), a VEGFR-2 kinase inhibitor (Bristol-Myers Squibb), XL-647 (Exelixis), a KDR inhibitor from Abbott Laboratories, sorafenib tosylate, and an analogue or derivative thereof.
7870. The method of item 7837 wherein the agent is an endotoxin antagonist.
7871. The method of item 7837 wherein the agent is an epothilone and tubulin binder.
7872. The method of item 7837 wherein the agent is an estrogen receptor antagonist. 7873. The method of item 7837 wherein the agent is an FGF inhibitor.
7874. The method of item 7837 wherein the agent is a farnexyl transferase inhibitor.
7875. The method of item 7837 wherein the agent is farnesyltransferase inhibitor selected from the group of A-197574 (Abbott), a famesyltransferase inhibitor from Servier, FPTIII (Strathclyde Institute for Drug R), LB-42908 (LG Life Sciences), Pharmaprojects No. 5063 (Genzyme), Pharmaprojects No. 5597 (Ipsen), Yissum Project No. B-1055 (Yissum), and an analogue or derivative thereof.
7876. The method of item 7837 wherein the agent is an FLT-3 kinase inhibitor.
7877. The method of item 7837 wherein the agent is an FGF receptor kinase inhibitor.
7878. The method of item 7837 wherein the agent is a fibrinogen antagonist selected from the group consisting of AUV-201 (Auvation), MG-13926 (Sanofi-Aventis), plasminogen activator (CAS No. 105913-11-9) (from Sanofi-Aventis or UCB), plasminogen activator-2 (tPA- 2) (Sanofi-Aventis), pro-urokinase (CAS No. 82657-92-9) (Sanofi-Aventis), and an analogue or derivative thereof.
7879. The method of item 7837 wherein the agent is a heat shock protein 90 antagonist selected from the group consisting of SRN-005 (Sirenade), geldanamycin, NSC-33050 (17-allylaminogeldanamycin; 17- AAG), 17-dimethylaminoethylamino-17-demethoxy-geldanamycin (17- DMAG), rifabutin (rifamycin XIV, 1',4-didehydro-1-deoxy-1 ,4-dihydro-5'-(2- methylpropyl)-1-oxo-), and an analogue or derivative thereof.
7880. The method of item 7837 wherein the agent is a histone deacetylase inhibitor.
7881. The method of item 7837 wherein the agent is an HMGCoA reductase inhibitor selected from the group consisting of an atherosclerosis therapeutic from Lipid Sciences, ATI-16000 (ARYx Therapeutics), KS-01-019 (Kos Pharmaceuticals), Pharmaprojects No. 2197 (Sanofi-Aventi), RP 61969 (Sanofi-Aventis), and an analogue or derivative thereof.
7882. The method of item 7837 wherein the agent is an ICAM inhibitor.
7883. The method of item 7837 wherein the agent is an IL, ICE and IRAK antagonist, wherein the antagonist is a CJ-14877, CP-424174 (Pfizer), NF-61 (Negma-Lerads), and an analogue or derivative thereof.
7884. The method of item 7837 wherein the agent is an IL-2 inhibitor.
7885. The method of item 7837 wherein the agent is an immunosuppressant selected from the group consisting of teriflunomide (Sanofi Aventis), chlorsulfaquinoxalone (NSC-339004), chlorsulfaquinoxalone sulfate, CS-712 (Sankyo), ismomultin alfa (CAS No. 457913-93-8) (Akzo Nobel), antiallergics from GenPat77, antiinflammatories or AT-005 (Androclus Therapeutics), autoimmune disease therapeutics from EpiVax, BN-007 (Bone), budesonide (CAS No. 51333-22- 3) (MAP Pharmaceuticals), CO-14 (Genzyme), edratide (CAS No. 433922- 67-9) (Teva), EP-314 (Enanta), eprovafen (CAS No. 101335-99-3) (Sanofi- Aventis), HWA-131 (CAS No. 118788-41-3) (Sanofi-Aventis), immunomodulators from MerLion Pharmaceuticals, immunosuppressives from Alchemia, IPL-12 (Inflazyme), MDL-9563 (CAS No. 27086-86-8) (Sanofi-Aventis), Pharmaprojects No. 2330 (Sanofi-Aventis), Pharmaprojects No. 6426 (Abgenix), PXS-25 (Pharmaxis), rosmarinic acid (CAS No. 20283-92-5) (Sanofi-Aventis), RP 42927 or RP 54745 (CAS No. 135330-08-4) (Sanofi-Aventis), SGN-35 (Seattle Genetics), ST-1959 (Sigma-Tau), type I diabetes therapy from SYNX Pharma, UNIL-88 (Debiopharm), VP-025 (Vasogen), VR-694 (Vectura), PRTX-001 (Protalex), and an analogue or derivative thereof.
7886. The method of item 7837 wherein the agent is an IMPDH (inosine monophosphate).
- 7887. The method of item 7837 wherein the agent is an integrin antagonist.
7888. The method of item 7837 wherein the agent is an interleukin antagonist.
7889. The method of item 7837 wherein the agent is an inhibitor of type III receptor tyrosine kinase.
7890. The method of item 7837 wherein the agent is an irreversible inhibitor of enzyme methionine aminopeptidase type 2.
7891. The method of item 7837 wherein the agent is an isozyme selective delta protein kinase C inhibitor. 7892. The method of item 7837 wherein the agent a JAK3 enzyme inhibitor.
7893. The method of item 7837 wherein the agent is a JNK inhibitor.
7894. The method of item 7837 wherein the agent is a kinase inhibitor.
7895. The method of item 7837 wherein the agent is kinesin antagonist.
7896. The method of item 7837 wherein the agent is a kinesin antagonist.
7897. The method of item 7837 wherein the agent is a leukotriene inhibitor and antagonist selected from the group consisting of ambicromil (CAS No. 58805-38-2) (Sanofi-Aventis), amelubant (CAS No. 346735-24-8) (Boehringer Ingelheim), DW-1141 (Dong Wha), ebselen (Daiichi Pharmaceutical), ibudilast (Kyorin), leucotriene inhibitors from Sanofi-Aventis, lymphotoxin-beta receptor (LT-D) from Biogen Idee, Pharmaprojects No. 1535 and 2728 (CAS No. 119340-33-9) (Sanofi- Aventis), R-112 (Rigel), Rev-5367 (CAS No. 92532-05-3) (Sanofi-Aventis), RG-14893 (CAS No. 141835-49-6) (Sanofi-Aventis), RG-5901-A (CAS No. 101910-24-1), 92532-23-5, RP 66153 (CAS No. 142422-79-5), RP 66364 (CAS No.186916-92-5), RP 69698 (CAS No. 141748-00-7) (Sanofi-Aventis), SC-411930 (Pfizer), SC-41930 (CAS No. 120072-59-5) (Pfizer), SC-50605 (CAS No. 138828-39-4) (Pfizer), SC-51146 (CAS No. 141059-52-1), SC- 53228 (CAS No. 153633-01-3) (Pfizer), spaglumic acid (ZY-15106) (CAS No.3106-85-2), 80619-64-3 (Novartis), tipredane (CAS No. 85197-77-9) (Bristol-Myers Squibb), U-75302 (CAS No. 119477-85-9) (Pfizer), and analogue or derivative thereof.
7898. The method of item 7837 wherein the agent is an MAP kinase inhibitor.
7899. The method of item 7837 wherein the agent is a matrix metalloproteinase inhibitor.
7900. The method of item 7837 wherein the agent is an MCP- CCR2 inhibitor.
7901. The method of item 7837 wherein the agent is an mTOR inhibitor.
- - - 7902. The method of item 7837 wherein the agent is an mTOR kinase inhibitor.
7903. The method of item 7837 wherein the agent is a microtubule inhibitor selected from the group consisting of antibody- maytansinoid conjugates from Biogen Idee, colchicines (MantiCore Pharmaceuticals), anticancer immunoconjugates from Johnson & Johnson, DIME from Octamer, gni-1f (GNI), huC242-DM4, huMy9-6-DM1 (ImmunoGen), IDN-5404 (Indena), IMO-098, IMOderm (Imotep), mebendazole (Introgen Therapeutics), microtubule poisons from Cambridge Enterprise, paclitaxel such as LOTAX from Aphios (CAS No. 33069-62-4), Genexol-PM from Samyang, Pharmaprojects No. 6383 (Tapestry Pharmaceuticals), RPR-112378 (Sanofi-Aventis), SGN-75 (Seattle Genetics), SPL-7435 (Starpharma), SSR-250411 (Sanofi-Aventis), trastuzumab-DM1 (Genentech), vinorelbine, and an analogue or derivative thereof. 7904. The method of item 7837 wherein the agent is an MIF inhibitor.
7905. The method of item 7837 wherein the agent is an MMP inhibitor.
7906. The method of item 7837 wherein the agent is a neurokinin (NK) antagonist selected from the group consisting of anthrotainin (CAS No. 148084-40-6) (Sanofi-Aventis), an IBS thereapeutic from ArQuIe, MDL-105212A (CAS No. 167261-60-1) (Ssanofi-Aventis), Pharmaprojects No. 2744, 3258 (CAS No. 139167-47-8) 4006, 4201 , or 5986 (Sanofi-Aventis), RP 67580 (CAS No. 135911-02-3), SR-144190 (CAS No. 201152-86-5), SSR-240600, SSR-241586 (Sanofi-Aventis), TKA-457 (Novartis), vestipitant mesylate (CAS No. 334476-64-1) (GlaxoSmithKline), Win-64821 (Sanofi-Aventis), PRX-96026 (Predix Pharmaceuticals), and an analogue or derivative thereof.
7907. The method of item 7837 wherein the agent is an NF kappa B inhibitor selected from the group consisting of AVE-0545 or AVE- 0547 (Sanofi-Aventis), bortezomib (CAS No. 179324-69-7) (Millennium Pharmaceuticals), dexanabinol (CAS No. 112924-45-5) (Pharmos), dexlipotam (Viatris), Pharmaprojects No. 6283 (INDRA) (OXiGENE)1 IPL- 576092 (CAS No. 137571-30-3) (Inflazyme), NFKB decoy (Corgentech), NFKB decoy oligo (AnGes MG), NFKB's from Ariad, osteoporosis treatments or S5 (F005) from Fulcrum Pharmaceuticals, P61 (Phytopharm), R-flurbiprofen (CAS No. 5104-49-4) (Encore Pharmaceuticals), and an analogue or derivative thereof.
7908. The method of item 7837 wherein the agent is a nitric oxide agonist. 7909. The method of item 7837 wherein the agent is an ornithine decarboxylase inhibitor.
7910. The method of item 7837 wherein the agent is a p38 MAP kinase inhibitor selected from the group consisting of AZD-6703 (AstraZeneca), JX-401 (Jexys Pharmaceuticals), BMS-2 (Bristol-Myers Squibb), a p38 MAP kinase inhibitor from Novartis, a p38-alpha MAP kinase inhibitor from Amphora, Pharmaprojects No. 5704 (Pharmacopeia), RPR- 200765A (Sanofi-Aventis), SD-282 (Johnson & Johnson), TAK-715 (Takeda), and an analogue or derivative thereof.
7911. The method of item 7837 wherein the agent is a palmitoyl-protein thϊoesterase inhibitor.
7912. The method of item 7837 wherein the agent is a PDGF receptor kinase inhibitor selected from the group consisting of AAL-993, AMN-107, or ABP-309 (Novartis), AMG-706 (Amgen), BAY-57-9352 (Bayer), CDP-860 (UCB), E-7080 (Eisai), imatinib (CAS No. 152459-95-5) (Novartis), OSI-930 (OSI Pharmaceuticals), RPR-127963E (Sanofi-Aventis), RWJ-540973 (Johnson & Johnson), sorafenib tosylate (Bayer), SU-11657 (Pfizer), tandutinib (CAS No. 387867-13-2) (Millennium Pharmaceuticals), vatalanib (Novartis), ZK-CDK (Schering AG), and an analogue or derivative thereof.
7913. The method of item 7837 wherein the agent is (-)- halofenate (Metabolex), AMG-131 (Amgen), antidiabetics from Japan Tobacco, AZD-4619, AZD-8450, AZD-8677 (AstraZeneca), DRF-10945, balaglitazone (Dr Reddy's), CS-00088, CS-00098 (Chipscreen Biosciences), E-3030 (Eisai), etalocib (CAS No. 161172-51-6) (EIi Lilly), GSK-641597 (Ligand), GSK-677954 (GlaxoSmithKline), GW-409544 (Ligand), GW- 590735 (GlaxoSmithKline), K-111 (Hoffmann-La Roche), LY-518674 (EIi Lilly), LY-674 (Ligand), LY-929 (Ligand), MC-3001 , MC-3002 (MaxoCore Pharmaceuticals), metformin HCI + pioglitazone (CAS No. 1115-70-4 and 112529-15-4), ACTOPLUS MET from Andrx), muraglitazar (CAS No. 331741-94-7) (Bristol-Myers Squibb), naveglitazar (Ligand), oleoylethanolamide (Kadmus Pharmaceuticals), ONO-5129, pioglitazone hydrochloride (CAS No. 111025-46-8 and 112529-15-4) (Takeda), PLX-204 (Plexxikon), PPAR agonists from Genfit, PPAR delta agonists from EIi Lilly, PPAR-alpha agonists from CrystalGenomics, PPAR-gamma modulators and PPAR-β modulators from CareX, rosiglitazone maleate (CAS No. 122324- 73-4 or 155141-29-0) (GlaxoSmithKline), rosiglitazone maleate/glimepir (CAS No. 155141-29-0 and 93479-97-1), AVANDARYL, rosiglitazone maleate/metformin extend (CAS No. 155141-29-0 and 657-24-9), AVANDAMET, rosiglitazone maleate+metformin, AVANDAMET (GlaxoSmithKline), tesaglitazar (AstraZeneca), and an analogue or derivative thereof.
7914. The method of item 7837 wherein the agent is a phosphatase inhibitor.
7915. The method of item 7837 wherein the agent is a phosphodiesterase (PDE) inhibitor selected from the group consisting of avanafil (Tanabe Seiyaku), dasantafil (CAS No. 569351-91-3) (Schering- Plough), A-906119 (CAS No. 134072-58-5), DL-850 (Sanofi-Aventis), GRC- 3015, GRC-3566, GRC-3886 (Glenmark), HWA-153 (CAS No. 56395-66-5) (Sanofi-Aventis), hydroxypumafentrine (Altana), IBFB-130011 , IBFB-14-016, IBFB-140301 , IBFB-150007, IBFB-211913 (IBFB Pharma), L-826141 (Merck & Co), medorinone (CAS No. 88296-61-1) (Sanofi-Aventis), MEM- 1917 (Memory Pharmaceuticals), ND-1251 (Neuro3d), PDE inhibitors from ICOS, PDE IV inhibitors from Memory Pharmaceuticals and CrystalGenomics, Pharmaprojects No. 2742 and 6141 (Sanofi-Aventis), QAD-171 (Novartis), RHC-2963 (CAS No. 76993-12-9 and 76993-14-1), RPR-117658, RPR-122818 derivatives, SR-24870 , and RPR-132294 (Sanofi-Aventis), SK-350 (ln2Gen), stroke therapy agents from deCODE Genetics, TAS-203 (Taiho), tofimilast (CAS No. 185954-27-2) (Pfizer), UK- 371800 (Pfizer), WIN-65579 (CAS No. 158020-82-7) (Sanofi-Aventis), IBFB- 130020 (IBFB Pharma), OPC-6535 (CAS No. 145739-56-6) (Otsuka), a phosphodiesterase III inhibitor, enoximone, a phosphodiesterase IV inhibitor, fosfosal, Atopik (Barrier Therapeutics), triflusal, a phosphodiesterase V inhibitor, and an analogue or derivative thereof.
7916. The method of item 7837 wherein the agent is a PKC inhibitor.
7917. The method of item 7837 wherein the agent is a platelet activating factor antagonist.
- - - - 7918. The method of item 7837 wherein-the agent is a platelet-derived growth factor receptor kinase inhibitor.
7919. The method of item 7837 wherein the agent is a prolyl hydroxylase inhibitor.
7920. The method of item 7837 wherein the agent is a polymorphonuclear neutrophil inhibitor.
7921. The method of item 7837 wherein the agent is a protein kinase B inhibitor.
7922. The method of item 7837 wherein the agent is a protein kinase C stimulant. 7923. The method of item 7837 wherein the agent is a purine nucleoside analogue.
7924. The method of item 7837 wherein the agent is a purinoreceptor P2X antagonist.
7925. The method of item 7837 wherein the agent is a Raf kinase inhibitor.
7926. The method of item 7837 wherein the agent is a reversible inhibitor of ErbB1 and ErbB2.
7927. The method of item 7837 wherein the agent is a ribonucleoside triphosphate reductase inhibitor.
- - - 7928. The method of item 7837 wherein the agent is an SDF- 1 antagonist.
7929. The method of item 7837 wherein the agent is a sheddase inhibitor.
7930. The method of item 7837 wherein the agent is an SRC inhibitor.
7931. The method of item 7837 wherein the agent is a stromelysin inhibitor.
7932. The method of item 7837 wherein the agent is an Syk kinase inhibitor. 7933. The method of item 7837 wherein the agent is a telomerase inhibitor.
7934. The method of item 7837 wherein the agent is a TGF beta inhibitor selected from the group consisting of pirfenidone (CAS No. 53179-13-8) (MARNAC)1 tranilast (CAS No. 53902-12-8) (Kissei), IN-1130 (ln2Gen), mannose-6-phosphate (BTG), TGF-β antagonists from Inflazyme (Pharmaprojects No. 6075), TGF-β antagonists from Sydney, non-industrial source), TGF-β I receptor kinase inhibitors from EIi Lilly, TGF-β receptor inhibitors from Johnson & Johnson, and an analogue or derivative thereof.
7935. The method of item 7837 wherein the agent is a TNFα antagonist or TACE inhibitor selected from the group consisting of adalimumab (CAS No. 331731-18-1) (Cambridge Antibody Technology), AGIX-4207 (AtheroGenics), AGT-1 (Advanced Biotherapy), an antiinflammatory from Borean Pharma, Cellzome, or Paradigm Therapeutics, anti-inflammatory vaccine (TNF-alpha kinoid) from Neovacs, humanized anti-TNF antibody or an anti-TNF MAb (CB0006) Celltech (UCB), apratastat (CAS No. 287445-51-0) (Wyeth), BMS-561392 (Bristol-Myers Squibb), BN- 006 (Bone), certolizumab pegol (CAS No. 428863-50-7 or CH-138 (UCB), cilomilast (CAS No. 153259-65-5) (GlaxoSmithKline), CR-1 (Nuada Pharmaceuticals), CRx-119 (CombinatoRx), D-5410 (UCB), dacopafant (CAS No. 125372-33-0) (Sanofi-Aventis), dersalazine (CAS No. 188913-57- 7/188913-58-8) (Uriach), etanercept (CAS No. 185243-69-0) (Amgen), ethyl pyruvate (Critical (Critical Therapeutics), golimumab (CAS No. 476181-74-5) (Johnson & Johnson), hormono-immunotherapy from Ipsen, CDP571 (e.g., Humicade from UCB), IC-485 (ICOS), infliximab (CAS No. 170277-31-3) (Johnson & Johnson), IP-751 (Manhattan Pharmaceuticals), ISIS-104838 (CAS No. 250755-32-9) (ISIS Pharmaceuticals), lenalidomide (CAS No. 191732-72-6) (Celgene), lentinan (CAS No. 37339-90-5) (Ajinomoto), MDL- 201112 (CAS No. 142130-73-2) (Sanofi-Aventis), medroxyprogesterone (CAS No. 520-85-4) (InKine Pharmaceutical), N-acetylcysteine (CAS No. 616-91-1) (Zambon), NBE-P2 (DIREVO Biotech), nerelimomab (CAS No. 162774-06-3) (Chiron), OM-294DP (OM PHARMA), onercept (CAS No. 199685-57-9) (Yeda), PASSTNF-alpha (Verigen), pentoxifylline or oxypentifylline (Sanofi-Aventis), Pharmaprojects No. 4091 , 4241 , 4295, or 4488 (Sanofi-Aventis), Pharmaprojects No. 5480 (Amgen), Pharmaprojects No. 6749 (Cengent), pirfenidone (CAS No. 53179-13-8) (MARNAC), RPR- 132294 (Sanofi-Aventis), S5 (F002) (Fulcrum Pharmaceuticals), simvastatin (CAS No. 79902-63-9) (Merck & Co), STA-6292 (Synta Pharmaceuticals), tacrolimus (CAS No. 104987-11-3) (Fujisawa LifeCycle Pharma), talactoferrin alfa (CAS No. 308240-58-6) (Agennix), thalidomide (CAS No. 50-35-1) (Celgene), TNF antagonists form ProStrakan, and Synergen, TNF inhibitors (Amgen), TNF-alpha antagonists from Dynavax Technologies and Jerina AG (Germany), TNF-alpha inhibitors from IBFB Pharma and Xencor (Xencor), torbafylline (CAS No. 105102-21-4) (Sanofi-Aventis), UR-1505 (Uriaeh), VT-346 (Viron Therapeutics), YSIL6 (Y's Therapeutics), YSTH2 (Y's Therapeutics), NPI-1302a-3 (Nereus Pharmaceuticals, a TNF antagonist from Jerina AG (Germany), dersalazine, and an analogue or derivative thereof.
7936. The method of item 7837 wherein the agent is a Toll receptor inhibitor.
7937. The method of item 7837 wherein the agent is a tubulin antagonist.
7938. The method of item 7837 wherein the agent is a tyrosine kinase inhibitor selected from the group consisting of SU-011248, SUTENT from Pfizer Inc. (New York, NY), BMS-354825, PN-355 (Paracelsian Pharmaceuticals), AGN-199659 (Allergan), AAL-993 or ABP- 309 (Novartis), adaphostin (NIH), AEE-788 (Novartis), AG-013736 (OSI Pharmaceuticals), AG-13736 (Pfizer), ALT-110 (Alteris Therapeutics), AMG- 706 (Amgen), anticancer MAbs from Xencor, anti-EGFrvlll MAbs from Abgenix, anti-HER2 MAb from Abiogen, AZD-2171 or AZD-9935 (AstraZeneca), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), CEP-5214 (Cephalon), CEP-7055 (Cephalon), cetuximab (ImCIone Systems), CHIR-200131 and CHIR-258 (Chiron), CP-547632 (OSI Pharmaceuticals), CP-724714 (Pfizer), CT-301 (Creabilis Therapeutics), D- 69491 (Baxter International), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EGFR/ErbB2 inhibitors from Array BioPharma, erlotinib (CAS No. 183319- 69-9) (OSI Pharmaceuticals), EXEL-2880 (Exelixis), FK-778 (Sanofi- Aventis) , gefitinib (CAS No. 184475-35-2) (AstraZeneca), GW-2286 or GW- 654652 (GlaxoSmithKline), her2/neu antigen from AlphaVax, HER-2/neu inhibitor from Generex, Herzyme (Medipad) (Sirna Therapeutics), HKI-272 (Wyeth), HuMax-EGFr (Genmab), idronoxil (CAS No. 81267-65-4) (Novogen), IGF-1 inhibitors from Ontogen, IMC-11 F8 (ImCIone Systems), kahalalide F (CAS No. 149204-42-2) (PharmaMar), KDR inhibitor from LG Life Sciences, KDR inhibitors from Abbott Laboratories, KDR kinase inhibitors (UCB), Kdr kinase inhibitors from Merck & Co, KRN-633 and KRN- 951 (Kirin Brewery), KSB-102 (Xenova), lapatinib ditosylate (CAS No. 388082-78-8) (GlaxoSmithKline), matuzumab (Merck KGaA), MDX-214 (Medarex), ME-103 (Pharmexa), MED-A300 (Gerolymatos), MNAC-13 (Lay Line Genomics), nimotuzumab (Center of Molecular Immunology), NSC- 330507 or NSC-707545 (NlH), NV-50 (Novogen), OSI-930 (OSi Pharmaceuticals), panitumumab (Abgenix), pelitinib (CAS No. 287933-82-7) (Wyeth), pertuzumab (CAS No. 380610-27-5) (Genentech), Pharmaprojects No. 3985 (Sanofi-Aventis), prostate cancer therapeutics from Sequenom (SQPC35, SQPC36, SQPC90), removab and remoxab (Trion Pharma), RG- 13022 (CAS No. 136831-48-6), RG-13291 (CAS No. 138989-50-1), or RG- 14620 (CAS No. 136831-49-7) (Sanofi-Aventis), RM-6427 (Romark), RNAi breast cancer therapy from Benitec, RP 53801 (CAS No. 125882-88-4) (Sanofi-Aventis), sorafenib tosylate (Bayer), SU-11657 (Pfizer), Tie-2 antagonists from Semaia (Hybrigenics), Tie-2 inhibitors from Ontogen, trastuzumab (CAS No. 180288-69-1) (Genentech), tyrosine kinase inhibitors from Sanofi-Aventis, U3-1287, U3-1565, U3-1784, U3-1800 (U3 Pharma), vatalanib (Novartis), VEGFR-2 kinase inhibitor from Bristol-Myers Squibb, XL-647 (Exelixis), ZD-6474 (AstraZeneca), ZK-CDK (Schering AG), an EGFR tyrosine kinase inhibitor, EKB-569 (Wyeth), and an analogue or derivative thereof.
7939. The method of item 7837 wherein the agent is a VEGF inhibitor.
7940. The method of item 7837 wherein the agent is a vitamin D receptor agonist.
7941. The method of item 7837 wherein the agent is ZD-6474 (an angiogenesis inhibitor).
7942. The method of item 7837 wherein the agent is AP- 23573 (an mTOR inhibitor).
7943. The method of item 7837 wherein the agent is synthadotin (a tubulin antagonist).
7944. The method of item 7837 wherein the agent is S-0885 (a collagenase inhibitor).
7945. The method of item 7837 wherein the agent is aplidine (an elongation factor-1 alpha inhibitor).
7946. The method of item 7837 wherein the agent is ixabepilone (an epithilone). 7947. The method of item 7837 wherein the agent is IDN- 5390 (an angiogenesis inhibitor).
7948. The method of item 7837 wherein the agent is SB- 2723005 (an angiogenesis inhibitor).
7949. The method of item 7837 wherein the agent is ABT-518 (an angiogenesis inhibitor).
7950. The method of item 7837 wherein the agent is combretastatin (an angiogenesis inhibitor).
7951. The method of item 7837 wherein the agent is anecortave acetate (an angiogenesis inhibitor).
7952. The method of item 7837 wherein the agent is SB- 715992 (a kinesin antagonist).
7953. The method of item 7837 wherein the agent is temsirolimus (an mTOR inhibitor).
7954. The method of item 7837 wherein the agent is adalimumab (a TNFα antagonist).
7955. The method of item 7837, wherein the composition comprises a polymer.
7956. The method of item 7837, wherein the composition comprises a polymer, and the polymer is, or comprises, a copolymer. 7957. The method of item 7837, wherein the composition comprises a polymer, and the polymer is, or comprises, a block copolymer.
7958. The method of item 7837, wherein the composition comprises a polymer, and the polymer is, or comprises, a random copolymer.
7959. The method of item 7837, wherein the composition comprises a polymer, and the polymer is, or comprises, a biodegradable polymer.
7960. The method of item 7837, wherein the composition comprises a polymer, and the polymer is, or comprises, a nonbiodegradable polymer.
-7961. The method of item 7837, wherein the composition comprises a polymer, and the polymer is, or comprises, a hydrophilic polymer.
7962. The method of item 7837, wherein the composition comprises a polymer, and the polymer is, or comprises, a hydrophobic polymer.
7963. The method of item 7837, wherein the composition comprises a polymer, and the polymer is, or comprises, a polymer having hydrophilic domains.
7964. The method of item 7837, wherein the composition comprises a polymer, and the polymer is, or comprises, a polymer having hydrophobic domains. 7965. The method of item 7837, wherein the composition comprises a polymer, and the polymer is, or comprises, a non-conductive polymer.
7966. The method of item 7837, wherein the composition comprises a polymer, and the polymer is, or comprises, an elastomer.
7967. The method of item 7837, wherein the composition comprises a polymer, and the polymer is, or comprises, a hydrogel.
7968. The method of item 7837, wherein the composition comprises a polymer, and the polymer is, or comprises, a silicone polymer.
7969. The method of item 7837, wherein the composition comprises a polymer, and the polymer is, or comprises, a hydrocarbon polymer.
7970. The method of item 7837, wherein the composition comprises a polymer, and the polymer is, or comprises, a styrene-derived polymer.
7971. The method of item 7837, wherein the composition comprises a polymer, and the polymer is, or comprises, a butadiene-derived polymer.
7972. The method of item 7837, wherein the composition comprises a polymer, and the polymer is, or comprises, a macromer.
7973. The method of item 7837, wherein the composition comprises a polymer, and the polymer is, or comprises, a poly(ethylene glycol) polymer. 7974. The method of item 7837, wherein the composition comprises a polymer, and the polymer is, or comprises, an amorphous polymer.
7975. The method of item 7837, wherein the composition further comprises a second pharmaceutically active agent.
7976. The method of item 7837, wherein the composition further comprises an anti-inflammatory agent.
7977. The method of item 7837, wherein the composition further comprises an agent that inhibits infection.
7978. The method of item 7837, wherein the composition further comprises an anthracycline.
7979. The method of item 7837, wherein the composition further comprises doxorubicin.
7980. The method of item 7837 wherein the composition further comprises mitoxantrone.
7981. The method of item 7837 wherein the composition further comprises a fluoropyrimidine.
7982. The method of item 7837, wherein the composition further comprises 5-fluorouracil (5-FU).
7983. The method of item 7837, wherein the composition further comprises a folic acid antagonist. 7984. The method of item 7837, wherein the composition further comprises methotrexate.
7985. The method of item 7837, wherein the composition further comprises a podophylotoxin.
7986. The method of item 7837, wherein the composition further comprises etoposide.
7987. The method of item 7837, wherein the composition further comprises camptothecin.
7988. The method of item 7837, wherein the composition further comprises a hydroxyurea.
7989. The method of item 7837, wherein the composition further comprises a platinum complex.
7990. The method of item 7837, wherein the composition further comprises cisplatin.
7991. The method of item 7837 wherein the composition further comprises an anti-thrombotic agent.
7992. The method of item 7837, wherein the composition further comprises a visualization agent.
7993. The method of item 7837, wherein the composition further comprises a visualization agent, and the visualization agent is a radiopaque material, wherein the radiopaque material comprises a metal, a halogenated compound, or a barium containing compound. 7994. The method of item 7837, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, barium, tantalum, or technetium.
7995. The method of item 7837, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, an MRI responsive material.
7996. The method of item 7837, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, a gadolinium chelate.
7997. The method of item 7837, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, iron, magnesium, manganese, copper, or chromium.
7998. The method of item 7837, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, iron oxide compound.
7999. The method of item 7837, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, a dye, pigment, or colorant.
8000. The method of item 7837 wherein the agent is released in effective concentrations from the composition comprising the agent by diffusion over a period ranging from the time of administration to about 90 days.
8001. The method of item 7837 wherein the agent is released in effective concentrations from the composition comprising the agent by erosion of the composition over a period ranging from the time of administration to about 90 days.
8002. The method of item 7837 wherein the composition further comprises an inflammatory cytokine.
8003. The method of item 7837 wherein the composition further comprises an agent that stimulates cell proliferation.
8004. The method of item 7837 wherein the composition further comprises a polymeric carrier.
8005. The method of item 7837 wherein the composition is in the form of a gel, paste, or spray.
8006. The method of item 7837 wherein the implant is partially constructed with the agent or the composition.
8007. The method of item 7837 wherein the implant is fully constructed with the agent or the composition.
8008. The method of item 7837 wherein the implant is impregnated with the agent or the composition.
8009. The method of item 7837, wherein the agent or the composition forms a coating, and the coating directly contacts the implant.
8010. The method of item 7837, wherein the agent or the composition forms a coating, and the coating indirectly contacts the implant. 8011. The method of item 7837 wherein the agent or the composition forms a coating, and the coating partially covers the implant.
8012. The method of item 7837, wherein the agent or the composition forms a coating, and the coating completely covers the implant.
8013. The method of item 7837 wherein the agent or the composition is located within pores or holes of the implant.
8014. The method of item 7837 wherein the agent or the composition is located within a channel, lumen, or divet of the implant.
8015. The method of item 7837 wherein the implant further comprising an echogenic material.
8016. The method of item 7837 wherein the implant further comprises an echogenic material, wherein the echogenic material is in the form of a coating.
8017. The method of item 7837 wherein the implant is sterile.
8018. The method of item 7837 wherein the agent is delivered from the implant, wherein the agent is released into tissue in the vicinity of the implant after deployment of the implant.
8019. The method of Item 7837 wherein the agent is delivered from the implant, wherein the agent is released into tissue in the vicinity of the implant after deployment of the implant, wherein the tissue is connective tissue. 8020. The method of item 7837 wherein the agent is delivered from the implant, wherein the agent is released into tissue in the vicinity of the implant after deployment of the implant, wherein the tissue is muscle tissue.
8021. The method of item 7837 wherein the agent is delivered from the implant, wherein the agent is released into tissue in the vicinity of the implant after deployment of the implant, wherein the tissue is nerve tissue.
8022. The method of item 7837 wherein the agent is delivered from the implant, wherein the agent is released into tissue in the vicinity of the implant after deployment of the implant, wherein the tissue is epithelium tissue.
8023. The method of item 7837 wherein the agent is delivered from the implant, wherein the agent is released in effective concentrations from the implant over a period ranging from the time of deployment of the implant to about 1 year.
8024. The method of item 7837 wherein the agent is delivered from the implant, wherein the agent is released in effective concentrations from the implant over a period ranging from about 1 month to 6 months.
8025. The method of item 7837 wherein the agent is delivered from the implant, wherein the agent is released in effective concentrations from the implant over a period ranging from about 1 - 90 days.
8026. The method of item 7837 wherein the agent is delivered from the implant, wherein the agent is released in effective concentrations from the implant at a constant rate. 8027. The method of item 7837 wherein the agent is delivered from the implant, wherein the agent is released in effective concentrations from the implant at an increasing rate.
8028. The method of item 7837 wherein the agent is delivered from the implant, wherein the agent is released in effective concentrations from the implant at a decreasing rate.
8029. The method of item 7837 wherein the agent is delivered from the implant, wherein the implant comprises about 0.01 Dg to about 10 Dg of the agent.
8030. The method of item 7837 wherein the agent is delivered from the implant, wherein the implant comprises about 10 Dg to about 10 mg of the agent.
8031. The method of item 7837 wherein the agent is delivered from the implant, wherein the implant comprises about 10 mg to about 250 mg of the agent.
8032. The method of item 7837 wherein the agent is delivered from the implant, wherein the implant comprises about 250 mg to about 1000 mg of the agent.
8033. The method of item 7837 wherein the agent is delivered from the implant, wherein the implant comprises about 1000 mg to about 2500 mg of the agent.
8034. The method of item 7837 wherein the agent is delivered from the implant, wherein a surface of the implant comprises less than 0.01 Dg of the agent per mm2 of implant surface to which the agent is applied. 8035. The method of item 7837 wherein the agent is delivered from the implant, wherein a surface of the implant comprises about 0.01 Dg to about 1 Dg of the agent per mm2 of implant surface to which the agent is applied.
8036. The method of item 7837 wherein the agent is delivered from the implant, wherein a surface of the implant comprises about 1 Dg to about 10 Dg of the agent per mm2 of implant surface to which the agent is applied.
8037. The method of item 7837 wherein the agent is delivered from the implant, wherein a surface of the implant comprises about 10 Dg to about 250 Dg of the agent per mm2 of implant surface to which the agent is applied.
- - 8038. The method of item 7837 wherein the agent is delivered from the implant, wherein a surface of the implant comprises about 250 Dg to about 1000 Dg of the agent per mm2 of implant surface to which the agent is applied.
8039. The method of item 7837 wherein the agent is delivered from the implant, wherein a surface of the implant comprises about 1000 Dg to about 2500 Dg of the agent per mm2 of implant surface to which the agent is applied.
8040. The method of item 7837, wherein the implant further comprises a coating, and the coating is a uniform coating.
8041. The method of item 7837, wherein the implant further comprises a coating, and the coating is a non-uniform coating. 8042. The method of item 7837, wherein the implant further comprises a coating, and the coating is a discontinuous coating.
8043. The method of item 7837, wherein the implant further comprises a coating, and the coating is a patterned coating.
8044. The method of item 7837, wherein the implant further comprises a coating, and the coating has a thickness of 100 Dm or less.
8045. The method of item 7837, wherein the implant further comprises a coating, and the coating has a thickness of 10 Dm or less.
8046. The method of item 7837, wherein the implant further comprises a coating, and the coating adheres to the surface of the implant upon deployment of the implant.
8047. The method of item 7837, wherein the implant further comprises a coating, and the coating is stable at room temperature for a period of at least 1 year.
8048. The method of item 7837, wherein the implant further comprises a coating, and the agent is present in the coating in an amount ranging between about 0.0001% to about 1% by weight.
8049. The method of item 7837, wherein the implant further comprises a coating, and the agent is present in the coating in an amount ranging between about 1 % to about 10% by weight.
8050. The method of item 7837, wherein the implant further comprises a coating, and the agent is present in the coating in an amount ranging between about 10% to about 25% by weight. 8051. The method of item 7837, wherein the implant further comprises a coating, and the agent is present in the coating in an amount ranging between about 25% to about 70% by weight.
8052. The method of item 7837, wherein the implant further comprises a coating, and the coating comprises a polymer.
8053. The method of item 7837, wherein the implant comprises a first coating having a first composition and a second coating having a second composition.
8054. The method of item 7837, wherein the implant comprises a first coating having a first composition and a second coating having a second composition, wherein the first composition and the second composition are different.
8055. A method as in any one of items 7837-8054, wherein the device is an AV fistula graft.
8056. A method as in any one of items 7837-8054, wherein the device is an AV access graft.
8057. A method as in any one of items 7837-8054, wherein the device is a venous catheter.
8058. A method as in any one of items 7837-8054, wherein the device is a vascular graft.
8059. A method as in any one of items 7837-8054, wherein the device is an implantable port. 8060. A method as in any one of items 7837-8054, wherein the device is an AV shunt.
8061. A method for inhibiting scarring comprising placing a device that comprises a medical device comprising a film or a mesh (e.g., an implant) and an anti-scarring agent or a composition comprising an anti- scarring agent into an animal host, wherein the agent inhibits scarring.
8062. The method of item 8061 wherein the agent is an adensosine A2A receptor antagonist.
8063. The method of item 8061 wherein the agent is an AKT inhibitor.
8064. The method of item 8061 wherein the agent is an alpha 2 integrin antagonist, wherein the alpha 2 integrin antagonist is Pharmaprojects No. 5754 (Merck KgaA).
8065. The method of item 8061 wherein the agent is an alpha 4 integrin antagonist.
8066. The method of item 8061 wherein the agent is an alpha 7 nicotinic receptor agonist.
8067. The method of item 8061 wherein the agent is an angiogenesis inhibitor selected from the group consisting of AG-12,958 (Pfizer), ATN-161 (Attenuon LLC), neovastat, an angiogenesis inhibitor from Jerina AG (Germany), NM-3 (Mercian), VGA-1155 (Taisho), FCE-26644 (Pfizer), FCE-26950 (Pfizer), FPMA (Meiji Daries), FR-111142 (Fujisawa), GGTI-298, GM-1306 (Ligand), GPA-1734 (Novartis), NNC-47-0011 (Novo Nordisk), herbamycin (Nippon Kayaku), lenalidomide (Celegene), 1P-10 (NIH), ABT-828 (Abbott), KIN-841 (Tokushima University, Japan), SF-1126 (Semafore Pharmaceuticals), laminin technology (NIH), CHIR-258 (Chiron), NVP-AEW541 (Novartis), NVP-AEW541 (Novartis), Vt16907 (Alchemia), OXI-8007 (Oxigene), EG-3306 (Ark Therapeutics), Maspin (Arriva), ABT- 567 (Abbott), PPI-2458 (Praecis Pharmaceuticals), CC-5079, CC-4089 (Celgene), HIF-1 alpha inhibitors (Xenova), S-247 (Pfizer), AP-23573 (Ariad), AZD-9935 (Astra Zeneca), mebendazole (Introgen Therapeutics), MetAP-2 inhibitors (GlaxoSmithKline), AG-615 (Angiogene Pharmaceuticals), Tie-2 antagonists (Hybrigenics), NC-381 , CYC-381, NC-169, NC-219, NC-383, NC-384, NC-407 (Lorus Therapeutics), ATN-224 (Attenuon), ON-01370 (Onconova), Vitronectin antagonists (Amgen), SDX-103 (Salmedix), Vitronectin antagonists (Shire), CHP (Riemser), TEK (Amgen), Anecortave acetate (Alcon), T46.2 (Matrix Therapeutics), HG-2 (Heptagen), TEM antagonists (Genzyme), Oxi-4500 (Oxigene), ATN-161 (Attenuon), WX-293 (Wilex), M-2025 (Metris Therapeutics), Alphastatin (BioActa), YH-16 (Yantai Rongchang), BIBF-1120 (Boehringer Ingelheim), BAY-57-9352 (Bayer), AS- 1404 (Cancer Research Technology), SC-77964 (Pfizer), glycomimetics (BioTie Therapies), TIE-2 Inhibitors (Ontogen), DIMI, Octamer (Octamer), ABR-215050 (Active Biotech), ABT-518 (Abbott), KDR inhibitors (Abbott), BSF-466895 (Abbott), SCH-221153 (Schering-Plough), DAC:antiangiogenic (ConjuChem), TFPI (EntreMed), AZD-2171 (Astra-Zenaca), CDC-394 (Celgene), LY290293 (EIi Lilly), IDN-5390 (Indena), Kdr Kinase Inhibitors (Merck), CT-113020, CT-116433, CT-116563, CT-31890, CT-32228) (Cell Therapeutics), A-299620 (Abbott), TWEAK Inhibitor (Amgen), VEGF modulators (Johnson and Johnson), Tum-N53, tumstatin (Genzyme), Thios- 1 , Thios-2 (Thios Pharmaceuticals), MV-6401 (Miravant Medical Technologies), Spisulosine (PharmaMar), CEP-7055 (Cephalon), AUV-201 (Auvation), LM-609 (EIi Lilly), SKF-106615 (AnorMED), Oglufanide disodium (Cytran), BW-114 (Pharminox), Calreticulin (NIH), WX-678 (Wilex), SD-7784 (Pfizer), WX-UK1 (Wilex), SH-268 (Schering AG), 2-Me-PGA (Celgene), S- 137 (Pfizer), ZD-6126 (Angiogene Pharmaceuticals), SG-292 (SignalGen), Benefin (Lane Labs), A6, A36 (Angstrom), SB-2723005 (GlaxoSmithKline), SC-7 (Cell Therapeutics), ZEN-014 (AEterna Zentaris), 2-methoxyestradiol (EntreMed), NK-130119 (Nippon Kayaku), CC-10004 (Celgene), AVE- 8062A (Ajinomoto), Tacedinaline (Pfizer), Actinonin (Tokyo Metropolitan Institute of Medical Science), Lenalidomide (Celgene), VGA-1155, BTO-956 (SRI International), ER-68203-00 (Eisai), CT-6685 (UCB), JKC-362 (Phoenix Pharmaceuticals), DMI-3798 (DMI Biosciences, Angiomate (Ipsen), ZD-6474 (AstraZeneca), CEP-5214 (Cephalon), Canstatin (Genzyme), NM-3 (Mercian), Oridigm (MediQuest Therapeutics), Exherin (Adherex), BLS-0597 (Boston Life Sciences), PTC-299 (PTC Therapeutics), NPI-2358 (Nereus Pharmaceuticals), CGP-79787 (Novartis), AEE-788 (Novartis), CKD-732 (Chong Kun Dang), CP-564959 (OSI Pharmaceuticals), CM-101 (CarboMed), CT-2584, CT3501 (Cell Therapeutics), combretastatin and analogues and derivatives thereof (Oxigene), Rebimastat (Bristol-Meyers Squibb), Dextrin 2-sulfate (ML Laboratories), Cilengitide (Merk KGaA), NSC-706704 (Pharminox), KRN- 951 (Kirin Brewery), Ukrain, NSC-631570 (Nowicky Pharma), Tecogalan sodium (Daiichi Pharmaceutical), Tz-93 (Tsumura), TBC-1635 (Encysive Pharmaceuticals), TAN-1120 (Takeda), Semaxanib (Pfizer), BDI-7800 (Biopharmacopae), SD-186, SD-983 (Bristol-Meyers Squibb), SB-223245 (GlaxoSmithKline), SC-236 (Pfizer), RWJ-590973 (Johnson and Johnson), ILX-1850 (Genzyme), SC-68488, S-836 (Pfizer), CG-55069-11 (CuraGen), Ki-23057 (Kirin Brewery), CCX-700 (Chemoentryx), Pegaptanib octasodium (Giled Sciences), ANGIOCOL (available from Biostratum Inc.), or an analogue or derivative thereof.
8068. The method of item 8061 wherein the agent is an apoptosis antagonist.
8069. The method of item 8061 wherein the agent is an apoptosis activator. 8070. The method of item 8061 wherein the agent is a beta 1 integrin antagonist.
8071. The method of item 8061 wherein the agent is a beta tubulin inhibitor.
8072. The method of item 8061 wherein the agent is a blocker of enzyme production in Hepatitis C.
8073. The method of item 8061 wherein the agent is a Bruton's tyrosine kinase inhibitor.
8074. The method of item 8061 wherein the agent is a calcineurin inhibitor.
8075. The method of item 8061 wherein the agent is a caspase 3 inhibitor.
8076. The method of item 8061 wherein the agent is a CC chemokine receptor antagonist.
8077. The method of item 8061 wherein the agent is a cell cycle inhibitor selected from the group consisting of SNS-595 (Sunesis), synthadotin, KRX-0403, and an analogue or derivative thereof.
8078. The method of item 8061 wherein the agent is a cathepsin B inhibitor.
8079. The method of item 8061 wherein the agent is a cathepsin K inhibitor, wherein the cathepsin K inhibitor is 462795 (GlaxoSmithKline), INPL-022-D6 (Amura Therapeutics), or an analogue or derivative thereof.
8080. The method of item 8061 wherein the agent is a cathepsin L inhibitor.
8081. The method of item 8061 wherein the agent is a CD40 antagonist.
8082. The method of item 8061 wherein the agent is a chemokine receptor agonist.
8083. The method of item 8061 wherein the agent is a chymase inhibitor.
8084. The method of item 8061 wherein the agent is a collagenase antagonist.
8085. The method of item 8061 wherein the agent is a CXCR antagonist.
8086. The method of item 8061 wherein the agent is a cyclin dependent kinase inhibitor selected from the group consisting of a CDK-1 inhibitor, a CDK-2 inhibitor, a CDK- 4 inhibitor, a CDK-6 inhibitor, a CAK1 inhibitor from GPC Biotech and Bristol-Myers Squibb, RGB-286199 (GPC Biotech), an anticancer agent from Astex Technology, a CAK1 inhibitor from GPC Biotech, a CDK inhibitor from Sanofi-Aventis, a CDK1/CDK2 inhibitor from Amgen, a CDK2 inhibitor from SUGEN-2 (Pfizer), a hearing loss therapy agent (Sound Pharmaceuticals), PD-0332991 (Pfizer), RGB-286199 (GPC Biotech), Ro-0505124 (Hoffmann-La Roche), a Ser/Thr kinase inhibitor from Lilly (EIi Lilly), CVT-2584 (CAS No. 199986-75-9) (CV Therapeutics), and an analogue or derivative thereof.
8087. The method of item 8061 wherein the agent is a cyclooxygenase 1 inhibitor.
8088. The method of item 8061 wherein the agent is a DHFR inhibitor.
8089. The method of item 8061 wherein the agent is a dual integrin inhibitor.
8090. The method of item 8061 wherein the agent is an elastase inhibitor.
8091. The method of item 8061 -wherein the~agent is an elongation factor-1 alpha inhibitor.
8092. The method of item 8061 wherein the agent is an endothelial growth factor antagonist.
8093. The method of item 8061 wherein the agent is an endothelial growth factor receptor kinase inhibitor selected from the group consisting of sorafenib tosylate (Bayer), AAL-993 (Novartis), ABP-309 (Novartis), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), E- 7080 (Eisal), EG-3306 (Ark Therapeutics), EXEL-2880 (Exelixis), GW- 654652 (GlaxoSmithKline), a KDR inhibitor from LG Life Sciences, CT-6685 and CT-6729 (UCB), KRN-633 and KRN-951 (Kirin Brewery), OSI-930 (OSI Pharmaceuticals), SP-5.2 (Supratek Pharma), SU-11657 (Pfizer), a Tie-2 antagonist (Hybrigenics), a VEGFR-2 kinase inhibitor (Bristol-Myers Squibb), XL-647 (Exelixis), a KDR inhibitor from Abbott Laboratories, sorafenib tosylate, and an analogue or derivative thereof.
8094. The method of item 8061 wherein the agent is an endotoxin antagonist.
8095. The method of item 8061 wherein the agent is an epothilone and tubulin binder.
8096. The method of item 8061 wherein the agent is an estrogen receptor antagonist.
8097. The method of item 8061 wherein the agent is an FGF inhibitor.
8098. The method of item 8061 wherein the agent is a farnexyl transferase inhibitor.
8099. The method of item 8061 wherein the agent is farnesyltransferase inhibitor selected from the group of A-197574 (Abbott), a famesyltransferase inhibitor from Servier, FPTIII (Strathclyde Institute for Drug R), LB-42908 (LG Life Sciences), Pharmaprojects No. 5063 (Genzyme), Pharmaprojects No. 5597 (Ipsen), Yissum Project No. B-1055 (Yissum), and an analogue or derivative thereof.
8100. The method of item 8061 wherein the agent is an FLT-3 kinase inhibitor.
8101. The method of item 8061 wherein the agent is an FGF receptor kinase inhibitor. 8102. The method of item 8061 wherein the agent is a fibrinogen antagonist selected from the group consisting of AUV-201 (Auvation), MG-13926 (Sanofi-Aventis), plasminogen activator (CAS No. 105913-11-9) (from Sanofi-Aventis or UCB), plasminogen activator-2 (tPA- 2) (Sanofi-Aventis), pro-urokinase (CAS No. 82657-92-9) (Sanofi-Aventis), and an analogue or derivative thereof.
8103. The method of item 8061 wherein the agent is a heat shock protein 90 antagonist selected from the group consisting of SRN-005 (Sirenade), geldanamycin, NSC-33050 (17-allylaminogeldanamycin; 17- AAG), 17-dimethylaminoethylamino-17-demethoxy-geldanamycin (17- DMAG), rifabutin (rifamycin XIV, 1',4-didehydro-1-deoxy-1 ,4-dihydro-5'-(2- methylpropyl)-1-oxo-), and an analogue or derivative thereof.
8104. The method of item 8061 wherein the agent is a histone deacetylase inhibitor. -
8105. The method of item 8061 wherein the agent is an HMGCoA reductase inhibitor selected from the group consisting of an atherosclerosis therapeutic from Lipid Sciences, ATI-16000 (ARYx Therapeutics), KS-01-019 (Kos Pharmaceuticals), Pharmaprojects No. 2197 (Sanofi-Aventi), RP 61969 (Sanofi-Aventis), and an analogue or derivative thereof.
8106. The method of item 8061 wherein the agent is an ICAM inhibitor.
8107. The method of item 8061 wherein the agent is an IL, ICE and IRAK antagonist, wherein the antagonist is a CJ-14877, CP-424174 (Pfizer), NF-61 (Negma-Lerads), and an analogue or derivative thereof. 8108. The method of item 8061 wherein the agent is an IL-2 inhibitor.
8109. The method of item 8061 wherein the agent is an immunosuppressant selected from the group consisting of teriflunomide (Sanofi Aventis), chlorsulfaquinoxalone (NSC-339004), chlorsulfaquinoxalone sulfate, CS-712 (Sankyo), ismomultin alfa (CAS No. 457913-93-8) (Akzo Nobel), antiallergics from GenPat77, antiinflammatories or AT-005 (Androclus Therapeutics), autoimmune disease therapeutics from EpiVax, BN-007 (Bone), budesonide (CAS No. 51333-22- 3) (MAP Pharmaceuticals), CO-14 (Genzyme), edratide (CAS No. 433922- 67-9) (Teva), EP-314 (Enanta), eprovafen (CAS No. 101335-99-3) (Sanofi- Aventis), HWA-131 (CAS No. 118788-41-3) (Sanofi-Aventis), immunomodulators from MerLion Pharmaceuticals, immunosuppressives from Alchemia, IPL-12 (Inflazyme), MDL-9563 (CAS No. 27086-86-8) (Sanofi-Aventis), Pharmaprojects No. 2330 (Sanofi-Aventis), Pharmaprojects No. 6426 (Abgenix), PXS-25 (Pharmaxis), rosmarinic acid (CAS No. 20283-92-5) (Sanofi-Aventis), RP 42927 or RP 54745 (CAS No. 135330-08-4) (Sanofi-Aventis), SGN-35 (Seattle Genetics), ST-1959 (Sigma-Tau), type I diabetes therapy from SYNX Pharma, UNIL-88 (Debiopharm), VP-025 (Vasogen), VR-694 (Vectura), PRTX-001 (Protalex), and an analogue or derivative thereof.
8110. The method of item 8061 wherein the agent is an IMPDH (inosine monophosphate).
8111. The method of item 8061 wherein the agent is an integrin antagonist.
8112. The method of item 8061 wherein the agent is an interleukin antagonist. 8113. The method of item 8061 wherein the agent is an inhibitor of type III receptor tyrosine kinase.
8114. The method of item 8061 wherein the agent is an irreversible inhibitor of enzyme methionine aminopeptidase type 2.
8115. The method of item 8061 wherein the agent is an isozyme selective delta protein kinase C inhibitor.
8116. The method of item 8061 wherein the agent a JAK3 enzyme inhibitor.
8117. The method of item 8061 wherein the agent is a JNK inhibitor.
8118. The method of-item 8061 wherein the agent is a kinase inhibitor.
8119. The method of item 8061 wherein the agent is kinesin antagonist.
8120. The method of item 8061 wherein the agent is a kinesin antagonist.
8121. The method of item 8061 wherein the agent is a leukotriene inhibitor and antagonist selected from the group consisting of ambicromil (CAS No. 58805-38-2) (Sanofi-Aventis), amelubant (CAS No. 346735-24-8) (Boehringer Ingelheim), DW-1141 (Dong Wha), ebselen (Daiichi Pharmaceutical), ibudilast (Kyorin), leucotriene inhibitors from Sanofi-Aventis, lymphotoxin-beta receptor (LT-D) from Biogen Idee, Pharmaprojects No. 1535 and 2728 (CAS No. 119340-33-9) (Sanofi- Aventis), R-112 (Rigel), Rev-5367 (CAS No. 92532-05-3) (Sanofi-Aventis), RG-14893 (CAS No. 141835-49-6) (Sanofi-Aventis), RG-5901-A (CAS No. 101910-24-1), 92532-23-5, RP 66153 (CAS No. 142422-79-5), RP 66364 (CAS No.186916-92-5), RP 69698 (CAS No. 141748-00-7) (Sanofi-Aventis), SC-411930 (Pfizer), SC-41930 (CAS No. 120072-59-5) (Pfizer), SC-50605 (CAS No. 138828-39-4) (Pfizer), SC-51146 (CAS No. 141059-52-1), SC- 53228 (CAS No. 153633-01-3) (Pfizer), spaglumic acid (ZY-15106) (CAS No.3106-85-2), 80619-64-3 (Novartis), tipredane (CAS No. 85197-77-9) (Bristol-Myers Squibb), U-75302 (CAS No. 119477-85-9) (Pfizer), and analogue or derivative thereof.
8122. The method of item 8061 wherein the agent is an MAP kinase inhibitor.
8123. The method of item 8061 wherein the agent is a matrix metalloproteinase inhibitor.
8124. The method of item 8061 wherein the agent is an MCP- CCR2 inhibitor.
8125. The method of item 8061 wherein the agent is an mTOR inhibitor.
8126. The method of item 8061 wherein the agent is an mTOR kinase inhibitor.
8127. The method of item 8061 wherein the agent is a microtubule inhibitor selected from the group consisting of antibody- maytansinoid conjugates from Biogen Idee, colchicines (MantiCore Pharmaceuticals), anticancer immunoconjugates from Johnson & Johnson, DIME from Octamer, gni-1f (GNI), huC242-DM4, huMy9-6-DM1 (ImmunoGen), IDN-5404 (Indena), IMO-098, IMOderm (Imotep), mebendazole (Introgen Therapeutics), microtubule poisons from Cambridge Enterprise, paclitaxel such as LOTAX from Aphios (CAS No. 33069-62-4), Genexol-PM from Samyang, Pharmaprojects No. 6383 (Tapestry Pharmaceuticals), RPR-112378 (Sanofi-Aventis), SGN-75 (Seattle Genetics), SPL-7435 (Starpharma), SSR-250411 (Sanofi-Aventis), trastuzumab-DM1 (Genentech), vinorelbine, and an analogue or derivative thereof.
8128. The method of item 8061 wherein the agent is an MIF inhibitor.
8129. The method of item 8061 wherein the agent is an MMP inhibitor.
8130. The method of item 8061 wherein the agent is a neurokinin (NK) antagonist selected from the group consisting of anthrotainin (CAS No. 148084-40-6) (Sanofi-Aventis), an IBS thereapeutic from ArQuIe, MDL-105212A (CAS No. 167261-60-1) (Ssanofi-Aventis), Pharmaprojects No. 2744, 3258 (CAS No. 139167-47-8) 4006, 4201, or 5986 (Sanofi-Aventis), RP 67580 (CAS No. 135911-02-3), SR-144190 (CAS No. 201152-86-5), SSR-240600, SSR-241586 (Sanofi-Aventis), TKA-457 (Novartis), vestipitant mesylate (CAS No. 334476-64-1) (GlaxoSmithKline), Win-64821 (Sanofi-Aventis), PRX-96026 (Predix Pharmaceuticals), and an analogue or derivative thereof.
8131. The method of item 8061 wherein the agent is an NF kappa B inhibitor selected from the group consisting of AVE-0545 or AVE- 0547 (Sanofi-Aventis), bortezomib (CAS No. 179324-69-7) (Millennium Pharmaceuticals), dexanabinol (CAS No. 112924-45-5) (Pharmos), dexlipotam (Viatris), Pharmaprojects No. 6283 (INDRA) (OXiGENE), IPL- 576092 (CAS No. 137571-30-3) (Inflazyme), NFKB decoy (Corgentech), NFKB decoy oligo (AnGes MG), NFKB's from Ariad, osteoporosis treatments or S5 (F005) from Fulcrum Pharmaceuticals, P61 (Phytopharm), R-flurbiprofen (CAS No. 5104-49-4) (Encore Pharmaceuticals), and an analogue or derivative thereof.
8132. The method of item 8061 wherein the agent is a nitric oxide agonist.
8133. The method of item 8061 wherein the agent is an ornithine decarboxylase inhibitor.
8134. The method of item 8061 wherein the agent is a p38 MAP kinase inhibitor selected from the group consisting of AZD-6703 (AstraZeneca), JX-401 (Jexys Pharmaceuticals), BMS-2 (Bristol-Myers Squibb), a p38 MAP kinase inhibitor from Novartis, a p38-alpha MAP kinase- inhibitor from Amphora, Pharmaprojects No. 5704 (Pharmacopeia), RPR- 200765A (Sanofi-Aventis), SD-282 (Johnson & Johnson), TAK-715 (Takeda), and an analogue or derivative thereof.
8135. The method of item 8061 wherein the agent is a palmitoyl-protein thioesterase inhibitor.
8136. The method of item 8061 wherein the agent is a PDGF receptor kinase inhibitor selected from the group consisting of AAL-993, AMN-107, or ABP-309 (Novartis), AMG-706 (Amgen), BAY-57-9352 (Bayer), CDP-860 (UCB), E-7080 (Eisai), imatinib (CAS No. 152459-95-5) (Novartis), OSI-930 (OSI Pharmaceuticals), RPR-127963E (Sanofi-Aventis), RWJ-540973 (Johnson & Johnson), sorafenib tosylate (Bayer), SU-11657 (Pfizer), tandutinib (CAS No. 387867-13-2) (Millennium Pharmaceuticals), vatalanib (Novartis), ZK-CDK (Schering AG), and an analogue or derivative thereof.
8137. The method of item 8061 wherein the agent is (-)- halofenate (Metabolex), AMG-131 (Amgen), antidiabetics from Japan Tobacco, AZD-4619, AZD-8450, AZD-8677 (AstraZeneca), DRF-10945, balaglitazone (Dr Reddy's), CS-00088, CS-00098 (Chipscreen Biosciences), E-3030 (Eisai), etalocib (CAS No. 161172-51-6) (Eli Lilly), GSK-641597 (Ligand), GSK-677954 (GlaxoSmithKline), GW-409544 (Ligand), GW- 590735 (GlaxoSmithKline), K-111 (Hoffmann-La Roche), LY-518674 (EIi Lilly), LY-674 (Ligand), LY-929 (Ligand), MC-3001 , MC-3002 (MaxoCore Pharmaceuticals), metformin HCI + pioglitazone (CAS No. 1115-70-4 and
112529-15-4), ACTOPLUS MET from Andrx), muraglitazar (CAS No. 331741-94-7) (Bristol-Myers Squibb), naveglitazar (Ligand), oleoylethanolamide (Kadmus Pharmaceuticals), ONO-5129, pioglitazone hydrochloride (CAS No. 111025-46-8 and -1-12529-15-4) (Takeda), PLX-204 (Plexxikon), PPAR agonists from Genfit, PPAR delta agonists from EIi Lilly, PPAR-alpha agonists from CrystalGenomics, PPAR-gamma modulators and PPAR-β modulators from CareX, rosiglitazone maleate (CAS No. 122324- 73-4 or 155141-29-0) (GlaxoSmithKline), rosiglitazone maleate/glimepir (CAS No. 155141-29-0 and 93479-97-1), AVANDARYL, rosiglitazone maleate/metformin extend (CAS No. 155141-29-0 and 657-24-9), AVANDAMET, rosiglitazone maleate+metformin, AVANDAMET (GlaxoSmithKline), tesaglitazar (AstraZeneca), and an analogue or derivative thereof.
8138. The method of item 8061 wherein the agent is a phosphatase inhibitor.
8139. The method of item 8061 wherein the agent is a phosphodiesterase (PDE) inhibitor selected from the group consisting of avanafil (Tanabe Seiyaku), dasantafil (CAS No. 569351-91-3) (Schering- Plough), A-906119 (CAS No. 134072-58-5), DL-850 (Sanofi-Aventis), GRC- 3015, GRC-3566, GRC-3886 (Glenmark), HWA-153 (CAS No. 56395-66-5) (Sanofi-Aventis), hydroxypumafentrine (Altana), IBFB-130011 , IBFB-14-016, IBFB-140301 , IBFB-150007, IBFB-211913 (IBFB Pharma), L-826141 (Merck & Co), medorinone (CAS No. 88296-61-1) (Sanofi-Aventis), MEM- 1917 (Memory Pharmaceuticals), ND-1251 (Neuro3d), PDE inhibitors from ICOS, PDE IV inhibitors from Memory Pharmaceuticals and CrystalGenomics, Pharmaprojects No. 2742 and 6141 (Sanofi-Aventis), QAD-171 (Novartis), RHC-2963 (CAS No. 76993-12-9 and 76993-14-1), RPR-117658, RPR-122818 derivatives, SR-24870 , and RPR-132294 (Sanofi-Aventis), SK-350 (ln2Gen), stroke therapy agents from deCODE Genetics, TAS-203 (Taiho), tofimilast (CAS No. 185954-27-2) (Pfizer), UK- 371800 (Pfizer), WIN-65579 (CAS No. 158020-82-7) (Sanofi-Aventis), IBFB- 130020 (IBFB Pharma), OPC-6535 (CAS No. 145739-56-6) (Otsuka), a phosphodiesterase III inhibitor, enoximone, a phosphodiesterase IV inhibitor, fosfosal, Atopik (Barrier Therapeutics), triflusal, a phosphodiesterase V inhibitor, and an analogue or derivative thereof.
8140. The method of item 8061 wherein the agent is a PKC inhibitor.
8141. The method of item 8061 wherein the agent is a platelet activating factor antagonist.
8142. The method of item 8061 wherein the agent is a platelet-derived growth factor receptor kinase inhibitor.
8143. The method of item 8061 wherein the agent is a prolyl hydroxylase inhibitor. 8144. The method of item 8061 wherein the agent is a polymorphonuclear neutrophil inhibitor.
8145. The method of item 8061 wherein the agent is a protein kinase B inhibitor.
8146. The method of item 8061 wherein the agent is a protein kinase C stimulant.
8147. The method of item 8061 wherein the agent is a purine nucleoside analogue.
8148. The method of item 8061 wherein the agent is a purinoreceptor P2X antagonist.
8149. The method of item 8061 wherein the agent is a Raf kinase inhibitor.
8150. The method of item 8061 wherein the agent is a reversible inhibitor of ErbB1 and ErbB2.
8151. The method of item 8061 wherein the agent is a ribonucleoside triphosphate reductase inhibitor.
8152. The method of item 8061 wherein the agent is an SDF- 1 antagonist.
8153. The method of item 8061 wherein the agent is a sheddase inhibitor. 8154. The method of item 8061 wherein the agent is an SRC inhibitor.
8155. The method of item 8061 wherein the agent is a stromelysin inhibitor.
8156. The method of item 8061 wherein the agent is an Syk kinase inhibitor.
8157. The method of item 8061 wherein the agent is a telomerase inhibitor.
8158. The method of item 8061 wherein the agent is a TGF beta inhibitor selected from the group consisting of pirfenidone (CAS No. 53179-13-8) (MARNAC), tranilast (CAS No. 53902-12-8) (Kissei), IN-1130 (ln2Gen), mannose-6-phosphate (BTG), TGF-β antagonists from Inflazyme (Pharmaprojects No. 6075), TGF-β antagonists from Sydney, non-industrial source), TGF-β I receptor kinase inhibitors from EIi Lilly, TGF-β receptor inhibitors from Johnson & Johnson, and an analogue or derivative thereof.
8159. The method of item 8061 wherein the agent is a TNFα antagonist or TACE inhibitor selected from the group consisting of adalimumab (CAS No. 331731-18-1) (Cambridge Antibody Technology), AGIX-4207 (AtheroGenics), AGT-1 (Advanced Biotherapy), an antiinflammatory from Borean Pharma, Cellzome, or Paradigm Therapeutics, anti-inflammatory vaccine (TNF-alpha kinoid) from Neovacs, humanized anti-TNF antibody or an anti-TNF MAb (CB0006) Celltech (UCB), apratastat (CAS No. 287445-51-0) (Wyeth), BMS-561392 (Bristol-Myers Squibb), BN- 006 (Bone), certolizumab pegol (CAS No. 428863-50-7 or CH-138 (UCB), cilomilast (CAS No. 153259-65-5) (GlaxoSmithKline), CR-1 (Nuada Pharmaceuticals), CRx-119 (CombinatoRx), D-5410 (UCB), dacopafant (CAS No. 125372-33-0) (Sanofi-Aventis), dersalazine (CAS No. 188913-57- 7/188913-58-8) (Uriach), etanercept (CAS No. 185243-69-0) (Amgen), ethyl pyruvate (Critical (Critical Therapeutics), golimumab (CAS No. 476181-74-5) (Johnson & Johnson), hormono-immunotherapy from Ipsen, CDP571 (e.g., Humicade from UCB), IC-485 (ICOS), infliximab (CAS No. 170277-31-3) (Johnson & Johnson), IP-751 (Manhattan Pharmaceuticals), ISIS-104838 (CAS No. 250755-32-9) (ISIS Pharmaceuticals), lenalidomide (CAS No. 191732-72-6) (Celgene), lentinan (CAS No. 37339-90-5) (Ajinomoto), MDL- 201112 (CAS No. 142130-73-2) (Sanofi-Aventis), medroxyprogesterone (CAS No. 520-85-4) (InKine Pharmaceutical), N-acetylcysteine (CAS No. 616-91-1) (Zambon), NBE-P2 (DIREVO Biotech), nerelimomab (CAS No. 162774-06-3) (Chiron), OM-294DP (OM PHARMA), onercept (CAS No. 199685-57-9) (Yeda), PASSTNF-alpha (Verigen), pentoxifylline or oxypentifylline (Sanofi-Aventis), Pharmaprojects No. 4091 , 4241 , 4295, or 4488 (Sanofi-Aventis), Pharmaprojects No. 5480 (Amgen), Pharmaprojects No. 6749 (Cengent), pirfenidone (CAS No. 53179-13-8) (MARNAC), RPR- 132294 (Sanofi-Aventis), S5 (F002) (Fulcrum Pharmaceuticals), simvastatin (CAS No. 79902-63-9) (Merck & Co), STA-6292 (Synta Pharmaceuticals), tacrolimus (CAS No. 104987-11-3) (Fujisawa LifeCycle Pharma), talactoferrin alfa (CAS No. 308240-58-6) (Agennix), thalidomide (CAS No. 50-35-1) (Celgene), TNF antagonists form ProStrakan, and Synergen, TNF inhibitors (Amgen), TNF-alpha antagonists from Dynavax Technologies and Jerina AG (Germany), TNF-alpha inhibitors from IBFB Pharma and Xencor (Xencor), torbafylline (CAS No. 105102-21-4) (Sanofi-Aventis), UR-1505 (Uriach), VT-346 (Viron Therapeutics), YSIL6 (Y1S Therapeutics), YSTH2 (Y's Therapeutics), NPI-1302a-3 (Nereus Pharmaceuticals, a TNF antagonist from Jerina AG (Germany), dersalazine, and an analogue or derivative thereof.
8160. The method of item 8061 wherein the agent is a Toll receptor inhibitor. 8161. The method of item 8061 wherein the agent is a tubulin antagonist.
8162. The method of item 8061 wherein the agent is a tyrosine kinase inhibitor selected from the group consisting of SU-011248, SUTENT from Pfizer Inc. (New York, NY), BMS-354825, PN-355 (Paracelsian Pharmaceuticals), AGN-199659 (Allergan), AAL-993 or ABP- 309 (Novartis), adaphostin (NIH)1 AEE-788 (Novartis), AG-013736 (OSI Pharmaceuticals), AG-13736 (Pfizer), ALT-110 (Alteris Therapeutics), AMG- 706 (Amgen), anticancer MAbs from Xencor, anti-EGFrvlll MAbs from Abgenix, anti-HER2 MAb from Abiogen, AZD-2171 or AZD-9935 (AstraZeneca), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), CEP-5214 (Cephalon), CEP-7055 (Cephalon), cetuximab (ImClone Systems), CHIR-200131 and CHIR-258 (Chiron), CP-547632 (OSI Pharmaceuticals), CP-724714 (Pfizer), CT-301 (Creabilis Therapeutics), D- 69491 (Baxter International), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EGFR/ErbB2 inhibitors from Array BioPharma, erlotinib (CAS No. 183319- 69-9) (OSI Pharmaceuticals), EXEL-2880 (Exelixis), FK-778 (Sanofi- Aventis) , gefitinib (CAS No. 184475-35-2) (AstraZeneca), GW-2286 or GW- 654652 (GlaxoSmithKline), her2/neu antigen from AlphaVax, HER-2/neu inhibitor from Generex, Herzyme (Medipad) (Sirna Therapeutics), HKI-272 (Wyeth), HuMax-EGFr (Genmab), idronoxil (CAS No. 81267-65-4) (Novogen), IGF-1 inhibitors from Ontogen, IMC-11 F8 (ImClone Systems), kahalalide F (CAS No. 149204-42-2) (PharmaMar), KDR inhibitor from LG Life Sciences, KDR inhibitors from Abbott Laboratories, KDR kinase inhibitors (UCB), Kdr kinase inhibitors from Merck & Co, KRN-633 and KRN- 951 (Kirin Brewery), KSB-102 (Xenova), lapatinib ditosylate (CAS No. 388082-78-8) (GlaxoSmithKline), matuzumab (Merck KGaA), MDX-214 (Medarex), ME-103 (Pharmexa), MED-A300 (Gerolymatos), MNAC-13 (Lay Line Genomics), nimotuzumab (Center of Molecular Immunology), NSC- 330507 or NSC-707545 (NIH), NV-50 (Novogen), OSI-930 (OSI Pharmaceuticals), panitumumab (Abgenix), pelitinib (CAS No. 287933-82-7) (Wyeth), pertuzumab (CAS No. 380610-27-5) (Genentech), Pharmaprojects No. 3985 (Sanofi-Aventis), prostate cancer therapeutics from Sequenom (SQPC35, SQPC36, SQPC90), removab and remoxab (Trion Pharma), RG- 13022 (CAS No. 136831-48-6), RG-13291 (CAS No. 138989-50-1), or RG- 14620 (CAS No. 136831-49-7) (Sanofi-Aventis), RM-6427 (Romark), RNAi breast cancer therapy from Benitec, RP 53801 (CAS No. 125882-88-4) (Sanofi-Aventis), sorafenib tosylate (Bayer), SU-11657 (Pfizer), Tie-2 antagonists from Semaia (Hybrigenics), Tie-2 inhibitors from Ontogen, trastuzumab (CAS No. 180288-69-1) (Genentech), tyrosine kinase inhibitors from Sanofi-Aventis, U3-1287, U3-1565, U3-1784, U3-1800 (U3 Pharma), vatalanib (Novartis), VEGFR-2 kinase inhibitor from Bristol-Myers Squibb, XL-647 (Exelixis), ZD-6474 (AstraZeneca), ZK-CDK (Schering AG), an EGFR tyrosine kinase inhibitor, EKB-569 (Wyeth), and an analogue or derivative thereof.
8163. The method of item 8061 wherein the agent is a VEGF inhibitor.
8164. The method of item 8061 wherein the agent is a vitamin D receptor agonist.
8165. The method of item 8061 wherein the agent is ZD-6474 (an angiogenesis inhibitor).
8166. The method of item 8061 wherein the agent is AP- 23573 (an mTOR inhibitor).
8167. The method of item 8061 wherein the agent is synthadotin (a tubulin antagonist). 8168. The method of item 8061 wherein the agent is S-0885 (a collagenase inhibitor).
8169. The method of item 8061 wherein the agent is aplidine (an elongation factor-1 alpha inhibitor).
8170. The method of item 8061 wherein the agent is ixabepilone (an epithilone).
8171. The method of item 8061 wherein the agent is IDN- 5390 (an angiogenesis inhibitor).
8172. The method of item 8061 wherein the agent is SB- 2723005 (an angiogenesis inhibitor).
8173. The method of item 8061 wherein the agent is ABT-518 (an angiogenesis inhibitor).
8174. The method of item 8061 wherein the agent is combretastatin (an angiogenesis inhibitor).
8175. The method of item 8061 wherein the agent is anecortave acetate (an angiogenesis inhibitor).
8176. The method of item 8061 wherein the agent is SB- 715992 (a kinesin antagonist).
8177. The method of item 8061 wherein the agent is temsirolimus (an mTOR inhibitor). 8178. The method of item 8061 wherein the agent is adalimumab (a TNFα antagonist).
8179. The method of item 8061 , wherein the composition comprises a polymer.
8180. The method of item 8061 , wherein the composition comprises a polymer, and the polymer is, or comprises, a copolymer.
8181. The method of item 8061 , wherein the composition comprises a polymer, and the polymer is, or comprises, a block copolymer.
8182. The method of item 8061 , wherein the composition comprises a polymer, and the polymer is, or comprises, a random copolymer.
8183. The method of item 8061 , wherein the composition comprises a polymer, and the polymer is, or comprises, a biodegradable polymer.
8184. The method of item 8061 , wherein the composition comprises a polymer, and the polymer is, or comprises, a nonbiodegradable polymer.
8185. The method of item 8061 , wherein the composition comprises a polymer, and the polymer is, or comprises, a hydrophilic polymer.
8186. The method of item 8061 , wherein the composition comprises a polymer, and the polymer is, or comprises, a hydrophobic polymer. 8187. The method of item 8061 , wherein the composition comprises a polymer, and the polymer is, or comprises, a polymer having hydrophilic domains.
8188. The method of item 8061 , wherein the composition comprises a polymer, and the polymer is, or comprises, a polymer having hydrophobic domains.
8189. The method of item 8061 , wherein the composition comprises a polymer, and the polymer is, or comprises, a non-conductive polymer.
8190. The method of item 8061 , wherein the composition comprises a polymer, and the polymer is, or comprises, an elastomer.
8191. The method of item 8061 , wherein the composition comprises a polymer, and the polymer is, or comprises, a hydrogel.
8192. The method of item 8061 , wherein the composition comprises a polymer, and the polymer is, or comprises, a silicone polymer.
8193. The method of item 8061 , wherein the composition comprises a polymer, and the polymer is, or comprises, a hydrocarbon polymer.
8194. The method of item 8061 , wherein the composition comprises a polymer, and the polymer is, or comprises, a styrene-derived polymer. 8195. The method of item 8061 , wherein the composition comprises a polymer, and the polymer is, or comprises, a butadiene-derived polymer.
8196. The method of item 8061 , wherein the composition comprises a polymer, and the polymer is, or comprises, a macromer.
8197. The method of item 8061 , wherein the composition comprises a polymer, and the polymer is, or comprises, a poly(ethylene glycol) polymer.
8198. The method of item 8061 , wherein the composition comprises a polymer, and the polymer is, or comprises, an amorphous polymer.
8199. The method of item 8061, wherein the composition further comprises a second pharmaceutically active agent.
8200. The method of item 8061 , wherein the composition further comprises an anti-inflammatory agent.
8201. The method of item 8061 , wherein the composition further comprises an agent that inhibits infection.
8202. The method of item 8061 , wherein the composition further comprises an anthracycline.
8203. The method of item 8061 , wherein the composition further comprises doxorubicin. 8204. The method of item 8061 wherein the composition further comprises mitoxantrone.
8205. The method of item 8061 wherein the composition further comprises a fluoropyrimidine.
8206. The method of item 8061 , wherein the composition further comprises 5-fluorouracil (5-FU).
8207. The method of item 8061 , wherein the composition further comprises a folic acid antagonist.
8208. The method of item 8061, wherein the composition further comprises methotrexate.
8209. The method of item 8061 , wherein the composition further comprises a podophylotoxin.
8210. The method of item 8061 , wherein the composition further comprises etoposide.
8211. The method of item 8061 , wherein the composition further comprises camptothecin.
8212. The method of item 8061 , wherein the composition further comprises a hydroxyurea.
8213. The method of item 8061 , wherein the composition further comprises a platinum complex. 8214. The method of item 8061 , wherein the composition further comprises cisplatin.
8215. The method of item 8061 wherein the composition further comprises an anti-thrombotic agent.
8216. The method of item 8061 , wherein the composition further comprises a visualization agent.
8217. The method of item 8061 , wherein the composition further comprises a visualization agent, and the visualization agent is a radiopaque material, wherein the radiopaque material comprises a metal, a halogenated compound, or a barium containing compound.
8218. The method of item 8061 , wherein the composition further comprises a visualization agent, and the visualization-agent is, or comprises, barium, tantalum, or technetium.
8219. The method of item 8061 , wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, an MRI responsive material.
8220. The method of item 8061 , wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, a gadolinium chelate.
8221. The method of item 8061 , wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, iron, magnesium, manganese, copper, or chromium. 8222. The method of item 8061 , wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, iron oxide compound.
8223. The method of item 8061 , wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, a dye, pigment, or colorant.
8224. The method of item 8061 wherein the agent is released in effective concentrations from the composition comprising the agent by diffusion over a period ranging from the time of administration to about 90 days.
8225. The method of item 8061 wherein the agent is released in effective concentrations from the composition comprising the agent by erosion of the composition over a period ranging from the time of administration to about 90 days.
8226. The method of item 8061 wherein the composition further comprises an inflammatory cytokine.
8227. The method of item 8061 wherein the composition further comprises an agent that stimulates cell proliferation.
8228. The method of item 8061 wherein the composition further comprises a polymeric carrier.
8229. The method of item 8061 wherein the composition is in the form of a gel, paste, or spray. 8230. The method of item 8061 wherein the implant is partially constructed with the agent or the composition.
8231. The method of item 8061 wherein the implant is fully constructed with the agent or the composition.
8232. The method of item 8061 wherein the implant is impregnated with the agent or the composition.
8233. The method of item 8061 , wherein the agent or the composition forms a coating, and the coating directly contacts the implant.
8234. The method of item 8061, wherein the agent or the composition forms a coating, and the coating indirectly contacts the implant.
8235. The method of item 8061 wherein the agent or the composition forms a coating, and the coating partially covers the implant.
8236. The method of item 8061 , wherein the agent or the composition forms a coating, and the coating completely covers the implant.
8237. The method of item 8061 wherein the agent or the composition is located within pores or holes of the implant.
8238. The method of item 8061 wherein the agent or the composition is located within a channel, lumen, or divet of the implant.
8239. The method of item 8061 wherein the implant further comprising an echogenic material. 8240. The method of item 8061 wherein the implant further comprises an echogenic material, wherein the echogenic material is in the form of a coating.
8241. The method of item 8061 wherein the implant is sterile.
8242. The method of item 8061 wherein the agent is delivered from the implant, wherein the agent is released into tissue in the vicinity of the implant after deployment of the implant.
8243. The method of item 8061 wherein the agent is delivered from the implant, wherein the agent is released into tissue in the vicinity of the implant after deployment of the implant, wherein the tissue is connective tissue.
8244. The method of item 8061 wherein the agent is delivered from the implant, wherein the agent is released into tissue in the vicinity of the implant after deployment of the implant, wherein the tissue is muscle tissue.
8245. The method of iterτκ8061 wherein the agent is delivered from the implant, wherein the agent is released into tissue in the vicinity of the implant after deployment of the implant, wherein the tissue is nerve tissue.
8246. The method of item 8061 wherein the agent is delivered from the implant, wherein the agent is released into tissue in the vicinity of the implant after deployment of the implant, wherein the tissue is epithelium tissue. 8247. The method of item 8061 wherein the agent is delivered from the implant, wherein the agent is released in effective concentrations from the implant over a period ranging from the time of deployment of the implant to about 1 year.
8248. The method of item 8061 wherein the agent is delivered from the implant, wfierein the agent is released in effective concentrations from the implant over a period ranging from about 1 month to 6 months.
8249. The method of item 8061 wherein the agent is delivered from the implant, wherein the agent is released in effective concentrations from the implant over a period ranging from about 1 - 90 days.
8250. The method of item 8061 wherein the agent is delivered from the implant, wherein the agent is released in effective concentrations from the implant at a constant rate. . . . _ . .
8251. The method of item 8061 wherein the agent is delivered from the implant, wherein the agent is released in effective concentrations from the implant at an increasing rate.
8252. The method of item 8061 wherein the agent is delivered from the implant, wherein the agent is released in effective concentrations from the implant at a decreasing rate.
8253. The method of item 8061 wherein the agent is delivered from the implant, wherein the implant comprises about 0.01 Dg to about 10 Dg of the agent. 8254. The method of item 8061 wherein the agent is delivered from the implant, wherein the implant comprises about 10 Og to about 10 mg of the agent.
8255. The method of item 8061 wherein the agent is delivered from the implant, wherein the implant comprises about 10 mg to about 250 mg of the agent.
8256. The method of item 8061 wherein the agent is delivered from the implant, wherein the implant comprises about 250 mg to about 1000 mg of the agent.
8257. The method of item 8061 wherein the agent is delivered from the implant, wherein the implant comprises about 1000 mg to about 2500 mg of the agent.
8258. The method of item 8061 wherein the agent is delivered from the implant, wherein a surface of the implant comprises less than 0.01 Dg of the agent per mm2 of implant surface to which the agent is applied.
8259. The method of item 8061 wherein the agent is delivered from the implant, wherein a surface of the implant comprises about 0.01 Dg to about 1 Dg of the agent per mm2 of implant surface to which the agent is applied.
8260. The method of item 8061 wherein the agent is delivered from the implant, wherein a surface of the implant comprises about 1 Dg to about 10 Dg of the agent per mm2 of implant surface to which the agent is applied. 8261. The method of item 8061 wherein the agent is delivered from the implant, wherein a surface of the implant comprises about 10 Dg to about 250 Dg of the agent per mm2 of implant surface to which the agent is applied.
8262. The method of item 8061 wherein the agent is delivered from the implant, wherein a surface of the implant comprises about 250 Dg to about 1000 Dg of the agent per mm2 of implant surface to which the agent is applied.
8263. The method of item 8061 wherein the agent is delivered from the implant, wherein a surface of the implant comprises about 1000 Dg to about 2500 Dg of the agent per mm2 of implant surface to which the agent is applied.
8264. The method of item 8061 , wherein the implant further comprises a coating, and the coating is a uniform coating.
8265. The method of item 8061, wherein the implant further comprises a coating, and the coating is a non-uniform coating.
8266. The method of item 8061 , wherein the implant further comprises a coating, and the coating is a discontinuous coating.
8267. The method of item 8061, wherein the implant further comprises a coating, and the coating is a patterned coating.
8268. The method of item 8061 , wherein the implant further comprises a coating, and the coating has a thickness of 100 Dm or less. 8269. The method of item 8061 , wherein the implant further comprises a coating, and the coating has a thickness of 10 Dm or less.
8270. The method of item 8061 , wherein the implant further comprises a coating, and the coating adheres to the surface of the implant upon deployment of the implant.
8271. The method of item 8061, wherein the implant further comprises a coating, and the coating is stable at room temperature for a period of at least 1 year.
8272. The method of item 8061, wherein the implant further comprises a coating, and the agent is present in the coating in an amount ranging between about 0.0001% to about 1% by weight.
- 8273. The method of item 8061 , wherein the implant further _ comprises a coating, and the agent is present in the coating in an amount ranging between about 1% to about 10% by weight.
8274. The method of item 8061, wherein the implant further comprises a coating, and the agent is present in the coating in an amount ranging between about 10% to about 25% by weight.
8275. The method of item 8061 , wherein the implant further comprises a coating, and the agent is present in the coating in an amount ranging between about 25% to about 70% by weight.
8276. The method of item 8061 , wherein the implant further comprises a coating, and the coating comprises a polymer. 8277. The method of item 8061 , wherein the implant comprises a first coating having a first composition and a second coating having a second composition.
8278. The method of item 8061 , wherein the implant comprises a first coating having a first composition and a second coating having a second composition, wherein the first composition and the second composition are different.
8279. A method as in any one of items 8061-8278, wherein the device is a surgical barrier.
8280. A method as in any one of items 8061-8278, wherein the device is a surgical adhesion barrier.
8281. A method as in any one of items 8061-8278, wherein the device is a surgical sheet.
8282. A method as in any one of items 8061-8278, wherein the device is a surgical patch.
8283. A method as in any one of items 8061-8278, wherein the device is a surgical wrap.
8284. A method as in any one of items 8061-8278, wherein the device is a vascular wrap.
8285. A method as in any one of items 8061-8278, wherein the device is a perivascular wrap. 8286. A method as in any one of items 8061-8278, wherein the device is an adventitial wrap.
8287. A method as in any one of items 8061-8278, wherein the device is a periadventitial wrap.
8288. A method as in any one of items 8061-8278, wherein the device is an adventitial sheet.
8289. A method as in any one of items 8061-8278, wherein the device is a perivascular mesh.
8290. A method as in any one of items 8061-8278, wherein the device is a bandage.
8291. A method as in any one of items 8061 -8278, wherein the device is liquid bandage.
8292. A method as in any one of items 8061-8278, wherein the device is a surgical dressing.
8293. A method as in any one of items 8061-8278, wherein the device is a gauze.
8294. A method as in any one of items 8061-8278, wherein the device is a fabric.
8295. A method as in any one of items 8061,-8278, wherein the device is tape. 8296. A method as in any one of items 8061-8278, wherein the device is a surgical membrane.
8297. A method as in any one of items 8061-8278, wherein the device is polymer matrix.
8298. A method as in any one of items 8061-8278, wherein the device is tissue covering.
8299. A method as in any one of items 8061-8278, wherein the device is surgical matrix.
8300. A method as in any one of items 8061-8278, wherein the device is an envelope.
8301. A method as in any one of items 8061-8278, wherein the device is a tissue covering.
8302. A method for inhibiting scarring comprising placing a device that comprises a glaucoma drainage device (e.g., an implant) and an anti-scarring agent or a composition comprising an anti-scarring agent into an animal host, wherein the agent inhibits scarring.
8303. The method of item 8302 wherein the agent is an adensosine A2A receptor antagonist.
8304. The method of item 8302 wherein the agent is an AKT inhibitor. 8305. The method of item 8302 wherein the agent is an alpha 2 integrin antagonist, wherein the alpha 2 integrin antagonist is Pharmaprojects No. 5754 (Merck KgaA).
8306. The method of item 8302 wherein the agent is an alpha 4 integrin antagonist.
8307. The method of item 8302 wherein the agent is an alpha 7 nicotinic receptor agonist.
8308. The method of item 8302 wherein the agent is an angiogenesis inhibitor selected from the group consisting of AG-12,958 (Pfizer), ATN-161 (Attention LLC), neovastat, an angiogenesis inhibitor from Jerina AG (Germany), NM-3 (Mercian), VGA-1155 (Taisho), FCE-26644 (Pfizer), FCE-26950 (Pfizer), FPMA (Meiji Daries), FR-111142 (Fujisawa), GGTI-298, GM-1306 (Ligand), GPA-1734 (Novartis), NNC-47-0011 (Novo Nordisk), herbamycin (Nippon Kayaku), lenalidomide (Celegene), IP-10 (NIH), ABT-828 (Abbott), KIN-841 (Tokushima University, Japan), SF-1126 (Semafore Pharmaceuticals), laminin technology (NIH), CHIR-258 (Chiron), NVP-AEW541 (Novartis), NVP-AEW541 (Novartis), Vt16907 (Alchemia), OXI-8007 (Oxigene), EG-3306 (Ark Therapeutics), Maspin (Arriva), ABT- 567 (Abbott), PPI-2458 (Praecis Pharmaceuticals), CC-5079, CC-4089 (Celgene), HIF-1 alpha inhibitors (Xenova), S-247 (Pfizer), AP-23573 (Ariad), AZD-9935 (Astra Zeneca), mebendazole (Introgen Therapeutics), MetAP-2 inhibitors (GlaxoSmithKline), AG-615 (Angiogene Pharmaceuticals), Tie-2 antagonists (Hybrigenics), NC-381, CYC-381, NC-169, NC-219, NC-383, NC-384, NC-407 (Lorus Therapeutics), ATN-224 (Attenuon), ON-01370 (Onconova), Vitronectin antagonists (Amgen), SDX-103 (Salmedix), Vitronectin antagonists (Shire), CHP (Riemser), TEK (Amgen), Anecortave acetate (Alcon), T46.2 (Matrix Therapeutics), HG-2 (Heptagen), TEM antagonists (Genzyme), Oxi-4500 (Oxigene), ATN-161 (Attenuon), WX-293 (Wilex), M-2025 (Metris Therapeutics), Alphastatin (BioActa), YH-16 (Yantai Rongchang), BIBF-1120 (Boehringer Ingelheim), BAY-57-9352 (Bayer), AS- 1404 (Cancer Research Technology), SC-77964 (Pfizer), glycomimetics (BioTie Therapies), TIE-2 Inhibitors (Ontogen), DIMI, Octamer (Octamer), ABR-215050 (Active Biotech), ABT-518 (Abbott), KDR inhibitors (Abbott), BSF-466895 (Abbott), SCH-221153 (Schering-Plough), DAC:antiangiogenic (ConjuChem), TFPI (EntreMed), AZD-2171 (Astra-Zenaca), CDC-394 (Celgene), LY290293 (EIi Lilly), IDN-5390 (Indena), Kdr Kinase Inhibitors (Merck), CT-113020, CT-116433, CT-116563, CT-31890, CT-32228) (Cell Therapeutics), A-299620 (Abbott), TWEAK Inhibitor (Amgen), VEGF modulators (Johnson and Johnson), Tum-N53, tumstatin (Genzyme), Thios- 1 , Thios-2 (Thios Pharmaceuticals), MV-6401 (Miravant Medical Technologies), Spisulosine (PharmaMar), CEP-7055 (Cephalon), AUV-201 (Auvation), LM-609 (EIi Lilly), SKF-106615 (AnorMED), Oglufanide disodium (Cytran), BW-114 (Pharminox), Calreticulin (NIH), WX-678 (Wilex), SD-7784 (Pfizer), WX-UK1 (Wilex), SH-268 (Schering AG), 2.-Me-PGA (Celgene), S- 137 (Pfizer), ZD-6126 (Angiogene Pharmaceuticals), SG-292 (SignalGen), Benefin (Lane Labs), A6, A36 (Angstrom), SB-2723005 (GlaxoSmithKline), SC-7 (Cell Therapeutics), ZEN-014 (AEterna Zentaris), 2-methoxyestradiol (EntreMed), NK-130119 (Nippon Kayaku), CC-10004 (Celgene), AVE- 8062A (Ajinomoto), Tacedinaline (Pfizer), Actinonin (Tokyo Metropolitan Institute of Medical Science), Lenalidomide (Celgene), VGA-1155, BTO-956 (SRI International), ER-68203-00 (Eisai), CT-6685 (UCB), JKC-362 (Phoenix Pharmaceuticals), DMI-3798 (DMI Biosciences, Angiomate (Ipsen), ZD-6474 (AstraZeneca), CEP-5214 (Cephalon), Canstatin (Genzyme), NM-3 (Mercian), Oridigm (MediQuest Therapeutics), Exherin (Adherex), BLS-0597 (Boston Life Sciences), PTC-299 (PTC Therapeutics), NPI-2358 (Nereus Pharmaceuticals), CGP-79787 (Novartis), AEE-788 (Novartis), CKD-732 (Chong Kun Dang), CP-564959 (OSI Pharmaceuticals), CM-101 (CarboMed), CT-2584, CT3501 (Cell Therapeutics), combretastatin and analogues and derivatives thereof (Oxigene), Rebimastat (Bristol-Meyers Squibb), Dextrin 2-sulfate (ML Laboratories), Cilengitide (Merk KGaA), NSC-706704 (Pharminox), KRN- 951 (Kirin Brewery), Ukrain, NSC-631570 (Nowicky Pharma), Tecogalan sodium (Daiichi Pharmaceutical), Tz-93 (Tsumura), TBC-1635 (Encysive Pharmaceuticals), TAN-1120 (Takeda), Semaxanib (Pfizer), BDI-7800 (Biopharmacopae), SD-186, SD-983 (Bristol-Meyers Squibb), SB-223245 (GlaxoSmithKline), SC-236 (Pfizer), RWJ-590973 (Johnson and Johnson), ILX-1850 (Genzyme), SC-68488, S-836 (Pfizer), CG-55069-11 (CuraGen), Ki-23057 (Kirin Brewery), CCX-700 (Chemoentryx), Pegaptanib octasodium (Giled Sciences), ANGIOCOL (available from Biostratum Inc.), or an analogue or derivative thereof.
8309. The method of item 8302 wherein the agent is an apoptosis antagonist.
8310. The method of item 8302 wherein the agent is_an apoptosis activator.
8311. The method of item 8302 wherein the agent is a beta 1 integrin antagonist.
8312. The method of item 8302 wherein the agent is a beta tubulin inhibitor.
8313. The method of item 8302 wherein the agent is a blocker of enzyme production in Hepatitis C.
8314. The method of item 8302 wherein the agent is a Bruton's tyrosine kinase inhibitor. 8315. The method of item 8302 wherein the agent is a calcineurin inhibitor.
8316. The method of item 8302 wherein the agent is a caspase 3 inhibitor.
8317. The method of item 8302 wherein the agent is a CC chemokine receptor antagonist.
8318. The method of item 8302 wherein the agent is a ceil cycle inhibitor selected from the group consisting of SNS-595 (Sunesis), synthadotin, KRX-0403, and an analogue or derivative thereof.
8319. The method of item 8302 wherein the agent is a cathepsin B inhibitor.
8320. The method of item 8302 wherein the agent is a cathepsin K inhibitor, wherein the cathepsin K inhibitor is 462795 (GlaxoSmithKline), INPL-022-D6 (Amura Therapeutics), or an analogue or derivative thereof.
8321. The method of item 8302 wherein the agent is a cathepsin L inhibitor.
8322. The method of item 8302 wherein the agent is a CD40 antagonist.
8323. The method of item 8302 wherein the agent is a chemokine receptor agonist. 8324. The method of item 8302 wherein the agent is a chymase inhibitor.
8325. The method of item 8302 wherein the agent is a collagenase antagonist.
8326. The method of item 8302 wherein the agent is a CXCR antagonist.
8327. The method of item 8302 wherein the agent is a cyclin dependent kinase inhibitor selected from the group consisting of a CDK-1 inhibitor, a CDK-2 inhibitor, a CDK- 4 inhibitor, a CDK-6 inhibitor, a CAK1 inhibitor from GPC Biotech and Bristol-Myers Squibb, RGB-286199 (GPC Biotech), an anticancer agent from Astex Technology, a CAK1 inhibitor from GPC Biotech, a CDK inhibitor from Sanofi-Aventis, a CDK1/CDK2 inhibitor from Amgen, a CDK2 inhibitor from SUGENU2 (Pfizer), a hearing loss _ therapy agent (Sound Pharmaceuticals), PD-0332991 (Pfizer), RGB-286199 (GPC Biotech), Ro-0505124 (Hoffmann-La Roche), a Ser/Thr kinase inhibitor from Lilly (EIi Lilly), CVT-2584 (CAS No. 199986-75-9) (CV Therapeutics), and an analogue or derivative thereof.
8328. The method of item 8302 wherein the agent is a cyclooxygenase 1 inhibitor.
8329. The method of item 8302 wherein the agent is a DHFR inhibitor.
8330. The method of item 8302 wherein the agent is a dual integrin inhibitor. 8331. The method of item 8302 wherein the agent is an elastase inhibitor.
8332. The method of item 8302 wherein the agent is an elongation factor-1 alpha inhibitor.
8333. The method of item 8302 wherein the agent is an endothelial growth factor antagonist.
8334. The method of item 8302 wherein the agent is an endothelial growth factor receptor kinase inhibitor selected from the group consisting of sorafenib tosylate (Bayer), AAL-993 (Novartis), ABP-309 (Novartis), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), E- 7080 (Eisai), EG-3306 (Ark Therapeutics), EXEL-2880 (Exelixis), GW- 654652 (GlaxoSmithKline), a KDR inhibitor from LG Life Sciences, CT-6685 and CT-6729 (UCB), KRN-633 and KRN-95.1 (Kirin Brewery), OSI.930 (OSI Pharmaceuticals), SP-5.2 (Supratek Pharma), SU-11657 (Pfizer), a Tie-2 antagonist (Hybrigenics), a VEGFR-2 kinase inhibitor (Bristol-Myers Squibb), XL-647 (Exelixis), a KDR inhibitor from Abbott Laboratories, sorafenib tosylate, and an analogue or derivative thereof.
8335. The method of item 8302 wherein the agent is an endotoxin antagonist.
8336. The method of item 8302 wherein the agent is an epothilone and tubulin binder.
8337. The method of item 8302 wherein the agent is an estrogen receptor antagonist. 8338. The method of item 8302 wherein the agent is an FGF inhibitor.
8339. The method of item 8302 wherein the agent is a farnexyl transferase inhibitor.
8340. The method of item 8302 wherein the agent is farnesyltransferase inhibitor selected from the group of A-197574 (Abbott), a famesyltransferase inhibitor from Servier, FPTIII (Strathclyde Institute for Drug R), LB-42908 (LG Life Sciences), Pharmaprojects No. 5063 (Genzyme), Pharmaprojects No. 5597 (Ipsen), Yissum Project No. B-1055 (Yissum), and an analogue or derivative thereof.
8341. The method of item 8302 wherein the agent is an FLT-3 kinase inhibitor.
8342. The method of item 8302 wherein the agent is an FGF receptor kinase inhibitor.
8343. The method of item 8302 wherein the agent is a fibrinogen antagonist selected from the group consisting of AUV-201 (Auvationj, MG-13926 (Sanofi-Aventis), plasminogen activator (CAS No. 105913-11-9) (from Sanofi-Aventis or UCB), plasminogen activator-2 (tPA- 2) (Sanofi-Aventis), pro-urokinase (CAS No. 82657-92-9) (Sanofi-Aventis), and an analogue or derivative thereof.
8344. The method of item 8302 wherein the agent is a heat shock protein 90 antagonist selected from the group consisting of SRN-005 (Sirenade), geldanamycin, NSC-33050 (17-allylaminogeldanamycin; 17- AAG), 17-dimethylaminoethylamino-17-demethoxy-geldanamycin (17- DMAG), rifabutin (rifamycin XlV, 1',4-didehydro-1-deoxy-1 ,4-dihydro-5'-(2- methylpropyl)-1-oxo-), and an analogue or derivative thereof.
8345. The method of item 8302 wherein the agent is a histone deacetylase inhibitor.
8346. The method of item 8302 wherein the agent is an HMGCoA reductase inhibitor selected from the group consisting of an atherosclerosis therapeutic from Lipid Sciences, ATI-16000 (ARYx Therapeutics), KS-01-019 (Kos Pharmaceuticals), Pharmaprojects No. 2197 (Sanofi-Aventi), RP 61969 (Sanofi-Aventis), and an analogue or derivative thereof.
8347. The method of item 8302 wherein the agent is an ICAM inhibitor.
8348. The method of item 8302 wherein the agent is an IL, ICE and IRAK antagonist, wherein the antagonist is a CJ-14877, CP-424174 (Pfizer), NF-61 (Negma-Lerads), and an analogue or derivative thereof.
8349. The method of item 8302 wherein the agent is an IL-2 inhibitor.
8350. The method of item 8302 wherein the agent is an immunosuppressant selected from the group consisting of teriflunomide (Sanofi Aventis), chlorsulfaquinoxalone (NSC-339004), chlorsulfaquinoxalone sulfate, CS-712 (Sankyo), ismomultin alfa (CAS No. 457913-93-8) (Akzo Nobel), antiallergics from GenPat77, antiinflammatories or AT-005 (Androclus Therapeutics), autoimmune disease therapeutics from EpiVax, BN-007 (Bone), budesonide (CAS No. 51333-22- 3) (MAP Pharmaceuticals), CO-14 (Genzyme), edratide (CAS No. 433922- 67-9) (Teva), EP-314 (Enanta), eprovafen (CAS No. 101335-99-3) (Sanofi- Aventis), HWA-131 (CAS No. 118788-41-3) (Sanofi-Aventis), immunomodulators from MerLion Pharmaceuticals, immunosuppressives from Alchemia, IPL-12 (Inflazyme), MDL-9563 (CAS No. 27086-86-8) (Sanofi-Aventis), Pharmaprojects No. 2330 (Sanofi-Aventis), Pharmaprojects No. 6426 (Abgenix), PXS-25 (Pharmaxis), rosmarinic acid (CAS No. 20283-92-5) (Sanofi-Aventis), RP 42927 or RP 54745 (CAS No. 135330-08-4) (Sanofi-Aventis), SGN-35 (Seattle Genetics), ST-1959 (Sigma-Tau), type I diabetes therapy from SYNX Pharma, UNIL-88 (Debiopharm), VP-025 (Vasogen), VR-694 (Vectura), PRTX-001 (Protalex), and an analogue or derivative thereof.
8351. The method of item 8302 wherein the agent is an IMPDH (inosine monophosphate).
8352. The method of item 8302 whereinJhe agent is an - integrin antagonist.
8353. The method of item 8302 wherein the agent is an interleukin antagonist.
8354. The method of item 8302 wherein the agent is an inhibitor of type III receptor tyrosine kinase.
8355. The method of item 8302 wherein the agent is an irreversible inhibitor of enzyme methionine aminopeptidase type 2.
8356. The method of item 8302 wherein the agent is an isozyme selective delta protein kinase C inhibitor. 8357. The method of item 8302 wherein the agent a JAK3 enzyme inhibitor.
8358. The method of item 8302 wherein the agent is a JNK inhibitor.
8359. The method of item 8302 wherein the agent is a kinase inhibitor.
8360. The method of item 8302 wherein the agent is kinesin antagonist.
8361. The method of item 8302 wherein the agent is a kinesin antagonist.
8362. The method of item 8302 wherein the agent-is a leukotriene inhibitor and antagonist selected from the group consisting of ambicromil (CAS No. 58805-38-2) (Sanofi-Aventis), amelubant (CAS No. 346735-24-8) (Boehringer Ingelheim), DW-1141 (Dong Wha), ebselen (Daiichi Pharmaceutical), ibudilast (Kyorin), leucotriene inhibitors from Sanofi-Aventis, lymphotoxin-beta receptor (LT-D) from Biogen Idee, Pharmaprojects No. 1535 and 2728 (CAS No. 119340-33-9) (Sanofi- Aventis), R-112 (Rigel), Rev-5367 (CAS No. 92532-05-3) (Sanofi-Aventis), RG-14893 (CAS No. 141835-49-6) (Sanofi-Aventis), RG-5901-A (CAS No. 101910-24-1), 92532-23-5, RP 66153 (CAS No. 142422-79-5), RP 66364 (CAS No.186916-92-5), RP 69698 (CAS No. 141748-00-7) (Sanofi-Aventis), SC-411930 (Pfizer), SC-41930 (CAS No. 120072-59-5) (Pfizer), SC-50605 (CAS No. 138828-39-4) (Pfizer), SC-51146 (CAS No. 141059-52-1), SC- 53228 (CAS No. 153633-01-3) (Pfizer), spaglumic acid (ZY-15106) (CAS No.3106-85-2), 80619-64-3 (Novartis), tipredane (CAS No. 85197-77-9) (Bristol-Myers Squibb), U-75302 (CAS No. 119477-85-9) (Pfizer), and analogue or derivative thereof.
8363. The method of item 8302 wherein the agent is an MAP kinase inhibitor.
8364. The method of item 8302 wherein the agent is a matrix metalloproteinase inhibitor.
8365. The method of item 8302 wherein the agent is an MCP- CCR2 inhibitor.
8366. The method of item 8302 wherein the agent is an mTOR inhibitor.
8367. The method of item 8302 wherein the agent is an mTOR kinase inhibitor.
8368. The method of item 8302 wherein the agent is a microtubule inhibitor selected from the group consisting of antibody- maytansinoid conjugates from Biogen Idee, colchicines (MantiCore Pharmaceuticals), anticancer immunoconjugates from Johnson & Johnson, DIME from Octamer, gni-1f (GNI), huC242-DM4, huMy9-6-DM1 (ImmunoGen), IDN-5404 (Indena), IMO-098, IMOderm (Imotep), mebendazole (Introgen Therapeutics), microtubule poisons from Cambridge Enterprise, paclitaxel such as LOTAX from Aphios (CAS No. 33069-62-4), Genexol-PM from Samyang, Pharmaprojects No. 6383 (Tapestry Pharmaceuticals), RPR-112378 (Sanofi-Aventis), SGN-75 (Seattle Genetics), SPL-7435 (Starpharma), SSR-250411 (Sanofi-Aventis), trastuzumab-DM1 (Genentech), vinorelbine, and an analogue or derivative thereof. 8369. The method of item 8302 wherein the agent is an MIF inhibitor.
8370. The method of item 8302 wherein the agent is an MMP inhibitor.
8371. The method of item 8302 wherein the agent is a neurokinin (NK) antagonist selected from the group consisting of anthrotainin (CAS No. 148084-40-6) (Sanofi-Aventis), an IBS thereapeutic from ArQuIe, MDL-105212A (CAS No. 167261-60-1) (Ssanofi-Aventis), Pharmaprojects No. 2744, 3258 (CAS No. 139167-47-8) 4006, 4201 , or 5986 (Sanofi-Aventis), RP 67580 (CAS No. 135911-02-3), SR-144190 (CAS No. 201152-86-5), SSR-240600, SSR-241586 (Sanofi-Aventis), TKA-457 (Novartis), vestipitant mesylate (CAS No. 334476-64-1) (GlaxoSmithKline), Win-64821 (Sanofi-Aventis), PRX-96026 (Predix Pharmaceuticals), and an analogue or derivative thereof.
8372. The method of item 8302 wherein the agent is an NF kappa B inhibitor selected from the group consisting of AVE-0545 or AVE- 0547 (Sanofi-Aventis), bortezomib (CAS No. 179324-69-7) (Millennium Pharmaceuticals), dexanabinol (CAS No. 112924-45-5) (Pharmos), dexlipotam (Viatris), Pharmaprojects No. 6283 (INDRA) (OXiGENE), IPL- 576092 (CAS No. 137571-30-3) (Inflazyme), NFKB decoy (Corgentech), NFKB decoy oligo (AnGes MG), NFKB's from Ariad, osteoporosis treatments or S5 (F005) from Fulcrum Pharmaceuticals, P61 (Phytopharm), R-flurbiprofen (CAS No. 5104-49-4) (Encore Pharmaceuticals), and an analogue or derivative thereof.
8373. The method of item 8302 wherein the agent is a nitric oxide agonist. 8374. The method of item 8302 wherein the agent is an ornithine decarboxylase inhibitor.
8375. The method of item 8302 wherein the agent is a p38 MAP kinase inhibitor selected from the group consisting of AZD-6703 (AstraZeneca), JX-401 (Jexys Pharmaceuticals), BMS-2 (Bristol-Myers Squibb), a p38 MAP kinase inhibitor from Novartis, a p38-alpha MAP kinase inhibitor from Amphora, Pharmaprojects No. 5704 (Pharmacopeia), RPR- 200765A (Sanofi-Aventis), SD-282 (Johnson & Johnson), TAK-715 (Takeda), and an analogue or derivative thereof.
8376. The method of item 8302 wherein the agent is a palmitoyl-protein thioesterase inhibitor.
8377. The method of item 8302 wherein the agent is a PDGF receptor kinase inhibitor selected from the group consisting of AAL-993, AMN-107, or ABP-309 (Novartis), AMG-706 (Amgen), BAY-57-9352 (Bayer), CDP-860 (UCB), E-7080 (Eisai), imatinib (CAS No. 152459-95-5) (Novartis), OSI-930 (OSI Pharmaceuticals), RPR-127963E (Sanofi-Aventis), RWJ-540973 (Johnson & Johnson), sorafenib tosylate (Bayer), SU-11657 (Pfizer), tandutinib (CAS No. 387867-13-2) (Millennium Pharmaceuticals), vatalanib (Novartis), ZK-CDK (Schering AG), and an analogue or derivative thereof.
8378. The method of item 8302 wherein the agent is (-)- halofenate (Metabolex), AMG-131 (Amgen), antidiabetics from Japan Tobacco, AZD-4619, AZD-8450, AZD-8677 (AstraZeneca), DRF-10945, balaglitazone (Dr Reddy's), CS-00088, CS-00098 (Chipscreen Biosciences), E-3030 (Eisai), etalocib (CAS No. 161172-51-6) (EIi Lilly), GSK-641597 (Ligand), GSK-677954 (GlaxoSmithKline), GW-409544 (Ligand), GW- 590735 (GlaxoSmithKline), K-111 (Hoffmann-La Roche), LY-518674 (EIi Lilly), LY-674 (Ligand), LY-929 (Ligand), MC-3001 , MC-3002 (MaxoCore Pharmaceuticals), metformin HCI + pioglitazone (CAS No. 1115-70-4 and 112529-15-4), ACTOPLUS MET from Andrx), muraglitazar (CAS No. 331741-94-7) (Bristol-Myers Squibb), naveglitazar (Ligand), oleoylethanolamide (Kadmus Pharmaceuticals), ONO-5129, pioglitazone hydrochloride (CAS No. 111025-46-8 and 112529-15-4) (Takeda), PLX-204 (Plexxikon), PPAR agonists from Genfit, PPAR delta agonists from EIi Lilly, PPAR-alpha agonists from CrystalGenomics, PPAR-gamma modulators and PPAR-β modulators from CareX, rosiglitazone maleate (CAS No. 122324- 73-4 or 155141-29-0) (GlaxoSmithKline), rosiglitazone maleate/glimepir (CAS No. 155141-29-0 and 93479-97-1), AVANDARYL, rosiglitazone maleate/metformin extend (CAS No. 155141-29-0 and 657-24-9), AVANDAMET, rosiglitazone maleate+metformin, AVANDAMET (GlaxoSmithKline), tesaglitazar (AstraZeneca), and an analogue or derivative thereof.
8379. The method of item 8302 wherein the agent is a phosphatase inhibitor.
8380. The method of item 8302 wherein the agent is a phosphodiesterase (PDE) inhibitor selected from the group consisting of avanafil (Tanabe Seiyaku), dasantafil (CAS No. 569351-91-3) (Schering- Plough), A-906119 (CAS No. 134072-58-5), DL-850 (Sanofi-Aventis), GRC- 3015, GRC-3566, GRC-3886 (Glenmark), HWA-153 (CAS No. 56395-66-5) (Sanofi-Aventis), hydroxypumafentrine (Altana), IBFB-130011 , IBFB-14-016, IBFB-140301 , IBFB-150007, IBFB-211913 (IBFB Pharma), L-826141 (Merck & Co), medorinone (CAS No. 88296-61-1) (Sanofi-Aventis), MEM- 1917 (Memory Pharmaceuticals), ND-1251 (Neuro3d), PDE inhibitors from ICOS, PDE IV inhibitors from Memory Pharmaceuticals and CrystalGenomics, Pharmaprojects No. 2742 and 6141 (Sanofi-Aventis), QAD-171 (Novartis), RHC-2963 (CAS No. 76993-12-9 and 76993-14-1), RPR-117658, RPR-122818 derivatives, SR-24870 , and RPR-132294 (Sanofi-Aventis), SK-350 (ln2Gen), stroke therapy agents from deCODE Genetics, TAS-203 (Taiho), tofimilast (CAS No. 185954-27-2) (Pfizer), UK- 371800 (Pfizer), WIN-65579 (CAS No. 158020-82-7) (Sanofi-Aventis), IBFB- 130020 (IBFB Pharma), OPC-6535 (CAS No. 145739-56-6) (Otsuka), a phosphodiesterase III inhibitor, enoximone, a phosphodiesterase IV inhibitor, fosfosal, Atopik (Barrier Therapeutics), triflusal, a phosphodiesterase V inhibitor, and an analogue or derivative thereof.
8381. The method of item 8302 wherein the agent is a PKC inhibitor.
8382. The method of item 8302 wherein the agent is a platelet activating factor antagonist.
8383. The method of item 8302 wherein the agent is a platelet-derived growth factor receptor kinase inhibitor.
8384. The method of item 8302 wherein the agent is a prolyl hydroxylase inhibitor.
8385. The method of item 8302 wherein the agent is a polymorphonuclear neutrophil inhibitor.
8386. The method of item 8302 wherein the agent is a protein kinase B inhibitor.
8387. The method of item 8302 wherein the agent is a protein kinase C stimulant. 8388. The method of item 8302 wherein the agent is a purine nucleoside analogue.
8389. The method of item 8302 wherein the agent is a purinoreceptor P2X antagonist.
8390. The method of item 8302 wherein the agent is a Raf kinase inhibitor.
8391. The method of item 8302 wherein the agent is a reversible inhibitor of ErbB1 and ErbB2.
8392. The method of item 8302 wherein the agent is a ribonucleoside triphosphate reductase inhibitor.
8393. The method of item 8302 wherein the agent is an SDF- 1 antagonist.
8394. The method of item 8302 wherein the agent is a sheddase inhibitor.
8395. The method of item 8302 wherein the agent is an SRC inhibitor.
8396. The method of item 8302 wherein the agent is a stromelysin inhibitor.
8397. The method of item 8302 wherein the agent is an Syk kinase inhibitor. 8398. The method of item 8302 wherein the agent is a telomerase inhibitor.
8399. The method of item 8302 wherein the agent is a TGF beta inhibitor selected from the group consisting of pirfenidone (CAS No. 53179-13-8) (MARNAC), tranilast (CAS No. 53902-12-8) (Kissei), IN-1130 (ln2Gen), mannose-6-phosphate (BTG), TGF-β antagonists from Inflazyme (Pharmaprojects No. 6075), TGF-β antagonists from Sydney, non-industrial source), TGF-β I receptor kinase inhibitors from EIi Lilly, TGF-β receptor inhibitors from Johnson & Johnson, and an analogue or derivative thereof.
8400. The method of item 8302 wherein the agent is a TN Fa antagonist or TACE inhibitor selected from the group consisting of adalimumab (CAS No. 331731-18-1) (Cambridge Antibody Technology), AGIX-4207 (AtheroGenics), AGT-1 (Advanced Biotherapy), an antiinflammatory from Borean Pharma, Cellzome, or Paradigm Therapeutics, anti-inflammatory vaccine (TNF-alpha kinoid) from Neovacs, humanized anti-TNF antibody or an anti-TNF MAb (CB0006) Celltech (UCB), apratastat (CAS No. 287445-51-0) (Wyeth), BMS-561392 (Bristol-Myers Squibb), BN- 006 (Bone), certolizumab pegol (CAS No. 428863-50-7 or CH-138 (UCB), cilomilast (CAS No. 153259-65-5) (GlaxoSmithKline), CR-1 (Nuada Pharmaceuticals), CRx-119 (CombinatoRx), D-5410 (UCB), dacopafant (CAS No. 125372-33-0) (Sanofi-Aventis), dersalazine (CAS No. 188913-57- 7/188913-58-8) (Uriach), etanercept (CAS No. 185243-69-0) (Amgen), ethyl pyruvate (Critical (Critical Therapeutics), golimumab (CAS No. 476181-74-5) (Johnson & Johnson), hormono-immunotherapy from Ipsen, CDP571 (e.g., Humicade from UCB), IC-485 (ICOS), infliximab (CAS No. 170277-31-3) (Johnson & Johnson), IP-751 (Manhattan Pharmaceuticals), ISIS-104838 (CAS No. 250755-32-9) (ISIS Pharmaceuticals), lenalidomide (CAS No. 191732-72-6) (Celgene), lentinan (CAS No. 37339-90-5) (Ajinomoto), MDL- 201112 (CAS No. 142130-73-2) (Sanofi-Aventis), medroxyprogesterone (CAS No. 520-85-4) (InKine Pharmaceutical), N-acetylcysteine (CAS No. 616-91-1) (Zambon), NBE-P2 (DIREVO Biotech), nerelimomab (CAS No. 162774-06-3) (Chiron), OM-294DP (OM PHARMA), onercept (CAS No. 199685-57-9) (Yeda), PASSTNF-alpha (Verigen), pentoxifylline or oxypentifylline (Sanofi-Aventis), Pharmaprojects No. 4091 , 4241 , 4295, or 4488 (Sanofi-Aventis), Pharmaprojects No. 5480 (Amgen), Pharmaprojects No. 6749 (Cengent), pirfenidone (CAS No. 53179-13-8) (MARNAC), RPR- 132294 (Sanofi-Aventis), S5 (F002) (Fulcrum Pharmaceuticals), simvastatin (CAS No. 79902-63-9) (Merck & Co), STA-6292 (Synta Pharmaceuticals), tacrolimus (CAS No. 104987-11-3) (Fujisawa LifeCycle Pharma), talactoferrin alfa (CAS No. 308240-58-6) (Agennix), thalidomide (CAS No. 50-35-1) (Celgene), TNF antagonists form ProStrakan, and Synergen, TNF inhibitors (Amgen), TNF-alpha antagonists from Dynavax Technologies and Jerina AG (Germany), TNF-alpha inhibitors from IBFB Pharma and Xencor (Xencor), torbafylline (CAS No. 105102-21-4) (Sanofi-Aventis), UR-1505 (Uriach), VT-346 (Viron Therapeutics), YSIL6 (Y's Therapeutics), YSTH2 (Y's Therapeutics), NPI-1302a-3 (Nereus Pharmaceuticals, a TNF antagonist from Jerina AG (Germany), dersalazine, and an analogue or derivative thereof.
8401. The method of item 8302 wherein the agent is a Toll receptor inhibitor.
8402. The method of item 8302 wherein the agent is a tubulin antagonist.
8403. The method of item 8302 wherein the agent is a tyrosine kinase inhibitor selected from the group consisting of SU-011248, SUTENT from Pfizer Inc. (New York, NY), BMS-354825, PN-355 (Paracelsian Pharmaceuticals), AGN-199659 (Allergan), AAL-993 or ABP- 309 (Novartis), adaphostin (NIH), AEE-788 (Novartis), AG-013736 (OSI Pharmaceuticals), AG-13736 (Pfizer), ALT-110 (Alteris Therapeutics), AMG- 706 (Amgen), anticancer MAbs from Xencor, anti-EGFrvlll MAbs from Abgenix, anti-HER2 MAb from Abiogen, AZD-2171 or AZD-9935 (AstraZeneca), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), CEP-5214 (Cephalon), CEP-7055 (Cephalon), cetuximab (ImCIone Systems), CHIR-200131 and CHIR-258 (Chiron), CP-547632 (OSI Pharmaceuticals), CP-724714 (Pfizer), CT-301 (Creabilis Therapeutics), D- 69491 (Baxter International), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EGFR/ErbB2 inhibitors from Array BioPharma, erlotinib (CAS No. 183319- 69-9) (OSI Pharmaceuticals), EXEL-2880 (Exelixis), FK-778 (Sanofi- Aventis) , gefitinib (CAS No. 184475-35-2) (AstraZeneca), GW-2286 or GW- 654652 (GlaxoSmithKline), her2/neu antigen from AlphaVax, HER-2/neu inhibitor from Generex, Herzyme (Medipad) (Sirna Therapeutics), HKI-272 (Wyeth), HuMax-EGFr (Genmab), idronoxil (CAS No. 81267-65-4) (Novogen), IGF-1 inhibitors from Ontogen, IMC-11 F8 (ImCIone Systems), kahalalide F (CAS-No. 149204-42-2) (PharmaMar), KDR inhibitor from LG Life Sciences, KDR inhibitors from Abbott Laboratories, KDR kinase inhibitors (UCB), Kdr kinase inhibitors from Merck & Co, KRN-633 and KRN- 951 (Kirin Brewery), KSB-102 (Xenova), lapatinib ditosylate (CAS No. 388082-78-8) (GlaxoSmithKline), matuzumab (Merck KGaA), MDX-214 (Medarex), ME-103 (Pharmexa), MED-A300 (Gerolymatos), MNAC-13 (Lay Line Genomics), nimotuzumab (Center of Molecular Immunology), NSC- 330507 or NSC-707545 (NIH), NV-50 (Novogen), OSI-930 (OSI Pharmaceuticals), panitumumab (Abgenix), pelitinib (CAS No. 287933-82-7) (Wyeth), pertuzumab (CAS No. 380610-27-5) (Genentech), Pharmaprojects No. 3985 (Sanofi-Aventis), prostate cancer therapeutics from Sequenom (SQPC35, SQPC36, SQPC90), removab and remoxab (Trion Pharma), RG- 13022 (CAS No. 136831-48-6), RG-13291 (CAS No. 138989-50-1), or RG- 14620 (CAS No. 136831-49-7) (Sanofi-Aventis), RM-6427 (Romark), RNAi breast cancer therapy from Benitec, RP 53801 (CAS No. 125882-88-4) (Sanofi-Aventis), sorafenib tosylate (Bayer), SU-11657 (Pfizer), Tie-2 antagonists from Semaia (Hybrigenics), Tie-2 inhibitors from Ontogen, trastuzumab (CAS No. 180288-69-1) (Genentech), tyrosine kinase inhibitors from Sanofi-Aventis, U3-1287, U3-1565, U3-1784, U3-1800 (U3 Pharma), vatalanib (Novartis), VEGFR-2 kinase inhibitor from Bristol-Myers Squibb, XL-647 (Exelixis), ZD-6474 (AstraZeneca), ZK-CDK (Schering AG), an EGFR tyrosine kinase inhibitor, EKB-569 (Wyeth), and an analogue or derivative thereof.
8404. The method of item 8302 wherein the agent is a VEGF inhibitor.
8405. The method of item 8302 wherein the agent is a vitamin D receptor agonist.
8406. The method of item 8302 wherein the agent is ZD-6474 (an angiogenesis inhibitor).
8407. The method of item 8302 wherein the agent is AP- 23573 (an mTOR inhibitor).
8408. The method of item 8302 wherein the agent is synthadotin (a tubulin antagonist).
8409. The method of item 8302 wherein the agent is S-0885 (a collagenase inhibitor).
8410. The method of item 8302 wherein the agent is aplidine (an elongation factor-1 alpha inhibitor).
8411. The method of item 8302 wherein the agent is ixabepilone (an epithilone). 8412. The method of item 8302 wherein the agent is IDN- 5390 (an angiogenesis inhibitor).
8413. The method of item 8302 wherein the agent is SB- 2723005 (an angiogenesis inhibitor).
8414. The method of item 8302 wherein the agent is ABT-518 (an angiogenesis inhibitor).
8415. The method of item 8302 wherein the agent is combretastatin (an angiogenesis inhibitor).
8416. The method of item 8302 wherein the agent is anecortave acetate (an angiogenesis inhibitor).
8417. The method of item 8302 wherein the agent is SB- 715992 (a kinesin antagonist).
8418. The method of item 8302 wherein the agent is temsirolimus (an mTOR inhibitor).
8419. The method of item 8302 wherein the agent is adalimumab (a TNFα antagonist).
8420. The method of item 8302, wherein the composition comprises a polymer.
8421. The method of item 8302, wherein the composition comprises a polymer, and the polymer is, or comprises, a copolymer. 8422. The method of item 8302, wherein the composition comprises a polymer, and the polymer is, or comprises, a block copolymer.
8423. The method of item 8302, wherein the composition comprises a polymer, and the polymer is, or comprises, a random copolymer.
8424. The method of item 8302, wherein the composition comprises a polymer, and the polymer is, or comprises, a biodegradable polymer.
8425. The method of item 8302, wherein the composition comprises a polymer, and the polymer is, or comprises, a nonbiodegradable polymer.
8426. The method of item 8302, whereiη the composition comprises a polymer, and the polymer is, or comprises, a hydrophilic polymer.
8427. The method of item 8302, wherein the composition comprises a polymer, and the polymer is, or comprises, a hydrophobic polymer.
8428. The method of item 8302, wherein the composition comprises a polymer, and the polymer is, or comprises, a polymer having hydrophilic domains.
8429. The method of item 8302, wherein the composition comprises a polymer, and the polymer is, or comprises, a polymer having hydrophobic domains. 8430. The method of item 8302, wherein the composition comprises a polymer, and the polymer is, or comprises, a non-conductive polymer.
8431. The method of item 8302, wherein the composition comprises a polymer, and the polymer is, or comprises, an elastomer.
8432. The method of item 8302, wherein the composition comprises a polymer, and the polymer is, or comprises, a hydrogel.
8433. The method of item 8302, wherein the composition comprises a polymer, and the polymer is, or comprises, a silicone polymer.
8434. The method of item 8302, wherein the composition comprises a polymer, and the polymer is, or comprises, a hydrocarbon polymer.
8435. The method of item 8302, wherein the composition comprises a polymer, and the polymer is, or comprises, a styrene-derived polymer.
8436. The method of item 8302, wherein the composition comprises a polymer, and the polymer is, or comprises, a butadiene-derived polymer.
8437. The method of item 8302, wherein the composition comprises a polymer, and the polymer is, or comprises, a macromer.
8438. The method of item 8302, wherein the composition comprises a polymer, and the polymer is, or comprises, a poly(ethylene glycol) polymer. 8439. The method of item 8302, wherein the composition comprises a polymer, and the polymer is, or comprises, an amorphous polymer.
8440. The method of item 8302, wherein the composition further comprises a second pharmaceutically active agent.
8441. The method of item 8302, wherein the composition further comprises an anti-inflammatory agent.
8442. The method of item 8302, wherein the composition further comprises an agent that inhibits infection.
8443. The method of item 8302, wherein the composition further comprises an anthracycline.
8444. The method of item 8302, wherein the composition further comprises doxorubicin.
8445. The method of item 8302 wherein the composition further comprises mitoxantrone.
8446. The method of item 8302 wherein the composition further comprises a fluoropyrimidine.
8447. The method of item 8302, wherein the composition further comprises 5-fluorouracil (5-FU).
8448. The method of item 8302, wherein the composition further comprises a folic acid antagonist. 8449. The method of item 8302, wherein the composition further comprises methotrexate.
8450. The method of item 8302, wherein the composition further comprises a podophylotoxin.
8451. The method of item 8302, wherein the composition further comprises etoposide.
8452. The method of item 8302, wherein the composition further comprises camptothecin.
8453. The method of item 8302, wherein the composition further comprises a hydroxyurea.
8454. The method of item 8302, wherein the composition further comprises a platinum complex.
8455. The method of item 8302, wherein the composition further comprises cisplatin.
8456. The method of item 8302 wherein the composition further comprises an anti-thrombotic agent.
8457. The method of item 8302, wherein the composition further comprises a visualization agent.
8458. The method of item 8302, wherein the composition further comprises a visualization agent, and the visualization agent is a radiopaque material, wherein the radiopaque material comprises a metal, a halogenated compound, or a barium containing compound. 8459. The method of item 8302, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, barium, tantalum, or technetium.
8460. The method of item 8302, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, an MRI responsive material.
8461. The method of item 8302, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, a gadolinium chelate.
8462. The method of item 8302, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, iron, magnesium, manganese, copper, or chromium.
8463. The method of item 8302, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, iron oxide compound.
8464. The method of item 8302, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, a dye, pigment, or colorant.
8465. The method of item 8302 wherein the agent is released in effective concentrations from the composition comprising the agent by diffusion over a period ranging from the time of administration to about 90 days.
8466. The method of item 8302 wherein the agent is released in effective concentrations from the composition comprising the agent by erosion of the composition over a period ranging from the time of administration to about 90 days.
8467. The method of item 8302 wherein the composition further comprises an inflammatory cytokine.
8468. The method of item 8302 wherein the composition further comprises an agent that stimulates cell proliferation.
8469. The method of item 8302 wherein the composition further comprises a polymeric carrier.
8470. The method of item 8302 wherein the composition is in the form of a gel, paste, or spray.
-- 8471. The method of item 8302 wherein. the implant is partially constructed with the agent or the composition.
8472. The method of item 8302 wherein the implant is fully constructed with the agent or the composition.
8473. The method of item 8302 wherein the implant is impregnated with the agent or the composition.
8474. The method of item 8302, wherein the agent or the composition forms a coating, and the coating directly contacts the implant.
8475. The method of item 8302, wherein the agent or the composition forms a coating, and the coating indirectly contacts the implant. 8476. The method of item 8302 wherein the agent or the composition forms a coating, and the coating partially covers the implant.
8477. The method of item 8302, wherein the agent or the composition forms a coating, and the coating completely covers the implant.
8478. The method of item 8302 wherein the agent or the composition is located within pores or holes of the implant.
8479. The method of item 8302 wherein the agent or the composition is located within a channel, lumen, or divet of the implant.
8480. The method of item 8302 wherein the implant further comprising an echogenic material.
8481. The method of item 8302 wherein the implant further comprises an echogenic material, wherein the echogenic material is in the form of a coating.
8482. The method of item 8302 wherein the implant is sterile.
8483. The method of item 8302 wherein the agent is delivered from the implant, wherein the agent is released into tissue in the vicinity of the implant after deployment of the implant.
8484. The method of item 8302 wherein the agent is delivered from the implant, wherein the agent is released into tissue in the vicinity of the implant after deployment of the implant, wherein the tissue is connective tissue. 8485. The method of item 8302 wherein the agent is delivered from the implant, wherein the agent is released into tissue in the vicinity of the implant after deployment of the implant, wherein the tissue is muscle tissue.
8486. The method of item 8302 wherein the agent is delivered from the implant, wherein the agent is released into tissue in the vicinity of the implant after deployment of the implant, wherein the tissue is nerve tissue.
8487. The method of item 8302 wherein the agent is delivered from the implant, wherein the agent is released into tissue in the vicinity of the implant after deployment of the implant, wherein the tissue is epithelium tissue.
8488. The method of item 8302 wherein the agent is delivered from the implant, wherein the agent is released in effective concentrations from the implant over a period ranging from the time of deployment of the implant to about 1 year.
8489. The method of item 8302 wherein the agent is delivered from the implant, wherein the agent is released in effective concentrations from the implant over a period ranging from about 1 month to 6 months.
8490. The method of item 8302 wherein the agent is delivered from the implant, wherein the agent is released in effective concentrations from the implant over a period ranging from about 1 - 90 days.
8491. The method of item 8302 wherein the agent is delivered from the implant, wherein the agent is released in effective concentrations from the implant at a constant rate. 8492. The method of item 8302 wherein the agent is delivered from the implant, wherein the agent is released in effective concentrations from the implant at an increasing rate.
8493. The method of item 8302 wherein the agent is delivered from the implant, wherein the agent is released in effective concentrations from the implant at a decreasing rate.
8494. The method of item 8302 wherein the agent is delivered from the implant, wherein the implant comprises about 0.01 Dg to about 10 Dg of the agent.
8495. The method of item 8302 wherein the agent is delivered from the implant, wherein the implant comprises about 10 Dg to about 10 mg of the agent.
8496. The method of item 8302 wherein the agent is delivered from the implant, wherein the implant comprises about 10 mg to about 250 mg of the agent. ,
8497. The method of item 8302 wherein the agent is delivered from the implant, wherein the implant comprises about 250 mg to about
1000 mg of the agent.
8498. The method of item 8302 wherein the agent is delivered from the implant, wherein the implant comprises about 1000 mg to about 2500 mg of the agent.
8499. The method of item 8302 wherein the agent is delivered from the implant, wherein a surface of the implant comprises less than 0.01 Dg of the agent per mm2 of implant surface to which the agent is applied. 8500. The method of item 8302 wherein the agent is delivered from the implant, wherein a surface of the implant comprises about 0.01 Dg to about 1 Og of the agent per mm2 of implant surface to which the agent is applied.
8501. The method of item 8302 wherein the agent is delivered from the implant, wherein a surface of the implant comprises about 1 Dg to about 10 Dg of the agent per mm2 of implant surface to which the agent is applied.
8502. The method of item 8302 wherein the agent is delivered from the implant, wherein a surface of the implant comprises about 10 Dg to about 250 Dg of the agent per mm2 of implant surface to which the agent is applied.
8503. The method of item 8302 wherein the agent is delivered from the implant, wherein a surface of the implant comprises about 250 Dg to about 1000 Dg of the agent per mm2 of implant surface to which the agent is applied.
8504. The method of item 8302 wherein the agent is delivered from the implant, wherein a surface of the implant comprises about 1000 Dg to about 2500 Dg of the agent per mm2 of implant surface to which the agent is applied.
8505. The method of item 8302, wherein the implant further comprises a coating, and the coating is a uniform coating.
8506. The method of item 8302, wherein the implant further comprises a coating, and the coating is a non-uniform coating. 8507. The method of item 8302, wherein the implant further comprises a coating, and the coating is a discontinuous coating.
8508. The method of item 8302, wherein the implant further comprises a coating, and the coating is a patterned coating.
8509. The method of item 8302, wherein the implant further comprises a coating, and the coating has a thickness of 100 Dm or less.
8510. The method of item 8302, wherein the implant further comprises a coating, and the coating has a thickness of 10 Dm or less.
8511. The method of item 8302, wherein the implant further comprises a coating, and the coating adheres to the surface of the implant upon deployment of the implant.
8512. The method of item 8302, wherein the implant further comprises a coating, and the coating is stable at room temperature for a period of at least 1 year.
8513. The method of item 8302, wherein the implant further comprises a coating, and the agent is present in the coating in an amount ranging between about 0.0001% to about 1% by weight.
8514. The method of item 8302, wherein the implant further comprises a coating, and the agent is present in the coating in an amount ranging between about 1% to about 10% by weight.
8515. The method of item 8302, wherein the implant further comprises a coating, and the agent is present in the coating in an amount ranging between about 10% to about 25% by weight. 8516. The method of item 8302, wherein the implant further comprises a coating, and the agent is present in the coating in an amount ranging between about 25% to about 70% by weight.
8517. The method of item 8302, wherein the implant further comprises a coating, and the coating comprises a polymer.
8518. The method of item 8302, wherein the implant comprises a first coating having a first composition and a second coating having a second composition.
8519. The method of item 8302, wherein the implant comprises a first coating having a first composition and a second coating having a second composition, wherein the first composition and the second composition are different.
8520. A method as in any one of items 8302-8519, wherein the device is a glaucoma drainage device comprising a plate and a tube.
8521. A method for inhibiting scarring comprising placing a device that comprises a prosthetic heart valve or component thereof (e.g., an implant) and an anti-scarring agent or a composition comprising an anti- scarring agent into an animal host, wherein the agent inhibits scarring.
8522. The method of item 8521 wherein the agent is an adensosine A2A receptor antagonist.
8523. The method of item 8521 wherein the agent is an AKT inhibitor. 8524. The method of item 8521 wherein the agent is an alpha 2 integrin antagonist, wherein the alpha 2 integrin antagonist is Pharmaprojects No. 5754 (Merck KgaA).
8525. The method of item 8521 wherein the agent is an alpha 4 integrin antagonist.
8526. The method of item 8521 wherein the agent is an alpha 7 nicotinic receptor agonist.
8527. The method of item 8521 wherein the agent is an angiogenesis inhibitor selected from the group consisting of AG-12,958 (Pfizer), ATN-161 (Attention LLC), neovastat, an angiogenesis inhibitor from Jerina AG (Germany), NM-3 (Mercian), VGA-1155 (Taisho), FCE-26644 (Pfizer), FCE-26950 (Pfizer), FPMA (Meiji Daries), FR-111142 (Fujisawa), GGTI-298, GM-1306 (Ligand), GPA-1734 (Novartis), NNC-47-O011 (Novo Nordisk), herbamycin (Nippon Kayaku), lenalidomide (Celegene), IP-10 (NIH), ABT-828 (Abbott), KIN-841 (Tokushima University, Japan), SF-1126 (Semafore Pharmaceuticals), laminin technology (NIH), CHIR-258 (Chiron), NVP-AEW541 (Novartis), NVP-AEW541 (Novartis), Vt16907 (Alchemia), OXI-8007 (Oxigene), EG-3306 (Ark Therapeutics), Maspin (Arriva), ABT- 567 (Abbott), PPI-2458 (Praecis Pharmaceuticals), CC-5079, CC-4089 (Celgene), HIF-1 alpha inhibitors (Xenova), S-247 (Pfizer), AP-23573 (Ariad), AZD-9935 (Astra Zeneca), mebendazole (Introgen Therapeutics), MetAP-2 inhibitors (GlaxoSmithKline), AG-615 (Angiogene Pharmaceuticals), Tie-2 antagonists (Hybrigenics), NC-381 , CYC-381, NC-169, NC-219, NC-383, NC-384, NC-407 (Lorus Therapeutics), ATN-224 (Attenuon), ON-01370 (Onconova), Vitronectin antagonists (Amgen), SDX-103 (Salmedix), Vitronectin antagonists (Shire), CHP (Riemser), TEK (Amgen), Anecortave acetate (Alcon), T46.2 (Matrix Therapeutics), HG-2 (Heptagen), TEM antagonists (Genzyme), Oxi-4500 (Oxigene), ATN-161 (Attenuon), WX-293 (Wilex), M-2025 (Metris Therapeutics), Alphastatin (BioActa), YH-16 (Yantai Rongchang), BIBF-1120 (Boehringer Ingelheim), BAY-57-9352 (Bayer), AS- 1404 (Cancer Research Technology), SC-77964 (Pfizer), glycomimetics (BioTie Therapies), TIE-2 Inhibitors (Ontogen), DIMI, Octamer (Octamer), ABR-215050 (Active Biotech), ABT-518 (Abbott), KDR inhibitors (Abbott), BSF-466895 (Abbott), SCH-221153 (Schering-Plough), DAC:antiangiogenic (ConjuChem), TFPI (EntreMed), AZD-2171 (Astra-Zenaca), CDC-394 (Celgene), LY290293 (EIi Lilly), IDN-5390 (Indena), Kdr Kinase Inhibitors (Merck), CT-113020, CT-116433, CT-116563, CT-31890, CT-32228) (Cell Therapeutics), A-299620 (Abbott), TWEAK Inhibitor (Amgen), VEGF modulators (Johnson and Johnson), Tum-N53, tumstatin (Genzyme), Thios- 1, Thios-2 (Thios Pharmaceuticals), MV-6401 (Miravant Medical Technologies), Spisulosine (PharmaMar), CEP-7055 (Cephalon), AUV-201 (Auvation), LM-609 (EIi Lilly), SKF-106615 (AnorMED), Oglufanide disodium (Cytran), BW-114 (Pharminox), Calreticulin (NIH), WX-678 (Wilex), SD-7784 (Pfizer), WX-UK1 (Wilex), SH-268 (Schering AG), 2-Me-PGA (Celgene), S- 137 (Pfizer), ZD-6126 (Angiogene Pharmaceuticals), SG-292 (SignalGen), Benefin (Lane Labs), A6, A36 (Angstrom), SB-2723005 (GlaxoSmithKline), SC-7 (Cell Therapeutics), ZEN-014 (AEterna Zentaris), 2-methoxyestradiol (EntreMed), NK-130119 (Nippon Kayaku), CC-10004 (Celgene), AVE- 8062A (Ajinomoto), Tacedinaline (Pfizer), Actinonin (Tokyo Metropolitan Institute of Medical Science), Lenalidomide (Celgene), VGA-1155, BTO-956 (SRI International), ER-68203-00 (Eisai), CT-6685 (UCB), JKC-362 (Phoenix Pharmaceuticals), DMI-3798 (DMI Biosciences, Angiomate (Ipsen), ZD-6474 (AstraZeneca), CEP-5214 (Cephalon), Canstatin (Genzyme), NM-3 (Mercian), Oridigm (MediQuest Therapeutics), Exherin (Adherex), BLS-0597 (Boston Life Sciences), PTC-299 (PTC Therapeutics), NPI-2358 (Nereus Pharmaceuticals), CGP-79787 (Novartis), AEE-788 (Novartis), CKD-732 (Chong Kun Dang), CP-564959 (OSI Pharmaceuticals), CM-101 (CarboMed), CT-2584, CT3501 (Cell Therapeutics), combretastatin and analogues and derivatives thereof (Oxigene), Rebimastat (Bristol-Meyers Squibb), Dextrin 2-sulfate (ML Laboratories), Cilengitide (Merk KGaA), NSC-706704 (Pharminox), KRN- 951 (Kirin Brewery), Ukrain, NSC-631570 (Nowicky Pharma), Tecogalan sodium (Daiichi Pharmaceutical), Tz-93 (Tsumura), TBC-1635 (Encysive Pharmaceuticals), TAN-1120 (Takeda), Semaxanib (Pfizer), BDI-7800 (Biopharmacopae), SD-186, SD-983 (Bristol-Meyers Squibb), SB-223245 (GlaxoSmithKline), SC-236 (Pfizer), RWJ-590973 (Johnson and Johnson), ILX-1850 (Genzyme), SC-68488, S-836 (Pfizer), CG-55069-11 (CuraGen), Ki-23057 (Kirin Brewery), CCX-700 (Chemoentryx), Pegaptanib octasodium (Giled Sciences), ANGIOCOL (available from Biostratum Inc.), or an analogue or derivative thereof.
8528. The method of item 8521 wherein the agent is an apoptosis antagonist.
8529. The method of item 8521 wherein the agent is an _ . apoptosis activator.
8530. The method of item 8521 wherein the agent is a beta 1 integrin antagonist.
8531. The method of item 8521 wherein the agent is a beta tubulin inhibitor.
8532. The method of item 8521 wherein the agent is a blocker of enzyme production in Hepatitis C.
8533. The method of item 8521 wherein the agent is a Bruton's tyrosine kinase inhibitor. 8534. The method of item 8521 wherein the agent is a calcineurin inhibitor.
8535. The method of item 8521 wherein the agent is a caspase 3 inhibitor.
8536. The method of item 8521 wherein the agent is a CC chemokine receptor antagonist.
8537. The method of item 8521 wherein the agent is a cell cycle inhibitor selected from the group consisting of SNS-595 (Sunesis), synthadotin, KRX-0403, and an analogue or derivative thereof.
8538. The method of item 8521 wherein the agent is a cathepsin B inhibitor.
8539. The method of item 8521 wherein the agent is a cathepsin K inhibitor, wherein the cathepsin K inhibitor is 462795 (GlaxoSmithKline), INPL-022-D6 (Amura Therapeutics), or an analogue or derivative thereof.
8540. The method of item 8521 wherein the agent is a cathepsin L inhibitor.
8541. The method of item 8521 wherein the agent is a CD40 antagonist.
8542. The method of item 8521 wherein the agent is a chemokine receptor agonist. 8543. The method of item 8521 wherein the agent is a chymase inhibitor.
8544. The method of item 8521 wherein the agent is a collagenase antagonist.
8545. The method of item 8521 wherein the agent is a CXCR antagonist.
8546. The method of item 8521 wherein the agent is a cyclin dependent kinase inhibitor selected from the group consisting of a CDK-1 inhibitor, a CDK-2 inhibitor, a CDK- 4 inhibitor, a CDK-6 inhibitor, a CAK1 inhibitor from GPC Biotech and Bristol-Myers Squibb, RGB-286199 (GPC Biotech), an anticancer agent from Astex Technology, a CAK1 inhibitor from GPC Biotech, a CDK inhibitor from Sanofi-Aventis, a CDK1/CDK2 inhibitor from Amgen, a CDK2 inhibitor from SUGEN-2 (Pfizer), a hearingJoss. .. therapy agent (Sound Pharmaceuticals), PD-0332991 (Pfizer), RGB-286199 (GPC Biotech), Ro-0505124 (Hoffmann-La Roche), a Ser/Thr kinase inhibitor from Lilly (EIi Lilly), CVT-2584 (CAS No. 199986-75-9) (CV Therapeutics), and an analogue or derivative thereof.
8547. The method of item 8521 wherein the agent is a cyclooxygenase 1 inhibitor.
8548. The method of item 8521 wherein the agent is a DHFR inhibitor.
8549. The method of item 8521 wherein the agent is a dual integrin inhibitor. 8550. The method of item 8521 wherein the agent is an elastase inhibitor.
8551. The method of item 8521 wherein the agent is an elongation factor-1 alpha inhibitor.
8552. The method of item 8521 wherein the agent is an endothelial growth factor antagonist.
8553. The method of item 8521 wherein the agent is an endothelial growth factor receptor kinase inhibitor selected from the group consisting of sorafenib tosylate (Bayer), AAL-993 (Novartis), ABP-309 (Novartis), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), E- 7080 (Eisai), EG-3306 (Ark Therapeutics), EXEL-2880 (Exelixis), GW- 654652 (GlaxoSmithKline), a KDR inhibitor from LG Life Sciences, CT-6685 and CT-6729 (UCB), KRN-633 and KRN-951 (Kirin Brewery), OSI-930 (QSI Pharmaceuticals), SP-5.2 (Supratek Pharma), SU-11657 (Pfizer), a Tie-2 antagonist (Hybrigenics), a VEGFR-2 kinase inhibitor (Bristol-Myers Squibb), XL-647 (Exelixis), a KDR inhibitor from Abbott Laboratories, sorafenib tosylate, and an analogue or derivative thereof.
8554. The method of item 8521 wherein the agent is an endotoxin antagonist.
8555. The method of item 8521 wherein the agent is an epothilone and tubulin binder.
8556. The method of item 8521 wherein the agent is an estrogen receptor antagonist. 8557. The method of item 8521 wherein the agent is an FGF inhibitor.
8558. The method of item 8521 wherein the agent is a farnexyl transferase inhibitor.
8559. The method of item 8521 wherein the agent is farnesyltransferase inhibitor selected from the group of A-197574 (Abbott), a farnesyltransferase inhibitor from Servier, FPTIII (Strathclyde Institute for Drug R), LB-42908 (LG Life Sciences), Pharmaprojects No. 5063 (Genzyme), Pharmaprojects No. 5597 (Ipsen), Yissum Project No. B-1055 (Yissum), and an analogue or derivative thereof.
8560. The method of item 8521 wherein the agent is an FLT-3 kinase inhibitor.
8561. The method of item 8521 wherein the agent is an FGF receptor kinase inhibitor.
8562. The method of item 8521 wherein the agent is a fibrinogen antagonist selected from the group consisting of AUV-201 (Auvation), MG-13926 (Sanofi-Aventis), plasminogen activator (CAS No. 105913-11-9) (from Sanofi-Aventis or UCB), plasminogen activator-2 (tPA- 2) (Sanofi-Aventis), pro-urokinase (CAS No. 82657-92-9) (Sanofi-Aventis), and an analogue or derivative thereof.
8563. The method of item 8521 wherein the agent is a heat shock protein 90 antagonist selected from the group consisting of SRN-005 (Sirenade), geldanamycin, NSC-33050 (17-allylaminogeldanamycin; 17- AAG), 17-dimethylaminoethylamino-17-demethoxy-geldanamycin (17- DMAG), rifabutin (rifamycin XIV, 1',4-didehydro-1-deoxy-1 ,4-dihydro-5'-(2- methylpropyl)-1-oxo-), and an analogue or derivative thereof.
8564. The method of item 8521 wherein the agent is a histone deacetylase inhibitor.
8565. The method of item 8521 wherein the agent is an HMGCoA reductase inhibitor selected from the group consisting of an atherosclerosis therapeutic from Lipid Sciences, ATI-16000 (ARYx Therapeutics), KS-01-019 (Kos Pharmaceuticals), Pharmaprojects No. 2197 (Sanofi-Aventi), RP 61969 (Sanofi-Aventis), and an analogue or derivative thereof.
8566. The method of item 8521 wherein the agent is an ICAM inhibitor.
8567. The method of item 8521 wherein the agent is an IL, ICE and IRAK antagonist, wherein the antagonist is a CJ-14877, CP-424174 (Pfizer), NF-61 (Negma-Lerads), and an analogue or derivative thereof.
8568. The method of item 8521 wherein the agent is an IL-2 inhibitor.
8569. The method of item 8521 wherein the agent is an immunosuppressant selected from the group consisting of teriflunomide (Sanofi Aventis), chlorsulfaquinoxalone (NSC-339004), chlorsulfaquinoxalone sulfate, CS-712 (Sankyo), ismomultin alfa (CAS No. 457913-93-8) (Akzo Nobel), antiallergics from GenPat77, antiinflammatories or AT-005 (Androclus Therapeutics), autoimmune disease therapeutics from EpiVax, BN-007 (Bone), budesonide (CAS No. 51333-22- 3) (MAP Pharmaceuticals), CO-14 (Genzyme), edratide (CAS No. 433922- 67-9) (Teva), EP-314 (Enanta), eprovafen (CAS No. 101335-99-3) (Sanofi- Aventis), HWA-131 (CAS No. 118788-41-3) (Sanofi-Aventis), immunomodulators from MerLion Pharmaceuticals, immunosuppressives from Alchemia, IPL-12 (Inflazyme), MDL-9563 (CAS No. 27086-86-8) (Sanofi-Aventis), Pharmaprojects No. 2330 (Sanofi-Aventis), Pharmaprojects No. 6426 (Abgenix), PXS-25 (Pharmaxis), rosmarinic acid (CAS No. 20283-92-5) (Sanofi-Aventis), RP 42927 or RP 54745 (CAS No. 135330-08-4) (Sanofi-Aventis), SGN-35 (Seattle Genetics), ST-1959 (Sigma-Tau), type I diabetes therapy from SYNX Pharma, UNIL-88 (Debiopharm), VP-025 (Vasogen), VR-694 (Vectura), PRTX-001 (Protaiex), and an analogue or derivative thereof.
8570. The method of item 8521 wherein the agent is an IMPDH (inosine monophosphate).
8571. The method of item 8521 wherein tha agent is an integrin antagonist.
8572. The method of item 8521 wherein the agent is an interleukin antagonist.
8573. The method of item 8521 wherein the agent is an inhibitor of type III receptor tyrosine kinase.
8574. The method of item 8521 wherein the agent is an irreversible inhibitor of enzyme methionine aminopeptidase type 2.
8575. The method of item 8521 wherein the agent is an isozyme selective delta protein kinase C inhibitor. 8576. The method of item 8521 wherein the agent a JAK3 enzyme inhibitor.
8577. The method of item 8521 wherein the agent is a JNK inhibitor.
8578. The method of item 8521 wherein the agent is a kinase inhibitor.
8579. The method of item 8521 wherein the agent is kinesin antagonist.
8580. The method of item 8521 wherein the agent is a kinesin antagonist.
8581. The method of item 8521 whereinihe agent is. a leukotriene inhibitor and antagonist selected from the group consisting of ambicromil (CAS No. 58805-38-2) (Sanofi-Aventis), amelubant (CAS No. 346735-24-8) (Boehringer Ingelheim), DW-1141 (Dong Wha), ebselen (Daiichi Pharmaceutical), ibudilast (Kyorin), leucotriene inhibitors from Sanofi-Aventis, lymphotoxin-beta receptor (LT-D) from Biogen Idee, Pharmaprojects No. 1535 and 2728 (CAS No. 119340-33-9) (Sanofi- Aventis), R-112 (Rigel), Rev-5367 (CAS No. 92532-05-3) (Sanofi-Aventis), RG-14893 (CAS No. 141835-49-6) (Sanofi-Aventis), RG-5901-A (CAS No. 101910-24-1), 92532-23-5, RP 66153 (CAS No. 142422-79-5), RP 66364 (CAS No.186916-92-5), RP 69698 (CAS No. 141748-00-7) (Sanofi-Aventis), SC-411930 (Pfizer), SC-41930 (CAS No. 120072-59-5) (Pfizer), SC-50605 (CAS No. 138828-39-4) (Pfizer), SC-51146 (CAS No. 141059-52-1), SC- 53228 (CAS No. 153633-01-3) (Pfizer), spaglumic acid (ZY-15106) (CAS No.3106-85-2), 80619-64-3 (Novartis), tipredane (CAS No. 85197-77-9) (Bristol-Myers Squibb), U-75302 (CAS No. 119477-85-9) (Pfizer), and analogue or derivative thereof.
8582. The method of item 8521 wherein the agent is an MAP kinase inhibitor.
8583. The method of item 8521 wherein the agent is a matrix metalloproteinase inhibitor.
8584. The method of item 8521 wherein the agent is an MCP- CCR2 inhibitor.
8585. The method of item 8521 wherein the agent is an mTOR inhibitor.
8586. The method of item 8521 wherein the agent is an mTOR kinase inhibitor.
8587. The method of item 8521 wherein the agent is a microtubule inhibitor selected from the group consisting of antibody- maytansinoid conjugates from Biogen Idee, colchicines (MantiCore Pharmaceuticals), anticancer immunoconjugates from Johnson & Johnson, DIME from Octamer, gni-1f (GNI), huC242-DM4, huMy9-6-DM1 (ImmunoGen), IDN-5404 (Indena), IMO-098, IMOderm (Imotep), mebendazole (Introgen Therapeutics), microtubule poisons from Cambridge Enterprise, paclitaxel such as LOTAX from Aphios (CAS No. 33069-62-4), Genexol-PM from Samyang, Pharmaprojects No. 6383 (Tapestry Pharmaceuticals), RPR-112378 (Sanofi-Aventis), SGN-75 (Seattle Genetics), SPL-7435 (Starpharma), SSR-250411 (Sanofi-Aventis), trastuzumab-DM1 (Genentech), vinorelbine, and an analogue or derivative thereof. 8588. The method of item 8521 wherein the agent is an MIF inhibitor.
8589. The method of item 8521 wherein the agent is an MMP inhibitor.
8590. The method of item 8521 wherein the agent is a neurokinin (NK) antagonist selected from the group consisting of anthrotainin (CAS No. 148084-40-6) (Sanofi-Aventis), an IBS thereapeutic from ArQuIe, MDL-105212A (CAS No. 167261-60-1) (Ssanofi-Aventis), Pharmaprojects No. 2744, 3258 (CAS No. 139167-47-8) 4006, 4201, or 5986 (Sanofi-Aventis), RP 67580 (CAS No. 135911-02-3), SR-144190 (CAS No. 201152-86-5), SSR-240600, SSR-241586 (Sanofi-Aventis), TKA-457 (Novartis), vestipitant mesylate (CAS No. 334476-64-1) (GlaxoSmithKline), Win-64821 (Sanofi-Aventis), PRX-96026 (Predix Pharmaceuticals), and an analogue or derivative thereof. ~
8591. The method of item 8521 wherein the agent is an NF kappa B inhibitor selected from the group consisting of AVE-0545 or AVE- 0547 (Sanofi-Aventis), bortezomib (CAS No. 179324-69-7) (Millennium Pharmaceuticals), dexanabinol (CAS No. 112924-45-5) (Pharmos), dexlipotam (Viatris), Pharmaprojects No. 6283 (INDRA) (OXiGENE), IPL- 576092 (CAS No. 137571-30-3) (Inflazyme), NFKB decoy (Corgentech), NFKB decoy oligo (AnGes MG), NFKB's from Ariad, osteoporosis treatments or S5 (F005) from Fulcrum Pharmaceuticals, P61 (Phytopharm), R-flurbiprofen (CAS No. 5104-49-4) (Encore Pharmaceuticals), and an analogue or derivative thereof.
8592. The method of item 8521 wherein the agent is a nitric oxide agonist. 8593. The method of item 8521 wherein the agent is an ornithine decarboxylase inhibitor.
8594. The method of item 8521 wherein the agent is a p38 MAP kinase inhibitor selected from the group consisting of AZD-6703 (AstraZeneca), JX-401 (Jexys Pharmaceuticals), BMS-2 (Bristol-Myers Squibb), a p38 MAP kinase inhibitor from Novartis, a p38-alpha MAP kinase inhibitor from Amphora, Pharmaprojects No. 5704 (Pharmacopeia), RPR- 200765A (Sanofi-Aventis), SD-282 (Johnson & Johnson), TAK-715 (Takeda), and an analogue or derivative thereof.
8595. The method of item 8521 wherein the agent is a palmitoyl-protein thioesterase inhibitor.
8596. The method of item 8521 wherein the agent is a PDGF receptor kinase inhibitor selected from the group consisting^of AAL-993, .. AMN-107, or ABP-309 (Novartis), AMG-706 (Amgen), BAY-57-9352 (Bayer), CDP-860 (UCB), E-7080 (Eisai), imatinib (CAS No. 152459-95-5) (Novartis), OSI-930 (OSI Pharmaceuticals), RPR-127963E (Sanofi-Aventis), RWJ-540973 (Johnson & Johnson), sorafenib tosylate (Bayer), SU-11657 (Pfizer), tandutinib (CAS No. 387867-13-2) (Millennium Pharmaceuticals), vatalanib (Novartis), ZK-CDK (Schering AG), and an analogue or derivative thereof.
8597. The method of item 8521 wherein the agent is (-)- halofenate (Metabolex), AMG-131 (Amgen), antidiabetics from Japan Tobacco, AZD-4619, AZD-8450, AZD-8677 (AstraZeneca), DRF-10945, balaglitazone (Dr Reddy's), CS-00088, CS-00098 (Chipscreen Biosciences), E-3030 (Eisai), etalocib (CAS No. 161172-51-6) (EIi Lilly), GSK-641597 (Ligand), GSK-677954 (GlaxoSmithKline), GW-409544 (Ligand), GW- 590735 (GlaxoSmithKline), K-111 (Hoffmann-La Roche), LY-518674 (EIi Lilly), LY-674 (Ligand), LY-929 (Ligand), MC-3001 , MC-3002 (MaxoCore Pharmaceuticals), metformin HCI + pioglitazone (CAS No. 1115-70-4 and 112529-15-4), ACTOPLUS MET from Andrx), muraglitazar (CAS No. 331741-94-7) (Bristol-Myers Squibb), naveglitazar (Ligand), oleoylethanolamide (Kadmus Pharmaceuticals), ONO-5129, pioglitazone hydrochloride (CAS No. 111025-46-8 and 112529-15-4) (Takeda), PLX-204 (Plexxikon), PPAR agonists from Genfit, PPAR delta agonists from EIi Lilly, PPAR-alpha agonists from CrystalGenomics, PPAR-gamma modulators and PPAR-β modulators from CareX, rosiglitazone maleate (CAS No. 122324- 73-4 or 155141-29-0) (GlaxoSmithKline), rosiglitazone maleate/glimepir (CAS No. 155141-29-0 and 93479-97-1), AVANDARYL, rosiglitazone maleate/metformin extend (CAS No. 155141-29-0 and 657-24-9), AVANDAMET, rosiglitazone maleate+metformin, AVANDAMET (GlaxoSmithKline), tesaglitazar (AstraZeneca), and an analogue or derivative thereof.
8598. The method of item 8521 wherein the agent is a phosphatase inhibitor.
8599. The method of item 8521 wherein the agent is a phosphodiesterase (PDE) inhibitor selected from the group consisting of avanafil (Tanabe Seiyaku), dasantafil (CAS No. 569351-91-3) (Schering- Plough), A-906119 (CAS No. 134072-58-5), DL-850 (Sanofi-Aventis), GRC- 3015, GRC-3566, GRC-3886 (Glenmark), HWA-153 (CAS No. 56395-66-5) (Sanofi-Aventis), hydroxypumafentrine (Altana), IBFB-130011 , IBFB-14-016, IBFB-140301, IBFB-150007, IBFB-211913 (IBFB Pharma), L-826141 (Merck & Co), medorinone (CAS No. 88296-61-1) (Sanofi-Aventis), MEM- 1917 (Memory Pharmaceuticals), ND-1251 (Neuro3d), PDE inhibitors from ICOS, PDE IV inhibitors from Memory Pharmaceuticals and CrystalGenomics, Pharmaprojects No. 2742 and 6141 (Sanofi-Aventis), QAD-171 (Novartis), RHC-2963 (CAS No. 76993-12-9 and 76993-14-1), RPR-117658, RPR-122818 derivatives, SR-24870 , and RPR-132294 (Sanofi-Aventis), SK-350 (ln2Gen), stroke therapy agents from deCODE Genetics, TAS-203 (Taiho), tofimilast (CAS No. 185954-27-2) (Pfizer), UK- 371800 (Pfizer), WIN-65579 (CAS No. 158020-82-7) (Sanofi-Aventis), IBFB- 130020 (IBFB Pharma), OPC-6535 (CAS No. 145739-56-6) (Otsuka), a phosphodiesterase III inhibitor, enoximone, a phosphodiesterase IV inhibitor, fosfosal, Atopik (Barrier Therapeutics), triflusal, a phosphodiesterase V inhibitor, and an analogue or derivative thereof.
8600. The method of item 8521 wherein the agent is a PKC inhibitor.
8601. The method of item 8521 wherein the agent is a platelet activating factor antagonist.
8602. The method of item 8521 wherein the agent is a platelet-derived growth factor receptor kinase inhibitor.
8603. The method of item 8521 wherein the agent is a prolyl hydroxylase inhibitor.
8604. The method of item 8521 wherein the agent is a polymorphonuclear neutrophil inhibitor.
8605. The method of item 8521 wherein the agent is a protein kinase B inhibitor.
8606. The method of item 8521 wherein the agent is a protein kinase C stimulant. 8607. The method of item 8521 wherein the agent is a purine nucleoside analogue.
8608. The method of item 8521 wherein the agent is a purinoreceptor P2X antagonist.
8609. The method of item 8521 wherein the agent is a Raf kinase inhibitor.
8610. The method of item 8521 wherein the agent is a reversible inhibitor of ErbB1 and ErbB2.
8611. The method of item 8521 wherein the agent is a ribonucleoside triphosphate reductase inhibitor.
8612. The method of item 8521 wherein the agent is an SDF- 1 antagonist.
8613. The method of item 8521 wherein the agent is a sheddase inhibitor.
8614. The method of item 8521 wherein the agent is an SRC inhibitor.
8615. The method of item 8521 wherein the agent is a stromelysin inhibitor.
8616. The method of item 8521 wherein the agent is an Syk kinase inhibitor. 8617. The method of item 8521 wherein the agent is a telomerase inhibitor.
8618. The method of item 8521 wherein the agent is a TGF beta inhibitor selected from the group consisting of pirfenidone (CAS No. 53179-13-8) (MARNAC), tranilast (CAS No. 53902-12-8) (Kissei), IN-1130 (ln2Gen), mannose-6-phosphate (BTG), TGF-β antagonists from Inflazyme (Pharmaprojects No. 6075), TGF-β antagonists from Sydney, non-industrial source), TGF-β I receptor kinase inhibitors from EIi Lilly, TGF-β receptor inhibitors from Johnson & Johnson, and an analogue or derivative thereof.
8619. The method of item 8521 wherein the agent is a TNFα antagonist or TACE inhibitor selected from the group consisting of adalimumab (CAS No. 331731-18-1) (Cambridge Antibody Technology), AGIX-4207 (AtheroGenics), AGT-1 (Advanced Biotherapy), an antiinflammatory from Borean Pharma, Cellzome, or Paradigm-Therapeutics, anti-inflammatory vaccine (TNF-alpha kinoid) from Neovacs, humanized anti-TNF antibody or an anti-TNF MAb (CB0006) Celltech (UCB), apratastat (CAS No. 287445-51-0) (Wyeth), BMS-561392 (Bristol-Myers Squibb), BN- 006 (Bone), certolizumab pegol (CAS No. 428863-50-7 or CH-138 (UCB), cilomilast (CAS No. 153259-65-5) (GlaxoSmithKline), CR-1 (Nuada Pharmaceuticals), CRx-119 (CombinatoRx), D-5410 (UCB), dacopafant (CAS No. 125372-33-0) (Sanofi-Aventis), dersalazine (CAS No. 188913-57- 7/188913-58-8) (Uriach), etanercept (CAS No. 185243-69-0) (Amgen), ethyl pyruvate (Critical (Critical Therapeutics), golimumab (CAS No. 476181-74-5) (Johnson & Johnson), hormono-immunotherapy from Ipsen, CDP571 (e.g., Humicade from UCB), IC-485 (ICOS), infliximab (CAS No. 170277-31-3) (Johnson & Johnson), IP-751 (Manhattan Pharmaceuticals), ISIS-104838 (CAS No. 250755-32-9) (ISIS Pharmaceuticals), lenalidomide (CAS No. 191732-72-6) (Celgene), lentinan (CAS No. 37339-90-5) (Ajinomoto), MDL- 201112 (CAS No. 142130-73-2) (Sanofi-Aventis), medroxyprogesterone (CAS No. 520-85-4) (InKine Pharmaceutical), N-acetylcysteine (CAS No. 616-91-1) (Zambon), NBE-P2 (DIREVO Biotech), nerelimomab (CAS No. 162774-06-3) (Chiron), OM-294DP (OM PHARMA), onercept (CAS No. 199685-57-9) (Yeda), PASSTNF-alpha (Verigen), pentoxifylline or oxypentifylline (Sanofi-Aventis), Pharmaprojects No. 4091, 4241, 4295, or 4488 (Sanofi-Aventis), Pharmaprojects No. 5480 (Amgen), Pharmaprojects No. 6749 (Cengent), pirfenidone (CAS No. 53179-13-8) (MARNAC), RPR- 132294 (Sanofi-Aventis), S5 (F002) (Fulcrum Pharmaceuticals), simvastatin (CAS No. 79902-63-9) (Merck & Co), STA-6292 (Synta Pharmaceuticals), tacrolimus (CAS No. 104987-11-3) (Fujisawa LifeCycle Pharma), talactoferrin alfa (CAS No. 308240-58-6) (Agennix), thalidomide (CAS No. 50-35-1) (Celgene), TNF antagonists form ProStrakan, and Synergen, TNF inhibitors (Amgen), TNF-alpha antagonists from Dynavax Technologies and Jerina AG (Germany), TNF-alpha inhibitors from IBFB Pharma and Xencor (Xencor), torbafylline (CAS No. 105102-21-4) (Sanofi-Aventis), UR-1505 (Uriach), VT-346 (Viron Therapeutics), YSIL6 (Vs Therapeutics), YSTH2 (Y's Therapeutics), NPI-1302a-3 (Nereus Pharmaceuticals, a TNF antagonist from Jerina AG (Germany), dersalazine, and an analogue or derivative thereof.
8620. The method of item 8521 wherein the agent is a Toll receptor inhibitor.
8621. The method of item 8521 wherein the agent is a tubulin antagonist.
8622. The method of item 8521 wherein the agent is a tyrosine kinase inhibitor selected from the group consisting of SU-011248, SUTENT from Pfizer Inc. (New York, NY), BMS-354825, PN-355 (Paracelsian Pharmaceuticals), AGN-199659 (Allergan), AAL-993 or ABP- 309 (Novartis), adaphostin (NIH), AEE-788 (Novartis), AG-013736 (OSI Pharmaceuticals), AG-13736 (Pfizer), ALT-110 (Alteris Therapeutics), AMG- 706 (Amgen), anticancer MAbs from Xencor, anti-EGFrvlll MAbs from Abgenix, anti-HER2 MAb from Abiogen, AZD-2171 or AZD-9935 (AstraZeneca), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), CEP-5214 (Cephalon), CEP-7055 (Cephalon), cetuximab (ImCIone Systems), CHIR-200131 and CHIR-258 (Chiron), CP-547632 (OSI Pharmaceuticals), CP-724714 (Pfizer), CT-301 (Creabilis Therapeutics), D- 69491 (Baxter International), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EGFR/ErbB2 inhibitors from Array BioPharma, erlotinib (CAS No. 183319- 69-9) (OSI Pharmaceuticals), EXEL-2880 (Exelixis), FK-778 (Sanofi- Aventis) , gefitinib (CAS No. 184475-35-2) (AstraZeneca), GW-2286 or GW- 654652 (GlaxoSmithKline), her2/neu antigen from AlphaVax, HER-2/neu inhibitor from Generex, Herzyme (Medipad) (Sirna Therapeutics), HKI-272 (Wyeth), HuMax-EGFr (Genmab), idronoxil (CAS No. 81267-65-4) (Novogen), IGF-1 inhibitors from Ontogen, IMC-11 F8 (ImCIone Systems), kahalalide F (CAS No. 149204-42-2) (PharmaMar), KDR inhibitor from LG Life Sciences, KDR inhibitors from Abbott Laboratories, KDR kinase inhibitors (UCB), Kdr kinase inhibitors from Merck & Co, KRN-633 and KRN- 951 (Kirin Brewery), KSB-102 (Xenova), lapatinib ditosylate (CAS No. 388082-78-8) (GlaxoSmithKline), matuzumab (Merck KGaA), MDX-214 (Medarex), ME-103 (Pharmexa), MED-A300 (Gerolymatos), MNAC-13 (Lay Line Genomics), nimotuzumab (Center of Molecular Immunology), NSC- 330507 or NSC-707545 (NIH), NV-50 (Novogen), OSI-930 (OSI Pharmaceuticals), panitumumab (Abgenix), pelitinib (CAS No. 287933-82-7) (Wyeth), pertuzumab (CAS No. 380610-27-5) (Genentech), Pharmaprojects No. 3985 (Sanofi-Aventis), prostate cancer therapeutics from Sequenom (SQPC35, SQPC36, SQPC90), removab and remoxab (Trion Pharma), RG- 13022 (CAS No. 136831-48-6), RG-13291 (CAS No. 138989-50-1), or RG- 14620 (CAS No. 136831-49-7) (Sanofi-Aventis), RM-6427 (Romark), RNAi breast cancer therapy from Benitec, RP 53801 (CAS No. 125882-88-4) (Sanofi-Aventis), sorafenib tosylate (Bayer), SU-11657 (Pfizer), Tie-2 antagonists from Semaia (Hybrigenics), Tie-2 inhibitors from Ontogen, trastuzumab (CAS No. 180288-69-1) (Genentech), tyrosine kinase inhibitors from Sanofi-Aventis, U3-1287, U3-1565, U3-1784, U3-1800 (U3 Pharma), vatalanib (Novartis), VEGFR-2 kinase inhibitor from Bristol-Myers Squibb, XL-647 (Exelixis), ZD-6474 (AstraZeneca), ZK-CDK (Schering AG), an EGFR tyrosine kinase inhibitor, EKB-569 (Wyeth), and an analogue or derivative thereof.
8623. The method of item 8521 wherein the agent is a VEGF inhibitor.
8624. The method of item 8521 wherein the agent is a vitamin D receptor agonist.
8625. The method of item 8521 wherein the agent is ZD-6474 (an angiogenesis inhibitor). .
8626. The method of item 8521 wherein the agent is AP- 23573 (an mTOR inhibitor).
8627. The method of item 8521 wherein the agent is synthadotin (a tubulin antagonist).
8628. The method of item 8521 wherein the agent is S-0885 (a collagenase inhibitor).
8629. The method of item 8521 wherein the agent is aplidine (an elongation factor-1 alpha inhibitor).
8630. The method of item 8521 wherein the agent is ixabepilone (an epithilone). 8631. The method of item 8521 wherein the agent is IDN- 5390 (an angiogenesis inhibitor).
8632. The method of item 8521 wherein the agent is SB- 2723005 (an angiogenesis inhibitor).
8633. The method of item 8521 wherein the agent is ABT-518 (an angiogenesis inhibitor).
8634. The method of item 8521 wherein the agent is combretastatin (an angiogenesis inhibitor).
8635. The method of item 8521 wherein the agent is anecortave acetate (an angiogenesis inhibitor).
8636. The method of item 8521 wherein the agent is SB- . 715992 (a kinesin antagonist).
8637. The method of item 8521 wherein the agent is temsirolimus (an mTOR inhibitor).
8638. The method of item 8521 wherein the agent is adalimumab (a TNFα antagonist).
8639. The method of item 8521, wherein the composition comprises a polymer.
8640. The method of item 8521, wherein the composition comprises a polymer, and the polymer is, or comprises, a copolymer. 8641. The method of item 8521 , wherein the composition comprises a polymer, and the polymer is, or comprises, a block copolymer.
8642. The method of item 8521 , wherein the composition comprises a polymer, and the polymer is, or comprises, a random copolymer.
8643. The method of item 8521 , wherein the composition comprises a polymer, and the polymer is, or comprises, a biodegradable polymer.
8644. The method of item 8521, wherein the composition comprises a polymer, and the polymer is, or comprises, a nonbiodegradable polymer.
8645. The method of item 8521 , wherein the composition comprises a polymer, and the polymer is, or comprises, a hydrophilic polymer.
8646. The method of item 8521, wherein the composition comprises a polymer, and the polymer is, or comprises, a hydrophobic polymer.
8647. The method of item 8521 , wherein the composition comprises a polymer, and the polymer is, or comprises, a polymer having hydrophilic domains.
8648. The method of item 8521, wherein the composition comprises a polymer, and the polymer is, or comprises, a polymer having hydrophobic domains. 8649. The method of item 8521 , wherein the composition comprises a polymer, and the polymer is, or comprises, a non-conductive polymer.
8650. The method of item 8521 , wherein the composition comprises a polymer, and the polymer is, or comprises, an elastomer.
8651. The method of item 8521 , wherein the composition comprises a polymer, and the polymer is, or comprises, a hydrogel.
8652. The method of item 8521 , wherein the composition comprises a polymer, and the polymer is, or comprises, a silicone polymer.
8653. The method of item 8521 , wherein the composition comprises a polymer, and the polymer is, or comprises, a hydrocarbon polymer.
8654. The method of item 8521, wherein the composition comprises a polymer, and the polymer is, or comprises, a styrene-derived polymer.
8655. The method of item 8521 , wherein the composition comprises a polymer, and the polymer is, or comprises, a butadiene-derived polymer.
8656. The method of item 8521, wherein the composition comprises a polymer, and the polymer is, or comprises, a macromer.
8657. The method of item 8521, wherein the composition comprises a polymer, and the polymer is, or comprises, a poly(ethylene glycol) polymer. 8658. The method of item 8521 , wherein the composition comprises a polymer, and the polymer is, or comprises, an amorphous polymer.
8659. The method of item 8521, wherein the composition further comprises a second pharmaceutically active agent.
8660. The method of item 8521, wherein the composition further comprises an anti-inflammatory agent.
8661. The method of item 8521, wherein the composition further comprises an agent that inhibits infection.
8662. The method of item 8521, wherein the composition further comprises an anthracycline.
8663. The method of item 8521, wherein the composition further comprises doxorubicin.
8664. The method of item 8521 wherein the composition further comprises mitoxantrone.
8665. The method of item 8521 wherein the composition further comprises a fluoropyrimidine.
8666. The method of item 8521, wherein the composition further comprises 5-fluorouracil (5-FU).
8667. The method of item 8521, wherein the composition further comprises a folic acid antagonist. 8668. The method of item 8521, wherein the composition further comprises methotrexate.
8669. The method of item 8521 , wherein the composition further comprises a podophylotoxin.
8670. The method of item 8521, wherein the composition further comprises etoposide.
8671. The method of item 8521 , wherein the composition further comprises camptothecin.
8672. The method of item 8521 , wherein the composition further comprises a hydroxyurea.
8673. The method of item 8521 , wherein the composition further comprises a platinum complex.
8674. The method of item 8521, wherein the composition further comprises cisplatin.
8675. The method of item 8521 wherein the composition further comprises an anti-thrombotic agent.
8676. The method of item 8521 , wherein the composition further comprises a visualization agent.
8677. The method of item 8521 , wherein the composition further comprises a visualization agent, and the visualization agent is a radiopaque material, wherein the radiopaque material comprises a metal, a halogenated compound, or a barium containing compound. 8678. The method of item 8521 , wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, barium, tantalum, or technetium.
8679. The method of item 8521, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, an MRI responsive material.
8680. The method of item 8521, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, a gadolinium chelate.
8681. The method of item 8521 , wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, iron, magnesium, manganese, copper, or chromium.
8682. The method of item 8521 , wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, iron oxide compound.
8683. The method of item 8521 , wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, a dye, pigment, or colorant.
8684. The method of item 8521 wherein the agent is released in effective concentrations from the composition comprising the agent by diffusion over a period ranging from the time of administration to about 90 days.
8685. The method of item 8521 wherein the agent is released in effective concentrations from the composition comprising the agent by erosion of the composition over a period ranging from the time of administration to about 90 days.
8686. The method of item 8521 wherein the composition further comprises an inflammatory cytokine.
8687. The method of item 8521 wherein the composition further comprises an agent that stimulates cell proliferation.
8688. The method of item 8521 wherein the composition further comprises a polymeric carrier.
8689. The method of item 8521 wherein the composition is in the form of a gel, paste, or spray.
8690. The method of item 8521 wherein the implant is partially constructed with the agent or the composition.
8691. The method of item 8521 wherein the implant is fully constructed with the agent or the composition.
8692. The method of item 8521 wherein the implant is impregnated with the agent or the composition.
8693. The method of item 8521 , wherein the agent or the composition forms a coating, and the coating directly contacts the implant.
8694. The method of item 8521 , wherein the agent or the composition forms a coating, and the coating indirectly contacts the implant. 8695. The method of item 8521 wherein the agent or the composition forms a coating, and the coating partially covers the implant.
8696. The method of item 8521 , wherein the agent or the composition forms a coating, and the coating completely covers the implant.
8697. The method of item 8521 wherein the agent or the composition is located within pores or holes of the implant.
8698. The method of item 8521 wherein the agent or the composition is located within a channel, lumen, or divet of the implant.
8699. The method of item 8521 wherein the implant further comprising an echogenic material.
8700. The method of item 8521 wherein the implant further comprises an echogenic material, wherein the echogenic material is in the form of a coating.
8701. The method of item 8521 wherein the implant is sterile.
8702. The method of item 8521 wherein the agent is delivered from the implant, wherein the agent is released into tissue in the vicinity of the implant after deployment of the implant.
8703. The method of item 8521 wherein the agent is delivered from the implant, wherein the agent is released into tissue in the vicinity of the implant after deployment of the implant, wherein the tissue is connective tissue. 8704. The method of item 8521 wherein the agent is delivered from the implant, wherein the agent is released into tissue in the vicinity of the implant after deployment of the implant, wherein the tissue is muscle tissue.
8705. The method of item 8521 wherein the agent is delivered from the implant, wherein the agent is released into tissue in the vicinity of the implant after deployment of the implant, wherein the tissue is nerve tissue.
8706. The method of item 8521 wherein the agent is delivered from the implant, wherein the agent is released into tissue in the vicinity of the implant after deployment of the implant, wherein the tissue is epithelium tissue.
8707. The method of item 8521 wherein the agent is delivered from the implant, wherein the agent is released in effective concentrations from the implant over a period ranging from the time of deployment of the implant to about 1 year.
8708. The method of item 8521 wherein the agent is delivered from the implant, wherein the agent is released in effective concentrations from the implant over a period ranging from about 1 month to 6 months.
8709. The method of item 8521 wherein the agent is delivered from the implant, wherein the agent is released in effective concentrations from the implant over a period ranging from about 1 - 90 days.
8710. The method of item 8521 wherein the agent is delivered from the implant, wherein the agent is released in effective concentrations from the implant at a constant rate. 8711. The method of item 8521 wherein the agent is delivered from the implant, wherein the agent is released in effective concentrations from the implant at an increasing rate.
8712. The method of item 8521 wherein the agent is delivered from the implant, wherein the agent is released in effective concentrations from the implant at a decreasing rate.
8713. The method of item 8521 wherein the agent is delivered from the implant, wherein the implant comprises about 0.01 Dg to about 10 Dg of the agent.
8714. The method of item 8521 wherein the agent is delivered from the implant, wherein the implant comprises about 10 Dg to about 10 mg of the agent.
8715. The method of item 8521 wherein the agent is delivered from the implant, wherein the implant comprises about 10 mg to about 250 mg of the agent.
8716. The method of item 8521 wherein the agent is delivered from the implant, wherein the implant comprises about 250 mg to about
1000 mg of the agent.
8717. The method of item 8521 wherein the agent is delivered from the implant, wherein the implant comprises about 1000 mg to about 2500 mg of the agent.
8718. The method of item 8521 wherein the agent is delivered from the implant, wherein a surface of the implant comprises less than 0.01 Dg of the agent per mm2 of implant surface to which the agent is applied. 8719. The method of item 8521 wherein the agent is delivered from the implant, wherein a surface of the implant comprises about 0.01 Dg to about 1 Dg of the agent per mm2 of implant surface to which the agent is applied.
8720. The method of item 8521 wherein the agent is delivered from the implant, wherein a surface of the implant comprises about 1 Dg to about 10 Dg of the agent per mm2 of implant surface to which the agent is applied.
8721. The method of item 8521 wherein the agent is delivered from the implant, wherein a surface of the implant comprises about 10 Dg to about 250 Dg of the agent per mm2 of implant surface to which the agent is applied.
8722. The method of item 8521 wherein the agent is delivered from the implant, wherein a surface of the implant comprises about 250 Dg to about 1000 Dg of the agent per mm2 of implant surface to which the agent is applied.
8723. The method of item 8521 wherein the agent is delivered from the implant, wherein a surface of the implant comprises about 1000 Dg to about 2500 Dg of the agent per mm2 of implant surface to which the agent is applied.
8724. The method of item 8521, wherein the implant further comprises a coating, and the coating is a uniform coating.
8725. The method of item 8521 , wherein the implant further comprises a coating, and the coating is a non-uniform coating. 8726. The method of item 8521 , wherein the implant further comprises a coating, and the coating is a discontinuous coating.
8727. The method of item 8521, wherein the implant further comprises a coating, and the coating is a patterned coating.
8728. The method of item 8521, wherein the implant further comprises a coating, and the coating has a thickness of 100 Dm or less.
8729. The method of item 8521, wherein the implant further comprises a coating, and the coating has a thickness of 10 Dm or less.
8730. The method of item 8521 , wherein the implant further comprises a coating, and the coating adheres to the surface of the implant upon deployment of the implant.
8731. The method of item 8521, wherein the implant further comprises a coating, and the coating is stable at room temperature for a period of at least 1 year.
8732. The method of item 8521, wherein the implant further comprises a coating, and the agent is present in the coating in an amount ranging between about 0.0001% to about 1% by weight.
8733. The method of item 8521 , wherein the implant further comprises a coating, and the agent is present in the coating in an amount ranging between about 1 % to about 10% by weight.
8734. The method of item 8521 , wherein the implant further comprises a coating, and the agent is present in the coating in an amount ranging between about 10% to about 25% by weight. 8735. The method of item 8521 , wherein the implant further comprises a coating, and the agent is present in the coating in an amount ranging between about 25% to about 70% by weight.
8736. The method of item 8521 , wherein the implant further comprises a coating, and the coating comprises a polymer.
8737. The method of item 8521 , wherein the implant comprises a first coating having a first composition and a second coating having a second composition.
8738. The method of item 8521 , wherein the implant comprises a first coating having a first composition and a second coating having a second composition, wherein the first composition and the second composition are different.
8739. A method as in any one of items 8521-8738, wherein the device is a mechanical prosthetic heart valve.
8740. A method as in any one of items 8521-8738, wherein the device is a bioprosthetic heart valve.
8741. A method as in any one of items 8521-8738, wherein the device is an implantable annular ring for receiving a prosthetic heart valve.
8742. A method as in any one of items 8521-8738, wherein the device is a suture ring having an outer peripheral tapered thread for attaching a heart valve prosthesis. 8743. A method as in any one of items 8521-8738, wherein the device is a suture ring for a mechanical heart valve.
8744. A method for inhibiting scarring comprising placing a device that comprises a penile implant and an anti-scarring agent or a composition comprising an anti-scarring agent into an animal host, wherein the agent inhibits scarring.
8745. The method of item 8744 wherein the agent is an adensosine A2A receptor antagonist.
8746. The method of item 8744 wherein the agent is an AKT inhibitor.
8747. The method of item 8744 wherein the agent is an alpha 2 integrin antagonist, wherein the alpha 2 integrin antagonist is Pharmaprojects No. 5754 (Merck KgaA).
8748. The method of item 8744 wherein the agent is an alpha 4 integrin antagonist.
8749. The method of item 8744 wherein the agent is an alpha 7 nicotinic receptor agonist.
8750. The method of item 8744 wherein the agent is an angiogenesis inhibitor selected from the group consisting of AG-12,958 (Pfizer), ATN-161 (Attenuon LLC), neovastat, an angiogenesis inhibitor from Jerina AG (Germany), NM-3 (Mercian), VGA-1155 (Taisho), FCE-26644 (Pfizer), FCE-26950 (Pfizer), FPMA (Meiji Daries), FR-111142 (Fujisawa), GGTI-298, GM-1306 (Ligand), GPA-1734 (Novartis), NNC-47-0011 (Novo Nordisk), herbamycin (Nippon Kayaku), lenalidomide (Celegene), IP-10 (NIH), ABT-828 (Abbott), KIN-841 (Tokushima University, Japan), SF-1126 (Semafore Pharmaceuticals), laminin technology (NIH), CHIR-258 (Chiron), NVP-AEW541 (Novartis), NVP-AEW541 (Novartis), Vt16907 (Alchemia), OXI-8007 (Oxigene), EG-3306 (Ark Therapeutics), Maspin (Arriva), ABT- 567 (Abbott), PPI-2458 (Praecis Pharmaceuticals), CC-5079, CC-4089 (Celgene), HIF-1 alpha inhibitors (Xenova), S-247 (Pfizer), AP-23573 (Ariad), AZD-9935 (Astra Zeneca), mebendazole (Introgen Therapeutics), MetAP-2 inhibitors (GlaxoSmithKline), AG-615 (Angiogene Pharmaceuticals), Tie-2 antagonists (Hybrigenics), NC-381 , CYC-381, NC-169, NC-219, NC-383, NC-384, NC-407 (Lorus Therapeutics), ATN-224 (Attenuon), ON-01370 (Onconova), Vitronectin antagonists (Amgen), SDX-103 (Salmedix), Vitronectin antagonists (Shire), CHP (Riemser), TEK (Amgen), Anecortave acetate (Alcon), T46.2 (Matrix Therapeutics), HG-2 (Heptagen), TEM antagonists (Genzyme), Oxi-4500 (Oxigene), ATN-161 (Attenuon), WX-293 (Wilex), M-2025 (Metris Therapeutics), Alphastatin (BioActa), YH-16 (Yantai Rongchang), BIBF-1120 (Boehringer Ingelheim), BAY-57-9352 (Bayer), AS- 1404 (Cancer Research Technology), SC-77964 (Pfizer), glycomimetics (BioTie Therapies), TIE-2 Inhibitors (Ontogen), DIMI, Octamer (Octamer), ABR-215050 (Active Biotech), ABT-518 (Abbott), KDR inhibitors (Abbott), BSF-466895 (Abbott), SCH-221153 (Schering-Plough), DAC:antiangiogenic (ConjuChem), TFPI (EntreMed), AZD-2171 (Astra-Zenaca), CDC-394 (Celgene), LY290293 (EIi Lilly), IDN-5390 (Indena), Kdr Kinase Inhibitors (Merck), CT-113020, CT-116433, CT-116563, CT-31890, CT-32228) (Cell Therapeutics), A-299620 (Abbott), TWEAK Inhibitor (Amgen), VEGF modulators (Johnson and Johnson), Tum-N53, tumstatin (Genzyme), Thios- 1 , Thios-2 (Thios Pharmaceuticals), MV-6401 (Miravant Medical Technologies), Spisulosine (PharmaMar), CEP-7055 (Cephalon), AUV-201 (Auvation), LM-609 (EIi Lilly), SKF-106615 (AnorMED), Oglufanide disodium (Cytran), BW-114 (Pharminox), Calreticulin (NIH), WX-678 (Wilex), SD-7784 (Pfizer), WX-UK1 (Wilex), SH-268 (Schering AG), 2-Me-PGA (Celgene), S- 137 (Pfizer), ZD-6126 (Angiogene Pharmaceuticals), SG-292 (SignalGen), Benefin (Lane Labs), A6, A36 (Angstrom), SB-2723005 (GlaxoSmithKline), SC-7 (Cell Therapeutics), ZEN-014 (AEterna Zentaris), 2-methoxyestradiol (EntreMed), NK-130119 (Nippon Kayaku), CC-10004 (Celgene), AVE- 8062A (Ajinomoto), Tacedinaline (Pfizer), Actinonin (Tokyo Metropolitan Institute of Medical Science), Lenalidomide (Celgene), VGA-1155, BTO-956 (SRI International), ER-68203-00 (Eisai), CT-6685 (UCB), JKC-362 (Phoenix Pharmaceuticals), DMI-3798 (DMI Biosciences, Angiomate (Ipsen), ZD-6474 (AstraZeneca), CEP-5214 (Cephalon), Canstatin (Genzyme), NM-3 (Mercian), Oridigm (MediQuest Therapeutics), Exherin (Adherex), BLS-0597 (Boston Life Sciences), PTC-299 (PTC Therapeutics), NPI-2358 (Nereus Pharmaceuticals), CGP-79787 (Novartis), AEE-788 (Novartis), CKD-732 (Chong Kun Dang), CP-564959 (OSI Pharmaceuticals), CM-101 (CarboMed), CT-2584, CT3501 (Cell Therapeutics), combretastatin and analogues and derivatives thereof (Oxigene), Rebimastat (Bristol-Meyers Squibb), Dextrin 2-sulfate (ML Laboratories), Cilengitide (Merk KGaA), NSC-706704 (Pharminox), KRN- 951 (Kirin Brewery), Ukrain, NSC-631570 (Nowicky Pharma), Tecogalan sodium (Daiichi Pharmaceutical), Tz-93 (Tsumura), TBC-1635 (Encysive Pharmaceuticals), TAN-1120 (Takeda), Semaxanib (Pfizer), BDI-7800 (Biopharmacopae), SD-186, SD-983 (Bristol-Meyers Squibb), SB-223245 (GlaxoSmithKline), SC-236 (Pfizer), RWJ-590973 (Johnson and Johnson), ILX-1850 (Genzyme), SC-68488, S-836 (Pfizer), CG-55069-11 (CuraGen), Ki-23057 (Kirin Brewery), CCX-700 (Chemoentryx), Pegaptanib octasodium (Giled Sciences), ANGIOCOL (available from Biostratum Inc.), or an analogue or derivative thereof.
8751. The method of item 8744 wherein the agent is an apoptosis antagonist.
8752. The method of item 8744 wherein the agent is an apoptosis activator. 8753. The method of item 8744 wherein the agent is a beta 1 integrin antagonist.
8754. The method of item 8744 wherein the agent is a beta tubulin inhibitor.
8755. The method of item 8744 wherein the agent is a blocker of enzyme production in Hepatitis C.
8756. The method of item 8744 wherein the agent is a Bruton's tyrosine kinase inhibitor.
8757. The method of item 8744 wherein the agent is a calcineurin inhibitor.
8758. The method of item 8744 wherein the agent is a caspase 3 inhibitor.
8759. The method of item 8744 wherein the agent is a CC chemokine receptor antagonist.
8760. The method of item 8744 wherein the agent is a cell cycle inhibitor selected from the group consisting of SNS-595 (Sunesis), synthadotin, KRX-0403, and an analogue or derivative thereof.
8761. The method of item 8744 wherein the agent is a cathepsin B inhibitor.
8762. The method of item 8744 wherein the agent is a cathepsin K inhibitor, wherein the cathepsin K inhibitor is 462795 (GlaxoSmithKline), INPL-022-D6 (Amura Therapeutics), or an analogue or derivative thereof.
8763. The method of item 8744 wherein the agent is a cathepsin L inhibitor.
8764. The method of item 8744 wherein the agent is a CD40 antagonist.
8765. The method of item 8744 wherein the agent is a chemokine receptor agonist.
8766. The method of item 8744 wherein the agent is a chymase inhibitor.
8767. The method of item 8744 wherein the agent is a collagenase antagonist.
8768. The method of item 8744 wherein the agent is a CXCR antagonist.
8769. The method of item 8744 wherein the agent is a cyclin dependent kinase inhibitor selected from the group consisting of a CDK-1 inhibitor, a CDK-2 inhibitor, a CDK- 4 inhibitor, a CDK-6 inhibitor, a CAK1 inhibitor from GPC Biotech and Bristol-Myers Squibb, RGB-286199 (GPC Biotech), an anticancer agent from Astex Technology, a CAK1 inhibitor from GPC Biotech, a CDK inhibitor from Sanofi-Aventis, a CDK1/CDK2 inhibitor from Amgen, a CDK2 inhibitor from SUGEN-2 (Pfizer), a hearing loss therapy agent (Sound Pharmaceuticals), PD-0332991 (Pfizer), RGB-286199 (GPC Biotech), Ro-0505124 (Hoffmann-La Roche), a Ser/Thr kinase inhibitor from Lilly (EIi Lilly), CVT-2584 (CAS No. 199986-75-9) (CV Therapeutics), and an analogue or derivative thereof.
8770. The method of item 8744 wherein the agent is a cyclooxygenase 1 inhibitor.
8771. The method of item 8744 wherein the agent is a DHFR inhibitor.
8772. The method of item 8744 wherein the agent is a dual integrin inhibitor.
8773. The method of item 8744 wherein the agent is an elastase inhibitor.
8774. The method of item 8744 wherein the agent is an elongation factor-1 alpha inhibitor.
8775. The method of item 8744 wherein the agent is an endothelial growth factor antagonist.
8776. The method of item 8744 wherein the agent is an endothelial growth factor receptor kinase inhibitor selected from the group consisting of sorafenib tosylate (Bayer), AAL-993 (Novartis), ABP-309 (Novartis), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), E- 7080 (Eisai), EG-3306 (Ark Therapeutics), EXEL-2880 (Exelixis), GW- 654652 (GlaxoSmithKline), a KDR inhibitor from LG Life Sciences, CT-6685 and CT-6729 (UCB), KRN-633 and KRN-951 (Kirin Brewery), OSI-930 (OSI Pharmaceuticals), SP-5.2 (Supratek Pharma), SLM 1657 (Pfizer), a Tie-2 antagonist (Hybrigenics), a VEGFR-2 kinase inhibitor (Bristol-Myers Squibb), XL-647 (Exelixis), a KDR inhibitor from Abbott Laboratories, sorafenib tosylate, and an analogue or derivative thereof.
8777. The method of item 8744 wherein the agent is an endotoxin antagonist.
8778. The method of item 8744 wherein the agent is an epothilone and tubulin binder.
8779. The method of item 8744 wherein the agent is an estrogen receptor antagonist.
8780. The method of item 8744 wherein the agent is an FGF inhibitor.
8781. The method of item 8744 wherein the agent is a famexyl transferase inhibitor.
8782. The method of item 8744 wherein the agent is famesyltransferase inhibitor selected from the group of A-197574 (Abbott), a famesyltransferase inhibitor from Servier, FPTIII (Strathclyde Institute for Drug R), LB-42908 (LG Life Sciences), Pharmaprojects No. 5063 (Genzyme), Pharmaprojects No. 5597 (Ipsen), Yissum Project No. B-1055 (Yissum), and an analogue or derivative thereof.
8783. The method of item 8744 wherein the agent is an FLT-3 kinase inhibitor.
8784. The method of item 8744 wherein the agent is an FGF receptor kinase inhibitor. 8785. The method of item 8744 wherein the agent is a fibrinogen antagonist selected from the group consisting of AUV-201 (Auvation), MG-13926 (Sanofi-Aventis), plasminogen activator (CAS No. 105913-11-9) (from Sanofi-Aventis or UCB), plasminogen activator-2 (tPA- 2) (Sanofi-Aventis), pro-urokinase (CAS No. 82657-92-9) (Sanofi-Aventis), and an analogue or derivative thereof.
8786. The method of item 8744 wherein the agent is a heat shock protein 90 antagonist selected from the group consisting of SRN-005 (Sirenade), geldanamycin, NSC-33050 (17-allylaminogeldanamycin; 17- AAG), 17-dimethylaminoethylamino-17-demethoxy-geldanamycin (17- DMAG), rifabutin (rifamycin XIV, 1',4-didehydro-1-deoxy-1 ,4-dihydro-5'-(2- methylpropyl)-1-oxo-), and an analogue or derivative thereof.
8787. The method of item 8744 wherein the agent is a histone deacetylase inhibitor. . . .
8788. The method of item 8744 wherein the agent is an HMGCoA reductase inhibitor selected from the group consisting of an atherosclerosis therapeutic from Lipid Sciences, ATI-16000 (ARYx Therapeutics), KS-01-019 (Kos Pharmaceuticals), Pharmaprojects No. 2197 (Sanofi-Aventi), RP 61969 (Sanofi-Aventis), and an analogue or derivative thereof.
8789. The method of item 8744 wherein the agent is an ICAM inhibitor.
8790. The method of item 8744 wherein the agent is an IL, ICE and IRAK antagonist, wherein the antagonist is a CJ-14877, CP-424174 (Pfizer), NF-61 (Negma-Lerads), and an analogue or derivative thereof. 8791. The method of item 8744 wherein the agent is an IL-2 inhibitor.
8792. The method of item 8744 wherein the agent is an immunosuppressant selected from the group consisting of teriflunomide (Sanofi Aventis), chlorsulfaquinoxalone (NSC-339004), chlorsulfaquinoxalone sulfate, CS-712 (Sankyo), ismomultin alfa (CAS No. 457913-93-8) (Akzo Nobel), antiallergics from GenPat77, antiinflammatories or AT-005 (Androclus Therapeutics), autoimmune disease therapeutics from EpiVax, BN-007 (Bone), budesonide (CAS No. 51333-22- 3) (MAP Pharmaceuticals), CO-14 (Genzyme), edratide (CAS No. 433922- 67-9) (Teva), EP-314 (Enanta), eprovafen (CAS No. 101335-99-3) (Sanofi- Aventis), HWA-131 (CAS No. 118788-41-3) (Sanofi-Aventis), immunomodulators from MerLion Pharmaceuticals, immunosuppressives from Alchemia, IPL-12 (Inflazyme), MDL-9563 (CAS No. 27086-86-8) (Sanofi-Aventis), Pharmaprojects No. 2330 (Sanofi-Aventis), Pharmaprojects No. 6426 (Abgenix), PXS-25 (Pharmaxis), rosmarinic acid (CAS No. 20283-92-5) (Sanofi-Aventis), RP 42927 or RP 54745 (CAS No. 135330-08-4) (Sanofi-Aventis), SGN-35 (Seattle Genetics), ST-1959 (Sigma-Tau), type I diabetes therapy from SYNX Pharma, UNIL-88 (Debiopharm), VP-025 (Vasogen), VR-694 (Vectura), PRTX-001 (Protalex), and an analogue or derivative thereof.
8793. The method of item 8744 wherein the agent is an IMPDH (inosine monophosphate).
8794. The method of item 8744 wherein the agent is an integrin antagonist.
8795. The method of item 8744 wherein the agent is an interleukin antagonist. 8796. The method of item 8744 wherein the agent is an inhibitor of type III receptor tyrosine kinase.
8797. The method of item 8744 wherein the agent is an irreversible inhibitor of enzyme methionine aminopeptidase type 2.
8798. The method of item 8744 wherein the agent is an isozyme selective delta protein kinase C inhibitor.
8799. The method of item 8744 wherein the agent a JAK3 enzyme inhibitor.
8800. The method of item 8744 wherein the agent is a JNK inhibitor.
8801. The method of item 8744 wherein the agent is a kinase inhibitor.
8802. The method of item 8744 wherein the agent is kinesin antagonist.
8803. The method of item 8744 wherein the agent is a kinesin antagonist.
8804. The method of item 8744 wherein the agent is a leukotriene inhibitor and antagonist selected from the group consisting of ambicromil (CAS No. 58805-38-2) (Sanofi-Aventis), amelubant (CAS No. 346735-24-8) (Boehringer Ingelheim), DW-1141 (Dong Wha), ebselen (Daiichi Pharmaceutical), ibudilast (Kyorin), leucotriene inhibitors from Sanofi-Aventis, lymphotoxin-beta receptor (LT-D) from Biogen Idee, Pharmaprojects No. 1535 and 2728 (CAS No. 119340-33-9) (Sanofi- Aventis), R-112 (Rigel), Rev-5367 (CAS No. 92532-05-3) (Sanofi-Aventis), RG-14893 (CAS No. 141835-49-6) (Sanofi-Aventis), RG-5901-A (CAS No. 101910-24-1), 92532-23-5, RP 66153 (CAS No. 142422-79-5), RP 66364 (CAS No.186916-92-5), RP 69698 (CAS No. 141748-00-7) (Sanofi-Aventis), SC-411930 (Pfizer), SC-41930 (CAS No. 120072-59-5) (Pfizer), SC-50605 (CAS No. 138828-39-4) (Pfizer), SC-51146 (CAS No. 141059-52-1), SC- 53228 (CAS No. 153633-01-3) (Pfizer), spaglumic acid (ZY-15106) (CAS No.3106-85-2), 80619-64-3 (Novartis), tipredane (CAS No. 85197-77-9) (Bristol-Myers Squibb), U-75302 (CAS No. 119477-85-9) (Pfizer), and analogue or derivative thereof.
8805. The method of item 8744 wherein the agent is an MAP kinase inhibitor.
8806. The method of item 8744 wherein the agent is a matrix metalloproteinase inhibitor.
8807. The method of item 8744 wherein the agent is an MCP- CCR2 inhibitor.
8808. The method of item 8744 wherein the agent is an mTOR inhibitor.
8809. The method of item 8744 wherein the agent is an mTOR kinase inhibitor.
8810. The method of item 8744 wherein the agent is a microtubule inhibitor selected from the group consisting of antibody- maytansinoid conjugates from Biogen Idee, colchicines (MantiCore Pharmaceuticals), anticancer immunoconjugates from Johnson & Johnson, DIME from Octamer, gni-1f (GNI), huC242-DM4, huMy9-6-DM1 (ImmunoGen), IDN-5404 (Indena), IMO-098, IMOderm (Imotep), mebendazole (Introgen Therapeutics), microtubule poisons from Cambridge Enterprise, paclitaxel such as LOTAX from Aphios (CAS No. 33069-62-4), Genexol-PM from Samyang, Pharmaprojects No. 6383 (Tapestry Pharmaceuticals), RPR-112378 (Sanofi-Aventis), SGN-75 (Seattle Genetics), SPL-7435 (Starpharma), SSR-250411 (Sanofi-Aventis), trastuzumab-DM1 (Genentech), vinorelbine, and an analogue or derivative thereof.
8811. The method of item 8744 wherein the agent is an MIF inhibitor.
8812. The method of item 8744 wherein the agent is an MMP inhibitor.
8813: The method of item 8744 wherein the agent is a neurokinin (NK) antagonist selected from the group consisting of anthrotainin (CAS No. 148084-40-6) (Sanofi-Aventis), an IBS thereapeutic from ArQuIe, MDL-105212A (CAS No. 167261-60-1) (Ssanofi-Aventis), Pharmaprojects No. 2744, 3258 (CAS No. 139167-47-8) 4006, 4201 , or 5986 (Sanofi-Aventis), RP 67580 (CAS No. 135911-02-3), SR-144190 (CAS No. 201152-86-5), SSR-240600, SSR-241586 (Sanofi-Aventis), TKA-457 (Novartis), vestipitant mesylate (CAS No. 334476-64-1) (GlaxoSmithKline), Win-64821 (Sanofi-Aventis), PRX-96026 (Predix Pharmaceuticals), and an analogue or derivative thereof.
8814. The method of item 8744 wherein the agent is an NF kappa B inhibitor selected from the group consisting of AVE-0545 or AVE- 0547 (Sanofi-Aventis), bortezomib (CAS No. 179324-69-7) (Millennium Pharmaceuticals), dexanabinol (CAS No. 112924-45-5) (Pharmos), dexlipotam (Viatris), Pharmaprojects No. 6283 (INDRA) (OXiGENE), IPL- 576092 (CAS No. 137571-30-3) (Inflazyme), NFKB decoy (Corgentech), NFKB decoy oligo (AnGes MG), NFKB's from Ariad, osteoporosis treatments or S5 (F005) from Fulcrum Pharmaceuticals, P61 (Phytopharm), R-flurbiprofen (CAS No. 5104-49-4) (Encore Pharmaceuticals), and an analogue or derivative thereof.
8815. The method of item 8744 wherein the agent is a nitric oxide agonist.
8816. The method of item 8744 wherein the agent is an ornithine decarboxylase inhibitor.
8817. The method of item 8744 wherein the agent is a p38 MAP kinase inhibitor selected from the group consisting of AZD-6703 (AstraZeneca), JX-401 (Jexys Pharmaceuticals), BMS-2 (Bristol-Myers Squibb), a p38 MAP kinase inhibitor from Novartis, a p38-alpha MAP kinase inhibitor from Amphora, Pharmaprojects No. 5704 (Pharmacopeia), RPR- 200765A (Sanofi-Aventis), SD-282 (Johnson & Johnson), TAK-715 (Takeda), and an analogue or derivative thereof.
8818. The method of item 8744 wherein the agent is a palmitoyl-protein thioesterase inhibitor.
8819. The method of item 8744 wherein the agent is a PDGF receptor kinase inhibitor selected from the group consisting of AAL-993, AMN-107, or ABP-309 (Novartis), AMG-706 (Amgen), BAY-57-9352 (Bayer), CDP-860 (UCB), E-7080 (Eisai), imatinib (CAS No. 152459-95-5) (Novartis), OSI-930 (OSI Pharmaceuticals), RPR-127963E (Sanofi-Aventis), RWJ-540973 (Johnson & Johnson), sorafenib tosylate (Bayer), SU-11657 (Pfizer), tandutinib (CAS No. 387867-13-2) (Millennium Pharmaceuticals), vatalanib (Novartis), ZK-CDK (Schering AG)1 and an analogue or derivative thereof.
8820. The method of item 8744 wherein the agent is (-)- halofenate (Metabolex), AMG-131 (Amgen), antidiabetics from Japan Tobacco, AZD-4619, AZD-8450, AZD-8677 (AstraZeneca), DRF-10945, balaglitazone (Dr Reddy's), CS-00088, CS-00098 (Chipscreen Biosciences), E-3030 (Eisai), etalocib (CAS No. 161172-51-6) (EIi Lilly), GSK-641597 (Ligand), GSK-677954 (GlaxoSmithKline), GW-409544 (Ligand), GW- 590735 (GlaxoSmithKline), K-111 (Hoffmann-La Roche), LY-518674 (EIi Lilly), LY-674 (Ligand), LY-929 (Ligand), MC-3001 , MC-3002 (MaxoCore Pharmaceuticals), metformin HCI + pioglitazone (CAS No. 1115-70-4 and 112529-15-4), ACTOPLUS MET from Andrx), muraglitazar (CAS No. 331741-94-7) (Bristol-Myers Squibb), naveglitazar (Ligand), oleoylethanolamide (Kadmus Pharmaceuticals), ONO-5129, pioglitazone hydrochloride (CAS No. 111025-46-8 and 112529-15-4) (Takeda), PLX-204 (Plexxikon), PPAR agonists from Genfit, PPAR delta agonists from EIi Lilly, PPAR-alpha agonists from CrystalGenomics, PPAR-gamma modulators and PPAR-β modulators from CareX, rosiglitazone maleate (CAS No. 122324- 73-4 or 155141-29-0) (GlaxoSmithKline), rosiglitazone maleate/glimepir (CAS No. 155141-29-0 and 93479-97-1), AVANDARYL, rosiglitazone maleate/metformin extend (CAS No. 155141-29-0 and 657-24-9), AVANDAMET, rosiglitazone maleate+metformin, AVANDAMET (GlaxoSmithKline), tesaglitazar (AstraZeneca), and an analogue or derivative thereof.
8821. The method of item 8744 wherein the agent is a phosphatase inhibitor.
8822. The method of item 8744 wherein the agent is a phosphodiesterase (PDE) inhibitor selected from the group consisting of avanafil (Tanabe Seiyaku), dasantafil (CAS No. 569351-91-3) (Schering- Plough), A-906119 (CAS No. 134072-58-5), DL-850 (Sanofi-Aventis), GRC- 3015, GRC-3566, GRC-3886 (Glenmark), HWA-153 (CAS No. 56395-66-5) (Sanofi-Aventis), hydroxypumafentrine (Altana), IBFB-130011 , IBFB-14-016, IBFB-140301, IBFB-150007, IBFB-211913 (IBFB Pharma), L-826141 (Merck & Co), medorinone (CAS No. 88296-61-1) (Sanofi-Aventis), MEM- 1917 (Memory Pharmaceuticals), ND-1251 (Neuro3d), PDE inhibitors from ICOS, PDE IV inhibitors from Memory Pharmaceuticals and CrystalGenomics, Pharmaprojects No. 2742 and 6141 (Sanofi-Aventis), QAD-171 (Novartis), RHC-2963 (CAS No. 76993-12-9 and 76993-14-1), RPR-117658, RPR-122818 derivatives, SR-24870 , and RPR-132294 (Sanofi-Aventis), SK-350 (ln2Gen), stroke therapy agents from deCODE Genetics, TAS-203 (Taiho), tofimilast (CAS No. 185954-27-2) (Pfizer), UK- 371800 (Pfizer), WIN-65579 (CAS No. 158020-82-7) (Sanofi-Aventis), IBFB- 130020 (IBFB Pharma), OPC-6535 (CAS No. 145739-56-6) (Otsuka), a phosphodiesterase III inhibitor, enoximone, a phosphodiesterase IV inhibitor, fosfosal, Atopik (Barrier Therapeutics), triflusal, a phosphodiesterase V inhibitor, and an analogue or derivative thereof.
8823. The method of item 8744 wherein the agent is a PKC inhibitor.
8824. The method of item 8744 wherein the agent is a platelet activating factor antagonist.
8825. The method of item 8744 wherein the agent is a platelet-derived growth factor receptor kinase inhibitor.
8826. The method of item 8744 wherein the agent is a prolyl hydroxylase inhibitor. 8827. The method of item 8744 wherein the agent is a polymorphonuclear neutrophil inhibitor.
8828. The method of item 8744 wherein the agent is a protein kinase B inhibitor.
8829. The method of item 8744 wherein the agent is a protein kinase C stimulant.
8830. The method of item 8744 wherein the agent is a purine nucleoside analogue.
8831. The method of item 8744 wherein the agent is a purinoreceptor P2X antagonist.
8832. The method of item 8744 wherein the agent is a Raf kinase inhibitor.
8833. The method of item 8744 wherein the agent is a reversible inhibitor of ErbB1 and ErbB2.
8834. The method of item 8744 wherein the agent is a ribonucleoside triphosphate reductase inhibitor.
8835. The method of item 8744 wherein the agent is an SDF- 1 antagonist.
8836. The method of item 8744 wherein the agent is a sheddase inhibitor. 8837. The method of item 8744 wherein the agent is an SRC inhibitor.
8838. The method of item 8744 wherein the agent is a stromelysin inhibitor.
8839. The method of item 8744 wherein the agent is an Syk kinase inhibitor.
8840. The method of item 8744 wherein the agent is a telomerase inhibitor.
8841. The method of item 8744 wherein the agent is a TGF beta inhibitor selected from the group consisting of pirfenidone (CAS No. 53179-13-8) (MARNAC), tranilast (CAS No. 53902-12-8) (Kissei), IN-1130 (ln2Gen), mannose-6-phosphate (BTG), TGF-β antagonists from Inflazyme (Pharmaprojects No. 6075), TGF-β antagonists from Sydney, non-industrial source), TGF-β I receptor kinase inhibitors from EIi Lilly, TGF-β receptor inhibitors from Johnson & Johnson, and an analogue or derivative thereof.
8842. The method of item 8744 wherein the agent is a TNFα antagonist or TACE inhibitor selected from the group consisting of adalimumab (CAS No. 331731-18-1) (Cambridge Antibody Technology), AGIX-4207 (AtheroGenics), AGT-1 (Advanced Biotherapy), an antiinflammatory from Borean Pharma, Cellzome, or Paradigm Therapeutics, anti-inflammatory vaccine (TNF-alpha kinoid) from Neovacs, humanized anti-TNF antibody or an anti-TNF MAb (CB0006) Celltech (UCB), apratastat (CAS No. 287445-51-0) (Wyeth), BMS-561392 (Bristol-Myers Squibb), BN- 006 (Bone), certolizumab pegol (CAS No. 428863-50-7 or CH-138 (UCB), cilomilast (CAS No. 153259-65-5) (GlaxoSmithKline), CR-1 (Nuada Pharmaceuticals), CRx-119 (CombinatoRx), D-5410 (UCB), dacopafant (CAS No. 125372-33-0) (Sanofi-Aventis), dersalazine (CAS No. 188913-57- 7/188913-58-8) (Uriach), etanercept (CAS No. 185243-69-0) (Amgen), ethyl pyruvate (Critical (Critical Therapeutics), golimumab (CAS No. 476181-74-5) (Johnson & Johnson), hormono-immunotherapy from Ipsen, CDP571 (e.g., Humicade from UCB), IC-485 (ICOS), infliximab (CAS No. 170277-31-3) (Johnson & Johnson), IP-751 (Manhattan Pharmaceuticals), ISIS-104838 (CAS No. 250755-32-9) (ISIS Pharmaceuticals), lenalidomide (CAS No. 191732-72-6) (Celgene), lentinan (CAS No. 37339-90-5) (Ajinomoto), MDL- 201112 (CAS No. 142130-73-2) (Sanofi-Aventis), medroxyprogesterone (CAS No. 520-85-4) (InKine Pharmaceutical), N-acetylcysteine (CAS No. 616-91-1) (Zambon), NBE-P2 (DIREVO Biotech), nerelimomab (CAS No. 162774-06-3) (Chiron), OM-294DP (OM PHARMA), onercept (CAS No. 199685-57-9) (Yeda), PASSTNF-alpha (Verigen), pentoxifylline or oxypentifylline (Sanofi-Aventis), Pharmaprojects No. 4091 , 4241 , 4295, or 4488 (Sanofi-Aventis), Pharmaprojects No. 5480 (Amgen), Pharmaprojects No. 6749 (Cengent), pirfenidone (CAS No. 53179-13-8) (MARNAC), RPR- 132294 (Sanofi-Aventis), S5 (F002) (Fulcrum Pharmaceuticals), simvastatin (CAS No. 79902-63-9) (Merck & Co), STA-6292 (Synta Pharmaceuticals), tacrolimus (CAS No. 104987-11-3) (Fujisawa LifeCycle Pharma), talactoferrin alfa (CAS No. 308240-58-6) (Agennix), thalidomide (CAS No. 50-35-1) (Celgene), TNF antagonists form ProStrakan, and Synergen, TNF inhibitors (Amgen), TNF-alpha antagonists from Dynavax Technologies and Jerina AG (Germany), TNF-alpha inhibitors from IBFB Pharma and Xencor (Xencor), torbafylline (CAS No. 105102-21-4) (Sanofi-Aventis), UR-1505 (Uriach), VT-346 (Viron Therapeutics), YSIL6 (Ys Therapeutics), YSTH2 (Y's Therapeutics), NPI-1302a-3 (Nereus Pharmaceuticals, a TNF antagonist from Jerina AG (Germany), dersalazine, and an analogue or derivative thereof.
8843. The method of item 8744 wherein the agent is a Toll receptor inhibitor. 8844. The method of item 8744 wherein the agent is a tubulin antagonist.
8845. The method of item 8744 wherein the agent is a tyrosine kinase inhibitor selected from the group consisting of SU-011248, SUTENT from Pfizer Inc. (New York, NY), BMS-354825, PN-355 (Paracelsian Pharmaceuticals), AGN-199659 (Allergan), AAL-993 or ABP- 309 (Novartis), adaphostin (NIH), AEE-788 (Novartis), AG-013736 (OSI Pharmaceuticals), AG-13736 (Pfizer), ALT-110 (Alteris Therapeutics), AMG- 706 (Amgen), anticancer MAbs from Xencor, anti-EGFrvlll MAbs from Abgenix, anti-HER2 MAb from Abiogen, AZD-2171 or AZD-9935 (AstraZeneca), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), CEP-5214 (Cephalon), CEP-7055 (Cephalon), cetuximab (ImClone Systems), CHIR-200131 and CHIR-258 (Chiron), CP-547632 (OSI Pharmaceuticals), CP-724714 (Pfizer), CT-301 (Creabilis Therapeutics), D- 69491 (Baxter International), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EGFR/ErbB2 inhibitors from Array BioPharma, erlotinib (CAS No. 183319- 69-9) (OSI Pharmaceuticals), EXEL-2880 (Exelixis), FK-778 (Sanofi- Aventis) , gefitinib (CAS No. 184475-35-2) (AstraZeneca), GW-2286 or GW- 654652 (GlaxoSmithKline), her2/neu antigen from AlphaVax, HER-2/neu inhibitor from Generex, Herzyme (Medipad) (Sirna Therapeutics), HKI-272 (Wyeth), HuMax-EGFr (Genmab), idronoxil (CAS No. 81267-65-4) (Novogen), IGF-1 inhibitors from Ontogen, IMC-11F8 (ImClone Systems), kahalalide F (CAS No. 149204-42-2) (PharmaMar), KDR inhibitor from LG Life Sciences, KDR inhibitors from Abbott Laboratories, KDR kinase inhibitors (UCB), Kdr kinase inhibitors from Merck & Co, KRN-633 and KRN- 951 (Kirin Brewery), KSB-102 (Xenova), lapatinib ditosylate (CAS No. 388082-78-8) (GlaxoSmithKline), matuzumab (Merck KGaA), MDX-214 (Medarex), ME-103 (Pharmexa), MED-A300 (Gerolymatos), MNAC-13 (Lay Line Genomics), nimotuzumab (Center of Molecular Immunology), NSC- 330507 or NSC-707545 (NIH), NV-50 (Novogen), OSI-930 (OSI Pharmaceuticals), panitumumab (Abgenix), pelitinib (CAS No. 287933-82-7) (Wyeth), pertuzumab (CAS No. 380610-27-5) (Genentech), Pharmaprojects No. 3985 (Sanofi-Aventis), prostate cancer therapeutics from Sequenom (SQPC35, SQPC36, SQPC90), removab and remoxab (Trion Pharma), RG- 13022 (CAS No. 136831-48-6), RG-13291 (CAS No. 138989-50-1), or RG- 14620 (CAS No. 136831-49-7) (Sanofi-Aventis), RM-6427 (Romark), RNAi breast cancer therapy from Benitec, RP 53801 (CAS No. 125882-88-4) (Sanofi-Aventis), sorafenib tosylate (Bayer), SU-11657 (Pfizer), Tie-2 antagonists from Semaia (Hybrigenics), Tie-2 inhibitors from Ontogen, trastuzumab (CAS No. 180288-69-1) (Genentech), tyrosine kinase inhibitors from Sanofi-Aventis, U3-1287, U3-1565, U3-1784, U3-1800 (U3 Pharma), vatalanib (Novartis), VEGFR-2 kinase inhibitor from Bristol-Myers Squibb, XL-647 (Exelixis), ZD-6474 (AstraZeneca), ZK-CDK (Schering AG), an EGFR tyrosine kinase inhibitor, EKB-569 (Wyeth), and an analogue or derivative thereof.
8846. The method of item 8744 wherein the agent is a VEGF inhibitor.
8847. The method of item 8744 wherein the agent is a vitamin D receptor agonist.
8848. The method of item 8744 wherein the agent is ZD-6474 (an angiogenesis inhibitor).
8849. The method of item 8744 wherein the agent is AP- 23573 (an mTOR inhibitor).
8850. The method of item 8744 wherein the agent is synthadotin (a tubulin antagonist). 8851. The method of item 8744 wherein the agent is S-0885 (a collagenase inhibitor).
8852. The method of item 8744 wherein the agent is aplidine (an elongation factor-1 alpha inhibitor).
8853. The method of item 8744 wherein the agent is ixabepilone (an epithilone).
8854. The method of item 8744 wherein the agent is IDN- 5390 (an angiogenesis inhibitor).
8855. The method of item 8744 wherein the agent is SB- 2723005 (an angiogenesis inhibitor).
8856. The method of item 8744 wherein the agent is ABT-518 (an angiogenesis inhibitor).
8857. The method of item 8744 wherein the agent is combretastatin (an angiogenesis inhibitor).
8858. The method of item 8744 wherein the agent is anecortave acetate (an angiogenesis inhibitor).
8859. The method of item 8744 wherein the agent is SB- 715992 (a kinesin antagonist).
8860. The method of item 8744 wherein the agent is temsirolimus (an mTOR inhibitor). 8861. The method of item 8744 wherein the agent is adalimumab (a TNFα antagonist).
8862. The method of item 8744, wherein the composition comprises a polymer.
8863. The method of item 8744, wherein the composition comprises a polymer, and the polymer is, or comprises, a copolymer.
8864. The method of item 8744, wherein the composition comprises a polymer, and the polymer is, or comprises, a block copolymer.
8865. The method of item 8744, wherein the composition comprises a polymer, and the polymer is, or comprises, a random copolymer.
8866. The method of item 8744, wherein the composition comprises a polymer, and the polymer is, or comprises, a biodegradable polymer.
8867. The method of item 8744, wherein the composition comprises a polymer, and the polymer is, or comprises, a nonbiodegradable polymer.
8868. The method of item 8744, wherein the composition comprises a polymer, and the polymer is, or comprises, a hydrophilic polymer.
8869. The method of item 8744, wherein the composition comprises a polymer, and the polymer is, or comprises, a hydrophobic polymer. 8870. The method of item 8744, wherein the composition comprises a polymer, and the polymer is, or comprises, a polymer having hydrophilic domains.
8871. The method of item 8744, wherein the composition comprises a polymer, and the polymer is, or comprises, a polymer having hydrophobic domains.
8872. The method of item 8744, wherein the composition comprises a polymer, and the polymer is, or comprises, a non-conductive polymer.
8873. The method of item 8744, wherein the composition comprises a polymer, and the polymer is, or comprises, an elastomer.
8874. The method of item 8744, wherein the composition comprises a polymer, and the polymer is, or comprises, a hydrogel.
8875. The method of item 8744, wherein the composition comprises a polymer, and the polymer is, or comprises, a silicone polymer.
8876. The method of item 8744, wherein the composition comprises a polymer, and the polymer is, or comprises, a hydrocarbon polymer.
8877. The method of item 8744, wherein the composition comprises a polymer, and the polymer is, or comprises, a styrene-derived polymer. 8878. The method of item 8744, wherein the composition comprises a polymer, and the polymer is, or comprises, a butadiene-derived polymer.
8879. The method of item 8744, wherein the composition comprises a polymer, and the polymer is, or comprises, a macromer.
8880. The method of item 8744, wherein the composition comprises a polymer, and the polymer is, or comprises, a poly(ethylene glycol) polymer.
8881. The method of item 8744, wherein the composition comprises a polymer, and the polymer is, or comprises, an amorphous polymer.
8882. The method of item 8744, wherein the composition further comprises a second pharmaceutically active agent.
8883. The method of item 8744, wherein the composition further comprises an anti-inflammatory agent.
8884. The method of item 8744, wherein the composition further comprises an agent that inhibits infection.
8885. The method of item 8744, wherein the composition further comprises an anthracycline.
8886. The method of item 8744, wherein the composition further comprises doxorubicin. 8887. The method of item 8744 wherein the composition further comprises mitoxantrone.
8888. The method of item 8744 wherein the composition further comprises a fluoropyrimidine.
8889. The method of item 8744, wherein the composition further comprises 5-fluorouracil (5-FU).
8890. The method of item 8744, wherein the composition further comprises a folic acid antagonist.
8891. The method of item 8744, wherein the composition further comprises methotrexate.
8892. The method of item 8744, wherein the composition further comprises a podophylotoxin.
8893. The method of item 8744, wherein the composition further comprises etoposide.
8894. The method of item 8744, wherein the composition further comprises camptothecin.
8895. The method of item 8744, wherein the composition further comprises a hydroxyurea.
8896. The method of item 8744, wherein the composition further comprises a platinum complex. 8897. The method of item 8744, wherein the composition further comprises cisplatin.
8898. The method of item 8744 wherein the composition further comprises an anti-thrombotic agent.
8899. The method of item 8744, wherein the composition further comprises a visualization agent.
8900. The method of item 8744, wherein the composition further comprises a visualization agent, and the visualization agent is a radiopaque material, wherein the radiopaque material comprises a metal, a halogenated compound, or a barium containing compound.
8901. The method of item 8744, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, barium, tantalum, or technetium.
8902. The method of item 8744, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, an MRI responsive material.
8903. The method of item 8744, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, a gadolinium chelate.
8904. The method of item 8744, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, iron, magnesium, manganese, copper, or chromium. 8905. The method of item 8744, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, iron oxide compound.
8906. The method of item 8744, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, a dye, pigment, or colorant.
8907. The method of item 8744 wherein the agent is released in effective concentrations from the composition comprising the agent by diffusion over a period ranging from the time of administration to about 90 days.
8908. The method of item 8744 wherein the agent is released in effective concentrations from the composition comprising the agent by erosion of the composition over a period ranging from the time of administration to about 90 days.
8909. The method of item 8744 wherein the composition further comprises an inflammatory cytokine.
8910. The method of item 8744 wherein the composition further comprises an agent that stimulates cell proliferation.
8911. The method of item 8744 wherein the composition further comprises a polymeric carrier.
8912. The method of item 8744 wherein the composition is in the form of a gel, paste, or spray. 8913. The method of item 8744 wherein the implant is partially constructed with the agent or the composition.
8914. The method of item 8744 wherein the implant is fully constructed with the agent or the composition.
8915. The method of item 8744 wherein the implant is impregnated with the agent or the composition.
8916. The method of item 8744, wherein the agent or the composition forms a coating, and the coating directly contacts the implant.
8917. The method of item 8744, wherein the agent or the composition forms a coating, and the coating indirectly contacts the implant.
8918. The method of item 8744 wherein the agent or the - composition forms a coating, and the coating partially covers the implant.
8919. The method of item 8744, wherein the agent or the composition forms a coating, and the coating completely covers the implant.
8920. The method of item 8744 wherein the agent or the composition is located within pores or holes of the implant.
8921. The method of item 8744 wherein the agent or the composition is located within a channel, lumen, or divet of the implant.
8922. The method of item 8744 wherein the implant further comprising an echogenic material. 8923. The method of item 8744 wherein the implant further comprises an echogenic material, wherein the echogenic material is in the form of a coating.
8924. The method of item 8744 wherein the implant is sterile.
8925. The method of item 8744 wherein the agent is delivered from the implant, wherein the agent is released into tissue in the vicinity of the implant after deployment of the implant.
8926. The method of item 8744 wherein the agent is delivered from the implant, wherein the agent is released into tissue in the vicinity of the implant after deployment of the implant, wherein the tissue is connective tissue.
8927. The method of item 8744 wherein the agent is delivered from the implant, wherein the agent is released into tissue in the vicinity of the implant after deployment of the implant, wherein the tissue is muscle tissue.
8928. The method of item 8744 wherein the agent is delivered from the implant, wherein the agent is released into tissue in the vicinity of the implant after deployment of the implant, wherein the tissue is nerve tissue.
8929. The method of item 8744 wherein the agent is delivered from the implant, wherein the agent is released into tissue in the vicinity of the implant after deployment of the implant, wherein the tissue is epithelium tissue. 8930. The method of item 8744 wherein the agent is delivered from the implant, wherein the agent is released in effective concentrations from the implant over a period ranging from the time of deployment of the implant to about 1 year.
8931. The method of item 8744 wherein the agent is delivered from the implant, wherein the agent is released in effective concentrations from the implant over a period ranging from about 1 month to 6 months.
8932. The method of item 8744 wherein the agent is delivered from the implant, wherein the agent is released in effective concentrations from the implant over a period ranging from about 1 - 90 days.
8933. The method of item 8744 wherein the agent is delivered from the implant, wherein the agent is released in effective concentrations from the implant at a constant rate. . . .. __ „_
8934. The method of item 8744 wherein the agent is delivered from the implant, wherein the agent is released in effective concentrations from the implant at an increasing rate.
8935. The method of item 8744 wherein the agent is delivered from the implant, wherein the agent is released in effective concentrations from the implant at a decreasing rate.
8936. The method of item 8744 wherein the agent is delivered from the implant, wherein the implant comprises about 0.01 Dg to about 10 Dg of the agent. 8937. The method of item 8744 wherein the agent is delivered from the implant, wherein the implant comprises about 10 Dg to about 10 mg of the agent.
8938. The method of item 8744 wherein the agent is delivered from the implant, wherein the implant comprises about 10 mg to about 250 mg of the agent.
8939. The method of item 8744 wherein the agent is delivered from the implant, wherein the implant comprises about 250 mg to about 1000 mg of the agent.
8940. The method of item 8744 wherein the agent is delivered from the implant, wherein the implant comprises about 1000 mg to about 2500 mg of the agent.
8941. The method of item 8744 wherein the agent is delivered from the implant, wherein a surface of the implant comprises less than 0.01 Dg of the agent per mm2 of implant surface to which the agent is applied.
8942. The method of item 8744 wherein the agent is delivered from the implant, wherein a surface of the implant comprises about 0.01 Dg to about 1 Dg of the agent per mm2 of implant surface to which the agent is applied.
8943. The method of item 8744 wherein the agent is delivered from the implant, wherein a surface of the implant comprises about 1 Dg to about 10 Dg of the agent per mm2 of implant surface to which the agent is applied. 8944. The method of item 8744 wherein the agent is delivered from the implant, wherein a surface of the implant comprises about 10 Dg to about 250 Dg of the agent per mm2 of implant surface to which the agent is applied.
8945. The method of item 8744 wherein the agent is delivered from the implant, wherein a surface of the implant comprises about 250 Dg to about 1000 Dg of the agent per mm2 of implant surface to which the agent is applied.
8946. The method of item 8744 wherein the agent is delivered from the implant, wherein a surface of the implant comprises about 1000 Dg to about 2500 Dg of the agent per mm2 of implant surface to which the agent is applied.
8947. The method of item 8744, wherein the-implant further comprises a coating, and the coating is a uniform coating.
8948. The method of item 8744, wherein the implant further comprises a coating, and the coating is a non-uniform coating.
8949. The method of item 8744, wherein the implant further comprises a coating, and the coating is a discontinuous coating.
8950. The method of item 8744, wherein the implant further comprises a coating, and the coating is a patterned coating.
8951. The method of item 8744, wherein the implant further comprises a coating, and the coating has a thickness of 100 Dm or less. 8952. The method of item 8744, wherein the implant further comprises a coating, and the coating has a thickness of 10 Dm or less.
8953. The method of item 8744, wherein the implant further comprises a coating, and the coating adheres to the surface of the implant upon deployment of the implant.
8954. The method of item 8744, wherein the implant further comprises a coating, and the coating is stable at room temperature for a period of at least 1 year.
8955. The method of item 8744, wherein the implant further comprises a coating, and the agent is present in the coating in an amount ranging between about 0.0001% to about 1 % by weight.
8956. The method of item 8744, wherein the implant further comprises a coating, and the agent is present in the coating in an amount ranging between about 1% to about 10% by weight.
8957. The method of item 8744, wherein the implant further comprises a coating, and the agent is present in the coating in an amount ranging between about 10% to about 25% by weight.
8958. The method of item 8744, wherein the implant further comprises a coating, and the agent is present in the coating in an amount ranging between about 25% to about 70% by weight.
8959. The method of item 8744, wherein the implant further comprises a coating, and the coating comprises a polymer. 8960. The method of item 8744, wherein the implant comprises a first coating having a first composition and a second coating having a second composition.
8961. The method of item 8744, wherein the implant comprises a first coating having a first composition and a second coating having a second composition, wherein the first composition and the second composition are different.
8962. A method as in any one of items 8744-8961 , wherein the device is a penile implant that is a flexible rod.
8963. A method as in any one of items 8744-8961, wherein the device is a penile implant that is a hinged rod.
8964. A method as in any one of items 8744-8961, wherein the device is a penile implant that is an inflatable device with a pump.
8965. A method for inhibiting scarring comprising placing a device that comprises an endotracheal or tracheostomy tube (e.g., an implant) and an anti-scarring agent or composition comprising an anti- scarring agent into an animal host, wherein the agent inhibits scarring.
8966. The method of item 8965 wherein the agent is an adensosine A2A receptor antagonist.
8967. The method of item 8965 wherein the agent is an AKT inhibitor. 8968. The method of item 8965 wherein the agent is an alpha 2 integrin antagonist, wherein the alpha 2 integrin antagonist is Pharmaprojects No. 5754 (Merck KgaA).
8969. The method of item 8965 wherein the agent is an alpha 4 integrin antagonist.
8970. The method of item 8965 wherein the agent is an alpha 7 nicotinic receptor agonist.
8971. The method of item 8965 wherein the agent is an angiogenesis inhibitor selected from the group consisting of AG-12,958 (Pfizer), ATN-161 (Attention LLC), neovastat, an angiogenesis inhibitor from Jerina AG (Germany), NM-3 (Mercian), VGA-1155 (Taisho), FCE-26644 (Pfizer), FCE-26950 (Pfizer), FPMA (Meiji Daries), FR-111142 (Fujisawa), GGTI-298, GM-1306 (Ligand), GPA-1734 (Novartis), NNC-47-001-1 (Novo Nordisk), herbamycin (Nippon Kayaku), lenalidomide (Celegene), IP-10 (NIH), ABT-828 (Abbott), KIN-841 (Tokushima University, Japan), SF-1126 (Semafore Pharmaceuticals), laminin technology (NIH), CHIR-258 (Chiron), NVP-AEW541 (Novartis), NVP-AEW541 (Novartis), Vt16907 (Alchemia), OXI-8007 (Oxigene), EG-3306 (Ark Therapeutics), Maspin (Arriva), ABT- 567 (Abbott), PPI-2458 (Praecis Pharmaceuticals), CC-5079, CC-4089 (Celgene), HIF-1 alpha inhibitors (Xenova), S-247 (Pfizer), AP-23573 (Ariad), AZD-9935 (Astra Zeneca), mebendazole (Introgen Therapeutics), MetAP-2 inhibitors (GlaxoSmithKline), AG-615 (Angiogene Pharmaceuticals), Tie-2 antagonists (Hybrigenics), NC-381, CYC-381 , NC-169, NC-219, NC-383, NC-384, NC-407 (Lorus Therapeutics), ATN-224 (Attenuon), ON-01370 (Onconova), Vitronectin antagonists (Amgen), SDX-103 (Salmedix), Vitronectin antagonists (Shire), CHP (Riemser), TEK (Amgen), Anecortave acetate (Alcon), T46.2 (Matrix Therapeutics), HG-2 (Heptagen), TEM antagonists (Genzyme), Oxi-4500 (Oxigene), ATN-161 (Attenuon), WX-293 (Wilex), M-2025 (Metris Therapeutics), Alphastatin (BioActa), YH-16 (Yantai Rongchang), BIBF-1120 (Boehringer Ingelheim), BAY-57-9352 (Bayer), AS- 1404 (Cancer Research Technology), SC-77964 (Pfizer), glycomimetics (BioTie Therapies), TIE-2 Inhibitors (Ontogen), DIMI, Octamer (Octamer), ABR-215050 (Active Biotech), ABT-518 (Abbott), KDR inhibitors (Abbott), BSF-466895 (Abbott), SCH-221153 (Schering-Plough), DAC:antiangiogenic (ConjuChem), TFPI (EntreMed), AZD-2171 (Astra-Zenaca), CDC-394 (Celgene), LY290293 (EIi Lilly), IDN-5390 (Indena), Kdr Kinase Inhibitors (Merck), CT-113020, CT-116433, CT-116563, CT-31890, CT-32228) (Cell Therapeutics), A-299620 (Abbott), TWEAK Inhibitor (Amgen), VEGF modulators (Johnson and Johnson), Tum-N53, tumstatin (Genzyme), Thios- 1, Thios-2 (Thios Pharmaceuticals), MV-6401 (Miravant Medical Technologies), Spisulosine (PharmaMar), CEP-7055 (Cephalon), AUV-201 (Auvation), LM-609 (EIi Lilly), SKF-106615 (AnorMED), Oglufanide disodium (Cytran), BW-114 (Pharminox), Calreticulin (NIH), WX-678 (Wilex), SD-7784 (Pfizer), WX-UK1 (Wilex), SH-268 (Schering AG), 2-Me-PGA (Celgene), S- 137 (Pfizer), ZD-6126 (Angiogene Pharmaceuticals), SG-292 (SignalGen), Benefin (Lane Labs), A6, A36 (Angstrom), SB-2723005 (GlaxoSmithKline), SC-7 (Cell Therapeutics), ZEN-014 (AEterna Zentaris), 2-methoxyestradiol (EntreMed), NK-130119 (Nippon Kayaku), CC-10004 (Celgene), AVE- 8062A (Ajinomoto), Tacedinaline (Pfizer), Actinonin (Tokyo Metropolitan Institute of Medical Science), Lenalidomide (Celgene), VGA-1155, BTO-956 (SRI International), ER-68203-00 (Eisai), CT-6685 (UCB), JKC-362 (Phoenix Pharmaceuticals), DMI-3798 (DMI Biosciences, Angiomate (Ipsen), ZD-6474 (AstraZeneca), CEP-5214 (Cephalon), Canstatin (Genzyme), NM-3 (Mercian), Oridigm (MediQuest Therapeutics), Exherin (Adherex), BLS-0597 (Boston Life Sciences), PTC-299 (PTC Therapeutics), NPI-2358 (Nereus Pharmaceuticals), CGP-79787 (Novartis), AEE-788 (Novartis), CKD-732 (Chong Kun Dang), CP-564959 (OSI Pharmaceuticals), CM-101 (CarboMed), CT-2584, CT3501 (Cell Therapeutics), combretastatin and analogues and derivatives thereof (Oxigene), Rebimastat (Bristol-Meyers Squibb), Dextrin 2-sulfate (ML Laboratories), Cilengitide (Merk KGaA)1 NSC-706704 (Pharminox), KRN- 951 (Kirin Brewery), Ukrain, NSC-631570 (Nowicky Pharma), Tecogalan sodium (Daiichi Pharmaceutical), Tz-93 (Tsumura), TBC-1635 (Encysive Pharmaceuticals), TAN-1120 (Takeda), Semaxanib (Pfizer), BDI-7800 (Biopharmacopae), SD-186, SD-983 (Bristol-Meyers Squibb), SB-223245 (GlaxoSmithKline), SC-236 (Pfizer), RWJ-590973 (Johnson and Johnson), ILX-1850 (Genzyme), SC-68488, S-836 (Pfizer), CG-55069-11 (CuraGen), Ki-23057 (Kirin Brewery), CCX-700 (Chemoentryx), Pegaptanib octasodium (Giled Sciences), ANGIOCOL (available from Biostratum Inc.), or an analogue or derivative thereof.
8972. The method of item 8965 wherein the agent is an apoptosis antagonist.
8973. The method of item 8965 wherein the agent is an apoptosis activator.
8974. The method of item 8965 wherein the agent is a beta 1 integrin antagonist.
8975. The method of item 8965 wherein the agent is a beta tubulin inhibitor.
8976. The method of item 8965 wherein the agent is a blocker of enzyme production in Hepatitis C.
8977. The method of item 8965 wherein the agent is a Bruton's tyrosine kinase inhibitor. 8978. The method of item 8965 wherein the agent is a calcineurin inhibitor.
8979. The method of item 8965 wherein the agent is a caspase 3 inhibitor.
8980. The method of item 8965 wherein the agent is a CC chemokine receptor antagonist.
8981. The method of item 8965 wherein the agent is a cell cycle inhibitor selected from the group consisting of SNS-595 (Sunesis), synthadotin, KRX-0403, and an analogue or derivative thereof.
8982. The method of item 8965 wherein the agent is a cathepsin B inhibitor.
8983. The method of item 8965 wherein the agent is a cathepsin K inhibitor, wherein the cathepsin K inhibitor is 462795 (GlaxoSmithKline), INPL-022-D6 (Amura Therapeutics), or an analogue or derivative thereof.
8984. The method of item 8965 wherein the agent is a cathepsin L inhibitor.
8985. The method of item 8965 wherein the agent is a CD40 antagonist.
8986. The method of item 8965 wherein the agent is a chemokine receptor agonist. 8987. The method of item 8965 wherein the agent is a chymase inhibitor.
8988. The method of item 8965 wherein the agent is a collagenase antagonist.
8989. The method of item 8965 wherein the agent is a CXCR antagonist.
8990. The method of item 8965 wherein the agent is a cyclin dependent kinase inhibitor selected from the group consisting of a CDK-1 inhibitor, a CDK-2 inhibitor, a CDK- 4 inhibitor, a CDK-6 inhibitor, a CAK1 inhibitor from GPC Biotech and Bristol-Myers Squibb, RGB-286199 (GPC Biotech), an anticancer agent from Astex Technology, a CAK1 inhibitor from GPC Biotech, a CDK inhibitor from Sanofi-Aventis, a CDK1/CDK2 inhibitor from Amgen, a CDK2 inhibitor from SUGEN-2 (Pfizer), a hearing loss therapy agent (Sound Pharmaceuticals), PD-0332991 (Pfizer), RGB-286199 (GPC Biotech), Ro-0505124 (Hoffmann-La Roche), a Ser/Thr kinase inhibitor from Lilly (EIi Lilly), CVT-2584 (CAS No. 199986-75-9) (CV Therapeutics), and an analogue or derivative thereof.
8991. The method of item 8965 wherein the agent is a cyclooxygenase 1 inhibitor.
8992. The method of item 8965 wherein the agent is a DHFR inhibitor.
8993. The method of item 8965 wherein the agent is a dual integrin inhibitor. 8994. The method of item 8965 wherein the agent is an elastase inhibitor.
8995. The method of item 8965 wherein the agent is an elongation factor-1 alpha inhibitor.
8996. The method of item 8965 wherein the agent is an endothelial growth factor antagonist.
8997. The method of item 8965 wherein the agent is an endothelial growth factor receptor kinase inhibitor selected from the group consisting of sorafenib tosylate (Bayer), AAL-993 (Novartis), ABP-309 (Novartis), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), E- 7080 (Eisai), EG-3306 (Ark Therapeutics), EXEL-2880 (Exelixis), GW- 654652 (GlaxoSmithKline), a KDR inhibitor from LG Life Sciences, CT-6685 and CT-6729 (UCB), KRN-633 and KRN-951 (Kirin Brewery), OSL930 (OSI Pharmaceuticals), SP-5.2 (Supratek Pharma), SU-11657 (Pfizer), a Tie-2 antagonist (Hybrigenics), a VEGFR-2 kinase inhibitor (Bristol-Myers Squibb), XL-647 (Exelixis), a KDR inhibitor from Abbott Laboratories, sorafenib tosylate, and an analogue or derivative thereof.
8998. The method of item 8965 wherein the agent is an endotoxin antagonist.
8999. The method of item 8965 wherein the agent is an epothilone and tubulin binder.
9000. The method of item 8965 wherein the agent is an estrogen receptor antagonist. 9001. The method of item 8965 wherein the agent is an FGF inhibitor.
9002. The method of item 8965 wherein the agent is a farnexyl transferase inhibitor.
9003. The method of item 8965 wherein the agent is farnesyltransferase inhibitor selected from the group of A-197574 (Abbott), a farnesyltransferase inhibitor from Servier, FPTIII (Strathclyde Institute for Drug R), LB-42908 (LG Life Sciences), Pharmaprojects No. 5063 (Genzyme), Pharmaprojects No. 5597 (Ipsen), Yissum Project No. B-1055 (Yissum), and an analogue or derivative thereof.
9004. The method of item 8965 wherein the agent is an FLT-3 kinase inhibitor.
9005. The method of item 8965 wherein the agent is an FGF receptor kinase inhibitor.
9006. The method of item 8965 wherein the agent is a fibrinogen antagonist selected from the group consisting of AUV-201 (Auvation), MG-13926 (Sanofi-Aventis), plasminogen activator (CAS No. 105913-11-9) (from Sanofi-Aventis or UCB), plasminogen activator-2 (tPA- 2) (Sanofi-Aventis), pro-urokinase (CAS No. 82657-92-9) (Sanofi-Aventis), and an analogue or derivative thereof.
9007. The method of item 8965 wherein the agent is a heat shock protein 90 antagonist selected from the group consisting of SRN-005 (Sirenade), geldanamycin, NSC-33050 (17-allylaminogeldanamycin; 17- AAG), 17-dimethylaminoethylamino-17-demethoxy-geldanamycin (17- DMAG), rifabutin (rifamycin XIV, 1',4-didehydro-1-deoxy-1 ,4-dihydro-5'-(2- methylpropyl)-1-oxo-), and an analogue or derivative thereof.
9008. The method of item 8965 wherein the agent is a histone deacetylase inhibitor.
9009. The method of item 8965 wherein the agent is an HMGCoA reductase inhibitor selected from the group consisting of an atherosclerosis therapeutic from Lipid Sciences, ATI-16000 (ARYx Therapeutics), KS-01-019 (Kos Pharmaceuticals), Pharmaprojects No. 2197 (Sanofi-Aventi), RP 61969 (Sanofi-Aventis), and an analogue or derivative thereof.
9010. The method of item 8965 wherein the agent is an ICAM inhibitor.
9011. The method of item 8965 wherein the agent is an IL, ICE and IRAK antagonist, wherein the antagonist is a CJ-14877, CP-424174 (Pfizer), NF-61 (Negma-Lerads), and an analogue or derivative thereof.
9012. The method of item 8965 wherein the agent is an IL-2 inhibitor.
9013. The method of item 8965 wherein the agent is an immunosuppressant selected from the group consisting of teriflunomide (Sanofi Aventis), chlorsulfaquinoxalone (NSC-339004), chlorsulfaquinoxalone sulfate, CS-712 (Sankyo), ismomultin alfa (CAS No. 457913-93-8) (Akzo Nobel), antiallergics from GenPat77, antiinflammatories or AT-005 (Androclus Therapeutics), autoimmune disease therapeutics from EpiVax, BN-007 (Bone), budesonide (CAS No. 51333-22- 3) (MAP Pharmaceuticals), CO-14 (Genzyme), edratide (CAS No. 433922- 67-9) (Teva), EP-314 (Enanta), eprovafen (CAS No. 101335-99-3) (Sanofi- Aventis), HWA-131 (CAS No. 118788-41-3) (Sanofi-Aventis), immunomodulators from MerLion Pharmaceuticals, immunosuppressives from Alchemia, IPL-12 (Inflazyme), MDL-9563 (CAS No. 27086-86-8) (Sanofi-Aventis), Pharmaprojects No. 2330 (Sanofi-Aventis), Pharmaprojects No. 6426 (Abgenix), PXS-25 (Pharmaxis), rosmarinic acid (CAS No. 20283-92-5) (Sanofi-Aventis), RP 42927 or RP 54745 (CAS No. 135330-08-4) (Sanofi-Aventis), SGN-35 (Seattle Genetics), ST-1959 (Sigma-Tau), type I diabetes therapy from SYNX Pharma, UNIL-88 (Debiopharm), VP-025 (Vasogen), VR-694 (Vectura), PRTX-001 (Protalex), and an analogue or derivative thereof.
9014. The method of item 8965 wherein the agent is an IMPDH (inosine monophosphate).
9015. The method of item 8965 wherein the agent is an integrin antagonist.
9016. The method of item 8965 wherein the agent is an interleukin antagonist.
9017. The method of item 8965 wherein the agent is an inhibitor of type III receptor tyrosine kinase.
9018. The method of item 8965 wherein the agent is an irreversible inhibitor of enzyme methionine aminopeptidase type 2.
9019. The method of item 8965 wherein the agent is an isozyme selective delta protein kinase C inhibitor. 9020. The method of item 8965 wherein the agent a JAK3 enzyme inhibitor.
9021. The method of item 8965 wherein the agent is a JNK inhibitor.
9022. The method of item 8965 wherein the agent is a kinase inhibitor.
9023. The method of item 8965 wherein the agent is kinesin antagonist.
9024. The method of item 8965 wherein the agent is a kinesin antagonist.
9025. The method of item 8965 wherein the agent is a leukotriene inhibitor and antagonist selected from the group consisting of ambicromil (CAS No. 58805-38-2) (Sanofi-Aventis), amelubant (CAS No. 346735-24-8) (Boehringer Ingelheim), DW-1141 (Dong Wha), ebselen (Daiichi Pharmaceutical), ibudilast (Kyorin), leucotriene inhibitors from Sanofi-Aventis, lymphotoxin-beta receptor (LT-D) from Biogen Idee, Pharmaprojects No. 1535 and 2728 (CAS No. 119340-33-9) (Sanofi- Aventis), R-112 (Rigel), Rev-5367 (CAS No. 92532-05-3) (Sanofi-Aventis), RG-14893 (CAS No. 141835-49-6) (Sanofi-Aventis), RG-5901-A (CAS No. 101910-24-1), 92532-23-5, RP 66153 (CAS No. 142422-79-5), RP 66364 (CAS No.186916-92-5), RP 69698 (CAS No. 141748-00-7) (Sanofi-Aventis), SC-411930 (Pfizer), SC-41930 (CAS No. 120072-59-5) (Pfizer), SC-50605 (CAS No. 138828-39-4) (Pfizer), SC-51146 (CAS No. 141059-52-1), SC- 53228 (CAS No. 153633-01-3) (Pfizer), spaglumic acid (ZY-15106) (CAS No.3106-85-2), 80619-64-3 (Novartis), tipredane (CAS No. 85197-77-9) (Bristol-Myers Squibb), U-75302 (CAS No. 119477-85-9) (Pfizer), and analogue or derivative thereof.
9026. The method of item 8965 wherein the agent is an MAP kinase inhibitor.
9027. The method of item 8965 wherein the agent is a matrix metalloproteinase inhibitor.
9028. The method of item 8965 wherein the agent is an MCP- CCR2 inhibitor.
9029. The method of item 8965 wherein the agent is an mTOR inhibitor.
9030. The method of item 8965 wherein the agent is an mTOR kinase inhibitor.
9031. The method of item 8965 wherein the agent is a microtubule inhibitor selected from the group consisting of antibody- maytansinoid conjugates from Biogen Idee, colchicines (MantiCore Pharmaceuticals), anticancer immunoconjugates from Johnson & Johnson, DIME from Octamer, gni-1f (GNI), huC242-DM4, huMy9-6-DM1 (ImmunoGen), IDN-5404 (Indena), IMO-098, IMOderm (Imotep), mebendazole (Introgen Therapeutics), microtubule poisons from Cambridge Enterprise, paclitaxel such as LOTAX from Aphios (CAS No. 33069-62-4), Genexol-PM from Samyang, Pharmaprojects No. 6383 (Tapestry Pharmaceuticals), RPR-112378 (Sanofi-Aventis), SGN-75 (Seattle Genetics), SPL-7435 (Starpharma), SSR-250411 (Sanofi-Aventis), trastuzumab-DM1 (Genentech), vinorelbine, and an analogue or derivative thereof. 9032. The method of item 8965 wherein the agent is an MIF inhibitor.
9033. The method of item 8965 wherein the agent is an MMP inhibitor.
9034. The method of item 8965 wherein the agent is a neurokinin (NK) antagonist selected from the group consisting of anthrotainin (CAS No. 148084-40-6) (Sanofi-Aventis), an IBS thereapeutic from ArQuIe, MDL-105212A (CAS No. 167261-60-1) (Ssanofi-Aventis), Pharmaprojects No. 2744, 3258 (CAS No. 139167-47-8) 4006, 4201 , or 5986 (Sanofi-Aventis), RP 67580 (CAS No. 135911-02-3), SR-144190 (CAS No. 201152-86-5), SSR-240600, SSR-241586 (Sanofi-Aventis), TKA-457 (Novartis), vestipitant mesylate (CAS No. 334476-64-1) (GlaxoSmithKline), Win-64821 (Sanofi-Aventis), PRX-96026 (Predix Pharmaceuticals), and an analogue or derivative thereof. -
9035. The method of item 8965 wherein the agent is an NF kappa B inhibitor selected from the group consisting of AVE-0545 or AVE- 0547 (Sanofi-Aventis), bortezomib (CAS No. 179324-69-7) (Millennium Pharmaceuticals), dexanabinol (CAS No. 112924-45-5) (Pharmos), dexlipotam (Viatris), Pharmaprojects No. 6283 (INDRA) (OXiGENE), IPL- 576092 (CAS No. 137571-30-3) (Inflazyme), NFKB decoy (Corgentech), NFKB decoy oligo (AnGes MG), NFKB's from Ariad, osteoporosis treatments or S5 (F005) from Fulcrum Pharmaceuticals, P61 (Phytopharm), R-flurbiprofen (CAS No. 5104-49-4) (Encore Pharmaceuticals), and an analogue or derivative thereof.
9036. The method of item 8965 wherein the agent is a nitric oxide agonist. 9037. The method of item 8965 wherein the agent is an ornithine decarboxylase inhibitor.
9038. The method of item 8965 wherein the agent is a p38 MAP kinase inhibitor selected from the group consisting of AZD-6703 (AstraZeneca), JX-401 (Jexys Pharmaceuticals), BMS-2 (Bristol-Myers Squibb), a p38 MAP kinase inhibitor from Novartis, a p38-alpha MAP kinase inhibitor from Amphora, Pharmaprojects No. 5704 (Pharmacopeia), RPR- 200765A (Sanofi-Aventis), SD-282 (Johnson & Johnson), TAK-715 (Takeda), and an analogue or derivative thereof.
9039. The method of item 8965 wherein the agent is a palmitoyl-protein thioesterase inhibitor.
9040. The method of item 8965 wherein the agent is a PDGF receptor kinase inhibitor selected from the group consisting of AAL-993, AMN-107, or ABP-309 (Novartis), AMG-706 (Amgen), BAY-57-9352 (Bayer), CDP-860 (UCB), E-7080 (Eisai), imatinib (CAS No. 152459-95-5) (Novartis), OSI-930 (OSl Pharmaceuticals), RPR-127963E (Sanofi-Aventis), RWJ-540973 (Johnson & Johnson), sorafenib tosylate (Bayer), SU-11657 (Pfizer), tandutinib (CAS No. 387867-13-2) (Millennium Pharmaceuticals), vatalanib (Novartis), ZK-CDK (Schering AG), and an analogue or derivative thereof.
9041. The method of item 8965 wherein the agent is (-)- halofenate (Metabolex), AMG-131 (Amgen), antidiabetics from Japan Tobacco, AZD-4619, AZD-8450, AZD-8677 (AstraZeneca), DRF-10945, balaglitazone (Dr Reddy's), CS-00088, CS-00098 (Chipscreen Biosciences), E-3030 (Eisai), etalocib (CAS No. 161172-51-6) (EIi Lilly), GSK-641597 (Ligand), GSK-677954 (GlaxoSmithKIine), GW-409544 (Ligand), GW- 590735 (GlaxoSmithKIine), K-111 (Hoffmann-La Roche), LY-518674 (EIi Lilly), LY-674 (Ligand), LY-929 (Ligand), MC-3001 , MC-3002 (MaxoCore Pharmaceuticals), metformin HCI + pioglitazone (CAS No. 1115-70-4 and 112529-15-4), ACTOPLUS MET from Andrx), muraglitazar (CAS No. 331741-94-7) (Bristol-Myers Squibb), naveglitazar (Ligand), oleoylethanolamide (Kadmus Pharmaceuticals), ONO-5129, pioglitazone hydrochloride (CAS No. 111025-46-8 and 112529-15-4) (Takeda), PLX-204 (Plexxikon), PPAR agonists from Genfit, PPAR delta agonists from EIi Lilly, PPAR-alpha agonists from CrystalGenomics, PPAR-gamma modulators and PPAR-β modulators from CareX, rosiglitazone maleate (CAS No. 122324- 73-4 or 155141-29-0) (GlaxoSmithKline), rosiglitazone maleate/glimepir (CAS No. 155141-29-0 and 93479-97-1), AVANDARYL, rosiglitazone maleate/metformin extend (CAS No. 155141-29-0 and 657-24-9), AVANDAMET, rosiglitazone maleate+metformin, AVANDAMET (GlaxoSmithKline), tesaglitazar (AstraZeneca), and an analogue or derivative thereof.
9042. The method of item 8965 wherein the agent is a phosphatase inhibitor.
9043. The method of item 8965 wherein the agent is a phosphodiesterase (PDE) inhibitor selected from the group consisting of avanafil (Tanabe Seiyaku), dasantafil (CAS No. 569351-91-3) (Schering- Plough), A-906119 (CAS No. 134072-58-5), DL-850 (Sanofi-Aventis), GRC- 3015, GRC-3566, GRC-3886 (Glenmark), HWA-153 (CAS No. 56395-66-5) (Sanofi-Aventis), hydroxypumafentrine (Altana), IBFB-130011, IBFB-14-016, IBFB-140301, IBFB-150007, IBFB-211913 (IBFB Pharma), L-826141 (Merck & Co), medorinone (CAS No. 88296-61-1) (Sanofi-Aventis), MEM- 1917 (Memory Pharmaceuticals), ND-1251 (Neuro3d), PDE inhibitors from ICOS, PDE IV inhibitors from Memory Pharmaceuticals and CrystalGenomics, Pharmaprojects No. 2742 and 6141 (Sanofi-Aventis), QAD-171 (Novartis), RHC-2963 (CAS No. 76993-12-9 and 76993-14-1), RPR-117658, RPR-122818 derivatives, SR-24870 , and RPR-132294 (Sanofi-Aventis), SK-350 (ln2Gen), stroke therapy agents from deCODE Genetics, TAS-203 (Taiho), tofimilast (CAS No. 185954-27-2) (Pfizer), UK- 371800 (Pfizer), WIN-65579 (CAS No. 158020-82-7) (Sanofi-Aventis), IBFB- 130020 (IBFB Pharma), OPC-6535 (CAS No. 145739-56-6) (Otsuka), a phosphodiesterase III inhibitor, enoximone, a phosphodiesterase IV inhibitor, fosfosal, Atopik (Barrier Therapeutics), triflusal, a phosphodiesterase V inhibitor, and an analogue or derivative thereof.
9044. The method of item 8965 wherein the agent is a PKC inhibitor.
9045. The method of item 8965 wherein the agent is a platelet activating factor antagonist.
9046. The method of item 8965 wherein the agent is a _ platelet-derived growth factor receptor kinase inhibitor.
9047. The method of item 8965 wherein the agent is a prolyl hydroxylase inhibitor.
9048. The method of item 8965 wherein the agent is a polymorphonuclear neutrophil inhibitor.
9049. The method of item 8965 wherein the agent is a protein kinase B inhibitor.
9050. The method of item 8965 wherein the agent is a protein kinase C stimulant. 9051. The method of item 8965 wherein the agent is a purine nucleoside analogue.
9052. The method of item 8965 wherein the agent is a purinoreceptor P2X antagonist.
9053. The method of item 8965 wherein the agent is a Raf kinase inhibitor.
9054. The method of item 8965 wherein the agent is a reversible inhibitor of ErbB1 and ErbB2.
9055. The method of item 8965 wherein the agent is a ribonucleoside triphosphate reductase inhibitor.
9056. The method of item 8965 wherein the agent is an SDF- 1 antagonist.
9057. The method of item 8965 wherein the agent is a sheddase inhibitor.
9058. The method of item 8965 wherein the agent is an SRC inhibitor.
9059. The method of item 8965 wherein the agent is a stromelysin inhibitor.
9060. The method of item 8965 wherein the agent is an Syk kinase inhibitor. 9061. The method of item 8965 wherein the agent is a telomerase inhibitor.
9062. The method of item 8965 wherein the agent is a TGF beta inhibitor selected from the group consisting of pirfenidone (CAS No. 53179-13-8) (MARNAC), tranilast (CAS No. 53902-12-8) (Kissei), IN-1130 (ln2Gen), mannose-6-phosphate (BTG), TGF-β antagonists from Inflazyme (Pharmaprojects No. 6075), TGF-β antagonists from Sydney, non-industrial source), TGF-β I receptor kinase inhibitors from EIi Lilly, TGF-β receptor inhibitors from Johnson & Johnson, and an analogue or derivative thereof.
9063. The method of item 8965 wherein the agent is a TNFα antagonist or TACE inhibitor selected from the group consisting of adalimumab (CAS No. 331731-18-1) (Cambridge Antibody Technology), AGIX-4207 (AtheroGenics), AGT-1 (Advanced Biotherapy), an antiinflammatory from Borean Pharma, Cellzome, or Paradigm Therapeutics, anti-inflammatory vaccine (TNF-alpha kinoid) from Neovacs, humanized anti-TNF antibody or an anti-TNF MAb (CB0006) Celltech (UCB), apratastat (CAS No. 287445-51-0) (Wyeth), BMS-561392 (Bristol-Myers Squibb), BN- 006 (Bone), certolizumab pegol (CAS No. 428863-50-7 or CH-138 (UCB), cilomilast (CAS No. 153259-65-5) (GlaxoSmithKline), CR-1 (Nuada Pharmaceuticals), CRx-119 (CombinatoRx), D-5410 (UCB), dacopafant (CAS No. 125372-33-0) (Sanofi-Aventis), dersalazine (CAS No. 188913-57- 7/188913-58-8) (Uriach), etanercept (CAS No. 185243-69-0) (Amgen), ethyl pyruvate (Critical (Critical Therapeutics), golimumab (CAS No. 476181-74-5) (Johnson & Johnson), hormono-immunotherapy from Ipsen, CDP571 (e.g., Humicade from UCB), IC-485 (ICOS), infliximab (CAS No. 170277-31-3) (Johnson & Johnson), IP-751 (Manhattan Pharmaceuticals), ISIS-104838 (CAS No. 250755-32-9) (ISIS Pharmaceuticals), lenalidomide (CAS No. 191732-72-6) (Celgene), lentinan (CAS No. 37339-90-5) (Ajinomoto), MDL- 201112 (CAS No. 142130-73-2) (Sanofi-Aventis), medroxyprogesterone (CAS No. 520-85-4) (InKine Pharmaceutical), N-acetylcysteine (CAS No. 616-91-1) (Zambon), NBE-P2 (DIREVO Biotech), nerelimomab (CAS No. 162774-06-3) (Chiron), OM-294DP (OM PHARMA), onercept (CAS No. 199685-57-9) (Yeda), PASSTNF-alpha (Verigen), pentoxifylline or oxypentifylline (Sanofi-Aventis), Pharmaprojects No. 4091 , 4241 , 4295, or 4488 (Sanofi-Aventis), Pharmaprojects No. 5480 (Amgen), Pharmaprojects No. 6749 (Cengent), pirfenidone (CAS No. 53179-13-8) (MARNAC), RPR- 132294 (Sanofi-Aventis), S5 (F002) (Fulcrum Pharmaceuticals), simvastatin (CAS No. 79902-63-9) (Merck & Co), STA-6292 (Synta Pharmaceuticals), tacrolimus (CAS No. 104987-11-3) (Fujisawa LifeCycle Pharma), talactoferrin alfa (CAS No. 308240-58-6) (Agennix), thalidomide (CAS No. 50-35-1) (Celgene), TNF antagonists form ProStrakan, and Synergen, TNF inhibitors (Amgen), TNF-alpha antagonists from Dynavax Technologies and Jerina AG (Germany), TNF-alpha inhibitors from IBFB Pharma and Xencor (Xencor), torbafylline (CAS No. 105102-21-4) (Sanofi-Aventis), UR-1505 (Uriach), VT-346 (Viron Therapeutics), YSIL6 (Y1S Therapeutics), YSTH2 . . (Y's Therapeutics), NPI-1302a-3 (Nereus Pharmaceuticals, a TNF antagonist from Jerina AG (Germany), dersalazine, and an analogue or derivative thereof.
9064. The method of item 8965 wherein the agent is a Toll receptor inhibitor.
9065. The method of item 8965 wherein the agent is a tubulin antagonist.
9066. The method of item 8965 wherein the agent is a tyrosine kinase inhibitor selected from the group consisting of SU-011248, SUTENT from Pfizer Inc. (New York, NY), BMS-354825, PN-355 (Paracelsian Pharmaceuticals), AGN-199659 (Allergan), AAL-993 or ABP- 309 (Novartis), adaphostin (NIH), AEE-788 (Novartis), AG-013736 (OSI Pharmaceuticals), AG-13736 (Pfizer), ALT-110 (Alteris Therapeutics), AMG- 706 (Amgen), anticancer MAbs from Xencor, anti-EGFrvlll MAbs from Abgenix, anti-HER2 MAb from Abiogen, AZD-2171 or AZD-9935 (AstraZeneca), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), CEP-5214 (Cephalon), CEP-7055 (Cephalon), cetuximab (ImCIone Systems), CHIR-200131 and CHIR-258 (Chiron), CP-547632 (OSI Pharmaceuticals), CP-724714 (Pfizer), CT-301 (Creabilis Therapeutics), D- 69491 (Baxter International), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EGFR/ErbB2 inhibitors from Array BioPharma, erlotinib (CAS No. 183319- 69-9) (OSI Pharmaceuticals), EXEL-2880 (Exelixis), FK-778 (Sanofi- Aventis) , gefitinib (CAS No. 184475-35-2) (AstraZeneca), GW-2286 or GW- 654652 (GlaxoSmithKline), her2/neu antigen from AlphaVax, HER-2/neu inhibitor from Generex, Herzyme (Medipad) (Sirna Therapeutics), HKI-272 (Wyeth), HuMax-EGFr (Genmab), idronoxil (CAS No. 81267-65-4) (Novogen), IGF-1 inhibitors from Ontogen, IMC-11 F8 (ImCIone Systems), kahalalide F (CAS No. 149204-42-2) (PharmaMar), KDR inhibitor from LG Life Sciences, KDR inhibitors from Abbott Laboratories, KDR kinase inhibitors (UCB), Kdr kinase inhibitors from Merck & Co, KRN-633 and KRN- 951 (Kirin Brewery), KSB-102 (Xenova), lapatinib ditosylate (CAS No. 388082-78-8) (GlaxoSmithKline), matuzumab (Merck KGaA), MDX-214 (Medarex), ME-103 (Pharmexa), MED-A300 (Gerolymatos), MNAC-13 (Lay Line Genomics), nimotuzumab (Center of Molecular Immunology), NSC- 330507 or NSC-707545 (NIH), NV-50 (Novogen), OSI-930 (OSI Pharmaceuticals), panitumumab (Abgenix), pelitinib (CAS No. 287933-82-7) (Wyeth), pertuzumab (CAS No. 380610-27-5) (Genentech), Pharmaprojects No. 3985 (Sanofi-Aventis), prostate cancer therapeutics from Sequenom (SQPC35, SQPC36, SQPC90), removab and remoxab (Trion Pharma), RG- 13022 (CAS No. 136831-48-6), RG-13291 (CAS No. 138989-50-1), or RG- 14620 (CAS No. 136831-49-7) (Sanofi-Aventis), RM-6427 (Romark), RNAi breast cancer therapy from Benitec, RP 53801 (CAS No. 125882-88-4) (Sanofi-Aventis), sorafenib tosylate (Bayer), SU-11657 (Pfizer), Tie-2 antagonists from Semaia (Hybrigenics), Tie-2 inhibitors from Ontogen, trastuzumab (CAS No. 180288-69-1) (Genentech), tyrosine kinase inhibitors from Sanofi-Aventis, U3-1287, U3-1565, U3-1784, U3-1800 (U3 Pharma), vatalanib (Novartis), VEGFR-2 kinase inhibitor from Bristol-Myers Squibb, XL-647 (Exelixis), ZD-6474 (AstraZeneca), ZK-CDK (Schering AG), an EGFR tyrosine kinase inhibitor, EKB-569 (Wyeth), and an analogue or derivative thereof.
9067. The method of item 8965 wherein the agent is a VEGF inhibitor.
9068. The method of item 8965 wherein the agent is a vitamin D receptor agonist.
9069. The method of item 8965 wherein the agent is ZD-6474 (an angiogenesis inhibitor). - _ . .. .
9070. The method of item 8965 wherein the agent is AP- 23573 (an imTOR inhibitor).
9071. The method of item 8965 wherein the agent is synthadotin (a tubulin antagonist).
9072. The method of item 8965 wherein the agent is S-0885 (a collagenase inhibitor).
9073. The method of item 8965 wherein the agent is aplidine (an elongation factor-1 alpha inhibitor).
9074. The method of item 8965 wherein the agent is ixabepilone (an epithilone). 9075. The method of item 8965 wherein the agent is IDN- 5390 (an angiogenesis inhibitor).
9076. The method of item 8965 wherein the agent is SB- 2723005 (an angiogenesis inhibitor).
9077. The method of item 8965 wherein the agent is ABT-518 (an angiogenesis inhibitor).
9078. The method of item 8965 wherein the agent is combretastatin (an angiogenesis inhibitor).
9079. The method of item 8965 wherein the agent is anecortave acetate (an angiogenesis inhibitor).
9080. The method of item 8965 wherein the agent is SB- 715992 (a kinesin antagonist).
9081. The method of item 8965 wherein the agent is temsirolimus (an mTOR inhibitor).
9082. The method of item 8965 wherein the agent is adalimumab (a TNFα antagonist).
9083. The method of item 8965, wherein the composition comprises a polymer.
9084. The method of item 8965, wherein the composition comprises a polymer, and the polymer is, or comprises, a copolymer. 9085. The method of item 8965, wherein the composition comprises a polymer, and the polymer is, or comprises, a block copolymer.
9086. The method of item 8965, wherein the composition comprises a polymer, and the polymer is, or comprises, a random copolymer.
9087. The method of item 8965, wherein the composition comprises a polymer, and the polymer is, or comprises, a biodegradable polymer.
9088. The method of item 8965, wherein the composition comprises a polymer, and the polymer is, or comprises, a nonbiodegradable polymer.
9089. The method of item 8965, wherein the composition comprises a polymer, and the polymer is, or comprises, a hydrophilic polymer.
9090. The method of item 8965, wherein the composition comprises a polymer, and the polymer is, or comprises, a hydrophobic polymer.
9091. The method of item 8965, wherein the composition comprises a polymer, and the polymer is, or comprises, a polymer having hydrophilic domains.
9092. The method of item 8965, wherein the composition comprises a polymer, and the polymer is, or comprises, a polymer having hydrophobic domains. 9093. The method of item 8965, wherein the composition comprises a polymer, and the polymer is, or comprises, a non-conductive polymer.
9094. The method of item 8965, wherein the composition comprises a polymer, and the polymer is, or comprises, an elastomer.
9095. The method of item 8965, wherein the composition comprises a polymer, and the polymer is, or comprises, a hydrogel.
9096. The method of item 8965, wherein the composition comprises a polymer, and the polymer is, or comprises, a silicone polymer.
9097. The method of item 8965, wherein the composition comprises a polymer, and the polymer is, or comprises, a hydrocarbon polymer. - - -
9098. The method of item 8965, wherein the composition comprises a polymer, and the polymer is, or comprises, a styrene-derived polymer.
9099. The method of item 8965, wherein the composition comprises a polymer, and the polymer is, or comprises, a butadiene-derived polymer.
9100. The method of item 8965, wherein the composition comprises a polymer, and the polymer is, or comprises, a macromer.
9101. The method of item 8965, wherein the composition comprises a polymer, and the polymer is, or comprises, a poly(ethylene glycol) polymer. 9102. The method of item 8965, wherein the composition comprises a polymer, and the polymer is, or comprises, an amorphous polymer.
9103. The method of item 8965, wherein the composition further comprises a second pharmaceutically active agent.
9104. The method of item 8965, wherein the composition further comprises an anti-inflammatory agent.
9105. The method of item 8965, wherein the composition further comprises an agent that inhibits infection.
9106. The method of item 8965, wherein the composition further comprises an anthracycline.
9107. The method of item 8965, wherein the composition further comprises doxorubicin.
9108. The method of item 8965 wherein the composition further comprises mitoxantrone.
9109. The method of item 8965 wherein the composition further comprises a fluoropyrimidine.
9110. The method of item 8965, wherein the composition further comprises 5-fluorouracil (5-FU).
9111. The method of item 8965, wherein the composition further comprises a folic acid antagonist. 9112. The method of item 8965, wherein the composition further comprises methotrexate.
9113. The method of item 8965, wherein the composition further comprises a podophylotoxin.
9114. The method of item 8965, wherein the composition further comprises etoposide.
9115. The method of item 8965, wherein the composition further comprises camptothecin.
9116. The method of item 8965, wherein the composition further comprises a hydroxyurea.
9117. The method of item 8965, wherein the composition further comprises a platinum complex.
9118. The method of item 8965, wherein the composition further comprises cisplatin.
9119. The method of item 8965 wherein the composition further comprises an anti-thrombotic agent.
9120. The method of item 8965, wherein the composition further comprises a visualization agent.
9121. The method of item 8965, wherein the composition further comprises a visualization agent, and the visualization agent is a radiopaque material, wherein the radiopaque material comprises a metal, a halogenated compound, or a barium containing compound. 9122. The method of item 8965, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, barium, tantalum, or technetium.
9123. The method of item 8965, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, an MRI responsive material.
9124. The method of item 8965, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, a gadolinium chelate.
9125. The method of item 8965, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, iron, magnesium, manganese, copper, or chromium.
9126. The method of item 8965, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, iron oxide compound.
9127. The method of item 8965, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, a dye, pigment, or colorant.
9128. The method of item 8965 wherein the agent is released in effective concentrations from the composition comprising the agent by diffusion over a period ranging from the time of administration to about 90 days.
9129. The method of item 8965 wherein the agent is released in effective concentrations from the composition comprising the agent by erosion of the composition over a period ranging from the time of administration to about 90 days.
9130. The method of item 8965 wherein the composition further comprises an inflammatory cytokine.
9131. The method of item 8965 wherein the composition further comprises an agent that stimulates cell proliferation.
9132. The method of item 8965 wherein the composition further comprises a polymeric carrier.
9133. The method of item 8965 wherein the composition is in the form of a gel, paste, or spray.
9134. The method of item 8965 wherein the implant is partially constructed with the agent or the composition.
9135. The method of item 8965 wherein the implant is fully constructed with the agent or the composition.
9136. The method of item 8965 wherein the implant is impregnated with the agent or the composition.
9137. The method of item 8965, wherein the agent or the composition forms a coating, and the coating directly contacts the implant.
9138. The method of item 8965, wherein the agent or the composition forms a coating, and the coating indirectly contacts the implant. 9139. The method of item 8965 wherein the agent or the composition forms a coating, and the coating partially covers the implant.
9140. The method of item 8965, wherein the agent or the composition forms a coating, and the coating completely covers the implant.
9141. The method of item 8965 wherein the agent or the composition is located within pores or holes of the implant.
9142. The method of item 8965 wherein the agent or the composition is located within a channel, lumen, or divet of the implant.
9143. The method of item 8965 wherein the implant further comprising an echogenic material.
9144. The method of item 8965 wherein the implant further - comprises an echogenic material, wherein the echogenic material is in the form of a coating.
9145. The method of item 8965 wherein the implant is sterile.
9146. The method of item 8965 wherein the agent is delivered from the implant, wherein the agent is released into tissue in the vicinity of the implant after deployment of the implant.
9147. The method of item 8965 wherein the agent is delivered from the implant, wherein the agent is released into tissue in the vicinity of the implant after deployment of the implant, wherein the tissue is connective tissue. 9148. The method of item 8965 wherein the agent is delivered from the implant, wherein the agent is released into tissue in the vicinity of the implant after deployment of the implant, wherein the tissue is muscle tissue.
9149. The method of item 8965 wherein the agent is delivered from the implant, wherein the agent is released into tissue in the vicinity of the implant after deployment of the implant, wherein the tissue is nerve tissue.
9150. The method of item 8965 wherein the agent is delivered from the implant, wherein the agent is released into tissue in the vicinity of the implant after deployment of the implant, wherein the tissue is epithelium tissue.
9151. The method of item 8965 wherein the agent is delivered from the implant, wherein the agent is released in effective concentrations from the implant over a period ranging from the time of deployment of the implant to about 1 year.
9152. The method of item 8965 wherein the agent is delivered from the implant, wherein the agent is released in effective concentrations from the implant over a period ranging from about 1 month to 6 months.
9153. The method of item 8965 wherein the agent is delivered from the implant, wherein the agent is released in effective concentrations from the implant over a period ranging from about 1 - 90 days.
9154. The method of item 8965 wherein the agent is delivered from the implant, wherein the agent is released in effective concentrations from the implant at a constant rate. 9155. The method of item 8965 wherein the agent is delivered from the implant, wherein the agent is released in effective concentrations from the implant at an increasing rate.
9156. The method of item 8965 wherein the agent is delivered from the implant, wherein the agent is released in effective concentrations from the implant at a decreasing rate.
9157. The method of item 8965 wherein the agent is delivered from the implant, wherein the implant comprises about 0.01 Dg to about 10 Dg of the agent.
9158. The method of item 8965 wherein the agent is delivered from the implant, wherein the implant comprises about 10 Dg to about 10 mg of the agent.
9159. The method of item 8965 wherein the agent is delivered from the implant, wherein the implant comprises about 10 mg to about 250 mg of the agent.
9160. The method of item 8965 wherein the agent is delivered from the implant, wherein the implant comprises about 250 mg to about 1000 mg of the agent.
9161. The method of item 8965 wherein the agent is delivered from the implant, wherein the implant comprises about 1000 mg to about 2500 mg of the agent.
9162. The method of item 8965 wherein the agent is delivered from the implant, wherein a surface of the implant comprises less than 0.01 Dg of the agent per mm2 of implant surface to which the agent is applied. 9163. The method of item 8965 wherein the agent is delivered from the implant, wherein a surface of the implant comprises about 0.01 Dg to about 1 Dg of the agent per mm2 of implant surface to which the agent is applied.
9164. The method of item 8965 wherein the agent is delivered from the implant, wherein a surface of the implant comprises about 1 Dg to about 10 Dg of the agent per mm2 of implant surface to which the agent is applied.
9165. The method of item 8965 wherein the agent is delivered from the implant, wherein a surface of the implant comprises about 10 Dg to about 250 Dg of the agent per mm2 of implant surface to which the agent is applied.
9166. The method of item 8965 wherein the agent is delivered from the implant, wherein a surface of the implant comprises about 250 Dg to about 1000 Dg of the agent per mm2 of implant surface to which the agent is applied.
9167. The method of item 8965 wherein the agent is delivered from the implant, wherein a surface of the implant comprises about 1000 Dg to about 2500 Dg of the agent per mm2 of implant surface to which the agent is applied.
9168. The method of item 8965, wherein the implant further comprises a coating, and the coating is a uniform coating.
9169. The method of item 8965, wherein the implant further comprises a coating, and the coating is a non-uniform coating. 9170. The method of item 8965, wherein the implant further comprises a coating, and the coating is a discontinuous coating.
9171. The method of item 8965, wherein the implant further comprises a coating, and the coating is a patterned coating.
9172. The method of item 8965, wherein the implant further comprises a coating, and the coating has a thickness of 100 Dm or less.
9173. The method of item 8965, wherein the implant further comprises a coating, and the coating has a thickness of 10 Dm or less.
9174. The method of item 8965, wherein the implant further comprises a coating, and the coating adheres to the surface of the implant upon deployment of the implant.
9175. The method of item 8965, wherein the implant further comprises a coating, and the coating is stable at room temperature for a period of at least 1 year.
9176. The method of item 8965, wherein the implant further comprises a coating, and the agent is present in the coating in an amount ranging between about 0.0001% to about 1% by weight.
9177. The method of item 8965, wherein the implant further comprises a coating, and the agent is present in the coating in an amount ranging between about 1% to about 10% by weight.
9178. The method of item 8965, wherein the implant further comprises a coating, and the agent is present in the coating in an amount ranging between about 10% to about 25% by weight. 9179. The method of item 8965, wherein the implant further comprises a coating, and the agent is present in the coating in an amount ranging between about 25% to about 70% by weight.
9180. The method of item 8965, wherein the implant further comprises a coating, and the coating comprises a polymer.
9181. The method of item 8965, wherein the implant comprises a first coating having a first composition and a second coating having a second composition.
9182. The method of item 8965, wherein the implant comprises a first coating having a first composition and a second coating having a second composition, wherein the first composition and the second composition are different.
9183. A method as in any one of items 8965-9182, wherein the device is an endotracheal tube.
9184. A method as in any one of items 8965-9182, wherein the device is an endotracheal tube with a single lumen.
9185. A method as in any one of items 8965-9182, wherein the device is an endotracheal tube with double lumens.
9186. A method as in any one of items 8965-9182, wherein the device is a tracheostomy tube.
9187. A method for inhibiting scarring comprising placing a device that comprises a peritoneal dialysis catheter (e.g., an implant) and an anti-scarring agent or a composition comprising an anti-scarring agent into an animal host, wherein the agent inhibits scarring.
9188. The method of item 9187 wherein the agent is an adensosine A2A receptor antagonist.
9189. The method of item 9187 wherein the agent is an AKT inhibitor.
9190. The method of item 9187 wherein the agent is an alpha 2 integrin antagonist, wherein the alpha 2 integrin antagonist is Pharmaprojects No. 5754 (Merck KgaA).
9191. The method of item 9187 wherein the agent is an alpha 4 integrin antagonist.
9192. The method of item 9187 wherein the agent is an alpha 7 nicotinic receptor agonist.
9193. The method of item 9187 wherein the agent is an angiogenesis inhibitor selected from the group consisting of AG-12,958 (Pfizer), ATN-161 (Attention LLC), neovastat, an angiogenesis inhibitor from Jerina AG (Germany), NM-3 (Mercian), VGA-1155 (Taisho), FCE-26644 (Pfizer), FCE-26950 (Pfizer), FPMA (Meiji Daries), FR-111142 (Fujisawa), GGTI-298, GM-1306 (Ligand), GPA-1734 (Novartis), NNC-47-0011 (Novo Nordisk), herbamycin (Nippon Kayaku), lenalidomide (Celegene), IP-10 (NIH), ABT-828 (Abbott), KIN-841 (Tokushima University, Japan), SF-1126 (Semafore Pharmaceuticals), laminin technology (NIH), CHIR-258 (Chiron), NVP-AEW541 (Novartis), NVP-AEW541 (Novartis), Vt16907 (Alchemia), OXI-8007 (Oxigene), EG-3306 (Ark Therapeutics), Maspin (Arriva), ABT- 567 (Abbott), PPI-2458 (Praecis Pharmaceuticals), CC-5079, CC-4089 (Celgene), HIF-1 alpha inhibitors (Xenova), S-247 (Pfizer), AP-23573 (Ariad), AZD-9935 (Astra Zeneca), mebendazole (Introgen Therapeutics), MetAP-2 inhibitors (GlaxoSmithKline), AG-615 (Angiogene Pharmaceuticals), Tie-2 antagonists (Hybrigenics), NC-381 , CYC-381 , NC-169, NC-219, NC-383, NC-384, NC-407 (Lorus Therapeutics), ATN-224 (Attenuon), ON-01370 (Onconova), Vitronectin antagonists (Amgen), SDX-103 (Salmedix), Vitronectin antagonists (Shire), CHP (Riemser), TEK (Amgen), Anecortave acetate (Alcon), T46.2 (Matrix Therapeutics), HG-2 (Heptagen), TEM antagonists (Genzyme), Oxi-4500 (Oxigene), ATN-161 (Attenuon), WX-293 (Wilex), M-2025 (Metris Therapeutics), Alphastatin (BioActa), YH-16 (Yantai Rongchang), BIBF-1120 (Boehringer Ingelheim), BAY-57-9352 (Bayer), AS- 1404 (Cancer Research Technology), SC-77964 (Pfizer), glycomimetics (BioTie Therapies), TIE-2 Inhibitors (Ontogen), DIMI, Octamer (Octamer), ABR-215050 (Active Biotech), ABT-518 (Abbott), KDR inhibitors (Abbott), BSF-466895 (Abbott), SCH-221153 (Schering-Plough), DAC:antiangiogenic (ConjuGhem), TFPI (EntreMed), AZD-2171 (Astra-Zenaca), CDC-394 . .. - (Celgene), LY290293 (EIi Lilly), IDN-5390 (Indena), Kdr Kinase Inhibitors (Merck), CT-113020, CT-116433, CT-116563, CT-31890, CT-32228) (Cell Therapeutics), A-299620 (Abbott), TWEAK Inhibitor (Amgen), VEGF modulators (Johnson and Johnson), Tum-N53, tumstatin (Genzyme), Thios- 1 , Thios-2 (Thios Pharmaceuticals), MV-6401 (Miravant Medical Technologies), Spisulosine (PharmaMar), CEP-7055 (Cephalon), AUV-201 (Auvation), LM-609 (EIi Lilly), SKF-106615 (AnorMED), Oglufanide disodium (Cytran), BW-114 (Pharminox), Calreticulin (NIH), WX-678 (Wilex), SD-7784 (Pfizer), WX-UK1 (Wilex), SH-268 (Schering AG), 2-Me-PGA (Celgene), S- 137 (Pfizer), ZD-6126 (Angiogene Pharmaceuticals), SG-292 (SignalGen), Benefin (Lane Labs), A6, A36 (Angstrom), SB-2723005 (GlaxoSmithKline), SC-7 (Cell Therapeutics), ZEN-014 (AEterna Zentaris), 2-methoxyestradiol (EntreMed), NK-130119 (Nippon Kayaku), CC-10004 (Celgene), AVE- 8062A (Ajinomoto), Tacedinaline (Pfizer), Actinonin (Tokyo Metropolitan Institute of Medical Science), Lenalidomide (Celgene), VGA-1155, BTO-956 (SRI International), ER-68203-00 (Eisai), CT-6685 (UCB), JKC-362 (Phoenix Pharmaceuticals), DMI-3798 (DMI Biosciences, Angiomate (Ipsen), ZD-6474 (AstraZeneca), CEP-5214 (Cephalon), Canstatin (Genzyme), NM-3 (Mercian), Oridigm (MediQuest Therapeutics), Exherin (Adherex), BLS-0597 (Boston Life Sciences), PTC-299 (PTC Therapeutics), NPI-2358 (Nereus Pharmaceuticals), CGP-79787 (Novartis), AEE-788 (Novartis), CKD-732 (Chong Kun Dang), CP-564959 (OSI Pharmaceuticals), CM-101 (CarboMed), CT-2584, CT3501 (Cell Therapeutics), combretastatin and analogues and derivatives thereof (Oxigene), Rebimastat (Bristol-Meyers Squibb), Dextrin 2-sulfate (ML Laboratories), Cilengitide (Merk KGaA), NSC-706704 (Pharminox), KRN- 951 (Kirin Brewery), Ukrain, NSC-631570 (Nowicky Pharma), Tecogalan sodium (Daiichi Pharmaceutical), Tz-93 (Tsumura), TBC-1635 (Encysive Pharmaceuticals), TAN-1120 (Takeda), Semaxanib (Pfizer), BDI-7800 (Biopharmacopae), SD-186, SD-983 (Bristol-Meyers Squibb), SB-223245 (GlaxoSmithKline), SC-236 (Pfizer), RWJ-590973 (Johnson and Johnson), ILX-1850 (Genzyme), SC-68488, S-836 (Pfizer), CG-55069-11 (CuraGen), Ki-23057 (Kirin Brewery), CCX-700 (Chemoentryx), Pegaptanib octasodium (Giled Sciences), ANGIOCOL (available from Biostratum Inc.), or an analogue or derivative thereof.
9194. The method of item 9187 wherein the agent is an apoptosis antagonist.
9195. The method of item 9187 wherein the agent is an apoptosis activator.
9196. The method of item 9187 wherein the agent is a beta 1 integrin antagonist. 9197. The method of item 9187 wherein the agent is a beta tubulin inhibitor.
9198. The method of item 9187 wherein the agent is a blocker of enzyme production in Hepatitis C.
9199. The method of item 9187 wherein the agent is a Bruton's tyrosine kinase inhibitor.
9200. The method of item 9187 wherein the agent is a calcineurin inhibitor.
9201. The method of item 9187 wherein the agent is a caspase 3 inhibitor.
9202. The method of item 9187 wherein the agent is a CC chemokine receptor antagonist.
9203. The method of item 9187 wherein the agent is a cell cycle inhibitor selected from the group consisting of SNS-595 (Sunesis), synthadotin, KRX-0403, and an analogue or derivative thereof.
9204. The method of item 9187 wherein the agent is a cathepsin B inhibitor.
9205. The method of item 9187 wherein the agent is a cathepsin K inhibitor, wherein the cathepsin K inhibitor is 462795 (GlaxoSmithKline), INPL-022-D6 (Amura Therapeutics), or an analogue or derivative thereof. 9206. The method of item 9187 wherein the agent is a cathepsin L inhibitor.
9207. The method of item 9187 wherein the agent is a CD40 antagonist.
9208. The method of item 9187 wherein the agent is a chemokine receptor agonist.
9209. The method of item 9187 wherein the agent is a chymase inhibitor.
9210. The method of item 9187 wherein the agent is a collagenase antagonist.
- 9211. The method of item 9187 wherein the agent is a CXCR antagonist.
9212. The method of item 9187 wherein the agent is a cyclin dependent kinase inhibitor selected from the group consisting of a CDK-1 inhibitor, a CDK-2 inhibitor, a CDK- 4 inhibitor, a CDK-6 inhibitor, a CAK1 inhibitor from GPC Biotech and Bristol-Myers Squibb, RGB-286199 (GPC Biotech), an anticancer agent from Astex Technology, a CAK1 inhibitor from GPC Biotech, a CDK inhibitor from Sanofi-Aventis, a CDK1/CDK2 inhibitor from Amgen, a CDK2 inhibitor from SUGEN-2 (Pfizer), a hearing loss therapy agent (Sound Pharmaceuticals), PD-0332991 (Pfizer), RGB-286199 (GPC Biotech), Ro-0505124 (Hoffmann-La Roche), a Ser/Thr kinase inhibitor from Lilly (EIi Lilly), CVT-2584 (CAS No. 199986-75-9) (CV Therapeutics), and an analogue or derivative thereof. 9213. The method of item 9187 wherein the agent is a cyclooxygenase 1 inhibitor.
9214. The method of item 9187 wherein the agent is a DHFR inhibitor.
9215. The method of item 9187 wherein the agent is a dual integrin inhibitor.
9216. The method of item 9187 wherein the agent is an elastase inhibitor.
9217. The method of item 9187 wherein the agent is an elongation factor-1 alpha inhibitor.
9218. The method of item 9187 wherein the agent is an endothelial growth factor antagonist.
9219. The method of item 9187 wherein the agent is an endothelial growth factor receptor kinase inhibitor selected from the group consisting of sorafenib tosylate (Bayer), AAL-993 (Novartis), ABP-309 (Novartis), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), E- 7080 (Eisai), EG-3306 (Ark Therapeutics), EXEL-2880 (Exelixis), GW- 654652 (GlaxoSmithKline), a KDR inhibitor from LG Life Sciences, CT-6685 and CT-6729 (UCB), KRN-633 and KRN-951 (Kirin Brewery), OSI-930 (OSI Pharmaceuticals), SP-5.2 (Supratek Pharma), SU-11657 (Pfizer), a Tie-2 antagonist (Hybrigenics), a VEGFR-2 kinase inhibitor (Bristol-Myers Squibb), XL-647 (Exelixis), a KDR inhibitor from Abbott Laboratories, sorafenib tosylate, and an analogue or derivative thereof. 9220. The method of item 9187 wherein the agent is an endotoxin antagonist.
9221. The method of item 9187 wherein the agent is an epothilone and tubulin binder.
9222. The method of item 9187 wherein the agent is an estrogen receptor antagonist.
9223. The method of item 9187 wherein the agent is an FGF inhibitor.
9224. The method of item 9187 wherein the agent is a farnexyl transferase inhibitor.
9225. The method of item 9187 wherein the agent is~ farnesyltransferase inhibitor selected from the group of A-197574 (Abbott), a farnesyltransferase inhibitor from Servier, FPTIII (Strathclyde Institute for Drug R), LB-42908 (LG Life Sciences), Pharmaprojects No. 5063 (Genzyme), Pharmaprojects No. 5597 (Ipsen), Yissum Project No. B-1055 (Yissum), and an analogue or derivative thereof.
9226. The method of item 9187 wherein the agent is an FLT-3 kinase inhibitor.
9227. The method of item 9187 wherein the agent is an FGF receptor kinase inhibitor.
9228. The method of item 9187 wherein the agent is a fibrinogen antagonist selected from the group consisting of AUV-201 (Auvation), MG-13926 (Sanofi-Aventis), plasminogen activator (CAS No. 105913-11-9) (from Sanofi-Aventis or UCB), plasminogen activator-2 (tPA- 2) (Sanofi-Aventis), pro-urokinase (CAS No. 82657-92-9) (Sanofi-Aventis), and an analogue or derivative thereof.
9229. The method of item 9187 wherein the agent is a heat shock protein 90 antagonist selected from the group consisting of SRN-005 (Sirenade), geldanamycin, NSC-33050 (17-allylaminogeldanamycin; 17- AAG), 17-dimethylaminoethylamino-17-demethoxy-geldanamycin (17- DMAG), rifabutin (rifamycin XIV, 1',4-didehydro-1-deoxy-1,4-dihydro-5'-(2- methylpropyl)-1-oxo~), and an analogue or derivative thereof.
9230. The method of item 9187 wherein the agent is a histone deacetylase inhibitor.
9231. The method of item 9187 wherein the agent is an HMGCoA reductase inhibitor selected from the group consisting of an atherosclerosis therapeutic from Lipid Sciences, ATI-16000 (ARYx Therapeutics), KS-01-019 (Kos Pharmaceuticals), Pharmaprojects No. 2197 (Sanofi-Aventi), RP 61969 (Sanofi-Aventis), and an analogue or derivative thereof.
9232. The method of item 9187 wherein the agent is an ICAM inhibitor.
9233. The method of item 9187 wherein the agent is an IL, ICE and IRAK antagonist, wherein the antagonist is a CJ-14877, CP-424174 (Pfizer), NF-61 (Negma-Lerads), and an analogue or derivative thereof.
9234. The method of item 9187 wherein the agent is an IL-2 inhibitor. 9235. The method of item 9187 wherein the agent is an immunosuppressant selected from the group consisting of teriflunomide (Sanofi Aventis), chlorsulfaquinoxalone (NSC-339004), chlorsulfaquinoxalone sulfate, CS-712 (Sankyo), ismomultin alfa (CAS No. 457913-93-8) (Akzo Nobel), antiallergics from GenPat77, antiinflammatories or AT-005 (Androclus Therapeutics), autoimmune disease therapeutics from EpiVax, BN-007 (Bone), budesonide (CAS No. 51333-22- 3) (MAP Pharmaceuticals), CO-14 (Genzyme), edratide (CAS No. 433922- 67-9) (Teva), EP-314 (Enanta), eprovafen (CAS No. 101335-99-3) (Sanofi- Aventis), HWA-131 (CAS No. 118788-41-3) (Sanofi-Aventis), immunomodulators from MerLion Pharmaceuticals, immunosuppressives from Alchemia, IPL-12 (Inflazyme), MDL-9563 (CAS No. 27086-86-8) (Sanofi-Aventis), Pharmaprojects No. 2330 (Sanofi-Aventis), Pharmaprojects No. 6426 (Abgenix), PXS-25 (Pharmaxis), rosmarinic acid (CAS No. 20283-92-5) (Sanofi-Aventis), RP 42927 or RP 54745 (CAS No.
135330-08-4) (Sanofi-Aventis), -SGN-35 (Seattle Genetics), ST-1959 . . (Sigma-Tau), type I diabetes therapy from SYNX Pharma, UNIL-88 (Debiopharm), VP-025 (Vasogen), VR-694 (Vectura), PRTX-001 (Protalex), and an analogue or derivative thereof.
9236. The method of item 9187 wherein the agent is an IMPDH (inosine monophosphate).
9237. The method of item 9187 wherein the agent is an integrin antagonist.
9238. The method of item 9187 wherein the agent is an interleukin antagonist.
9239. The method of item 9187 wherein the agent is an inhibitor of type III receptor tyrosine kinase. 9240. The method of item 9187 wherein the agent is an irreversible inhibitor of enzyme methionine aminopeptidase type 2.
9241. The method of item 9187 wherein the agent is an isozyme selective delta protein kinase C inhibitor.
9242. The method of item 9187 wherein the agent a JAK3 enzyme inhibitor.
9243. The method of item 9187 wherein the agent is a JNK inhibitor.
9244. The method of item 9187 wherein the agent is a kinase inhibitor.
9245. The method of item 9187 wherein the agent is kinesin antagonist.
9246. The method of item 9187 wherein the agent is a kinesin antagonist.
9247. The method of item 9187 wherein the agent is a leukotriene inhibitor and antagonist selected from the group consisting of ambicromil (CAS No. 58805-38-2) (Sanofi-Aventis), amelubant (CAS No. 346735-24-8) (Boehringer Ingelheim), DW-1141 (Dong Wha), ebselen (Daiichi Pharmaceutical), ibudilast (Kyorin), leucotriene inhibitors from Sanofi-Aventis, lymphotoxin-beta receptor (LT-D) from Biogen Idee, Pharmaprojects No. 1535 and 2728 (CAS No. 119340-33-9) (Sanofi- Aventis), R-112 (Rigel), Rev-5367 (CAS No. 92532-05-3) (Sanofi-Aventis), RG-14893 (CAS No. 141835-49-6) (Sanofi-Aventis), RG-5901-A (CAS No. 101910-24-1), 92532-23-5, RP 66153 (CAS No. 142422-79-5), RP 66364 (CAS No.186916-92-5), RP 69698 (CAS No. 141748-00-7) (Sanofi-Aventis), SC-411930 (Pfizer), SC-41930 (CAS No. 120072-59-5) (Pfizer), SC-50605 (CAS No. 138828-39-4) (Pfizer), SC-51146 (CAS No. 141059-52-1), SC- 53228 (CAS No. 153633-01-3) (Pfizer), spaglumic acid (ZY-15106) (CAS No.3106-85-2), 80619-64-3 (Novartis), tipredane (CAS No. 85197-77-9) (Bristol-Myers Squibb), U-75302 (CAS No. 119477-85-9) (Pfizer), and analogue or derivative thereof.
9248. The method of item 9187 wherein the agent is an MAP kinase inhibitor.
9249. The method of item 9187 wherein the agent is a matrix metalloproteinase inhibitor.
9250. The method of item 9187 wherein the agent is an MCP- GCR2 inhibitor. . . . ..
9251. The method of item 9187 wherein the agent is an mTOR inhibitor.
9252. The method of item 9187 wherein the agent is an mTOR kinase inhibitor.
9253. The method of item 9187 wherein the agent is a microtubule inhibitor selected from the group consisting of antibody- maytansinoid conjugates from Biogen Idee, colchicines (MantiCore Pharmaceuticals), anticancer immunoconjugates from Johnson & Johnson, DIME from Octamer, gni-1f (GNI), huC242-DM4, huMy9-6-DM1 (ImmunoGen), IDN-5404 (Indena), IMO-098, IMOderm (Imotep), mebendazole (Introgen Therapeutics), microtubule poisons from Cambridge Enterprise, paclitaxel such as LOTAX from Aphios (CAS No. 33069-62-4), Genexol-PM from Samyang, Pharmaprojects No. 6383 (Tapestry Pharmaceuticals), RPR-112378 (Sanofi-Aventis), SGN-75 (Seattle Genetics), SPL-7435 (Starpharma), SSR-250411 (Sanofi-Aventis), trastuzumab-DM1 (Genentech), vinorelbine, and an analogue or derivative thereof.
9254. The method of item 9187 wherein the agent is an MIF inhibitor.
9255. The method of item 9187 wherein the agent is an MMP inhibitor.
9256. The method of item 9187 wherein the agent is a neurokinin (NK) antagonist selected from the group consisting of anthrotainin (CAS No. 148084-40-6) (Sanofi-Aventis), an IBS thereapeutic from ArQuIe1 MDL-105212A (CAS No. 167261-60-1) (Ssanofi-Aventis), Pharmaprojects No. 2744, 3258 (CAS No. 139167-47-8) 4006, 4201 , or 5986 (Sanofi-Aventis), RP 67580 (CAS No. 135911-02-3), SR-144190 (CAS No. 201152-86-5), SSR-240600, SSR-241586 (Sanofi-Aventis), TKA-457 (Novartis), vestipitant mesylate (CAS No. 334476-64-1) (GlaxoSmithKline), Win-64821 (Sanofi-Aventis), PRX-96026 (Predix Pharmaceuticals), and an analogue or derivative thereof.
9257. The method of item 9187 wherein the agent is an NF kappa B inhibitor selected from the group consisting of AVE-0545 or AVE- 0547 (Sanofi-Aventis), bortezomib (CAS No. 179324-69-7) (Millennium Pharmaceuticals), dexanabinol (CAS No. 112924-45-5) (Pharmos), dexlipotam (Viatris), Pharmaprojects No. 6283 (INDRA) (OXiGENE), IPL- 576092 (CAS No. 137571-30-3) (Inflazyme), NFKB decoy (Corgentech), NFKB decoy oligo (AnGes MG), NFKB's from Ariad, osteoporosis treatments or S5 (F005) from Fulcrum Pharmaceuticals, P61 (Phytopharm), R-flurbiprofen (CAS No. 5104-49-4) (Encore Pharmaceuticals), and an analogue or derivative thereof.
9258. The method of item 9187 wherein the agent is a nitric oxide agonist.
9259. The method of item 9187 wherein the agent is an ornithine decarboxylase inhibitor.
9260. The method of item 9187 wherein the agent is a p38 MAP kinase inhibitor selected from the group consisting of AZD-6703 (AstraZeneca), JX-401 (Jexys Pharmaceuticals), BMS-2 (Bristol-Myers Squibb), a p38 MAP kinase inhibitor from Novartis, a p38-alpha MAP kinase inhibitor from Amphora, Pharmaprojects No. 5704 (Pharmacopeia), RPR- 200765A (Sanofi-Aventis), SD-282 (Johnson & Johnson), TAK-715 (Takeda), and an analogue or derivative thereof. - _ . _
9261. The method of item 9187 wherein the agent is a palmitoyl-protein thioesterase inhibitor.
9262. The method of item 9187 wherein the agent is a PDGF receptor kinase inhibitor selected from the group consisting of AAL-993, AMN-107, or ABP-309 (Novartis), AMG-706 (Amgen), BAY-57-9352 (Bayer), CDP-860 (UCB), E-7080 (Eisai), imatinib (CAS No. 152459-95-5) (Novartis), OSI-930 (OSI Pharmaceuticals), RPR-127963E (Sanofi-Aventis), RWJ-540973 (Johnson & Johnson), sorafenib tosylate (Bayer), SU-11657 (Pfizer), tandutinib (CAS No. 387867-13-2) (Millennium Pharmaceuticals), vatalanib (Novartis), ZK-CDK (Schering AG), and an analogue or derivative thereof. 9263. The method of item 9187 wherein the agent is (-)- halofenate (Metabolex), AMG-131 (Amgen), antidiabetics from Japan Tobacco, AZD-4619, AZD-8450, AZD-8677 (AstraZeneca), DRF-10945, balaglitazone (Dr Reddy's), CS-00088, CS-00098 (Chipscreen Biosciences), E-3030 (Eisai), etalocib (CAS No. 161172-51-6) (EIi Lilly), GSK-641597 (Ligand), GSK-677954 (GlaxoSmithKline), GW-409544 (Ligand), GW- 590735 (GlaxoSmithKline), K-111 (Hoffmann-La Roche), LY-518674 (EIi Lilly), LY-674 (Ligand), LY-929 (Ligand), MC-3001 , MC-3002 (MaxoCore Pharmaceuticals), metformin HCI + pioglitazone (CAS No. 1115-70-4 and
112529-15-4), ACTOPLUS MET from Andrx), muraglitazar (CAS No. 331741-94-7) (Bristol-Myers Squibb), naveglitazar (Ligand), oleoylethanolamide (Kadmus Pharmaceuticals), ONO-5129, pioglitazone hydrochloride (CAS No. 111025-46-8 and 112529-15-4) (Takeda), PLX-204 (Plexxikon), PPAR agonists from Genfit, PPAR delta agonists from EIi Lilly, PPAR-alpha agonists from CrystalGenomics, PPAR-gamma modulators and PPAR-β modulators from CareX, rosiglitazone maleate (CAS No. 122324- 73-4 or 155141-29-0) (GlaxoSmithKline), rosiglitazone maleate/glimepir (CAS No. 155141-29-0 and 93479-97-1), AVANDARYL, rosiglitazone maleate/metformin extend (CAS No. 155141-29-0 and 657-24-9), AVANDAMET, rosiglitazone maleate+metformin, AVANDAMET (GlaxoSmithKline), tesaglitazar (AstraZeneca), and an analogue or derivative thereof.
9264. The method of item 9187 wherein the agent is a phosphatase inhibitor.
9265. The method of item 9187 wherein the agent is a phosphodiesterase (PDE) inhibitor selected from the group consisting of avanafil (Tanabe Seiyaku), dasantafil (CAS No. 569351-91-3) (Schering- Plough), A-906119 (CAS No. 134072-58-5), DL-850 (Sanofi-Aventis), GRC- 3015, GRC-3566, GRC-3886 (Glenmark), HWA-153 (CAS No. 56395-66-5) (Sanofi-Aventis), hydroxypumafentrine (Altana), IBFB-130011 , IBFB-14-016, IBFB-140301 , IBFB-150007, IBFB-211913 (IBFB Pharma), L-826141 (Merck & Co), medorinone (CAS No. 88296-61-1) (Sanofi-Aventis), MEM- 1917 (Memory Pharmaceuticals), ND-1251 (Neuro3d), PDE inhibitors from ICOS, PDE IV inhibitors from Memory Pharmaceuticals and CrystalGenomics, Pharmaprojects No. 2742 and 6141 (Sanofi-Aventis), QAD-171 (Novartis), RHC-2963 (CAS No. 76993-12-9 and 76993-14-1), RPR-117658, RPR-122818 derivatives, SR-24870 , and RPR-132294 (Sanofi-Aventis), SK-350 (ln2Gen), stroke therapy agents from deCODE Genetics, TAS-203 (Taiho), tofimilast (CAS No. 185954-27-2) (Pfizer), UK- 371800 (Pfizer), WIN-65579 (CAS No. 158020-82-7) (Sanofi-Aventis), IBFB- 130020 (IBFB Pharma), OPC-6535 (CAS No. 145739-56-6) (Otsuka), a phosphodiesterase III inhibitor, enoximone, a phosphodiesterase IV inhibitor, fosfosal, Atopik (Barrier Therapeutics), triflusal, a phosphodiesterase V inhibitor, and an analogue or derivative thereof.
9266. The method of item 9187 wherein the agent is a PKC inhibitor.
9267. The method of item 9187 wherein the agent is a platelet activating factor antagonist.
9268. The method of item 9187 wherein the agent is a platelet-derived growth factor receptor kinase inhibitor.
9269. The method of item 9187 wherein the agent is a prolyl hydroxylase inhibitor.
9270. The method of item 9187 wherein the agent is a polymorphonuclear neutrophil inhibitor. 9271. The method of item 9187 wherein the agent is a protein kinase B inhibitor.
9272. The method of item 9187 wherein the agent is a protein kinase C stimulant.
9273. The method of item 9187 wherein the agent is a purine nucleoside analogue.
9274. The method of item 9187 wherein the agent is a purinoreceptor P2X antagonist.
9275. The method of item 9187 wherein the agent is a Raf kinase inhibitor.
9276. The method of item 9187 wherein the agent is a . reversible inhibitor of ErbB1 and ErbB2.
9277. The method of item 9187 wherein the agent is a ribonucleoside triphosphate reductase inhibitor.
9278. The method of item 9187 wherein the agent is an SDF- 1 antagonist.
9279. The method of item 9187 wherein the agent is a sheddase inhibitor.
9280. The method of item 9187 wherein the agent is an SRC inhibitor. 9281. The method of item 9187 wherein the agent is a stromelysin inhibitor.
9282. The method of item 9187 wherein the agent is an Syk kinase inhibitor.
9283. The method of item 9187 wherein the agent is a telomerase inhibitor.
9284. The method of item 9187 wherein the agent is a TGF beta inhibitor selected from the group consisting of pirfenidone (CAS No. 53179-13-8) (MARNAC), tranilast (CAS No. 53902-12-8) (Kissei), IN-1130 (ln2Gen), mannose-6-phosphate (BTG), TGF-β antagonists from Inflazyme (Pharmaprojects No. 6075), TGF-β antagonists from Sydney, non-industrial source), TGF-β I receptor kinase inhibitors from EIi Lilly, TGF-β receptor inhibitors from Johnson & Johnson, and an analogue or derivative thereof.
9285. The method of item 9187 wherein the agent is a TNFα antagonist or TACE inhibitor selected from the group consisting of adalimumab (CAS No. 331731-18-1) (Cambridge Antibody Technology), AGIX-4207 (AtheroGenics), AGT-1 (Advanced Biotherapy), an antiinflammatory from Borean Pharma, Cellzome, or Paradigm Therapeutics, anti-inflammatory vaccine (TNF-alpha kinoid) from Neovacs, humanized anti-TNF antibody or an anti-TNF MAb (CB0006) Celltech (UCB), apratastat (CAS No. 287445-51-0) (Wyeth), BMS-561392 (Bristol-Myers Squibb), BN- 006 (Bone), certolizumab pegol (CAS No. 428863-50-7 or CH-138 (UCB), cilomilast (CAS No. 153259-65-5) (GlaxoSmithKline), CR-1 (Nuada Pharmaceuticals), CRx-119 (CombinatoRx), D-5410 (UCB), dacopafant (CAS No. 125372-33-0) (Sanofi-Aventis), dersalazine (CAS No. 188913-57- 7/188913-58-8) (Uriach), etanercept (CAS No. 185243-69-0) (Amgen), ethyl pyruvate (Critical (Critical Therapeutics), golimumab (CAS No. 476181-74-5) (Johnson & Johnson), hormono-immunotherapy from Ipsen, CDP571 (e.g., Humicade from UCB), IC-485 (ICOS), infliximab (CAS No. 170277-31-3) (Johnson & Johnson), IP-751 (Manhattan Pharmaceuticals), ISIS-104838 (CAS No. 250755-32-9) (ISIS Pharmaceuticals), lenalidomide (CAS No. 191732-72-6) (Celgene), lentinan (CAS No. 37339-90-5) (Ajinomoto), MDL- 201112 (CAS No. 142130-73-2) (Sanofi-Aventis), medroxyprogesterone (CAS No. 520-85-4) (InKine Pharmaceutical), N-acetylcysteine (CAS No. 616-91-1) (Zambon), NBE-P2 (DIREVO Biotech), nerelimomab (CAS No. 162774-06-3) (Chiron), OM-294DP (OM PHARMA), onercept (CAS No. 199685-57-9) (Yeda), PASSTNF-alpha (Verigen), pentoxifylline or oxypentifylline (Sanofi-Aventis), Pharmaprojects No. 4091, 4241 , 4295, or 4488 (Sanofi-Aventis), Pharmaprojects No. 5480 (Amgen), Pharmaprojects No. 6749 (Cengent), pirfenidone (CAS No. 53179-13-8) (MARNAC), RPR- 132294 (Sanofi-Aventis), S5 (F002) (Fulcrum Pharmaceuticals), simvastatin (CAS No. 79902-63-9) (Merck & Co), STA-6292 (Synta Pharmaceuticals), tacrolimus (CAS No. 104987-11-3) (Fujisawa LifeCycle Pharma), talactoferrin alfa (CAS No. 308240-58-6) (Agennix), thalidomide (CAS No. 50-35-1) (Celgene), TNF antagonists form ProStrakan, and Synergen, TNF inhibitors (Amgen), TNF-alpha antagonists from Dynavax Technologies and Jerina AG (Germany), TNF-alpha inhibitors from IBFB Pharma and Xencor (Xencor), torbafylline (CAS No. 105102-21-4) (Sanofi-Aventis), UR-1505 (Uriach), VT-346 (Won Therapeutics), YSIL6 (Vs Therapeutics), YSTH2 (Y's Therapeutics), NPI-1302a-3 (Nereus Pharmaceuticals, a TNF antagonist from Jerina AG (Germany), dersalazine, and an analogue or derivative thereof.
9286. The method of item 9187 wherein the agent is a Toll receptor inhibitor.
9287. The method of item 9187 wherein the agent is a tubulin antagonist. 9288. The method of item 9187 wherein the agent is a tyrosine kinase inhibitor selected from the group consisting of SU-011248, SUTENT from Pfizer Inc. (New York, NY), BMS-354825, PN-355 (Paracelsian Pharmaceuticals), AGN-199659 (Allergan), AAL-993 or ABP- 309 (Novartis), adaphostin (NIH), AEE-788 (Novartis), AG-013736 (OSI Pharmaceuticals), AG-13736 (Pfizer), ALT-110 (Alteris Therapeutics), AMG- 706 (Amgen), anticancer MAbs from Xencor, anti-EGFrvlll MAbs from Abgenix, anti-HER2 MAb from Abiogen, AZD-2171 or AZD-9935 (AstraZeneca), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), CEP-5214 (Cephalon), CEP-7055 (Cephalon), cetuximab (ImClone Systems), CHIR-200131 and CHIR-258 (Chiron), CP-547632 (OSI Pharmaceuticals), CP-724714 (Pfizer), CT-301 (Creabilis Therapeutics), D- 69491 (Baxter International), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EGFR/ErbB2 inhibitors from Array BioPharma, erlotinib (CAS No. 183319- 69-9) (OSI Pharmaceuticals), EXEL-2880 (Exelixis), FK-778 (Sanofi- Aventis) , gefitinib (CAS No. 184475-35-2) (AstraZeneca), GW-2286.or GW- 654652 (GlaxoSmithKline), her2/neu antigen from AlphaVax, HER-2/neu inhibitor from Generex, Herzyme (Medipad) (Sirna Therapeutics), HKI-272 (Wyeth), HuMax-EGFr (Genmab), idronoxil (CAS No. 81267-65-4) (Novogen), IGF-1 inhibitors from Ontogen, IMC-11F8 (ImClone Systems), kahalalide F (CAS No. 149204-42-2) (PharmaMar), KDR inhibitor from LG Life Sciences, KDR inhibitors from Abbott Laboratories, KDR kinase inhibitors (UCB), Kdr kinase inhibitors from Merck & Co, KRN-633 and KRN- 951 (Kirin Brewery), KSB-102 (Xenova), lapatinib ditosylate (CAS No. 388082-78-8) (GlaxoSmithKline), matuzumab (Merck KGaA), MDX-214 (Medarex), ME-103 (Pharmexa), MED-A300 (Gerolymatos), MNAC-13 (Lay Line Genomics), nimotuzumab (Center of Molecular Immunology), NSC- 330507 or NSC-707545 (NIH), NV-50 (Novogen), OSI-930 (OSI Pharmaceuticals), panitumumab (Abgenix), pelitinib (CAS No. 287933-82-7) (Wyeth), pertuzumab (CAS No. 380610-27-5) (Genentech), Pharmaprojects No. 3985 (Sanofi-Aventis), prostate cancer therapeutics from Sequenom (SQPC35, SQPC36, SQPC90), removab and remoxab (Trion Pharma), RG- 13022 (CAS No. 136831-48-6), RG-13291 (CAS No. 138989-50-1), or RG- 14620 (CAS No. 136831-49-7) (Sanofi-Aventis), RM-6427 (Romark), RNAi breast cancer therapy from Benitec, RP 53801 (CAS No. 125882-88-4) (Sanofi-Aventis), sorafenib tosylate (Bayer), SU-11657 (Pfizer), Tie-2 antagonists from Semaia (Hybrigenics), Tie-2 inhibitors from Ontogen, trastuzumab (CAS No. 180288-69-1) (Genentech), tyrosine kinase inhibitors from Sanofi-Aventis, U3-1287, U3-1565, U3-1784, U3-1800 (U3 Pharma), vatalanib (Novartis), VEGFR-2 kinase inhibitor from Bristol-Myers Squibb, XL-647 (Exelixis), ZD-6474 (AstraZeneca), ZK-CDK (Schering AG), an EGFR tyrosine kinase inhibitor, EKB-569 (Wyeth), and an analogue or derivative thereof.
9289. The method of item 9187 wherein the agent is a VEGF inhibitor.
9290. The method of item 9187 wherein the agent is a vitamin D receptor agonist.
9291. The method of item 9187 wherein the agent is ZD-6474 (an angiogenesis inhibitor).
9292. The method of item 9187 wherein the agent is AP- 23573 (an mTOR inhibitor).
9293. The method of item 9187 wherein the agent is synthadotin (a tubulin antagonist).
9294. The method of item 9187 wherein the agent is S-0885 (a collagenase inhibitor). 9295. The method of item 9187 wherein the agent is aplidine (an elongation factor-1 alpha inhibitor).
9296. The method of item 9187 wherein the agent is ixabepilone (an epithilone).
9297. The method of item 9187 wherein the agent is IDN- 5390 (an angiogenesis inhibitor).
9298. The method of item 9187 wherein the agent is SB- 2723005 (an angiogenesis inhibitor).
9299. The method of item 9187 wherein the agent is ABT-518 (an angiogenesis inhibitor).
9300. The method of item 9187 wherein the agent is combretastatin (an angiogenesis inhibitor).
9301. The method of item 9187 wherein the agent is anecortave acetate (an angiogenesis inhibitor).
9302. The method of item 9187 wherein the agent is SB- 715992 (a kinesin antagonist).
9303. The method of item 9187 wherein the agent is temsirolimus (an mTOR inhibitor).
9304. The method of item 9187 wherein the agent is adalimumab (a TNFα antagonist). 9305. The method of item 9187, wherein the composition comprises a polymer.
9306. The method of item 9187, wherein the composition comprises a polymer, and the polymer is, or comprises, a copolymer.
9307. The method of item 9187, wherein the composition comprises a polymer, and the polymer is, or comprises, a block copolymer.
9308. The method of item 9187, wherein the composition comprises a polymer, and the polymer is, or comprises, a random copolymer.
9309. The method of item 9187, wherein the composition comprises a polymer, and the polymer is, or comprises, a biodegradable polymer. . . .
9310. The method of item 9187, wherein the composition comprises a polymer, and the polymer is, or comprises, a nonbiodegradable polymer.
9311. The method of item 9187, wherein the composition comprises a polymer, and the polymer is, or comprises, a hydrophilic polymer.
9312. The method of item 9187, wherein the composition comprises a polymer, and the polymer is, or comprises, a hydrophobic polymer. 9313. The method of item 9187, wherein the composition comprises a polymer, and the polymer is, or comprises, a polymer having hydrophilic domains.
9314. The method of item 9187, wherein the composition comprises a polymer, and the polymer is, or comprises, a polymer having hydrophobic domains.
9315. The method of item 9187, wherein the composition comprises a polymer, and the polymer is, or comprises, a non-conductive polymer.
9316. The method of item 9187, wherein the composition comprises a polymer, and the polymer is, or comprises, an elastomer.
9317. The method of item 9187, wherein the composition . comprises a polymer, and the polymer is, or comprises, a hydrogel.
9318. The method of item 9187, wherein the composition comprises a polymer, and the polymer is, or comprises, a silicone polymer.
9319. The method of item 9187, wherein the composition comprises a polymer, and the polymer is, or comprises, a hydrocarbon polymer.
9320. The method of item 9187, wherein the composition comprises a polymer, and the polymer is, or comprises, a styrene-derived polymer. 9321. The method of item 9187, wherein the composition comprises a polymer, and the polymer is, or comprises, a butadiene-derived polymer.
9322. The method of item 9187, wherein the composition comprises a polymer, and the polymer is, or comprises, a macromer.
9323. The method of item 9187, wherein the composition comprises a polymer, and the polymer is, or comprises, a poly(ethylene glycol) polymer.
9324. The method of item 9187, wherein the composition comprises a polymer, and the polymer is, or comprises, an amorphous polymer.
9325. The method of item 9187, wherein the composition further comprises a second pharmaceutically active agent.
9326. The method of item 9187, wherein the composition further comprises an anti-inflammatory agent.
9327. The method of item 9187, wherein the composition further comprises an agent that inhibits infection.
9328. The method of item 9187, wherein the composition further comprises an anthracycline.
9329. The method of item 9187, wherein the composition further comprises doxorubicin. 9330. The method of item 9187 wherein the composition further comprises mitoxantrone.
9331. The method of item 9187 wherein the composition further comprises a fluoropyrimidine.
9332. The method of item 9187, wherein the composition further comprises 5-fluorouracil (5-FU).
9333. The method of item 9187, wherein the composition further comprises a folic acid antagonist.
9334. The method of item 9187, wherein the composition further comprises methotrexate.
9335. The method of item 9187, wherein the composition further comprises a podophylotoxin.
9336. The method of item 9187, wherein the composition further comprises etoposide.
9337. The method of item 9187, wherein the composition further comprises camptothecin.
9338. The method of item 9187, wherein the composition further comprises a hydroxyurea.
9339. The method of item 9187, wherein the composition further comprises a platinum complex. 9340. The method of item 918,7, wherein the composition further comprises cisplatin.
9341. The method of item 9187 wherein the composition further comprises an anti-thrombotic agent.
9342. The method of item 9187, wherein the composition further comprises a visualization agent.
9343. The method of item 9187, wherein the composition further comprises a visualization agent, and the visualization agent is a radiopaque material, wherein the radiopaque material comprises a metal, a halogenated compound, or a barium containing compound.
9344. The method of item 9187, wherein the composition further comprises a visualization agent, and the visualization agentis, or . comprises, barium, tantalum, or technetium.
9345. The method of item 9187, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, an MRI responsive material.
9346. The method of item 9187, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, a gadolinium chelate.
9347. The method of item 9187, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, iron, magnesium, manganese, copper, or chromium. 9348. The method of item 9187, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, iron oxide compound.
9349. The method of item 9187, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, a dye, pigment, or colorant.
9350. The method of item 9187 wherein the agent is released in effective concentrations from the composition comprising the agent by diffusion over a period ranging from the time of administration to about 90 days.
9351. The method of item 9187 wherein the agent is released in effective concentrations from the composition comprising the agent by erosion of the composition over a period ranging from the time of administration to about 90 days.
9352. The method of item 9187 wherein the composition further comprises an inflammatory cytokine.
9353. The method of item 9187 wherein the composition further comprises an agent that stimulates cell proliferation.
9354. The method of item 9187 wherein the composition further comprises a polymeric carrier.
9355. The method of item 9187 wherein the composition is in the form of a gel, paste, or spray. 9356. The method of item 9187 wherein the implant is partially constructed with the agent or the composition.
9357. The method of item 9187 wherein the implant is fully constructed with the agent or the composition.
9358. The method of item 9187 wherein the implant is impregnated with the agent or the composition.
9359. The method of item 9187, wherein the agent or the composition forms a coating, and the coating directly contacts the implant.
9360. The method of item 9187, wherein the agent or the composition forms a coating, and the coating indirectly contacts the implant.
9361. The method of item 9187 wherein the agent or the composition forms a coating, and the coating partially covers the implant.
9362. The method of item 9187, wherein the agent or the composition forms a coating, and the coating completely covers the implant.
9363. The method of item 9187 wherein the agent or the composition is located within pores or holes of the implant.
9364. The method of item 9187 wherein the agent or the composition is located within a channel, lumen, or divet of the implant.
9365. The method of item 9187 wherein the implant further comprising an echogenic material. 9366. The method of item 9187 wherein the implant further comprises an echogenic material, wherein the echogenic material is in the form of a coating.
9367. The method of item 9187 wherein the implant is sterile.
9368. The method of item 9187 wherein the agent is delivered from the implant, wherein the agent is released into tissue in the vicinity of the implant after deployment of the implant.
9369. The method of item 9187 wherein the agent is delivered from the implant, wherein the agent is released into tissue in the vicinity of the implant after deployment of the implant, wherein the tissue is connective tissue.
9370. The method of item 9187 wherein the agent is delivered from the implant, wherein the agent is released into tissue in the vicinity of the implant after deployment of the implant, wherein the tissue is muscle tissue.
9371. The method of item 9187 wherein the agent is delivered from the implant, wherein the agent is released into tissue in the vicinity of the implant after deployment of the implant, wherein the tissue is nerve tissue.
9372. The method of item 9187 wherein the agent is delivered from the implant, wherein the agent is released into tissue in the vicinity of the implant after deployment of the implant, wherein the tissue is epithelium tissue. 9373. The method of item 9187 wherein the agent is delivered from the implant, wherein the agent is released in effective concentrations from the implant over a period ranging from the time of deployment of the implant to about 1 year.
9374. The method of item 9187 wherein the agent is delivered from the implant, wherein the agent is released in effective concentrations from the implant over a period ranging from about 1 month to 6 months.
9375. The method of item 9187 wherein the agent is delivered from the implant, wherein the agent is released in effective concentrations from the implant over a period ranging from about 1 - 90 days.
9376. The method of item 9187 wherein the agent is delivered from the implant, wherein the agent is released in effective concentrations from the implant at a constant rate.
9377. The method of item 9187 wherein the agent is delivered from the implant, wherein the agent is released in effective concentrations from the implant at an increasing rate.
9378. The method of item 9187 wherein the agent is delivered from the implant, wherein the agent is released in effective concentrations from the implant at a decreasing rate.
9379. The method of item 9187 wherein the agent is delivered from the implant, wherein the implant comprises about 0.01 Dg to about 10 Dg of the agent. 9380. The method of item 9187 wherein the agent is delivered from the implant, wherein the implant comprises about 10 Dg to about 10 mg of the agent.
9381. The method of item 9187 wherein the agent is delivered from the implant, wherein the implant comprises about 10 mg to about 250 mg of the agent.
9382. The method of item 9187 wherein the agent is delivered from the implant, wherein the implant comprises about 250 mg to about
1000 mg of the agent.
9383. The method of item 9187 wherein the agent is delivered from the implant, wherein the implant comprises about 1000 mg to about 2500 mg of the agent.
9384. The method of item 9187 wherein the agent is delivered from the implant, wherein a surface of the implant comprises less than 0.01 Dg of the agent per mm2 of implant surface to which the agent is applied.
9385. The method of item 9187 wherein the agent is delivered from the implant, wherein a surface of the implant comprises about 0.01 Dg to about 1 Dg of the agent per mm2 of implant surface to which the agent is applied.
9386. The method of item 9187 wherein the agent is delivered from the implant, wherein a surface of the implant comprises about 1 Dg to about 10 Dg of the agent per mm2 of implant surface to which the agent is applied. 9387. The method of item 9187 wherein the agent is delivered from the implant, wherein a surface of the implant comprises about 10 Dg to about 250 Dg of the agent per mm2 of implant surface to which the agent is applied.
9388. The method of item 9187 wherein the agent is delivered from the implant, wherein a surface of the implant comprises about 250 Dg to about 1000 Dg of the agent per mm2 of implant surface to which the agent is applied.
9389. The method of item 9187 wherein the agent is delivered from the implant, wherein a surface of the implant comprises about 1000 Dg to about 2500 Dg of the agent per mm2 of implant surface to which the agent is applied.
9390. The method of item 9187, wherein the implant further comprises a coating, and the coating is a uniform coating.
9391. The method of item 9187, wherein the implant further comprises a coating, and the coating is a non-uniform coating.
9392. The method of item 9187, wherein the implant further comprises a coating, and the coating is a discontinuous coating.
9393. The method of item 9187, wherein the implant further comprises a coating, and the coating is a patterned coating.
9394. The method of item 9187, wherein the implant further comprises a coating, and the coating has a thickness of 100 Dm or less. 9395. The method of item 9187, wherein the implant further comprises a coating, and the coating has a thickness of 10 Dm or less.
9396. The method of item 9187, wherein the implant further comprises a coating, and the coating adheres to the surface of the implant upon deployment of the implant.
9397. The method of item 9187, wherein the implant further comprises a coating, and the coating is stable at room temperature for a period of at least 1 year.
9398. The method of item 9187, wherein the implant further comprises a coating, and the agent is present in the coating in an amount ranging between about 0.0001% to about 1% by weight.
9399. The method of item 9187, wherein, the implant further comprises a coating, and the agent is present in the coating in an amount ranging between about 1% to about 10% by weight.
9400. The method of item 9187, wherein the implant further comprises a coating, and the agent is present in the coating in an amount ranging between about 10% to about 25% by weight.
9401. The method of item 9187, wherein the implant further comprises a coating, and the agent is present in the coating in an amount ranging between about 25% to about 70% by weight.
9402. The method of item 9187, wherein the implant further comprises a coating, and the coating comprises a polymer. 9403. The method of item 9187, wherein the implant comprises a first coating having a first composition and a second coating having a second composition.
9404. The method of item 9187, wherein the implant comprises a first coating having a first composition and a second coating having a second composition, wherein the first composition and the second composition are different.
9405. A method as in any one of items 9187-9404, wherein the device is a peritoneal dialysis catheter adapted for delivering a drug to the peritoneum.
9406. A method for inhibiting scarring comprising placing a device that comprises a central nervous system shunt or pressure monitor device (e.g., an implant) and an anti-scarring agent or a composition comprising an anti-scarring agent into an animal host, wherein the agent inhibits scarring.
9407. The method of item 9406 wherein the agent is an adensosine A2A receptor antagonist.
9408. The method of item 9406 wherein the agent is an AKT inhibitor.
9409. The method of item 9406 wherein the agent is an alpha 2 integrin antagonist, wherein the alpha 2 integrin antagonist is Pharmaprojects No. 5754 (Merck KgaA).
9410. The method of item 9406 wherein the agent is an alpha 4 integrin antagonist. 9411. The method of item 9406 wherein the agent is an alpha 7 nicotinic receptor agonist.
9412. The method of item 9406 wherein the agent is an angiogenesis inhibitor selected from the group consisting of AG-12,958 (Pfizer), ATN-161 (Attenuon LLC), neovastat, an angiogenesis inhibitor from Jerina AG (Germany), NM-3 (Mercian), VGA-1155 (Taisho), FCE-26644 (Pfizer), FCE-26950 (Pfizer), FPMA (Meiji Daries), FR-111142 (Fujisawa), GGTI-298, GM-1306 (Ligand), GPA-1734 (Novartis), NNC-47-0011 (Novo Nordisk), herbamycin (Nippon Kayaku), lenalidomide (Celegene), IP-10 (NIH), ABT-828 (Abbott), KIN-841 (Tokushima University, Japan), SF-1126 (Semafore Pharmaceuticals), laminin technology (NIH), CHIR-258 (Chiron), NVP-AEW541 (Novartis), NVP-AEW541 (Novartis), Vt16907 (Alchemia), OXI-8007 (Oxigene), EG-3306 (Ark Therapeutics), Maspin (Arriva), ABT- 567 (Abbott), PPI-2458 (Praecis Pharmaceuticals), CC-5079, CC-4089 (Celgene), HIF-1 alpha inhibitors (Xenova), S-247 (Pfizer), AP-23573 (Ariad), AZD-9935 (Astra Zeneca), mebendazole (Introgen Therapeutics), MetAP-2 inhibitors (GlaxoSmithKline), AG-615 (Angiogene Pharmaceuticals), Tie-2 antagonists (Hybrigenics), NC-381, CYC-381, NC-169, NC-219, NC-383, NC-384, NC-407 (Lorus Therapeutics), ATN-224 (Attenuon), ON-01370 (Onconova), Vitronectin antagonists (Amgen), SDX-103 (Salmedix), Vitronectin antagonists (Shire), CHP (Riemser), TEK (Amgen), Anecortave acetate (Alcon), T46.2 (Matrix Therapeutics), HG-2 (Heptagen), TEM antagonists (Genzyme), Oxi-4500 (Oxigene), ATN-161 (Attenuon), WX-293 (Wilex), M-2025 (Metris Therapeutics), Alphastatin (BioActa), YH-16 (Yantai Rongchang), BIBF-1120 (Boehringer Ingelheim), BAY-57-9352 (Bayer), AS- 1404 (Cancer Research Technology), SC-77964 (Pfizer), glycomimetics (BioTie Therapies), TIE-2 Inhibitors (Ontogen), DIMI, Octamer (Octamer), ABR-215050 (Active Biotech), ABT-518 (Abbott), KDR inhibitors (Abbott), BSF-466895 (Abbott), SCH-221153 (Schering-Plough), DAC:antiangiogenic (ConjuChem), TFPI (EntreMed), AZD-2171 (Astra-Zenaca), CDC-394 (Celgene), LY290293 (EIi Lilly), IDN-5390 (Indena), Kdr Kinase Inhibitors (Merck), CT-113020, CT-116433, CT-116563, CT-31890, CT-32228) (Cell Therapeutics), A-299620 (Abbott), TWEAK Inhibitor (Amgen), VEGF modulators (Johnson and Johnson), Tum-N53, tumstatin (Genzyme), Thios- 1 , Thios-2 (Thios Pharmaceuticals), MV-6401 (Miravant Medical Technologies), Spisulosine (PharmaMar), CEP-7055 (Cephalon), AUV-201 (Auvation), LM-609 (EIi Lilly), SKF-106615 (AnorMED), Oglufanide disodium (Cytran), BW-114 (Pharminox), Calreticulin (NIH), WX-678 (Wilex), SD-7784 (Pfizer), WX-UK1 (Wilex), SH-268 (Schering AG), 2-Me-PGA (Celgene), S- 137 (Pfizer), ZD-6126 (Angiogene Pharmaceuticals), SG-292 (SignalGen), Benefin (Lane Labs), A6, A36 (Angstrom), SB-2723005 (GlaxoSmithKline), SC-7 (Cell Therapeutics), ZEN-014 (AEterna Zentaris), 2-methoxyestradiol (EntreMed), NK-130119 (Nippon Kayaku), CC-10004 (Celgene), AVE- 8062A (Ajinomoto), Tacedinaline (Pfizer), Actinonin (Tokyo Metropolitan Institute of Medical Science), Lenalidomide (Celgene), VGA-1155, BTO-956 (SRI International), ER-68203-00 (Eisai), CT-6685 (UCB), JKC-362 (Phoenix Pharmaceuticals), DMI-3798 (DMI Biosciences, Angiomate (Ipsen), ZD-6474 (AstraZeneca), CEP-5214 (Cephalon), Canstatin (Genzyme), NM-3 (Mercian), Oridigm (MediQuest Therapeutics), Exherin (Adherex), BLS-0597 (Boston Life Sciences), PTC-299 (PTC Therapeutics), NPI-2358 (Nereus Pharmaceuticals), CGP-79787 (Novartis), AEE-788 (Novartis), CKD-732 (Chong Kun Dang), CP-564959 (OSI Pharmaceuticals), CM-101 (CarboMed), CT-2584, CT3501 (Cell Therapeutics), combretastatin and analogues and derivatives thereof (Oxigene), Rebimastat (Bristol-Meyers Squibb), Dextrin 2-sulfate (ML Laboratories), Cilengitide (Merk KGaA), NSC-706704 (Pharminox), KRN- 951 (Kirin Brewery), Ukrain, NSC-631570 (Nowicky Pharma), Tecogalan sodium (Daiichi Pharmaceutical), Tz-93 (Tsumura), TBC-1635 (Encysive Pharmaceuticals), TAN-1120 (Takeda), Semaxanib (Pfizer), BDI-7800 (Biopharmacopae), SD-186, SD-983 (Bristol-Meyers Squibb), SB-223245 (GlaxoSmithKline), SC-236 (Pfizer), RWJ-590973 (Johnson and Johnson), ILX-1850 (Genzyme), SC-68488, S-836 (Pfizer), CG-55069-11 (CuraGen), Ki-23057 (Kirin Brewery), CCX-700 (Chemoentryx), Pegaptanib octasodium (Giled Sciences), ANGIOCOL (available from Biostratum Inc.), or an analogue or derivative thereof.
9413. The method of item 9406 wherein the agent is an apoptosis antagonist.
9414. The method of item 9406 wherein the agent is an apoptosis activator.
9415. The method of item 9406 wherein the agent is a beta 1 integrin antagonist.
9416. The method of item 9406 wherein the agent is a beta tubulin inhibitor. . _ _ . . . .
9417. The method of item 9406 wherein the agent is a blocker of enzyme production in Hepatitis C.
9418. The method of item 9406 wherein the agent Is a Bruton's tyrosine kinase inhibitor.
9419. The method of item 9406 wherein the agent is a calcineurin inhibitor.
9420. The method of item 9406 wherein the agent is a caspase 3 inhibitor.
9421. The method of item 9406 wherein the agent is a CC chemokine receptor antagonist. 9422. The method of item 9406 wherein the agent is a cell cycle inhibitor selected from the group consisting of SNS-595 (Sunesis), synthadotin, KRX-0403, and an analogue or derivative thereof.
9423. The method of item 9406 wherein the agent is a cathepsin B inhibitor.
9424. The method of item 9406 wherein the agent is a cathepsin K inhibitor, wherein the cathepsin K inhibitor is 462795 (GlaxoSmithKline), INPL-022-D6 (Amura Therapeutics), or an analogue or derivative thereof.
9425. The method of item 9406 wherein the agent is a cathepsin L inhibitor.
9426. The method of item 9406 wherein the.agent is a CD40 antagonist.
9427. The method of item 9406 wherein the agent is a chemokine receptor agonist.
9428. The method of item 9406 wherein the agent is a chymase inhibitor.
9429. The method of item 9406 wherein the agent is a collagenase antagonist.
9430. The method of item 9406 wherein the agent is a CXCR antagonist. 9431. The method of item 9406 wherein the agent is a cyclin dependent kinase inhibitor selected from the group consisting of a CDK-1 inhibitor, a CDK-2 inhibitor, a CDK- 4 inhibitor, a CDK-6 inhibitor, a CAK1 inhibitor from GPC Biotech and Bristol-Myers Squibb, RGB-286199 (GPC Biotech), an anticancer agent from Astex Technology, a CAK1 inhibitor from GPC Biotech, a CDK inhibitor from Sanofi-Aventis, a CDK1/CDK2 inhibitor from Amgen, a CDK2 inhibitor from SUGEN-2 (Pfizer), a hearing loss therapy agent (Sound Pharmaceuticals), PD-0332991 (Pfizer), RGB-286199 (GPC Biotech), Ro-0505124 (Hoffmann-La Roche), a Ser/Thr kinase inhibitor from Lilly (EIi Lilly), CVT-2584 (CAS No. 199986-75-9) (CV Therapeutics), and an analogue or derivative thereof.
9432. The method of item 9406 wherein the agent is a cyclooxygenase 1 inhibitor.
9433. The method of item 9406 wherein the agentis a.DHFR inhibitor.
9434. The method of item 9406 wherein the agent is a dual integrin inhibitor.
9435. The method of item 9406 wherein the agent is an elastase inhibitor.
9436. The method of item 9406 wherein the agent is an elongation factor-1 alpha inhibitor.
9437. The method of item 9406 wherein the agent is an endothelial growth factor antagonist. 9438. The method of item 9406 wherein the agent is an endothelial growth factor receptor kinase inhibitor selected from the group consisting of sorafenib tosylate (Bayer), AAL-993 (Novartis), ABP-309 (Novartis), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), E- 7080 (Eisai), EG-3306 (Ark Therapeutics), EXEL-2880 (Exelixis), GW- 654652 (GlaxoSmithKline), a KDR inhibitor from LG Life Sciences, CT-6685 and CT-6729 (UCB), KRN-633 and KRN-951 (Kirin Brewery), OSI-930 (OSI Pharmaceuticals), SP-5.2 (Supratek Pharma), SU-11657 (Pfizer), a Tie-2 antagonist (Hybrigenics), a VEGFR-2 kinase inhibitor (Bristol-Myers Squibb), XL-647 (Exelixis), a KDR inhibitor from Abbott Laboratories, sorafenib tosylate, and an analogue or derivative thereof.
9439. The method of item 9406 wherein the agent is an endotoxin antagonist.
9440. The method of item 9406 wherein the agent is an epothilone and tubulin binder.
9441. The method of item 9406 wherein the agent is an estrogen receptor antagonist.
9442. The method of item 9406 wherein the agent is an FGF inhibitor.
9443. The method of item 9406 wherein the agent is a famexyl transferase inhibitor.
9444. The method of item 9406 wherein the agent is famesyltransferase inhibitor selected from the group of A-197574 (Abbott), a farnesyltransferase inhibitor from Servier, FPTIII (Strathclyde Institute for Drug R), LB-42908 (LG Life Sciences), Pharmaprojects No. 5063 (Genzyme), Pharmaprojects No. 5597 (Ipsen), Yissum Project No. B-1055 (Yissum), and an analogue or derivative thereof.
9445. The method of item 9406 wherein the agent is an FLT-3 kinase inhibitor.
9446. The method of item 9406 wherein the agent is an FGF receptor kinase inhibitor.
9447. The method of item 9406 wherein the agent is a fibrinogen antagonist selected from the group consisting of AUV-201 (Auvation), MG-13926 (Sanofi-Aventis), plasminogen activator (CAS No. 105913-11-9) (from Sanofi-Aventis or UCB), plasminogen activator-2 (tPA- 2) (Sanofi-Aventis), pro-urokinase (CAS No. 82657-92-9) (Sanofi-Aventis), and an analogue or derivative thereof.
9448. The method of item 9406 wherein the agent is a heat shock protein 90 antagonist selected from the group consisting of SRN-005 (Sirenade), geldanamycin, NSC-33050 (17-allylaminogeldanamycin; 17- AAG), ^-dimethylaminoethylamino-^-demethoxy-geldanamycin (17- DMAG), rifabutin (rifamycin XIV, 1',4-didehydro-1-deoxy-1,4-dihydro-5'-(2- methylpropyl)-1-oxo-), and an analogue or derivative thereof.
9449. The method of item 9406 wherein the agent is a histone deacetylase inhibitor.
9450. The method of item 9406 wherein the agent is an HMGCoA reductase inhibitor selected from the group consisting of an atherosclerosis therapeutic from Lipid Sciences, ATI-16000 (ARYx Therapeutics), KS-01-019 (Kos Pharmaceuticals), Pharmaprojects No. 2197 (Sanofi-Aventi), RP 61969 (Sanofi-Aventis), and an analogue or derivative thereof.
9451. The method of item 9406 wherein the agent is an ICAM inhibitor.
9452. The method of item 9406 wherein the agent is an IL, ICE and IRAK antagonist, wherein the antagonist is a CJ-14877, CP-424174 (Pfizer), NF-61 (Negma-Lerads), and an analogue or derivative thereof.
9453. The method of item 9406 wherein the agent is an IL-2 inhibitor.
9454. The method of item 9406 wherein the agent is an immunosuppressant selected from the group consisting of teriflunomide (Sanofi Aventis), chlorsulfaquinoxalone (NSC-339004), chlorsulfaquinoxalone sulfate, CS-712 (Sankyo), ismomultin alfa (CAS No. 457913-93-8) (Akzo Nobel), antiallergics from GenPat77, antiinflammatories or AT-005 (Androclus Therapeutics), autoimmune disease therapeutics from EpiVax, BN-007 (Bone), budesonide (CAS No. 51333-22- 3) (MAP Pharmaceuticals), CO-14 (Genzyme), edratide (CAS No. 433922- 67-9) (Teva), EP-314 (Enanta), eprovafen (CAS No. 101335-99-3) (Sanofi- Aventis), HWA-131 (CAS No. 118788-41-3) (Sanofi-Aventis), immunomodulators from MerLion Pharmaceuticals, immunosuppressives from Alchemia, IPL-12 (Inflazyme), MDL-9563 (CAS No. 27086-86-8) (Sanofi-Aventis), Pharmaprojects No. 2330 (Sanofi-Aventis), Pharmaprojects No. 6426 (Abgenϊx), PXS-25 (Pharmaxis), rosmarinic acid (CAS No. 20283-92-5) (Sanofi-Aventis), RP 42927 or RP 54745 (CAS No. 135330-08-4) (Sanofi-Aventis), SGN-35 (Seattle Genetics), ST-1959 (Sigma-Tau), type I diabetes therapy from SYNX Pharma, UNIL-88 (Debiopharm), VP-025 (Vasogen), VR-694 (Vectura), PRTX-001 (Protalex), and an analogue or derivative thereof.
9455. The method of item 9406 wherein the agent is an IMPDH (inosine monophosphate).
9456. The method of item 9406 wherein the agent is an integrin antagonist.
9457. The method of item 9406 wherein the agent is an interleukin antagonist.
9458. The method of item 9406 wherein the agent is an inhibitor of type III receptor tyrosine kinase.
9459. The method of item 9406 wherein the_agent is an irreversible inhibitor of enzyme methionine aminopeptidase type 2.
9460. The method of item 9406 wherein the agent is an isozyme selective delta protein kinase C inhibitor.
9461. The method of item 9406 wherein the agent a JAK3 enzyme inhibitor.
9462. The method of item 9406 wherein the agent is a JNK inhibitor.
9463. The method of item 9406 wherein the agent is a kinase inhibitor. 9464. The method of item 9406 wherein the agent is kinesin antagonist.
9465. The method of item 9406 wherein the agent is a kinesin antagonist.
9466. The method of item 9406 wherein the agent is a leukotriene inhibitor and antagonist selected from the group consisting of ambicromil (CAS No. 58805-38-2) (Sanofi-Aventis), ameiubant (CAS No. 346735-24-8) (Boehringer Ingelheim), DW-1141 (Dong Wha), ebselen (Daiichi Pharmaceutical), ibudilast (Kyorin), leucotriene inhibitors from Sanofi-Aventis, lymphotoxin-beta receptor (LT-D) from Biogen Idee, Pharmaprojects No. 1535 and 2728 (CAS No. 119340-33-9) (Sanofi- Aventis), R-112 (Rigel), Rev-5367 (CAS No. 92532-05-3) (Sanofi-Aventis), RG-14893 (CAS No. 141835-49-6) (Sanofi-Aventis), RG-5901-A (CAS No. 101910-24-1), 92532-23-5, RP 66153 (CAS No. 142422-79-5), RP 66364 (CAS No.186916-92-5), RP 69698 (CAS No. 141748-00-7) (Sanofi-Aventis), SC-411930 (Pfizer), SC-41930 (CAS No. 120072-59-5) (Pfizer), SC-50605 (CAS No. 138828-39-4) (Pfizer), SC-51146 (CAS No. 141059-52-1), SC- 53228 (CAS No. 153633-01-3) (Pfizer), spaglumic acid (ZY-15106) (CAS No.3106-85-2), 80619-64-3 (Novartis), tipredane (CAS No. 85197-77-9) (Bristol-Myers Squibb), U-75302 (CAS No. 119477-85-9) (Pfizer), and analogue or derivative thereof.
9467. The method of item 9406 wherein the agent is an MAP kinase inhibitor.
9468. The method of item 9406 wherein the agent is a matrix metalloproteinase inhibitor. 9469. The method of item 9406 wherein the agent is an MCP- CCR2 inhibitor.
9470. The method of item 9406 wherein the agent is an mTOR inhibitor.
9471. The method of item 9406 wherein the agent is an mTOR kinase inhibitor.
9472. The method of item 9406 wherein the agent is a microtubule inhibitor selected from the group consisting of antibody- maytansinoid conjugates from Biogen Idee, colchicines (MantiCore Pharmaceuticals), anticancer immunoconjugates from Johnson & Johnson, DIME from Octamer, gni-1f (GNI), huC242-DM4, huMy9-6-DM1 (ImmunoGen), IDN-5404 (Indena), IMO-098, IMOderm (Imotep), mebendazole (Introgen Therapeutics), microtubule poisons from Cambridge Enterprise, paclitaxel such as LOTAX from Aphios (CAS No. 33069-62-4), Genexol-PM from Samyang, Pharmaprojects No. 6383 (Tapestry Pharmaceuticals), RPR-112378 (Sanofi-Aventis), SGN-75 (Seattle Genetics), SPL-7435 (Starpharma), SSR-250411 (Sanofi-Aventis), trastuzumab-DM1 (Genentech), vinorelbine, and an analogue or derivative thereof.
9473. The method of item 9406 wherein the agent is an MIF inhibitor.
9474. The method of item 9406 wherein the agent is an MMP inhibitor.
9475. The method of item 9406 wherein the agent is a neurokinin (NK) antagonist selected from the group consisting of anthrotainin (CAS No. 148084-40-6) (Sanofi-Aventis), an IBS thereapeutic from ArQuIe, MDL-105212A (CAS No. 167261-60-1) (Ssanofi-Aventis), Pharmaprojects No. 2744, 3258 (CAS No. 139167-47-8) 4006, 4201, or 5986 (Sanofi-Aventis), RP 67580 (CAS No. 135911-02-3), SR-144190 (CAS No. 201152-86-5), SSR-240600, SSR-241586 (Sanofi-Aventis), TKA-457 (Novartis), vestipitant mesylate (CAS No. 334476-64-1) (GlaxoSmithKline), Wϊn-64821 (Sanofi-Aventis), PRX-96026 (Predix Pharmaceuticals), and an analogue or derivative thereof.
9476. The method of item 9406 wherein the agent is an NF kappa B inhibitor selected from the group consisting of AVE-0545 or AVE- 0547 (Sanofi-Aventis), bortezomib (CAS No. 179324-69-7) (Millennium Pharmaceuticals), dexanabinol (CAS No. 112924-45-5) (Pharmos), dexlipotam (Viatris), Pharmaprojects No. 6283 (INDRA) (OXiGENE), IPL- 576092 (CAS No. 137571-30-3) (Inflazyme), NFKB decoy (Corgentech), NFKB decoy oligo (AnGes MG), NFKB's from Ariad, osteoporosis treatments or S5 (F005) from Fulcrum Pharmaceuticals, P61 (Phytopharm), R-flurbiprofen (CAS No. 5104-49-4) (Encore Pharmaceuticals), and an analogue or derivative thereof.
9477. The method of item 9406 wherein the agent is a nitric oxide agonist.
9478. The method of item 9406 wherein the agent is an ornithine decarboxylase inhibitor.
9479. The method of item 9406 wherein the agent is a p38 MAP kinase inhibitor selected from the group consisting of AZD-6703 (AstraZeneca), JX-401 (Jexys Pharmaceuticals), BMS-2 (Bristol-Myers Squibb), a p38 MAP kinase inhibitor from Novartis, a p38-alpha MAP kinase inhibitor from Amphora, Pharmaprojects No. 5704 (Pharmacopeia), RPR- 200765A (Sanofi-Aventis), SD-282 (Johnson & Johnson), TAK-715 (Takeda), and an analogue or derivative thereof.
9480. The method of item 9406 wherein the agent is a palmitoyl-protein thioesterase inhibitor.
9481. The method of item 9406 wherein the agent is a PDGF receptor kinase inhibitor selected from the group consisting of AAL-993, AMN-107, or ABP-309 (Novartis), AMG-706 (Amgen), BAY-57-9352 (Bayer), CDP-860 (UCB), E-7080 (Eisai), imatinib (CAS No. 152459-95-5) (Novartis), OSI-930 (OSI Pharmaceuticals), RPR-127963E (Sanofi-Aventis), RWJ-540973 (Johnson & Johnson), sorafenib tosylate (Bayer), SU-11657 (Pfizer), tandutinib (CAS No. 387867-13-2) (Millennium Pharmaceuticals), vatalanib (Novartis), ZK-CDK (Schering AG), and an analogue or derivative thereof.
9482. The method of item 9406 wherein the agent is (-)- halofenate (Metabolex), AMG-131 (Amgen), antidiabetics from Japan Tobacco, AZD-4619, AZD-8450, AZD-8677 (AstraZeneca), DRF-10945, balaglitazone (Dr Reddy's), CS-00088, CS-00098 (Chipscreen Biosciences), E-3030 (Eisai), etalocib (CAS No. 161172-51-6) (EIi Lilly), GSK-641597 (Ligand), GSK-677954 (GlaxoSmithKline), GW-409544 (Ligand), GW- 590735 (GlaxoSmithKline), K-111 (Hoffmann-La Roche), LY-518674 (EIi Lilly), LY-674 (Ligand), LY-929 (Ligand), MC-3001, MC-3002 (MaxoCore Pharmaceuticals), metformin HCi + pioglitazone (CAS No. 1115-70-4 and 112529-15-4), ACTOPLUS MET from Andrx), muraglitazar (CAS No. 331741-94-7) (Bristol-Myers Squibb), naveglitazar (Ligand), oleoylethanolamide (Kadmus Pharmaceuticals), ONO-5129, pioglitazone hydrochloride (CAS No. 111025-46-8 and 112529-15-4) (Takeda), PLX-204 (Plexxikon), PPAR agonists from Genfit, PPAR delta agonists from EIi Lilly, PPAR-alpha agonists from CrystalGenomics, PPAR-gamma modulators and PPAR-β modulators from CareX, rosiglitazone maleate (CAS No. 122324- 73-4 or 155141-29-0) (GlaxoSmithKline), rosiglitazone maleate/glimepir (CAS No. 155141-29-0 and 93479-97-1), AVANDARYL, rosiglitazone maleate/metformin extend (CAS No. 155141-29-0 and 657-24-9), AVANDAMET, rosiglitazone maleate+metformin, AVANDAMET (GlaxoSmithKline), tesaglitazar (AstraZeneca), and an analogue or derivative thereof.
9483. The method of item 9406 wherein the agent is a phosphatase inhibitor.
9484. The method of item 9406 wherein the agent is a phosphodiesterase (PDE) inhibitor selected from the group consisting of avanafil (Tanabe Seiyaku), dasantafil (CAS No. 569351-91-3) (Schering- Plough), A-906119 (CAS No. 134072-58-5), DL-850 (Sanofi-Aventis), GRC- 3015, GRC-3566, GRC-3886 (Glenmark), HWA-153 (CAS No. 56395-66-5) (Sanofi-Aventis), hydroxypumafentrine (Altana), IBFB-130011, IBFB-14-016, IBFB-140301 , IBFB-150007, IBFB-211913 (IBFB Pharma), L-826141 (Merck & Co), medorinone (CAS No. 88296-61-1) (Sanofi-Aventis), MEM- 1917 (Memory Pharmaceuticals), ND-1251 (Neuro3d), PDE inhibitors from ICOS, PDE IV inhibitors from Memory Pharmaceuticals and CrystalGenomics, Pharmaprojects No. 2742 and 6141 (Sanofi-Aventis), QAD-171 (Novartis), RHC-2963 (CAS No. 76993-12-9 and 76993-14-1), RPR-117658, RPR-122818 derivatives, SR-24870 , and RPR-132294 (Sanofi-Aventis), SK-350 (ln2Gen), stroke therapy agents from deCODE Genetics, TAS-203 (Taiho), tofimilast (CAS No. 185954-27-2) (Pfizer), UK- 371800 (Pfizer), WIN-65579 (CAS No. 158020-82-7) (Sanofi-Aventis), IBFB- 130020 (IBFB Pharma), OPC-6535 (CAS No. 145739-56-6) (Otsuka), a phosphodiesterase III inhibitor, enoximone, a phosphodiesterase IV inhibitor, fosfosal, Atopik (Barrier Therapeutics), triflusal, a phosphodiesterase V inhibitor, and an analogue or derivative thereof. 9485. The method of item 9406 wherein the agent is a PKC inhibitor.
9486. The method of item 9406 wherein the agent is a platelet activating factor antagonist.
9487. The method of item 9406 wherein the agent is a platelet-derived growth factor receptor kinase inhibitor.
9488. The method of item 9406 wherein the agent is a prolyl hydroxylase inhibitor.
9489. The method of item 9406 wherein the agent is a polymorphonuclear neutrophil inhibitor.
9490. The method of item 9406 wherein the agent is a protein kinase B inhibitor.
9491. The method of item 9406 wherein the agent is a protein kinase C stimulant.
9492. The method of item 9406 wherein the agent is a purine nucleoside analogue.
9493. The method of item 9406 wherein the agent is a purinoreceptor P2X antagonist.
9494. The method of item 9406 wherein the agent is a Raf kinase inhibitor. 9495. The method of item 9406 wherein the agent is a reversible inhibitor of ErbB1 and ErbB2.
9496. The method of item 9406 wherein the agent is a ribonucleoside triphosphate reductase inhibitor.
9497. The method of item 9406 wherein the agent is an SDF- 1 antagonist.
9498. The method of item 9406 wherein the agent is a sheddase inhibitor.
9499. The method of item 9406 wherein the agent is an SRC inhibitor.
9500. The method of item 9406 wherein the agent is a stromelysin inhibitor.
9501. The method of item 9406 wherein the agent is an Syk kinase inhibitor.
9502. The method of item 9406 wherein the agent is a telomerase inhibitor.
9503. The method of item 9406 wherein the agent is a TGF beta inhibitor selected from the group consisting of pirfenidone (CAS No. 53179-13-8) (MARNAC), tranilast (CAS No. 53902-12-8) (Kissei), IN-1130 (ln2Gen), mannose-6-phosphate (BTG), TGF-β antagonists from Inflazyme (Pharmaprojects No. 6075), TGF-β antagonists from Sydney, non-industrial source), TGF-β I receptor kinase inhibitors from EIi Lilly, TGF-β receptor inhibitors from Johnson & Johnson, and an analogue or derivative thereof. 9504. The method of item 9406 wherein the agent is a TNFα antagonist or TACE inhibitor selected from the group consisting of adalimumab (CAS No. 331731-18-1) (Cambridge Antibody Technology), AGlX-4207 (AtheroGenics), AGT-1 (Advanced Biotherapy), an antiinflammatory from Borean Pharma, Cellzome, or Paradigm Therapeutics, anti-inflammatory vaccine (TNF-alpha kinoid) from Neovacs, humanized anti-TNF antibody or an anti-TNF MAb (CB0006) Celltech (UCB), apratastat (CAS No. 287445-51-0) (Wyeth), BMS-561392 (Bristol-Myers Squibb), BN- 006 (Bone), certolizumab pegol (CAS No. 428863-50-7 or CH-138 (UCB), cilomilast (CAS No. 153259-65-5) (GlaxoSmithKline), CR-1 (Nuada Pharmaceuticals), CRx-119 (CombinatoRx), D-5410 (UCB), dacopafant (CAS No. 125372-33-0) (Sanofi-Aventis), dersalazine (CAS No. 188913-57- 7/188913-58-8) (Uriach), etanercept (CAS No. 185243-69-0) (Amgen), ethyl pyruvate (Critical (Critical Therapeutics), golimumab (CAS No. 476181-74-5) (Johnson & Johnson), hormono-immunotherapy from Ipsen, CDP571 (e.g., Humicade from UCB), IC-485 (ICOS), infliximab (CAS No. 17O277-31-3) . (Johnson & Johnson), IP-751 (Manhattan Pharmaceuticals), ISIS-104838 (CAS No. 250755-32-9) (ISIS Pharmaceuticals), lenalidomide (CAS No. 191732-72-6) (Celgene), lentinan (CAS No. 37339-90-5) (Ajinomoto), MDL- 201112 (CAS No. 142130-73-2) (Sanofi-Aventis), medroxyprogesterone (CAS No. 520-85-4) (InKine Pharmaceutical), N-acetylcysteine (CAS No. 616-91-1) (Zambon), NBE-P2 (DIREVO Biotech), nerelimomab (CAS No. 162774-06-3) (Chiron), OM-294DP (OM PHARMA), onercept (CAS No. 199685-57-9) (Yeda), PASSTNF-alpha (Verigen), pentoxifylline or oxypentifylline (Sanofi-Aventis), Pharmaprojects No. 4091 , 4241 , 4295, or 4488 (Sanofi-Aventis), Pharmaprojects No. 5480 (Amgen), Pharmaprojects No. 6749 (Cengent), pirfenidone (CAS No. 53179-13-8) (MARNAC), RPR- 132294 (Sanofi-Aventis), S5 (F002) (Fulcrum Pharmaceuticals), simvastatin (CAS No. 79902-63-9) (Merck & Co), STA-6292 (Synta Pharmaceuticals), tacrolimus (CAS No. 104987-11-3) (Fujisawa LifeCycle Pharma), talactoferrin alfa (CAS No. 308240-58-6) (Agennix), thalidomide (CAS No. 50-35-1) (Celgene), TNF antagonists form ProStrakan, and Synergen, TNF inhibitors (Amgen), TNF-alpha antagonists from Dynavax Technologies and Jerina AG (Germany), TNF-alpha inhibitors from IBFB Pharma and Xencor (Xencor), torbafylline (CAS No. 105102-21-4) (Sanofi-Aventis), UR-1505 (Uriach), VT-346 (Viron Therapeutics), YSIL6 (Y1S Therapeutics), YSTH2 (Y's Therapeutics), NPI-1302a-3 (Nereus Pharmaceuticals, a TNF antagonist from Jerina AG (Germany), dersalazine, and an analogue or derivative thereof.
9505. The method of item 9406 wherein the agent is a Toll receptor inhibitor.
9506. The method of item 9406 wherein the agent is a tubulin antagonist.
9507. The method of item 9406 wherein the agent is .a. tyrosine kinase inhibitor selected from the group consisting of SU-011248, SUTENT from Pfizer Inc. (New York, NY), BMS-354825, PN-355 (Paracelsian Pharmaceuticals), AGN-199659 (Allergan), AAL-993 or ABP- 309 (Novartis), adaphostin (NIH), AEE-788 (Novartis), AG-013736 (OSI Pharmaceuticals), AG-13736 (Pfizer), ALT-110 (Alteris Therapeutics), AMG- 706 (Amgen), anticancer MAbs from Xencor, anti-EGFrvlll MAbs from Abgenix, anti-HER2 MAb from Abiogen, AZD-2171 or AZD-9935 (AstraZeneca), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), CEP-5214 (Cephalon), CEP-7055 (Cephalon), cetuximab (ImClone Systems), CHIR-200131 and CHIR-258 (Chiron), CP-547632 (OSI Pharmaceuticals), CP-724714 (Pfizer), CT-301 (Creabilis Therapeutics), D- 69491 (Baxter International), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EGFR/ErbB2 inhibitors from Array BioPharma, erlotinib (CAS No. 183319- 69-9) (OSI Pharmaceuticals), EXEL-2880 (Exelixis), FK-778 (Sanofi- Aventis) , gefitinib (CAS No. 184475-35-2) (AstraZeneca), GW-2286 or GW- 654652 (GlaxoSmithKline), her2/neu antigen from AlphaVax, HER-2/neu inhibitor from Generex, Herzyme (Medipad) (Sirna Therapeutics), HKI-272 (Wyeth), HuMax-EGFr (Genmab), idronoxil (CAS No. 81267-65-4) (Novogen), IGF-1 inhibitors from Ontogen, IMC-11F8 (ImCIone Systems), kahalalide F (CAS No. 149204-42-2) (PharmaMar), KDR inhibitor from LG Life Sciences, KDR inhibitors from Abbott Laboratories, KDR kinase inhibitors (UCB), Kdr kinase inhibitors from Merck & Co, KRN-633 and KRN- 951 (Kirin Brewery), KSB-102 (Xenova), lapatinib ditosylate (CAS No. 388082-78-8) (GlaxoSmithKline), matuzumab (Merck KGaA), MDX-214 (Medarex), ME-103 (Pharmexa), MED-A300 (Gerolymatos), MNAC-13 (Lay Line Genomics), nimotuzumab (Center of Molecular Immunology), NSC- 330507 or NSC-707545 (NIH), NV-50 (Novogen), OSI-930 (OSI Pharmaceuticals), panitumumab (Abgenix), pelitinib (CAS No. 287933-82-7) (Wyeth), pertuzumab (CAS No. 380610-27-5) (Genentech), Pharmaprojects No. 3985 (Sanofi-Aventis), prostate cancer therapeutics from Sequenom (SQPC35, SQPC36, SQPC90), removab and re.mox.ab (Trion Pharma), RG- 13022 (CAS No. 136831-48-6), RG-13291 (CAS No. 138989-50-1), or RG- 14620 (CAS No. 136831-49-7) (Sanofi-Aventis), RM-6427 (Romark), RNAi breast cancer therapy from Benitec, RP 53801 (CAS No. 125882-88-4) (Sanofi-Aventis), sorafenib tosylate (Bayer), SU-11657 (Pfizer), Tie-2 antagonists from Semaia (Hybrigenics), Tie-2 inhibitors from Ontogen, trastuzumab (CAS No. 180288-69-1) (Genentech), tyrosine kinase inhibitors from Sanofi-Aventis, U3-1287, U3-1565, U3-1784, U3-1800 (U3 Pharma), vatalanib (Novartis), VEGFR-2 kinase inhibitor from Bristol-Myers Squibb, XL-647 (Exelixis), ZD-6474 (AstraZeneca), ZK-CDK (Schering AG), an EGFR tyrosine kinase inhibitor, EKB-569 (Wyeth), and an analogue or derivative thereof.
9508. The method of item 9406 wherein the agent is a VEGF inhibitor. 9509. The method of item 9406 wherein the agent is a vitamin D receptor agonist.
9510. The method of item 9406 wherein the agent is ZD-6474 (an angiogenesis inhibitor).
9511. The method of item 9406 wherein the agent is AP- 23573 (an mTOR inhibitor).
9512. The method of item 9406 wherein the agent is synthadotin (a tubulin antagonist).
9513. The method of item 9406 wherein the agent is S-0885 (a collagenase inhibitor).
9514. The method of item 9406 wherein the agent is apϋdine (an elongation factor-1 alpha inhibitor).
9515. The method of item 9406 wherein the agent is ixabepilone (an epithilone).
9516. The method of item 9406 wherein the agent is IDN- 5390 (an angiogenesis inhibitor).
9517. The method of item 9406 wherein the agent is SB- 2723005 (an angiogenesis inhibitor).
9518. The method of item 9406 wherein the agent is ABT-518 (an angiogenesis inhibitor). 9519. The method of item 9406 wherein the agent is combretastatin (an angiogenesis inhibitor).
9520. The method of item 9406 wherein the agent is anecortave acetate (an angiogenesis inhibitor).
9521. The method of item 9406 wherein the agent is SB- 715992 (a kinesin antagonist).
9522. The method of item 9406 wherein the agent is temsirolimus (an mTOR inhibitor).
9523. The method of item 9406 wherein the agent is adalimumab (a TNFα antagonist).
9524. The method of item 9406, wherein the composition comprises a polymer.
9525. The method of item 9406, wherein the composition comprises a polymer, and the polymer is, or comprises, a copolymer.
9526. The method of item 9406, wherein the composition comprises a polymer, and the polymer is, or comprises, a block copolymer.
9527. The method of item 9406, wherein the composition comprises a polymer, and the polymer is, or comprises, a random copolymer.
9528. The method of item 9406, wherein the composition comprises a polymer, and the polymer is, or comprises, a biodegradable polymer. 9529. The method of item 9406, wherein the composition comprises a polymer, and the polymer is, or comprises, a nonbiodegradable polymer.
9530. The method of item 9406, wherein the composition comprises a polymer, and the polymer is, or comprises, a hydrophilic polymer.
9531. The method of item 9406, wherein the composition comprises a polymer, and the polymer is, or comprises, a hydrophobic polymer.
9532. The method of item 9406, wherein the composition comprises a polymer, and the polymer is, or comprises, a polymer having hydrophilic domains.
9533. The method of item 9406, wherein the composition comprises a polymer, and the polymer is, or comprises, a polymer having hydrophobic domains.
9534. The method of item 9406, wherein the composition comprises a polymer, and the polymer is, or comprises, a non-conductive polymer.
9535. The method of item 9406, wherein the composition comprises a polymer, and the polymer is, or comprises, an elastomer.
9536. The method of item 9406, wherein the composition comprises a polymer, and the polymer is, or comprises, a hydrogel. 9537. The method of item 9406, wherein the composition comprises a polymer, and the polymer is, or comprises, a silicone polymer.
9538. The method of item 9406, wherein the composition comprises a polymer, and the polymer is, or comprises, a hydrocarbon polymer.
9539. The method of item 9406, wherein the composition comprises a polymer, and the polymer is, or comprises, a styrene-derived polymer.
9540. The method of item 9406, wherein the composition comprises a polymer, and the polymer is, or comprises, a butadiene-derived polymer.
9541. The method of item 9406, wherein the composition, comprises a polymer, and the polymer is, or comprises, a macromer.
9542. The method of item 9406, wherein the composition comprises a polymer, and the polymer is, or comprises, a poly(ethylene glycol) polymer.
9543. The method of item 9406, wherein the composition comprises a polymer, and the polymer is, or comprises, an amorphous polymer.
9544. The method of item 9406, wherein the composition further comprises a second pharmaceutically active agent.
9545. The method of item 9406, wherein the composition further comprises an anti-inflammatory agent. 9546. The method of item 9406, wherein the composition further comprises an agent that inhibits infection.
9547. The method of item 9406, wherein the composition further comprises an anthracycline.
9548. The method of item 9406, wherein the composition further comprises doxorubicin.
9549. The method of item 9406 wherein the composition further comprises mitoxantrone.
9550. The method of item 9406 wherein the composition further comprises a fluoropyrimidine.
9551. The method of item 9406, wherein the composition further comprises 5-fluorouracil (5-FU).
9552. The method of item 9406, wherein the composition further comprises a folic acid antagonist.
9553. The method of item 9406, wherein the composition further comprises methotrexate.
9554. The method of item 9406, wherein the composition further comprises a podophylotoxin.
9555. The method of item 9406, wherein the composition further comprises etoposide. 9556. The method of item 9406, wherein the composition further comprises camptothecin.
9557. The method of item 9406, wherein the composition further comprises a hydroxyurea.
9558. The method of item 9406, wherein the composition further comprises a platinum complex.
9559. The method of item 9406, wherein the composition further comprises cisplatin.
9560. The method of item 9406 wherein the composition further comprises an anti-thrombotic agent.
9561. The method of item 9406, wherein the composition further comprises a visualization agent.
9562. The method of item 9406, wherein the composition further comprises a visualization agent, and the visualization agent is a radiopaque material, wherein the radiopaque material comprises a metal, a halogenated compound, or a barium containing compound.
9563. The method of item 9406, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, barium, tantalum, or technetium.
9564. The method of item 9406, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, an MRI responsive material. 9565. The method of item 9406, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, a gadolinium chelate.
9566. The method of item 9406, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, iron, magnesium, manganese, copper, or chromium.
9567. The method of item 9406, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, iron oxide compound.
9568. The method of item 9406, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, a dye, pigment, or colorant.
9569. The method of item 9406 wherein the agent is released in effective concentrations from the composition comprising the agent by diffusion over a period ranging from the time of administration to about 90 days.
9570. The method of item 9406 wherein the agent is released in effective concentrations from the composition comprising the agent by erosion of the composition over a period ranging from the time of administration to about 90 days.
9571. The method of item 9406 wherein the composition further comprises an inflammatory cytokine.
9572. The method of item 9406 wherein the composition further comprises an agent that stimulates cell proliferation. 9573. The method of item 9406 wherein the composition further comprises a polymeric carrier.
9574. The method of item 9406 wherein the composition is in the form of a gel, paste, or spray.
9575. The method of item 9406 wherein the implant is partially constructed with the agent or the composition.
9576. The method of item 9406 wherein the implant is fully constructed with the agent or the composition.
9577. The method of item 9406 wherein the implant is impregnated with the agent or the composition.
9578. The method of item 9406, wherein the agent or the composition forms a coating, and the coating directly contacts the implant.
9579. The method of item 9406, wherein the agent or the composition forms a coating, and the coating indirectly contacts the implant.
9580. The method of item 9406 wherein the agent or the composition forms a coating, and the coating partially covers the implant.
9581. The method of item 9406, wherein the agent or the composition forms a coating, and the coating completely covers the implant.
9582. The method of item 9406 wherein the agent or the composition is located within pores or holes of the implant. 9583. The method of item 9406 wherein the agent or the composition is located within a channel, lumen, or divet of the implant.
9584. The method of item 9406 wherein the implant further comprising an echogenic material.
9585. The method of item 9406 wherein the implant further comprises an echogenic material, wherein the echogenic material is in the form of a coating.
9586. The method of item 9406 wherein the implant is sterile.
9587. The method of item 9406 wherein the agent is delivered from the implant, wherein the agent is released into tissue in the vicinity of the implant after deployment of the implant.
9588. The method of item 9406 wherein the agent is delivered from the implant, wherein the agent is released into tissue in the vicinity of the implant after deployment of the implant, wherein the tissue is connective tissue.
9589. The method of item 9406 wherein the agent is delivered from the implant, wherein the agent is released into tissue in the vicinity of the implant after deployment of the implant, wherein the tissue is muscle tissue.
9590. The method of item 9406 wherein the agent is delivered from the implant, wherein the agent is released into tissue in the vicinity of the implant after deployment of the implant, wherein the tissue is nerve tissue. 9591. The method of item 9406 wherein the agent is delivered from the implant, wherein the agent is released into tissue in the vicinity of the implant after deployment of the implant, wherein the tissue is epithelium tissue.
9592. The method of item 9406 wherein the agent is delivered from the implant, wherein the agent is released in effective concentrations from the implant over a period ranging from the time of deployment of the implant to about 1 year.
9593. The method of item 9406 wherein the agent is delivered from the implant, wherein the agent is released in effective concentrations from the implant over a period ranging from about 1 month to 6 months.
9594. The method of item 9406 wherein the agent is delivered from the implant, wherein the agent is released in effective concentrations from the implant over a period ranging from about 1 - 90 days.
9595. The method of item 9406 wherein the agent is delivered from the implant, wherein the agent is released in effective concentrations from the implant at a constant rate.
9596. The method of item 9406 wherein the agent is delivered from the implant, wherein the agent is released in effective concentrations from the implant at an increasing rate.
9597. The method of item 9406 wherein the agent is delivered from the implant, wherein the agent is released in effective concentrations from the implant at a decreasing rate. 9598. The method of item 9406 wherein the agent is delivered from the implant, wherein the implant comprises about 0.01 Dg to about 10 Dg of the agent.
9599. The method of item 9406 wherein the agent is delivered from the implant, wherein the implant comprises about 10 Dg to about 10 mg of the agent.
9600. The method of item 9406 wherein the agent is delivered from the implant, wherein the implant comprises about 10 mg to about 250 mg of the agent.
9601. The method of item 9406 wherein the agent is delivered from the implant, wherein the implant comprises about 250 mg to about
1000 mg of the agent.
" '" - - - - - - ^ - -
9602. The method of item 9406 wherein the agent is delivered from the implant, wherein the implant comprises about 1000 mg to about 2500 mg of the agent.
9603. The method of item 9406 wherein the agent is delivered from the implant, wherein a surface of the implant comprises less than 0.01 Dg of the agent per mm2 of implant surface to which the agent is applied.
9604. The method of item 9406 wherein the agent is delivered from the implant, wherein a surface of the implant comprises about 0.01 Dg to about 1 Dg of the agent per mm2 of implant surface to which the agent is applied.
9605. The method of item 9406 wherein the agent is delivered from the implant, wherein a surface of the implant comprises about 1 Dg to about 10 Dg of the agent per mm2 of implant surface to which the agent is applied.
9606. The method of item 9406 wherein the agent is delivered from the implant, wherein a surface of the implant comprises about 10 Dg to about 250 Dg of the agent per mm2 of implant surface to which the agent is applied.
9607. The method of item 9406 wherein the agent is delivered from the implant, wherein a surface of the implant comprises about 250 Dg to about 1000 Dg of the agent per mm2 of implant surface to which the agent is applied.
9608. The method of item 9406 wherein the agent is delivered from the implant, wherein a surface of the implant comprises about 1000 Dg to about 2500 Dg of the agent per mm2 of implant surface to which the agent is applied.
9609. The method of item 9406, wherein the implant further comprises a coating, and the coating is a uniform coating.
9610. The method of item 9406, wherein the implant further comprises a coating, and the coating is a non-uniform coating.
9611. The method of item 9406, wherein the implant further comprises a coating, and the coating is a discontinuous coating.
9612. The method of item 9406, wherein the implant further comprises a coating, and the coating is a patterned coating. 9613. The method of item 9406, wherein the implant further comprises a coating, and the coating has a thickness of 100 Dm or less.
9614. The method of item 9406, wherein the implant further comprises a coating, and the coating has a thickness of 10 Dm or less.
9615. The method of item 9406, wherein the implant further comprises a coating, and the coating adheres to the surface of the implant upon deployment of the implant.
9616. The method of item 9406, wherein the implant further comprises a coating, and the coating is stable at room temperature for a period of at least 1 year.
9617. The method of item 9406, wherein the implant further comprises a coating, and the agent is present in the coating in an amount ranging between about 0.0001% to about 1% by weight.
9618. The method of item 9406, wherein the implant further comprises a coating, and the agent is present in the coating in an amount ranging between about 1% to about 10% by weight.
9619. The method of item 9406, wherein the implant further comprises a coating, and the agent is present in the coating in an amount ranging between about 10% to about 25% by weight.
9620. The method of item 9406, wherein the implant further comprises a coating, and the agent is present in the coating in an amount ranging between about 25% to about 70% by weight. 9621. The method of item 9406, wherein the implant further comprises a coating, and the coating comprises a polymer.
9622. The method of item 9406, wherein the implant comprises a first coating having a first composition and a second coating having a second composition.
9623. The method of item 9406, wherein the implant comprises a first coating having a first composition and a second coating having a second composition, wherein the first composition and the second composition are different.
9624. A method as in any one of items 9406-9623, wherein the device is a ventriculopleural shunt.
9625. A method as in any one of items 9406:9623, wherein the device is a jugular vein shunt.
9626. A method as in any one of items 9406-9623, wherein the device is a vena cava shunt.
9627. A method as in any one of items 9406-9623, wherein the device is a ventriculoperitoneal shunt.
9628. A method as in any one of items 9406-9623, wherein the device is a gallbladder shunt.
9629. A method as in any one of items 9406-9623, wherein the device is a peritoneum shunt. 9630. A method as in any one of items 9406-9623, wherein the device is an external ventricular drainage device.
9631. A method as in any one of items 9406-9623, wherein the device is an intracranial pressure monitoring device.
9632. A method as in any one of items 9406-9623, wherein the device is a dural patch.
9633. A method as in any one of items 9406-9623, wherein the device is an implant to prevent epidural fibrosis post-laminectomy.
9634. A method as in any one of items 9406-9623, wherein the device is a device for continuous subarachnoid infusion.
9635. A method as in any one of items 9406-9623, wherein the device is a drainage shunt useful for draining fluids in the brain.
9636. A method as in any one of items 9406-9623, wherein the device is a pressure monitoring device.
9637. A method for inhibiting scarring comprising placing a device that comprises an inferior vena cava filter (e.g., an implant) and an anti-scarring agent or a composition comprising an anti-scarring agent into an animal host, wherein the agent inhibits scarring.
9638. The method of item 9637 wherein the agent is an adensosine A2A receptor antagonist.
9639. The method of item 9637 wherein the agent is an AKT inhibitor. 9640. The method of item 9637 wherein the agent is an alpha 2 integrin antagonist, wherein the alpha 2 integrin antagonist is Pharmaprojects No. 5754 (Merck KgaA).
9641. The method of item 9637 wherein the agent is an alpha 4 integrin antagonist.
9642. The method of item 9637 wherein the agent is an alpha 7 nicotinic receptor agonist.
9643. The method of item 9637 wherein the agent is an angiogenesis inhibitor selected from the group consisting of AG-12,958 (Pfizer), ATN-161 (Attention LLC), neovastat, an angiogenesis inhibitor from Jerina AG (Germany), NM-3 (Mercian), VGA-1155 (Taisho), FCE-26644 (Pfizer), FCE-26950 (Pfizer), FPMA (Meiji Daries), FR-111142 (Fujisawa), GGTI-298, GM-1306 (Ligand), GPA-1734 (Novartis), NNC-47-0011 (Novo Nordisk), herbamycin (Nippon Kayaku), lenalidomide (Celegene), IP-10 (NIH), ABT-828 (Abbott), KIN-841 (Tokushima University, Japan), SF-1126 (Semafore Pharmaceuticals), laminin technology (NIH), CHIR-258 (Chiron), NVP-AEW541 (Novartis), NVP-AEW541 (Novartis), Vt16907 (Alchemia), OXI-8007 (Oxigene), EG-3306 (Ark Therapeutics), Maspin (Arriva), ABT- 567 (Abbott), PPI-2458 (Praecis Pharmaceuticals), CC-5079, CC-4089 (Celgene), HIF-1 alpha inhibitors (Xenova), S-247 (Pfizer), AP-23573 (Ariad), AZD-9935 (Astra Zeneca), mebendazole (Introgen Therapeutics), MetAP-2 inhibitors (GlaxoSmithKline), AG-615 (Angiogene Pharmaceuticals), Tie-2 antagonists (Hybrigenics), NC-381, CYC-381 , NC-169, NC-219, NC-383, NC-384, NC-407 (Lorus Therapeutics), ATN-224 (Attenuon), ON-01370 (Onconova), Vitronectin antagonists (Amgen), SDX-103 (Salmedix), Vitronectin antagonists (Shire), CHP (Riemser), TEK (Amgen), Anecortave acetate (Alcon), T46.2 (Matrix Therapeutics), HG-2 (Heptagen), TEM antagonists (Genzyme), Oxi-4500 (Oxigene), ATN-161 (Attenuon), WX-293 (Wilex), M-2025 (Metris Therapeutics), Alphastatin (BioActa), YH-16 (Yantai Rongchang), BIBF-1120 (Boehringer Ingelheim), BAY-57-9352 (Bayer), AS- 1404 (Cancer Research Technology), SC-77964 (Pfizer), glycomimetics (BioTie Therapies), TIE-2 Inhibitors (Ontogen), DIMI, Octamer (Octamer), ABR-215050 (Active Biotech), ABT-518 (Abbott), KDR inhibitors (Abbott), BSF-466895 (Abbott), SCH-221153 (Schering-Plough), DAC.antiangiogenic (ConjuChem), TFPI (EntreMed), AZD-2171 (Astra-Zenaca), CDC-394 (Celgene), LY290293 (EIi Lilly), IDN-5390 (Indena), Kdr Kinase Inhibitors (Merck), CT-113020, CT-116433, CT-116563, CT-31890, CT-32228) (Cell Therapeutics), A-299620 (Abbott), TWEAK Inhibitor (Amgen), VEGF modulators (Johnson and Johnson), Tum-N53, tumstatin (Genzyme), Thios- 1 , Thios-2 (Thios Pharmaceuticals), MV-6401 (Miravant Medical Technologies), Spisulosine (PharmaMar), CEP-7055 (Cephalon), AUV-201 (Auvation), LM-609 (EIi Lilly), SKF-106615 (AnorMED), Oglufanide disodium (Cytran), BW-114 (Pharminox), Calreticulin (NIH), WX-678 (Wilex), SD-7784 (Pfizer), WX-UK1 (Wilex), SH-268 (Schering AG), 2-Me-PGA (Celgene), S- 137 (Pfizer), ZD-6126 (Angiogene Pharmaceuticals), SG-292 (SignalGen), Benefin (Lane Labs), A6, A36 (Angstrom), SB-2723005 (GlaxoSmithKline), SC-7 (Cell Therapeutics), ZEN-014 (AEterna Zentaris), 2-methoxyestradiol (EntreMed), NK-130119 (Nippon Kayaku), CC-10004 (Celgene), AVE- 8062A (Ajinomoto), Tacedinaline (Pfizer), Actinonin (Tokyo Metropolitan Institute of Medical Science), Lenalidomide (Celgene), VGA-1155, BTO-956 (SRI International), ER-68203-00 (Eisai), CT-6685 (UCB), JKC-362 (Phoenix Pharmaceuticals), DMI-3798 (DMI Biosciences, Angiomate (Ipsen), ZD-6474 (AstraZeneca), CEP-5214 (Cephalon), Canstatin (Genzyme), NM-3 (Mercian), Oridigm (MediQuest Therapeutics), Exherin (Adherex), BLS-0597 (Boston Life Sciences), PTC-299 (PTC Therapeutics), NPI-2358 (Nereus Pharmaceuticals), CGP-79787 (Novartis), AEE-788 (Novartis), CKD-732 (Chong Kun Dang), CP-564959 (OSI Pharmaceuticals), CM-101 (CarboMed), CT-2584, CT3501 (Cell Therapeutics), combretastatin and analogues and derivatives thereof (Oxigene), Rebimastat (Bristol-Meyers Squibb), Dextrin 2-sulfate (ML Laboratories), Cilengitide (Merk KGaA), NSC-706704 (Pharminox), KRN- 951 (Kirin Brewery), Ukrain, NSC-631570 (Nowicky Pharma), Tecogalan sodium (Daiichi Pharmaceutical), Tz-93 (Tsumura), TBC-1635 (Encysive Pharmaceuticals), TAN-1120 (Takeda), Semaxanib (Pfizer), BDI-7800 (Biopharmacopae), SD-186, SD-983 (Bristol-Meyers Squibb), SB-223245 (GlaxoSmithKline), SC-236 (Pfizer), RWJ-590973 (Johnson and Johnson), ILX-1850 (Genzyme), SC-68488, S-836 (Pfizer), CG-55069-11 (CuraGen), Ki-23057 (Kirin Brewery), CCX-700 (Chemoentryx), Pegaptanib octasodium (Giled Sciences), ANGIOCOL (available from Biostratum Inc.), or an analogue or derivative thereof.
9644. The method of item 9637 wherein the agent is an apoptosis antagonist.
9645. The method of item 9637 wherein the agent is.an apoptosis activator.
9646. The method of item 9637 wherein the agent is a beta 1 integrin antagonist.
9647. The method of item 9637 wherein the agent is a beta tubulin inhibitor.
9648. The method of item 9637 wherein the agent is a blocker of enzyme production in Hepatitis C.
9649. The method of item 9637 wherein the agent is a Bruton's tyrosine kinase inhibitor. 9650. The method of item 9637 wherein the agent is a calcineurin inhibitor.
9651. The method of item 9637 wherein the agent is a caspase 3 inhibitor.
9652. The method of item 9637 wherein the agent is a CC chemokine receptor antagonist.
9653. The method of item 9637 wherein the agent is a cell cycle inhibitor selected from the group consisting of SNS-595 (Sunesis), synthadotin, KRX-0403, and an analogue or derivative thereof.
9654. The method of item 9637 wherein the agent is a cathepsin B inhibitor.
9655. The method of item 9637 wherein the agent is a cathepsin K inhibitor, wherein the cathepsin K inhibitor is 462795 (GlaxoSmithKline), INPL-022-D6 (Amura Therapeutics), or an analogue or derivative thereof.
9656. The method of item 9637 wherein the agent is a cathepsin L inhibitor.
9657. The method of item 9637 wherein the agent is a CD40 antagonist.
9658. The method of item 9637 wherein the agent is a chemokine receptor agonist. 9659. The method of item 9637 wherein the agent is a chymase inhibitor.
9660. The method of item 9637 wherein the agent is a collagenase antagonist.
9661. The method of item 9637 wherein the agent is a CXCR antagonist.
9662. The method of item 9637 wherein the agent is a cyclin dependent kinase inhibitor selected from the group consisting of a CDK-1 inhibitor, a CDK-2 inhibitor, a CDK- 4 inhibitor, a CDK-6 inhibitor, a CAK1 inhibitor from GPC Biotech and Bristol-Myers Squibb, RGB-286199 (GPC Biotech), an anticancer agent from Astex Technology, a CAK1 inhibitor from GPC Biotech, a CDK inhibitor from Sanofi-Aventis, a CDK1/CDK2 inhibitor from Amgen, a CDK2 inhibitor from SUGEN-2 (Pfizer), a hearing Loss., therapy agent (Sound Pharmaceuticals), PD-0332991 (Pfizer), RGB-286199 (GPC Biotech), Ro-0505124 (Hoffmann-La Roche), a Ser/Thr kinase inhibitor from Lilly (EIi Lilly), CVT-2584 (CAS No. 199986-75-9) (CV Therapeutics), and an analogue or derivative thereof.
9663. The method of item 9637 wherein the agent is a cyclooxygenase 1 inhibitor.
9664. The method of item 9637 wherein the agent is a DHFR inhibitor.
9665. The method of item 9637 wherein the agent is a dual integrin inhibitor. 9666. The method of item 9637 wherein the agent is an elastase inhibitor.
9667. The method of item 9637 wherein the agent is an elongation factor-1 alpha inhibitor.
9668. The method of item 9637 wherein the agent is an endothelial growth factor antagonist.
9669. The method of item 9637 wherein the agent is an endothelial growth factor receptor kinase inhibitor selected from the group consisting of sorafenib tosylate (Bayer), AAL-993 (Novartis), ABP-309 (Novartis), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), E- 7080 (Eisai), EG-3306 (Ark Therapeutics), EXEL-2880 (Exelixis), GW- 654652 (GlaxoSmithKline), a KDR inhibitor from LG Life Sciences, CT-6685 and CT-6729 (UCB), KRN-633 and KRN-951 (Kirin Brewery), ..081-.9.3D-(OSI Pharmaceuticals), SP-5.2 (Supratek Pharma), SlM 1657 (Pfizer), a Tie-2 antagonist (Hybrlgenics), a VEGFR-2 kinase inhibitor (Bristol-Myers Squibb), XL-647 (Exelixis), a KDR inhibitor from Abbott Laboratories, sorafenib tosylate, and an analogue or derivative thereof.
9670. The method of item 9637 wherein the agent is an endotoxin antagonist.
9671. The method of item 9637 wherein the agent is an epothilone and tubulin binder.
9672. The method of item 9637 wherein the agent is an estrogen receptor antagonist. 9673. The method of item 9637 wherein the agent is an FGF inhibitor.
9674. The method of item 9637 wherein the agent is a famexyl transferase inhibitor.
9675. The method of item 9637 wherein the agent is farnesyltransferase inhibitor selected from the group of A-197574 (Abbott), a famesyltransferase inhibitor from Servier, FPTIII (Strathclyde Institute for Drug R), LB-42908 (LG Life Sciences), Pharmaprojects No. 5063 (Genzyme), Pharmaprojects No. 5597 (Ipsen), Yissum Project No. B-1055 (Yissum), and an analogue or derivative thereof.
9676. The method of item 9637 wherein the agent is an FLT-3 kinase inhibitor.
9677. The method of item 9637 wherein the agent is an FGF receptor kinase inhibitor.
9678. The method of item 9637 wherein the agent is a fibrinogen antagonist selected from the group consisting of AUV-201 (Auvation), MG-13926 (Sanofi-Aventis), plasminogen activator (CAS No. 105913-11-9) (from Sanofi-Aventis or UCB), plasminogen activator-2 (tPA- 2) (Sanofi-Aventis), pro-urokinase (CAS No. 82657-92-9) (Sanofi-Aventis), and an analogue or derivative thereof.
9679. The method of item 9637 wherein the agent is a heat shock protein 90 antagonist selected from the group consisting of SRN-005 (Sirenade), geldanamycin, NSC-33050 (17-allylaminogeldanamycin; 17- AAG), 17-dimethylaminoethylamino-17-demethoxy-geldanamycin (17- DMAG)1 rifabutin (rifamycin XIV1 1',4-didehydro-1-deoxy-1 ,4-dihydro-5'-(2- methylpropyl)-1-oxo-), and an analogue or derivative thereof.
9680. The method of item 9637 wherein the agent is a histone deacetylase inhibitor.
9681. The method of item 9637 wherein the agent is an HMGCoA reductase inhibitor selected from the group consisting of an atherosclerosis therapeutic from Lipid Sciences, ATI-16000 (ARYx Therapeutics), KS-01-019 (Kos Pharmaceuticals), Pharmaprojects No. 2197 (Sanofi-Aventi), RP 61969 (Sanofi-Aventis), and an analogue or derivative thereof.
9682. The method of item 9637 wherein the agent is an ICAM inhibitor.
9683. The method of item 9637 wherein the agent is an IL, ICE and IRAK antagonist, wherein the antagonist is a CJ-14877, CP-424174 (Pfizer), NF-61 (Negma-Lerads), and an analogue or derivative thereof.
9684. The method of item 9637 wherein the agent is an IL-2 inhibitor.
9685. The method of item 9637 wherein the agent is an immunosuppressant selected from the group consisting of teriflunomide (Sanofi Aventis), chlorsulfaquinoxalone (NSC-339004), chlorsulfaquinoxalone sulfate, CS-712 (Sankyo), ismomultin alfa (CAS No. 457913-93-8) (Akzo Nobel), antiallergics from GenPat77, antiinflammatories or AT-005 (Androclus Therapeutics), autoimmune disease therapeutics from EpiVax, BN-007 (Bone), budesonide (CAS No. 51333-22- 3) (MAP Pharmaceuticals), CO-14 (Genzyme), edratide (CAS No. 433922- 67-9) (Teva), EP-314 (Enanta), eprovafen (CAS No. 101335-99-3) (Sanofi- Aventis), HWA-131 (CAS No. 118788-41-3) (Sanofi-Aventis), immunomodulators from MerLion Pharmaceuticals, immunosuppressives from Alchemia, IPL-12 (Inflazyme), MDL-9563 (CAS No. 27086-86-8) (Sanofi-Aventis), Pharmaprojects No. 2330 (Sanofi-Aventis), Pharmaprojects No. 6426 (Abgenix), PXS-25 (Pharmaxis), rosmarinic acid (CAS No. 20283-92-5) (Sanofi-Aventis), RP 42927 or RP 54745 (CAS No. 135330-08-4) (Sanofi-Aventis), SGN-35 (Seattle Genetics), ST-1959 (Sigma-Tau), type I diabetes therapy from SYNX Pharma, UNIL-88 (Debiopharm), VP-025 (Vasogen), VR-694 (Vectura), PRTX-001 (Protalex), and an analogue or derivative thereof.
9686. The method of item 9637 wherein the agent is an IMPDH (inosine monophosphate).
9687. The method of item 9637 wherein the agent is an integrin antagonist.
9688. The method of item 9637 wherein the agent is an interleukin antagonist.
9689. The method of item 9637 wherein the agent is an inhibitor of type III receptor tyrosine kinase.
9690. The method of item 9637 wherein the agent is an irreversible inhibitor of enzyme methionine aminopeptidase type 2.
9691. The method of item 9637 wherein the agent is an isozyme selective delta protein kinase C inhibitor. 9692. The method of item 9637 wherein the agent a JAK3 enzyme inhibitor.
9693. The method of item 9637 wherein the agent is a JNK inhibitor.
9694. The method of item 9637 wherein the agent is a kinase inhibitor.
9695. The method of item 9637 wherein the agent is kinesin antagonist.
9696. The method of item 9637 wherein the agent is a kinesin antagonist.
9697. The method of item 9637 wherein the agent is a leukotriene inhibitor and antagonist selected from the group consisting of ambicromil (CAS No. 58805-38-2) (Sanofi-Aventis), amelubant (CAS No. 346735-24-8) (Boehringer Ingelheim), DW-1141 (Dong Wha), ebselen (Daiichi Pharmaceutical), ibudilast (Kyorin), leucotriene inhibitors from Sanofi-Aventis, lymphotoxin-beta receptor (LT-D) from Biogen Idee, Pharmaprojects No. 1535 and 2728 (CAS No. 119340-33-9) (Sanofi- Aventis), R-112 (Rigel), Rev-5367 (CAS No. 92532-05-3) (Sanofi-Aventis), RG-14893 (CAS No. 141835-49-6) (Sanofi-Aventis), RG-5901-A (CAS No. 101910-24-1), 92532-23-5, RP 66153 (CAS No. 142422-79-5), RP 66364 (CAS No.186916-92-5), RP 69698 (CAS No. 141748-00-7) (Sanofi-Aventis), SC-411930 (Pfizer), SC-41930 (CAS No. 120072-59-5) (Pfizer), SC-50605 (CAS No. 138828-39-4) (Pfizer), SC-51146 (CAS No. 141059-52-1), SC- 53228 (CAS No. 153633-01-3) (Pfizer), spaglumic acid (ZY-15106) (CAS No.3106-85-2), 80619-64-3 (Novartis), tipredane (CAS No. 85197-77-9) (Bristol-Myers Squibb), U-75302 (CAS No. 119477-85-9) (Pfizer), and analogue or derivative thereof.
9698. The method of item 9637 wherein the agent is an MAP kinase inhibitor.
9699. The method of item 9637 wherein the agent is a matrix metalloproteinase inhibitor.
9700. The method of item 9637 wherein the agent is an MCP- CCR2 inhibitor.
9701. The method of item 9637 wherein the agent is an mTOR inhibitor.
9702. The method of item 9637 wherein the agent is an mTOR kinase inhibitor.
9703. The method of item 9637 wherein the agent is a microtubule inhibitor selected from the group consisting of antibody- maytansinoid conjugates from Biogen Idee, colchicines (MantiCore Pharmaceuticals), anticancer immunoconjugates from Johnson & Johnson, DIME from Octamer, gni-1f (GNI), huC242-DM4, huMy9-6-DM1 (ImmunoGen), IDN-5404 (Indena), IMO-098, IMOderm (Imotep), mebendazole (Introgen Therapeutics), microtubule poisons from Cambridge Enterprise, paclitaxel such as LOTAX from Aphios (CAS No. 33069-62-4), Genexol-PM from Samyang, Pharmaprojects No. 6383 (Tapestry Pharmaceuticals), RPR-112378 (Sanofi-Aventis), SGN-75 (Seattle Genetics), SPL-7435 (Starpharma), SSR-250411 (Sanofi-Aventis), trastuzumab-DM1 (Genentech), vinorelbine, and an analogue or derivative thereof. 9704. The method of item 9637 wherein the agent is an MIF inhibitor.
9705. The method of item 9637 wherein the agent is an MMP inhibitor.
9706. The method of item 9637 wherein the agent is a neurokinin (NK) antagonist selected from the group consisting of anthrotainin (CAS No. 148084-40-6) (Sanofi-Aventis), an IBS thereapeutic from ArQuIe, MDL-105212A (CAS No. 167261-60-1) (Ssanofi-Aventis), Pharmaprojects No. 2744, 3258 (CAS No. 139167-47-8) 4006, 4201 , or 5986 (Sanofi-Aventis), RP 67580 (CAS No. 135911-02-3), SR-144190 (CAS No. 201152-86-5), SSR-240600, SSR-241586 (Sanofi-Aventis), TKA-457 (Novartis), vestipitant mesylate (CAS No. 334476-64-1) (GlaxoSmithKline), Win-64821 (Sanofi-Aventis), PRX-96026 (Predix Pharmaceuticals), and an analogue or derivative thereof.
9707. The method of item 9637 wherein the agent is an NF kappa B inhibitor selected from the group consisting of AVE-0545 or AVE- 0547 (Sanofi-Aventis), bortezomib (CAS No. 179324-69-7) (Millennium Pharmaceuticals), dexanabinol (CAS No. 112924-45-5) (Pharmos), dexlipotam (Viatris), Pharmaprojects No. 6283 (INDRA) (OXiGENE), IPL- 576092 (CAS No. 137571-30-3) (Inflazyme), NFKB decoy (Corgentech), NFKB decoy oligo (AnGes MG), NFKB's from Ariad, osteoporosis treatments or S5 (F005) from Fulcrum Pharmaceuticals, P61 (Phytopharm), R-flurbiprofen (CAS No. 5104-49-4) (Encore Pharmaceuticals), and an analogue or derivative thereof.
9708. The method of item 9637 wherein the agent is a nitric oxide agonist. 9709. The method of item 9637 wherein the agent is an ornithine decarboxylase inhibitor.
9710. The method of item 9637 wherein the agent is a p38 MAP kinase inhibitor selected from the group consisting of AZD-6703 (AstraZeneca), JX-401 (Jexys Pharmaceuticals), BMS-2 (Bristol-Myers Squibb), a p38 MAP kinase inhibitor from Novartis, a p38-alpha MAP kinase inhibitor from Amphora, Pharmaprojects No. 5704 (Pharmacopeia), RPR- 200765A (Sanofi-Aventis), SD-282 (Johnson & Johnson), TAK-715 (Takeda), and an analogue or derivative thereof.
9711. The method of item 9637 wherein the agent is a palmitoyl-protein thioesterase inhibitor.
9712. The method of item 9637 wherein the agent is a PDGF receptor kinase inhibitor selected from the group consisting of AAL-993, AMN-107, or ABP-309 (Novartis), AMG-706 (Amgen), BAY-57-9352 (Bayer), CDP-860 (UCB), E-7080 (Eisai), imatinib (CAS No. 152459-95-5) (Novartis), OSI-930 (OSI Pharmaceuticals), RPR-127963E (Sanofi-Aventis), RWJ-540973 (Johnson & Johnson), sorafenib tosylate (Bayer), SU-11657 (Pfizer), tandutinib (CAS No. 387867-13-2) (Millennium Pharmaceuticals), vatalanib (Novartis), ZK-CDK (Schering AG), and an analogue or derivative thereof.
9713. The method of item 9637 wherein the agent is (-)- halofenate (Metabolex), AMG-131 (Amgen), antidiabetics from Japan Tobacco, AZD-4619, AZD-8450, AZD-8677 (AstraZeneca), DRF-10945, balaglitazone (Dr Reddy's), CS-00088, CS-00098 (Chipscreen Biosciences), E-3030 (Eisai), etalocib (CAS No. 161172-51-6) (EIi Lilly), GSK-641597 (Ligand), GSK-677954 (GlaxoSmithKline), GW-409544 (Ligand), GW- 590735 (GlaxoSmithKline), K-111 (Hoffmann-La Roche), LY-518674 (EIi Lilly), LY-674 (Ligand), LY-929 (Ligand), MC-3001 , MC-3002 (MaxoCore Pharmaceuticals), metformin HCl + pioglitazone (CAS No. 1115-70-4 and 112529-15-4), ACTOPLUS MET from Andrx), muraglitazar (CAS No. 331741-94-7) (Bristol-Myers Squibb), naveglitazar (Ligand), oleoylethanolamide (Kadmus Pharmaceuticals), ONO-5129, pioglitazone hydrochloride (CAS No. 111025-46-8 and 112529-15-4) (Takeda), PLX-204 (Plexxikon), PPAR agonists from Genfit, PPAR delta agonists from EIi Lilly, PPAR-alpha agonists from CrystalGenomics, PPAR-gamma modulators and PPAR-β modulators from CareX, rosiglitazone maleate (CAS No. 122324- 73-4 or 155141-29-0) (GlaxoSmithKline), rosiglitazone maleate/glimepir (CAS No. 155141-29-0 and 93479-97-1), AVANDARYL, rosiglitazone maleate/metformin extend (CAS No. 155141-29-0 and 657-24-9), AVANDAMET, rosiglitazone maleate+metformin, AVANDAMET (GlaxoSmithKline), tesaglitazar (AstraZeneca), and an analogue or derivative thereof.
9714. The method of item 9637 wherein the agent is a phosphatase inhibitor.
9715. The method of item 9637 wherein the agent is a phosphodiesterase (PDE) inhibitor selected from the group consisting of avanafil (Tanabe Seiyaku), dasantafil (CAS No. 569351-91-3) (Schering- Plough), A-906119 (CAS No. 134072-58-5), DL-850 (Sanofi-Aventis), GRC- 3015, GRC-3566, GRC-3886 (Glenmark), HWA-153 (CAS No. 56395-66-5) (Sanofi-Aventis), hydroxypumafentrine (Altana), IBFB-130011 , IBFB-14-016, IBFB-140301 , IBFB-150007, IBFB-211913 (IBFB Pharma), L-826141 (Merck & Co), medorinone (CAS No. 88296-61-1) (Sanofi-Aventis), MEM- 1917 (Memory Pharmaceuticals), ND-1251 (Neuro3d), PDE inhibitors from ICOS, PDE IV inhibitors from Memory Pharmaceuticals and CrystalGenomics, Pharmaprojects No. 2742 and 6141 (Sanofi-Aventis), QAD-171 (Novartis), RHC-2963 (CAS No. 76993-12-9 and 76993-14-1), RPR-117658, RPR-122818 derivatives, SR-24870 , and RPR-132294 (Sanofi-Aventis), SK-350 (ln2Gen), stroke therapy agents from deCODE Genetics, TAS-203 (Taiho), tofimilast (CAS No. 185954-27-2) (Pfizer), UK- 371800 (Pfizer), WIN-65579 (CAS No. 158020-82-7) (Sanofi-Aventis), IBFB- 130020 (IBFB Pharma), OPC-6535 (CAS No. 145739-56-6) (Otsuka), a phosphodiesterase III inhibitor, enoximone, a phosphodiesterase IV inhibitor, fosfosal, Atopik (Barrier Therapeutics), triflusal, a phosphodiesterase V inhibitor, and an analogue or derivative thereof.
9716. The method of item 9637 wherein the agent is a PKC inhibitor.
9717. The method of item 9637 wherein the agent is a platelet activating factor antagonist.
9718. The method of item 9637 wherein the agent is a platelet-derived growth factor receptor kinase inhibitor.
9719. The method of item 9637 wherein the agent is a prolyl hydroxylase inhibitor.
9720. The method of item 9637 wherein the agent is a polymorphonuclear neutrophil inhibitor.
9721. The method of item 9637 wherein the agent is a protein kinase B inhibitor.
9722. The method of item 9637 wherein the agent is a protein kinase C stimulant. 9723. The method of item 9637 wherein the agent is a purine nucleoside analogue.
9724. The method of item 9637 wherein the agent is a purinoreceptor P2X antagonist.
9725. The method of item 9637 wherein the agent is a Raf kinase inhibitor.
9726. The method of item 9637 wherein the agent is a reversible inhibitor of ErbB1 and ErbB2.
9727. The method of item 9637 wherein the agent is a ribonucleoside triphosphate reductase inhibitor.
9728. The method of item 9637 wherein the agent is an SDF- 1 antagonist.
9729. The method of item 9637 wherein the agent is a sheddase inhibitor.
9730. The method of item 9637 wherein the agent is an SRC inhibitor.
9731. The method of item 9637 wherein the agent is a stromelysin inhibitor.
9732. The method of item 9637 wherein the agent is an Syk kinase inhibitor. 9733. The method of item 9637 wherein the agent is a telomerase inhibitor.
9734. The method of item 9637 wherein the agent is a TGF beta inhibitor selected from the group consisting of pirfenidone (CAS No. 53179-13-8) (MARNAC), tranilast (CAS No. 53902-12-8) (Kissei), IN-1130 (ln2Gen), mannose-6-phosphate (BTG), TGF-β antagonists from Inflazyme (Pharmaprojects No. 6075), TGF-β antagonists from Sydney, non-industrial source), TGF-β I receptor kinase inhibitors from EIi Lilly, TGF-β receptor inhibitors from Johnson & Johnson, and an analogue or derivative thereof.
9735. The method of item 9637 wherein the agent is a TNFα antagonist or TACE inhibitor selected from the group consisting of adalimumab (CAS No. 331731-18-1) (Cambridge Antibody Technology), AGIX-4207 (AtheroGenics), AGT-1 (Advanced Biotherapy), an antiinflammatory from Borean Pharma, Cellzome, or Paradigm Therapeutics, anti-inflammatory vaccine (TNF-alpha kinoid) from Neovacs, humanized anti-TNF antibody or an anti-TNF MAb (CB0006) Celltech (UCB), apratastat (CAS No. 287445-51-0) (Wyeth), BMS-561392 (Bristol-Myers Squibb), BN- 006 (Bone), certolizumab pegol (CAS No. 428863-50-7 or CH-138 (UCB), cilomilast (CAS No. 153259-65-5) (GlaxoSmithKline), CR-1 (Nuada Pharmaceuticals), CRx-119 (CombinatoRx), D-5410 (UCB), dacopafant (CAS No. 125372-33-0) (Sanofi-Aventis), dersalazine (CAS No. 188913-57- 7/188913-58-8) (Uriach), etanercept (CAS No. 185243-69-0) (Amgen), ethyl pyruvate (Critical (Critical Therapeutics), golimumab (CAS No. 476181-74-5) (Johnson & Johnson), hormono-immunotherapy from Ipsen, CDP571 (e.g., Humicade from UCB), IC-485 (ICOS), infliximab (CAS No. 170277-31-3) (Johnson & Johnson), IP-751 (Manhattan Pharmaceuticals), ISIS-104838 (CAS No. 250755-32-9) (ISIS Pharmaceuticals), lenalϊdomide (CAS No. 191732-72-6) (Celgene), Ientinan (CAS No. 37339-90-5) (Ajinomoto), MDL- 201112 (CAS No. 142130-73-2) (Sanofi-Aventis), medroxyprogesterone (CAS No. 520-85-4) (InKine Pharmaceutical), N-acetylcysteine (CAS No. 616-91-1) (Zambon), NBE-P2 (DIREVO Biotech), nerelimomab (CAS No. 162774-06-3) (Chiron), OM-294DP (OM PHARMA), onercept (CAS No. 199685-57-9) (Yeda), PASSTNF-alpha (Verigen), pentoxifylline or oxypentifylline (Sanofi-Aventis), Pharmaprojects No. 4091 , 4241 , 4295, or 4488 (Sanofi-Aventis), Pharmaprojects No. 5480 (Amgen), Pharmaprojects No. 6749 (Cengent), pirfenidone (CAS No. 53179-13-8) (MARNAC), RPR- 132294 (Sanofi-Aventis), S5 (F002) (Fulcrum Pharmaceuticals), simvastatin (CAS No. 79902-63-9) (Merck & Co), STA-6292 (Synta Pharmaceuticals), tacrolimus (CAS No. 104987-11-3) (Fujisawa LifeCycle Pharma), talactoferrin alfa (CAS No. 308240-58-6) (Agennix), thalidomide (CAS No. 50-35-1) (Celgene), TNF antagonists form ProStrakan, and Synergen, TNF inhibitors (Amgen), TNF-alpha antagonists from Dynavax Technologies and Jerina AG (Germany), TNF-alpha inhibitors from IBFB Pharma and Xencor (Xencor), torbafylline (CAS No. 105102-21-4) (Sanofi-Aventis), UR-1505 (Uriach), VT-346 (Viron Therapeutics), YSIL6 (Y's Therapeutics), YSTH2 (Y's Therapeutics), NPI-1302a-3 (Nereus Pharmaceuticals, a TNF antagonist from Jerina AG (Germany), dersalazine, and an analogue or derivative thereof.
9736. The method of item 9637 wherein the agent is a Toll receptor inhibitor.
9737. The method of item 9637 wherein the agent is a tubulin antagonist.
9738. The method of item 9637 wherein the agent is a tyrosine kinase inhibitor selected from the group consisting of SU-011248, SUTENT from Pfizer Inc. (New York, NY), BMS-354825, PN-355 (Paracelsian Pharmaceuticals), AGN-199659 (Allergan), AAL-993 or ABP- 309 (Novartis), adaphostin (NIH), AEE-788 (Novartis), AG-013736 (OSI Pharmaceuticals), AG-13736 (Pfizer), ALT-110 (Alteris Therapeutics), AMG- 706 (Amgen), anticancer MAbs from Xencor, anti-EGFrvlll MAbs from Abgenix, anti-HER2 MAb from Abiogen, AZD-2171 or AZD-9935 (AstraZeneca), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), CEP-5214 (Cephalon), CEP-7055 (Cephalon), cetuximab (ImCIone Systems), CHIR-200131 and CHIR-258 (Chiron), CP-547632 (OSI Pharmaceuticals), CP-724714 (Pfizer), CT-301 (Creabilis Therapeutics), D- 69491 (Baxter International), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EGFR/ErbB2 inhibitors from Array BioPharma, erlotinib (CAS No. 183319- 69-9) (OSI Pharmaceuticals), EXEL-2880 (Exelixis), FK-778 (Sanofi- Aventis) , gefitinib (CAS No. 184475-35-2) (AstraZeneca), GW-2286 or GW- 654652 (GlaxoSmithKline), her2/neu antigen from AlphaVax, HER-2/neu inhibitor from Generex, Herzyme (Medipad) (Sirna Therapeutics), HKI-272 (Wyeth), HuMax-EGFr (Genmab), idronoxil (CAS No. 81267-65-4) (Novogen), IGF-1 inhibitors from Ontogen, IMC-11F8 (ImCIone Systems), kahalalide F (CAS No. 149204-42-2) (PharmaMar), KDR inhibitor from LG Life Sciences, KDR inhibitors from Abbott Laboratories, KDR kinase inhibitors (UCB), Kdr kinase inhibitors from Merck & Co, KRN-633 and KRN- 951 (Kirin Brewery), KSB-102 (Xenova), lapatinib ditosylate (CAS No. 388082-78-8) (GlaxoSmithKline), matuzumab (Merck KGaA)1 MDX-214 (Medarex), ME-103 (Pharmexa), MED-A300 (Gerolymatos), MNAC-13 (Lay Line Genomics), nimotuzumab (Center of Molecular Immunology), NSC- 330507 or NSC-707545 (NIH), NV-50 (Novogen), OSI-930 (OSI Pharmaceuticals), panitumumab (Abgenix), pelitinib (CAS No. 287933-82-7) (Wyeth), pertuzumab (CAS No. 380610-27-5) (Genentech), Pharmaprojects No. 3985 (Sanofi-Aventis), prostate cancer therapeutics from Sequenom (SQPC35, SQPC36, SQPC90), removab and remoxab (Trion Pharma), RG- 13022 (CAS No. 136831-48-6), RG-13291 (CAS No. 138989-50-1), or RG- 14620 (CAS No. 136831-49-7) (Sanofi-Aventis), RM-6427 (Romark), RNAi breast cancer therapy from Benitec, RP 53801 (CAS No. 125882-88-4) (Sanofi-Aventis), sorafenib tosylate (Bayer), SU-11657 (Pfizer), Tie-2 antagonists from Semaia (Hybrigenics), Tie-2 inhibitors from Ontogen, trastuzumab (CAS No. 180288-69-1) (Genentech), tyrosine kinase inhibitors from Sanofi-Aventis, U3-1287, U3-1565, U3-1784, U3-1800 (U3 Pharma), vatalanib (Novartis), VEGFR-2 kinase inhibitor from Bristol-Myers Squibb, XL-647 (Exelixis), ZD-6474 (AstraZeneca), ZK-CDK (Schering AG), an EGFR tyrosine kinase inhibitor, EKB-569 (Wyeth), and an analogue or derivative thereof.
9739. The method of item 9637 wherein the agent is a VEGF inhibitor.
9740. The method of item 9637 wherein the agent is a vitamin D receptor agonist.
9741. The method of item 9637 wherein the agent is ZD-6474 (an angiogenesis inhibitor).
9742. The method of item 9637 wherein the agent is AP- 23573 (an mTOR inhibitor).
9743. The method of item 9637 wherein the agent is synthadotin (a tubulin antagonist).
9744. The method of item 9637 wherein the agent is S-0885 (a collagenase inhibitor).
9745. The method of item 9637 wherein the agent is aplidine (an elongation factor-1 alpha inhibitor).
9746. The method of item 9637 wherein the agent is ixabepilone (an epithilone). 9747. The method of item 9637 wherein the agent is IDN- 5390 (an angiogenesis inhibitor).
9748. The method of item 9637 wherein the agent is SB- 2723005 (an angiogenesis inhibitor).
9749. The method of item 9637 wherein the agent is ABT-518 (an angiogenesis inhibitor).
9750. The method of item 9637 wherein the agent is combretastatin (an angiogenesis inhibitor).
9751. The method of item 9637 wherein the agent is anecortave acetate (an angiogenesis inhibitor).
9752. The method of item 9637 wherein the agent is SB- 715992 (a kinesin antagonist).
9753. The method of item 9637 wherein the agent is temsirolimus (an mTOR inhibitor).
9754. The method of item 9637 wherein the agent is adalimumab (a TNFα antagonist).
9755. The method of item 9637, wherein the composition comprises a polymer.
9756. The method of item 9637, wherein the composition comprises a polymer, and the polymer is, or comprises, a copolymer. 9757. The method of item 9637, wherein the composition comprises a polymer, and the polymer is, or comprises, a block copolymer.
9758. The method of item 9637, wherein the composition comprises a polymer, and the polymer is, or comprises, a random copolymer.
9759. The method of item 9637, wherein the composition comprises a polymer, and the polymer is, or comprises, a biodegradable polymer.
9760. The method of item 9637, wherein the composition comprises a polymer, and the polymer is, or comprises, a nonbiodegradable polymer.
9761. The method of item 9637, wherein the composition comprises a polymer, and the polymer is, or comprises, a hydrophilic polymer.
9762. The method of item 9637, wherein the composition comprises a polymer, and the polymer is, or comprises, a hydrophobic polymer.
9763. The method of item 9637, wherein the composition comprises a polymer, and the polymer is, or comprises, a polymer having hydrophilic domains.
9764. The method of item 9637, wherein the composition comprises a polymer, and the polymer is, or comprises, a polymer having hydrophobic domains. 9765. The method of item 9637, wherein the composition comprises a polymer, and the polymer is, or comprises, a non-conductive polymer.
9766. The method of item 9637, wherein the composition comprises a polymer, and the polymer is, or comprises, an elastomer.
9767. The method of item 9637, wherein the composition comprises a polymer, and the polymer is, or comprises, a hydrogel.
9768. The method of item 9637, wherein the composition comprises a polymer, and the polymer is, or comprises, a silicone polymer.
9769. The method of item 9637, wherein the composition comprises a polymer, and the polymer is, or comprises, a hydrocarbon polymer.
9770. The method of item 9637, wherein the composition comprises a polymer, and the polymer is, or comprises, a styrene-derived polymer.
9771. The method of item 9637, wherein the composition comprises a polymer, and the polymer is, or comprises, a butadiene-derived polymer.
9772. The method of item 9637, wherein the composition comprises a polymer, and the polymer is, or comprises, a macromer.
9773. The method of item 9637, wherein the composition comprises a polymer, and the polymer is, or comprises, a poly(ethylene glycol) polymer. 9774. The method of item 9637, wherein the composition comprises a polymer, and the polymer is, or comprises, an amorphous polymer.
9775. The method of item 9637, wherein the composition further comprises a second pharmaceutically active agent.
9776. The method of item 9637, wherein the composition further comprises an anti-inflammatory agent.
9777. The method of item 9637, wherein the composition further comprises an agent that inhibits infection.
9778. The method of item 9637, wherein the composition further comprises an anthracycline.
9779. The method of item 9637, wherein the composition further comprises doxorubicin.
9780. The method of item 9637 wherein the composition further comprises mitoxantrone.
9781. The method of item 9637 wherein the composition further comprises a fiuoropyrimidine.
9782. The method of item 9637, wherein the composition further comprises 5-fluorouracil (5-FU).
9783. The method of item 9637, wherein the composition further comprises a folic acid antagonist. 9784. The method of item 9637, wherein the composition further comprises methotrexate.
9785. The method of item 9637, wherein the composition further comprises a podophylotoxin.
9786. The method of item 9637, wherein the composition further comprises etoposide.
9787. The method of item 9637, wherein the composition further comprises camptothecin.
9788. The method of item 9637, wherein the composition further comprises a hydroxyurea.
9789. The method of item 9637, wherein the composition further comprises a platinum complex.
9790. The method of item 9637, wherein the composition further comprises cisplatin.
9791. The method of item 9637 wherein the composition further comprises an anti-thrombotic agent.
9792. The method of item 9637, wherein the composition further comprises a visualization agent.
9793. The method of item 9637, wherein the composition further comprises a visualization agent, and the visualization agent is a radiopaque material, wherein the radiopaque material comprises a metal, a halogenated compound, or a barium containing compound. 9794. The method of item 9637, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, barium, tantalum, or technetium.
9795. The method of item 9637, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, an MRI responsive material.
9796. The method of item 9637, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, a gadolinium chelate.
9797. The method of item 9637, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, iron, magnesium, manganese, copper, or chromium.
9798. The method of item 9637, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, iron oxide compound.
9799. The method of item 9637, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, a dye, pigment, or colorant.
9800. The method of item 9637 wherein the agent is released in effective concentrations from the composition comprising the agent by diffusion over a period ranging from the time of administration to about 90 days.
9801. The method of item 9637 wherein the agent is released in effective concentrations from the composition comprising the agent by erosion of the composition over a period ranging from the time of administration to about 90 days.
9802. The method of item 9637 wherein the composition further comprises an inflammatory cytokine.
9803. The method of item 9637 wherein the composition further comprises an agent that stimulates cell proliferation.
9804. The method of item 9637 wherein the composition further comprises a polymeric carrier.
9805. The method of item 9637 wherein the composition is in the form of a gel, paste, or spray.
9806. The method of item 9637 wherein the jmplant is partially constructed with the agent or the composition.
9807. The method of item 9637 wherein the implant is fully constructed with the agent or the composition.
9808. The method of item 9637 wherein the implant is impregnated with the agent or the composition.
9809. The method of item 9637, wherein the agent or the composition forms a coating, and the coating directly contacts the implant.
9810. The method of item 9637, wherein the agent or the composition forms a coating, and the coating indirectly contacts the implant. 9811. The method of item 9637 wherein the agent or the composition forms a coating, and the coating partially covers the implant.
9812. The method of item 9637, wherein the agent or the composition forms a coating, and the coating completely covers the implant.
9813. The method of item 9637 wherein the agent or the composition is located within pores or holes of the implant.
9814. The method of item 9637 wherein the agent or the composition is located within a channel, lumen, or divet of the implant.
9815. The method of item 9637 wherein the implant further comprising an echogenic material.
9816. The method of item 9637 wherein the implant further comprises an echogenic material, wherein the echogenic material is in the form of a coating.
9817. The method of item 9637 wherein the implant is sterile.
9818. The method of item 9637 wherein the agent is delivered from the implant, wherein the agent is released into tissue in the vicinity of the implant after deployment of the implant.
9819. The method of item 9637 wherein the agent is delivered from the implant, wherein the agent is released into tissue in the vicinity of the implant after deployment of the implant, wherein the tissue is connective tissue. 9820. The method of item 9637 wherein the agent is delivered from the implant, wherein the agent is released into tissue in the vicinity of the implant after deployment of the implant, wherein the tissue is muscle tissue.
9821. The method of item 9637 wherein the agent is delivered from the implant, wherein the agent is released into tissue in the vicinity of the implant after deployment of the implant, wherein the tissue is nerve tissue.
9822. The method of item 9637 wherein the agent is delivered from the implant, wherein the agent is released into tissue in the vicinity of the implant after deployment of the implant, wherein the tissue is epithelium tissue.
9823. The method of item 9637 wherein the agent is delivered from the implant, wherein the agent is released in effective concentrations from the implant over a period ranging from the time of deployment of the implant to about 1 year.
9824. The method of item 9637 wherein the agent is delivered from the implant, wherein the agent is released in effective concentrations from the implant over a period ranging from about 1 month to 6 months.
9825. The method of item 9637 wherein the agent is delivered from the implant, wherein the agent is released in effective concentrations from the implant over a period ranging from about 1 - 90 days.
9826. The method of item 9637 wherein the agent is delivered from the implant, wherein the agent is released in effective concentrations from the implant at a constant rate. 9827. The method of item 9637 wherein the agent is delivered from the implant, wherein the agent is released in effective concentrations from the implant at an increasing rate.
9828. The method of item 9637 wherein the agent is delivered from the implant, wherein the agent is released in effective concentrations from the implant at a decreasing rate.
9829. The method of item 9637 wherein the agent is delivered from the implant, wherein the implant comprises about 0.01 Dg to about 10 Dg of the agent.
9830. The method of item 9637 wherein the agent is delivered from the implant, wherein the implant comprises about 10 Dg to about 10 mg of the agent.
9831. The method of item 9637 wherein the agent is delivered from the implant, wherein the implant comprises about 10 mg to about 250 mg of the agent.
9832. The method of item 9637 wherein the agent is delivered from the implant, wherein the implant comprises about 250 mg to about
1000 mg of the agent.
9833. The method of item 9637 wherein the agent is delivered from the implant, wherein the implant comprises about 1000 mg to about 2500 mg of the agent.
9834. The method of item 9637 wherein the agent is delivered from the implant, wherein a surface of the implant comprises less than 0.01 Dg of the agent per mm2 of implant surface to which the agent is applied. 9835. The method of item 9637 wherein the agent is delivered from the implant, wherein a surface of the implant comprises about 0.01 Dg to about 1 Dg of the agent per mm2 of implant surface to which the agent is applied.
9836. The method of item 9637 wherein the agent is delivered from the implant, wherein a surface of the implant comprises about 1 Dg to about 10 Dg of the agent per mm2 of implant surface to which the agent is applied.
9837. The method of item 9637 wherein the agent is delivered from the implant, wherein a surface of the implant comprises about 10 Dg to about 250 Dg of the agent per mm2 of implant surface to which the agent is applied.
9838. The method of item 9637 wherein the agent is delivered from the implant, wherein a surface of the implant comprises about 250 Dg to about 1000 Dg of the agent per mm2 of implant surface to which the agent is applied.
9839. The method of item 9637 wherein the agent is delivered from the implant, wherein a surface of the implant comprises about 1000 Dg to about 2500 Dg of the agent per mm2 of implant surface to which the agent is applied.
9840. The method of item 9637, wherein the implant further comprises a coating, and the coating is a uniform coating.
9841. The method of item 9637, wherein the implant further comprises a coating, and the coating is a non-uniform coating. 9842. The method of item 9637, wherein the implant further comprises a coating, and the coating is a discontinuous coating.
9843. The method of item 9637, wherein the implant further comprises a coating, and the coating is a patterned coating.
9844. The method of item 9637, wherein the implant further comprises a coating, and the coating has a thickness of 100 Dm or less.
9845. The method of item 9637, wherein the implant further comprises a coating, and the coating has a thickness of 10 Dm or less.
9846. The method of item 9637, wherein the implant further comprises a coating, and the coating adheres to the surface of the implant upon deployment of the implant.
9847. The method of item 9637, wherein the implant further comprises a coating, and the coating is stable at room temperature for a period of at least 1 year.
9848. The method of item 9637, wherein the implant further comprises a coating, and the agent is present in the coating in an amount ranging between about 0.0001% to about 1% by weight.
9849. The method of item 9637, wherein the implant further comprises a coating, and the agent is present in the coating in an amount ranging between about 1% to about 10% by weight.
9850. The method of item 9637, wherein the implant further comprises a coating, and the agent is present in the coating in an amount ranging between about 10% to about 25% by weight. 9851. The method of item 9637, wherein the implant further comprises a coating, and the agent is present in the coating in an amount ranging between about 25% to about 70% by weight.
9852. The method of item 9637, wherein the implant further comprises a coating, and the coating comprises a polymer.
9853. The method of item 9637, wherein the implant comprises a first coating having a first composition and a second coating having a second composition.
9854. The method of item 9637, wherein the implant comprises a first coating having a first composition and a second coating having a second composition, wherein the first composition and the second composition are different.
9855. A method as in any one of items 9637-9854, wherein the device is a vascular filter.
9856. A method as in any one of items 9637-9854, wherein the device is a blood filter.
9857. A method as in any one of items 9637-9854, wherein the device is a caval filter.
9858. A method as in any one of items 9637-9854, wherein the device is a vena cava filter.
9859. A method as in any one of items 9637-9854, wherein the device is a thrombus filter. 9860. A method as in any one of items 9637-9854, wherein the device is a antimigration filter.
9861. A method as in any one of items 9637-9854, wherein the device is a percutaneous filter system.
9862. A method as in any one of items 9637-9854, wherein the device is an intravascular trap.
9863. A method as in any one of items 9637-9854, wherein the device is an intravascular filter.
9864. A method as in any one of items 9637-9854, wherein the device is a clot filter.
9865. A method as in any one of items 9637-9854, wherein the device is a vein filter.
9866. A method as in any one of items 9637-9854, wherein the device is a body vessel filter.
9867. A method for inhibiting scarring comprising placing a device that comprises a gastrointestinal device (e.g., an implant) and an anti-scarring agent or a composition comprising an anti-scarring agent into an animal host, wherein the agent inhibits scarring.
9868. The method of item 9867 wherein the agent is an adensosine A2A receptor antagonist.
9869. The method of item 9867 wherein the agent is an AKT inhibitor. 9870. The method of item 9867 wherein the agent is an alpha 2 integrin antagonist, wherein the alpha 2 integrin antagonist is Pharmaprojects No. 5754 (Merck KgaA).
9871. The method of item 9867 wherein the agent is an alpha 4 integrin antagonist.
9872. The method of item 9867 wherein the agent is an alpha 7 nicotinic receptor agonist.
9873. The method of item 9867 wherein the agent is an angiogenesis inhibitor selected from the group consisting of AG-12,958 (Pfizer), ATN-161 (Attention LLC), neovastat, an angiogenesis inhibitor from Jerina AG (Germany), NM-3 (Mercian), VGA-1155 (Taisho), FCE-26644 (Pfizer), FCE-26950 (Pfizer), FPMA (Meiji Daries), FR-111142 (Fujisawa), GGTI-298, GM-1306 (Ligand), GPA-1734 (Novartis), NNC-47-0011 (Novo Nordisk), herbamycin (Nippon Kayaku), lenalidomide (Celegene), IP-10 (NIH), ABT-828 (Abbott), KIN-841 (Tokushima University, Japan), SF-1126 (Semafore Pharmaceuticals), laminin technology (NIH), CHlR-258 (Chiron), NVP-AEW541 (Novartis), NVP-AEW541 (Novartis), Vt16907 (Alchemia), OXI-8007 (Oxigene), EG-3306 (Ark Therapeutics), Maspin (Arriva), ABT- 567 (Abbott), PPI-2458 (Praecis Pharmaceuticals), CC-5079, CC-4089 (Celgene), HlF-1alpha inhibitors (Xenova), S-247 (Pfizer), AP-23573 (Ariad), AZD-9935 (Astra Zeneca), mebendazole (Introgen Therapeutics), MetAP-2 inhibitors (GlaxoSmithKline), AG-615 (Angiogene Pharmaceuticals), Tie-2 antagonists (Hybrigenics), NC-381, CYC-381, NC-169, NC-219, NC-383, NC-384, NC-407 (Lorus Therapeutics), ATN-224 (Attenuon), ON-01370 (Onconova), Vitronectin antagonists (Amgen), SDX-103 (Salmedix), Vitronectin antagonists (Shire), CHP (Riemser), TEK (Amgen), Anecortave acetate (Alcon), T46.2 (Matrix Therapeutics), HG-2 (Heptagen), TEM antagonists (Genzyme), Oxi-4500 (Oxigene), ATN-161 (Attenuon), WX-293 (Wilex), M-2025 (Metris Therapeutics), Alphastatin (BioActa), YH-16 (Yantai Rongchang), BIBF-1120 (Boehringer Ingelheim), BAY-57-9352 (Bayer), AS- 1404 (Cancer Research Technology), SC-77964 (Pfizer), glycomimetics (BioTie Therapies), TIE-2 Inhibitors (Ontogen), DIMI, Octamer (Octamer), ABR-215050 (Active Biotech), ABT-518 (Abbott), KDR inhibitors (Abbott), BSF-466895 (Abbott), SCH-221153 (Schering-Plough), DAC:antiangiogenic (ConjuChem), TFPI (EntreMed), AZD-2171 (Astra-Zenaca), CDC-394 (Celgene), LY290293 (EIi Lilly), IDN-5390 (Indena), Kdr Kinase Inhibitors (Merck), CT-113020, CT-116433, CT-116563, CT-31890, CT-32228) (Cell Therapeutics), A-299620 (Abbott), TWEAK Inhibitor (Amgen), VEGF modulators (Johnson and Johnson), Tum-N53, tumstatin (Genzyme), Thios- 1 , Thios-2 (Thios Pharmaceuticals), MV-6401 (Miravant Medical Technologies), Spisulosine (PharmaMar), CEP-7055 (Cephalon), AUV-201 (Auvation), LM-609 (EIi Lilly), SKF-106615 (AnorMED), Oglufanide disodium (Cytran), BW-114 (Pharminox), Calreticulin (NIH), WX-678 (Wilex), SD-7784 (Pfizer), WX-UK1 (Wilex), SH-268 (Schering AG), 2-Me-PGA (Celgene), S- 137 (Pfizer), ZD-6126 (Angiogene Pharmaceuticals), SG-292 (SignalGen), Benefin (Lane Labs), A6, A36 (Angstrom), SB-2723005 (GlaxoSmithKline), SC-7 (Cell Therapeutics), ZEN-014 (AEterna Zentaris), 2-methoxyestradiol (EntreMed), NK-130119 (Nippon Kayaku), CC-10004 (Celgene), AVE- 8062A (Ajinomoto), Tacedinaline (Pfizer), Actinonin (Tokyo Metropolitan Institute of Medical Science), Lenalidomide (Celgene), VGA-1155, BTO-956 (SRI International), ER-68203-00 (Eisai), CT-6685 (UCB), JKC-362 (Phoenix Pharmaceuticals), DMI-3798 (DMI Biosciences, Angiomate (Ipsen), ZD-6474 (AstraZeneca), CEP-5214 (Cephalon), Canstatin (Genzyme), NM-3 (Mercian), Oridigm (MediQuest Therapeutics), Exherin (Adherex), BLS-0597 (Boston Life Sciences), PTC-299 (PTC Therapeutics), NPI-2358 (Nereus Pharmaceuticals), CGP-79787 (Novartis), AEE-788 (Novartis), CKD-732 (Chong Kun Dang), CP-564959 (OSI Pharmaceuticals), CM-101 (CarboMed), CT-2584, CT3501 (Cell Therapeutics), combretastatin and analogues and derivatives thereof (Oxigene), Rebimastat (Bristol-Meyers Squibb), Dextrin 2-sulfate (ML Laboratories), Cilengitide (Merk KGaA), NSC-706704 (Pharminox), KRN- 951 (Kirin Brewery), Ukrain, NSC-631570 (Nowicky Pharma), Tecogalan sodium (Daiichi Pharmaceutical), Tz-93 (Tsumura), TBC-1635 (Encysive Pharmaceuticals), TAN-1120 (Takeda), Semaxanib (Pfizer), BDI-7800 (Biopharmacopae), SD-186, SD-983 (Bristol-Meyers Squibb), SB-223245 (GlaxoSmithKline), SC-236 (Pfizer), RWJ-590973 (Johnson and Johnson), ILX-1850 (Genzyme), SC-68488, S-836 (Pfizer), CG-55069-11 (CuraGen), Ki-23057 (Kirin Brewery), CCX-700 (Chemoentryx), Pegaptanib octasodium (Giled Sciences), ANGIOCOL (available from Biostratum Inc.), or an analogue or derivative thereof.
9874. The method of item 9867 wherein the agent is an apoptosis antagonist.
9875. The method of item 9867 wherein the agent is an apoptosis activator.
9876. The method of item 9867 wherein the agent is a beta 1 integrin antagonist.
9877. The method of item 9867 wherein the agent is a beta tubulin inhibitor.
9878. The method of item 9867 wherein the agent is a blocker of enzyme production in Hepatitis C.
9879. The method of item 9867 wherein the agent is a Bruton's tyrosine kinase inhibitor. 9880. The method of item 9867 wherein the agent is a calcineurin inhibitor.
9881. The method of item 9867 wherein the agent is a caspase 3 inhibitor.
9882. The method of item 9867 wherein the agent is a CC chemokine receptor antagonist.
9883. The method of item 9867 wherein the agent is a cell cycle inhibitor selected from the group consisting of SNS-595 (Sunesis), synthadotin, KRX-0403, and an analogue or derivative thereof.
9884. The method of item 9867 wherein the agent is a cathepsin B inhibitor.
9885. The method of item 9867 wherein the agent is a cathepsin K inhibitor, wherein the cathepsin K inhibitor is 462795 (GlaxoSmithKline), INPL-022-D6 (Amura Therapeutics), or an analogue or derivative thereof.
9886. The method of item 9867 wherein the agent is a cathepsin L inhibitor.
9887. The method of item 9867 wherein the agent is a CD40 antagonist.
9888. The method of item 9867 wherein the agent is a chemokine receptor agonist. 9889. The method of item 9867 wherein the agent is a chymase inhibitor.
9890. The method of item 9867 wherein the agent is a collagenase antagonist.
9891. The method of item 9867 wherein the agent is a CXCR antagonist.
9892. The method of item 9867 wherein the agent is a cyclin dependent kinase inhibitor selected from the group consisting of a CDK-1 inhibitor, a CDK-2 inhibitor, a CDK- 4 inhibitor, a CDK-6 inhibitor, a CAK1 inhibitor from GPC Biotech and Bristol-Myers Squibb, RGB-286199 (GPC Biotech), an anticancer agent from Astex Technology, a CAK1 inhibitor from GPC Biotech, a CDK inhibitor from Sanofi-Aventis, a CDK1/CDK2 inhibitor from Amgen, a CDK2 inhibitor from SUGEN-2 (Pfizer), a hearing loss therapy agent (Sound Pharmaceuticals), PD-0332991 (Pfizer), RGB-286199 (GPC Biotech), Ro-0505124 (Hoffmann-La Roche), a Ser/Thr kinase inhibitor from Lilly (EIi Lilly), CVT-2584 (CAS No. 199986-75-9) (CV Therapeutics), and an analogue or derivative thereof.
9893. The method of item 9867 wherein the agent is a cyclooxygenase 1 inhibitor.
9894. The method of item 9867 wherein the agent is a DHFR inhibitor.
9895. The method of item 9867 wherein the agent is a dual integrin inhibitor. 9896. The method of item 9867 wherein the agent is an elastase inhibitor.
9897. The method of item 9867 wherein the agent is an elongation factor-1 alpha inhibitor.
9898. The method of item 9867 wherein the agent is an endothelial growth factor antagonist.
9899. The method of item 9867 wherein the agent is an endothelial growth factor receptor kinase inhibitor selected from the group consisting of sorafenib tosylate (Bayer), AAL-993 (Novartis), ABP-309 (Novartis), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), E- 7080 (Eisai), EG-3306 (Ark Therapeutics), EXEL-2880 (Exelixis), GW- 654652 (GlaxoSmithKline), a KDR inhibitor from LG Life Sciences, CT-6685 and CT-6729 (UCB), KRN-633 and KRN-951 (Kirin Brewery), OSI-930 (OSI Pharmaceuticals), SP-5.2 (Supratek Pharma), SU-11657 (Pfizer), a Tie-2 antagonist (Hybrigenics), a VEGFR-2 kinase inhibitor (Bristol-Myers Squibb), XL-647 (Exelixis), a KDR inhibitor from Abbott Laboratories, sorafenib tosylate, and an analogue or derivative thereof.
9900. The method of item 9867 wherein the agent is an endotoxin antagonist.
9901. The method of item 9867 wherein the agent is an epothilone and tubulin binder.
9902. The method of item 9867 wherein the agent is an estrogen receptor antagonist. 9903. The method of item 9867 wherein the agent is an FGF inhibitor.
9904. The method of item 9867 wherein the agent is a farnexyl transferase inhibitor.
9905. The method of item 9867 wherein the agent is farnesyltransferase inhibitor selected from the group of A-197574 (Abbott), a famesyltransferase inhibitor from Servier, FPTIII (Strathclyde Institute for Drug R), LB-42908 (LG Life Sciences), Pharmaprojects No. 5063 (Genzyme), Pharmaprojects No. 5597 (Ipsen), Yissum Project No. B-1055 (Yissum), and an analogue or derivative thereof.
9906. The method of item 9867 wherein the agent is an FLT-3 kinase inhibitor.
9907. The method of item 9867 wherein the agent is an FGF receptor kinase inhibitor.
9908. The method of item 9867 wherein the agent is a fibrinogen antagonist selected from the group consisting of AUV-201 (Auvation), MG-13926 (Sanofi-Aventis), plasminogen activator (CAS No. 105913-11-9) (from Sanofi-Aventis or UCB), plasminogen activator-2 (tPA- 2) (Sanofi-Aventis), pro-urokinase (CAS No. 82657-92-9) (Sanofi-Aventis), and an analogue or derivative thereof.
9909. The method of item 9867 wherein the agent is a heat shock protein 90 antagonist selected from the group consisting of SRN-005 (Sirenade), geldanamycin, NSC-33050 (17-allylaminogeldanamycin; 17- AAG), 17-dimethylaminoethylamino-17-demethoxy-geldanamycin (17- DMAG), rifabutin (rifamycin XiV, 1',4-didehydro-1-deoxy-1 ,4-dihydro-5'-(2- methylpropyl)-1-oxo-), and an analogue or derivative thereof.
9910. The method of item 9867 wherein the agent is a histone deacetylase inhibitor.
9911. The method of item 9867 wherein the agent is an HMGCoA reductase inhibitor selected from the group consisting of an atherosclerosis therapeutic from Lipid Sciences, ATI-16000 (ARYx Therapeutics), KS-01-019 (Kos Pharmaceuticals), Pharmaprojects No. 2197 (Sanofi-Aventi), RP 61969 (Sanofi-Aventis), and an analogue or derivative thereof.
9912. The method of item 9867 wherein the agent is an ICAM inhibitor.
9913. The method of item 9867 wherein the agent is an IL, ICE and IRAK antagonist, wherein the antagonist is a CJ-14877, CP-424174 (Pfizer), NF-61 (Negma-Lerads), and an analogue or derivative thereof.
9914. The method of item 9867 wherein the agent is an IL-2 inhibitor.
9915. The method of item 9867 wherein the agent is an immunosuppressant selected from the group consisting of teriflunomide (Sanofi Aventis), chlorsulfaquinoxalone (NSC-339004), chlorsulfaquinoxalone sulfate, CS-712 (Sankyo), ismomultin alfa (CAS No. 457913-93-8) (Akzo Nobel), antiallergics from GenPat.77, antiinflammatories or AT-005 (Androclus Therapeutics), autoimmune disease therapeutics from EpiVax, BN-007 (Bone), budesonide (CAS No. 51333-22- 3) (MAP Pharmaceuticals), CO-14 (Genzyme), edratide (CAS No. 433922- 67-9) (Teva), EP-314 (Enanta), eprovafen (CAS No. 101335-99-3) (Sanofi- Aventis), HWA-131 (CAS No. 118788-41-3) (Sanofi-Aventis), immunomodulators from MerLion Pharmaceuticals, immunosuppressives from Alchemia, IPL-12 (Inflazyme), MDL-9563 (CAS No. 27086-86-8) (Sanofi-Aventis), Pharmaprojects No. 2330 (Sanofi-Aventis), Pharmaprojects No. 6426 (Abgenix), PXS-25 (Pharmaxis), rosmarinic acid (CAS No. 20283-92-5) (Sanofi-Aventis), RP 42927 or RP 54745 (CAS No. 135330-08-4) (Sanofi-Aventis), SGN-35 (Seattle Genetics), ST-1959 (Sigma-Tau), type I diabetes therapy from SYNX Pharma, UNIL-88 (Debiopharm), VP-025 (Vasogen), VR-694 (Vectura), PRTX-001 (Protalex), and an analogue or derivative thereof.
9916. The method of item 9867 wherein the agent is an IMPDH (inosine monophosphate).
9917. The method of item 9867 wherein the agent is an integrin antagonist.
9918. The method of item 9867 wherein the agent is an interleukin antagonist.
9919. The method of item 9867 wherein the agent is an inhibitor of type III receptor tyrosine kinase.
9920. The method of item 9867 wherein the agent is an irreversible inhibitor of enzyme methionine aminopeptidase type 2.
9921. The method of item 9867 wherein the agent is an isozyme selective delta protein kinase C inhibitor. 9922. The method of item 9867 wherein the agent a JAK3 enzyme inhibitor.
9923. The method of item 9867 wherein the agent is a JNK inhibitor.
9924. The method of item 9867 wherein the agent is a kinase inhibitor.
9925. The method of item 9867 wherein the agent is kinesin antagonist.
9926. The method of item 9867 wherein the agent is a kinesin antagonist.
9927. The method of item 9867 wherein the agent is a leukotriene inhibitor and antagonist selected from the group consisting of ambicromil (CAS No. 58805-38-2) (Sanofi-Aventis), amelubant (CAS No. 346735-24-8) (Boehringer Ingelheim), DW-1141 (Dong Wha), ebselen (Daiichi Pharmaceutical), ibudilast (Kyorin), leucotriene inhibitors from Sanofi-Aventis, lymphotoxin-beta receptor (LT-D) from Biogen Idee, Pharmaprojects No. 1535 and 2728 (CAS No. 119340-33-9) (Sanofi- Aventis), R-112 (Rigel), Rev-5367 (CAS No. 92532-05-3) (Sanofi-Aventis), RG-14893 (CAS No. 141835-49-6) (Sanofi-Aventis), RG-5901-A (CAS No. 101910-24-1), 92532-23-5, RP 66153 (CAS No. 142422-79-5), RP 66364 (CAS No.186916-92-5), RP 69698 (CAS No. 141748-00-7) (Sanofi-Aventis), SC-411930 (Pfizer), SC-41930 (CAS No. 120072-59-5) (Pfizer), SC-50605 (CAS No. 138828-39-4) (Pfizer), SC-51146 (CAS No. 141059-52-1), SC- 53228 (CAS No. 153633-01-3) (Pfizer), spaglumic acid (ZY-15106) (CAS No.3106-85-2), 80619-64-3 (Novartis), tipredane (CAS No. 85197-77-9) (Bristol-Myers Squibb), U-75302 (CAS No. 119477-85-9) (Pfizer), and analogue or derivative thereof.
9928. The method of item 9867 wherein the agent is an MAP kinase inhibitor.
9929. The method of item 9867 wherein the agent is a matrix metalloproteinase inhibitor.
9930. The method of item 9867 wherein the agent is an MCP- CCR2 inhibitor.
9931. The method of item 9867 wherein the agent is an mTOR inhibitor.
9932. The method of item 9867 wherein the agent is an mf OR kinase inhibitor.
9933. The method of item 9867 wherein the agent is a microtubule inhibitor selected from the group consisting of antibody- maytansinoid conjugates from Biogen Idee, colchicines (MantiCore Pharmaceuticals), anticancer immunoconjugates from Johnson & Johnson, DIME from Octamer, gni-1f (GNI), huC242-DM4, huMy9-6-DM1 (ImmunoGen), IDN-5404 (Indena), IMO-098, IMOderm (Imotep), mebendazole (Introgen Therapeutics), microtubule poisons from Cambridge Enterprise, paclitaxel such as LOTAX from Aphios (CAS No. 33069-62-4), Genexol-PM from Samyang, Pharmaprojects No. 6383 (Tapestry Pharmaceuticals), RPR-112378 (Sanofi-Aventis), SGN-75 (Seattle Genetics), SPL-7435 (Starpharma), SSR-250411 (Sanofi-Aventis), trastuzumab-DM1 (Genentech), vinorelbine, and an analogue or derivative thereof. 9934. The method of item 9867 wherein the agent is an MIF inhibitor.
9935. The method of item 9867 wherein the agent is an MMP inhibitor.
9936. The method of item 9867 wherein the agent is a neurokinin (NK) antagonist selected from the group consisting of anthrotainin (CAS No. 148084-40-6) (Sanofi-Aventis), an IBS thereapeutic from ArQuIe, MDL-105212A (CAS No. 167261-60-1) (Ssanofi-Aventis), Pharmaprojects No. 2744, 3258 (CAS No. 139167-47-8) 4006, 4201, or 5986 (Sanofi-Aventis), RP 67580 (CAS No. 135911-02-3), SR-144190 (CAS No. 201152-86-5), SSR-240600, SSR-241586 (Sanofi-Aventis), TKA-457 (Novartis), vestipitant mesylate (CAS No. 334476-64-1) (GlaxoSmithKline), Win-64821 (Sanofi-Aventis), PRX-96026 (Predix Pharmaceuticals), and an analogue or derivative thereof.
9937. The method of item 9867 wherein the agent is an NF kappa B inhibitor selected from the group consisting of AVE-0545 or AVE- 0547 (Sanofi-Aventis), bortezomib (CAS No. 179324-69-7) (Millennium Pharmaceuticals), dexanabinol (CAS No. 112924-45-5) (Pharmos), dexlipotam (Viatris), Pharmaprojects No. 6283 (INDRA) (OXiGENE), IPL- 576092 (CAS No. 137571-30-3) (Inflazyme), NFKB decoy (Corgentech), NFKB decoy oligo (AnGes MG), NFKB's from Ariad, osteoporosis treatments or S5 (F005) from Fulcrum Pharmaceuticals, P61 (Phytopharm), R-flurbiprofen (CAS No. 5104-49-4) (Encore Pharmaceuticals), and an analogue or derivative thereof.
9938. The method of item 9867 wherein the agent is a nitric oxide agonist. 9939. The method of item 9867 wherein the agent is an ornithine decarboxylase inhibitor.
9940. The method of item 9867 wherein the agent is a p38 MAP kinase inhibitor selected from the group consisting of AZD-6703 (AstraZeneca), JX-401 (Jexys Pharmaceuticals), BMS-2 (Bristol-Myers Squibb), a p38 MAP kinase inhibitor from Novartis, a p38-alpha MAP kinase inhibitor from Amphora, Pharmaprojects No. 5704 (Pharmacopeia), RPR- 200765A (Sanofi-Aventis), SD-282 (Johnson & Johnson), TAK-715 (Takeda), and an analogue or derivative thereof.
9941. The method of item 9867 wherein the agent is a palmitoyl-protein thioesterase inhibitor.
9942. The method of item 9867 wherein the agent is a PDGF receptor kinase inhibitor selected from the group consisting of AAL-993, AMN-107, or ABP-309 (Novartis), AMG-706 (Amgen), BAY-57-9352 (Bayer), CDP-860 (UCB), E-7080 (Eisai), imatinib (CAS No. 152459-95-5) (Novartis), OSI-930 (OSI Pharmaceuticals), RPR-127963E (Sanofi-Aventis), RWJ-540973 (Johnson & Johnson), sorafenib tosylate (Bayer), SU-11657 (Pfizer), tandutinib (CAS No. 387867-13-2) (Millennium Pharmaceuticals), vatalanib (Novartis), ZK-CDK (Schering AG), and an analogue or derivative thereof.
9943. The method of item 9867 wherein the agent is (-)- halofenate (Metabolex), AMG-131 (Amgen), antidiabetics from Japan Tobacco, AZD-4619, AZD-8450, AZD-8677 (AstraZeneca), DRF-10945, balaglitazone (Dr Reddy's), CS-00088, CS-00098 (Chipscreen Biosciences), E-3030 (Eisai), etalocib (CAS No. 161172-51-6) (EIi Lilly), GSK-641597 (Ligand), GSK-677954 (GlaxoSmithKline), GW-409544 (Ligand), GW- 590735 (GlaxoSmithKline), K-111 (Hoffmann-La Roche), LY-518674 (EIi Lilly), LY-674 (Ligand), LY-929 (Ligand), MC-3001 , MC-3002 (MaxoCore Pharmaceuticals), metformin HCI + pioglitazone (CAS No. 1115-70-4 and 112529-15-4), ACTOPLUS MET from Andrx), muraglitazar (CAS No. 331741-94-7) (Bristol-Myers Squibb), naveglitazar (Ligand), oleoylethanolamide (Kadmus Pharmaceuticals), ONO-5129, pioglitazone hydrochloride (CAS No. 111025-46-8 and 112529-15-4) (Takeda), PLX-204 (Plexxikon), PPAR agonists from Genfit, PPAR delta agonists from EIi Lilly, PPAR-alpha agonists from CrystalGenomics, PPAR-gamma modulators and PPAR-β modulators from CareX, rosiglitazone maleate (CAS No. 122324- 73-4 or 155141-29-0) (GlaxoSmithKline), rosiglitazone maleate/glimepir (CAS No. 155141-29-0 and 93479-97-1), AVANDARYL, rosiglitazone maleate/metformin extend (CAS No. 155141-29-0 and 657-24-9), AVANDAMET, rosiglitazone maleate+metformin, AVANDAMET (GlaxoSmithKline), tesaglitazar (AstraZeneca), and an analogue or derivative thereof.
9944. The method of item 9867 wherein the agent is a phosphatase inhibitor.
9945. The method of item 9867 wherein the agent is a phosphodiesterase (PDE) inhibitor selected from the group consisting of avanafil (Tanabe Seiyaku), dasantafil (CAS No. 569351-91-3) (Schering- Plough), A-906119 (CAS No. 134072-58-5), DL-850 (Sanofi-Aventis), GRC- 3015, GRC-3566, GRC-3886 (Glenmark), HWA-153 (CAS No. 56395-66-5) (Sanofi-Aventis), hydroxypumafentrine (Altana), IBFB-130011 , IBFB-14-016, IBFB-140301, IBFB-150007, IBFB-211913 (IBFB Pharma), L-826141 (Merck & Co), medorinone (CAS No. 88296-61-1) (Sanofi-Aventis), MEM- 1917 (Memory Pharmaceuticals), ND-1251 (Neuro3d), PDE inhibitors from ICOS, PDE IV inhibitors from Memory Pharmaceuticals and CrystalGenomics, Pharmaprojects No. 2742 and 6141 (Sanofi-Aventis), QAD-171 (Novartis), RHC-2963 (CAS No. 76993-12-9 and 76993-14-1), RPR-117658, RPR-122818 derivatives, SR-24870 , and RPR-132294 (Sanofi-Aventis), SK-350 (ln2Gen), stroke therapy agents from deCODE Genetics, TAS-203 (Taiho), tofimilast (CAS No. 185954-27-2) (Pfizer), UK- 371800 (Pfizer), WIN-65579 (CAS No. 158020-82-7) (Sanofi-Aventis), IBFB- 130020 (IBFB Pharma), OPC-6535 (CAS No. 145739-56-6) (Oteuka), a phosphodiesterase III inhibitor, enoximone, a phosphodiesterase IV inhibitor, fosfosal, Atopik (Barrier Therapeutics), triflusal, a phosphodiesterase V inhibitor, and an analogue or derivative thereof.
9946. The method of item 9867 wherein the agent is a PKC inhibitor.
9947. The method of item 9867 wherein the agent is a platelet activating factor antagonist.
9948. The method of item 9867 wherein the agent is a platelet-derived growth factor receptor kinase inhibitor.
9949. The method of item 9867 wherein the agent is a prolyl hydroxylase inhibitor.
9950. The method of item 9867 wherein the agent is a polymorphonuclear neutrophil inhibitor.
9951. The method of item 9867 wherein the agent is a protein kinase B inhibitor.
9952. The method of item 9867 wherein the agent is a protein kinase C stimulant. 9953. The method of item 9867 wherein the agent is a purine nucleoside analogue.
9954. The method of item 9867 wherein the agent is a purinoreceptor P2X antagonist.
9955. The method of item 9867 wherein the agent is a Raf kinase inhibitor.
9956. The method of item 9867 wherein the agent is a reversible inhibitor of ErbB1 and ErbB2.
9957. The method of item 9867 wherein the agent is a ribonucleoside triphosphate reductase inhibitor.
9958. The method of item 9867 wherein the agent is an SDF- 1 antagonist.
9959. The method of item 9867 wherein the agent is a sheddase inhibitor.
9960. The method of item 9867 wherein the agent is an SRC inhibitor.
9961. The method of item 9867 wherein the agent is a stromelysin inhibitor.
9962. The method of item 9867 wherein the agent is an Syk kinase inhibitor. 9963. The method of item 9867 wherein the agent is a telomerase inhibitor.
9964. The method of item 9867 wherein the agent is a TGF beta inhibitor selected from the group consisting of pirfenidone (CAS No. 53179-13-8) (MARNAC)1 tranilast (CAS No. 53902-12-8) (Kissei), IN-1130 (ln2Gen), mannose-6-phosphate (BTG), TGF-β antagonists from Inflazyme (Pharmaprojects No. 6075), TGF-β antagonists from Sydney, non-industrial source), TGF-β I receptor kinase inhibitors from EIi Lilly, TGF-β receptor inhibitors from Johnson & Johnson, and an analogue or derivative thereof.
9965. The method of item 9867 wherein the agent is a TNFα antagonist or TACE inhibitor selected from the group consisting of adalimumab (CAS No. 331731-18-1) (Cambridge Antibody Technology), AGIX-4207 (AtheroGenics), AGT-1 (Advanced Biotherapy), an antiinflammatory from Borean Pharma, Cellzome, or Paradigm Therapeutics, anti-inflammatory vaccine (TNF-alpha kinoid) from Neovacs, humanized anti-TNF antibody or an anti-TNF MAb (CB0006) Celltech (UCB), apratastat (CAS No. 287445-51-0) (Wyeth), BMS-561392 (Bristol-Myers Squibb), BN- 006 (Bone), certolizumab pegol (CAS No. 428863-50-7 or CH-138 (UCB), cilomilast (CAS No. 153259-65-5) (GlaxoSmithKline), CR-1 (Nuada Pharmaceuticals), CRx-119 (CombinatoRx), D-5410 (UCB), dacopafant (CAS No. 125372-33-0) (Sanofi-Aventis), dersalazine (CAS No. 188913-57- 7/188913-58-8) (Uriach), etanercept (CAS No. 185243-69-0) (Amgen), ethyl pyruvate (Critical (Critical Therapeutics), golimumab (CAS No. 476181-74-5) (Johnson & Johnson), hormono-immunotherapy from Ipsen, CDP571 (e.g., Humicade from UCB), IC-485 (ICOS), infliximab (CAS No. 170277-31-3) (Johnson & Johnson), IP-751 (Manhattan Pharmaceuticals), ISIS-104838 (CAS No. 250755-32-9) (ISIS Pharmaceuticals), lenalidomide (CAS No. 191732-72-6) (Celgene), lentinan (CAS No. 37339-90-5) (Ajinomoto), MDL- 201112 (CAS No. 142130-73-2) (Sanofi-Aventis), medroxyprogesterone (CAS No. 520-85-4) (InKine Pharmaceutical), N-acetylcysteine (CAS No. 616-91-1) (Zambon), NBE-P2 (DIREVO Biotech), nerelimomab (CAS No. 162774-06-3) (Chiron), OM-294DP (OM PHARMA), onercept (CAS No. 199685-57-9) (Yeda), PASSTNF-alpha (Verigen), pentoxifylline or oxypentifylline (Sanofi-Aventis), Pharmaprojects No. 4091 , 4241, 4295, or 4488 (Sanofi-Aventis), Pharmaprojects No. 5480 (Amgen), Pharmaprojects No. 6749 (Cengent), pirfenidone (CAS No. 53179-13-8) (MARNAC), RPR- 132294 (Sanofi-Aventis), S5 (F002) (Fulcrum Pharmaceuticals), simvastatin (CAS No. 79902-63-9) (Merck & Co), STA-6292 (Synta Pharmaceuticals), tacrolimus (CAS No. 104987-11-3) (Fujisawa LifeCycle Pharma), talactoferrin alfa (CAS No. 308240-58-6) (Agennix), thalidomide (CAS No. 50-35-1) (Celgene), TNF antagonists form ProStrakan, and Synergen, TNF inhibitors (Amgen), TNF-alpha antagonists from Dynavax Technologies and Jerina AG (Germany), TNF-alpha inhibitors from IBFB Pharma and Xencor (Xencor), torbafylline (CAS No. 105102-21-4) (Sanofi-Aventis), UR-1505 (Uriach), VT-346 (Viron Therapeutics), YSIL6 (Y's Therapeutics), YSTH2 (Y's Therapeutics), NPI-1302a-3 (Nereus Pharmaceuticals, a TNF antagonist from Jerina AG (Germany), dersalazine, and an analogue or derivative thereof.
9966. The method of item 9867 wherein the agent is a Toll receptor inhibitor.
9967. The method of item 9867 wherein the agent is a tubulin antagonist.
9968. The method of item 9867 wherein the agent is a tyrosine kinase inhibitor selected from the group consisting of SU-011248, SUTENT from Pfizer Inc. (New York, NY), BMS-354825, PN-355 (Paracelsian Pharmaceuticals), AGN-199659 (Allergan), AAL-993 or ABP- 309 (Novartis), adaphostin (NIH), AEE-788 (Novartis), AG-013736 (OSI Pharmaceuticals), AG-13736 (Pfizer), ALT-110 (Alteris Therapeutics), AMG- 706 (Amgen), anticancer MAbs from Xencor, anti-EGFrvlll MAbs from Abgenix, anti-HER2 MAb from Abiogen, AZD-2171 or AZD-9935 (AstraZeneca), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), CEP-5214 (Cephalon), CEP-7055 (Cephalon), cetuximab (ImCIone Systems), CHIR-200131 and CHIR-258 (Chiron), CP-547632 (OSI Pharmaceuticals), CP-724714 (Pfizer), CT-301 (Creabilis Therapeutics), D- 69491 (Baxter International), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EGFR/ErbB2 inhibitors from Array BioPharma, erlotinib (CAS No. 183319- 69-9) (OSI Pharmaceuticals), EXEL-2880 (Exelixis), FK-778 (Sanofi- Aventis) , gefitinib (CAS No. 184475-35-2) (AstraZeneca), GW-2286 or GW- 654652 (GlaxoSmithKline), her2/neu antigen from AlphaVax, HER-2/neu inhibitor from Generex, Herzyme (Medipad) (Sirna Therapeutics), HKI-272 (Wyeth), HuMax-EGFr (Genmab), idronoxil (CAS No. 81267-65-4) (Novogen), IGF-1 inhibitors from Ontogen, IMC-11 F8 (ImCIone Systems), kahalalide F (CAS No. 149204-42-2) (PharmaMar), KDR inhibitor from LG Life Sciences, KDR inhibitors from Abbott Laboratories, KDR kinase inhibitors (UCB), Kdr kinase inhibitors from Merck & Co, KRN-633 and KRN- 951 (Kirin Brewery), KSB-102 (Xenova), lapatinib ditosylate (CAS No. 388082-78-8) (GlaxoSmithKline), matuzumab (Merck KGaA), MDX-214 (Medarex), ME-103 (Pharmexa), MED-A300 (Gerolymatos), MNAC-13 (Lay Line Genomics), nimotuzumab (Center of Molecular Immunology), NSC- 330507 or NSC-707545 (NIH), NV-50 (Novogen), OSI-930 (OSI Pharmaceuticals), panitumumab (Abgenix), pelitinib (CAS No. 287933-82-7) (Wyeth), pertuzumab (CAS No. 380610-27-5) (Genentech), Pharmaprojects No. 3985 (Sanofi-Aventis), prostate cancer therapeutics from Sequenom (SQPC35, SQPC36, SQPC90), removab and remoxab (Trion Pharma), RG- 13022 (CAS No. 136831-48-6), RG-13291 (CAS No. 138989-50-1), or RG- 14620 (CAS No. 136831-49-7) (Sanofi-Aventis), RM-6427 (Romark), RNAi breast cancer therapy from Benitec, RP 53801 (CAS No. 125882-88-4) (Sanofi-Aventis), sorafenib tosylate (Bayer), SU-11657 (Pfizer), Tie-2 antagonists from Semaia (Hybrigenics), Tie-2 inhibitors from Ontogen, trastuzumab (CAS No. 180288-69-1) (Genentech), tyrosine kinase inhibitors from Sanofi-Aventis, U3-1287, U3-1565, U3-1784, U3-1800 (U3 Pharma), vatalanib (Novartis), VEGFR-2 kinase inhibitor from Bristol-Myers Squibb, XL-647 (Exelixis), ZD-6474 (AstraZeneca), ZK-CDK (Schering AG), an EGFR tyrosine kinase inhibitor, EKB-569 (Wyeth), and an analogue or derivative thereof.
9969. The method of item 9867 wherein the agent is a VEGF inhibitor.
9970. The method of item 9867 wherein the agent is a vitamin D receptor agonist.
9971. The method of item 9867 wherein the agent is ZD-6474 (an angiogenesis inhibitor).
9972. The method of item 9867 wherein the agent is AP- 23573 (an mTOR inhibitor).
9973. The method of item 9867 wherein the agent is synthadotin (a tubulin antagonist).
9974. The method of item 9867 wherein the agent is S-0885 (a collagenase inhibitor).
9975. The method of item 9867 wherein the agent is aplidine (an elongation factor-1 alpha inhibitor).
9976. The method of item 9867 wherein the agent is ixabepilone (an epithilone). 9977. The method of item 9867 wherein the agent is IDN- 5390 (an angiogenesis inhibitor).
9978. The method of item 9867 wherein the agent is SB- 2723005 (an angiogenesis inhibitor).
9979. The method of item 9867 wherein the agent is ABT-518 (an angiogenesis inhibitor).
9980. The method of item 9867 wherein the agent is combretastatin (an angiogenesis inhibitor).
9981. The method of item 9867 wherein the agent is anecortave acetate (an angiogenesis inhibitor).
9982. The method of item 9867 wherein the agent is SB- 715992 (a kinesin antagonist).
9983. The method of item 9867 wherein the agent is temsirolimus (an mTOR inhibitor).
9984. The method of item 9867 wherein the agent is adalimumab (a TNFα antagonist).
9985. The method of item 9867, wherein the composition comprises a polymer.
9986. The method of item 9867, wherein the composition comprises a polymer, and the polymer is, or comprises, a copolymer. 9987. The method of item 9867, wherein the composition comprises a polymer, and the polymer is, or comprises, a block copolymer.
9988. The method of item 9867, wherein the composition comprises a polymer, and the polymer is, or comprises, a random copolymer.
9989. The method of item 9867, wherein the composition comprises a polymer, and the polymer is, or comprises, a biodegradable polymer.
9990. The method of item 9867, wherein the composition comprises a polymer, and the polymer is, or comprises, a nonbiodegradable polymer.
9991. The method of item 9867, wherein the composition comprises a polymer, and the polymer is, or comprises, a hydrophilic polymer.
9992. The method of item 9867, wherein the composition comprises a polymer, and the polymer is, or comprises, a hydrophobic polymer.
9993. The method of item 9867, wherein the composition comprises a polymer, and the polymer is, or comprises, a polymer having hydrophilic domains.
9994. The method of item 9867, wherein the composition comprises a polymer, and the polymer is, or comprises, a polymer having hydrophobic domains. 9995. The method of item 9867, wherein the composition comprises a polymer, and the polymer is, or comprises, a non-conductive polymer.
9996. The method of item 9867, wherein the composition comprises a polymer, and the polymer is, or comprises, an elastomer.
9997. The method of item 9867, wherein the composition comprises a polymer, and the polymer is, or comprises, a hydrogel.
9998. The method of item 9867, wherein the composition comprises a polymer, and the polymer is, or comprises, a silicone polymer.
9999. The method of item 9867, wherein the composition comprises a polymer, and the polymer is, or comprises, a hydrocarbon polymer.
10000. The method of item 9867, wherein the composition comprises a polymer, and the polymer is, or comprises, a styrene-derived polymer.
10001. The method of item 9867, wherein the composition comprises a polymer, and the polymer is, or comprises, a butadiene-derived polymer.
10002. The method of item 9867, wherein the composition comprises a polymer, and the polymer is, or comprises, a macromer. 10003. The method of item 9867, wherein the composition comprises a polymer, and the polymer is, or comprises, a poly(ethylene glycol) polymer.
10004. The method of item 9867, wherein the composition comprises a polymer, and the polymer is, or comprises, an amorphous polymer.
10005. The method of item 9867, wherein the composition further comprises a second pharmaceutically active agent.
10006. The method of item 9867, wherein the composition further comprises an anti-inflammatory agent.
10007. The method of item 9867, wherein the composition further comprises an agent that inhibits infection.
10008. The method of item 9867, wherein the composition further comprises an anthracycline.
10009. The method of item 9867, wherein the composition further comprises doxorubicin.
10010. The method of item 9867 wherein the composition further comprises mitoxantrone.
10011. The method of item 9867 wherein the composition further comprises a fluoropyrimldine.
10012. The method of item 9867, wherein the composition further comprises 5-fluorouracil (5-FU). 10013. The method of item 9867, wherein the composition further comprises a folic acid antagonist.
10014. The method of item 9867, wherein the composition further comprises methotrexate.
10015. The method of item 9867, wherein the composition further comprises a podophylotoxin.
10016. The method of item 9867, wherein the composition further comprises etoposide.
10017. The method of item 9867, wherein the composition further comprises camptothecin.
10018. The method of item 9867, wherein the composition further comprises a hydroxyurea.
10019. The method of item 9867, wherein the composition further comprises a platinum complex.
10020. The method of item 9867, wherein the composition further comprises cisplatin.
10021. The method of item 9867 wherein the composition further comprises an anti-thrombotic agent.
10022. The method of item 9867, wherein the composition further comprises a visualization agent. 10023. The method of item 9867, wherein the composition further comprises a visualization agent, and the visualization agent is a radiopaque material, wherein the radiopaque material comprises a metal, a halogenated compound, or a barium containing compound.
10024. The method of item 9867, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, barium, tantalum, or technetium.
10025. The method of item 9867, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, an MRI responsive material.
10026. The method of item 9867, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, a gadolinium chelate.
10027. The method of item 9867, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, iron, magnesium, manganese, copper, or chromium.
10028. The method of item 9867, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, iron oxide compound.
10029. The method of item 9867, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, a dye, pigment, or colorant.
10030. The method of item 9867 wherein the agent is released in effective concentrations from the composition comprising the agent by diffusion over a period ranging from the time of administration to about 90 days.
10031. The method of item 9867 wherein the agent is released in effective concentrations from the composition comprising the agent by erosion of the composition over a period ranging from the time of administration to about 90 days.
10032. The method of item 9867 wherein the composition further comprises an inflammatory cytokine.
10033. The method of item 9867 wherein the composition further comprises an agent that stimulates cell proliferation.
10034. The method of item 9867 wherein the composition further comprises a polymeric carrier.
10035. The method of item 9867 wherein the composition is in the form of a gel, paste, or spray.
10036. The method of item 9867 wherein the implant is partially constructed with the agent or the composition.
10037. The method of item 9867 wherein the implant is fully constructed with the agent or the composition.
10038. The method of item 9867 wherein the implant is impregnated with the agent or the composition. 10039. The method of item 9867, wherein the agent or the composition forms a coating, and the coating directly contacts the implant.
10040. The method of item 9867, wherein the agent or the composition forms a coating, and the coating indirectly contacts the implant.
10041. The method of item 9867 wherein the agent or the composition forms a coating, and the coating partially covers the implant.
10042. The method of item 9867, wherein the agent or the composition forms a coating, and the coating completely covers the implant.
10043. The method of item 9867 wherein the agent or the composition is located within pores or holes of the implant.
10044. The method of item 9867 wherein the agent or the composition is located within a channel, lumen, or divet of the implant.
10045. The method of item 9867 wherein the implant further comprising an echogenic material.
10046. The method of item 9867 wherein the implant further comprises an echogenic material, wherein the echogenic material is in the form of a coating.
10047. The method of item 9867 wherein the implant is sterile. 10048. The method of item 9867 wherein the agent is delivered from the implant, wherein the agent is released into tissue in the vicinity of the implant after deployment of the implant.
10049. The method of item 9867 wherein the agent is delivered from the implant, wherein the agent is released into tissue in the vicinity of the implant after deployment of the implant, wherein the tissue is connective tissue.
10050. The method of item 9867 wherein the agent is delivered from the implant, wherein the agent is released into tissue in the vicinity of the implant after deployment of the implant, wherein the tissue is muscle tissue.
10051. The method of item 9867 wherein the agent is delivered from the implant, wherein the agent is released into tissue in the vicinity of the implant after deployment of the implant, wherein the tissue is nerve tissue.
10052. The method of item 9867 wherein the agent is delivered from the implant, wherein the agent is released into tissue in the vicinity of the implant after deployment of the implant, wherein the tissue is epithelium tissue.
10053. The method of item 9867 wherein the agent is delivered from the implant, wherein the agent is released in effective concentrations from the implant over a period ranging from the time of deployment of the implant to about 1 year.
10054. The method of item 9867 wherein the agent is delivered from the implant, wherein the agent is released in effective concentrations from the implant over a period ranging from about 1 month to 6 months.
10055. The method of item 9867 wherein the agent is delivered from the implant, wherein the agent is released in effective concentrations from the implant over a period ranging from about 1 - 90 days.
10056. The method of item 9867 wherein the agent is delivered from the implant, wherein the agent is released in effective concentrations from the implant at a constant rate.
10057. The method of item 9867 wherein the agent is delivered from the implant, wherein the agent is released in effective concentrations from the implant at an increasing rate.
10058. The method of item 9867 wherein the agent is delivered from the implant, wherein the agent is released in effective concentrations from the implant at a decreasing rate.
10059. The method of item 9867 wherein the agent is delivered from the implant, wherein the implant comprises about 0.01 Dg to about 10 Dg of the agent.
10060. The method of item 9867 wherein the agent is delivered from the implant, wherein the implant comprises about 10 Dg to about 10 mg of the agent.
10061. The method of item 9867 wherein the agent is delivered from the implant, wherein the implant comprises about 10 mg to about 250 mg of the agent. 10062. The method of item 9867 wherein the agent is delivered from the implant, wherein the implant comprises about 250 mg to about 1000 mg of the agent.
10063. The method of item 9867 wherein the agent is delivered from the implant, wherein the implant comprises about 1000 mg to about 2500 mg of the agent.
10064. The method of item 9867 wherein the agent is delivered from the implant, wherein a surface of the implant comprises less than 0.01 Dg of the agent per mm2 of implant surface to which the agent is applied.
10065. The method of item 9867 wherein the agent is delivered from the implant, wherein a surface of the implant comprises about 0.01 Dg to about 1 Dg of the agent per mm2 of implant surface to which the agent is applied.
10066. The method of item 9867 wherein the agent is delivered from the implant, wherein a surface of the implant comprises about 1 Dg to about 10 Dg of the agent per mm2 of implant surface to which the agent is applied.
10067. The method of item 9867 wherein the agent is delivered from the implant, wherein a surface of the implant comprises about 10 Dg to about 250 Dg of the agent per mm2 of implant surface to which the agent is applied.
10068. The method of item 9867 wherein the agent is delivered from the implant, wherein a surface of the implant comprises about 250 Dg to about 1000 Dg of the agent per mm2 of implant surface to which the agent is applied.
10069. The method of item 9867 wherein the agent is delivered from the implant, wherein a surface of the implant comprises about 1000 Dg to about 2500 Dg of the agent per mm2 of implant surface to which the agent is applied.
10070. The method of item 9867, wherein the implant further comprises a coating, and the coating is a uniform coating.
10071. The method of item 9867, wherein the implant further comprises a coating, and the coating is a non-uniform coating.
10072. The method of item 9867, wherein the implant further comprises a coating, and the coating is a discontinuous coating.
10073. The method of item 9867, wherein the implant further comprises a coating, and the coating is a patterned coating.
10074. The method of item 9867, wherein the implant further comprises a coating, and the coating has a thickness of 100 Dm or less.
10075. The method of item 9867, wherein the implant further comprises a coating, and the coating has a thickness of 10 Dm or less.
10076. The method of item 9867, wherein the implant further comprises a coating, and the coating adheres to the surface of the implant upon deployment of the implant. 10077. The method of item 9867, wherein the implant further comprises a coating, and the coating is stable at room temperature for a period of at least 1 year.
10078. The method of item 9867, wherein the implant further comprises a coating, and the agent is present in the coating in an amount ranging between about 0.0001% to about 1% by weight.
10079. The method of item 9867, wherein the implant further comprises a coating, and the agent is present in the coating in an amount ranging between about 1 % to about 10% by weight.
10080. The method of item 9867, wherein the implant further comprises a coating, and the agent is present in the coating in an amount ranging between about 10% to about 25% by weight.
10081. The method of item 9867, wherein the implant further comprises a coating, and the agent is present in the coating in an amount ranging between about 25% to about 70% by weight.
10082. The method of item 9867, wherein the implant further comprises a coating, and the coating comprises a polymer.
10083. The method of item 9867, wherein the implant comprises a first coating having a first composition and a second coating having a second composition.
10084. The method of item 9867, wherein the implant comprises a first coating having a first composition and a second coating having a second composition, wherein the first composition and the second composition are different. 10085. A method as in any one of items 9867-10084, wherein the device is a drainage tube.
10086. A method as in any one of items 9867-10084, wherein the device is a feeding tube.
10087. A method as in any one of items 9867-10084, wherein the device is a portosystemic shunt.
10088. A method as in any one of items 9867-10084, wherein the device is a shunt for ascite.
10089. A method as in any one of items 9867-10084, wherein the device is a nasogastric or nasoenteral tube.
10090. A method as in any one of items 9867-10084, wherein the device is a gastrostomy or percutaneous feeding tube.
10091. A method as in any one of items 9867-10084, wherein the device is a jejunostomy endoscopic tube.
10092. A method as in any one of items 9867-10084, wherein the device is a colostomy device.
10093. A method as in any one of items 9867-10084, wherein the device is a biliary T-tube.
10094. A method as in any one of items 9867-10084, wherein the device is a biopsy forceps. 10095. A method as in any one of items 9867-10084, wherein the device is a biliary stone removal device.
10096. A method as in any one of items 9867-10084, wherein the device is an endoscopic retrograde cholangiopancretography device.
10097. A method as in any one of items 9867-10084, wherein the device is a dilation balloon.
10098. A method as in any one of items 9867-10084, wherein the device is an enteral feeding device.
10099. A method as in any one of items 9867-10084, wherein the device is a stent.
10100. A method as in any one of items 9867-10084, wherein the device is a low profile device.
10101. A method as in any one of items 9867-10084, wherein the device is a virtual colonoscopy device.
10102. A method as in any one of items 9867-10084, wherein the device is a capsule endoscope.
10103. A method as in any one of items 9867-10084, wherein the device is a retrieval device.
10104. A method as in any one of items 9867-10084, wherein the device is a gastrointestinal device adapted for examining the interior of the gastrointestinal tract. 10105. A method as in any one of items 9867-10084, wherein the device is a gastrointestinal device adapted for irrigation or aspiration of the gastrointestinal tract.
10106. A method as in any one of items 9867-10084, wherein the device is a colostomy device.
10107. A method as in any one of items 9867-10084, wherein the device is a mechanical hemostatic device adapted for control of gastrointestinal bleeding.
10108. A method as in any one of items 9867-10084, wherein the device is a gastrointestinal device adapted for cleaning blocked gastrointestinal tract.
10109. A method as in any one of items 9867-10084, wherein the device is a gastrointestinal device for providing communication between two bodily systems.
10110. A method as in any one of items 9867-10084, wherein the device is a portosystemic shunt.
10111. A method as in any one of items 9867-10084, wherein the device is a dilatation catheter.
10112. A method for inhibiting scarring comprising placing a device that comprises a central venous catheter (e.g., an implant) and an anti-scarring agent or a composition comprising an anti-scarring agent into an animal host, wherein the agent inhibits scarring. 10113. The method of item 10112 wherein the agent is an adensosine A2A receptor antagonist.
10114. The method of item 10112 wherein the agent is an AKT inhibitor.
10115. The method of item 10112 wherein the agent ϊs an alpha 2 integrin antagonist, wherein the alpha 2 integrin antagonist is Pharmaprojects No. 5754 (Merck KgaA).
10116. The method of item 10112 wherein the agent is an alpha 4 integrin antagonist.
10117. The method of item 10112 wherein the agent is an alpha 7 nicotinic receptor agonist.
10118. The method of item 10112 wherein the agent is an angiogenesis inhibitor selected from the group consisting of AG-12,958 (Pfizer), ATN-161 (Attention LLC), neovastat, an angiogenesis inhibitor from Jerina AG (Germany), NM-3 (Mercian), VGA-1155 (Taisho), FCE-26644 (Pfizer), FCE-26950 (Pfizer), FPMA (Meiji Daries), FR-111142 (Fujisawa), GGTI-298, GM-1306 (Ligand), GPA-1734 (Novartis), NNC-47-0011 (Novo Nordisk), herbamycin (Nippon Kayaku), lenalidomide (Celegene), IP-10 (NIH), ABT-828 (Abbott), KIN-841 (Tokushima University, Japan), SF-1126 (Semafore Pharmaceuticals), laminin technology (NIH), CHIR-258 (Chiron), NVP-AEW541 (Novartis), NVP-AEW541 (Novartis), Vt16907 (Alchemia), OXI-8007 (Oxigene), EG-3306 (Ark Therapeutics), Maspin (Arriva), ABT- 567 (Abbott), PPI-2458 (Praecis Pharmaceuticals), CC-5079, CC-4089 (Celgene), HIF-1 alpha inhibitors (Xenova), S-247 (Pfizer), AP-23573 (Ariad), AZD-9935 (Astra Zeneca), mebendazole (Introgen Therapeutics), MetAP-2 inhibitors (GlaxoSmithKline), AG-615 (Angiogene Pharmaceuticals), Tie-2 antagonists (Hybrigenics), NC-381, CYC-381, NC-169, NC-219, NC-383, NC-384, NC-407 (Lorus Therapeutics), ATN-224 (Attention), ON-01370 (Onconova), Vitronectin antagonists (Amgen), SDX-103 (Salmedix), Vitronectin antagonists (Shire), CHP (Riemser), TEK (Amgen), Anecortave acetate (Alcon), T46.2 (Matrix Therapeutics), HG-2 (Heptagen), TEM antagonists (Genzyme), Oxi-4500 (Oxigene), ATN-161 (Attenuon), WX-293 (Wilex), M-2025 (Metris Therapeutics), Alphastatin (BioActa), YH-16 (Yantai Rongchang), BIBF-1120 (Boehringer Ingelheim), BAY-57-9352 (Bayer), AS- 1404 (Cancer Research Technology), SC-77964 (Pfizer), glycomimetics (BioTie Therapies), TIE-2 Inhibitors (Ontogen), DIMI, Octamer (Octamer), ABR-215050 (Active Biotech), ABT-518 (Abbott), KDR inhibitors (Abbott), BSF-466895 (Abbott), SCH-221153 (Schering-Plough), DAC:antiangiogenic (ConjuChem), TFPI (EntreMed), AZD-2171 (Astra-Zenaca), CDC-394 (Celgene), LY290293 (EIi Lilly), IDN-5390 (Indena), Kdr Kinase Inhibitors (Merck), CT-113020, CT- 116433, CT-116563, CT-31890, CT-32228) (Cell Therapeutics), A-299620 (Abbott), TWEAK Inhibitor (Amgen), VEGF modulators (Johnson and Johnson), Tum-N53, tumstatin (Genzyme), Thios- 1 , Thios-2 (Thios Pharmaceuticals), MV-6401 (Miravant Medical Technologies), Spisulosine (PharmaMar), CEP-7055 (Cephalon), AUV-201 (Auvation), LM-609 (EIi Lilly), SKF-106615 (AnorMED), Oglufanide disodium (Cytran), BW-114 (Pharminox), Calreticulin (NIH), WX-678 (Wilex), SD-7784 (Pfizer), WX-UK1 (Wilex), SH-268 (Schering AG), 2-Me-PGA (Celgene), S- 137 (Pfizer), ZD-6126 (Angiogene Pharmaceuticals), SG-292 (SignalGen), Benefin (Lane Labs), A6, A36 (Angstrom), SB-2723005 (GlaxoSmithKline), SC-7 (Cell Therapeutics), ZEN-014 (AEterna Zentaris), 2-methoxyestradiol (EntreMed), NK-130119 (Nippon Kayaku), CC-10004 (Celgene), AVE- 8062A (Ajinomoto), Tacedinaline (Pfizer), Actinonin (Tokyo Metropolitan Institute of Medical Science), Lenalidomide (Celgene), VGA-1155, BTO-956 (SRI International), ER-68203-00 (Eisai), CT-6685 (UCB), JKC-362 (Phoenix Pharmaceuticals), DMI-3798 (DMI Biosciences, Angiomate (Ipsen), ZD-6474 (AstraZeneca), CEP-5214 (Cephalon), Canstatin (Genzyme), NM-3 (Mercian), Oridigm (MediQuest Therapeutics), Exherin (Adherex), BLS-0597 (Boston Life Sciences), PTC-299 (PTC Therapeutics), NPI-2358 (Nereus Pharmaceuticals), CGP-79787 (Novartis), AEE-788 (Novartis), CKD-732 (Chong Kun Dang), CP-564959 (OSI Pharmaceuticals), CM-101 (CarboMed), CT-2584, CT3501 (Cell Therapeutics), combretastatin and analogues and derivatives thereof (Oxigene), Rebimastat (Bristol-Meyers Squibb), Dextrin 2-sulfate (ML Laboratories), Cilengitide (Merk KGaA), NSC-706704 (Pharminox), KRN- 951 (Kirin Brewery), Ukrain, NSC-631570 (Nowicky Pharma), Tecogalan sodium (Daiichi Pharmaceutical), Tz-93 (Tsumura), TBC-1635 (Encysive Pharmaceuticals), TAN-1120 (Takeda), Semaxanib (Pfizer), BDI-7800 (Biopharmacopae), SD-186, SD-983 (Bristol-Meyers Squibb), SB-223245 (GlaxoSmithKline), SC-236 (Pfizer), RWJ-590973 (Johnson and Johnson), ILX-1850 (Genzyme), SC-68488, S-836 (Pfizer), CG-55069-11 (CuraGen), Ki-23057 (Kirin Brewery), CCX-700 (Chemoentryx), Pegaptanib octasodium (Giled Sciences), ANGIOCOL (available from Biostratum Inc.), or an analogue or derivative thereof.
10119. The method of item 10112 wherein the agent is an apoptosis antagonist.
10120. The method of item 10112 wherein the agent is an apoptosis activator.
10121. The method of item 10112 wherein the agent is a beta 1 integrin antagonist.
10122. The method of item 10112 wherein the agent is a beta tubulin inhibitor. 10123. The method of item 10112 wherein the agent is a blocker of enzyme production in Hepatitis C.
10124. The method of item 10112 wherein the agent is a Bruton's tyrosine kinase inhibitor.
10125. The method of item 10112 wherein the agent is a calcineurin inhibitor.
10126. The method of item 10112 wherein the agent is a caspase 3 inhibitor.
10127. The method of item 10112 wherein the agent is a CC chemokine receptor antagonist.
10128. The method of item 10112 wherein the agent is a cell cycle inhibitor selected from the group consisting of SNS-595 (Sunesis), synthadotin, KRX-0403, and an analogue or derivative thereof.
10129. The method of item 10112 wherein the agent is a cathepsin B inhibitor.
10130. The method of item 10112 wherein the agent is a cathepsin K inhibitor, wherein the cathepsin K inhibitor is 462795 (GlaxoSmithKline), INPL-022-D6 (Amura Therapeutics), or an analogue or derivative thereof.
10131. The method of item 10112 wherein the agent is a cathepsin L inhibitor. 10132. The method of item 10112 wherein the agent is a CD40 antagonist.
10133. The method of item 10112 wherein the agent is a chemokine receptor agonist.
10134. The method of item 10112 wherein the agent is a chymase inhibitor.
10135. The method of item 10112 wherein the agent is a collagenase antagonist.
10136. The method of item 10112 wherein the agent is a CXCR antagonist.
10137. The method of item 10112 wherein the agent is a cyclin dependent kinase inhibitor selected from the group consisting of a CDK-1 inhibitor, a CDK-2 inhibitor, a CDK- 4 inhibitor, a CDK-6 inhibitor, a CAK1 inhibitor from GPC Biotech and Bristol-Myers Squibb, RGB-286199 (GPC Biotech), an anticancer agent from Astex Technology, a CAK1 inhibitor from GPC Biotech, a CDK inhibitor from Sanofi-Aventis, a CDK1/CDK2 inhibitor from Amgen, a CDK2 inhibitor from SUGEN-2 (Pfizer), a hearing loss therapy agent (Sound Pharmaceuticals), PD-0332991 (Pfizer), RGB-286199 (GPC Biotech), Ro-0505124 (Hoffmann-La Roche), a Ser/Thr kinase inhibitor from Lilly (EIi Lilly), CVT-2584 (CAS No. 199986-75- 9) (CV Therapeutics), and an analogue or derivative thereof.
10138. The method of item 10112 wherein the agent is a cyclooxygenase 1 inhibitor. 10139. The method of item 10112 wherein the agent is a DHFR inhibitor.
10140. The method of item 10112 wherein the agent is a dual integrin inhibitor.
10141. The method of item 10112 wherein the agent is an eiastase inhibitor.
10142. The method of item 10112 wherein the agent is an elongation factor-1 alpha inhibitor.
10143. The method of item 10112 wherein the agent is an endothelial growth factor antagonist.
10144. The method of item 10112 wherein the agent is an endothelial growth factor receptor kinase inhibitor selected from the group consisting of sorafenib tosylate (Bayer), AAL-993 (Novartis), ABP-309 (Novartis), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), E- 7080 (Eisai), EG-3306 (Ark Therapeutics), EXEL-2880 (Exelixis), GW- 654652 (GlaxoSmithKline), a KDR inhibitor from LG Life Sciences, CT-6685 and CT-6729 (UCB), KRN-633 and KRN-951 (Kirin Brewery), OSI-930 (OSI Pharmaceuticals), SP-5.2 (Supratek Pharma), SU-11657 (Pfizer), a Tie-2 antagonist (Hybrigenics), a VEGFR-2 kinase inhibitor (Bristol-Myers Squibb), XL-647 (Exelixis), a KDR inhibitor from Abbott Laboratories, sorafenib tosylate, and an analogue or derivative thereof.
10145. The method of item 10112 wherein the agent is an endotoxin antagonist. 10146. The method of item 10112 wherein the agent is an epothilone and tubulin binder.
10147. The method of item 10112 wherein the agent is an estrogen receptor antagonist.
10148. The method of item 10112 wherein the agent is an FGF inhibitor.
10149. The method of item 10112 wherein the agent is a famexyl transferase inhibitor.
10150. The method of item 10112 wherein the agent is famesyltransferase inhibitor selected from the group of A-197574 (Abbott), a famesyltransferase inhibitor from Servier, FPTIII (Strathclyde Institute for Drug R), LB-42908 (LG Life Sciences), Pharmaprojects No. 5063 (Genzyme), Pharmaprojects No. 5597 (Ipsen), Yissum Project No. B-1055 (Yissum), and an analogue or derivative thereof.
10151. The method of item 10112 wherein the agent is an FLT-3 kinase inhibitor.
10152. The method of item 10112 wherein the agent is an FGF receptor kinase inhibitor.
10153. The method of item 10112 wherein the agent is a fibrinogen antagonist selected from the group consisting of AUV-201 (Auvation), MG-13926 (Sanofi-Aventis), plasminogen activator (CAS No. 105913-11-9) (from Sanofi-Aventis or UCB), plasminogen activator-2 (tPA- 2) (Sanofi-Aventis), pro-urokinase (CAS No. 82657-92-9) (Sanofi-Aventis), and an analogue or derivative thereof. 10154. The method of item 10112 wherein the agent is a heat shock protein 90 antagonist selected from the group consisting of SRN- 005 (Sirenade), geldanamycin, NSC-33050 (17-allylaminogeldanamycin; 17- AAG), ^-dimethylaminoethylamino-^-demethoxy-geldanamycin (17- DMAG), rifabutin (rifamycin XIV, 1l,4-didehydro-1-deoxy-1 ,4-dihydro-5'-(2- methylpropyl)-1-oxo-), and an analogue or derivative thereof.
10155. The method of item 10112 wherein the agent is a histone deacetylase inhibitor.
10156. The method of item 10112 wherein the agent is an HMGCoA reductase inhibitor selected from the group consisting of an atherosclerosis therapeutic from Lipid Sciences, ATI-16000 (ARYx Therapeutics), KS-01-019 (Kos Pharmaceuticals), Pharmaprojects No. 2197 (Sanofi-Aventi), RP 61969 (Sanofi-Aventis), and an analogue or derivative thereof.
10157. The method of item 10112 wherein the agent is an ICAM inhibitor.
10158. The method of item 10112 wherein the agent is an IL, ICE and IRAK antagonist, wherein the antagonist is a CJ-14877, CP- 424174 (Pfizer), NF-61 (Negma-Lerads), and an analogue or derivative thereof.
10159. The method of item 10112 wherein the agent is an IL-2 inhibitor.
10160. The method of item 10112 wherein the agent is an immunosuppressant selected from the group consisting of teriflunomide (Sanofi Aventis), chlorsulfaquinoxalone (NSC-339004), chlorsulfaquinoxalone sulfate, CS-712 (Sankyo), ismomultin alfa (CAS No. 457913-93-8) (Akzo Nobel), antiallergics from GenPat77, antiinflammatories or AT-005 (Androclus Therapeutics), autoimmune disease therapeutics from EpiVax, BN-007 (Bone), budesonide (CAS No. 51333-22- 3) (MAP Pharmaceuticals), CO-14 (Genzyme), edratide (CAS No. 433922- 67-9) (Teva), EP-314 (Enanta), eprovafen (CAS No. 101335-99-3) (Sanofi- Aventis), HWA-131 (CAS No. 118788-41-3) (Sanofi-Aventis), immunomodulators from MerLion Pharmaceuticals, immunosuppressives from Alchemia, IPL-12 (Inflazyme), MDL-9563 (CAS No. 27086-86-8) (Sanofi-Aventis), Pharmaprojects No. 2330 (Sanofi-Aventis), Pharmaprojects No. 6426 (Abgenix), PXS-25 (Pharmaxis), rosmarinic acid (CAS No. 20283-92-5) (Sanofi-Aventis), RP 42927 or RP 54745 (CAS No. 135330-08-4) (Sanofi-Aventis), SGN-35 (Seattle Genetics), ST-1959 (Sigma-Tau), type I diabetes therapy from SYNX Pharma, UNIL-88 (Debiopharm), VP-025 (Vasogen), VR-694 (Vectura), PRTX-001 (Protalex), and an analogue or derivative thereof.
10161. The method of item 10112 wherein the agent is an IMPDH (inosine monophosphate).
10162. The method of item 10112 wherein the agent is an integrin antagonist.
10163. The method of item 10112 wherein the agent is an interleukin antagonist.
10164. The method of item 10112 wherein the agent is an inhibitor of type III receptor tyrosine kinase.
10165. The method of item 10112 wherein the agent is an irreversible inhibitor of enzyme methionine aminopeptidase type 2. 10166. The method of item 10112 wherein the agent is an isozyme selective delta protein kinase C inhibitor.
10167. The method of item 10112 wherein the agent a JAK3 enzyme inhibitor.
10168. The method of item 10112 wherein the agent is a
JNK inhibitor.
10169. The method of item 10112 wherein the agent is a kinase inhibitor.
10170. The method of item 10112 wherein the agent is kinesin antagonist.
10171. The method of item 10112 wherein the agent is a kinesin antagonist.
10172. The method of item 10112 wherein the agent is a leukotriene inhibitor and antagonist selected from the group consisting of ambicromil (CAS No. 58805-38-2) (Sanofi-Aventis), amelubant (CAS No. 346735-24-8) (Boehringer Ingelheim), DW-1141 (Dong Wha), ebselen (Daiichi Pharmaceutical), ibudilast (Kyorin), leucotriene inhibitors from Sanofi-Aventis, lymphotoxin-beta receptor (LT-D) from Biogen Idee, Pharmaprojects No. 1535 and 2728 (CAS No. 119340-33-9) (Sanofi- Aventis), R-112 (Rigel), Rev-5367 (CAS No. 92532-05-3) (Sanofi-Aventis), RG-14893 (CAS No. 141835-49-6) (Sanofi-Aventis), RG-5901-A (CAS No. 101910-24-1), 92532-23-5, RP 66153 (CAS No. 142422-79-5), RP 66364 (CAS No.186916-92-5), RP 69698 (CAS No. 141748-00-7) (Sanofi-Aventis), SC-411930 (Pfizer), SC-41930 (CAS No. 120072-59-5) (Pfizer), SC-50605 (CAS No. 138828-39-4) (Pfizer), SC-51146 (CAS No. 141059-52-1), SC- 53228 (CAS No. 153633-01-3) (Pfizer), spaglumic acid (ZY-15106) (CAS No.3106-85-2), 80619-64-3 (Novartis), tipredane (CAS No. 85197-77-9) (Bristol-Myers Squibb), U-75302 (CAS No. 119477-85-9) (Pfizer), and analogue or derivative thereof.
10173. The method of item 10112 wherein the agent is an MAP kinase inhibitor.
10174. The method of item 10112 wherein the agent is a matrix metalloproteinase inhibitor.
10175. The method of item 10112 wherein the agent is an MCP-CCR2 inhibitor.
10176. The method of item 10112 wherein the agent is an mTOR inhibitor.
10177. The method of item 10112 wherein the agent is an mTOR kinase inhibitor.
10178. The method of item 10112 wherein the agent is a microtubule inhibitor selected from the group consisting of antibody- maytansinoid conjugates from Biogen Idee, colchicines (MantiCore Pharmaceuticals), anticancer immunoconjugates from Johnson & Johnson, DIME from Octamer, gni-1f (GNI), huC242-DM4, huMy9-6-DM1 (ImmunoGen), IDN-5404 (Indena), IMO-098, IMOderm (Imotep), mebendazole (Introgen Therapeutics), microtubule poisons from Cambridge Enterprise, paclitaxel such as LOTAX from Aphios (CAS No. 33069-62-4), Genexol-PM from Samyang, Pharmaprojects No. 6383 (Tapestry Pharmaceuticals), RPR-112378 (Sanofi-Aventis), SGN-75 (Seattle Genetics), SPL-7435 (Starpharma), SSR-250411 (Sanofi-Aventis), trastuzumab-DM1 (Genentech), vinorelbine, and an analogue or derivative thereof.
10179. The method of item 10112 wherein the agent is an MIF inhibitor.
10180. The method of item 10112 wherein the agent is an MMP inhibitor.
10181. The method of item 10112 wherein the agent is a neurokinin (NK) antagonist selected from the group consisting of anthrotainin (CAS No. 148084-40-6) (Sanofi-Aventis), an IBS tbereapeutic from ArQuIe, MDL-105212A (CAS No. 167261-60-1) (Ssanofi-Aventis), Pharmaprojects No. 2744, 3258 (CAS No. 139167-47-8) 4006, 4201, or 5986 (Sanofi-Aventis), RP 67580 (CAS No. 135911-02-3), SR-144190 (CAS No. 201152-86-5), SSR-240600, SSR-241586 (Sanofi-Aventis), TKA-457 (Novartis), vestipitant mesylate (CAS No. 334476-64-1) (GlaxoSmithKline), Win-64821 (Sanofi-Aventis), PRX-96026 (Predix Pharmaceuticals), and an analogue or derivative thereof.
10182. The method of item 10112 wherein the agent is an NF kappa B inhibitor selected from the group consisting of AVE-0545 or AVE-0547 (Sanofi-Aventis), bortezomib (CAS No. 179324-69-7) (Millennium Pharmaceuticals), dexanabinol (CAS No. 112924-45-5) (Pharmos), dexlipotam (Viatris), Pharmaprojects No. 6283 (INDRA) (OXiGENE), IPL- 576092 (CAS No. 137571-30-3) (Inflazyme), NFKB decoy (Corgentech), NFKB decoy oligo (AnGes MG), NFKB's from Ariad, osteoporosis treatments or S5 (F005) from Fulcrum Pharmaceuticals, P61 (Phytopharm), R-flurbiprofen (CAS No. 5104-49-4) (Encore Pharmaceuticals), and an analogue or derivative thereof. 10183. The method of item 10112 wherein the agent is a nitric oxide agonist.
10184. The method of item 10112 wherein the agent is an ornithine decarboxylase inhibitor.
10185. The method of item 10112 wherein the agent is a p38 MAP kinase inhibitor selected from the group consisting of AZD-6703 (AstraZeneca), JX-401 (Jexys Pharmaceuticals), BMS-2 (Bristol-Myers Squibb), a p38 MAP kinase inhibitor from Novartis, a p38-alpha MAP kinase inhibitor from Amphora, Pharmaprojects No. 5704 (Pharmacopeia), RPR- 200765A (Sanofi-Aventis), SD-282 (Johnson & Johnson), TAK-715 (Takeda), and an analogue or derivative thereof.
10186. The method of item 10112 wherein the agent is a palmitoyl-protein thioesterase inhibitor.
10187. The method of item 10112 wherein the agent is a PDGF receptor kinase inhibitor selected from the group consisting of AAL- 993, AMN-107, or ABP-309 (Novartis), AMG-706 (Amgen), BAY-57-9352 (Bayer), CDP-860 (UCB), E-7080 (Eisai), imatinib (CAS No. 152459-95-5) (Novartis), OSI-930 (OSI Pharmaceuticals), RPR-127963E (Sanofi-Aventis), RWJ-540973 (Johnson & Johnson), sorafenib tosylate (Bayer), SU-11657 (Pfizer), tandutinib (CAS No. 387867-13-2) (Millennium Pharmaceuticals), vatalanib (Novartis), ZK-CDK (Sobering AG), and an analogue or derivative thereof.
10188. The method of item 10112 wherein the agent is (-)-halofenate (Metabolex), AMG-131 (Amgen), antidiabetics from Japan Tobacco, AZD-4619, AZD-8450, AZD-8677 (AstraZeneca), DRF-10945, balaglitazone (Dr Reddy's), CS-00088, CS-00098 (Chipscreen Biosciences), E-3030 (Eisai), etalocib (CAS No. 161172-51-6) (EIi Lilly), GSK-641597 (Ligand), GSK-677954 (GlaxoSmithKline), GW-409544 (Ligaπd), GW- 590735 (GlaxoSmithKline), K-111 (Hoffmann-La Roche), LY-518674 (EIi Lilly), LY-674 (Ligand), LY-929 (Ligand), MC-3001 , MC-3002 (MaxoCore Pharmaceuticals), metformin HCI + pioglitazone (CAS No. 1115-70-4 and 112529-15-4), ACTOPLUS MET from Andrx), muraglitazar (CAS No. 331741-94-7) (Bristol-Myers Squibb), naveglitazar (Ligand), oleoylethanolamide (Kadmus Pharmaceuticals), ONO-5129, pioglitazone hydrochloride (CAS No. 111025-46-8 and 112529-15-4) (Takeda), PLX-204 (Plexxikon), PPAR agonists from Genfit, PPAR delta agonists from EIi Lilly, PPAR-alpha agonists from CrystalGenomics, PPAR-gamma modulators and PPAR-β modulators from CareX, rosiglitazone maleate (CAS No. 122324- 73-4 or 155141-29-0) (GlaxoSmithKline), rosiglitazone maleate/glimepir (CAS No. 155141-29-0 and 93479-97-1), AVANDARYL, rosiglitazone maleate/metformin extend (CAS No. 155141-29-0 and 657-24-9), AVANDAMET, rosiglitazone maleate+metformin, AVANDAMET (GlaxoSmithKline), tesaglitazar (AstraZeneca), and an analogue or derivative thereof.
10189. The method of item 10112 wherein the agent is a phosphatase inhibitor.
10190. The method of item 10112 wherein the agent is a phosphodiesterase (PDE) inhibitor selected from the group consisting of avanafil (Tanabe Seiyaku), dasantafil (CAS No. 569351-91-3) (Schering- Plough), A-906119 (CAS No. 134072-58-5), DL-850 (Sanofi-Aventis), GRC- 3015, GRC-3566, GRC-3886 (Glenmark), HWA-153 (CAS No. 56395-66-5) (Sanofi-Aventis), hydroxypumafentrine (Altana), IBFB-130011 , IBFB-14-016, IBFB-140301, IBFB-150007, IBFB-211913 (IBFB Pharma), L-826141 (Merck & Co), medorinone (CAS No. 88296-61-1) (Sanofi-Aventis), MEM- 1917 (Memory Pharmaceuticals), ND-1251 (Neuro3d), PDE inhibitors from ICOS, PDE IV inhibitors from Memory Pharmaceuticals and CrystalGenomics, Pharmaprojects No. 2742 and 6141 (Sanofi-Aventis), QAD-171 (Novartis), RHC-2963 (CAS No. 76993-12-9 and 76993-14-1), RPR-117658, RPR-122818 derivatives, SR-24870 , and RPFM 32294 (Sanofi-Aventis), SK-350 (ln2Gen), stroke therapy agents from deCODE Genetics, TAS-203 (Taiho), tofimilast (CAS No. 185954-27-2) (Pfizer), UK- 371800 (Pfizer), WIN-65579 (CAS No. 158020-82-7) (Sanofi-Aventis), IBFB- 130020 (IBFB Pharma), OPC-6535 (CAS No. 145739-56-6) (Otsuka), a phosphodiesterase III inhibitor, enoximone, a phosphodiesterase IV inhibitor, fosfosal, Atopik (Barrier Therapeutics), triflusal, a phosphodiesterase V inhibitor, and an analogue or derivative thereof.
10191. The method of item 10112 wherein the agent is a PKC inhibitor.
10192. The method of item 10112 wherein the agent is a platelet activating factor antagonist.
10193. The method of item 10112 wherein the agent is a platelet-derived growth factor receptor kinase inhibitor.
10194. The method of item 10112 wherein the agent is a prolyl hydroxylase inhibitor.
10195. The method of item 10112 wherein the agent is a polymorphonuclear neutrophil inhibitor.
10196. The method of item 10112 wherein the agent is a protein kinase B inhibitor. 10197. The method of item 10112 wherein the agent is a protein kinase C stimulant.
10198. The method of item 10112 wherein the agent is a purine nucleoside analogue.
10199. The method of item 10112 wherein the agent is a purinoreceptor P2X antagonist.
10200. The method of item 10112 wherein the agent is a Raf kinase inhibitor.
10201. The method of item 10112 wherein the agent is a reversible inhibitor of ErbB1 and ErbB2.
10202. The method of item 10112 wherein the agent is a ribonucleoside triphosphate reductase inhibitor.
10203. The method of item 10112 wherein the agent is an SDF-1 antagonist.
10204. The method of item 10112 wherein the agent is a sheddase inhibitor.
10205. The method of item 10112 wherein the agent is an SRC inhibitor.
10206. The method of item 10112 wherein the agent is a stromelysin inhibitor. 10207. The method of item 10112 wherein the agent is an Syk kinase inhibitor.
10208. The method of item 10112 wherein the agent is a telomerase inhibitor.
10209. The method of item 10112 wherein the agent is a TGF beta inhibitor selected from the group consisting of pirfenidone (CAS No. 53179-13-8) (MARNAC), tranilast (CAS No. 53902-12-8) (Kissei), IN- 1130 (ln2Gen), mannose-6-phosphate (BTG), TGF-β antagonists from Inflazyme (Pharmaprojects No. 6075), TGF-β antagonists from Sydney, non-industrial source), TGF-β I receptor kinase inhibitors from EIi Lilly, TGF- β receptor inhibitors from Johnson & Johnson, and an analogue or derivative thereof.
10210. The method of item 10112 wherein the agent is a TNFα antagonist or TACE inhibitor selected from the group consisting of adalimumab (CAS No. 331731-18-1) (Cambridge Antibody Technology), AGIX-4207 (AtheroGenics), AGT-1 (Advanced Biotherapy), an antiinflammatory from Borean Pharma, Cellzome, or Paradigm Therapeutics, anti-inflammatory vaccine (TNF-alpha kinoid) from Neovacs, humanized anti-TNF antibody or an anti-TNF MAb (CB0006) Celltech (UCB), apratastat (CAS No. 287445-51-0) (Wyeth), BMS-561392 (Bristol-Myers Squibb), BN- 006 (Bone), certolizumab pegol (CAS No. 428863-50-7 or CH-138 (UCB), cilomilast (CAS No. 153259-65-5) (GlaxoSmithKline), CR-1 (Nuada Pharmaceuticals), CRx-119 (CombinatoRx), D-5410 (UCB), dacopafant (CAS No. 125372-33-0) (Sanofi-Aventis), dersalazine (CAS No. 188913-57- 7/188913-58-8) (Uriach), etanercept (CAS No. 185243-69-0) (Amgen), ethyl pyruvate (Critical (Critical Therapeutics), golimumab (CAS No. 476181-74-5) (Johnson & Johnson), hormono-immunotherapy from Ipsen, CDP571 (e.g., Humicade from UCB), IC-485 (ICOS), infliximab (CAS No. 170277-31-3) (Johnson & Johnson), IP-751 (Manhattan Pharmaceuticals), ISIS-104838 (CAS No. 250755-32-9) (ISIS Pharmaceuticals), lenalidomide (CAS No. 191732-72-6) (Celgene), lentinan (CAS No. 37339-90-5) (Ajinomoto), MDL- 201112 (CAS No. 142130-73-2) (Sanofi-Aventis), medroxyprogesterone (CAS No. 520-85-4) (InKine Pharmaceutical), N-acetylcysteine (CAS No. 616-91-1) (Zambon), NBE-P2 (DIREVO Biotech), nerelimomab (CAS No. 162774-06-3) (Chiron), OM-294DP (OM PHARMA), onercept (CAS No. 199685-57-9) (Yeda), PASSTNF-alpha (Verigen), pentoxifylline or oxypentifylline (Sanofi-Aventis), Pharmaprojects No. 4091 , 4241, 4295, or 4488 (Sanofi-Aventis), Pharmaprojects No. 5480 (Amgen), Pharmaprojects No. 6749 (Cengent), pirfenidone (CAS No. 53179-13-8) (MARNAC)1 RPR- 132294 (Sanofi-Aventis), S5 (F002) (Fulcrum Pharmaceuticals), simvastatin (CAS No. 79902-63-9) (Merck & Co), STA-6292 (Synta Pharmaceuticals), tacrolimus (CAS No. 104987-11-3) (Fujisawa LifeCycle Pharma), talactoferrin alfa (CAS No. 308240-58-6) (Agennix), thalidomide (CAS No. 50-35-1) (Celgene), TNF antagonists form ProStrakan, and Synergen, TNF inhibitors (Amgen), TNF-alpha antagonists from Dynavax Technologies and Jerina AG (Germany), TNF-alpha inhibitors from IBFB Pharma and Xencor (Xencor), torbafylline (CAS No. 105102-21-4) (Sanofi-Aventis), UR-1505 (Uriach), VT-346 (Viron Therapeutics), YSIL6 (Y's Therapeutics), YSTH2 (Y's Therapeutics), NPI-1302a-3 (Nereus Pharmaceuticals, a TNF antagonist from Jerina AG (Germany), dersalazine, and an analogue or derivative thereof.
10211. The method of item 10112 wherein the agent is a Toll receptor inhibitor.
10212. The method of item 10112 wherein the agent is a tubulin antagonist. 10213. The method of item 10112 wherein the agent is a tyrosine kinase inhibitor selected from the group consisting of SU-011248, SUTENT from Pfizer Inc. (New York, NY), BMS-354825, PN-355 (Paracelsian Pharmaceuticals), AGN-199659 (Allergan), AAL-993 or ABP- 309 (Novartis), adaphostin (NIH), AEE-788 (Novartis), AG-013736 (OSI Pharmaceuticals), AG-13736 (Pfizer), ALT-110 (Alteris Therapeutics), AMG- 706 (Amgen), anticancer MAbs from Xencor, anti-EGFrvlll MAbs from Abgenix, anti-HER2 MAb from Abiogen, AZD-2171 or AZD-9935 (AstraZeneca), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), CEP-5214 (Cephalon), CEP-7055 (Cephalon), cetuximab (ImClone Systems), CHIR-200131 and CHIR-258 (Chiron), CP-547632 (OSI Pharmaceuticals), CP-724714 (Pfizer), CT-301 (Creabilis Therapeutics), D- 69491 (Baxter International), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EGFR/ErbB2 inhibitors from Array BioPharma, erlotinib (CAS No. 183319- 69-9) (OSI Pharmaceuticals), EXEL-2880 (Exelixis), FK-778 (Sanofi- Aventis) , gefitinib (CAS No. 184475-35-2) (AstraZeneca), GW-2286 or GW- 654652 (GlaxoSmithKline), her2/neu antigen from AlphaVax, HER-2/neu inhibitor from Generex, Herzyme (Medipad) (Sirna Therapeutics), HKI-272 (Wyeth), HuMax-EGFr (Genmab), idronoxil (CAS No. 81267-65-4) (Novogen), IGF-1 inhibitors from Ontogen, IMC-11 F8 (ImClone Systems), kahalalide F (CAS No. 149204-42-2) (PharmaMar), KDR inhibitor from LG Life Sciences, KDR inhibitors from Abbott Laboratories, KDR kinase inhibitors (UCB), Kdr kinase inhibitors from Merck & Co, KRN-633 and KRN- 951 (Kirin Brewery), KSB-102 (Xenova), lapatinib ditosylate (CAS No. 388082-78-8) (GlaxoSmithKline), matuzumab (Merck KGaA), MDX-214 (Medarex), ME-103 (Pharmexa), MED-A300 (Gerolymatos), MNAC-13 (Lay Line Genomics), nimotuzumab (Center of Molecular Immunology), NSC- 330507 or NSC-707545 (NIH), NV-50 (Novogen), OSI-930 (OSI Pharmaceuticals), panitumumab (Abgenix), pelitinib (CAS No. 287933-82-7) (Wyeth), pertuzumab (CAS No. 380610-27-5) (Genentech), Pharmaprojects No. 3985 (Sanofi-Aventis), prostate cancer therapeutics from Sequenom (SQPC35, SQPC36, SQPC90), removab and remoxab (Trion Pharma), RG- 13022 (CAS No. 136831-48-6), RG-13291 (CAS No. 138989-50-1), or RG- 14620 (CAS No. 136831-49-7) (Sanofi-Aventis), RM-6427 (Romark), RNAi breast cancer therapy from Benitec, RP 53801 (CAS No. 125882-88-4) (Sanofi-Aventis), sorafenib tosylate (Bayer), SU-11657 (Pfizer), Tie-2 antagonists from Semaia (Hybrigenics), Tie-2 inhibitors from Ontogen, trastuzumab (CAS No. 180288-69-1) (Genentech), tyrosine kinase inhibitors from Sanofi-Aventis, U3-1287, U3-1565, U3-1784, U3-1800 (U3 Pharma), vatalanib (Novartis), VEGFR-2 kinase inhibitor from Bristol-Myers Squibb, XL-647 (Exelixis), ZD-6474 (AstraZeneca), ZK-CDK (Schering AG), an EGFR tyrosine kinase inhibitor, EKB-569 (Wyeth), and an analogue or derivative thereof.
10214. The method of item 10112 wherein the agent is a VEGF inhibitor.
10215. The method of item 10112 wherein the agent is a vitamin D receptor agonist.
10216. The method of item 10112 wherein the agent is ZD-6474 (an angiogenesis inhibitor).
10217. The method of item 10112 wherein the agent is AP-23573 (an mTOR inhibitor).
10218. The method of item 10112 wherein the agent is synthadotin (a tubulin antagonist).
10219. The method of item 10112 wherein the agent is S-0885 (a collagenase inhibitor). 10220. The method of item 10112 wherein the agent is aplidine (an elongation factor-1 alpha inhibitor).
10221. The method of item 10112 wherein the agent is ixabepilone (an epithilone).
10222. The method of item 10112 wherein the agent is IDN-5390 (an angiogenesis inhibitor).
10223. The method of item 10112 wherein the agent is SB-2723005 (an angiogenesis inhibitor).
10224. The method of item 10112 wherein the agent is ABT-518 (an angiogenesis inhibitor).
10225. The method of item 10112 wherein the agent is combretastatin (an angiogenesis inhibitor).
10226. The method of item 10112 wherein the agent is anecortave acetate (an angiogenesis inhibitor).
10227. The method of item 10112 wherein the agent is SB-715992 (a kinesin antagonist).
10228. The method of item 10112 wherein the agent is temsirolimus (an mTOR inhibitor).
10229. The method of item 10112 wherein the agent is adalimumab (a TN Fa antagonist). 10230. The method of item 10112, wherein the composition comprises a polymer.
10231. The method of item 10112, wherein the composition comprises a polymer, and the polymer is, or comprises, a copolymer.
10232. The method of item 10112, wherein the composition comprises a polymer, and the polymer is, or comprises, a block copolymer.
10233. The method of item 10112, wherein the composition comprises a polymer, and the polymer is, or comprises, a random copolymer.
10234. The method of item 10112, wherein the composition comprises a polymer, and the polymer is, or comprises, a biodegradable polymer.
10235. The method of item 10112, wherein the composition comprises a polymer, and the polymer is, or comprises, a nonbiodegradable polymer.
10236. The method of item 10112, wherein the composition comprises a polymer, and the polymer is, or comprises, a hydrophilic polymer.
10237. The method of item 10112, wherein the composition comprises a polymer, and the polymer is, or comprises, a hydrophobic polymer. 10238. The method of item 10112, wherein the composition comprises a polymer, and the polymer is, or comprises, a polymer having hydrophilic domains.
10239. The method of item 10112, wherein the composition comprises a polymer, and the polymer is, or comprises, a polymer having hydrophobic domains.
10240. The method of item 10112, wherein the composition comprises a polymer, and the polymer is, or comprises, a non- conductive polymer.
10241. The method of item 10112, wherein the composition comprises a polymer, and the polymer is, or comprises, an elastomer.
10242. The method of item 10112, wherein the composition comprises a polymer, and the polymer is, or comprises, a hydrogel.
10243. The method of item 10112, wherein the composition comprises a polymer, and the polymer is, or comprises, a silicone polymer.
10244. The method of item 10112, wherein the composition comprises a polymer, and the polymer is, or comprises, a hydrocarbon polymer.
10245. The method of item 10112, wherein the composition comprises a polymer, and the polymer is, or comprises, a styrene-derived polymer. 10246. The method of item 10112, wherein the composition comprises a polymer, and the polymer is, or comprises, a butadiene-derived polymer.
10247. The method of item 10112, wherein the composition comprises a polymer, and the polymer is, or comprises, a macromer.
10248. The method of item 10112, wherein the composition comprises a polymer, and the polymer is, or comprises, a poly(ethylene glycol) polymer.
10249. The method of item 10112, wherein the composition comprises a polymer, and the polymer is, or comprises, an amorphous polymer.
10250. The method of item 10112, wherein the composition further comprises a second pharmaceutically active agent.
10251. The method of item 10112, wherein the composition further comprises an anti-inflammatory agent.
10252. The method of item 10112, wherein the composition further comprises an agent that inhibits infection.
10253. The method of item 10112, wherein the composition further comprises an anthracycline.
10254. The method of item 10112, wherein the composition further comprises doxorubicin. 10255. The method of item 10112 wherein the composition further comprises mitoxantrone.
10256. The method of item 10112 wherein the composition further comprises a fluoropyrimidine.
10257. The method of item 10112, wherein the composition further comprises 5-fluorouracil (5-FU).
10258. The method of item 10112, wherein the composition further comprises a folic acid antagonist.
10259. The method of item 10112, wherein the composition further comprises methotrexate.
10260. The method of item 10112, wherein the composition further comprises a podophylotoxin.
10261. The method of item 10112, wherein the composition further comprises etoposide.
10262. The method of item 10112, wherein the composition further comprises camptothecin.
10263. The method of item 10112, wherein the composition further comprises a hydroxyurea.
10264. The method of item 10112, wherein the composition further comprises a platinum complex. 10265. The method of item 10112, wherein the composition further comprises cisplatin.
10266. The method of item 10112 wherein the composition further comprises an antithrombotic agent.
10267. The method of item 10112, wherein the composition further comprises a visualization agent.
10268. The method of item 10112, wherein the composition further comprises a visualization agent, and the visualization agent is a radiopaque material, wherein the radiopaque material comprises a metal, a halogenated compound, or a barium containing compound.
10269. The method of item 10112, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, barium, tantalum, or technetium.
10270. The method of item 10112, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, an MRI responsive material.
10271. The method of item 10112, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, a gadolinium chelate.
10272. The method of item 10112, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, iron, magnesium, manganese, copper, or chromium. 10273. The method of item 10112, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, iron oxide compound.
10274. The method of item 10112, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, a dye, pigment, or colorant.
10275. The method of item 10112 wherein the agent is released in effective concentrations from the composition comprising the agent by diffusion over a period ranging from the time of administration to about 90 days.
10276. The method of item 10112 wherein the agent is released in effective concentrations from the composition comprising the agent by erosion of the composition over a period ranging from the time of administration to about 90 days.
10277. The method of item 10112 wherein the composition further comprises an inflammatory cytokine.
10278. The method of item 10112 wherein the composition further comprises an agent that stimulates cell proliferation.
10279. The method of item 10112 wherein the composition further comprises a polymeric carrier.
10280. The method of item 10112 wherein the composition is in the form of a gel, paste, or spray. 10281. The method of item 10112 wherein the implant is partially constructed with the agent or the composition.
10282. The method of item 10112 wherein the implant is fully constructed with the agent or the composition.
10283. The method of item 10112 wherein the implant is impregnated with the agent or the composition.
10284. The method of item 10112, wherein the agent or the composition forms a coating, and the coating directly contacts the implant.
10285. The method of item 10112, wherein the agent or the composition forms a coating, and the coating indirectly contacts the implant.
10286. The method of item 10112 wherein the agent or the composition forms a coating, and the coating partially covers the implant.
10287. The method of item 10112, wherein the agent or the composition forms a coating, and the coating completely covers the implant.
10288. The method of item 10112 wherein the agent or the composition is located within pores or holes of the implant.
10289. The method of item 10112 wherein the agent or the composition is located within a channel, lumen, or divet of the implant. 10290. The method of item 10112 wherein the implant further comprising an echogenic material.
10291. The method of item 10112 wherein the implant further comprises an echogenic material, wherein the echogenic material is in the form of a coating.
10292. The method of item 10112 wherein the implant is sterile.
10293. The method of item 10112 wherein the agent is delivered from the implant, wherein the agent is released into tissue in the vicinity of the implant after deployment of the implant.
10294. The method of item 10112 wherein the agent is delivered from the implant, wherein the agent is released into tissue in the vicinity of the implant after deployment of the implant, wherein the tissue is connective tissue.
10295. The method of item 10112 wherein the agent is delivered from the implant, wherein the agent is released into tissue in the vicinity of the implant after deployment of the implant, wherein the tissue is muscle tissue.
10296. The method of item 10112 wherein the agent is delivered from the implant, wherein the agent is released into tissue in the vicinity of the implant after deployment of the implant, wherein the tissue is nerve tissue.
10297. The method of item 10112 wherein the agent is delivered from the implant, wherein the agent is released into tissue in the vicinity of the implant after deployment of the implant, wherein the tissue is epithelium tissue.
10298. The method of item 10112 wherein the agent is delivered from the implant, wherein the agent is released in effective concentrations from the implant over a period ranging from the time of deployment of the implant to about 1 year.
10299. The method of item 10112 wherein the agent is delivered from the implant, wherein the agent is released in effective concentrations from the implant over a period ranging from about 1 month to 6 months.
10300. The method of item 10112 wherein the agent is delivered from the implant, wherein the agent is released in effective concentrations from the implant over a period ranging from about 1 - 90 days.
10301. The method of item 10112 wherein the agent is delivered from the implant, wherein the agent is released in effective concentrations from the implant at a constant rate.
10302. The method of item 10112 wherein the agent is delivered from the implant, wherein the agent is released in effective concentrations from the implant at an increasing rate.
10303. The method of item 10112 wherein the agent is delivered from the implant, wherein the agent is released in effective concentrations from the implant at a decreasing rate. 10304. The method of item 10112 wherein the agent is delivered from the implant, wherein the implant comprises about 0.01 Dg to about 10 Dg of the agent.
10305. The method of item 10112 wherein the agent is delivered from the implant, wherein the implant comprises about 10 Dg to about 10 mg of the agent.
10306. The method of item 10112 wherein the agent is delivered from the implant, wherein the implant comprises about 10 mg to about 250 mg of the agent.
10307. The method of item 10112 wherein the agent is delivered from the implant, wherein the implant comprises about 250 mg to about 1000 mg of the agent.
10308. The method of item 10112 wherein the agent is delivered from the implant, wherein the implant comprises about 1000 mg to about 2500 mg of the agent.
10309. The method of item 10112 wherein the agent is delivered from the implant, wherein a surface of the implant comprises less than 0.01 Dg of the agent per mm2 of implant surface to which the agent is applied.
10310. The method of item 10112 wherein the agent is delivered from the implant, wherein a surface of the implant comprises about 0.01 Dg to about 1 Dg of the agent per mm2 of implant surface to which the agent is applied. 10311. The method of item 10112 wherein the agent is delivered from the implant, wherein a surface of the implant comprises about 1 Dg to about 10 Dg of the agent per mm2 of implant surface to which the agent is applied.
10312. The method of item 10112 wherein the agent is delivered from the implant, wherein a surface of the implant comprises about 10 Dg to about 250 Dg of the agent per mm2 of implant surface to which the agent is applied.
10313. The method of item 10112 wherein the agent is delivered from the implant, wherein a surface of the implant comprises about 250 Dg to about 1000 Dg of the agent per mm2 of implant surface to which the agent is applied.
10314. The method of item 10112 wherein the agent is delivered from the implant, wherein a surface of the implant comprises about 1000 Dg to about 2500 Dg of the agent per mm2 of implant surface to which the agent is applied.
10315. The method of item 10112, wherein the implant further comprises a coating, and the coating is a uniform coating.
10316. The method of item 10112, wherein the implant further comprises a coating, and the coating is a non-uniform coating.
10317. The method of item 10112, wherein the implant further comprises a coating, and the coating is a discontinuous coating.
10318. The method of item 10112, wherein the implant further comprises a coating, and the coating is a patterned coating. 10319. The method of item 10112, wherein the implant further comprises a coating, and the coating has a thickness of 100 Dm or less.
10320. The method of item 10112, wherein the implant further comprises a coating, and the coating has a thickness of 10 Gm or less.
10321. The method of item 10112, wherein the implant further comprises a coating, and the coating adheres to the surface of the implant upon deployment of the implant.
10322. The method of item 10112, wherein the implant further comprises a coating, and the coating is stable at room temperature for a period of at least 1 year.
10323. The method of item 10112, wherein the implant further comprises a coating, and the agent is present in the coating in an amount ranging between about 0.0001% to about 1% by weight.
10324. The method of item 10112, wherein the implant further comprises a coating, and the agent is present in the coating in an amount ranging between about 1% to about 10% by weight.
10325. The method of item 10112, wherein the implant further comprises a coating, and the agent is present in the coating in an amount ranging between about 10% to about 25% by weight.
10326. The method of item 10112, wherein the implant further comprises a coating, and the agent is present in the coating in an amount ranging between about 25% to about 70% by weight. 10327. The method of item 10112, wherein the implant further comprises a coating, and the coating comprises a polymer.
10328. The method of item 10112, wherein the implant comprises a first coating having a first composition and a second coating having a second composition.
10329. The method of item 10112, wherein the implant comprises a first coating having a first composition and a second coating having a second composition, wherein the first composition and the second composition are different.
10330. A method as in any one of items 10112-10329, wherein the device is a central venous catheter with a cuff.
10331. A method as in any one of items 10112-10329, wherein the device is a central venous catheter without a cuff.
10332. A method as in any one of items 10112-10329, wherein the device is a central venous catheter with a flange.
10333. A method as in any one of items 10112-10329, wherein the device is a central venous catheter without a flange.
10334. A method as in any one of items 10112-10329, wherein the device is a central venous catheter adapted for providing access to the circulatory system.
10335. A method as in any one of items 10112-10329, wherein the device is a central venous catheter adapted for providing multiple conduits for accessing the circulatory system. 10336. A method as in any one of items 10112-10329, wherein the device is a central venous catheter comprising a means for preventing infection as a result of long term use.
10337. A method as in any one of items 10112-10329, wherein the device is a central venous catheter adaptable for being used with an apparatus that provides a means of controlling the injection or withdrawal of bodily fluids through the central venous catheter.
10338. A method as in any one of items 10112-10329, wherein the device is a parenteral nutrition catheter.
10339. A method as in any one of items 10112-10329, wherein the device is a peripherally inserted central venous catheter.
10340. A method as in any one of items 10112-10329, wherein the device is a flow directed balloon tipped pulmonary artery catheter.
10341. A method as in any one of items 10112-10329, wherein the device is a long term central venous access catheter.
10342. A method for inhibiting scarring comprising placing a device that comprises a ventricular assist device (e.g., an implant) and an anti-scarring agent or a composition comprising an anti-scarring agent into an animal host, wherein the agent inhibits scarring.
10343. The method of item 10342 wherein the agent is an adensosine A2A receptor antagonist. 10344. The method of item 10342 wherein the agent is an AKT inhibitor.
10345. The method of item 10342 wherein the agent is an alpha 2 integrin antagonist, wherein the alpha 2 integrin antagonist is Pharmaprojects No. 5754 (Merck KgaA).
10346. The method of item 10342 wherein the agent is an alpha 4 integrin antagonist.
10347. The method of item 10342 wherein the agent is an alpha 7 nicotinic receptor agonist.
10348. The method of item 10342 wherein the agent is an angiogenesis inhibitor selected from the group consisting of AG-12,958 (Pfizer), ATN-161 (Attenuon LLC), neovastat, an angiogenesis inhibitor from Jerina AG (Germany), NM-3 (Mercian), VGA-1155 (Taisho), FCE-26644 (Pfizer), FCE-26950 (Pfizer), FPMA (MeijF Daries), FR-111142 (Fujisawa), GGTI-298, GM-1306 (Ligand), GPA-1734 (Novartis), NNC-47-0011 (Novo Nordisk), herbamycin (Nippon Kayaku), lenalidomide (Celegene), IP-10 (NIH), ABT-828 (Abbott), KIN-841 (Tokushima University, Japan), SF-1126 (Semafore Pharmaceuticals), laminin technology (NIH), CHIR-258 (Chiron), NVP-AEW541 (Novartis), NVP-AEW541 (Novartis), Vt16907 (Alchemia), OXI-8007 (Oxigene), EG-3306 (Ark Therapeutics), Maspin (Arriva), ABT- 567 (Abbott), PPI-2458 (Praecis Pharmaceuticals), CC-5079, CC-4089 (Celgene), HIF-1 alpha inhibitors (Xenova), S-247 (Pfizer), AP-23573 (Ariad), AZD-9935 (Astra Zeneca), mebendazole (Introgen Therapeutics), MetAP-2 inhibitors (GlaxoSmithKline), AG-615 (Angiogene Pharmaceuticals), Tie-2 antagonists (Hybrigenics), NC-381 , CYC-381, NC-169, NC-219, NC-383, NC-384, NC-407 (Lorus Therapeutics), ATN-224 (Attenuon), ON-01370 (Onconova), Vitronectin antagonists (Amgen), SDX-103 (Salmedix), Vitronectin antagonists (Shire), CHP (Riemser), TEK (Amgen), Anecortave acetate (Alcon), T46.2 (Matrix Therapeutics), HG-2 (Heptagen), TEM antagonists (Genzyme), Oxi-4500 (Oxigene), ATN-161 (Attenuon), WX-293 (Wilex), M-2025 (Metris Therapeutics), Alphastatϊn (BioActa), YH-16 (Yantai Rongchang), BIBF-1120 (Boehringer Ingelheim), BAY-57-9352 (Bayer), AS- 1404 (Cancer Research Technology), SC-77964 (Pfizer), glycomimetics (BioTie Therapies), TIE-2 Inhibitors (Ontogen), DIMI, Octamer (Octamer), ABR-215050 (Active Biotech), ABT-518 (Abbott), KDR inhibitors (Abbott), BSF-466895 (Abbott), SCH-221153 (Schering-Plough), DAC:antiangiogenic (ConjuChem), TFPI (EntreMed), AZD-2171 (Astra-Zenaca), CDC-394 (Celgene), LY290293 (EIi Lilly), IDN-5390 (Indena), Kdr Kinase Inhibitors (Merck), CT-113020, CT-116433, CT-116563, CT-31890, CT-32228) (Cell Therapeutics), A-299620 (Abbott), TWEAK Inhibitor (Amgen), VEGF modulators (Johnson and Johnson), Tum-N53, tumstatin (Genzyme), Thios- 1 , Thios-2 (Thios Pharmaceuticals), MV-6401 (Miravant Medical Technologies), Spisulosine (PharmaMar), CEP-7055 (Cephalon), AUV-201 (Auvation), LM-609 (EIi Lilly), SKF-106615 (AnorMED), Oglufanide disodium (Cytran), BW-114 (Pharminox), Calreticulin (NIH), WX-678 (Wilex), SD-7784 (Pfizer), WX-UK1 (Wilex), SH-268 (Schering AG), 2-Me-PGA (Celgene), S- 137 (Pfizer), ZD-6126 (Angiogene Pharmaceuticals), SG-292 (SignalGen), Benefin (Lane Labs), A6, A36 (Angstrom), SB-2723005 (GlaxoSmithKline), SC-7 (Cell Therapeutics), ZEN-014 (AEterna Zentaris), 2-methoxyestradiol (EntreMed), NK-130119 (Nippon Kayaku), CC-10004 (Celgene), AVE- 8062A (Ajinomoto), Tacedinaline (Pfizer), Actinonin (Tokyo Metropolitan Institute of Medical Science), Lenalidomide (Celgene), VGA-1155, BTO-956 (SRI International), ER-68203-00 (Eisai), CT-6685 (UCB), JKC-362 (Phoenix Pharmaceuticals), DMI-3798 (DMI Biosciences, Angiomate (Ipsen), ZD-6474 (AstraZeneca), CEP-5214 (Cephalon), Canstatin (Genzyme), NM-3 (Mercian), Oridigm (MediQuest Therapeutics), Exherin (Adherex), BLS-0597 (Boston Life Sciences), PTC-299 (PTC Therapeutics), NPl-2358 (Nereus Pharmaceuticals), CGP-79787 (Novartis), AEE-788 (Novartis), CKD-732 (Chong Kun Dang), CP-564959 (OSI Pharmaceuticals), CM-101 (CarboMed), CT-2584, CT3501 (Cell Therapeutics), combretastatin and analogues and derivatives thereof (Oxigene), Rebimastat (Bristol-Meyers Squibb), Dextrin 2-sulfate (ML Laboratories), Cilengitide (Merk KGaA), NSC-706704 (Pharminox), KRN- 951 (Kirin Brewery), Ukrain, NSC-631570 (Nowicky Pharma), Tecogalan sodium (Daiichi Pharmaceutical), Tz-93 (Tsumura), TBC-1635 (Encysive Pharmaceuticals), TAN-1120 (Takeda), Semaxanib (Pfizer), BDI-7800 (Biopharmacopae), SD-186, SD-983 (Bristol-Meyers Squibb), SB-223245 (GlaxoSmithKline), SC-236 (Pfizer), RWJ-590973 (Johnson and Johnson), ILX-1850 (Genzyme), SC-68488, S-836 (Pfizer), CG-55069-11 (CuraGen), Ki-23057 (Kirin Brewery), CCX-700 (Chemoentryx), Pegaptanib octasodium (Giled Sciences), ANGIOCOL (available from Biostratum Inc.), or an analogue or derivative thereof.
10349. The method of item 10342 wherein the agent is an apoptosis antagonist.
10350. The method of item 10342 wherein the agent is an apoptosis activator.
10351. The method of item 10342 wherein the agent is a beta 1 integrin antagonist.
10352. The method of item 10342 wherein the agent is a beta tubulin inhibitor.
10353. The method of item 10342 wherein the agent is a blocker of enzyme production in Hepatitis C. 10354. The method of item 10342 wherein the agent is a Bruton's tyrosine kinase inhibitor.
10355. The method of item 10342 wherein the agent is a calcineurin inhibitor.
10356. The method of item 10342 wherein the agent is a caspase 3 inhibitor.
10357. The method of item 10342 wherein the agent is a CC chemokine receptor antagonist.
10358. The method of item 10342 wherein the agent is a cell cycle inhibitor selected from the group consisting of SNS-595 (Sunesis), synthadotin, KRX-0403, and an analogue or derivative thereof.
10359. The method of item 10342 wherein the agent is a cathepsin B inhibitor.
10360. The method of item 10342 wherein the agent is a cathepsin K inhibitor, wherein the cathepsin K inhibitor is 462795 (GlaxoSmithKline), INPL-022-D6 (Amura Therapeutics), or an analogue or derivative thereof.
10361. The method of item 10342 wherein the agent is a cathepsin L inhibitor.
10362. The method of item 10342 wherein the agent is a CD40 antagonist. 10363. The method of item 10342 wherein the agent is a chemokine receptor agonist.
10364. The method of item 10342 wherein the agent is a chymase inhibitor.
10365. The method of item 10342 wherein the agent is a collagenase antagonist.
10366. The method of item 10342 wherein the agent is a CXCR antagonist.
10367. The method of item 10342 wherein the agent is a cyclin dependent kinase inhibitor selected from the group consisting of a CDK-1 inhibitor, a CDK-2 inhibitor, a CDK- 4 inhibitor, a CDK-6 inhibitor, a CAK1 inhibitor from GPC Biotech and Bristol-Myers Squibb, RGB-286199 (GPC Biotech), an anticancer agent from Astex Technology, a CAK1 inhibitor from GPC Biotech, a CDK inhibitor from Sanofi-Aventis, a CDK1/CDK2 inhibitor from Amgen, a CDK2 inhibitor from SUGEN-2 (Pfizer), a hearing loss therapy agent (Sound Pharmaceuticals), PD-0332991 (Pfizer), RGB-286199 (GPC Biotech), Ro-0505124 (Hoffmann-La Roche), a Ser/Thr kinase inhibitor from Lilly (EIi Lilly), CVT-2584 (CAS No. 199986-75- 9) (CV Therapeutics), and an analogue or derivative thereof.
10368. The method of item 10342 wherein the agent is a cyclooxygenase 1 inhibitor.
10369. The method of item 10342 wherein the agent is a DHFR inhibitor. 10370. The method of item 10342 wherein the agent is a dual integrin inhibitor.
10371. The method of item 10342 wherein the agent is an elastase inhibitor.
10372. The method of item 10342 wherein the agent is an elongation factor-1 alpha inhibitor.
10373. The method of item 10342 wherein the agent is an endothelial growth factor antagonist.
10374. The method of item 10342 wherein the agent is an endothelial growth factor receptor kinase inhibitor selected from the group consisting of sorafenib tosylate (Bayer), AAL-993 (Novartis), ABP-309 (Novartis), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), E- 7080 (Eisai), EG-3306 (Ark Therapeutics), EXEL-2880 (Exelixis), GW- 654652 (GlaxoSmithKline), a KDR inhibitor from LG Life Sciences, CT-6685 and CT-6729 (UCB), KRN-633 and KRN-951 (Kirin Brewery), OSI-930 (OSI Pharmaceuticals), SP-5.2 (Supratek Pharma), SU-11657 (Pfizer), a Tie-2 antagonist (Hybrigenics), a VEGFR-2 kinase inhibitor (Bristol-Myers Squibb), XL-647 (Exelixis), a KDR inhibitor from Abbott Laboratories, sorafenib tosylate, and an analogue or derivative thereof.
10375. The method of item 10342 wherein the agent is an endotoxin antagonist.
10376. The method of item 10342 wherein the agent is an epothilone and tubulin binder. 10377. The method of item 10342 wherein the agent is an estrogen receptor antagonist.
10378. The method of item 10342 wherein the agent is an FGF inhibitor.
10379. The method of item 10342 wherein the agent is a famexyl transferase inhibitor.
10380. The method of item 10342 wherein the agent is farnesyltransferase inhibitor selected from the group of A-197574 (Abbott), a famesyltransferase inhibitor from Servier, FPTIIl (Strathclyde Institute for Drug R), LB-42908 (LG Life Sciences), Pharmaprojects No. 5063 (Genzyme), Pharmaprojects No. 5597 (Ipsen), Yissum Project No. B-1055 (Yissum), and an analogue or derivative thereof.
10381. The method of item 10342 wherein the agent is an FLT-3 kinase inhibitor.
10382. The method of item 10342 wherein the agent is an FGF receptor kinase inhibitor.
10383. The method of item 10342 wherein the agent is a fibrinogen antagonist selected from the group consisting of AUV-201 (Auvation), MG-13926 (Sanofi-Aventis), plasminogen activator (CAS No. 105913-11-9) (from Sanofi-Aventis or UCB), plasminogen activator-2 (tPA- 2) (Sanofi-Aventis), pro-urokinase (CAS No. 82657-92-9) (Sanofi-Aventis), and an analogue or derivative thereof.
10384. The method of item 10342 wherein the agent is a heat shock protein 90 antagonist selected from the group consisting of SRN- 005 (Sirenade), geldanamycin, NSC-33050 (17-allylaminogeldanamycin; 17- AAG), 17-dimethylaminoethylamino-17-demethoxy-geldanamycin (17- DMAG), rifabutin (rifamycin XIV, 1',4-didehydro-1-deoxy-1 ,4-dihydro-5'-(2- methylpropyl)-1-oxo-), and an analogue or derivative thereof.
10385. The method of item 10342 wherein the agent is a histone deacetylase inhibitor.
10386. The method of item 10342 wherein the agent is an HMGCoA reductase inhibitor selected from the group consisting of an atherosclerosis therapeutic from Lipid Sciences, AT1-16000 (ARYx Therapeutics), KS-01-019 (Kos Pharmaceuticals), Pharmaprojects No. 2197 (Sanofi-Aventi), RP 61969 (Sanofi-Aventis), and an analogue or derivative thereof.
10387. The method of item 10342 wherein the agent is an ICAM inhibitor.
10388. The method of item 10342 wherein the agent is an IL, ICE and IRAK antagonist, wherein the antagonist is a CJ-14877, CP- 424174 (Pfizer), NF-61 (Negma-Lerads), and an analogue or derivative thereof.
10389. The method of item 10342 wherein the agent is an IL-2 inhibitor.
10390. The method of item 10342 wherein the agent is an immunosuppressant selected from the group consisting of teriflunomide (Sanofi Aventis), chlorsulfaquinoxalone (NSC-339004), chlorsulfaquinoxalone sulfate, CS-712 (Sankyo), ismomultin alfa (CAS No. 457913-93-8) (Akzo Nobel), antiallergics from GenPat77, anti- inflammatories or AT-005 (Androclus Therapeutics), autoimmune disease therapeutics from EpiVax, BN-007 (Bone), budesonide (CAS No. 51333-22- 3) (MAP Pharmaceuticals), CO-14 (Genzyme), edratide (CAS No. 433922- 67-9) (Teva), EP-314 (Enanta), eprovafen (CAS No. 101335-99-3) (Sanofi- Aventis), HWA-131 (CAS No. 118788-41-3) (Sanofi-Aventis), immunomodulators from MerLion Pharmaceuticals, immunosuppressives from Alchemia, IPL-12 (Inflazyme), MDL-9563 (CAS No. 27086-86-8) (Sanofi-Aventis), Pharmaprojects No. 2330 (Sanofi-Aventis), Pharmaprojects No. 6426 (Abgenix), PXS-25 (Pharmaxis), rosmarinic acid (CAS No. 20283-92-5) (Sanofi-Aventis), RP 42927 or RP 54745 (CAS No. 135330-08-4) (Sanofi-Aventis), SGN-35 (Seattle Genetics), ST-1959 (Sigma-Tau), type I diabetes therapy from SYNX Pharma, UNIL-88 (Debiopharm), VP-025 (Vasogen), VR-694 (Vectura), PRTX-001 (Protalex), and an analogue or derivative thereof.
10391. The method of item 10342 wherein the agent is an IMPDH (inosine monophosphate).
10392. The method of item 10342 wherein the agent is an integrin antagonist.
10393. The method of item 10342 wherein the agent is an interleukin antagonist.
10394. The method of item 10342 wherein the agent is an inhibitor of type III receptor tyrosine kinase.
10395. The method of item 10342 wherein the agent is an irreversible inhibitor of enzyme methionine aminopeptidase type 2. 10396. The method of item 10342 wherein the agent is an isozyme selective delta protein kinase C inhibitor.
10397. The method of item 10342 wherein the agent a JAK3 enzyme inhibitor.
10398. The method of item 10342 wherein the agent is a
JNK inhibitor.
10399. The method of item 10342 wherein the agent is a kinase inhibitor.
10400. The method of item 10342 wherein the agent is kinesin antagonist.
10401. The method of item 10342 wherein the agent is a kinesin antagonist.
10402. The method of item 10342 wherein the agent is a leukotriene inhibitor and antagonist selected from the group consisting of ambicromil (CAS No. 58805-38-2) (Sanofi-Aventis), amelubant (CAS No. 346735-24-8) (Boehringer Ingelheim), DW-1141 (Dong Wha), ebselen (Daiichi Pharmaceutical), ibudilast (Kyorin), leucotriene inhibitors from Sanofi-Aventis, lymphotoxin-beta receptor (LT-D) from Biogen Idee, Pharmaprojects No. 1535 and 2728 (CAS No. 119340-33-9) (Sanofi- Aventis), R-112 (Rigel), Rev-5367 (CAS No. 92532-05-3) (Sanofi-Aventis), RG-14893 (CAS No. 141835-49-6) (Sanofi-Aventis), RG-5901-A (CAS No. 101910-24-1), 92532-23-5, RP 66153 (CAS No. 142422-79-5), RP 66364 (CAS No.186916-92-5), RP 69698 (CAS No. 141748-00-7) (Sanofi-Aventis), SC-411930 (Pfizer), SC-41930 (CAS No. 120072-59-5) (Pfizer), SC-50605 (CAS No. 138828-39-4) (Pfizer), SC-51146 (CAS No. 141059-52-1), SC- 53228 (CAS No. 153633-01-3) (Pfizer), spaglumic acid (ZY-15106) (CAS No.3106-85-2), 80619-64-3 (Novartis), tipredane (CAS No. 85197-77-9) (Bristol-Myers Squibb), U-75302 (CAS No. 119477-85-9) (Pfizer), and analogue or derivative thereof.
10403. The method of item 10342 wherein the agent is an MAP kinase inhibitor.
10404. The method of item 10342 wherein the agent is a matrix metalloproteinase inhibitor.
10405. The method of item 10342 wherein the agent is an MCP-CCR2 inhibitor.
10406. The method of item 10342 wherein the agent is an mTOR inhibitor.
10407. The method of item 10342 wherein the agent is an mTOR kinase inhibitor.
10408. The method of item 10342 wherein the agent is a microtubule inhibitor selected from the group consisting of antibody- maytansinoid conjugates from Biogen Idee, colchicines (MantiCore Pharmaceuticals), anticancer immunoconjugates from Johnson & Johnson, DIME from Octamer, gni-1f (GNI), huC242-DM4, huMy9-6-DM1 (ImmunoGen), IDN-5404 (Indena), IMO-098, IMOderm (Imotep), mebendazole (Introgen Therapeutics), microtubule poisons from Cambridge Enterprise, paclitaxel such as LOTAX from Aphios (CAS No. 33069-62-4), Genexol-PM from Samyang, Pharmaprojects No. 6383 (Tapestry Pharmaceuticals), RPR-112378 (Sanofi-Aventis), SGN-75 (Seattle Genetics), SPL-7435 (Starpharma), SSR-250411 (Sanofi-Aventis), trastuzumab-DM1 (Genentech), vinorelbine, and an analogue or derivative thereof.
10409. The method of item 10342 wherein the agent is an MIF inhibitor.
10410. The method of item 10342 wherein the agent is an MMP inhibitor.
10411. The method of item 10342 wherein the agent is a neurokinin (NK) antagonist selected from the group consisting of anthrotainin (CAS No. 148084-40-6) (Sanofi-Aventis), an IBS thereapeutic from ArQuIe, MDL-105212A (CAS No. 167261-60-1) (Ssanofi-Aventis), Pharmaprojects No. 2744, 3258 (CAS No. 139167-47-8) 4006, 4201, or 5986 (Sanofi-Aventis), RP 67580 (CAS No. 135911-02-3), SR-144190 (CAS No. 201152-86-5), SSR-240600, SSR-241586 (Sanofi-Aventis), TKA-457 (Novartis), vestipitant mesylate (CAS No. 334476-64-1) (GlaxoSmithKline), Win-64821 (Sanofi-Aventis), PRX-96026 (Predix Pharmaceuticals), and an analogue or derivative thereof.
10412. The method of item 10342 wherein the agent is an NF kappa B inhibitor selected from the group consisting of AVE-0545 or AVE-0547 (Sanofi-Aventis), bortezomib (CAS No. 179324-69-7) (Millennium Pharmaceuticals), dexanabinol (CAS No. 112924-45-5) (Pharmos), dexlipotam (Viatris), Pharmaprojects No. 6283 (INDRA) (OXiGENE), IPL- 576092 (CAS No. 137571-30-3) (Inflazyme), NFKB decoy (Corgentech), NFKB decoy oligo (AnGes MG), NFKB's from Ariad, osteoporosis treatments or S5 (F005) from Fulcrum Pharmaceuticals, P61 (Phytopharm), R-flurbiprofen (CAS No. 5104-49-4) (Encore Pharmaceuticals), and an analogue or derivative thereof. 10413. The method of item 10342 wherein the agent is a nitric oxide agonist.
10414. The method of item 10342 wherein the agent is an ornithine decarboxylase inhibitor.
10415. The method of item 10342 wherein the agent is a p38 MAP kinase inhibitor selected from the group consisting of AZD-6703 (AstraZeneca), JX-401 (Jexys Pharmaceuticals), BMS-2 (Bristol-Myers Squibb), a p38 MAP kinase inhibitor from Novartis, a p38-alpha MAP kinase inhibitor from Amphora, Pharmaprojects No. 5704 (Pharmacopeia), RPR- 200765A (Sanofi-Aventis), SD-282 (Johnson & Johnson), TAK-715 (Takeda), and an analogue or derivative thereof.
10416. The method of item 10342 wherein the agent is a palmitoyl-protein thioesterase inhibitor.
10417. The method of item 10342 wherein the agent is a PDGF receptor kinase inhibitor selected from the group consisting of AAL- 993, AMN-107, or ABP-309 (Novartis), AMG-706 (Amgen), BAY-57-9352 (Bayer), CDP-860 (UCB), E-7080 (Eisai), imatinib (CAS No. 152459-95-5) (Novartis), OSI-930 (OSI Pharmaceuticals), RPR-127963E (Sanofi-Aventis), RWJ-540973 (Johnson & Johnson), sorafenib tosylate (Bayer), SU-11657 (Pfizer), tandutinib (CAS No. 387867-13-2) (Millennium Pharmaceuticals), vatalanib (Novartis), ZK-CDK (Schering AG), and an analogue or derivative thereof.
10418. The method of item 10342 wherein the agent is (-)-halofenate (Metabolex), AMG-131 (Amgen), antidiabetics from Japan Tobacco, AZD-4619, AZD-8450, AZD-8677 (AstraZeneca), DRF-10945, balaglitazone (Dr Reddy's), CS-00088, CS-00098 (Chipscreen Biosciences), E-3030 (Eisai), etalocib (CAS No. 161172-51-6) (EIi Lilly), GSK-641597 (Ligand), GSK-677954 (GlaxoSmithKline), GW-409544 (Ligand), GW- 590735 (GlaxoSmithKline), K-111 (Hoffmann-La Roche), LY-518674 (EIi Lilly), LY-674 (Ligand), LY-929 (Ligand), MC-3001 , MC-3002 (MaxoCore Pharmaceuticals), metformin HCI + pioglitazone (CAS No. 1115-70-4 and 112529-15-4), ACTOPLUS MET from Andrx), muraglitazar (CAS No. 331741-94-7) (Bristol-Myers Squibb), naveglitazar (Ligand), oleoylethanolamide (Kadmus Pharmaceuticals), ONO-5129, pioglitazone hydrochloride (CAS No. 111025-46-8 and 112529-15-4) (Takeda), PLX-204 (Plexxikon), PPAR agonists from Genfit, PPAR delta agonists from EIi Lilly, PPAR-alpha agonists from CrystalGenomics, PPAR-gamma modulators and PPAR-β modulators from CareX, rosiglitazone maleate (CAS No. 122324- 73-4 or 155141-29-0) (GlaxoSmithKline), rosiglitazone maleate/glimepir (CAS No. 155141-29-0 and 93479-97-1), AVANDARYL, rosiglitazone maleate/metformin extend (CAS No. 155141-29-0 and 657-24-9), AVANDAMET, rosiglitazone maleate+metformin, AVANDAMET (GlaxoSmithKline), tesaglitazar (AstraZeneca), and an analogue or derivative thereof.
10419. The method of item 10342 wherein the agent is a phosphatase inhibitor.
10420. The method of item 10342 wherein the agent is a phosphodiesterase (PDE) inhibitor selected from the group consisting of avanafil (Tanabe Seiyaku), dasantafil (CAS No. 569351-91-3) (Schering- Plough), A-906119 (CAS No. 134072-58-5), DL-850 (Sanofi-Aventis), GRC- 3015, GRC-3566, GRC-3886 (Glenmark), HWA-153 (CAS No. 56395-66-5) (Sanofi-Aventis), hydroxypumafentrine (Altana), IBFB-130011 , IBFB-14-016, IBFB-140301 , IBFB-150007, IBFB-211913 (IBFB Pharma), L-826141 (Merck & Co), medorinone (CAS No. 88296-61-1) (Sanofi-Aventis), MEM- 1917 (Memory Pharmaceuticals), ND-1251 (Neuro3d), PDE inhibitors from ICOS, PDE IV inhibitors from Memory Pharmaceuticals and CrystalGenomics, Pharmaprojects No. 2742 and 6141 (Sanofi-Aventis), QAD-171 (Novartis), RHC-2963 (CAS No. 76993-12-9 and 76993-14-1), RPR-117658, RPR-122818 derivatives, SR-24870 , and RPR-132294 (Sanofi-Aventis), SK-350 (ln2Gen), stroke therapy agents from deCODE Genetics, TAS-203 (Taiho), tofimilast (CAS No. 185954-27-2) (Pfizer), UK- 371800 (Pfizer), WIN-65579 (CAS No. 158020-82-7) (Sanofi-Aventis), IBFB- 130020 (IBFB Pharma), OPC-6535 (CAS No. 145739-56-6) (Otsuka), a phosphodiesterase III inhibitor, enoximone, a phosphodiesterase IV inhibitor, fosfosal, Atopik (Barrier Therapeutics), triflusal, a phosphodiesterase V inhibitor, and an analogue or derivative thereof.
10421. The method of item 10342 wherein the agent is a PKC inhibitor.
10422. The method of item 10342 wherein the agent is a platelet activating factor antagonist.
10423. The method of item 10342 wherein the agent is a platelet-derived growth factor receptor kinase inhibitor.
10424. The method of item 10342 wherein the agent is a prolyl hydroxylase inhibitor.
10425. The method of item 10342 wherein the agent is a polymorphonuclear neutrophil inhibitor.
10426. The method of item 10342 wherein the agent is a protein kinase B inhibitor. 10427. The method of item 10342 wherein the agent is a protein kinase C stimulant.
10428. The method of item 10342 wherein the agent is a purine nucleoside analogue.
10429. The method of item 10342 wherein the agent is a purinoreceptor P2X antagonist.
10430. The method of item 10342 wherein the agent is a Raf kinase inhibitor.
10431. The method of item 10342 wherein the agent is a reversible inhibitor of ErbB1 and ErbB2.
10432. The method of item 10342 wherein the agent is a ribonucleoside triphosphate reductase inhibitor.
10433. The method of item 10342 wherein the agent is an SDF-1 antagonist.
10434. The method of item 10342 wherein the agent is a sheddase inhibitor.
10435. The method of item 10342 wherein the agent is an SRC inhibitor.
10436. The method of item 10342 wherein the agent is a stromelysin inhibitor. 10437. The method of item 10342 wherein the agent is an Syk kinase inhibitor.
10438. The method of item 10342 wherein the agent is a telomerase inhibitor.
10439. The method of item 10342 wherein the agent is a TGF beta inhibitor selected from the group consisting of pirfenidone (CAS No. 53179-13-8) (MARNAC), tranilast (CAS No. 53902-12-8) (Kissei), IN- 1130 (ln2Gen), mannose-6-phosphate (BTG), TGF-β antagonists from Inflazyme (Pharmaprojects No. 6075), TGF-β antagonists from Sydney, non-industrial source), TGF-β I receptor kinase inhibitors from EIi Lilly, TGF- β receptor inhibitors from Johnson & Johnson, and an analogue or derivative thereof.
10440. The method of item 10342 wherein the agent is a TNFα antagonist or TACE inhibitor selected from the group consisting of adalimumab (CAS No. 331731-18-1) (Cambridge Antibody Technology), AGIX-4207 (AtheroGenics), AGT-1 (Advanced Biotherapy), an antiinflammatory from Borean Pharma, Cellzome, or Paradigm Therapeutics, anti-inflammatory vaccine (TNF-alpha kinoid) from Neovacs, humanized anti-TNF antibody or an anti-TNF MAb (CB0006) Celltech (UCB), apratastat (CAS No. 287445-51-0) (Wyeth), BMS-561392 (Bristol-Myers Squibb), BN- 006 (Bone), certolizumab pegol (CAS No. 428863-50-7 or CH-138 (UCB), cilomilast (CAS No. 153259-65-5) (GlaxoSmithKline), CR-1 (Nuada Pharmaceuticals), CRx-119 (CombinatoRx), D-5410 (UCB), dacopafant (CAS No. 125372-33-0) (Sanofi-Aventis), dersalazine (CAS No. 188913-57- 7/188913-58-8) (Uriach), etanercept (CAS No. 185243-69-0) (Amgen), ethyl pyruvate (Critical (Critical Therapeutics), golimumab (CAS No. 476181-74-5) (Johnson & Johnson), hormono-immunotherapy from Ipsen, CDP571 (e.g., Humicade from UCB), IC-485 (ICOS), infliximab (CAS No. 170277-31-3) (Johnson & Johnson), IP-751 (Manhattan Pharmaceuticals), ISIS-104838 (CAS No. 250755-32-9) (ISIS Pharmaceuticals), lenalidomide (CAS No. 191732-72-6) (Celgene), lentinan (CAS No. 37339-90-5) (Ajinomoto), MDL- 201112 (CAS No. 142130-73-2) (Sanofi-Aventis), medroxyprogesterone (CAS No. 520-85-4) (InKine Pharmaceutical), N-acetylcysteine (CAS No. 616-91-1) (Zambon), NBE-P2 (DIREVO Biotech), nerelimomab (CAS No. 162774-06-3) (Chiron), OM-294DP (OM PHARMA), onercept (CAS No. 199685-57-9) (Yeda), PASSTNF-alpha (Verigen), pentoxifylline or oxypentifylline (Sanofi-Aventis), Pharmaprojects No. 4091 , 4241, 4295, or 4488 (Sanofi-Aventis), Pharmaprojects No. 5480 (Amgen), Pharmaprojects No. 6749 (Cengent), pirfenidone (CAS No. 53179-13-8) (MARNAC), RPR- 132294 (Sanofi-Aventis), S5 (F002) (Fulcrum Pharmaceuticals), simvastatin (CAS No. 79902-63-9) (Merck & Co), STA-6292 (Synta Pharmaceuticals), tacrolimus (CAS No. 104987-11-3) (Fujisawa LifeCycle Pharma), talactoferrin alfa (CAS No. 308240-58-6) (Agennix), thalidomide (CAS No. 50-35-1) (Celgene), TNF antagonists form ProStrakan, and Synergen, TNF inhibitors (Amgen), TNF-alpha antagonists from Dynavax Technologies and Jerina AG (Germany), TNF-alpha inhibitors from IBFB Pharma and Xencor (Xencor), torbafylline (CAS No. 105102-21-4) (Sanofi-Aventis), UR-1505 (Uriach), VT-346 (Viron Therapeutics), YSIL6 (Y's Therapeutics), YSTH2 (Y's Therapeutics), NPI-1302a-3 (Nereus Pharmaceuticals, a TNF antagonist from Jerina AG (Germany), dersalazine, and an analogue or derivative thereof.
10441. The method of item 10342 wherein the agent is a Toll receptor inhibitor.
10442. The method of item 10342 wherein the agent is a tubulin antagonist. 10443. The method of item 10342 wherein the agent is a tyrosine kinase inhibitor selected from the group consisting of SU-011248, SUTENT from Pfizer Inc. (New York, NY), BMS-354825, PN-355 (Paracelsian Pharmaceuticals), AGN-199659 (Allergan), AAL-993 or ABP- 309 (Novartis), adaphostin (NIH), AEE-788 (Novartis), AG-013736 (OSI Pharmaceuticals), AG-13736 (Pfizer), ALT-110 (Alteris Therapeutics), AMG- 706 (Amgen), anticancer MAbs from Xencor, anti-EGFrvlll MAbs from Abgenix, anti-HER2 MAb from Abiogen, AZD-2171 or AZD-9935 (AstraZeneca), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), CEP-5214 (Cephalon), CEP-7055 (Cephalon), cetuximab (ImClone Systems), CHIR-200131 and CHIR-258 (Chiron), CP-547632 (OSI Pharmaceuticals), CP-724714 (Pfizer), CT-301 (Creabilis Therapeutics), D- 69491 (Baxter International), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EGFR/ErbB2 inhibitors from Array BioPharma, erlotinib (CAS No. 183319- 69-9) (OSI Pharmaceuticals), EXEL-2880 (Exelixis), FK-778 (Sanofi- Aventis) , gefitinib (CAS No. 184475-35-2) (AstraZeneca), GW-2286 or GW- 654652 (GlaxoSmithKline), her2/neu antigen from AlphaVax, HER-2/neu inhibitor from Generex, Herzyme (Medipad) (Sirna Therapeutics), HKI-272 (Wyeth), HuMax-EGFr (Genmab), idronoxil (CAS No. 81267-65-4) (Novogen), IGF-1 inhibitors from Ontogen, IMC-11F8 (ImClone Systems), kahalalide F (CAS No. 149204-42-2) (PharmaMar), KDR inhibitor from LG Life Sciences, KDR inhibitors from Abbott Laboratories, KDR kinase inhibitors (UCB), Kdr kinase inhibitors from Merck & Co, KRN-633 and KRN- 951 (Kirin Brewery), KSB-102 (Xenova), lapatinib ditosylate (CAS No. 388082-78-8) (GlaxoSmithKline), matuzumab (Merck KGaA), MDX-214 (Medarex), ME-103 (Pharmexa), MED-A300 (Gerolymatos), MNAC-13 (Lay Line Genomics), nimotuzumab (Center of Molecular Immunology), NSC- 330507 or NSC-707545 (NIH), NV-50 (Novogen), OSI-930 (OSI Pharmaceuticals), panitumumab (Abgenix), pelitinib (CAS No. 287933-82-7) (Wyeth), pertuzumab (CAS No. 380610-27-5) (Genentech), Pharmaprojects No. 3985 (Sanofi-Aventis), prostate cancer therapeutics from Sequenom (SQPC35, SQPC36, SQPC90), removab and remoxab (Trion Pharma), RG- 13022 (CAS No. 136831-48-6), RG-13291 (CAS No. 138989-50-1), or RG- 14620 (CAS No. 136831-49-7) (Sanofi-Aventis), RM-6427 (Romark), RNAi breast cancer therapy from Benitec, RP 53801 (CAS No. 125882-88-4) (Sanofi-Aventis), sorafenib tosylate (Bayer), SU-11657 (Pfizer), Tie-2 antagonists from Semaia (Hybrigenics), Tie-2 inhibitors from Ontogen, trastuzumab (CAS No. 180288-69-1) (Genentech), tyrosine kinase inhibitors from Sanofi-Aventis, U3-1287, U3-1565, U3-1784, U3-1800 (U3 Pharma), vatalanib (Novartis), VEGFR-2 kinase inhibitor from Bristol-Myers Squibb, XL-647 (Exelixis), ZD-6474 (AstraZeneca), ZK-CDK (Schering AG), an EGFR tyrosine kinase inhibitor, EKB-569 (Wyeth), and an analogue or derivative thereof.
10444. The method of item 10342 wherein the agent is a VEGF inhibitor.
10445. The method of item 10342 wherein the agent is a vitamin D receptor agonist.
10446. The method of item 10342 wherein the agent is ZD-6474 (an angiogenesis inhibitor).
10447. The method of item 10342 wherein the agent is AP-23573 (an mTOR inhibitor).
10448. The method of item 10342 wherein the agent is synthadotin (a tubulin antagonist).
10449. The method of item 10342 wherein the agent is S-0885 (a collagenase inhibitor). 10450. The method of item 10342 wherein the agent is aplidine (an elongation factor-1 alpha inhibitor).
10451. The method of item 10342 wherein the agent is ixabepilone (an epithilone).
10452. The method of item 10342 wherein the agent is IDN-5390 (an angiogenesis inhibitor).
10453. The method of item 10342 wherein the agent is SB-2723005 (an angiogenesis inhibitor).
10454. The method of item 10342 wherein the agent is ABT-518 (an angiogenesis inhibitor).
10455. The method of item 10342 wherein the agent is combretastatin (an angiogenesis inhibitor).
10456. The method of item 10342 wherein the agent is anecortave acetate (an angiogenesis inhibitor).
10457. The method of item 10342 wherein the agent is SB-715992 (a kinesin antagonist).
10458. The method of item 10342 wherein the agent is temsirolimus (an mTOR inhibitor).
10459. The method of item 10342 wherein the agent is adalimumab (a TNFα antagonist). 10460. The method of item 10342, wherein the composition comprises a polymer.
10461. The method of item 10342, wherein the composition comprises a polymer, and the polymer is, or comprises, a copolymer.
10462. The method of item 10342, wherein the composition comprises a polymer, and the polymer is, or comprises, a block copolymer.
10463. The method of item 10342, wherein the composition comprises a polymer, and the polymer is, or comprises, a random copolymer.
10464. The method of item 10342, wherein the composition comprises a polymer, and the polymer is, or comprises, a biodegradable polymer.
10465. The method of item 10342, wherein the composition comprises a polymer, and the polymer is, or comprises, a nonbiodegradable polymer.
10466. The method of item 10342, wherein the composition comprises a polymer, and the polymer is, or comprises, a hydrophilic polymer.
10467. The method of item 10342, wherein the composition comprises a polymer, and the polymer is, or comprises, a hydrophobic polymer. 10468. The method of item 10342, wherein the composition comprises a polymer, and the polymer is, or comprises, a polymer having hydrophilic domains.
10469. The method of item 10342, wherein the composition comprises a polymer, and the polymer is, or comprises, a polymer having hydrophobic domains.
10470. The method of item 10342, wherein the composition comprises a polymer, and the polymer is, or comprises, a non- conductive polymer.
10471. The method of item 10342, wherein the composition comprises a polymer, and the polymer is, or comprises, an elastomer.
10472. The method of item 10342, wherein the composition comprises a polymer, and the polymer is, or comprises, a hydrogel.
10473. The method of item 10342, wherein the composition comprises a polymer, and the polymer is, or comprises, a silicone polymer.
10474. The method of item 10342, wherein the composition comprises a polymer, and the polymer is, or comprises, a hydrocarbon polymer.
10475. The method of item 10342, wherein the composition comprises a polymer, and the polymer is, or comprises, a styrene-derived polymer. 10476. The method of item 10342, wherein the composition comprises a polymer, and the polymer is, or comprises, a butadiene-derived polymer.
10477. The method of item 10342, wherein the composition comprises a polymer, and the polymer is, or comprises, a macromer.
10478. The method of item 10342, wherein the composition comprises a polymer, and the polymer is, or comprises, a poly(ethylene glycol) polymer.
10479. The method of item 10342, wherein the composition comprises a polymer, and the polymer is, or comprises, an amorphous polymer.
10480. The method of item 10342, wherein the composition further comprises a second pharmaceutically active agent.
10481. The method of item 10342, wherein the composition further comprises an anti-inflammatory agent.
10482. The method of item 10342, wherein the composition further comprises an agent that inhibits infection.
10483. The method of item 10342, wherein the composition further comprises an anthracycline.
10484. The method of item 10342, wherein the composition further comprises doxorubicin. 10485. The method of item 10342 wherein the composition further comprises mitoxantrone.
10486. The method of item 10342 wherein the composition further comprises a fluoropyrimidine.
10487. The method of item 10342, wherein the composition further comprises 5-fluorouracil (5-FU).
10488. The method of item 10342, wherein the composition further comprises a folic acid antagonist.
10489. The method of item 10342, wherein the composition further comprises methotrexate.
10490. The method of item 10342, wherein the composition further comprises a podophylotoxin.
10491. The method of item 10342, wherein the composition further comprises etoposide.
10492. The method of item 10342, wherein the composition further comprises camptothecin.
10493. The method of item 10342, wherein the composition further comprises a hydroxyurea.
10494. The method of item 10342, wherein the composition further comprises a platinum complex. 10495. The method of item 10342, wherein the composition further comprises cisplatin.
10496. The method of item 10342 wherein the composition further comprises an anti-thrombotic agent.
10497. The method of item 10342, wherein the composition further comprises a visualization agent.
10498. The method of item 10342, wherein the composition further comprises a visualization agent, and the visualization agent is a radiopaque material, wherein the radiopaque material comprises a metal, a halogenated compound, or a barium containing compound.
10499. The method of item 10342, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, barium, tantalum, or technetium.
10500. The method of item 10342, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, an MRI responsive material.
10501. The method of item 10342, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, a gadolinium chelate.
10502. The method of item 10342, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, iron, magnesium, manganese, copper, or chromium. 10503. The method of item 10342, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, iron oxide compound.
10504. The method of item 10342, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, a dye, pigment, or colorant.
10505. The method of item 10342 wherein the agent is released in effective concentrations from the composition comprising the agent by diffusion over a period ranging from the time of administration to about 90 days.
10506. The method of item 10342 wherein the agent is released in effective concentrations from the composition comprising the agent by erosion of the composition over a period ranging from the time of administration to about 90 days.
10507. The method of item 10342 wherein the composition further comprises an inflammatory cytokine.
10508. The method of item 10342 wherein the composition further comprises an agent that stimulates cell proliferation.
10509. The method of item 10342 wherein the composition further comprises a polymeric carrier.
10510. The method of item 10342 wherein the composition is in the form of a gel, paste, or spray. 10511. The method of item 10342 wherein the implant is partially constructed with the agent or the composition.
10512. The method of item 10342 wherein the implant is fully constructed with the agent or the composition.
10513. The method of item 10342 wherein the implant is impregnated with the agent or the composition.
10514. The method of item 10342, wherein the agent or the composition forms a coating, and the coating directly contacts the implant.
10515. The method of item 10342, wherein the agent or the composition forms a coating, and the coating indirectly contacts the implant.
10516. The method of item 10342 wherein the agent or the composition forms a coating, and the coating partially covers the implant.
10517. The method of item 10342, wherein the agent or the composition forms a coating, and the coating completely covers the implant.
10518. The method of item 10342 wherein the agent or the composition is located within pores or holes of the implant.
10519. The method of item 10342 wherein the agent or the composition is located within a channel, lumen, or divet of the implant. 10520. The method of item 10342 wherein the implant further comprising an echogenic material.
10521. The method of item 10342 wherein the implant further comprises an echogenic material, wherein the echogenic material is in the form of a coating.
10522. The method of item 10342 wherein the implant is sterile.
10523. The method of item 10342 wherein the agent is delivered from the implant, wherein the agent is released into tissue in the vicinity of the implant after deployment of the implant.
10524. The method of item 10342 wherein the agent is delivered from the implant, wherein the agent is released into tissue in the vicinity of the implant after deployment of the implant, wherein the tissue is connective tissue.
10525. The method of item 10342 wherein the agent is delivered from the implant, wherein the agent is released into tissue in the vicinity of the implant after deployment of the implant, wherein the tissue is muscle tissue.
10526. The method of item 10342 wherein the agent is delivered from the implant, wherein the agent is released into tissue in the vicinity of the implant after deployment of the implant, wherein the tissue is nerve tissue.
10527. The method of item 10342 wherein the agent is delivered from the implant, wherein the agent is released into tissue in the vicinity of the implant after deployment of the implant, wherein the tissue is epithelium tissue.
10528. The method of item 10342 wherein the agent is delivered from the implant, wherein the agent is released in effective concentrations from the implant over a period ranging from the time of deployment of the implant to about 1 year.
10529. The method of item 10342 wherein the agent is delivered from the implant, wherein the agent is released in effective concentrations from the implant over a period ranging from about 1 month to 6 months.
10530. The method of item 10342 wherein the agent is delivered from the implant, wherein the agent is released in effective concentrations from the implant over a period ranging from about 1 - 90 days.
10531. The method of item 10342 wherein the agent is delivered from the implant, wherein the agent is released in effective concentrations from the implant at a constant rate.
10532. The method of item 10342 wherein the agent is delivered from the implant, wherein the agent is released in effective concentrations from the implant at an increasing rate.
10533. The method of item 10342 wherein the agent is delivered from the implant, wherein the agent is released in effective concentrations from the implant at a decreasing rate. 10534. The method of item 10342 wherein the agent is delivered from the implant, wherein the implant comprises about 0.01 Dg to about 10 Dg of the agent.
10535. The method of item 10342 wherein the agent is delivered from the implant, wherein the implant comprises about 10 Dg to about 10 mg of the agent.
10536. The method of item 10342 wherein the agent is delivered from the implant, wherein the implant comprises about 10 mg to about 250 mg of the agent.
10537. The method of item 10342 wherein the agent is delivered from the implant, wherein the implant comprises about 250 mg to about 1000 mg of the agent.
10538. The method of item 10342 wherein the agent is delivered from the implant, wherein the implant comprises about 1000 mg to about 2500 mg of the agent.
10539. The method of item 10342 wherein the agent is delivered from the implant, wherein a surface of the implant comprises less than 0.01 Dg of the agent per mm2 of implant surface to which the agent is applied.
10540. The method of item 10342 wherein the agent is delivered from the implant, wherein a surface of the implant comprises about 0.01 Dg to about 1 Dg of the agent per mm2 of implant surface to which the agent is applied. 10541. The method of item 10342 wherein the agent is delivered from the implant, wherein a surface of the implant comprises about 1 Dg to about 10 Dg of the agent per mm2 of implant surface to which the agent is applied.
10542. The method of item 10342 wherein the agent is delivered from the implant, wherein a surface of the implant comprises about 10 Dg to about 250 Dg of the agent per mm2 of implant surface to which the agent is applied.
10543. The method of item 10342 wherein the agent is delivered from the implant, wherein a surface of the implant comprises about 250 Dg to about 1000 Dg of the agent per mm2 of implant surface to which the agent is applied.
10544. The method of item 10342 wherein the agent is delivered from the implant, wherein a surface of the implant comprises about 1000 Dg to about 2500 Dg of the agent per mm2 of implant surface to which the agent is applied.
10545. The method of item 10342r wherein the implant further comprises a coating, and the coating is a uniform coating.
10546. The method of item 10342, wherein the implant further comprises a coating, and the coating is a non-uniform coating.
10547. The method of item 10342, wherein the implant further comprises a coating, and the coating is a discontinuous coating.
10548. The method of item 10342, wherein the implant further comprises a coating, and the coating is a patterned coating. 10549. The method of item 10342, wherein the implant further comprises a coating, and the coating has a thickness of 100 Dm or less.
10550. The method of item 10342, wherein the implant further comprises a coating, and the coating has a thickness of 10 Dm or less.
10551. The method of item 10342, wherein the implant further comprises a coating, and the coating adheres to the surface of the implant upon deployment of the implant.
10552. The method of item 10342, wherein the implant further comprises a coating, and the coating is stable at room temperature for a period of at least 1 year.
10553. The method of item 10342, wherein the implant further comprises a coating, and the agent is present in the coating in an amount ranging between about 0.0001% to about 1% by weight.
10554. The method of item 10342, wherein the implant further comprises a coating, and the agent is present in the coating in an amount ranging between about 1% to about 10% by weight.
10555. The method of item 10342, wherein the implant further comprises a coating, and the agent is present in the coating in an amount ranging between about 10% to about 25% by weight.
10556. The method of item 10342, wherein the implant further comprises a coating, and the agent is present in the coating in an amount ranging between about 25% to about 70% by weight. 10557. The method of item 10342, wherein the implant further comprises a coating, and the coating comprises a polymer.
10558. The method of item 10342, wherein the implant comprises a first coating having a first composition and a second coating having a second composition.
10559. The method of item 10342, wherein the implant comprises a first coating having a first composition and a second coating having a second composition, wherein the first composition and the second composition are different.
10560. A method as in any one of items 10342-10559, wherein the device is a left ventricular assist device.
10561. A method as in any one of items 10342-10559, wherein the device is a right ventricular assist device.
10562. A method as in any one of items 10342-10559, wherein the device is a biventricular assist device.
10563. A method as in any one of items 10342-10559, wherein the device is a cardiac assist device.
10564. A method as in any one of items 10342-10559, wherein the device is a mechanical assist device.
10565. A method as in any one of items 10342-10559, wherein the device is an artificial cardiac assist device. 10566. A method as in any one of items 10342-10559, wherein the device is an implantable heart assist system.
10567. A method as in any one of items 10342-10559, wherein the device is a heart assist pump.
10568. A method as in any one of items 10342-10559, wherein the device is an intraventricular cardiac assist device.
10569. A method for inhibiting scarring comprising placing a device that comprises a spinal implant and an anti-scarring agent or a composition comprising an anti-scarring agent into an animal host, wherein the agent inhibits scarring.
10570. The method of item 10569 wherein the agent is an adensosine A2A receptor antagonist.
10571. The method of item 10569 wherein the agent is an AKT inhibitor.
10572. The method of item 10569 wherein the agent is an alpha 2 integrin antagonist, wherein the alpha 2 integrin antagonist is Pharmaprojects No. 5754 (Merck KgaA).
10573. The method of item 10569 wherein the agent is an alpha 4 integrin antagonist.
10574. The method of item 10569 wherein the agent is an alpha 7 nicotinic receptor agonist. 10575. The method of item 10569 wherein the agent is an angiogenesis inhibitor selected from the group consisting of AG-12,958 (Pfizer), ATN-161 (Attenuon LLC), neovastat, an angiogenesis inhibitor from Jerina AG (Germany), NM-3 (Mercian), VGA-1155 (Taisho), FCE-26644 (Pfizer), FCE-26950 (Pfizer), FPMA (Meiji Daries), FR-111142 (Fujisawa), GGTI-298, GM-1306 (Ligand), GPA-1734 (Novartis), NNC-47-0011 (Novo Nordisk), herbamycin (Nippon Kayaku), lenalidomide (Celegene), IP-10 (NIH), ABT-828 (Abbott), KIN-841 (Tokushima University, Japan), SF-1126 (Semafore Pharmaceuticals), laminin technology (NIH), CHIR-258 (Chiron), NVP-AEW541 (Novartis), NVP-AEW541 (Novartis), Vt16907 (Alchemia), OXI-8007 (Oxigene), EG-3306 (Ark Therapeutics), Maspin (Arriva), ABT- 567 (Abbott), PPI-2458 (Praecis Pharmaceuticals), CC-5079, CC-4089 (Celgene), HIF-1 alpha inhibitors (Xenova), S-247 (Pfizer), AP-23573 (Ariad), AZD-9935 (Astra Zeneca), mebendazole (Introgen Therapeutics), MetAP-2 inhibitors (GlaxoSmithKline), AG-615 (Angiogene Pharmaceuticals), Tie-2 antagonists (Hybrigenics), NC-381 , CYC-381 , NC-169, NC-219, NC-383, NC-384, NC-407 (Lorus Therapeutics), ATN-224 (Attenuon), ON-01370 (Onconova), Vitronectin antagonists (Amgen), SDX-103 (Salmedix), Vitronectin antagonists (Shire), CHP (Riemser), TEK (Amgen), Anecortave acetate (Alcon), T46.2 (Matrix Therapeutics), HG-2 (Heptagen), TEM antagonists (Genzyme), Oxi-4500 (Oxigene), ATN-161 (Attenuon), WX-293 (Wilex), M-2025 (Metris Therapeutics), Alphastatin (BioActa), YH-16 (Yantai Rongchang), BIBF-1120 (Boehringer Ingelheim), BAY-57-9352 (Bayer), AS- 1404 (Cancer Research Technology), SC-77964 (Pfizer), glycomimetics (BioTie Therapies), TIE-2 Inhibitors (Ontogen), DIMI1 Octamer (Octamer), ABR-215050 (Active Biotech), ABT-518 (Abbott), KDR inhibitors (Abbott), BSF-466895 (Abbott), SCH-221153 (Schering-Plough), DAC:antiangiogenic (ConjuChem), TFPI (EntreMed), AZD-2171 (Astra-Zenaca), CDC-394 (Celgene), LY290293 (EIi Lilly), IDN-5390 (Indena), Kdr Kinase Inhibitors (Merck), CT-113020, CT-116433, CT-116563, CT-31890, CT-32228) (Cell Therapeutics), A-299620 (Abbott), TWEAK Inhibitor (Amgen), VEGF modulators (Johnson and Johnson), Tum-N53, tumstatin (Genzyme), Thios- 1 , Thios-2 (Thios Pharmaceuticals), MV-6401 (Miravant Medical Technologies), Spisulosine (PharmaMar), CEP-7055 (Cephalon), AUV-201 (Auvation), LM-609 (EIi Lilly), SKF-106615 (AnorMED), Oglufanide disodium (Cytran), BW-114 (Pharminox), Calreticulin (NIH), WX-678 (Wilex), SD-7784 (Pfizer), WX-UK1 (Wilex), SH-268 (Schering AG), 2-Me-PGA (Celgene), S- 137 (Pfizer), ZD-6126 (Angiogene Pharmaceuticals), SG-292 (SignalGen), Benefin (Lane Labs), A6, A36 (Angstrom), SB-2723005 (GlaxoSmithKline), SC-7 (Cell Therapeutics), ZEN-014 (AEterna Zentaris), 2-methoxyestradiol (EntreMed), NK-130119 (Nippon Kayaku), CC-10004 (Celgene), AVE- 8062A (Ajinomoto), Tacedinaline (Pfizer), Actinonin (Tokyo Metropolitan Institute of Medical Science), Lenalidomide (Celgene), VGA-1155, BTO-956 (SRI International), ER-68203-00 (Eisai), CT-6685 (UCB), JKC-362 (Phoenix Pharmaceuticals), DMI-3798 (DMI Biosciences, Angiomate (Ipsen), ZD-6474 (AstraZeneca), CEP-5214 (Cephalon), Canstatin (Genzyme), NM-3 (Mercian), Oridigm (MediQuest Therapeutics), Exherin (Adherex), BLS-0597 (Boston Life Sciences), PTC-299 (PTC Therapeutics), NPI-2358 (Nereus Pharmaceuticals), CGP-79787 (Novartis), AEE-788 (Novartis), CKD-732 (Chong Kun Dang), CP-564959 (OSI Pharmaceuticals), CM-101 (CarboMed), CT-2584, CT3501 (Cell Therapeutics), combretastatin and analogues and derivatives thereof (Oxigene), Rebimastat (Bristol-Meyers Squibb), Dextrin 2-sulfate (ML Laboratories), Cilengitide (Merk KGaA), NSC-706704 (Pharminox), KRN- 951 (Kirin Brewery), Ukrain, NSC-631570 (Nowicky Pharma), Tecogalan sodium (Daiichi Pharmaceutical), Tz-93 (Tsumura), TBC-1635 (Encysive Pharmaceuticals), TAN-1120 (Takeda), Semaxanib (Pfizer), BDI-7800 (Biopharmacopae), SD-186, SD-983 (Bristol-Meyers Squibb), SB-223245 (GlaxoSmithKline), SC-236 (Pfizer), RWJ-590973 (Johnson and Johnson), ILX-1850 (Genzyme), SC-68488, S-836 (Pfizer), CG-55069-11 (CuraGen), Ki-23057 (Kirin Brewery), CCX-700 (Chemoentryx), Pegaptanib octasodium (Giled Sciences), ANGIOCOL (available from Biostratum Inc.), or an analogue or derivative thereof.
10576. The method of item 10569 wherein the agent is an apoptosis antagonist.
10577. The method of item 10569 wherein the agent is an apoptosis activator.
10578. The method of item 10569 wherein the agent is a beta 1 integrin antagonist.
10579. The method of item 10569 wherein the agent is a beta tubulin inhibitor.
10580. The method of item 10569 wherein the agent is a blocker of enzyme production in Hepatitis C.
10581. The method of item 10569 wherein the agent is a Bruton's tyrosine kinase inhibitor.
10582. The method of item 10569 wherein the agent is a calcineurin inhibitor.
10583. The method of item 10569 wherein the agent is a caspase 3 inhibitor.
10584. The method of item 10569 wherein the agent is a CC chemokine receptor antagonist. 10585. The method of item 10569 wherein the agent is a cell cycle inhibitor selected from the group consisting of SNS-595 (Sunesis), synthadotin, KRX-0403, and an analogue or derivative thereof.
10586. The method of item 10569 wherein the agent is a cathepsin B inhibitor.
10587. The method of item 10569 wherein the agent is a cathepsin K inhibitor, wherein the cathepsin K inhibitor is 462795 (GlaxoSmithKline), INPL-022-D6 (Amura Therapeutics), or an analogue or derivative thereof.
10588. The method of item 10569 wherein the agent is a cathepsin L inhibitor.
10589. The method of item 10569 wherein the agent is a CD40 antagonist.
10590. The method of item 10569 wherein the agent is a chemokine receptor agonist.
10591. The method of item 10569 wherein the agent is a chymase inhibitor.
10592. The method of item 10569 wherein the agent is a collagenase antagonist.
10593. The method of item 10569 wherein the agent is a CXCR antagonist. 10594. The method of item 10569 wherein the agent is a cyclin dependent kinase inhibitor selected from the group consisting of a CDK-1 inhibitor, a CDK-2 inhibitor, a CDK- 4 inhibitor, a CDK-6 inhibitor, a CAK1 inhibitor from GPC Biotech and Bristol-Myers Squibb, RGB-286199 (GPC Biotech), an anticancer agent from Astex Technology, a CAK1 inhibitor from GPC Biotech, a CDK inhibitor from Sanofi-Aventis, a CDK1/CDK2 inhibitor from Amgen, a CDK2 inhibitor from SUGEN-2 (Pfizer), a hearing loss therapy agent (Sound Pharmaceuticals), PD-0332991 (Pfizer), RGB-286199 (GPC Biotech), Ro-0505124 (Hoffmann-La Roche), a Ser/Thr kinase inhibitor from Lilly (EIi Lilly), CVT-2584 (CAS No. 199986-75- 9) (CV Therapeutics), and an analogue or derivative thereof.
10595. The method of item 10569 wherein the agent is a cyclooxygenase 1 inhibitor.
10596. The method of item 10569 wherein the agent is a DHFR inhibitor.
10597. The method of item 10569 wherein the agent is a dual integrin inhibitor.
10598. The method of item 10569 wherein the agent is an elastase inhibitor.
10599. The method of item 10569 wherein the agent is an elongation factor-1 alpha inhibitor.
10600. The method of item 10569 wherein the agent is an endothelial growth factor antagonist. 10601. The method of item 10569 wherein the agent is an endothelial growth factor receptor kinase inhibitor selected from the group consisting of sorafenib tosylate (Bayer), AAL-993 (Novartis), ABP-309 (Novartis), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), E- 7080 (Eisai), EG-3306 (Ark Therapeutics), EXEL-2880 (Exelixis), GW- 654652 (GlaxoSmithKline), a KDR inhibitor from LG Life Sciences, CT-6685 and CT-6729 (UCB), KRN-633 and KRN-951 (Kirin Brewery), OSI-930 (OSI Pharmaceuticals), SP-5.2 (Supratek Pharma), SU-11657 (Pfizer), a Tie-2 antagonist (Hybrigenics), a VEGFR-2 kinase inhibitor (Bristol-Myers Squibb), XL-647 (Exelixis), a KDR inhibitor from Abbott Laboratories, sorafenib tosylate, and an analogue or derivative thereof.
10602. The method of item 10569 wherein the agent is an endotoxin antagonist.
10603. The method of item 10569 wherein the agent is an epothilone and tubulin binder.
10604. The method of item 10569 wherein the agent is an estrogen receptor antagonist.
10605. The method of item 10569 wherein the agent is an FGF inhibitor.
10606. The method of item 10569 wherein the agent is a farnexyl transferase inhibitor.
10607. The method of item 10569 wherein the agent is famesyltransferase inhibitor selected from the group of A-197574 (Abbott), a famesyltransferase inhibitor from Servier, FPTIII (Strathclyde Institute for Drug R), LB-42908 (LG Life Sciences), Pharmaprojects No. 5063 (Genzyme), Pharmaprojects No. 5597 (Ipsen), Yissum Project No. B-1055 (Yissum), and an analogue or derivative thereof.
10608. The method of item 10569 wherein the agent is an FLT-3 kinase inhibitor.
10609. The method of item 10569 wherein the agent is an FGF receptor kinase inhibitor.
10610. The method of item 10569 wherein the agent is a fibrinogen antagonist selected from the group consisting of AUV-201 (Auvation), MG-13926 (Sanofi-Aventis), plasminogen activator (CAS No. 105913-11-9) (from Sanofi-Aventis or UCB), plasminogen activator-2 (tPA- 2) (Sanofi-Aventis), pro-urokinase (CAS No. 82657-92-9) (Sanofi-Aventis), and an analogue or derivative thereof.
10611. The method of item 10569 wherein the agent is a heat shock protein 90 antagonist selected from the group consisting of SRN- 005 (Sirenade), geldanamycin, NSC-33050 (17-allylaminogeldanamycin; 17- AAG), 17-dimethylaminoethylamino-17-demethoxy-geldanamycin (17- DMAG), rifabutin (rifamycin XIV, 1',4-didehydro-1-deoxy-1 ,4-dihydro-5'-(2- methylpropyl)-1-oxo-), and an analogue or derivative thereof.
10612. The method of item 10569 wherein the agent is a histone deacetylase inhibitor.
10613. The method of item 10569 wherein the agent is an HMGCoA reductase inhibitor selected from the group consisting of an atherosclerosis therapeutic from Lipid Sciences, ATI-16000 (ARYx Therapeutics), KS-01-019 (Kos Pharmaceuticals), Pharmaprojects No. 2197 (Sanofi-Aventi), RP 61969 (Sanofi-Aventis), and an analogue or derivative thereof.
10614. The method of item 10569 wherein the agent is an ICAM inhibitor.
10615. The method of item 10569 wherein the agent is an IL, ICE and IRAK antagonist, wherein the antagonist is a CJ-14877, CP- 424174 (Pfizer), NF-61 (Negma-Lerads), and an analogue or derivative thereof.
10616. The method of item 10569 wherein the agent is an IL-2 inhibitor.
10617. The method of item 10569 wherein the agent is an immunosuppressant selected from the group consisting of teriflunomide (Sanofi Aventis), chlorsulfaquinoxalone (NSC-339004), chlorsulfaquinoxalone sulfate, CS-712 (Sankyo), ismomultin alfa (CAS No. 457913-93-8) (Akzo Nobel), antiallergics from GenPat77, antiinflammatories or AT-005 (Androclus Therapeutics), autoimmune disease therapeutics from EpiVax, BN-007 (Bone), budesonide (CAS No. 51333-22- 3) (MAP Pharmaceuticals), CO-14 (Genzyme), edratide (CAS No. 433922- 67-9) (Teva), EP-314 (Enanta), eprovafen (CAS No. 101335-99-3) (Sanofi- Aventis), HWA-131 (CAS No. 118788-41-3) (Sanofi-Aventis), immunomodulators from MerLion Pharmaceuticals, immunosuppressives from Alchemia, IPL-12 (Inflazyme), MDL-9563 (CAS No. 27086-86-8) (Sanofi-Aventis), Pharmaprojects No. 2330 (Sanofi-Aventis), Pharmaprojects No. 6426 (Abgenix), PXS-25 (Pharmaxis), rosmarinic acid (CAS No. 20283-92-5) (Sanofi-Aventis), RP 42927 or RP 54745 (CAS No. 135330-08-4) (Sanofi-Aventis), SGN-35 (Seattle Genetics), ST-1959 (Sigma-Tau), type I diabetes therapy from SYNX Pharma, UNIL-88 (Debiopharm), VP-025 (Vasogen), VR-694 (Vectura), PRTX-001 (Protalex), and an analogue or derivative thereof.
10618. The method of item 10569 wherein the agent is an fMPDH (inosine monophosphate).
10619. The method of item 10569 wherein the agent is an integrin antagonist.
10620. The method of item 10569 wherein the agent is an interleukin antagonist.
10621. The method of item 10569 wherein the agent is an inhibitor of type III receptor tyrosine kinase.
10622. The method of item 10569 wherein the agent is an irreversible inhibitor of enzyme methionine aminopeptidase type 2.
10623. The method of item 10569 wherein the agent is an isozyme selective delta protein kinase C inhibitor.
10624. The method of item 10569 wherein the agent a JAK3 enzyme inhibitor.
10625. The method of item 10569 wherein the agent is a JNK inhibitor.
10626. The method of item 10569 wherein the agent is a kinase inhibitor. 10627. The method of item 10569 wherein the agent is kinesin antagonist.
10628. The method of item 10569 wherein the agent is a kinesin antagonist.
10629. The method of item 10569 wherein the agent is a leukotriene inhibitor and antagonist selected from the group consisting of ambicromil (CAS No. 58805-38-2) (Sanofi-Aventis), amelubant (CAS No. 346735-24-8) (Boehringer Ingelheim), DW-1141 (Dong Wha), ebselen (Daiichi Pharmaceutical), ibudilast (Kyorin), leucotriene inhibitors from Sanofi-Aventis, lymphotoxin-beta receptor (LT-D) from Biogen Idee, Pharmaprojects No. 1535 and 2728 (CAS No. 119340-33-9) (Sanofi- Aventis), R-112 (Rigel), Rev-5367 (CAS No. 92532-05-3) (Sanofi-Aventis), RG-14893 (CAS No. 141835-49-6) (Sanofi-Aventis), RG-5901-A (CAS No. 101910-24-1), 92532-23-5, RP 66153 (CAS No. 142422-79-5), RP 66364 (CAS No.186916-92-5), RP 69698 (CAS No. 141748-00-7) (Sanofi-Aventis), SC-411930 (Pfizer), SC-41930 (CAS No. 120072-59-5) (Pfizer), SC-50605 (CAS No. 138828-39-4) (Pfizer), SC-51146 (CAS No. 141059-52-1), SC- 53228 (CAS No. 153633-01-3) (Pfizer), spaglumic acid (ZY-15106) (CAS No.3106-85-2), 80619-64-3 (Novartis), tipredane (CAS No. 85197-77-9) (Bristol-Myers Squibb), U-75302 (CAS No. 119477-85-9) (Pfizer), and analogue or derivative thereof.
10630. The method of item 10569 wherein the agent is an MAP kinase inhibitor.
10631. The method of item 10569 wherein the agent is a matrix metalloproteinase inhibitor. 10632. The method of item 10569 wherein the agent is an MCP-CCR2 inhibitor.
10633. The method of item 10569 wherein the agent is an mTOR inhibitor.
10634. The method of item 10569 wherein the agent is an mTOR kinase inhibitor.
10635. The method of item 10569 wherein the agent is a microtubule inhibitor selected from the group consisting of antibody- maytansinoid conjugates from Biogen Idee, colchicines (MantiCore Pharmaceuticals), anticancer immunoconjugates from Johnson & Johnson, DIME from Octamer, gni-1f (GNI), huC242-DM4, huMy9-6-DM1 (ImmunoGen), IDN-5404 (Indena), IMO-098, IMOderm (Imotep), mebendazole (Introgen Therapeutics), microtubule poisons from Cambridge Enterprise, paclitaxel such as LOTAX from Aphios (CAS No. 33069-62-4), Genexol-PM from Samyang, Pharmaprojects No. 6383 (Tapestry Pharmaceuticals), RPR-112378 (Sanofi-Aventis), SGN-75 (Seattle Genetics), SPL-7435 (Starpharma), SSR-250411 (Sanofi-Aventis), trastuzumab-DM1 (Genentech), vinorelbine, and an analogue or derivative thereof.
10636. The method of item 10569 wherein the agent is an MIF inhibitor.
10637. The method of item 10569 wherein the agent is an MMP inhibitor.
10638. The method of item 10569 wherein the agent is a neurokinin (NK) antagonist selected from the group consisting of anthrotainin (CAS No. 148084-40-6) (Sanofi-Aventis), an IBS thereapeutic from ArQuIe1 MDL-105212A (CAS No. 167261-60-1) (Ssanofi-Aventis), Pharmaprojects No. 2744, 3258 (CAS No. 139167-47-8) 4006, 4201 , or 5986 (Sanofi-Aventis), RP 67580 (CAS No. 135911-02-3), SR-144190 (CAS No. 201152-86-5), SSR-240600, SSR-241586 (Sanofi-Aventis), TKA-457 (Novartis), vestipitant mesylate (CAS No. 334476-64-1) (GlaxoSmithKline), Win-64821 (Sanofi-Aventis), PRX-96026 (Predix Pharmaceuticals), and an analogue or derivative thereof.
10639. The method of item 10569 wherein the agent is an NF kappa B inhibitor selected from the group consisting of AVE-0545 or AVE-0547 (Sanofi-Aventis), bortezomib (CAS No. 179324-69-7) (Millennium Pharmaceuticals), dexanabinol (CAS No. 112924-45-5) (Pharmos), dexlipotam (Viatris), Pharmaprojects No. 6283 (INDRA) (OXiGENE), IPL- 576092 (CAS No. 137571-30-3) (Inflazyme), NFKB decoy (Corgentech), NFKB decoy oligo (AnGes MG), NFKB's from Ariad, osteoporosis treatments or S5 (F005) from Fulcrum Pharmaceuticals, P61 (Phytopharm), R-flurbiprofen (CAS No. 5104-49-4) (Encore Pharmaceuticals), and an analogue or derivative thereof.
10640. The method of item 10569 wherein the agent is a nitric oxide agonist.
10641. The method of item 10569 wherein the agent is an ornithine decarboxylase inhibitor.
10642. The method of item 10569 wherein the agent is a p38 MAP kinase inhibitor selected from the group consisting of AZD-6703 (AstraZeneca), JX-401 (Jexys Pharmaceuticals), BMS-2 (Bristol-Myers Squibb), a p38 MAP kinase inhibitor from Novartis, a p38-alpha MAP kinase inhibitor from Amphora, Pharmaprojects No. 5704 (Pharmacopeia), RPR- 200765A (Sanofi-Aventis), SD-282 (Johnson & Johnson), TAK-715 (Takeda), and an analogue or derivative thereof.
10643. The method of item 10569 wherein the agent is a palmitoyl-protein thioesterase inhibitor.
10644. The method of item 10569 wherein the agent is a PDGF receptor kinase inhibitor selected from the group consisting of AAL- 993, AMN-107, or ABP-309 (Novartis), AMG-706 (Amgen), BAY-57-9352 (Bayer), CDP-860 (UCB), E-7080 (Eisai), imatinib (CAS No. 152459-95-5) (Novartis), OSI-930 (OSI Pharmaceuticals), RPR-127963E (Sanofi-Aventis), RWJ-540973 (Johnson & Johnson), sorafenib tosylate (Bayer), SU-11657 (Pfizer), tandutinib (CAS No. 387867-13-2) (Millennium Pharmaceuticals), vatalanib (Novartis), ZK-CDK (Schering AG), and an analogue or derivative thereof.
10645. The method of item 10569 wherein the agent is (-)-halofenate (Metabolex), AMG-131 (Amgen), antidiabetics from Japan Tobacco, AZD-4619, AZD-8450, AZD-8677 (AstraZeneca), DRF-10945, balaglitazone (Dr Reddy's), CS-00088, CS-00098 (Chipscreen Biosciences), E-3030 (Eisai), etalocib (CAS No. 161172-51-6) (EIi Lilly), GSK-641597 (Ligand), GSK-677954 (GlaxoSmithKline), GW-409544 (Ligand), GW- 590735 (GlaxoSmithKline), K-111 (Hoffmann-La Roche), LY-518674 (EIi Lilly), LY-674 (Ligand), LY-929 (Ligand), MC-3001, MC-3002 (MaxoCore Pharmaceuticals), metformin HCI + pioglitazone (CAS No. 1115-70-4 and 112529-15-4), ACTOPLUS MET from Andrx), muraglitazar (CAS No. 331741-94-7) (Bristol-Myers Squibb), naveglitazar (Ligand), oleoylethanolamide (Kadmus Pharmaceuticals), ONO-5129, pioglitazone hydrochloride (CAS No. 111025-46-8 and 112529-15-4) (Takeda), PLX-204 (Plexxikon), PPAR agonists from Genfit, PPAR delta agonists from EIi Lilly, PPAR-alpha agonists from CrystalGenomics, PPAR-gamma modulators and PPAR-β modulators from CareX, rosiglitazone maleate (CAS No. 122324- 73-4 or 155141-29-0) (GlaxoSmithKline), rosiglitazone maleate/glimepir (CAS No. 155141-29-0 and 93479-97-1), AVANDARYL, rosiglitazone maleate/metformin extend (CAS No. 155141-29-0 and 657-24-9), AVANDAMET, rosiglitazone maleate+metformin, AVANDAMET (GlaxoSmithKline), tesaglitazar (AstraZeneca), and an analogue or derivative thereof.
10646. The method of item 10569 wherein the agent is a phosphatase inhibitor.
10647. The method of item 10569 wherein the agent is a phosphodiesterase (PDE) inhibitor selected from the group consisting of avanafil (Tanabe Seiyaku), dasantafil (CAS No. 569351-91-3) (Schering- Plough), A-906119 (CAS No. 134072-58-5), DL-850 (Sanofi-Aventis), GRC- 3015, GRC-3566, GRC-3886 (Glenmark), HWA-153 (CAS No. 56395-66-5) (Sanofi-Aventis), hydroxypumafentrine (Altana), IBFB-130011 , IBFB-14-016, IBFB-140301 , IBFB-150007, IBFB-211913 (IBFB Pharma), L-826141 (Merck & Co), medorinone (CAS No. 88296-61-1) (Sanofi-Aventis), MEM- 1917 (Memory Pharmaceuticals), ND-1251 (Neuro3d), PDE inhibitors from ICOS, PDE IV inhibitors from Memory Pharmaceuticals and CrystalGenomics, Pharmaprojects No. 2742 and 6141 (Sanofi-Aventis), QAD-171 (Novartis), RHC-2963 (CAS No. 76993-12-9 and 76993-14-1), RPR-117658, RPR-122818 derivatives, SR-24870 , and RPR-132294 (Sanofi-Aventis), SK-350 (ln2Gen), stroke therapy agents from deCODE Genetics, TAS-203 (Taiho), tofimilast (CAS No. 185954-27-2) (Pfizer), UK- 371800 (Pfizer), WIN-65579 (CAS No. 158020-82-7) (Sanofi-Aventis), IBFB- 130020 (IBFB Pharma), OPC-6535 (CAS No. 145739-56-6) (Otsuka), a phosphodiesterase III inhibitor, enoximone, a phosphodiesterase IV inhibitor, fosfosal, Atopik (Barrier Therapeutics), triflusal, a phosphodiesterase V inhibitor, and an analogue or derivative thereof. 10648. The method of item 10569 wherein the agent is a PKC inhibitor.
10649. The method of item 10569 wherein the agent is a platelet activating factor antagonist.
10650. The method of item 10569 wherein the agent is a platelet-derived growth factor receptor kinase inhibitor.
10651. The method of item 10569 wherein the agent is a prolyl hydroxylase inhibitor.
10652. The method of item 10569 wherein the agent is a polymorphonuclear neutrophil inhibitor.
10653. The method of item 10569 wherein the agent is a protein kinase B inhibitor.
10654. The method of item 10569 wherein the agent is a protein kinase C stimulant.
10655. The method of item 10569 wherein the agent is a purine nucleoside analogue.
10656. The method of item 10569 wherein the agent is a purinoreceptor P2X antagonist.
10657. The method of item 10569 wherein the agent is a Raf kinase inhibitor. 10658. The method of item 10569 wherein the agent is a reversible inhibitor of ErbB1 and ErbB2.
10659. The method of item 10569 wherein the agent is a ribonucleoside triphosphate reductase inhibitor.
10660. The method of item 10569 wherein the agent is an SDF-1 antagonist.
10661. The method of item 10569 wherein the agent is a sheddase inhibitor.
10662. The method of item 10569 wherein the agent is an SRC inhibitor.
10663. The method of item 10569 wherein the agent is a stromelysin inhibitor.
10664. The method of item 10569 wherein the agent is an Syk kinase inhibitor.
10665. The method of item 10569 wherein the agent is a telomerase inhibitor.
10666. The method of item 10569 wherein the agent is a TGF beta inhibitor selected from the group consisting of pirfenidone (CAS No. 53179-13-8) (MARNAC), tranilast (CAS No. 53902-12-8) (Kissei), IN- 1130 (ln2Gen), mannose-6-phosphate (BTG), TGF-β antagonists from Inflazyme (Pharmaprojects No. 6075), TGF-β antagonists from Sydney, non-industrial source), TGF-β I receptor kinase inhibitors from EIi Lilly, TGF- β receptor inhibitors from Johnson & Johnson, and an analogue or derivative thereof.
10667. The method of item 10569 wherein the agent is a
TNFα antagonist or TACE inhibitor selected from the group consisting of adalimumab (CAS No. 331731-18-1) (Cambridge Antibody Technology), AGIX-4207 (AtheroGenics), AGT-1 (Advanced Biotherapy), an antiinflammatory from Borean Pharma, Cellzome, or Paradigm Therapeutics, anti-inflammatory vaccine (TNF-alpha kinoid) from Neovacs, humanized anti-TNF antibody or an anti-TNF MAb (CB0006) Celltech (UCB), apratastat (CAS No. 287445-51-0) (Wyeth), BMS-561392 (Bristol-Myers Squibb), BN- 006 (Bone), certolizumab pegol (CAS No. 428863-50-7 or CH-138 (UCB), cilomilast (CAS No. 153259-65-5) (GlaxoSmithKline), CR-1 (Nuada Pharmaceuticals), CRx-119 (CombinatoRx), D-5410 (UCB), dacopafant (CAS No. 125372-33-0) (Sanofi-Aventis), dersalazine (CAS No. 188913-57- 7/188913-58-8) (Uriach), etanercept (CAS No. 185243-69-0) (Amgen), ethyl pyruvate (Critical (Critical Therapeutics), golimumab (CAS No. 476181-74-5) (Johnson & Johnson), hormono-immunotherapy from Ipsen, CDP571 (e.g., Humicade from UCB), IC-485 (ICOS), infliximab (CAS No. 170277-31-3) (Johnson & Johnson), IP-751 (Manhattan Pharmaceuticals), ISIS-104838 (CAS No. 250755-32-9) (ISIS Pharmaceuticals), lenalidomide (CAS No. 191732-72-6) (Celgene), lentinan (CAS No. 37339-90-5) (Ajinomoto), MDL- 201112 (CAS No. 142130-73-2) (Sanofi-Aventis), medroxyprogesterone (CAS No. 520-85-4) (InKine Pharmaceutical), N-acetylcysteine (CAS No. 616-91-1) (Zambon), NBE-P2 (DIREVO Biotech), nerelimomab (CAS No. 162774-06-3) (Chiron), OM-294DP (OM PHARMA), onercept (CAS No. 199685-57-9) (Yeda), PASSTNF-alpha (Verigen), pentoxifylline or oxypentifylline (Sanofi-Aventis), Pharmaprojects No. 4091, 4241, 4295, or 4488 (Sanofi-Aventis), Pharmaprojects No. 5480 (Amgen), Pharmaprojects No. 6749 (Cengent), pirfenidone (CAS No. 53179-13-8) (MARNAC), RPR- 132294 (Sanofi-Aventis), S5 (F002) (Fulcrum Pharmaceuticals), simvastatin (CAS No. 79902-63-9) (Merck & Co), STA-6292 (Synta Pharmaceuticals), tacrolimus (CAS No. 104987-11-3) (Fujisawa LifeCycle Pharma), talactoferrin alfa (CAS No. 308240-58-6) (Agennix), thalidomide (CAS No. 50-35-1) (Celgene), TNF antagonists form ProStrakan, and Synergen, TNF inhibitors (Amgen), TNF-alpha antagonists from Dynavax Technologies and Jerina AG (Germany), TNF-alpha inhibitors from IBFB Pharma and Xencor (Xencor), torbafylline (CAS No. 105102-21-4) (Sanofi-Aventis), UR-1505 (Uriach), VT-346 (Viron Therapeutics), YSIL6 (Y's Therapeutics), YSTH2 (Y's Therapeutics), NPI-1302a-3 (Nereus Pharmaceuticals, a TNF antagonist from Jerina AG (Germany), dersalazine, and an analogue or derivative thereof.
10668. The method of item 10569 wherein the agent is a Toll receptor inhibitor.
10669. The method of item 10569 wherein the agent is a tubulin antagonist.
10670. The method of item 10569 wherein the agent is a tyrosine kinase inhibitor selected from the group consisting of SU-011248, SUTENT from Pfizer Inc. (New York, NY), BMS-354825, PN-355 (Paracelsian Pharmaceuticals), AGN-199659 (Allergan), AAL-993 or ABP- 309 (Novartis), adaphostin (NIH), AEE-788 (Novartis), AG-013736 (OSI Pharmaceuticals), AG-13736 (Pfizer), ALT-110 (Alteris Therapeutics), AMG- 706 (Amgen), anticancer MAbs from Xencor, anti-EGFrvlll MAbs from Abgenix, anti-HER2 MAb from Abiogen, AZD-2171 or AZD-9935 (AstraZeneca), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), CEP-5214 (Cephalon), CEP-7055 (Cephalon), cetuximab (ImClone Systems), CHIR-200131 and CHIR-258 (Chiron), CP-547632 (OSI Pharmaceuticals), CP-724714 (Pfizer), CT-301 (Creabilis Therapeutics), D- 69491 (Baxter International), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EGFR/ErbB2 inhibitors from Array BioPharma, erlotinib (CAS No. 183319- 69-9) (OSI Pharmaceuticals), EXEL-2880 (Exelixis), FK-778 (Sanofi- Aventis) , gefitinib (CAS No. 184475-35-2) (AstraZeneca), GW-2286 or GW- 654652 (GlaxoSmithKline), her2/neu antigen from AlphaVax, HER-2/neu inhibitor from Generex, Herzyme (Medipad) (Sirna Therapeutics), HKI-272 (Wyeth), HuMax-EGFr (Genmab), idronoxil (CAS No. 81267-65-4) (Novogen), IGF-1 inhibitors from Ontogen, IMC-11 F8 (ImCIone Systems), kahalalide F (CAS No. 149204-42-2) (PharmaMar), KDR inhibitor from LG Life Sciences, KDR inhibitors from Abbott Laboratories, KDR kinase inhibitors (UCB), Kdr kinase inhibitors from Merck & Co, KRN-633 and KRN- 951 (Kirin Brewery), KSB-102 (Xenova), lapatinib ditosylate (CAS No. 388082-78-8) (GlaxoSmithKline), matuzumab (Merck KGaA), MDX-214 (Medarex), ME-103 (Pharmexa), MED-A300 (Gerolymatos), MNAC-13 (Lay Line Genomics), nimotuzumab (Center of Molecular Immunology), NSC- 330507 or NSC-707545 (NIH), NV-50 (Novogen), OSI-930 (OSI Pharmaceuticals), panitumumab (Abgenix), pelitinib (CAS No. 287933-82-7) (Wyeth), pertuzumab (CAS No. 380610-27-5) (Genentech), Pharmaprojects No. 3985 (Sanofi-Aventis), prostate cancer therapeutics from Sequenom (SQPC35, SQPC36, SQPC90), removab and remoxab (Trion Pharma), RG- 13022 (CAS No. 136831-48-6), RG-13291 (CAS No. 138989-50-1), or RG- 14620 (CAS No. 136831-49-7) (Sanofi-Aventis), RM-6427 (Romark), RNAi breast cancer therapy from Benitec, RP 53801 (CAS No. 125882-88-4) (Sanofi-Aventis), sorafenib tosylate (Bayer), SU-11657 (Pfizer), Tie-2 antagonists from Semaia (Hybrigenics), Tie-2 inhibitors from Ontogen, trastuzumab (CAS No. 180288-69-1) (Genentech), tyrosine kinase inhibitors from Sanofi-Aventis, U3-1287, U3-1565, U3-1784, U3-1800 (U3 Pharma), vatalanib (Novartis), VEGFR-2 kinase inhibitor from Bristol-Myers Squibb, XL-647 (Exelixis), ZD-6474 (AstraZeneca), ZK-CDK (Schering AG), an EGFR tyrosine kinase inhibitor, EKB-569 (Wyeth), and an analogue or derivative thereof. 10671. The method of item 10569 wherein the agent is a VEGF inhibitor.
10672. The method of item 10569 wherein the agent is a vitamin D receptor agonist.
10673. The method of item 10569 wherein the agent is ZD-6474 (an angiogenesis inhibitor).
10674. The method of item 10569 wherein the agent is AP-23573 (an mTOR inhibitor).
10675. The method of item 10569 wherein the agent is synthadotin (a tubulin antagonist).
10676. The method of item 10569 wherein the agent is S-0885 (a collagenase inhibitor).
10677. The method of item 10569 wherein the agent is aplidine (an elongation factor-1 alpha inhibitor).
10678. The method of item 10569 wherein the agent is ixabepilone (an epithilone).
10679. The method of item 10569 wherein the agent is IDN-5390 (an angiogenesis inhibitor).
10680. The method of item 10569 wherein the agent is SB-2723005 (an angiogenesis inhibitor). 10681. The method of item 10569 wherein the agent is ABT-518 (an angiogenesis inhibitor).
10682. The method of item 10569 wherein the agent is combretastatin (an angiogenesis inhibitor).
10683. The method of item 10569 wherein the agent is anecortave acetate (an angiogenesis inhibitor).
10684. The method of item 10569 wherein the agent is SB-715992 (a kinesin antagonist).
10685. The method of item 10569 wherein the agent is temsirolimus (an mTOR inhibitor).
10686. The method of item 10569 wherein the agent is adalimumab (a TNFα antagonist).
10687. The method of item 10569, wherein the composition comprises a polymer.
10688. The method of item 10569, wherein the composition comprises a polymer, and the polymer is, or comprises, a copolymer.
10689. The method of item 10569, wherein the composition comprises a polymer, and the polymer is, or comprises, a block copolymer. 10690. The method of item 10569, wherein the composition comprises a polymer, and the polymer is, or comprises, a random copolymer.
10691. The method of item 10569, wherein the composition comprises a polymer, and the polymer is, or comprises, a biodegradable polymer.
10692. The method of item 10569, wherein the composition comprises a polymer, and the polymer is, or comprises, a nonbiodegradable polymer.
10693. The method of item 10569, wherein the composition comprises a polymer, and the polymer is, or comprises, a hydrophilic polymer.
10694. The method of item 10569, wherein the composition comprises a polymer, and the polymer is, or comprises, a hydrophobic polymer.
10695. The method of item 10569, wherein the composition comprises a polymer, and the polymer is, or comprises, a polymer having hydrophilic domains.
10696. The method of item 10569, wherein the composition comprises a polymer, and the polymer is, or comprises, a polymer having hydrophobic domains.
10697. The method of item 10569, wherein the composition comprises a polymer, and the polymer is, or comprises, a non- conductive polymer. 10698. The method of item 10569, wherein the composition comprises a polymer, and the polymer is, or comprises, an elastomer.
10699. The method of item 10569, wherein the composition comprises a polymer, and the polymer is, or comprises, a hydrogel.
10700. The method of item 10569, wherein the composition comprises a polymer, and the polymer is, or comprises, a silicone polymer.
10701. The method of item 10569, wherein the composition comprises a polymer, and the polymer is, or comprises, a hydrocarbon polymer.
10702. The method of item 10569, wherein the composition comprises a polymer, and the polymer is, or comprises, a styrene-derived polymer.
10703. The method of item 10569, wherein the composition comprises a polymer, and the polymer is, or comprises, a butadiene-derived polymer.
10704. The method of item 10569, wherein the composition comprises a polymer, and the polymer is, or comprises, a macromer.
10705. The method of item 10569, wherein the composition comprises a polymer, and the polymer is, or comprises, a poly(ethylene glycol) polymer. 10706. The method of item 10569, wherein the composition comprises a polymer, and the polymer is, or comprises, an amorphous polymer.
10707. The method of item 10569, wherein the composition further comprises a second pharmaceutically active agent.
10708. The method of item 10569, wherein the composition further comprises an anti-inflammatory agent.
10709. The method of item 10569, wherein the composition further comprises an agent that inhibits infection.
10710. The method of item 10569, wherein the composition further comprises an anthracycline.
10711. The method of item 10569, wherein the composition further comprises doxorubicin.
10712. The method of item 10569 wherein the composition further comprises mitoxantrone.
10713. The method of item 10569 wherein the composition further comprises a fluoropyrimidine.
10714. The method of item 10569, wherein the composition further comprises 5-fluorouracil (5-FU).
10715. The method of item 10569, wherein the corriposition further comprises a folic acid antagonist. 10716. The method of item 10569, wherein the composition further comprises methotrexate.
10717. The method of item 10569, wherein the composition further comprises a podophylotoxin.
10718. The method of item 10569, wherein the composition further comprises etoposide.
10719. The method of item 10569, wherein the composition further comprises camptothecin.
10720. The method of item 10569, wherein the composition further comprises a hydroxyurea.
10721. The method of item 10569, wherein the composition further comprises a platinum complex.
10722. The method of item 10569, wherein the composition further comprises cisplatin.
10723. The method of item 10569 wherein the composition further comprises an antithrombotic agent.
10724. The method of item 10569, wherein the composition further comprises a visualization agent.
10725. The method of item 10569, wherein the composition further comprises a visualization agent, and the visualization agent is a radiopaque material, wherein the radiopaque material comprises a metal, a halogenated compound, or a barium containing compound. 10726. The method of item 10569, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, barium, tantalum, or technetium.
10727. The method of item 10569, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, an MRI responsive material.
10728. The method of item 10569, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, a gadolinium chelate.
10729. The method of item 10569, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, iron, magnesium, manganese, copper, or chromium.
10730. The method of item 10569, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, iron oxide compound.
10731. The method of item 10569, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, a dye, pigment, or colorant.
10732. The method of item 10569 wherein the agent is released in effective concentrations from the composition comprising the agent by diffusion over a period ranging from the time of administration to about 90 days.
10733. The method of item 10569 wherein the agent is released in effective concentrations from the composition comprising the agent by erosion of the composition over a period ranging from the time of administration to about 90 days.
10734. The method of item 10569 wherein the composition further comprises an inflammatory cytokine.
10735. The method of item 10569 wherein the composition further comprises an agent that stimulates cell proliferation.
10736. The method of item 10569 wherein the composition further comprises a polymeric carrier.
10737. The method of item 10569 wherein the composition is in the form of a gel, paste, or spray.
10738. The method of item 10569 wherein the implant is partially constructed with the agent or the composition.
10739. The method of item 10569 wherein the implant is fully constructed with the agent or the composition.
10740. The method of item 10569 wherein the implant is impregnated with the agent or the composition.
10741. The method of item 10569, wherein the agent or the composition forms a coating, and the coating directly contacts the implant.
10742. The method of item 10569, wherein the agent or the composition forms a coating, and the coating indirectly contacts the implant. 10743. The method of item 10569 wherein the agent or the composition forms a coating, and the coating partially covers the implant.
10744. The method of item 10569, wherein the agent or the composition forms a coating, and the coating completely covers the implant.
10745. The method of item 10569 wherein the agent or the composition is located within pores or holes of the implant.
10746. The method of item 10569 wherein the agent or the composition is located within a channel, lumen, or divet of the implant.
10747. The method of item 10569 wherein the implant further comprising an echogenic material.
10748. The method of item 10569 wherein the implant further comprises an echogenic material, wherein the echogenic material is in the form of a coating.
10749. The method of item 10569 wherein the implant is sterile.
10750. The method of item 10569 wherein the agent is delivered from the implant, wherein the agent is released into tissue in the vicinity of the implant after deployment of the implant.
10751. The method of item 10569 wherein the agent is delivered from the implant, wherein the agent is released into tissue in the vicinity of the implant after deployment of the implant, wherein the tissue is connective tissue.
10752. The method of item 10569 wherein the agent is delivered from the implant, wherein the agent is released into tissue in the vicinity of the implant after deployment of the implant, wherein the tissue is muscle tissue.
10753. The method of item 10569 wherein the agent is delivered from the implant, wherein the agent is released into tissue in the vicinity of the implant after deployment of the implant, wherein the tissue is nerve tissue.
10754. The method of item 10569 wherein the agent is delivered from the implant, wherein the agent is released into tissue in the vicinity of the implant after deployment of the implant, wherein the tissue is epithelium tissue.
10755. The method of item 10569 wherein the agent is delivered from the implant, wherein the agent is released in effective concentrations from the implant over a period ranging from the time of deployment of the implant to about 1 year.
10756. The method of item 10569 wherein the agent is delivered from the implant, wherein the agent is released in effective concentrations from the implant over a period ranging from about 1 month to 6 months.
10757. The method of item 10569 wherein the agent is delivered from the implant, wherein the agent is released in effective concentrations from the implant over a period ranging from about 1 - 90 days.
10758. The method of item 10569 wherein the agent is delivered from the implant, wherein the agent is released in effective concentrations from the implant at a constant rate.
10759. The method of item 10569 wherein the agent is delivered from the implant, wherein the agent is released in effective concentrations from the implant at an increasing rate.
10760. The method of item 10569 wherein the agent is delivered from the implant, wherein the agent is released in effective concentrations from the implant at a decreasing rate.
10761. The method of item 10569 wherein the agent is delivered from the implant, wherein the implant comprises about 0.01 Og to about 10 Dg of the agent.
10762. The method of item 10569 wherein the agent is delivered from the implant, wherein the implant comprises about 10 Dg to about 10 mg of the agent.
10763. The method of item 10569 wherein the agent is delivered from the implant, wherein the implant comprises about 10 mg to about 250 mg of the agent.
10764. The method of item 10569 wherein the agent is delivered from the implant, wherein the implant comprises about 250 mg to about 1000 mg of the agent. 10765. The method of item 10569 wherein the agent is delivered from the implant, wherein the implant comprises about 1000 mg to about 2500 mg of the agent.
10766. The method of item 10569 wherein the agent is delivered from the implant, wherein a surface of the implant comprises less than 0.01 Dg of the agent per mm2 of implant surface to which the agent is applied.
10767. The method of item 10569 wherein the agent is delivered from the implant, wherein a surface of the implant comprises about 0.01 Dg to about 1 Dg of the agent per mm2 of implant surface to which the agent is applied.
10768. The method of item 10569 wherein the agent is delivered from the implant, wherein a surface of the implant comprises about 1 Dg to about 10 Dg of the agent per mm2 of implant surface to which the agent is applied.
10769. The method of item 10569 wherein the agent is delivered from the implant, wherein a surface of the implant comprises about 10 Dg to about 250 Dg of the agent per mm2 of implant surface to which the agent is applied.
10770. The method of item 10569 wherein the agent is delivered from the implant, wherein a surface of the implant comprises about 250 Dg to about 1000 Dg of the agent per mm2 of implant surface to which the agent is applied.
10771. The method of item 10569 wherein the agent is delivered from the implant, wherein a surface of the implant comprises about 1000 Dg to about 2500 Dg of the agent per mm2 of implant surface to which the agent is applied.
10772. The method of item 10569, wherein the implant further comprises a coating, and the coating is a uniform coating.
10773. The method of item 10569, wherein the implant further comprises a coating, and the coating is a non-uniform coating.
10774. The method of item 10569, wherein the implant further comprises a coating, and the coating is a discontinuous coating.
10775. The method of item 10569, wherein the implant further comprises a coating, and the coating is a patterned coating.
10776. The method of item 10569, wherein the implant further comprises a coating, and the coating has a thickness of 100 Dm or less.
10777. The method of item 10569, wherein the implant further comprises a coating, and the coating has a thickness of 10 Dm or less.
10778. The method of item 10569, wherein the implant further comprises a coating, and the coating adheres to the surface of the implant upon deployment of the implant.
10779. The method of item 10569, wherein the implant further comprises a coating, and the coating is stable at room temperature for a period of at least 1 year. 10780. The method of item 10569, wherein the implant further comprises a coating, and the agent is present in the coating in an amount ranging between about 0.0001% to about 1% by weight.
10781. The method of item 10569, wherein the implant further comprises a coating, and the agent is present in the coating in an amount ranging between about 1% to about 10% by weight.
10782. The method of item 10569, wherein the implant further comprises a coating, and the agent is present in the coating in an amount ranging between about 10% to about 25% by weight.
10783. The method of item 10569, wherein the implant further comprises a coating, and the agent is present in the coating in an amount ranging between about 25% to about 70% by weight.
10784. The method of item 10569, wherein the implant further comprises a coating, and the coating comprises a polymer.
10785. The method of item 10569, wherein the implant comprises a first coating having a first composition and a second coating having a second composition.
10786. The method of item 10569, wherein the implant comprises a first coating having a first composition and a second coating having a second composition, wherein the first composition and the second composition are different.
10787. A method as in any one of items 10569-10786, wherein the device is a spinal disc. 10788. A method as in any one of items 10569-10786, wherein the device is a vertebral implant.
10789. A method as in any one of items 10569-10786, wherein the device is a vertebral disc prosthesis.
10790. A method as in any one of items 10569-10786, wherein the device is a lumbar disc implant.
10791. A method as in any one of items 10569-10786, wherein the device is a cervical disc implant.
10792. A method as in any one of items 10569-10786, wherein the device is an intervertebral implant.
10793. A method as in any one of items 10569-10786, wherein the device is a spinal prosthesis.
10794. A method as in any one of items 10569-10786, wherein the device is an artificial disc.
10795. A method as in any one of items 10569-10786, wherein the device is a spinal disc endoprosthesis.
10796. A method as in any one of items 10569-10786, wherein the device is an intervertebral implant.
10797. A method as in any one of items 10569-10786, wherein the device is an implantable spinal graft. 10798. A method as in any one of items 10569-10786, wherein the device is an implantable bone graft.
10799. A method as in any one of items 10569-10786, wherein the device is an artificial lumbar disc.
10800. A method as in any one of items 10569-10786, wherein the device is a spinal nucleus implant.
10801. A method as in any one of items 10569-10786, wherein the device is an intervertebral disc spacer.
10802. A method as in any one of items 10569-10786, wherein the device is a fusion cage.
10803. A method as in any one of items 10569-10786, wherein the device is a fusion basket.
10804. A method as in any one of items 10569-10786, wherein the device is a fusion cage apparatus.
10805. A method as in any one of items 10569-10786, wherein the device is an interbody cage.
10806. A method as in any one of items 10569-10786, wherein the device is an interbody implant.
10807. A method as in any one of items 10569-10786, wherein the device is a fusion cage anchoring device. 10808. A method as in any one of items 10569-10786, wherein the device is a bone fixation apparatus.
10809. A method as in any one of items 10569-10786, wherein the device is a fusion stabilization chamber.
10810. A method as in any one of items 10569-10786, wherein the device is an anchoring bone plate.
10811. A method as in any one of items 10569-10786, wherein the device is a bone screw.
10812. A method for inhibiting scarring comprising placing a device that comprises an implant that provides a surgical adhesion barrier and an anti-scarring agent or a composition comprising an anti- scarring agent into an animal host, wherein the agent inhibits scarring.
10813. The method of item 10812 wherein the agent is an adensosine A2A receptor antagonist.
10814. The method of item 10812 wherein the agent is an AKT inhibitor.
10815. The method of item 10812 wherein the agent is an alpha 2 integrin antagonist, wherein the alpha 2 integrin antagonist is Pharmaprojects No. 5754 (Merck KgaA).
10816. The method of item 10812 wherein the agent is an alpha 4 integrin antagonist. 10817. The method of item 10812 wherein the agent is an alpha 7 nicotinic receptor agonist.
10818. The method of item 10812 wherein the agent is an angiogenesis inhibitor selected from the group consisting of AG-12,958 (Pfizer), ATN-161 (Attenuon LLC), neovastat, an angiogenesis inhibitor from Jerina AG (Germany), NM-3 (Mercian), VGA-1155 (Taisho), FCE-26644 (Pfizer), FCE-26950 (Pfizer), FPMA (Meiji Daries), FR-111142 (Fujisawa), GGTI-298, GM-1306 (Ligand), GPA-1734 (Novartis), NNC-47-0011 (Novo Nordisk), herbamycin (Nippon Kayaku), lenalidomide (Celegene), IP-10 (NIH), ABT-828 (Abbott), KIN-841 (Tokushima University, Japan), SF-1126 (Semafore Pharmaceuticals), laminin technology (NIH), CHIR-258 (Chiron), NVP-AEW541 (Novartis), NVP-AEW541 (Novartis), Vt16907 (Alchemia), OXI-8007 (Oxigene), EG-3306 (Ark Therapeutics), Maspin (Arriva), ABT- 567 (Abbott), PPI-2458 (Praecis Pharmaceuticals), CC-5079, CC-4089 (Celgene), HIF-1 alpha inhibitors (Xenova), S-247 (Pfizer), AP-23573 (Ariad), AZD-9935 (Astra Zeneca), mebendazole (Introgen Therapeutics), MetAP-2 inhibitors (GlaxoSmithKline), AG-615 (Angiogene Pharmaceuticals), Tie-2 antagonists (Hybrigenics), NC-381 , CYC-381, NC-169, NC-219, NC-383, NC-384, NC-407 (Lorus Therapeutics), ATN-224 (Attenuon), ON-01370 (Onconova), Vitronectin antagonists (Amgen), SDX-103 (Salmedix), Vitronectin antagonists (Shire), CHP (Riemser), TEK (Amgen), Anecortave acetate (Alcon), T46.2 (Matrix Therapeutics), HG-2 (Heptagen), TEM antagonists (Genzyme), Oxi-4500 (Oxigene), ATN-161 (Attenuon), WX-293 (Wilex), M-2025 (Metris Therapeutics), Alphastatin (BioActa), YH-16 (Yantai Rongchang), BIBF-1120 (Boehringer Ingelheim), BAY-57-9352 (Bayer), AS- 1404 (Cancer Research Technology), SC-77964 (Pfizer), glycomimetics (BioTie Therapies), TIE-2 Inhibitors (Ontogen), DIMI, Octamer (Octamer), ABR-215050 (Active Biotech), ABT-518 (Abbott), KDR inhibitors (Abbott), BSF-466895 (Abbott), SCH-221153 (Schering-Plough), DAC:antiangiogenic (ConjuChem), TFPI (EntreMed), AZD-2171 (Astra-Zenaca), CDC-394 (Celgene), LY290293 (EIi Lilly), IDN-5390 (Indena), Kdr Kinase Inhibitors (Merck), CT-113020, CT-116433, CT-116563, CT-31890, CT-32228) (Cell Therapeutics), A-299620 (Abbott), TWEAK Inhibitor (Amgen), VEGF modulators (Johnson and Johnson), Tum-N53, tumstatin (Genzyme), Thios- 1 , Thios-2 (Thios Pharmaceuticals), MV-6401 (Miravant Medical Technologies), Spisulosine (PharmaMar), CEP-7055 (Cephalon), AUV-201 (Auvation), LM-609 (EIi Lilly), SKF-106615 (AnorMED), Oglufanide disodium (Cytran), BW-114 (Pharminox), Calreticulin (NIH), WX-678 (Wilex), SD-7784 (Pfizer), WX-UK1 (Wilex), SH-268 (Schering AG), 2-Me-PGA (Celgene), S- 137 (Pfizer), ZD-6126 (Angiogene Pharmaceuticals), SG-292 (SignalGen), Benefin (Lane Labs), A6, A36 (Angstrom), SB-2723005 (GlaxoSmithKline), SC-7 (Cell Therapeutics), ZEN-014 (AEterna Zentaris), 2-methoxyestradiol (EntreMed), NK-130119 (Nippon Kayaku), CC-10004 (Celgene), AVE- 8062A (Ajinomoto), Tacedinaline (Pfizer), Actinonin (Tokyo Metropolitan Institute of Medical Science), Lenalidomide (Celgene), VGA-1155, BTO-956 (SRI International), ER-68203-00 (Eisai), CT-6685 (UCB), JKC-362 (Phoenix Pharmaceuticals), DMI-3798 (DMI Biosciences, Angiomate (Ipsen), ZD-6474 (AstraZeneca), CEP-5214 (Cephalon), Canstatin (Genzyme), NM-3 (Mercian), Oridigm (MediQuest Therapeutics), Exherin (Adherex), BLS-0597 (Boston Life Sciences), PTC-299 (PTC Therapeutics), NPI-2358 (Nereus Pharmaceuticals), CGP-79787 (Novartis), AEE-788 (Novartis), CKD-732 (Chong Kun Dang), CP-564959 (OSI Pharmaceuticals), CM-101 (CarboMed), CT-2584, CT3501 (Cell Therapeutics), combretastatin and analogues and derivatives thereof (Oxigene), Rebimastat (Bristol-Meyers Squibb), Dextrin 2-sulfate (ML Laboratories), Cilengitide (Merk KGaA), NSC-706704 (Pharminox), KRN- 951 (Kirin Brewery), Ukrain, NSC-631570 (Nowicky Pharma), Tecogalan sodium (Daiichi Pharmaceutical), Tz-93 (Tsumura), TBC-1635 (Encysive Pharmaceuticals), TAN-1120 (Takeda), Semaxanib (Pfizer), BDI-7800 (Biopharmacopae), SD-186, SD-983 (Bristol-Meyers Squibb), SB-223245 (GlaxoSmithKline), SC-236 (Pfizer), RWJ-590973 (Johnson and Johnson), ILX-1850 (Genzyme), SC-68488, S-836 (Pfizer), CG-55069-11 (CuraGen), Ki-23057 (Kirin Brewery), CCX-700 (Chemoentryx), Pegaptanib octasodium (Giled Sciences), ANGIOCOL (available from Biostratum Inc.), or an analogue or derivative thereof.
10819. The method of item 10812 wherein the agent is an apoptosis antagonist.
10820. The method of item 10812 wherein the agent is an apoptosis activator.
10821. The method of item 10812 wherein the agent is a beta 1 integrin antagonist.
10822. The method of item 10812 wherein the agent is a beta tubulin inhibitor.
10823. The method of item 10812 wherein the agent is a blocker of enzyme production in Hepatitis C.
10824. The method of item 10812 wherein the agent is a Bruton's tyrosine kinase inhibitor.
10825. The method of item 10812 wherein the agent is a calcineurin inhibitor.
10826. The method of item 10812 wherein the agent is a caspase 3 inhibitor.
10827. The method of item 10812 wherein the agent is a CC chemokine receptor antagonist. 10828. The method of item 10812 wherein the agent is a cell cycle inhibitor selected from the group consisting of SNS-595 (Sunesis), synthadotin, KRX-0403, and an analogue or derivative thereof.
10829. The method of item 10812 wherein the agent is a cathepsin B inhibitor.
10830. The method of item 10812 wherein the agent is a cathepsin K inhibitor, wherein the cathepsin K inhibitor is 462795 (GlaxoSmithKline), INPL-022-D6 (Amura Therapeutics), or an analogue or derivative thereof.
10831. The method of item 10812 wherein the agent is a cathepsin L inhibitor.
10832. The method of item 10812 wherein the agent is a CD40 antagonist.
10833. The method of item 10812 wherein the agent is a chemokine receptor agonist.
10834. The method of item 10812 wherein the agent is a chymase inhibitor.
10835. The method of item 10812 wherein the agent is a collagenase antagonist.
10836. The method of item 10812 wherein the agent is a CXCR antagonist. 10837. The method of item 10812 wherein the agent is a cyclin dependent kinase inhibitor selected from the group consisting of a CDK-1 inhibitor, a CDK-2 inhibitor, a CDK- 4 inhibitor, a CDK-6 inhibitor, a CAK1 inhibitor from GPC Biotech and Bristol-Myers Squibb, RGB-286199 (GPC Biotech), an anticancer agent from Astex Technology, a CAK1 inhibitor from GPC Biotech, a CDK inhibitor from Sanofi-Aventis, a CDK1/CDK2 inhibitor from Amgen, a CDK2 inhibitor from SUGEN-2 (Pfizer), a hearing loss therapy agent (Sound Pharmaceuticals), PD-0332991 (Pfizer), RGB-286199 (GPC Biotech), Ro-0505124 (Hoffmann-La Roche), a Ser/Thr kinase inhibitor from Lilly (EIi Lilly), CVT-2584 (CAS No. 199986-75- 9) (CV Therapeutics), and an analogue or derivative thereof.
10838. The method of item 10812 wherein the agent is a cyclooxygenase 1 inhibitor.
10839. The method of item 10812 wherein the agent is a DHFR inhibitor.
10840. The method of item 10812 wherein the agent is a dual integrin inhibitor.
10841. The method of item 10812 wherein the agent is an elastase inhibitor.
10842. The method of item 10812 wherein the agent is an elongation factor-1 alpha inhibitor.
10843. The method of item 10812 wherein the agent is an endothelial growth factor antagonist. 10844. The method of item 10812 wherein the agent is an endothelial growth factor receptor kinase inhibitor selected from the group consisting of sorafenib tosylate (Bayer), AAL-993 (Novartis), ABP-309 (Novartis), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), E- 7080 (Eisai), EG-3306 (Ark Therapeutics), EXEL-2880 (Exelixis), GW- 654652 (GlaxoSmithKline), a KDR inhibitor from LG Life Sciences, CT-6685 and CT-6729 (UCB), KRN-633 and KRN-951 (Kirin Brewery), OSI-930 (OSI Pharmaceuticals), SP-5.2 (Supratek Pharma), SU-11657 (Pfizer), a Tie-2 antagonist (Hybrigenics), a VEGFR-2 kinase inhibitor (Bristol-Myers Squibb), XL-647 (Exelixis), a KDR inhibitor from Abbott Laboratories, sorafenib tosylate, and an analogue or derivative thereof.
10845. The method of item 10812 wherein the agent is an endotoxin antagonist.
10846. The method of item 10812 wherein the agent is an epothilone and tubulin binder.
10847. The method of item 10812 wherein the agent is an estrogen receptor antagonist.
10848. The method of item 10812 wherein the agent is an FGF inhibitor.
10849. The method of item 10812 wherein the agent is a farnexyl transferase inhibitor.
10850. The method of item 10812 wherein the agent is famesyltransferase inhibitor selected from the group of A-197574 (Abbott), a farnesyltransferase inhibitor from Servier, FPTIII (Strathclyde Institute for Drug R), LB-42908 (LG Life Sciences), Pharmaprojects No. 5063 (Genzyme), Pharmaprojects No. 5597 (Ipsen), Yissum Project No. B-1055 (Yissum), and an analogue or derivative thereof.
10851. The method of item 10812 wherein the agent is an FLT-3 kinase inhibitor.
10852. The method of item 10812 wherein the agent is an FGF receptor kinase inhibitor.
10853. The method of item 10812 wherein the agent is a fibrinogen antagonist selected from the group consisting of AUV-201 (Auvation), MG-13926 (Sanofi-Aventis), plasminogen activator (CAS No. 105913-11-9) (from Sanofi-Aventis or UCB), plasminogen activator-2 (tPA- 2) (Sanofi-Aventis), pro-urokinase (CAS No. 82657-92-9) (Sanofi-Aventis), and an analogue or derivative thereof.
10854. The method of item 10812 wherein the agent is a heat shock protein 90 antagonist selected from the group consisting of SRN- 005 (Sirenade), geldanamycin, NSC-33050 (17-allylaminogeldanamycin; 17- AAG), 17-dimethylaminoethylamino-17-demethoxy-geldanamycin (17- DMAG), rifabutin (rifamycin XIV, 1',4-didehydro-1-deoxy-1 ,4-dihydro-5'-(2- methylpropyl)-1~oxo-), and an analogue or derivative thereof.
10855. The method of item 10812 wherein the agent is a histone deacetylase inhibitor.
10856. The method of item 10812 wherein the agent is an HMGCoA reductase inhibitor selected from the group consisting of an atherosclerosis therapeutic from Lipid Sciences, ATM 6000 (ARYx Therapeutics), KS-01-019 (Kos Pharmaceuticals), Pharmaprojects No. 2197 (Sanofi-Aventi), RP 61969 (Sanofi-Aventis), and an analogue or derivative thereof.
10857. The method of item 10812 wherein the agent is an ICAM inhibitor.
10858. The method of item 10812 wherein the agent is an IL, ICE and IRAK antagonist, wherein the antagonist is a CJ-14877, CP- 424174 (Pfizer), NF-61 (Negma-Lerads), and an analogue or derivative thereof.
10859. The method of item 10812 wherein the agent is an IL-2 inhibitor.
10860. The method of item 10812 wherein the agent is an immunosuppressant selected from the group consisting of terifiunomide (Sanofi Aventis), chlorsulfaquinoxalone (NSC-339004), chlorsulfaquinoxalone sulfate, CS-712 (Sankyo), ismomultin alfa (CAS No. 457913-93-8) (Akzo Nobel), antiallergics from GenPat77, antiinflammatories or AT-005 (Androclus Therapeutics), autoimmune disease therapeutics from EpiVax, BN-007 (Bone), budesonide (CAS No. 51333-22- 3) (MAP Pharmaceuticals), CO-14 (Genzyme), edratide (CAS No. 433922- 67-9) (Teva), EP-314 (Enanta), eprovafen (CAS No. 101335-99-3) (Sanofi- Aventis), HWA-131 (CAS No. 118788-41-3) (Sanofi-Aventis), immunomodulators from MerLion Pharmaceuticals, immunosuppressives from Alchemia, IPL-12 (Inflazyme), MDL-9563 (CAS No. 27086-86-8) (Sanofi-Aventis), Pharmaprojects No. 2330 (Sanofi-Aventis), Pharmaprojects No. 6426 (Abgenix), PXS-25 (Pharmaxis), rosmarinic acid (CAS No. 20283-92-5) (Sanofi-Aventis), RP 42927 or RP 54745 (CAS No. 135330-08-4) (Sanofi-Aventis), SGN-35 (Seattle Genetics), ST-1959 (Sigma-Tau), type I diabetes therapy from SYNX Pharma, UNIL-88 (Debiopharm), VP-025 (Vasogen), VR-694 (Vectura), PRTX-001 (Protalex), and an analogue or derivative thereof.
10861. The method of item 10812 wherein the agent is an IMPDH (inosine monophosphate).
10862. The method of item 10812 wherein the agent is an integrin antagonist.
10863. The method of item 10812 wherein the agent is an interleukin antagonist.
10864. The method of item 10812 wherein the agent is an inhibitor of type III receptor tyrosine kinase.
10865. The method of item 10812 wherein the agent is an irreversible inhibitor of enzyme methionine aminopeptidase type 2.
10866. The method of item 10812 wherein the agent is an isozyme selective delta protein kinase C inhibitor.
10867. The method of item 10812 wherein the agent a JAK3 enzyme inhibitor.
10868. The method of item 10812 wherein the agent is a JNK inhibitor.
10869. The method of item 10812 wherein the agent is a kinase inhibitor. 10870. The method of item 10812 wherein the agent is kinesin antagonist.
10871. The method of item 10812 wherein the agent is a kinesin antagonist.
10872. The method of item 10812 wherein the agent is a leukotriene inhibitor and antagonist selected from the group consisting of ambicromil (CAS No. 58805-38-2) (Sanofi-Aventis), amelubant (CAS No. 346735-24-8) (Boehringer Ingelheim), DW-1141 (Dong Wha), ebselen (Daiichi Pharmaceutical), ibudilast (Kyorin), leucotriene inhibitors from Sanofi-Aventis, lymphotoxin-beta receptor (LT-D) from Biogen Idee, Pharmaprojects No. 1535 and 2728 (CAS No. 119340-33-9) (Sanofi- Aventis), R-112 (Rigel), Rev-5367 (CAS No. 92532-05-3) (Sanofi-Aventis), RG-14893 (CAS No. 141835-49-6) (Sanofi-Aventis), RG-5901-A (CAS No. 101910-24-1), 92532-23-5, RP 66153 (CAS No. 142422-79-5), RP 66364 (CAS No.186916-92-5), RP 69698 (CAS No. 141748-00-7) (Sanofi-Aventis), SC-411930 (Pfizer), SC-41930 (CAS No. 120072-59-5) (Pfizer), SC-50605 (CAS No. 138828-39-4) (Pfizer), SC-51146 (CAS No. 141059-52-1), SC- 53228 (CAS No. 153633-01-3) (Pfizer), spaglumic acid (ZY-15106) (CAS No.3106-85-2), 80619-64-3 (Novartis), tipredane (CAS No. 85197-77-9) (Bristol-Myers Squibb), U-75302 (CAS No. 119477-85-9) (Pfizer), and analogue or derivative thereof.
10873. The method of item 10812 wherein the agent is an MAP kinase inhibitor.
10874. The method of item 10812 wherein the agent is a matrix metalloproteinase inhibitor. 10875. The method of item 10812 wherein the agent is an MCP-CCR2 inhibitor.
10876. The method of item 10812 wherein the agent is an mTOR inhibitor.
10877. The method of item 10812 wherein the agent is an mTOR kinase inhibitor.
10878. The method of item 10812 wherein the agent is a microtubule inhibitor selected from the group consisting of antibody- maytansinoid conjugates from Biogen Idee, colchicines (MantiCore Pharmaceuticals), anticancer immunoconjugates from Johnson & Johnson, DIME from Octamer, gni-1f (GNI)1 huC242-DM4, huMy9-6-DM1 (ImmunoGen), IDN-5404 (Indena), IMO-098, IMOderm (Imotep), mebendazole (Introgen Therapeutics), microtubule poisons from Cambridge Enterprise, paclitaxel such as LOTAX from Aphios (CAS No. 33069-62-4), Genexol-PM from Samyang, Pharmaprojects No. 6383 (Tapestry Pharmaceuticals), RPR-112378 (Sanofi-Aventis), SGN-75 (Seattle Genetics), SPL-7435 (Starpharma), SSR-250411 (Sanofi-Aventis), trastuzumab-DM1 (Genentech), vinorelbine, and an analogue or derivative thereof.
10879. The method of item 10812 wherein the agent is an MIF inhibitor.
10880. The method of item 10812 wherein the agent is an MMP inhibitor.
10881. The method of item 10812 wherein the agent is a neurokinin (NK) antagonist selected from the group consisting of anthrotainin (CAS No. 148084-40-6) (Sanofi-Aventis), an IBS thereapeutic from ArQuIe, MDL-105212A (CAS No. 167261-60-1) (Ssanofi-Aventis), Pharmaprojects No. 2744, 3258 (CAS No. 139167-47-8) 4006, 4201 , or 5986 (Sanofi-Aventis), RP 67580 (CAS No. 135911-02-3), SR-144190 (CAS No. 201152-86-5), SSR-240600, SSR-241586 (Sanofi-Aventis), TKA-457 (Novartis), vestipitant mesylate (CAS No. 334476-64-1) (GlaxoSmithKline), Win-64821 (Sanofi-Aventis), PRX-96026 (Predix Pharmaceuticals), and an analogue or derivative thereof.
10882. The method of item 10812 wherein the agent is an NF kappa B inhibitor selected from the group consisting of AVE-0545 or AVE-0547 (Sanofi-Aventis), bortezomib (CAS No. 179324-69-7) (Millennium Pharmaceuticals), dexanabinol (CAS No. 112924-45-5) (Pharmos), dexlipotam (Viatris), Pharmaprojects No. 6283 (INDRA) (OXiGENE), IPL- 576092 (CAS No. 137571-30-3) (Inflazyme), NFKB decoy (Corgentech), NFKB decoy oligo (AnGes MG), NFKB's from Ariad, osteoporosis treatments or S5 (F005) from Fulcrum Pharmaceuticals, P61 (Phytopharm), R-flurbiprofen (CAS No. 5104-49-4) (Encore Pharmaceuticals), and an analogue or derivative thereof.
10883. The method of item 10812 wherein the agent is a nitric oxide agonist.
10884. The method of item 10812 wherein the agent is an ornithine decarboxylase inhibitor.
10885. The method of item 10812 wherein the agent is a p38 MAP kinase inhibitor selected from the group consisting of AZD-6703 (AstraZeneca), JX-401 (Jexys Pharmaceuticals), BMS-2 (Bristol-Myers Squibb), a p38 MAP kinase inhibitor from Novartis, a p38-alpha MAP kinase inhibitor from Amphora, Pharmaprojects No. 5704 (Pharmacopeia), RPR- 200765A (Sanofi-Aventis), SD-282 (Johnson & Johnson), TAK-715 (Takeda), and an analogue or derivative thereof.
10886. The method of item 10812 wherein the agent is a palmitoyl-protein thioesterase inhibitor.
10887. The method of item 10812 wherein the agent is a PDGF receptor kinase inhibitor selected from the group consisting of AAL- 993, AMN-107, or ABP-309 (Novartis), AMG-706 (Amgen), BAY-57-9352 (Bayer), CDP-860 (UCB), E-7080 (Eisai), imatinib (CAS No. 152459-95-5) (Novartis), OSl-930 (OSI Pharmaceuticals), RPR-127963E (Sanofi-Aventis), RWJ-540973 (Johnson & Johnson), sorafenib tosylate (Bayer), SU-11657 (Pfizer), tandutinib (CAS No. 387867-13-2) (Millennium Pharmaceuticals), vatalanib (Novartis), ZK-CDK (Schering AG), and an analogue or derivative thereof.
10888. The method of item 10812 wherein the agent is (-)-halofenate (Metabolex), AMG-131 (Amgen), antidiabetics from Japan Tobacco, AZD-4619, AZD-8450, AZD-8677 (AstraZeneca), DRF-10945, balagiitazone (Dr Reddy's), CS-00088, CS-00098 (Chipscreen Biosciences), E-3030 (Eisai), etalocib (CAS No. 161172-51-6) (EIi Lilly), GSK-641597 (Ligand), GSK-677954 (GlaxoSmithKline), GW-409544 (Ligand), GW- 590735 (GlaxoSmithKline), K-111 (Hoffmann-La Roche), LY-518674 (EIi Lilly), LY-674 (Ligand), LY-929 (Ligand), MC-3001, MC-3002 (MaxoCore Pharmaceuticals), metformin HCI + pioglitazone (CAS No. 1115-70-4 and 112529-15-4), ACTOPLUS MET from Andrx), muraglitazar (CAS No. 331741-94-7) (Bristol-Myers Squibb), naveglitazar (Ligand), oleoylethanolamide (Kadmus Pharmaceuticals), ONO-5129, pioglitazone hydrochloride (CAS No. 111025-46-8 and 112529-15-4) (Takeda), PLX-204 (Plexxikon), PPAR agonists from Genfit, PPAR delta agonists from EIi Lilly, PPAR-alpha agonists from CrystalGenomics, PPAR-gamma modulators and PPAR-β modulators from CareX, rosiglitazone maleate (CAS No. 122324- 73-4 or 155141-29-0) (GlaxoSmithKline), rosiglitazone maleate/glimepir (CAS No. 155141-29-0 and 93479-97-1), AVANDARYL, rosiglitazone maleate/metformin extend (CAS No. 155141-29-0 and 657-24-9), AVANDAMET, rosiglitazone maleate+metformin, AVANDAMET (GlaxoSmithKline), tesaglitazar (AstraZeneca), and an analogue or derivative thereof.
10889. The method of item 10812 wherein the agent is a phosphatase inhibitor.
10890. The method of item 10812 wherein the agent is a phosphodiesterase (PDE) inhibitor selected from the group consisting of avanafil (Tanabe Seiyaku), dasantafil (CAS No. 569351-91-3) (Schering- Plough), A-906119 (CAS No. 134072-58-5), DL-850 (Sanofi-Aventis), GRC- 3015, GRC-3566, GRC-3886 (Glenmark), HWA-153 (CAS No. 56395-66-5) (Sanofi-Aventis), hydroxypumafentrine (Altana), IBFB-130011 , IBFB-14-016, IBFB-140301 , IBFB-150007, IBFB-211913 (IBFB Pharma), L-826141 (Merck & Co), medorinone (CAS No. 88296-61-1) (Sanofi-Aventis), MEM- 1917 (Memory Pharmaceuticals), ND-1251 (Neuro3d), PDE inhibitors from ICOS, PDE IV inhibitors from Memory Pharmaceuticals and CrystalGenomics, Pharmaprojects No. 2742 and 6141 (Sanofi-Aventis), QAD-171 (Novartis), RHC-2963 (CAS No. 76993-12-9 and 76993-14-1), RPR-117658, RPR-122818 derivatives, SR-24870 , and RPR-132294 (Sanofi-Aventis), SK-350 (ln2Gen), stroke therapy agents from deCODE Genetics, TAS-203 (Taiho), tofimilast (CAS No. 185954-27-2) (Pfizer), UK- 371800 (Pfizer), WIN-65579 (CAS No. 158020-82-7) (Sanofi-Aventis), IBFB- 130020 (IBFB Pharma), OPC-6535 (CAS No. 145739-56-6) (Otsuka), a phosphodiesterase III inhibitor, enoximone, a phosphodiesterase IV inhibitor, fosfosal, Atopik (Barrier Therapeutics), triflusal, a phosphodiesterase V inhibitor, and an analogue or derivative thereof. 10891. The method of item 10812 wherein the agent is a PKC inhibitor.
10892. The method of item 10812 wherein the agent is a platelet activating factor antagonist.
10893. The method of item 10812 wherein the agent is a platelet-derived growth factor receptor kinase inhibitor.
10894. The method of item 10812 wherein the agent is a prolyl hydroxylase inhibitor.
10895. The method of item 10812 wherein the agent is a polymorphonuclear neutrophil inhibitor.
10896. The method of item 10812 wherein the agent is a protein kinase B inhibitor.
10897. The method of item 10812 wherein the agent is a protein kinase C stimulant.
10898. The method of item 10812 wherein the agent is a purine nucleoside analogue.
10899. The method of item 10812 wherein the agent is a purinoreceptor P2X antagonist.
10900. The method of item 10812 wherein the agent is a Raf kinase inhibitor. 10901. The method of item 10812 wherein the agent is a reversible inhibitor of ErbB1 and ErbB2.
10902. The method of item 10812 wherein the agent is a ribonucleoside triphosphate reductase inhibitor.
10903. The method of item 10812 wherein the agent is an SDF-1 antagonist.
10904. The method of item 10812 wherein the agent is a sheddase inhibitor.
10905. The method of item 10812 wherein the agent is an SRC inhibitor.
10906. The method of item 10812 wherein the agent is a stromelysin inhibitor.
10907. The method of item 10812 wherein the agent is an Syk kinase inhibitor.
10908. The method of item 10812 wherein" the agent is a telomerase inhibitor.
10909. The method of item 10812 wherein the agent is a TGF beta inhibitor selected from the group consisting of pirfenidone (CAS No. 53179-13-8) (MARNAC), tranilast (CAS No. 53902-12-8) (Kissei), IN- 1130 (ln2Gen), mannose-6-phosphate (BTG), TGF-β antagonists from Inflazyme (Pharmaprojects No. 6075), TGF-β antagonists from Sydney, non-industrial source), TGF-β I receptor kinase inhibitors from EIi Lilly, TGF- β receptor inhibitors from Johnson & Johnson, and an analogue or derivative thereof.
10910. The method of item 10812 wherein the agent is a
TNFα antagonist or TACE inhibitor selected from the group consisting of adalimumab (CAS No. 331731-18-1) (Cambridge Antibody Technology), AGIX-4207 (AtheroGenics), AGT-1 (Advanced Biotherapy), an antiinflammatory from Borean Pharma, Cellzome, or Paradigm Therapeutics, anti-inflammatory vaccine (TNF-alpha kinoid) from Neovacs, humanized anti-TNF antibody or an anti-TNF MAb (CB0006) Celltech (UCB), apratastat (CAS No. 287445-51-0) (Wyeth), BMS-561392 (Bristol-Myers Squibb), BN- 006 (Bone), certolizumab pegol (CAS No. 428863-50-7 or CH-138 (UCB), cilomilast (CAS No. 153259-65-5) (GlaxoSmithKline), CR-1 (Nuada Pharmaceuticals), CRx-119 (CombinatoRx), D-5410 (UCB), dacopafant (CAS No. 125372-33-0) (Sanofi-Aventis), dersalazine (CAS No. 188913-57- 7/188913-58-8) (Uriach), etanercept (CAS No. 185243-69-0) (Amgen), ethyl pyruvate (Critical (Critical Therapeutics), golimumab (CAS No. 476181-74-5) (Johnson & Johnson), hormono-immunotherapy from Ipsen, CDP571 (e.g., Humicade from UCB), IC-485 (ICOS), infliximab (CAS No. 170277-31-3) (Johnson & Johnson), IP-751 (Manhattan Pharmaceuticals), ISIS-104838 (CAS No. 250755-32-9) (ISIS Pharmaceuticals), lenalidomide (CAS No. 191732-72-6) (Celgene), lentinan (CAS No. 37339-90-5) (Ajinomoto), MDL- 201112 (CAS No. 142130-73-2) (Sanofi-Aventis), medroxyprogesterone (CAS No. 520-85-4) (InKine Pharmaceutical), N-acetylcysteine (CAS No. 616-91-1) (Zambon), NBE-P2 (DIREVO Biotech), nerelimomab (CAS No. 162774-06-3) (Chiron), OM-294DP (OM PHARMA), onercept (CAS No. 199685-57-9) (Yeda), PASSTNF-alpha (Verigen), pentoxifylline or oxypentifylline (Sanofi-Aventis), Pharmaprojects No. 4091, 4241, 4295, or 4488 (Sanofi-Aventis), Pharmaprojects No. 5480 (Amgen), Pharmaprojects No. 6749 (Cengent), pirfenidone (CAS No. 53179-13-8) (MARNAC), RPR- 132294 (Sanofi-Aventis), S5 (F002) (Fulcrum Pharmaceuticals), simvastatin (CAS No. 79902-63-9) (Merck & Co), STA-6292 (Synta Pharmaceuticals), tacrolimus (CAS No. 104987-11-3) (Fujisawa LifeCycle Pharma), talactoferrin alfa (CAS No. 308240-58-6) (Agennix), thalidomide (CAS No. 50-35-1) (Celgene), TNF antagonists form ProStrakan, and Synergen, TNF inhibitors (Amgen), TNF-alpha antagonists from Dynavax Technologies and Jerina AG (Germany), TNF-alpha inhibitors from IBFB Pharma and Xencor (Xencor), torbafylline (CAS No. 105102-21-4) (Sanofi-Aventis), UR-1505 (Uriach), VT-346 (Viron Therapeutics), YSIL6 (Y's Therapeutics), YSTH2 (Y's Therapeutics), NPI-1302a-3 (Nereus Pharmaceuticals, a TNF antagonist from Jerina AG (Germany), dersalazine, and an analogue or derivative thereof.
10911. The method of item 10812 wherein the agent is a Toll receptor inhibitor.
10912. The method of item 10812 wherein the agent is a tubulin antagonist.
10913. The method of item 10812 wherein the agent is a tyrosine kinase inhibitor selected from the group consisting of SU-011248, SUTENT from Pfizer Inc. (New York, NY), BMS-354825, PN-355 (Paracelsian Pharmaceuticals), AGN-199659 (Allergan), AAL-993 or ABP- 309 (Novartis), adaphostin (NlH), AEE-788 (Novartis), AG-013736 (OSI Pharmaceuticals), AG-13736 (Pfizer), ALT-110 (Alteris Therapeutics), AMG- 706 (Amgen), anticancer MAbs from Xencor, anti-EGFrvlll MAbs from Abgenix, anti-HER2 MAb from Abiogen, AZD-2171 or AZD-9935 (AstraZeneca), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), CEP-5214 (Cephalon), CEP-7055 (Cephalon), cetuximab (ImClone Systems), CHIR-200131 and CHIR-258 (Chiron), CP-547632 (OSI Pharmaceuticals), CP-724714 (Pfizer), CT-301 (Creabilis Therapeutics), D- 69491 (Baxter International), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EGFR/ErbB2 inhibitors from Array BioPharma, erlotinib (CAS No. 183319- 69-9) (OSI Pharmaceuticals), EXEL-2880 (Exelixis), FK-778 (Sanofi- Aventis) , gefitinib (CAS No. 184475-35-2) (AstraZeneca), GW-2286 or GW- 654652 (GlaxoSmithKline), her2/neu antigen from AlphaVax, HER-2/neu inhibitor from Generex, Herzyme (Medipad) (Sirna Therapeutics), HKI-272 (Wyeth), HuMax-EGFr (Genmab), idronoxil (CAS No. 81267-65-4) (Novogen), IGF-1 inhibitors from Ontogen, IMC-11F8 (ImCIone Systems), kahalalide F (CAS No. 149204-42-2) (PharmaMar), KDR inhibitor from LG Life Sciences, KDR inhibitors from Abbott Laboratories, KDR kinase inhibitors (UCB), Kdr kinase inhibitors from Merck & Co, KRN-633 and KRN- 951 (Kirin Brewery), KSB-102 (Xenova), lapatinib ditosylate (CAS No. 388082-78-8) (GlaxoSmithKline), matuzumab (Merck KGaA), MDX-214 (Medarex), ME-103 (Pharmexa), MED-A300 (Gerolymatos), MNAC-13 (Lay Line Genomics), nimotuzumab (Center of Molecular Immunology), NSC- 330507 or NSC-707545 (NIH), NV-50 (Novogen), OSI-930 (OSI Pharmaceuticals), panitumumab (Abgenix), pelitinib (CAS No. 287933-82-7) (Wyeth), pertuzumab (CAS No. 380610-27-5) (Genentech), Pharmaprojects No. 3985 (Sanofi-Aventis), prostate cancer therapeutics from Sequenom (SQPC35, SQPC36, SQPC90), removab and remoxab (Trion Pharma), RG- 13022 (CAS No. 136831-48-6), RG-13291 (CAS No. 138989-50-1), or RG- 14620 (CAS No. 136831-49-7) (Sanofi-Aventis), RM-6427 (Romark), RNAi breast cancer therapy from Benitec, RP 53801 (CAS No. 125882-88-4) (Sanofi-Aventis), sorafenib tosylate (Bayer), SU-11657 (Pfizer), Tie-2 antagonists from Semaia (Hybrigenics), Tie-2 inhibitors from Ontogen, trastuzumab (CAS No. 180288-69-1) (Genentech), tyrosine kinase inhibitors from Sanofi-Aventis, U3-1287, U3-1565, U3-1784, U3-1800 (U3 Pharma), vatalanib (Novartis), VEGFR-2 kinase inhibitor from Bristol-Myers Squibb, XL-647 (Exelixis), ZD-6474 (AstraZeneca), ZK-CDK (Schering AG), an EGFR tyrosine kinase inhibitor, EKB-569 (Wyeth), and an analogue or derivative thereof. 10914. The method of item 10812 wherein the agent is a VEGF inhibitor.
10915. The method of item 10812 wherein the agent is a vitamin D receptor agonist.
10916. The method of item 10812 wherein the agent is ZD-6474 (an angiogenesis inhibitor).
10917. The method of item 10812 wherein the agent is AP-23573 (an mTOR inhibitor).
10918. The method of item 10812 wherein the agent is synthadotin (a tubulin antagonist).
10919. The method of item 10812 wherein the agent is S-0885 (a collagenase inhibitor).
10920. The method of item 10812 wherein the agent is aplidine (an elongation factor-1 alpha inhibitor).
10921. The method of item 10812 wherein the agent is ixabepilone (an epithilone).
10922. The method of item 10812 wherein the agent is lDN-5390 (an angiogenesis inhibitor).
10923. The method of item 10812 wherein the agent is SB-2723005 (an angiogenesis inhibitor). 10924. The method of item 10812 wherein the agent is ABT-518 (an angiogenesis inhibitor).
10925. The method of item 10812 wherein the agent is combretastatin (an angiogenesis inhibitor).
10926. The method of item 10812 wherein the agent is anecortave acetate (an angiogenesis inhibitor).
10927. The method of item 10812 wherein the agent is SB-715992 (a kinesin antagonist).
10928. The method of item 10812 wherein the agent is temsirolimus (an mTOR inhibitor).
10929. The method of item 10812 wherein the agent is adalimumab (a TNFα antagonist).
10930. The method of item 10812, wherein the composition comprises a polymer.
10931. The method of item 10812, wherein the composition comprises a polymer, and the polymer is, or comprises, a copolymer.
10932. The method of item 10812, wherein the composition comprises a polymer, and the polymer is, or comprises, a block copolymer. 10933. The method of item 10812, wherein the composition comprises a polymer, and the polymer is, or comprises, a random copolymer.
10934. The method of item 10812, wherein the composition comprises a polymer, and the polymer is, or comprises, a biodegradable polymer.
10935. The method of item 10812, wherein the composition comprises a polymer, and the polymer is, or comprises, a nonbiodegradable polymer.
10936. The method of item 10812, wherein the composition comprises a polymer, and the polymer is, or comprises, a hydrophilic polymer.
10937. The method of item 10812, wherein the composition comprises a polymer, and the polymer is, or comprises, a hydrophobic polymer.
10938. The method of item 10812, wherein the composition comprises a polymer, and the polymer is, or comprises, a polymer having hydrophilic domains.
10939. The method of item 10812, wherein the composition comprises a polymer, and the polymer is, or comprises, a polymer having hydrophobic domains.
10940. The method of item 10812, wherein the composition comprises a polymer, and the polymer is, or comprises, a non- conductive polymer. 10941. The method of item 10812, wherein the composition comprises a polymer, and the polymer is, or comprises, an elastomer.
10942. The method of item 10812, wherein the composition comprises a polymer, and the polymer is, or comprises, a hydrogel.
10943. The method of item 10812, wherein the composition comprises a polymer, and the polymer is, or comprises, a silicone polymer.
10944. The method of item 10812, wherein the composition comprises a polymer, and the polymer is, or comprises, a hydrocarbon polymer.
10945. The method of item 10812, wherein the composition comprises a polymer, and the polymer is, or comprises, a styrene-derived polymer.
10946. The method of item 10812, wherein the composition comprises a polymer, and the polymer is, or comprises, a butadiene-derived polymer.
10947. The method of item 10812, wherein the composition comprises a polymer, and the polymer is, or comprises, a macromer.
10948. The method of item 10812, wherein the composition comprises a polymer, and the polymer is, or comprises, a poly(ethylene glycol) polymer. 10949. The method of item 10812, wherein the composition comprises a polymer, and the polymer is, or comprises, an amorphous polymer.
10950. The method of item 10812, wherein the composition further comprises a second pharmaceutically active agent.
10951. The method of item 10812, wherein the composition further comprises an anti-inflammatory agent.
10952. The method of item 10812, wherein the composition further comprises an agent that inhibits infection.
10953. The method of item 10812, wherein the composition further comprises an anthracycline.
10954. The method of item 10812, wherein the composition further comprises doxorubicin.
10955. The method of item 10812 wherein the composition further comprises mitoxantrone.
10956. The method of item 10812 wherein the composition further comprises a fluoropyrimidine.
10957. The method of item 10812, wherein the composition further comprises 5-fluorouracil (5-FU).
10958. The method of item 10812, wherein the composition further comprises a folic acid antagonist. 10959. The method of item 10812, wherein the composition further comprises methotrexate.
10960. The method of item 10812, wherein the composition further comprises a podophylotoxin.
10961. The method of item 10812, wherein the composition further comprises etoposide.
10962. The method of item 10812, wherein the composition further comprises camptothecin.
10963. The method of item 10812, wherein the composition further comprises a hydroxyurea.
10964. The method of item 10812, wherein the composition further comprises a platinum complex.
10965. The method of item 10812, wherein the composition further comprises cisplatin.
10966. The method of item 10812 wherein the composition further comprises an anti-thrombotic agent.
10967. The method of item 10812, wherein the composition further comprises a visualization agent.
10968. The method of item 10812, wherein the composition further comprises a visualization agent, and the visualization agent is a radiopaque material, wherein the radiopaque material comprises a metal, a halogenated compound, or a barium containing compound. 10969. The method of item 10812, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, barium, tantalum, or technetium.
10970. The method of item 10812, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, an MRI responsive material.
10971. The method of item 10812, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, a gadolinium chelate.
10972. The method of item 10812, wherein the
, composition further comprises a visualization agent, and the visualization agent is, or comprises, iron, magnesium, manganese, copper, or chromium.
10973. The method of item 10812, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, iron oxide compound.
10974. The method of item 10812, wherein the composition further comprises a visualization agent, and the visualization agent is, or comprises, a dye, pigment, or colorant.
10975. The method of item 10812 wherein the agent is released in effective concentrations from the composition comprising the agent by diffusion over a period ranging from the time of administration to about 90 days.
10976. The method of item 10812 wherein the agent is released in effective concentrations from the composition comprising the agent by erosion of the composition over a period ranging from the time of administration to about 90 days.
10977. The method of item 10812 wherein the composition further comprises an inflammatory cytokine.
10978. The method of item 10812 wherein the composition further comprises an agent that stimulates cell proliferation.
10979. The method of item 10812 wherein the composition further comprises a polymeric carrier.
10980. The method of item 10812 wherein the composition is in the form of a gel, paste, or spray.
10981. The method of item 10812 wherein the implant is partially constructed with the agent or the composition.
10982. The method of item 10812 wherein the implant is fully constructed with the agent or the composition.
10983. The method of item 10812 wherein the implant is impregnated with the agent or the composition.
10984. The method of item 10812, wherein the agent or the composition forms a coating, and the coating directly contacts the implant.
10985. The method of item 10812, wherein the agent or the composition forms a coating, and the coating indirectly contacts the implant. 10986. The method of item 10812 wherein the agent or the composition forms a coating, and the coating partially covers the implant.
10987. The method of item 10812, wherein the agent or the composition forms a coating, and the coating completely covers the implant.
10988. The method of item 10812 wherein the agent or the composition is located within pores or holes of the implant.
10989. The method of item 10812 wherein the agent or the composition is located within a channel, lumen, or divet of the implant.
10990. The method of item 10812 wherein the implant further comprising an echogenic material.
10991. The method of item 10812 wherein the implant further comprises an echogenic material, wherein the echogenic material is in the form of a coating.
10992. The method of item 10812 wherein the implant is sterile.
10993. The method of item 10812 wherein the agent is delivered from the implant, wherein the agent is released into tissue in the vicinity of the implant after deployment of the implant.
10994. The method of item 10812 wherein the agent is delivered from the implant, wherein the agent is released into tissue in the vicinity of the implant after deployment of the implant, wherein the tissue is connective tissue.
10995. The method of item 10812 wherein the agent is delivered from the implant, wherein the agent is released into tissue in the vicinity of the implant after deployment of the implant, wherein the tissue is muscle tissue.
10996. The method of item 10812 wherein the agent is delivered from the implant, wherein the agent is released into tissue in the vicinity of the implant after deployment of the implant, wherein the tissue is nerve tissue.
10997. The method of item 10812 wherein the agent is delivered from the implant, wherein the agent is released into tissue in the vicinity of the implant after deployment of the implant, wherein the tissue is epithelium tissue.
10998. The method of item 10812 wherein the agent is delivered from the implant, wherein the agent is released in effective concentrations from the implant over a period ranging from the time of deployment of the implant to about 1 year.
10999. The method of item 10812 wherein the agent is delivered from the implant, wherein the agent is released in effective concentrations from the implant over a period ranging from about 1 month to 6 months.
11000. The method of item 10812 wherein the agent is delivered from the implant, wherein the agent is released in effective concentrations from the implant over a period ranging from about 1 - 90 days.
11001. The method of item 10812 wherein the agent is delivered from the implant, wherein the agent is released in effective concentrations from the implant at a constant rate.
11002. The method of item 10812 wherein the agent is delivered from the implant, wherein the agent is released in effective concentrations from the implant at an increasing rate.
11003. The method of item 10812 wherein the agent is delivered from the implant, wherein the agent is released in effective concentrations from the implant at a decreasing rate.
11004. The method of item 10812 wherein the agent is delivered from the implant, wherein the implant comprises about 0.01 Dg to about 10 Dg of the agent.
11005. The method of item 10812 wherein the agent is delivered from the implant, wherein the implant comprises about 10 Dg to about 10 mg of the agent.
11006. The method of item 10812 wherein the agent is delivered from the implant, wherein the implant comprises about 10 mg to about 250 mg of the agent.
11007. The method of item 10812 wherein the agent is delivered from the implant, wherein the implant comprises about 250 mg to about 1000 mg of the agent. 11008. The method of item 10812 wherein the agent is delivered from the implant, wherein the implant comprises about 1000 mg to about 2500 mg of the agent.
11009. The method of item 10812 wherein the agent is delivered from the implant, wherein a surface of the implant comprises less than 0.01 Og of the agent per mm2 of implant surface to which the agent is applied.
11010. The method of item 10812 wherein the agent is delivered from the implant, wherein a surface of the implant comprises about 0.01 Dg to about 1 Dg of the agent per mm2 of implant surface to which the agent is applied.
11011. The method of item 10812 wherein the agent is delivered from the implant, wherein a surface of the implant comprises about 1 Dg to about 10 Dg of the agent per mm2 of implant surface to which the agent is applied.
11012. The method of item 10812 wherein the agent is delivered from the implant, wherein a surface of the implant comprises about 10 Dg to about 250 Dg of the agent per mm2 of implant surface to which the agent is applied.
11013. The method of item 10812 wherein the agent is delivered from the implant, wherein a surface of the implant comprises about 250 Dg to about 1000 Dg of the agent per mm2 of implant surface to which the agent is applied.
11014. The method of item 10812 wherein the agent is delivered from the implant, wherein a surface of the implant comprises about 1000 Dg to about 2500 Dg of the agent per mm2 of implant surface to which the agent is applied.
11015. The method of item 10812, wherein the implant further comprises a coating, and the coating is a uniform coating.
11016. The method of item 10812, wherein the implant further comprises a coating, and the coating is a non-uniform coating.
11017. The method of item 10812, wherein the implant further comprises a coating, and the coating is a discontinuous coating.
11018. The method of item 10812, wherein the implant further comprises a coating, and the coating is a patterned coating.
11019. The method of item 10812, wherein the implant further comprises a coating, and the coating has a thickness of 100 Dm or less.
11020. The method of item 10812, wherein the implant further comprises a coating, and the coating has a thickness of 10 Dm or less.
11021. The method of item 10812, wherein the implant further comprises a coating, and the coating adheres to the surface of the implant upon deployment of the implant.
11022. The method of item 10812, wherein the implant further comprises a coating, and the coating is stable at room temperature for a period of at least 1 year. 11023. The method of item 10812, wherein the implant further comprises a coating, and the agent is present in the coating in an amount ranging between about 0.0001% to about 1% by weight.
11024. The method of item 10812, wherein the implant further comprises a coating, and the agent is present in the coating in an amount ranging between about 1% to about 10% by weight.
11025. The method of item 10812, wherein the implant further comprises a coating, and the agent is present in the coating in an amount ranging between about 10% to about 25% by weight.
11026. The method of item 10812, wherein the implant further comprises a coating, and the agent is present in the coating in an amount ranging between about 25% to about 70% by weight.
11027. The method of item 10812, wherein the implant further comprises a coating, and the coating comprises a polymer.
11028. The method of item 10812, wherein the implant comprises a first coating having a first composition and a second coating having a second composition.
11029. The method of item 10812, wherein the implant comprises a first coating having a first composition and a second coating having a second composition, wherein the first composition and the second composition are different.
11030. A method of making a device comprising: combining a medical device (e.g., an implant) and an anti-scarring agent or a composition comprising an anti-scarring agent, wherein the agent inhibits scarring between the device and a host into which the device is implanted.
11031. The method of item 11030 wherein the agent is an adensosine A2A receptor antagonist.
11032. The method of item 11030 wherein the agent is an AKT inhibitor.
11033. The method of item 11030 wherein the agent is an alpha 2 integrin antagonist, wherein the alpha 2 integrin antagonist is Pharmaprojects No. 5754 (Merck KgaA).
11034. The method of item 11030 wherein the agent is an alpha 4 integrin antagonist.
11035. The method of item 11030 wherein the agent is an alpha 7 nicotinic receptor agonist.
11036. The method of item 11030 wherein the agent is an angiogenesis inhibitor selected from the group consisting of AG-12,958 (Pfizer), ATN-161 (Attenuon LLC), neovastat, an angiogenesis inhibitor from Jerina AG (Germany), NM-3 (Mercian), VGA-1155 (Taisho), FCE-26644 (Pfizer), FCE-26950 (Pfizer), FPMA (Meiji Daries), FR-111142 (Fujisawa), GGTI-298, GM-1306 (Ligand), GPA-1734 (Novartis), NNC-47-0011 (Novo Nordisk), herbamycin (Nippon Kayaku), lenalidomide (Celegene), IP-10 (NIH), ABT-828 (Abbott), KIN-841 (Tokushima University, Japan), SF-1126 (Semafore Pharmaceuticals), laminin technology (NIH), CHIR-258 (Chiron), NVP-AEW541 (Novartis), NVP-AEW541 (Novartis), Vt16907 (Alchemia), OXI-8007 (Oxigene), EG-3306 (Ark Therapeutics), Maspin (Arriva), ABT- 567 (Abbott), PPI-2458 (Praecis Pharmaceuticals), CC-5079, CC-4089 (Celgene), HIF-1 alpha inhibitors (Xenova), S-247 (Pfizer), AP-23573 (Ariad), AZD-9935 (Astra Zeneca), mebendazole (Introgen Therapeutics), MetAP-2 inhibitors (GlaxoSmithKline), AG-615 (Angiogene Pharmaceuticals), Tie-2 antagonists (Hybrigenics), NC-381 , CYC-381 , NC-169, NC-219, NC-383, NC-384, NC-407 (Lorus Therapeutics), ATN-224 (Attenuon), ON-01370 (Onconova), Vitronectin antagonists (Amgen), SDX-103 (Salmedix), Vitronectin antagonists (Shire), CHP (Riemser), TEK (Amgen), Anecortave acetate (Alcon), T46.2 (Matrix Therapeutics), HG-2 (Heptagen), TEM antagonists (Genzyme), Oxi-4500 (Oxigene), ATN-161 (Attenuon), WX-293 (Wilex), M-2025 (Metris Therapeutics), Alphastatin (BioActa), YH-16 (Yantai Rongchang), BIBF-1120 (Boehringer Ingelheim), BAY-57-9352 (Bayer), AS- 1404 (Cancer Research Technology), SC-77964 (Pfizer), glycomimetics (BioTie Therapies), TIE-2 Inhibitors (Ontogen), DIMI, Octamer (Octamer), ABR-215050 (Active Biotech), ABT-518 (Abbott), KDR inhibitors (Abbott), BSF-466895 (Abbott), SCH-221153 (Schering-Plough), DAC:antiangiogenic (ConjuChem), TFPI (EntreMed), AZD-2171 (Astra-Zenaca), CDC-394 (Celgene), LY290293 (EIi Lilly), IDN-5390 (Indena), Kdr Kinase Inhibitors (Merck), CT-113020, CT-116433, CT-116563, CT-31890, CT-32228) (Cell Therapeutics), A-299620 (Abbott), TWEAK Inhibitor (Amgen), VEGF modulators (Johnson and Johnson), Tum-N53, tumstatin (Genzyme), Thios- 1 , Thios-2 (Thios Pharmaceuticals), MV-6401 (Miravant Medical Technologies), Spisulosine (PharmaMar), CEP-7055 (Cephalon), AUV-201 (Auvation), LM-609 (EIi Lilly), SKF-106615 (AnorMED), Oglufanide disodium (Cytran), BW-114 (Pharminox), Calreticulin (NIH), WX-678 (Wilex), SD-7784 (Pfizer), WX-UK1 (Wilex), SH-268 (Schering AG), 2-Me-PGA (Celgene), S- 137 (Pfizer), ZD-6126 (Angiogene Pharmaceuticals), SG-292 (SignalGen), Benefin (Lane Labs), A6, A36 (Angstrom), SB-2723005 (GlaxoSmithKline), SC-7 (Cell Therapeutics), ZEN-014 (AEterna Zentaris), 2-methoxyestradiol (EntreMed), NK-130119 (Nippon Kayaku), CC-10004 (Celgene), AVE- 8062A (Ajinomoto), Tacedinaline (Pfizer), Actinonin (Tokyo Metropolitan Institute of Medical Science), Lenalidomide (Celgene), VGA-1155, BTO-956 (SRI International), ER-68203-00 (Eisai), CT-6685 (UCB), JKC-362 (Phoenix Pharmaceuticals), DMI-3798 (DMI Biosciences, Angiomate (Ipsen), ZD-6474 (AstraZeneca), CEP-5214 (Cephalon), Canstatin (Genzyme), NM-3 (Mercian), Oridigm (MediQuest Therapeutics), Exherin (Adherex), BLS-0597 (Boston Life Sciences), PTC-299 (PTC Therapeutics), NPI-2358 (Nereus Pharmaceuticals), CGP-79787 (Novartis), AEE-788 (Novartis), CKD-732 (Chong Kun Dang), CP-564959 (OSI Pharmaceuticals), CM-101 (CarboMed), CT-2584, CT3501 (Cell Therapeutics), combretastatin and analogues and derivatives thereof (Oxigene), Rebimastat (Bristol-Meyers Squibb), Dextrin 2-sulfate (ML Laboratories), Cilengitide (Merk KGaA), NSC-706704 (Pharminox), KRN- 951 (Kirin Brewery), Ukrain, NSC-631570 (Nowicky Pharma), Tecogalan sodium (Daiichi Pharmaceutical), Tz-93 (Tsumura), TBC-1635 (Encysive Pharmaceuticals), TAN-1120 (Takeda), Semaxanib (Pfizer), BDI-7800 (Biopharmacopae), SD-186, SD-983 (Bristol-Meyers Squibb), SB-223245 (GlaxoSmithKline), SC-236 (Pfizer), RWJ-590973 (Johnson and Johnson), ILX-1850 (Genzyme), SC-68488, S-836 (Pfizer), CG-55069-11 (CuraGen), Ki-23057 (Kirin Brewery), CCX-700 (Chemoentryx), Pegaptanib octasodium (Giled Sciences), ANGIOCOL (available from Biostratum Inc.), or an analogue or derivative thereof.
11037. The method of item 11030 wherein the agent is an apoptosis antagonist.
11038. The method of item 11030 wherein the agent is an apoptosis activator.
11039. The method of item 11030 wherein the agent is a beta 1 integrin antagonist. 11040. The method of item 11030 wherein the agent is a beta tubulin inhibitor.
11041. The method of item 11030 wherein the agent is a blocker of enzyme production in Hepatitis C.
11042. The method of item 11030 wherein the agent is a Bruton's tyrosine kinase inhibitor.
11043. The method of item 11030 wherein the agent is a calcineurin inhibitor.
11044. The method of item 11030 wherein the agent is a caspase 3 inhibitor.
11045. The method of item 11030 wherein the agent is a CC chemokine receptor antagonist.
11046. The method of item 11030 wherein the agent is a cell cycle inhibitor selected from the group consisting of SNS-595 (Sunesis), synthadotin, KRX-0403, and an analogue or derivative thereof.
11047. The method of item 11030 wherein the agent is a cathepsin B inhibitor.
11048. The method of item 11030 wherein the agent is a cathepsin K inhibitor, wherein the cathepsin K inhibitor is 462795 (GlaxoSmithKline), INPL-022-D6 (Amura Therapeutics), or an analogue or derivative thereof. 11049. The method of item 11030 wherein the agent is a cathepsin L inhibitor.
11050. The method of item 11030 wherein the agent is a CD40 antagonist.
11051. The method of item 11030 wherein the agent is a chemokine receptor agonist.
11052. The method of item 11030 wherein the agent is a chymase inhibitor.
11053. The method of item 11030 wherein the agent is a collagenase antagonist.
11054. The method of item 11030 wherein the agent is a CXCR antagonist.
11055. The method of item 11030 wherein the agent is a cyclin dependent kinase inhibitor selected from the group consisting of a CDK-1 inhibitor, a CDK-2 inhibitor, a CDK- 4 inhibitor, a CDK-6 inhibitor, a CAK1 inhibitor from GPC Biotech and Bristol-Myers Squibb, RGB-286199 (GPC Biotech), an anticancer agent from Astex Technology, a CAK1 inhibitor from GPC Biotech, a CDK inhibitor from Sanofi-Aventis, a CDK1/CDK2 inhibitor from Amgen, a CDK2 inhibitor from SUGEN-2 (Pfizer), a hearing loss therapy agent (Sound Pharmaceuticals), PD-0332991 (Pfizer), RGB-286199 (GPC Biotech), Ro-0505124 (Hoffmann-La Roche), a Ser/Thr kinase inhibitor from Lilly (EIi Lilly), CVT-2584 (CAS No. 199986-75- 9) (CV Therapeutics), and an analogue or derivative thereof. 11056. The method of item 11030 wherein the agent is a cyclooxygenase 1 inhibitor.
11057. The method of item 11030 wherein the agent is a DHFR inhibitor.
11058. The method of item 11030 wherein the agent is a dual integrin inhibitor.
11059. The method of item 11030 wherein the agent is an elastase inhibitor.
11060. The method of item 11030 wherein the agent is an elongation factor-1 alpha inhibitor.
11061. The method of item 11030 wherein the agent is an endothelial growth factor antagonist.
11062. The method of item 11030 wherein the agent is an endothelial growth factor receptor kinase inhibitor selected from the group consisting of sorafenib tosylate (Bayer), AAL-993 (Novartis), ABP-309 (Novartis), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), E- 7080 (Eisai), EG-3306 (Ark Therapeutics), EXEL-2880 (Exelixis), GW- 654652 (GlaxoSmithKline), a KDR inhibitor from LG Life Sciences, CT-6685 and CT-6729 (UCB), KRN-633 and KRN-951 (Kirin Brewery), OSI-930 (OSI Pharmaceuticals), SP-5.2 (Supratek Pharma), SU-11657 (Pfizer), a Tie-2 antagonist (Hybrigenics), a VEGFR-2 kinase inhibitor (Bristol-Myers Squibb), XL-647 (Exelixis), a KDR inhibitor from Abbott Laboratories, sorafenib tosylate, and an analogue or derivative thereof. 11063. The method of item 11030 wherein the agent is an endotoxin antagonist.
11064. The method of item 11030 wherein the agent is an epothilone and tubulin binder.
11065. The method of item 11030 wherein the agent is an estrogen receptor antagonist.
11066. The method of item 11030 wherein the agent is an FGF inhibitor.
11067. The method of item 11030 wherein the agent is a farnexyl transferase inhibitor.
11068. The method of item 11030 wherein the agent is farnesyltransferase inhibitor selected from the group of A-197574 (Abbott), a famesyltransferase inhibitor from Servier, FPTIII (Strathclyde Institute for Drug R), LB-42908 (LG Life Sciences), Pharmaprojects No. 5063 (Genzyme), Pharmaprojects No. 5597 (Ipsen), Yissum Project No. B-1055 (Yissum), and an analogue or derivative thereof.
11069. The method of item 11030 wherein the agent is an FLT-3 kinase inhibitor.
11070. The method of item 11030 wherein the agent is an FGF receptor kinase inhibitor.
11071. The method of item 11030 wherein the agent is a fibrinogen antagonist selected from the group consisting of AUV-201 (Auvation), MG-13926 (Sanofi-Aventis), plasminogen activator (CAS No. 105913-11-9) (from Sanofi-Aventis or UCB), plasminogen activator-2 (tPA- 2) (Sanofi-Aventis), pro-urokinase (CAS No. 82657-92-9) (Sanofi-Aventis), and an analogue or derivative thereof.
11072. The method of item 11030 wherein the agent is a heat shock protein 90 antagonist selected from the group consisting of SRN- 005 (Sirenade), geldanamycin, NSC-33050 (17-allylaminogeldanamycin; 17- AAG), ^-dimethylaminoethylamino-^-demethoxy-geldanamycin (17- DMAG), rifabutin (rifamycin XIV, 1',4-didehydro-1-deoxy-1 ,4-dihydro-5'-(2- methylpropyl)-1-oxo-), and an analogue or derivative thereof.
11073. The method of item 11030 wherein the agent is a histone deacetylase inhibitor.
11074. The method of item 11030 wherein the agent is an HMGCoA reductase inhibitor selected from the group consisting of an atherosclerosis therapeutic from Lipid Sciences, ATI-16000 (ARYx Therapeutics), KS-01-019 (Kos Pharmaceuticals), Pharmaprojects No. 2197 (Sanofi-Aventi), RP 61969 (Sanofi-Aventis), and an analogue or derivative thereof.
11075. The method of item 11030 wherein the agent is an ICAM inhibitor.
11076. The method of item 11030 wherein the agent is an IL, ICE and IRAK antagonist, wherein the antagonist is a CJ-14877, CP- 424174 (Pfizer), NF-61 (Negma-Lerads), and an analogue or derivative thereof.
11077. The method of item 11030 wherein the agent is an IL-2 inhibitor. 11078. The method of item 11030 wherein the agent is an immunosuppressant selected from the group consisting of teriflunomide (Sanofi Aventis), chlorsulfaquinoxalone (NSC-339004), chlorsulfaquinoxalone sulfate, CS-712 (Sankyo), ismomultin alfa (CAS No. 457913-93-8) (Akzo Nobel), antiallergics from GenPat77, antiinflammatories or AT-005 (Androclus Therapeutics), autoimmune disease therapeutics from EpiVax, BN-007 (Bone), budesonide (CAS No. 51333-22- 3) (MAP Pharmaceuticals), CO-14 (Genzyme), edratide (CAS No. 433922- 67-9) (Teva), EP-314 (Enanta), eprovafen (CAS No. 101335-99-3) (Sanofi- Aventis), HWA-131 (CAS No. 118788-41-3) (Sanofi-Aventis), immunomodulators from MerLion Pharmaceuticals, immunosuppressives from Alchemia, IPL-12 (Inflazyme), MDL-9563 (CAS No. 27086-86-8) (Sanofi-Aventis), Pharmaprojects No. 2330 (Sanofi-Aventis), Pharmaprojects No. 6426 (Abgenix), PXS-25 (Pharmaxis), rosmarinic acid (CAS No. 20283-92-5) (Sanofi-Aventis), RP 42927 or RP 54745 (CAS No.
135330-08-4) (Sanofi-Aventis), SGN-35 (Seattle Genetics), ST-1959 (Sigma-Tau), type I diabetes therapy from SYNX Pharma, UNIL-88 (Debiopharm), VP-025 (Vasogen), VR-694 (Vectura), PRTX-001 (Protalex), and an analogue or derivative thereof.
11079. The method of item 11030 wherein the agent is an IMPDH (inosine monophosphate).
11080. The method of item 11030 wherein the agent is an integrin antagonist.
11081. The method of item 11030 wherein the agent is an interleukin antagonist.
11082. The method of item 11030 wherein the agent is an inhibitor of type III receptor tyrosine kinase. 11083. The method of item 11030 wherein the agent is an irreversible inhibitor of enzyme methionine aminopeptidase type 2.
11084. The method of item 11030 wherein the agent is an isozyme selective delta protein kinase C inhibitor.
11085. The method of item 11030 wherein the agent a JAK3 enzyme inhibitor.
11086. The method of item 11030 wherein the agent is a
JNK inhibitor.
11087. The method of item 11030 wherein the agent is a kinase inhibitor.
11088. The method of item 11030 wherein the agent is kinesin antagonist.
11089. The method of item 11030 wherein the agent is a kinesin antagonist.
11090. The method of item 11030 wherein the agent is a leukotriene inhibitor and antagonist selected from the group consisting of ambicromil (CAS No. 58805-38-2) (Sanofi-Aventis), amelubant (CAS No. 346735-24-8) (Boehringer Ingelheim), DW-1141 (Dong Wha), ebselen (Daiichi Pharmaceutical), ibudilast (Kyorin), Ieucotriene inhibitors from Sanofi-Aventis, lymphotoxin-beta receptor (LT-D) from Biogen Idee, Pharmaprojects No. 1535 and 2728 (CAS No. 119340-33-9) (Sanofi- Aventis), R-112 (Rigel), Rev-5367 (CAS No. 92532-05-3) (Sanofi-Aventis), RG-14893 (CAS No. 141835-49-6) (Sanofi-Aventis), RG-5901-A (CAS No. 101910-24-1), 92532-23-5, RP 66153 (CAS No. 142422-79-5), RP 66364 (CAS No.186916-92-5), RP 69698 (CAS No. 141748-00-7) (Sanofi-Aventis), SC-411930 (Pfizer), SC-41930 (CAS No. 120072-59-5) (Pfizer), SC-50605 (CAS No. 138828-39-4) (Pfizer), SC-51146 (CAS No. 141059-52-1), SC- 53228 (CAS No. 153633-01-3) (Pfizer), spaglumic acid (ZY-15106) (CAS No.3106-85-2), 80619-64-3 (Novartis), tipredane (CAS No. 85197-77-9) (Bristol-Myers Squibb), U-75302 (CAS No. 119477-85-9) (Pfizer), and analogue or derivative thereof.
11091. The method of item 11030 wherein the agent is an MAP kinase inhibitor.
11092. The method of item 11030 wherein the agent is a matrix metalloproteinase inhibitor.
11093. The method of item 11030 wherein the agent is an MCP-CCR2 inhibitor.
11094. The method of item 11030 wherein the agent is an mTOR inhibitor.
11095. The method of item 11030 wherein the agent is an mTOR kinase inhibitor.
11096. The method of item 11030 wherein the agent is a microtubule inhibitor selected from the group consisting of antibody- maytansinoid conjugates from Biogen Idee, colchicines (MantiCore Pharmaceuticals), anticancer immunoconjugates from Johnson & Johnson, DIME from Octamer, gni-1f (GNI), huC242-DM4, huMy9-6-DM1 (immunoGen), lDN-5404 (Indena), IMO-098, IMOderm (Imotep), mebendazole (Introgen Therapeutics), microtubule poisons from Cambridge Enterprise, paclitaxel such as LOTAX from Aphios (CAS No. 33069-62-4), Genexol-PM from Samyang, Pharmaprojects No. 6383 (Tapestry Pharmaceuticals), RPR-112378 (Sanofi-Aventis), SGN-75 (Seattle Genetics), SPL-7435 (Starpharma), SSR-250411 (Sanofi-Aventis), trastuzumab-DM1 (Genentech), vinorelbine, and an analogue or derivative thereof.
11097. The method of item 11030 wherein the agent is an MIF inhibitor.
11098. The method of item 11030 wherein the agent is an MMP inhibitor.
11099. The method of item 11030 wherein the agent is a neurokinin (NK) antagonist selected from the group consisting of anthrotainin (CAS No. 148084-40-6) (Sanofi-Aventis), an IBS thereapeutic from ArQuIe, MDL-105212A (CAS No. 167261 -60-1 ) (Ssanofi-Aventis), Pharmaprojects No. 2744, 3258 (CAS No. 139167-47-8) 4006, 4201 , or 5986 (Sanofi-Aventis), RP 67580 (CAS No. 135911-02-3), SR-144190 (CAS No. 201152-86-5), SSR-240600, SSR-241586 (Sanofi-Aventis), TKA-457 (Novartis), vestipitant mesylate (CAS No. 334476-64-1) (GlaxoSmithKline), Win-64821 (Sanofi-Aventis), PRX-96026 (Predix Pharmaceuticals), and an analogue or derivative thereof.
11100. The method of item 11030 wherein the agent is an NF kappa B inhibitor selected from the group consisting of AVE-0545 or AVE-0547 (Sanofi-Aventis), bortezomib (CAS No. 179324-69-7) (Millennium Pharmaceuticals), dexanabinol (CAS No. 112924-45-5) (Pharmos), dexlipotam (Viatris), Pharmaprojects No. 6283 (INDRA) (OXiGENE), IPL- 576092 (CAS No. 137571-30-3) (Inflazyme), NFKB decoy (Corgentech), NFKB decoy oligo (AnGes MG), NFKB's from Ariad, osteoporosis treatments or S5 (F005) from Fulcrum Pharmaceuticals, P61 (Phytopharm), R-flurbiprofen (CAS No. 5104-49-4) (Encore Pharmaceuticals), and an analogue or derivative thereof.
11101. The method of item 11030 wherein the agent is a nitric oxide agonist.
11102. The method of item 11030 wherein the agent is an ornithine decarboxylase inhibitor.
11103. The method of item 11030 wherein the agent is a p38 MAP kinase inhibitor selected from the group consisting of AZD-6703 (AstraZeneca), JX-401 (Jexys Pharmaceuticals), BMS-2 (Bristol-Myers Squibb), a p38 MAP kinase inhibitor from Novartis, a p38-alpha MAP kinase inhibitor from Amphora, Pharmaprojects No. 5704 (Pharmacopeia), RPR- 200765A (Sanofi-Aventis), SD-282 (Johnson & Johnson), TAK-715 (Takeda), and an analogue or derivative thereof.
11104. The method of item 11030 wherein the agent is a palmitoyl-protein thioesterase inhibitor.
11105. The method of item 11030 wherein the agent is a PDGF receptor kinase inhibitor selected from the group consisting of AAL- 993, AMN-107, or ABP-309 (Novartis), AMG-706 (Amgen), BAY-57-9352 (Bayer), CDP-860 (UCB), E-7080 (Eisai), imatinib (CAS No. 152459-95-5) (Novartis), OSI-930 (OSI Pharmaceuticals), RPR-127963E (Sanofi-Aventis), RWJ-540973 (Johnson & Johnson), sorafenib tosylate (Bayer), SU-11657 (Pfizer), tandutinib (CAS No. 387867-13-2) (Millennium Pharmaceuticals), vatalanib (Novartis), ZK-CDK (Schering AG), and an analogue or derivative thereof. 11106. The method of item 11030 wherein the agent is (-)-halofenate (Metabolex), AMG-131 (Amgen), antidiabetics from Japan Tobacco, AZD-4619, AZD-8450, AZD-8677 (AstraZeneca), DRF-10945, balaglitazone (Dr Reddy's), CS-00088, CS-00098 (Chipscreen Biosciences), E-3030 (Eisai), etalocib (CAS No. 161172-51-6) (EIi Lilly), GSK-641597 (Ligand), GSK-677954 (GlaxoSmithKline), GW-409544 (Ligand), GW- 590735 (GlaxoSmithKline), K-111 (Hoffmann-La Roche), LY-518674 (EIi Lilly), LY-674 (Ligand), LY-929 (Ligand), MC-3001, MC-3002 (MaxoCore Pharmaceuticals), metformin HCI + pioglitazone (CAS No. 1115-70-4 and 112529-15-4), ACTOPLUS MET from Andrx), muraglitazar (CAS No. 331741-94-7) (Bristol-Myers Squibb), naveglitazar (Ligand), oleoylethanolamide (Kadmus Pharmaceuticals), ONO-5129, pioglitazone hydrochloride (CAS No. 111025-46-8 and 112529-15-4) (Takeda), PLX-204 (Plexxikon), PPAR agonists from Genfit, PPAR delta agonists from EIi Lilly, PPAR-alpha agonists from CrystalGenomics, PPAR-gamma modulators and PPAR-β modulators from CareX, rosiglitazone maleate (CAS No. 122324- 73-4 or 155141-29-0) (GlaxoSmithKline), rosiglitazone maleate/glimepir (CAS No. 155141-29-0 and 93479-97-1), AVANDARYL, rosiglitazone maleate/metformin extend (CAS No. 155141-29-0 and 657-24-9), AVANDAMET, rosiglitazone maleate+metformin, AVANDAMET (GlaxoSmithKline), tesaglitazar (AstraZeneca), and an analogue or derivative thereof.
11107. The method of item 11030 wherein the agent is a phosphatase inhibitor.
11108. The method of item 11030 wherein the agent is a phosphodiesterase (PDE) inhibitor selected from the group consisting of avanafil (Tanabe Seiyaku), dasantafil (CAS No. 569351-91-3) (Schering- Plough), A-906119 (CAS No. 134072-58-5), DL-850 (Sanofi-Aventis), GRC- 3015, GRC-3566, GRC-3886 (Glenmark), HWA-153 (CAS No. 56395-66-5) (Sanofi-Aventis), hydroxypumafentrine (Altana), IBFB-130011 , IBFB-14-016, IBFB-140301 , IBFB-150007, IBFB-211913 (IBFB Pharma), L-826141 (Merck & Co), medorinone (CAS No. 88296-61-1) (Sanofi-Aventis), MEM- 1917 (Memory Pharmaceuticals), ND-1251 (Neuro3d), PDE inhibitors from ICOS, PDE IV inhibitors from Memory Pharmaceuticals and CrystalGenomics, Pharmaprojects No. 2742 and 6141 (Sanofi-Aventis), QAD-171 (Novartis), RHC-2963 (CAS No. 76993-12-9 and 76993-14-1), RPR-117658, RPR-122818 derivatives, SR-24870 , and RPR-132294 (Sanofi-Aventis), SK-350 (ln2Gen), stroke therapy agents from deCODE Genetics, TAS-203 (Taiho), tofimilast (CAS No. 185954-27-2) (Pfizer), UK- 371800 (Pfizer), WIN-65579 (CAS No. 158020-82-7) (Sanofi-Aventis), IBFB- 130020 (IBFB Pharma), OPC-6535 (CAS No. 145739-56-6) (Otsuka), a phosphodiesterase III inhibitor, enoximone, a phosphodiesterase IV inhibitor, fosfosal, Atopik (Barrier Therapeutics), triflusal, a phosphodiesterase V inhibitor, and an analogue or derivative thereof.
11109. The method of item 11030 wherein the agent is a PKC inhibitor.
11110. The method of item 11030 wherein the agent is a platelet activating factor antagonist.
11111. The method of item 11030 wherein the agent is a platelet-derived growth factor receptor kinase inhibitor.
11112. The method of item 11030 wherein the agent is a prolyl hydroxylase inhibitor.
11113. The method of item 11030 wherein the agent is a polymorphonuclear neutrophil inhibitor. 11114. The method of item 11030 wherein the agent is a protein kinase B inhibitor.
11115. The method of item 11030 wherein the agent is a protein kinase C stimulant.
11116. The method of item 11030 wherein the agent is a purine nucleoside analogue.
11117. The method of item 11030 wherein the agent is a purinoreceptor P2X antagonist.
11118. The method of item 11030 wherein the agent is a Raf kinase inhibitor.
11119. The method of item 11030 wherein the agent is a reversible inhibitor of ErbB1 and ErbB2.
11120. The method of item 11030 wherein the agent is a ribonucleoside triphosphate reductase inhibitor.
11121. The method of item 11030 wherein the agent is an SDF-1 antagonist.
11122. The method of item 11030 wherein the agent is a sheddase inhibitor.
11123. The method of item 11030 wherein the agent is an SRC inhibitor. 11124. The method of item 11030 wherein the agent is a stromelysin inhibitor.
11125. The method of item 11030 wherein the agent is an Syk kinase inhibitor.
11126. The method of item 11030 wherein the agent is a telomerase inhibitor.
11127. The method of item 11030 wherein the agent is a TGF beta inhibitor selected from the group consisting of pirfenidone (CAS No. 53179-13-8) (MARNAC), tranilast (CAS No. 53902-12-8) (Kissei), IN- 1130 (ln2Gen), mannose-6-phosphate (BTG), TGF-β antagonists from Inflazyme (Pharmaprojects No. 6075), TGF-β antagonists from Sydney, non-industrial source), TGF-β I receptor kinase inhibitors from EIi Lilly, TGF- β receptor inhibitors from Johnson & Johnson, and an analogue or derivative thereof.
11128. The method of item 11030 wherein the agent is a TNFα antagonist or TACE inhibitor selected from the group consisting of adalimumab (CAS No. 331731-18-1) (Cambridge Antibody Technology), AGIX-4207 (AtheroGenics), AGT-1 (Advanced Biotherapy), an antiinflammatory from Borean Pharma, Cellzome, or Paradigm Therapeutics, anti-inflammatory vaccine (TNF-alpha kinoid) from Neovacs, humanized anti-TNF antibody or an anti-TNF MAb (CB0006) Celltech (UCB), apratastat (CAS No. 287445-51-0) (Wyeth), BMS-561392 (Bristol-Myers Squibb), BN- 006 (Bone), certolizumab pegol (CAS No. 428863-50-7 or CH-138 (UCB), cilomilast (CAS No. 153259-65-5) (GlaxoSmithKline), CR-1 (Nuada Pharmaceuticals), CRx-119 (CombinatoRx), D-5410 (UCB), dacopafant (CAS No. 125372-33-0) (Sanofi-Aventis), dersalazine (CAS No. 188913-57- 7/188913-58-8) (Uriach), etanercept (CAS No. 185243-69-0) (Amgen), ethyl pyruvate (Critical (Critical Therapeutics), golimumab (CAS No. 476181-74-5) (Johnson & Johnson), hormono-immunotherapy from Ipsen, CDP571 (e.g., Humicade from UCB), IC-485 (ICOS), infliximab (CAS No. 170277-31-3) (Johnson & Johnson), IP-751 (Manhattan Pharmaceuticals), ISIS-104838 (CAS No. 250755-32-9) (ISIS Pharmaceuticals), lenalidomide (CAS No. 191732-72-6) (Celgene), lentinan (CAS No. 37339-90-5) (Ajinomoto), MDL- 201112 (CAS No. 142130-73-2) (Sanofi-Aventis), medroxyprogesterone (CAS No. 520-85-4) (InKine Pharmaceutical), N-acetylcysteine (CAS No. 616-91-1) (Zambon), NBE-P2 (DIREVO Biotech), nerelimomab (CAS No. 162774-06-3) (Chiron), OM-294DP (OM PHARMA), onercept (CAS No. 199685-57-9) (Yeda), PASSTNF-alpha (Verigen), pentoxifylline or oxypentifylline (Sanofi-Aventis), Pharmaprojects No. 4091, 4241 , 4295, or 4488 (Sanofi-Aventis), Pharmaprojects No. 5480 (Amgen), Pharmaprojects No. 6749 (Cengent), pirfenidone (CAS No. 53179-13-8) (MARNAC), RPR- 132294 (Sanofi-Aventis), S5 (F002) (Fulcrum Pharmaceuticals), simvastatin (CAS No. 79902-63-9) (Merck & Co), STA-6292 (Synta Pharmaceuticals), tacrolimus (CAS No. 104987-11-3) (Fujisawa LifeCycle Pharma), talactoferrin alfa (CAS No. 308240-58-6) (Agennix), thalidomide (CAS No. 50-35-1) (Celgene), TNF antagonists form ProStrakan, and Synergen, TNF inhibitors (Amgen), TNF-alpha antagonists from Dynavax Technologies and Jerina AG (Germany), TNF-alpha inhibitors from IBFB Pharma and Xencor (Xencor), torbafylline (CAS No. 105102-21-4) (Sanofi-Aventis), UR-1505 (Uriach), VT-346 (Viron Therapeutics), YSIL6 (Y's Therapeutics), YSTH2 (Y's Therapeutics), NPI-1302a-3 (Nereus Pharmaceuticals, a TNF antagonist from Jerina AG (Germany), dersalazine, and an analogue or derivative thereof.
11129. The method of item 11030 wherein the agent is a
Toll receptor inhibitor. 11130. The method of item 11030 wherein the agent is a tubulin antagonist.
11131. The method of item 11030 wherein the agent is a tyrosine kinase inhibitor selected from the group consisting of SU-011248, SUTENT from Pfizer Inc. (New York, NY), BMS-354825, PN-355 (Paracelsian Pharmaceuticals), AGN-199659 (Allergan), AAL-993 or ABP- 309 (Novartis), adaphostin (NIH), AEE-788 (Novartis), AG-013736 (OSI Pharmaceuticals), AG-13736 (Pfizer), ALT-110 (Alteris Therapeutics), AMG- 706 (Amgen), anticancer MAbs from Xencor, anti-EGFrvlll MAbs from Abgenix, anti-HER2 MAb from Abiogen, AZD-2171 or AZD-9935 (AstraZeneca), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), CEP-5214 (Cephalon), CEP-7055 (Cephalon), cetuximab (ImClone Systems), CHIR-200131 and CHIR-258 (Chiron), CP-547632 (OSI Pharmaceuticals), CP-724714 (Pfizer), CT-301 (Creabilis Therapeutics), D- 69491 (Baxter International), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EGFR/ErbB2 inhibitors from Array BioPharma, erlotinib (CAS No. 183319- 69-9) (OSI Pharmaceuticals), EXEL-2880 (Exelixis), FK-778 (Sanofi- Aventis) , gefitinib (CAS No. 184475-35-2) (AstraZeneca), GW-2286 or GW- 654652 (GlaxoSmithKline), her2/neu antigen from AlphaVax, HER-2/neu inhibitor from Generex, Herzyme (Medipad) (Sirna Therapeutics), HKI-272 (Wyeth), HuMax-EGFr (Genmab), idronoxil (CAS No. 81267-65-4) (Novogen), IGF-1 inhibitors from Ontogen, IMC-11 F8 (ImClone Systems), kahalalide F (CAS No. 149204-42-2) (PharmaMar), KDR inhibitor from LG Life Sciences, KDR inhibitors from Abbott Laboratories, KDR kinase inhibitors (UCB), Kdr kinase inhibitors from Merck & Co, KRN-633 and KRN- 951 (Kirin Brewery), KSB-102 (Xenova), lapatinib ditosylate (CAS No. 388082-78-8) (GlaxoSmithKline), matuzumab (Merck KGaA), MDX-214 (Medarex), ME-103 (Pharmexa), MED-A300 (Gerolymatos), MNAC-13 (Lay Line Genomics), nimotuzumab (Center of Molecular Immunology), NSC- 330507 or NSC-707545 (NIH), NV-50 (Novogen), OSI-930 (OSI Pharmaceuticals), panitumumab (Abgenix), pelitinib (CAS No. 287933-82-7) (Wyeth), pertuzumab (CAS No. 380610-27-5) (Genentech), Pharmaprojects No. 3985 (Sanofi-Aventis), prostate cancer therapeutics from Sequenom (SQPC35, SQPC36, SQPC90), removab and remoxab (Trion Pharma), RG- 13022 (CAS No. 136831-48-6), RG-13291 (CAS No. 138989-50-1), or RG- 14620 (CAS No. 136831-49-7) (Sanofi-Aventis), RM-6427 (Romark), RNAi breast cancer therapy from Benitec, RP 53801 (CAS No. 125882-88-4) (Sanofi-Aventis), sorafenib tosylate (Bayer), SU-11657 (Pfizer), Tie-2 antagonists from Semaia (Hybrigenics), Tie-2 inhibitors from Ontogen, trastuzumab (CAS No. 180288-69-1) (Genentech), tyrosine kinase inhibitors from Sanofi-Aventis, U3-1287, U3-1565, U3-1784, U3-1800 (U3 Pharma), vatalanib (Novartis), VEGFR-2 kinase inhibitor from Bristol-Myers Squibb, XL-647 (Exelixis), ZD-6474 (AstraZeneca), ZK-CDK (Schering AG), an EGFR tyrosine kinase inhibitor, EKB-569 (Wyeth), and an analogue or derivative thereof.
11132. The method of item 11030 wherein the agent is a VEGF inhibitor.
11133. The method of item 11030 wherein the agent is a vitamin D receptor agonist.
11134. The method of item 11030 wherein the agent is ZD-6474 (an angiogenesis inhibitor).
11135. The method of item 11030 wherein the agent is AP-23573 (an mTOR inhibitor).
11136. The method of item 11030 wherein the agent is synthadotin (a tubulin antagonist). 11137. The method of item 11030 wherein the agent is S-0885 (a collagenase inhibitor).
11138. The method of item 11030 wherein the agent is aplidine (an elongation factor-1 alpha inhibitor).
11139. The method of item 11030 wherein the agent is ixabepilone (an epithilone).
11140. The method of item 11030 wherein the agent is IDN-5390 (an angiogenesis inhibitor).
11141. The method of item 11030 wherein the agent is SB-2723005 (an angiogenesis inhibitor).
11142. The method of item 11030 wherein the agent is ABT-518 (an angiogenesis inhibitor).
11143. The method of item 11030 wherein the agent is combretastatin (an angiogenesis inhibitor).
11144. The method of item 11030 wherein the agent is anecortave acetate (an angiogenesis inhibitor).
11145. The method of item 11030 wherein the agent is SB-715992 (a kinesin antagonist).
11146. The method of item 11030 wherein the agent is temsirolimus (an mTOR inhibitor). 11147. The method of item 11030 wherein the agent is adalimumab (a TNFα antagonist).
11148. The method of item 11030, wherein the composition comprises a polymer.
11149. The method of item 11030, wherein the composition comprises a polymeric carrier.
11150. The method of item 11030 wherein the anti- scarring agent inhibits adhesion between the device and a host into which the device is implanted.
11151. The method of item 11030 wherein the device delivers the anti-scarring agent locally to tissue proximate to the device.
11152. The method of item 11030 wherein the device has a coating that comprises the anti-scarring agent.
11153. The method of item 11030, wherein the device has a coating that comprises the agent and is disposed on a surface of the implant.
11154. The method of item 11030, wherein the device has a coating that comprises the agent and directly contacts the implant.
11155. The method of item 11030, wherein the device has a coating that comprises the agent and indirectly contacts the implant.
11156. The method of item 11030, wherein the device has a coating that comprises the agent and partially covers the implant. 11157. The method of item 11030, wherein the device has a coating that comprises the agent and completely covers the implant.
11158. The method of item 11030, wherein the device has a uniform coating.
11159. The method of item 11030, wherein the device has a non-uniform coating.
11160. The method of item 11030, wherein the device has a discontinuous coating.
11161. The method of item 11030, wherein the device has a patterned coating.
11162. The method of item 11030, wherein the device has a coating with a thickness of 100 Dm or less.
11163. The method of item 11030, wherein the device has a coating with a thickness of 10 Dm or less.
11164. The method of item 11030, wherein the device has a coating, and the coating adheres to the surface of the implant upon deployment of the implant.
11165. The method of item 11030, wherein the device has a coating, and wherein the coating is stable at room temperature for a period of 1 year. 11166. The method of item 11030, wherein the device has a coating, and wherein the anti-scarring agent is present in the coating in an amount ranging between about 0.0001% to about 1 % by weight.
11167. The method of item 11030, wherein the device has a coating, and wherein the anti-scarring agent is present in the coating in an amount ranging between about 1 % to about 10% by weight.
11168. The method of item 11030, wherein the device has a coating, and wherein the anti-scarring agent is present in the coating in an amount ranging between about 10% to about 25% by weight.
11169. The method of item 11030, wherein the device has a coating, and wherein the anti-scarring agent is present in the coating in an amount ranging between about 25% to about 70% by weight.
11170. The method of item 11030, wherein the device has a coating, and wherein the coating further comprises a polymer.
11171. The method of item 11030, wherein the device has a first coating having a first composition and a second coating having a second composition.
11172. The method of item 11030, wherein the device has a first coating having a first composition and a second coating having a second composition, wherein the first composition and the second composition are different.
11173. The method of item 11030, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a copolymer. 11174. The method of item 11030, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a block copolymer.
11175. The method of item 11030, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a random copolymer.
11176. The method of item 11030, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a biodegradable polymer.
11177. The method of item 11030, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a non-biodegradable polymer.
11178. The method of item 11030, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a hydrophilic polymer.
11179. The method of item 11030, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a hydrophobic polymer.
11180. The method of item 11030, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a polymer having hydrophilic domains.
11181. The method of item 11030, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a polymer having hydrophobic domains. 11182. The method of item 11030, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a non-conductive polymer.
11183. The method of item 11030, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises an elastomer.
11184. The method of item 11030, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a hydrogel.
11185. The method of item 11030, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a silicone polymer.
11186. The method of item 11030, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a hydrocarbon polymer.
11187. The method of item 11030, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a styrene-derived polymer.
11188. The method of item 11030, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a butadiene polymer.
11189. The method of item 11030, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a macromer. 11190. The method of item 11030, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a poly(ethylene glycol) polymer.
11191. The method of item 11030 wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises an amorphous polymer.
11192. The method of item 11030, wherein the device comprises a lubricious coating.
11193. The method of item 11030 wherein the anti- scarring agent is located within pores or holes of the device.
11194. The method of item 11030 wherein the anti- scarring agent is located within a channel, lumen, or divet of the device. _
11195. The method of item 11030, wherein the device comprises a second pharmaceutically active agent.
11196. The method of item 11030 wherein the device comprises an anti-inflammatory agent.
11197. The method of item 11030 wherein the device comprises an agent that inhibits infection.
11198. The method of item 11030 wherein the device comprises an agent that inhibits infection, and wherein the agent is an anthracycline. 11199. The method of item 11030 wherein the device comprises an agent that inhibits infection, and wherein the agent is doxorubicin.
11200. The method of item 11030 wherein the device comprises an agent that inhibits infection, and wherein the agent is mitoxantrone.
11201. The method of item 11030 wherein the device comprises an agent that inhibits infection, and wherein the agent is a fluoropyrimidine.
11202. The method of item 11030 wherein the device comprises an agent that inhibits infection, and wherein the agent is 5- fluorouracil (5-FU).
11203. The method of item 11030 wherein the device comprises an agent that inhibits infection, and wherein the agent is a folic acid antagonist.
11204. The method of item 11030 wherein the device comprises an agent that inhibits infection, and wherein the agent is methotrexate.
11205. The method of item 11030 wherein the device comprises an agent that inhibits infection, and wherein the agent is a podophylotoxin.
11206. The method of item 11030 wherein the device comprises an agent that inhibits infection, and wherein the agent is etoposide. 11207. The method of item 11030 wherein the device comprises an agent that inhibits infection, and wherein the agent is a camptothecin.
11208. The method of item 11030 wherein the device comprises an agent that inhibits infection, and wherein the agent is a hydroxyurea.
11209. The method of item 11030 wherein the device comprises an agent that inhibits infection, and wherein the agent is a platinum complex.
11210. The method of item 11030 wherein the device comprises an agent that inhibits infection, and wherein the agent is cisplatin.
11211. The method of item 11030, further comprising an antithrombotic agent.
11212. The method of item 11030 wherein the device comprises a visualization agent.
11213. The method of item 11030 wherein the device comprises a visualization agent, wherein the visualization agent is a radiopaque material, and wherein the radiopaque material comprises a metal, a halogenated compound, or a barium containing compound.
11214. The method of item 11030 wherein the device comprises a visualization agent, wherein the visualization agent is a radiopaque material, and wherein the radiopaque material comprises barium, tantalum, or technetium. 11215. The method of item 11030 wherein the device comprises a visualization agent, and wherein the visualization agent is a MRI responsive material.
11216. The method of item 11030 wherein the device comprises a visualization agent, and wherein the visualization agent comprises a gadolinium chelate.
11217. The method of item 11030 wherein the device comprises a visualization agent, and wherein the visualization agent comprises iron, magnesium, manganese, copper, or chromium.
11218. The method of item 11030 wherein the device comprises a visualization agent, and wherein the visualization agent comprises an iron oxide compound.
11219. The method of item 11030 wherein the device comprises a visualization agent, and wherein the visualization agent comprises a dye, pigment, or colorant.
11220. The method of item 11030 wherein the device comprises an echogenic material.
11221. The method of item 11030 wherein the device comprises an echogenic material, and wherein the echogenic material is in the form of a coating.
11222. The method of item 11030 wherein the device is sterile. 11223. The method of item 11030 wherein the anti- scarring agent is released into tissue in the vicinity of the device after deployment of the device.
11224. The method of item 11030 wherein the anti- scarring agent is released into tissue in the vicinity of the device after deployment of the device, and wherein the tissue is connective tissue.
11225. The method of item 11030 wherein the anti- scarring agent is released into tissue in the vicinity of the device after deployment of the device, and wherein the tissue is muscle tissue.
11226. The method of item 11030 wherein the anti- scarring agent is released into tissue in the vicinity of the device after deployment of the device, and wherein the tissue is nerve tissue.
11227. The method of item 11030 wherein the anti- scarring agent is released into tissue in the vicinity of the device after deployment of the device, and wherein the tissue is epithelium tissue.
11228. The method of item 11030 wherein the anti- scarring agent is released in effective concentrations from the device over a period ranging from the time of deployment of the device to about 1 year.
11229. The method of item 11030 wherein the anti- scarring agent is released in effective concentrations from the device over a period ranging from about 1 month to 6 months.
11230. The method of item 11030 wherein the anti- scarring agent is released in effective concentrations from the device over a period ranging from about 1 - 90 days. 11231. The method of item 11030 wherein the anti- scarring agent is released in effective concentrations from the device at a constant rate.
11232. The method of item 11030 wherein the anti- scarring agent is released in effective concentrations from the device at an increasing rate.
11233. The method of item 11030 wherein the anti- scarring agent is released in effective concentrations from the device at a decreasing rate.
11234. The method of item 11030 wherein the anti- scarring agent is released in effective concentrations from the composition comprising the anti-scarring agent by diffusion over a period ranging from the time of deployment of the device to about 90 days.
11235. The method of item 11030 wherein the anti- scarring agent is released in effective concentrations from the composition comprising the anti-scarring agent by erosion of the composition over a period ranging from the time of deployment of the device to about 90 days.
11236. The method of item 11030 wherein the device comprises about 0.01 Dg to about 10 Dg of the anti-scarring agent.
11237. The method of item 11030 wherein the device comprises about 10 Dg to about 10 mg of the anti-scarring agent.
11238. The method of item 11030 wherein the device comprises about 10 mg to about 250 mg of the anti-scarring agent. 11239. The method of item 11030 wherein the device comprises about 250 mg to about 1000 mg of the anti-scarring agent.
11240. The method of item 11030 wherein the device comprises about 1000 mg to about 2500 mg of the anti-scarring agent.
11241. The method of item 11030 wherein a surface of the device comprises less than 0.01 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
11242. The method of item 11030 wherein a surface of the device comprises about 0.01 Dg to about 1 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
11243. The method of item 11030 wherein a surface of the device comprises about 1 Dg to about 10 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
11244. The method of item 11030 wherein a surface of the device comprises about 10 Dg to about 250 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
11245. The method of item 11030 wherein a surface of the device comprises about 250 Dg to about 1000 Dg of the anti-scarring agent of anti-scarring agent per mm2 of device surface to which the anti- scarring agent is applied.
11246. The method of item 11030 wherein a surface of the device comprises about 1000 Dg to about 2500 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied. 11247. The method of item 11030 wherein the combining is performed by direct affixing the agent or the composition to the implant.
11248. The method of item 11030 wherein the combining is performed by spraying the agent or the component onto the implant.
11249. The method of item 11030 wherein the combining is performed by electrospraying the agent or the composition onto the implant.
11250. The method of item 11030 wherein the combining is performed by dipping the implant into a solution comprising the agent or the composition.
11251. The method of item 11030 wherein the combining is performed by covalently attaching the agent or the composition to the implant.
11252. The method of item 11030 wherein the combining is performed by non-covalently attaching the agent or the composition to the implant.
11253. The method of item 11030 wherein the combining is performed by coating the implant with a substance that contains the agent or the composition.
11254. The method of item 11030 wherein the combining is performed by coating the implant with a substance that absorbs the agent. 11255. The method of item 11030 wherein the combining is performed by interweaving a thread composed of, or coated with, the agent or the composition.
11256. The method of item 11030 wherein the combining is performed by covering all the implant with a sleeve that contains the agent or the composition.
11257. The method of item 11030 wherein the combining is performed by covering a portion of the implant with a sleeve that contains the agent or the composition.
11258. The method of item 11030 wherein the combining is performed by covering all the implant with a cover that contains the agent or the composition.
11259. The method of item 11030 wherein the combining is performed by covering a portion of the implant with a cover that contains the agent or the composition.
11260. The method of item 11030 wherein the combining is performed by covering all the implant with an electrospun fabric that contains the agent or the composition.
11261. The method of item 11030 wherein the combining is performed by covering a portion of the implant with an electrospun fabric that contains the agent or the composition.
11262. The method of item 11030 wherein the combining is performed by covering all the implant with a mesh that contains the agent or the composition. 11263. The method of item 11030 wherein the combining is performed by covering a portion of the implant with a mesh that contains the agent or the composition.
11264. The method of item 11030 wherein the combining is performed by constructing all the implant with the agent or the composition.
11265. The method of item 11030 wherein the combining is performed by constructing a portion of the implant with the agent or the composition.
11266. The method of item 11030 wherein the combining is performed by impregnating the implant with the agent or the composition.
11267. The method of item 11030 wherein the combining is performed by constructing all of the implant from a degradable polymer that releases the agent.
11268. The method of item 11030 wherein the combining is performed by constructing a portion of the implant from a degradable polymer that releases the agent.
11269. The method of item 11030 wherein the combining is performed by dipping the implant into a solution that comprise the agent and an inert solvent for the implant.
11270. The method of item 11030 wherein the combining is performed by dipping the implant into a solution that comprises the agent and a solvent that will swell the implant. 11271. The method of item 11030 wherein the combining is performed by dipping the implant into a solution that comprises the agent and a solvent that will dissolve the implant.
11272. The method of item 11030 wherein the combining is performed by dipping the implant into a solution that comprises the agent, a polymer and an inert solvent for the implant.
11273. The method of item 11030 wherein the combining is performed by dipping the implant into a solution that comprises the agent, a polymer and a solvent that will swell the implant.
11274. The method of item 11030 wherein the combining is performed by dipping the implant into a solution that comprises the agent, a polymer and a solvent that will dissolve the implant.
11275. The method of item 11030 wherein the combining is performed by spraying the implant into a solution that comprises the agent and an inert solvent for the implant.
11276. The method of item 11030 wherein the combining is performed by spraying the implant into a solution that comprises the agent and a solvent that will swell the implant.
11277. The method of item 11030 wherein the combining is performed by spraying the implant into a solution that comprises the agent and a solvent that will dissolve the implant.
11278. The method of item 11030 wherein the combining is performed by spraying the implant into a solution that comprises the agent, a polymer and an inert solvent for the implant. 11279. The method of item 11030 wherein the combining is performed by spraying the implant into a solution that comprises the agent, a polymer and a solvent that will swell the implant.
11280. The method of item 11030 wherein the combining is performed by spraying the implant into a solution that comprises the agent, a polymer and a solvent that will dissolve the implant.
11281. A method as in any one of items 11030-11280, wherein the device is an intravascular device.
11282. A method as in any one of items 11030-11280, wherein the device is a gastrointestinal stent.
11283. A method as in any one of items 11030-11280, wherein the device is a tracheal and bronchial stent.
11284. A method as in any one of items 11030-11280, wherein the device is a genital urinary stent.
11285. A method as in any one of items 11030-11280, wherein the device is an ear and nose stent.
11286. A method as in any one of items 11030-11280, wherein the device is an ear ventilation.
11287. A method as in any one of items 11030-11280, wherein the device is an intraocular implant.
11288. A method as in any one of items 11030-11280, wherein the device is a vascular graft. 11289. A method as in any one of items 11030-11280, wherein the device comprises a film or a mesh.
11290. A method as in any one of items 11030-11280, wherein the device is a glaucoma drainage device.
11291. A method as in any one of items 11030-11280, wherein the device is a prosthetic heart valve or a component thereof.
11292. A method as in any one of items 11030-11280, wherein the device is a penile implant.
11293. A method as in any one of items 11030-11280, wherein the device is an endotracheal or tracheostomy tube.
11294. A method as in any one of items 11030-11280, wherein the device is a peritoneal dialysis catheter.
11295. A method as in any one of items 11030-11280, wherein the device is a central nervous system shunt or a pressure monitoring device.
11296. A method as in any one of items 11030-11280, wherein the device is an inferior vena cava filter.
11297. A method as in any one of items 11030-11280, wherein the device is a gastrointestinal device.
11298. A method as in any one of items 11030-11280, wherein the device is a central venous catheter. 11299. A method as in any one of items 11030-11280, wherein the device is a ventricular assist device.
11300. A method as in any one of items 11030-11280, wherein the device is a spinal implant.
11301. A method as in any one of items 11030-11280, wherein the device is an implantable electrical device.
11302. A method as in any one of items 11030-11280, wherein the device is an implantable sensor.
11303. A method as in any one of items 11030-11280, wherein the device is an implantable pump.
11304. A method as in any one of items 11030-11280, wherein the device is a soft tissue implant.
11305. A method of making a medical device comprising: combining an intravascular implant and an anti-scarring agent or a composition comprising an anti-scarring agent, wherein the agent inhibits scarring between the device and a host into which the device is implanted.
11306. The method of item 11305 wherein the agent is an adensosine A2A receptor antagonist.
11307. The method of item 11305 wherein the agent is an AKT inhibitor. 11308. The method of item 11305 wherein the agent is an alpha 2 integrin antagonist, wherein the alpha 2 integrin antagonist is Pharmaprojects No. 5754 (Merck KgaA).
11309. The method of item 11305 wherein the agent is an alpha 4 integrin antagonist.
11310. The method of item 11305 wherein the agent is an alpha 7 nicotinic receptor agonist.
11311. The method of item 11305 wherein the agent is an angiogenesis inhibitor selected from the group consisting of AG-12,958 (Pfizer), ATN-161 (Attention LLC), neovastat, an angiogenesis inhibitor from Jerina AG (Germany), NM-3 (Mercian), VGA-1155 (Taisho), FCE-26644 (Pfizer), FCE-26950 (Pfizer), FPMA (Meiji Daries), FFM 11142 (Fujisawa), GGTI-298, GM-1306 (Ligand), GPA-1734 (Novartis), NNC-47-0011 (Noyo . Nordisk), herbamycin (Nippon Kayaku), lenalidomide (Celegene), IP-10 (NIH), ABT-828 (Abbott), KIN-841 (Tokushima University, Japan), SF-1126 (Semafore Pharmaceuticals), laminin technology (NIH), CHIR-258 (Chiron), NVP-AEW541 (Novartis), NVP-AEW541 (Novartis), Vt16907 (Alchemia), OXI-8007 (Oxigene), EG-3306 (Ark Therapeutics), Maspin (Arriva), ABT- 567 (Abbott), PPI-2458 (Praecis Pharmaceuticals), CC-5079, CC-4089 (Celgene), HIF-1 alpha inhibitors (Xenova), S-247 (Pfizer), AP-23573 (Ariad), AZD-9935 (Astra Zeneca), mebendazole (Introgen Therapeutics), MetAP-2 inhibitors (GlaxoSmithKline), AG-615 (Angiogene Pharmaceuticals), Tie-2 antagonists (Hybrigenics), NC-381, CYC-381, NC-169, NC-219, NC-383, NC-384, NC-407 (Lorus Therapeutics), ATN-224 (Attenuon), ON-01370 (Onconova), Vitronectin antagonists (Amgen), SDX-103 (Salmedix), Vitronectin antagonists (Shire), CHP (Riemser), TEK (Amgen), Anecortave acetate (Alcon), T46.2 (Matrix Therapeutics), HG-2 (Heptagen), TEM antagonists (Genzyme), Oxi-4500 (Oxigene), ATN-161 (Attenuon), WX-293 (Wilex), M-2025 (Metris Therapeutics), Alphastatin (BioActa), YH-16 (Yantai Rongchang), BIBF-1120 (Boehringer Ingelheim), BAY-57-9352 (Bayer), AS- 1404 (Cancer Research Technology), SC-77964 (Pfizer), glycomimetics (BioTie Therapies), TIE-2 Inhibitors (Ontogen), DIMI, Octamer (Octamer), ABR-215050 (Active Biotech), ABT-518 (Abbott), KDR inhibitors (Abbott), BSF-466895 (Abbott), SCH-221153 (Schering-Plough), DAC:antiangiogenic (ConjuChem), TFPI (EntreMed), AZD-2171 (Astra-Zenaca), CDC-394 (Celgene), LY290293 (EIi Lilly), IDN-5390 (Indena), Kdr Kinase Inhibitors (Merck), CT-113020, CT-116433, CT-116563, CT-31890, CT-32228) (Cell Therapeutics), A-299620 (Abbott), TWEAK Inhibitor (Amgen), VEGF modulators (Johnson and Johnson), Tum-N53, tumstatin (Genzyme), Thios- 1 , Thios-2 (Thios Pharmaceuticals), MV-6401 (Miravant Medical Technologies), Spisulosine (PharmaMar), CEP-7055 (Cephalon), AUV-201 (Auvation), LM-609 (EIi Lilly), SKF-106615 (AnorMED), Oglufanide disodium (Cytran), BW-114 (Pharminox), Calreticulin (NIH), WX-678 (Wilex), SD-7784 (Pfizer), WX-UK1 (Wilex), SH-268 (Schering AG), 2-Me-PGA (Celgene), S- 137 (Pfizer), ZD-6126 (Angiogene Pharmaceuticals), SG-292 (SignalGen), Benefin (Lane Labs), A6, A36 (Angstrom), SB-2723005 (GlaxoSmithKline), SC-7 (Cell Therapeutics), ZEN-014 (AEterna Zentaris), 2-methoxyestradiol (EntreMed), NK-130119 (Nippon Kayaku), CC-10004 (Celgene), AVE- 8062A (Ajinomoto), Tacedinaline (Pfizer), Actinonin (Tokyo Metropolitan Institute of Medical Science), Lenalidomide (Celgene), VGA-1155, BTO-956 (SRI International), ER-68203-00 (Eisai), CT-6685 (UCB), JKC-362 (Phoenix Pharmaceuticals), DMI-3798 (DMI Biosciences, Angiomate (Ipsen), ZD-6474 (AstraZeneca), CEP-5214 (Cephalon), Canstatin (Genzyme), NM-3 (Mercian), Oridigm (MediQuest Therapeutics), Exherin (Adherex), BLS-0597 (Boston Life Sciences), PTC-299 (PTC Therapeutics), NPI-2358 (Nereus Pharmaceuticals), CGP-79787 (Novartis), AEE-788 (Novartis), CKD-732 (Chong Kun Dang), CP-564959 (OSI Pharmaceuticals), CM-101 (CarboMed), CT-2584, CT3501 (Cell Therapeutics), combretastatin and analogues and derivatives thereof (Oxigene), Rebimastat (Bristol-Meyers Squibb), Dextrin 2-sulfate (ML Laboratories), Cilengitide (Merk KGaA), NSC-706704 (Pharminox), KRN- 951 (Kirin Brewery), Ukrain, NSC-631570 (Nowicky Pharma), Tecogalan sodium (Daiichi Pharmaceutical), Tz-93 (Tsumura), TBC-1635 (Encysive Pharmaceuticals), TAN-1120 (Takeda), Semaxanib (Pfizer), BDI-7800 (Biopharmacopae), SD-186, SD-983 (Bristol-Meyers Squibb), SB-223245 (GlaxoSmithKline), SC-236 (Pfizer), RWJ-590973 (Johnson and Johnson), ILX-1850 (Genzyme), SC-68488, S-836 (Pfizer), CG-55069-11 (CuraGen), Ki-23057 (Kirin Brewery), CCX-700 (Chemoentryx), Pegaptanib octasodium (Giled Sciences), ANGIOCOL (available from Biostratum Inc.), or an analogue or derivative thereof.
11312. The method of item 11305 wherein the agent is an apoptosis antagonist.
11313. The method of item 11305 wherein the agent is an apoptosis activator.
11314. The method of item 11305 wherein the agent is a beta 1 integrin antagonist.
11315. The method of item 11305 wherein the agent is a beta tubulin inhibitor.
11316. The method of item 11305 wherein the agent is a blocker of enzyme production in Hepatitis C.
11317. The method of item 11305 wherein the agent is a Bruton's tyrosine kinase inhibitor. 11318. The method of item 11305 wherein the agent is a calcineurin inhibitor.
11319. The method of item 11305 wherein the agent is a caspase 3 inhibitor.
11320. The method of item 11305 wherein the agent is a CC chemokine receptor antagonist.
11321. The method of item 11305 wherein the agent is a cell cycle inhibitor selected from the group consisting of SNS-595 (Sunesis), synthadotin, KRX-0403, and an analogue or derivative thereof.
11322. The method of item 11305 wherein the agent is a cathepsin B inhibitor.
11323. The method of item 11305 wherein the agent is a cathepsin K inhibitor, wherein the cathepsin K inhibitor is 462795 (GlaxoSmithKline), INPL-022-D6 (Amura Therapeutics), or an analogue or derivative thereof.
11324. The method of item 11305 wherein the agent is a cathepsin L inhibitor.
11325. The method of item 11305 wherein the agent is a CD40 antagonist.
11326. The method of item 11305 wherein the agent is a chemokine receptor agonist. 11327. The method of item 11305 wherein the agent is a chymase inhibitor.
11328. The method of item 11305 wherein the agent is a collagenase antagonist.
11329. The method of item 11305 wherein the agent is a CXCR antagonist.
11330. The method of item 11305 wherein the agent is a cyclin dependent kinase inhibitor selected from the group consisting of a CDK-1 inhibitor, a CDK-2 inhibitor, a CDK- 4 inhibitor, a CDK-6 inhibitor, a CAK1 inhibitor from GPC Biotech and Bristol-Myers Squibb, RGB-286199 (GPC Biotech), an anticancer agent from Astex Technology, a CAK1 inhibitor from GPC Biotech, a CDK inhibitor from Sanofi-Aventis, a CDK1/CDK2 inhibitor from Amgen, a CDK2 inhibitor from SUGEN-2 (Pfizer), a hearing loss therapy agent (Sound Pharmaceuticals), PD-0332991 (Pfizer), RGB-286199 (GPC Biotech), Ro-0505124 (Hoffmann-La Roche), a Ser/Thr kinase inhibitor from Lilly (EIi Lilly), CVT-2584 (CAS No. 199986-75- 9) (CV Therapeutics), and an analogue or derivative thereof.
11331. The method of item 11305 wherein the agent is a cyclooxygenase 1 inhibitor.
11332. The method of item 11305 wherein the agent is a DHFR inhibitor.
11333. The method of item 11305 wherein the agent is a dual integrin inhibitor. 11334. The method of item 11305 wherein the agent is an elastase inhibitor.
11335. The method of item 11305 wherein the agent is an elongation factor-1 alpha inhibitor.
11336. The method of item 11305 wherein the agent is an endothelial growth factor antagonist.
11337. The method of item 11305 wherein the agent is an endothelial growth factor receptor kinase inhibitor selected from the group consisting of sorafenib tosylate (Bayer), AAL-993 (Novartis), ABP-309 (Novartis), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), E- 7080 (Eisai), EG-3306 (Ark Therapeutics), EXEL-2880 (Exelixis), GW- 654652 (GlaxoSmithKline), a KDR inhibitor from LG Life Sciences, CT-6685 and CT-6729 (UCB), KRN-633 and KRN-951 (Kirjn Brewery), OSI-930 (OSI Pharmaceuticals), SP-5.2 (Supratek Pharma), SU-11657 (Pfizer), a Tie-2 antagonist (Hybrigenics), a VEGFR-2 kinase inhibitor (Bristol-Myers Squibb), XL-647 (Exelixis), a KDR inhibitor from Abbott Laboratories, sorafenib tosylate, and an analogue or derivative thereof.
11338. The method of item 11305 wherein the agent is an endotoxin antagonist.
11339. The method of item 11305 wherein the agent is an epothilone and tubulin binder.
11340. The method of item 11305 wherein the agent is an estrogen receptor antagonist. 11341. The method of item 11305 wherein the agent is an FGF inhibitor.
11342. The method of item 11305 wherein the agent is a farnexyl transferase inhibitor.
11343. The method of item 11305 wherein the agent is farnesyltransferase inhibitor selected from the group of A-197574 (Abbott), a famesyltransferase inhibitor from Servier, FPTIII (Strathclyde Institute for Drug R), LB-42908 (LG Life Sciences), Pharmaprojects No. 5063 (Genzyme), Pharmaprojects No. 5597 (Ipsen), Yissum Project No. B-1055 (Yissum), and an analogue or derivative thereof.
11344. The method of item 11305 wherein the agent is an FLT-3 kinase inhibitor.
11345. The method of item 11305 wherein the agent is an FGF receptor kinase inhibitor.
11346. The method of item 11305 wherein the agent is a fibrinogen antagonist selected from the group consisting of AUV-201 (Auvation), MG-13926 (Sanofi-Aventis), plasminogen activator (CAS No. 105913-11-9) (from Sanofi-Aventis or UCB), plasminogen activator-2 (tPA- 2) (Sanofi-Aventis), pro-urokinase (CAS No. 82657-92-9) (Sanofi-Aventis), and an analogue or derivative thereof.
11347. The method of item 11305 wherein the agent is a heat shock protein 90 antagonist selected from the group consisting of SRN- 005 (Sirenade), geldanamycin, NSC-33050 (17-allylaminogeldanamycin; 17- AAG), 17-dimethylaminoethylamino-17-demethoxy-geldanamycin (17- DMAG), rifabutin (rifamycin XIV1 1',4-didehydro-1-deoxy-1 ,4-dihydro-5'-(2- methylpropyl)-1-oxo-), and an analogue or derivative thereof.
11348. The method of item 11305 wherein the agent is a histone deacetylase inhibitor.
11349. The method of item 11305 wherein the agent is an HMGCoA reductase inhibitor selected from the group consisting of an atherosclerosis therapeutic from Lipid Sciences, ATI-16000 (ARYx Therapeutics), KS-01-019 (Kos Pharmaceuticals), Pharmaprojects No. 2197 (Sanofi-Aventi), RP 61969 (Sanofi-Aventis), and an analogue or derivative thereof.
11350. The method of item 11305 wherein the agent is an ICAM inhibitor.
11351. The method of item 11305 wherein the agent is an IL, ICE and IRAK antagonist, wherein the antagonist is a CJ-14877, CP- 424174 (Pfizer), NF-61 (Negma-Lerads), and an analogue or derivative thereof.
11352. The method of item 11305 wherein the agent is an IL-2 inhibitor.
11353. The method of item 11305 wherein the agent is an immunosuppressant selected from the group consisting of teriflunomide (Sanofi Aventis), chlorsulfaquinoxalone (NSC-339004), chlorsulfaquinoxalone sulfate, CS-712 (Sankyo), ismomultin alfa (CAS No. 457913-93-8) (Akzo Nobel), antiallergics from GenPat77, antiinflammatories or AT-005 (Androclus Therapeutics), autoimmune disease therapeutics from EpiVax, BN-007 (Bone), budesonide (CAS No. 51333-22- 3) (MAP Pharmaceuticals), CO-14 (Genzyme), edratide (CAS No. 433922- 67-9) (Teva), EP-314 (Enanta), eprovafen (CAS No. 101335-99-3) (Sanofi- Aventis), HWA-131 (CAS No. 118788-41-3) (Sanofi-Aventis), immunomodulators from MerLion Pharmaceuticals, immunosuppressives from Alchemia, IPL-12 (Inflazyme), MDL-9563 (CAS No. 27086-86-8) (Sanofi-Aventis), Pharmaprojects No. 2330 (Sanofi-Aventis), Pharmaprojects No. 6426 (Abgenix), PXS-25 (Pharmaxis), rosmarinic acid (CAS No. 20283-92-5) (Sanofi-Aventis), RP 42927 or RP 54745 (CAS No. 135330-08-4) (Sanofi-Aventis), SGN-35 (Seattle Genetics), ST-1959 (Sigma-Tau), type I diabetes therapy from SYNX Pharma, UNIL-88 (Debiopharm), VP-025 (Vasogen), VR-694 (Vectura), PRTX-001 (Protalex), and an analogue or derivative thereof.
11354. The method of item 11305 wherein the agent is an IMPDH (inosine monophosphate).
11355. The method of item 11305 wherein the agent is an integrin antagonist.
11356. The method of item 11305 wherein the agent is an interleukin antagonist.
11357. The method of item 11305 wherein the agent is an inhibitor of type III receptor tyrosine kinase.
11358. The method of item 11305 wherein the agent is an irreversible inhibitor of enzyme methionine aminopeptidase type 2.
11359. The method of item 11305 wherein the agent is an isozyme selective delta protein kinase C inhibitor. 11360. The method of item 11305 wherein the agent a
JAK3 enzyme inhibitor.
11361. The method of item 11305 wherein the agent is a
JNK inhibitor.
11362. The method of item 11305 wherein the agent is a kinase inhibitor.
11363. The method of item 11305 wherein the agent is kinesin antagonist.
11364. The method of item 11305 wherein the agent is a kinesin antagonist.
11365. The method of item 11305 wherein the agent is a leukotriene inhibitor and antagonist selected from the group consisting of ambicromil (CAS No. 58805-38-2) (Sanofi-Aventis), amelubant (CAS No. 346735-24-8) (Boehringer Ingelheim), DW-1141 (Dong Wha), ebselen (Daiichi Pharmaceutical), ibudilast (Kyorin), leucotriene inhibitors from Sanofi-Aventis, lymphotoxin-beta receptor (LT-D) from Biogen Idee, Pharmaprojects No. 1535 and 2728 (CAS No. 119340-33-9) (Sanofi- Aventis), R-112 (Rigel), Rev-5367 (CAS No. 92532-05-3) (Sanofi-Aventis), RG-14893 (CAS No. 141835-49-6) (Sanofi-Aventis), RG-5901-A (CAS No. 101910-24-1), 92532-23-5, RP 66153 (CAS No. 142422-79-5), RP 66364 (CAS No.186916-92-5), RP 69698 (CAS No. 141748-00-7) (Sanofi-Aventis), SC-411930 (Pfizer), SC-41930 (CAS No. 120072-59-5) (Pfizer), SC-50605 (CAS No. 138828-39-4) (Pfizer), SC-51146 (CAS No. 141059-52-1), SC- 53228 (CAS No. 153633-01-3) (Pfizer), spaglumic acid (ZY-15106) (CAS No.3106-85-2), 80619-64-3 (Novartis), tipredane (CAS No. 85197-77-9) (Bristol-Myers Squibb), U-75302 (CAS No. 119477-85-9) (Pfizer), and analogue or derivative thereof.
11366. The method of item 11305 wherein the agent is an MAP kinase inhibitor.
11367. The method of item 11305 wherein the agent is a matrix metalloproteinase inhibitor.
11368. The method of item 11305 wherein the agent is an MCP-CCR2 inhibitor.
11369. The method of item 11305 wherein the agent is an mTOR inhibitor.
11370. The method of item 11305 wherein the agent is an mTOR kinase inhibitor.
11371. The method of item 11305 wherein the agent is a microtubule inhibitor selected from the group consisting of antibody- maytansinoid conjugates from Biogen Idee, colchicines (MantiCore Pharmaceuticals), anticancer immunoconjugates from Johnson & Johnson, DIME from Octamer, gni-1f (GNI), huC242-DM4, huMy9-6-DM1 (ImmunoGen), IDN-5404 (Indena), IMO-098, IMOderm (Imotep), mebendazole (Introgen Therapeutics), microtubule poisons from Cambridge Enterprise, paclitaxel such as LOTAX from Aphios (CAS No. 33069-62-4), Genexol-PM from Samyang, Pharmaprojects No. 6383 (Tapestry Pharmaceuticals), RPR-112378 (Sanofi-Aventis), SGN-75 (Seattle Genetics), SPL-7435 (Starpharma), SSR-250411 (Sanofi-Aventis), trastuzumab-DM1 (Genentech), vinorelbine, and an analogue or derivative thereof. 11372. The method of item 11305 wherein the agent is an MIF inhibitor.
11373. The method of item 11305 wherein the agent is an MMP inhibitor.
11374. The method of item 11305 wherein the agent is a neurokinin (NK) antagonist selected from the group consisting of anthrotainin (CAS No. 148084-40-6) (Sanofi-Aventis), an IBS thereapeutic from ArQuIe1 MDL-105212A (CAS No. 167261-60-1) (Ssanofi-Aventis), Pharmaprojects No. 2744, 3258 (CAS No. 139167-47-8) 4006, 4201 , or 5986 (Sanofi-Aventis), RP 67580 (CAS No. 135911-02-3), SR-144190 (CAS No. 201152-86-5), SSR-240600, SSR-241586 (Sanofi-Aventis), TKA-457 (Novartis), vestipitant mesylate (CAS No. 334476-64-1) (GlaxoSmithKline), Win-64821 (Sanofi-Aventis), PRX-96026 (Predix Pharmaceuticals), and an analogue or derivative thereof.
11375. The method of item 11305 wherein the agent is an NF kappa B inhibitor selected from the group consisting of AVE-0545 or AVE-0547 (Sanofi-Aventis), bortezomib (CAS No. 179324-69-7) (Millennium Pharmaceuticals), dexanabinol (CAS No. 112924-45-5) (Pharmos), dexlipotam (Viatris), Pharmaprojects No. 6283 (INDRA) (OXiGENE), IPL- 576092 (CAS No. 137571-30-3) (Inflazyme), NFKB decoy (Corgentech), NFKB decoy oligo (AnGes MG), NFKB's from Ariad, osteoporosis treatments or S5 (F005) from Fulcrum Pharmaceuticals, P61 (Phytopharm), R-flurbiprofen (CAS No. 5104-49-4) (Encore Pharmaceuticals), and an analogue or derivative thereof.
11376. The method of item 11305 wherein the agent is a nitric oxide agonist. 11377. The method of item 11305 wherein the agent is an ornithine decarboxylase inhibitor.
11378. The method of item 11305 wherein the agent is a p38 MAP kinase inhibitor selected from the group consisting of AZD-6703 (AstraZeneca), JX-401 (Jexys Pharmaceuticals), BMS-2 (Bristol-Myers Squibb), a p38 MAP kinase inhibitor from Novartis, a p38-alpha MAP kinase inhibitor from Amphora, Pharmaprojects No. 5704 (Pharmacopeia), RPR- 200765A (Sanofi-Aventis), SD-282 (Johnson & Johnson), TAK-715 (Takeda), and an analogue or derivative thereof.
11379. The method of item 11305 wherein the agent is a palmitoyl-protein thioesterase inhibitor.
11380. The method of item 11305 wherein the agent is a PDGF receptor kinase inhibitor selected from the group consisting.of AAL- 993, AMN-107, or ABP-309 (Novartis), AMG-706 (Amgen), BAY-57-9352 (Bayer), CDP-860 (UCB), E-7080 (Eisai), imatinib (CAS No. 152459-95-5) (Novartis), OSI-930 (OSI Pharmaceuticals), RPR-127963E (Sanofi-Aventis), RWJ-540973 (Johnson & Johnson), sorafenib tosylate (Bayer), SU-11657 (Pfizer), tandutinib (CAS No. 387867-13-2) (Millennium Pharmaceuticals), vatalanib (Novartis), ZK-CDK (Schering AG), and an analogue or derivative thereof.
11381. The method of item 11305 wherein the agent is (-)-halofenate (Metabolex), AMG-131 (Amgen), antidiabetics from Japan Tobacco, AZD-4619, AZD-8450, AZD-8677 (AstraZeneca), DRF-10945, balaglitazone (Dr Reddy's), CS-00088, CS-00098 (Chipscreen Biosciences), E-3030 (Eisai), etalocib (CAS No. 161172-51-6) (EIi Lilly), GSK-641597 (Ligand), GSK-677954 (GlaxoSmithKline), GW-409544 (Ligand), GW- 590735 (GlaxoSmithKline), K-111 (Hoffmann-La Roche), LY-518674 (EIi Lilly), LY-674 (Ligand), LY-929 (Ligand), MC-3001 , MC-3002 (MaxoCore Pharmaceuticals), metformin HCI + pioglltazone (CAS No. 1115-70-4 and 112529-15-4), ACTOPLUS MET from Andrx), muraglitazar (CAS No. 331741-94-7) (Bristol-Myers Squibb), naveglitazar (Ligand), oleoylethanolamide (Kadmus Pharmaceuticals), ONO-5129, pioglitazone hydrochloride (CAS No. 111025-46-8 and 112529-15-4) (Takeda), PLX-204 (Plexxikon), PPAR agonists from Genfit, PPAR delta agonists from EIi Lilly, PPAR-alpha agonists from CrystalGenomics, PPAR-gamma modulators and PPAR-β modulators from CareX, rosiglitazone maleate (CAS No. 122324- 73-4 or 155141-29-0) (GlaxoSmithKline), rosiglitazone maleate/glimepir (CAS No. 155141-29-0 and 93479-97-1), AVANDARYL, rosiglitazone maleate/metformin extend (CAS No. 155141-29-0 and 657-24-9), AVANDAMET, rosiglitazone maleate+metformin, AVANDAMET (GlaxoSmithKline), tesaglitazar (AstraZeneca), and an analogue or derivative thereof.
11382. The method of item 11305 wherein the agent is a phosphatase inhibitor.
11383. The method of item 11305 wherein the agent is a phosphodiesterase (PDE) inhibitor selected from the group consisting of avanafil (Tanabe Seiyaku), dasantafil (CAS No. 569351-91-3) (Schering- Plough), A-906119 (CAS No. 134072-58-5), DL-850 (Sanofi-Aventis), GRC- 3015, GRC-3566, GRC-3886 (Glenmark), HWA-153 (CAS No. 56395-66-5) (Sanofi-Aventis), hydroxypumafentrine (Altana), IBFB-130011 , IBFB-14-016, IBFB-140301 , IBFB-150007, IBFB-211913 (IBFB Pharma), L-826141 (Merck & Co), medorinone (CAS No. 88296-61-1) (Sanofi-Aventis), MEM- 1917 (Memory Pharmaceuticals), ND-1251 (Neuro3d), PDE inhibitors from ICOS, PDE IV inhibitors from Memory Pharmaceuticals and CrystalGenomics, Pharmaprojects No. 2742 and 6141 (Sanofi-Aventis), QAD-171 (Novartis), RHC-2963 (CAS No. 76993-12-9 and 76993-14-1), RPR-117658, RPR-122818 derivatives, SR-24870 , and RPR-132294 (Sanofi-Aventis), SK-350 (ln2Gen), stroke therapy agents from deCODE Genetics, TAS-203 (Taiho), tofimilast (CAS No. 185954-27-2) (Pfizer), UK- 371800 (Pfizer), WIN-65579 (CAS No. 158020-82-7) (Sanofi-Aventis), IBFB- 130020 (IBFB Pharma), OPC-6535 (CAS No. 145739-56-6) (Otsuka), a phosphodiesterase III inhibitor, enoximone, a phosphodiesterase IV inhibitor, fosfosal, Atopik (Barrier Therapeutics), triflusal, a phosphodiesterase V inhibitor, and an analogue or derivative thereof.
11384. The method of item 11305 wherein the agent is a PKC inhibitor.
11385. The method of item 11305 wherein the agent is a platelet activating factor antagonist.
11386. The method of item 11305 wherein the agent is a platelet-derived growth factor receptor kinase inhibitor.
11387. The method of item 11305 wherein the agent is a prolyl hydroxylase inhibitor.
11388. The method of item 11305 wherein the agent is a polymorphonuclear neutrophil inhibitor.
11389. The method of item 11305 wherein the agent is a protein kinase B inhibitor.
11390. The method of item 11305 wherein the agent is a protein kinase C stimulant. 11391. The method of item 11305 wherein the agent is a purine nucleoside analogue.
11392. The method of item 11305 wherein the agent is a purinoreceptor P2X antagonist.
11393. The method of item 11305 wherein the agent is a Raf kinase inhibitor.
11394. The method of item 11305 wherein the agent is a reversible inhibitor of ErbB1 and ErbB2.
11395. The method of item 11305 wherein the agent is a ribonucleoside triphosphate reductase inhibitor.
11396. The method of item 11305 wherein the agent is an SDF-1 antagonist.
11397. The method of item 11305 wherein the agent is a sheddase inhibitor.
11398. The method of item 11305 wherein the agent is an SRC inhibitor.
11399. The method of item 11305 wherein the agent is a stromelysin inhibitor.
11400. The method of item 11305 wherein the agent is an Syk kinase inhibitor. 11401. The method of item 11305 wherein the agent is a telomerase inhibitor.
11402. The method of item 11305 wherein the agent is a TGF beta inhibitor selected from the group consisting of pirfenidone (CAS No. 53179-13-8) (MARNAC), tranilast (CAS No. 53902-12-8) (Kissei), IN- 1130 (ln2Gen), mannose-6-phosphate (BTG), TGF-β antagonists from Inflazyme (Pharmaprojects No. 6075), TGF-β antagonists from Sydney, non-industrial source), TGF-β I receptor kinase inhibitors from EIi Lilly, TGF- β receptor inhibitors from Johnson & Johnson, and an analogue or derivative thereof.
11403. The method of item 11305 wherein the agent is a TNFα antagonist or TACE inhibitor selected from the group consisting of adalimumab (CAS No. 331731-18-1) (Cambridge Antibody Technology), AGIX-4207 (AtheroGenics), AGT-1 (Advanced Biotherapy), an antiinflammatory from Borean Pharma, Cellzome, or Paradigm Therapeutics, anti-inflammatory vaccine (TNF-alpha kinoid) from Neovacs, humanized anti-TNF antibody or an anti-TNF MAb (CB0006) Celltech (UCB), apratastat (CAS No. 287445-51-0) (Wyeth), BMS-561392 (Bristol-Myers Squibb), BN- 006 (Bone), certolizumab pegol (CAS No. 428863-50-7 or CH-138 (UCB), cilomilast (CAS No. 153259-65-5) (GlaxoSmithKline), CR-1 (Nuada Pharmaceuticals), CRx-119 (CombinatoRx), D-5410 (UCB)1 dacopafant (CAS No. 125372-33-0) (Sanofi-Aventis), dersalazine (CAS No. 188913-57- 7/188913-58-8) (Uriach), etanercept (CAS No. 185243-69-0) (Amgen), ethyl pyruvate (Critical (Critical Therapeutics), golimumab (CAS No. 476181-74-5) (Johnson & Johnson), hormono-immunotherapy from Ipsen, CDP571 (e.g., Humicade from UCB), IC-485 (ICOS), infliximab (CAS No. 170277-31-3) (Johnson & Johnson), IP-751 (Manhattan Pharmaceuticals), ISIS-104838 (CAS No. 250755-32-9) (ISIS Pharmaceuticals), lenalidomide (CAS No. 191732-72-6) (Celgene), lentinan (CAS No. 37339-90-5) (Ajinomoto), MDL- 201112 (CAS No. 142130-73-2) (Sanofi-Aventis), medroxyprogesterone (CAS No. 520-85-4) (InKine Pharmaceutical), N-acetylcysteine (CAS No. 616-91-1) (Zambon), NBE-P2 (DIREVO Biotech), nerelimomab (CAS No. 162774-06-3) (Chiron), OM-294DP (OM PHARMA), onercept (CAS No. 199685-57-9) (Yeda), PASSTNF-alpha (Verigen), pentoxifylline or oxypentifylline (Sanofi-Aventis), Pharmaprojects No. 4091 , 4241 , 4295, or 4488 (Sanofi-Aventis), Pharmaprojects No. 5480 (Amgen), Pharmaprojects No. 6749 (Cengent), pirfenidone (CAS No. 53179-13-8) (MARNAC), RPR- 132294 (Sanofi-Aventis), S5 (F002) (Fulcrum Pharmaceuticals), simvastatin (CAS No. 79902-63-9) (Merck & Co), STA-6292 (Synta Pharmaceuticals), tacrolimus (CAS No. 104987-11-3) (Fujisawa LifeCycle Pharma), talactoferrin alfa (CAS No. 308240-58-6) (Agennix), thalidomide (CAS No. 50-35-1) (Celgene), TNF antagonists form ProStrakan, and Synergen, TNF inhibitors (Amgen), TNF-alpha antagonists from Dynavax Technologies and Jerina AG (Germany), TNF-alpha inhibitors from IBFB Pharma and Xencor (Xencor), torbafylline (CAS No, 105102-21-4) (Sanofi-Aventis), UR-1505 (Uriach), VT-346 (Viron Therapeutics), YSIL6 (Y1S Therapeutics), YSTH2 (Y's Therapeutics), NPI-1302a-3 (Nereus Pharmaceuticals, a TNF antagonist from Jerina AG (Germany), dersalazine, and an analogue or derivative thereof.
11404. The method of item 11305 wherein the agent is a Toll receptor inhibitor.
11405. The method of item 11305 wherein the agent is a tubulin antagonist.
11406. The method of item 11305 wherein the agent is a tyrosine kinase inhibitor selected from the group consisting of SU-011248, SUTENT from Pfizer Inc. (New York, NY)1 BMS-354825, PN-355 (Paracelsian Pharmaceuticals), AGN-199659 (Allergan), AAL-993 or ABP- 309 (Novartis), adaphostin (NIH)1 AEE-788 (Novartis), AG-013736 (OSI Pharmaceuticals), AG-13736 (Pfizer), ALT-110 (Alteris Therapeutics), AMG- 706 (Amgen), anticancer MAbs from Xencor, anti-EGFrvlll MAbs from Abgenix, anti-HER2 MAb from Abiogen, AZD-2171 or AZD-9935 (AstraZeneca), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), CEP-5214 (Cephalon), CEP-7055 (Cephalon), cetuximab (ImCIone Systems), CHIR-200131 and CHIR-258 (Chiron), CP-547632 (OSI Pharmaceuticals), CP-724714 (Pfizer), CT-301 (Creabilis Therapeutics), D- 69491 (Baxter International), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EGFR/ErbB2 inhibitors from Array BioPharma, erlotinib (CAS No. 183319- 69-9) (OSI Pharmaceuticals), EXEL-2880 (Exelixis), FK-778 (Sanofi- Aventis) , gefitinib (CAS No. 184475-35-2) (AstraZeneca), GW-2286 or GW- 654652 (GlaxoSmithKline), her2/neu antigen from AlphaVax, HER-2/neu inhibitor from Generex, Herzyme (Medipad) (Sirna Therapeutics), HKI-272 (Wyeth), HuMax-EGFr (Genmab), idronoxil (CAS No. 81267-65-4) (Novogen), IGF-1 inhibitors from Ontogen, IMC-11F8 (ImCIone Systems), kahalalide F (CAS No. 149204-42-2) (PharmaMar), KDR inhibitor from LG Life Sciences, KDR inhibitors from Abbott Laboratories, KDR kinase inhibitors (UCB), Kdr kinase inhibitors from Merck & Co, KRN-633 and KRN- 951 (Kirin Brewery), KSB-102 (Xenova), lapatinib ditosylate (CAS No. 388082-78-8) (GlaxoSmithKline), matuzumab (Merck KGaA), MDX-214 (Medarex), ME-103 (Pharmexa), MED-A300 (Gerolymatos), MNAC-13 (Lay Line Genomics), nimotuzumab (Center of Molecular Immunology), NSC- 330507 or NSC-707545 (NIH), NV-50 (Novogen), OSI-930 (OSI Pharmaceuticals), panitumumab (Abgenix), pelitinib (CAS No. 287933-82-7) (Wyeth), pertuzumab (CAS No. 380610-27-5) (Genentech), Pharmaprojects No. 3985 (Sanofi-Aventis), prostate cancer therapeutics from Sequenom (SQPC35, SQPC36, SQPC90), removab and remoxab (Trion Pharma), RG- 13022 (CAS No. 136831-48-6), RG-13291 (CAS No. 138989-50-1), or RG- 14620 (CAS No. 136831-49-7) (Sanofi-Aventis), RM-6427 (Romark), RNAi breast cancer therapy from Benitec, RP 53801 (CAS No. 125882-88-4) (Sanofi-Aventis), sorafenib tosylate (Bayer), SU-11657 (Pfizer), Tie-2 antagonists from Semaia (Hybrigenics), Tie-2 inhibitors from Ontogen, trastuzumab (CAS No. 180288-69-1) (Genentech), tyrosine kinase inhibitors from Sanofi-Aventis, U3-1287, U3-1565, U3-1784, U3-1800 (U3 Pharma), vatalanib (Novartis), VEGFR-2 kinase inhibitor from Bristol-Myers Squibb, XL-647 (Exelixis), ZD-6474 (AstraZeneca), ZK-CDK (Schering AG), an EGFR tyrosine kinase inhibitor, EKB-569 (Wyeth), and an analogue or derivative thereof.
11407. The method of item 11305 wherein the agent is a VEGF inhibitor.
11408. The method of item 11305 wherein the agent is a vitamin D receptor agonist.
11409. The method of item 11305 wherein the agent is ZD-6474 (an angiogenesis inhibitor).
11410. The method of item 11305 wherein the agent is AP-23573 (an mTOR inhibitor).
11411. The method of item 11305 wherein the agent is synthadotin (a tubulin antagonist).
11412. The method of item 11305 wherein the agent is S-0885 (a collagenase inhibitor).
11413. The method of item 11305 wherein the agent is aplidine (an elongation factor-1 alpha inhibitor). 11414. The method of item 11305 wherein the agent is ixabepilone (an epithilone).
11415. The method of item 11305 wherein the agent is IDN-5390 (an angiogenesis inhibitor).
11416. The method of item 11305 wherein the agent is SB-2723005 (an angiogenesis inhibitor).
11417. The method of item 11305 wherein the agent is ABT-518 (an angiogenesis inhibitor).
11418. The method of item 11305 wherein the agent is combretastatin (an angiogenesis inhibitor).
11419. The method of item 11305.wherein the agent is anecortave acetate (an angiogenesis inhibitor).
11420. The method of item 11305 wherein the agent is SB-715992 (a kinesin antagonist).
11421. The method of item 11305 wherein the agent is temsirolimus (an mTOR inhibitor).
11422. The method of item 11305 wherein the agent is adalimumab (a TNFα antagonist).
11423. The method of item 11305, wherein the composition comprises a polymer. 11424. The method of item 11305, wherein the composition comprises a polymeric carrier.
11425. The method of item 11305 wherein the anti- scarring agent inhibits adhesion between the device and a host into which the device is implanted.
11426. The method of item 11305 wherein the device delivers the anti-scarring agent locally to tissue proximate to the device.
11427. The method of item 11305 wherein the device has a coating that comprises the anti-scarring agent.
11428. The method of item 11305, wherein the device has a coating that comprises the agent and is disposed on a surface of the implant.
11429. The method of item 11305, wherein the device has a coating that comprises the agent and directly contacts the implant.
11430. The method of item 11305, wherein the device has a coating that comprises the agent and indirectly contacts the implant.
11431. The method of item 11305, wherein the device has a coating that comprises the agent and partially covers the implant.
11432. The method of item 11305, wherein the device has a coating that comprises the agent and completely covers the implant.
11433. The method of item 11305, wherein the device has a uniform coating. 11434. The method of item 11305, wherein the device has a non-uniform coating.
11435. The method of item 11305, wherein the device has a discontinuous coating.
11436. The method of item 11305, wherein the device has a patterned coating.
11437. The method of item 11305, wherein the device has a coating with a thickness of 100 Dm or less.
11438. The method of item 11305, wherein the device has a coating with a thickness of 10 Gm or less.
11439. The method of item 11305, wherein the device has a coating, and the coating adheres to the surface of the implant upon deployment of the implant.
11440. The method of item 11305, wherein the device has a coating, and wherein the coating is stable at room temperature for a period of 1 year.
11441. The method of item 11305, wherein the device has a coating, and wherein the anti-scarring agent is present in the coating in an amount ranging between about 0.0001% to about 1% by weight.
11442. The method of item 11305, wherein the device has a coating, and wherein the anti-scarring agent is present in the coating in an amount ranging between about 1 % to about 10% by weight. 11443. The method of item 11305, wherein the device has a coating, and wherein the anti-scarring agent is present in the coating in an amount ranging between about 10% to about 25% by weight.
11444. The method of item 11305, wherein the device has a coating, and wherein the anti-scarring agent is present in the coating in an amount ranging between about 25% to about 70% by weight.
11445. The method of item 11305, wherein the device has a coating, and wherein the coating further comprises a polymer.
11446. The method of item 11305, wherein the device has a first coating having a first composition and a second coating having a second composition.
11447. The method of item 11305, wherein the device has a first coating having a first composition and a second coating having a second composition, wherein the first composition and the second composition are different.
11448. The method of item 11305, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a copolymer.
11449. The method of item 11305, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a block copolymer.
11450. The method of item 11305, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a random copolymer. 11451. The method of item 11305, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a biodegradable polymer.
11452. The method of item 11305, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a non-biodegradable polymer.
11453. The method of item 11305, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a hydrophilic polymer.
11454. The method of item 11305, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a hydrophobic polymer.
11455. The method of Item 11305, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a polymer having hydrophilic domains.
11456. The method of item 11305, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a polymer having hydrophobic domains.
11457. The method of item 11305, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a non-conductive polymer.
11458. The method of item 11305, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises an elastomer. 11459. The method of item 11305, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a hydrogel.
11460. The method of item 11305, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a silicone polymer.
11461. The method of item 11305, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a hydrocarbon polymer.
11462. The method of item 11305, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a styrene-derived polymer.
11463. The method of item 11305, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a butadiene polymer.
11464. The method of item 11305, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a macromer.
11465. The method of item 11305, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a poly(ethylene glycol) polymer.
11466. The method of item 11305 wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises an amorphous polymer. 11467. The method of item 11305, wherein the device comprises a lubricious coating.
11468. The method of item 11305 wherein the anti- scarring agent is located within pores or holes of the device.
11469. The method of item 11305 wherein the anti- scarring agent is located within a channel, lumen, or divet of the device.
11470. The method of item 11305, wherein the device comprises a second pharmaceutically active agent.
11471. The method of item 11305 wherein the device comprises an anti-inflammatory agent.
11472. The method of item 11305 wherein the device comprises an agent that inhibits infection.
11473. The method of item 11305 wherein the device comprises an agent that inhibits infection, and wherein the agent is an anthracycline.
11474. The method of item 11305 wherein the device comprises an agent that inhibits infection, and wherein the agent is doxorubicin.
11475. The method of item 11305 wherein the device comprises an agent that inhibits infection, and wherein the agent is mitoxantrone. 11476. The method of item 11305 wherein the device comprises an agent that inhibits infection, and wherein the agent is a fluoropyrimidine.
11477. The method of item 11305 wherein the device comprises an agent that inhibits infection, and wherein the agent is 5- fluorouracil (5-FU).
11478. The method of item 11305 wherein the device comprises an agent that inhibits infection, and wherein the agent is a folic acid antagonist.
11479. The method of item 11305 wherein the device comprises an agent that inhibits infection, and wherein the agent is methotrexate.
11480. The method of item 11305 wherein the device comprises an agent that inhibits infection, and wherein the agent is a podophylotoxin.
11481. The method of item 11305 wherein the device comprises an agent that inhibits infection, and wherein the agent is etoposide.
11482. The method of item 11305 wherein the device comprises an agent that inhibits infection, and wherein the agent is a camptothecin.
11483. The method of item 11305 wherein the device comprises an agent that inhibits infection, and wherein the agent is a hydroxyurea. 11484. The method of item 11305 wherein the device comprises an agent that inhibits infection, and wherein the agent is a platinum complex.
11485. The method of item 11305 wherein the device comprises an agent that inhibits infection, and wherein the agent is cisplatin.
11486. The method of item 11305, further comprising an anti-thrombotic agent.
11487. The method of item 11305 wherein the device comprises a visualization agent.
11488. The method of item 11305 wherein the device comprises a visualization agent, wherein the visualization agent is a radiopaque material, and wherein the radiopaque material comprises a metal, a halogenated compound, or a barium containing compound.
11489. The method of item 11305 wherein the device comprises a visualization agent, wherein the visualization agent is a radiopaque material, and wherein the radiopaque material comprises barium, tantalum, or technetium.
11490. The method of item 11305 wherein the device comprises a visualization agent, and wherein the visualization agent is a MRI responsive material.
11491. The method of item 11305 wherein the device comprises a visualization agent, and wherein the visualization agent comprises a gadolinium chelate. 11492. The method of item 11305 wherein the device comprises a visualization agent, and wherein the visualization agent comprises iron, magnesium, manganese, copper, or chromium.
11493. The method of item 11305 wherein the device comprises a visualization agent, and wherein the visualization agent comprises an iron oxide compound.
11494. The method of item 11305 wherein the device comprises a visualization agent, and wherein the visualization agent comprises a dye, pigment, or colorant.
11495. The method of item 11305 wherein the device comprises an echogenic material.
11496. The method of item 11305 wherein the device comprises an echogenic material, and wherein the echogenic material is in the form of a coating.
11497. The method of item 11305 wherein the device is sterile.
11498. The method of item 11305 wherein the anti- scarring agent is released into tissue in the vicinity of the device after deployment of the device.
11499. The method of item 11305 wherein the anti- scarring agent is released into tissue in the vicinity of the device after deployment of the device, and wherein the tissue is connective tissue. 11500. The method of item 11305 wherein the anti- scarring agent is released into tissue in the vicinity of the device after deployment of the device, and wherein the tissue is muscle tissue.
11501. The method of item 11305 wherein the anti- scarring agent is released into tissue in the vicinity of the device after deployment of the device, and wherein the tissue is nerve tissue.
11502. The method of item 11305 wherein the anti- scarring agent is released into tissue in the vicinity of the device after deployment of the device, and wherein the tissue is epithelium tissue.
11503. The method of item 11305 wherein the anti- scarring agent is released in effective concentrations from the device over a period ranging from the time of deployment of the device to about 1 year.
11504. The method of item 11305 wherein the anti- scarring agent is released in effective concentrations from the device over a period ranging from about 1 month to 6 months.
11505. The method of item 11305 wherein the anti- scarring agent is released in effective concentrations from the device over a period ranging from about 1 - 90 days.
11506. The method of item 11305 wherein the anti- scarring agent is released in effective concentrations from the device at a constant rate.
11507. The method of item 11305 wherein the anti- scarring agent is released in effective concentrations from the device at an increasing rate. 11508. The method of item 11305 wherein the anti- scarring agent is released in effective concentrations from the device at a decreasing rate.
11509. The method of item 11305 wherein the anti- scarring agent is released in effective concentrations from the composition comprising the anti-scarring agent by diffusion over a period ranging from the time of deployment of the device to about 90 days.
11510. The method of item 11305 wherein the anti- scarring agent is released in effective concentrations from the composition comprising the anti-scarring agent by erosion of the composition over a period ranging from the time of deployment of the device to about 90 days.
11511. The method of item 11305 wherein the device comprises about 0.01 Dg to about 10 Gg of the anti-scarring agents - -
11512. The method of item 11305 wherein the device comprises about 10 Dg to about 10 mg of the anti-scarring agent.
11513. The method of item 11305 wherein the device comprises about 10 mg to about 250 mg of the anti-scarring agent.
11514. The method of item 11305 wherein the device comprises about 250 mg to about 1000 mg of the anti-scarring agent.
11515. The method of item 11305 wherein the device comprises about 1000 mg to about 2500 mg of the anti-scarring agent. 11516. The method of item 11305 wherein a surface of the device comprises less than 0.01 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
11517. The method of item 11305 wherein a surface of the device comprises about 0.01 Gg to about 1 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
11518. The method of item 11305 wherein a surface of the device comprises about 1 Dg to about 10 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
11519. The method of item 11305 wherein a surface of the device comprises about 10 Dg to about 250 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
11520. The method of item 11305 wherein a surface of the device comprises about 250 Dg to about 1000 Dg of the anti-scarring agent of anti-scarring agent per mm2 of device surface to which the anti- scarring agent is applied.
11521. The method of item 11305 wherein a surface of the device comprises about 1000 Dg to about 2500 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
11522. The method of item 11305 wherein the combining is performed by direct affixing the agent or the composition to the implant. 11523. The method of item 11305 wherein the combining is performed by spraying the agent or the component onto the implant.
11524. The method of item 11305 wherein the combining is performed by electrospraying the agent or the composition onto the implant.
11525. The method of item 11305 wherein the combining is performed by dipping the implant into a solution comprising the agent or the composition.
11526. The method of item 11305 wherein the combining is performed by covalently attaching the agent or the composition to the implant.
11527. The method of item 11305 wherein the combining is performed by non-covalently attaching the agent or the composition to the implant.
11528. The method of item 11305 wherein the combining is performed by coating the implant with a substance that contains the agent or the composition.
11529. The method of item 11305 wherein the combining is performed by coating the implant with a substance that absorbs the agent.
11530. The method of item 11305 wherein the combining is performed by interweaving a thread composed of, or coated with, the agent or the composition. 11531. The method of item 11305 wherein the combining is performed by covering all the implant with a sleeve that contains the agent or the composition.
11532. The method of item 11305 wherein the combining is performed by covering a portion of the implant with a sleeve that contains the agent or the composition.
11533. The method of item 11305 wherein the combining is performed by covering all the implant with a cover that contains the agent or the composition.
11534. The method of item 11305 wherein the combining is performed by covering a portion of the implant with a cover that contains the agent or the composition.
11535. The method of item 11305 wherein the combining is performed by covering all the implant with an electrospun fabric that contains the agent or the composition.
11536. The method of item 11305 wherein the combining is performed by covering a portion of the implant with an electrospun fabric that contains the agent or the composition.
11537. The method of item 11305 wherein the combining is performed by covering all the implant with a mesh that contains the agent or the composition.
11538. The method of item 11305 wherein the combining is performed by covering a portion of the implant with a mesh that contains the agent or the composition. 11539. The method of item 11305 wherein the combining is performed by constructing all the implant with the agent or the composition.
11540. The method of item 11305 wherein the combining is performed by constructing a portion of the implant with the agent or the composition.
11541. The method of item 11305 wherein the combining is performed by impregnating the implant with the agent or the composition.
11542. The method of item 11305 wherein the combining is performed by constructing all of the implant from a degradable polymer that releases the agent.
11543. The method of item 11305 wherein the combining is performed by constructing a portion of the implant from a degradable polymer that releases the agent.
11544. The method of item 11305 wherein the combining is performed by dipping the implant into a solution that comprise the agent and an inert solvent for the implant.
11545. The method of item 11305 wherein the combining is performed by dipping the implant into a solution that comprises the agent and a solvent that will swell the implant.
11546. The method of item 11305 wherein the combining is performed by dipping the implant into a solution that comprises the agent and a solvent that will dissolve the implant. 11547. The method of item 11305 wherein the combining is performed by dipping the implant into a solution that comprises the agent, a polymer and an inert solvent for the implant.
11548. The method of item 11305 wherein the combining is performed by dipping the implant into a solution that comprises the agent, a polymer and a solvent that will swell the implant.
11549. The method of item 11305 wherein the combining is performed by dipping the implant into a solution that comprises the agent, a polymer and a solvent that will dissolve the implant.
11550. The method of item 11305 wherein the combining is performed by spraying the implant into a solution that comprises the agent and an inert solvent for the implant.
11551. The method of item 11305 wherein the combining is performed by spraying the implant into a solution that comprises the agent and a solvent that will swell the implant.
11552. The method of item 11305 wherein the combining is performed by spraying the implant into a solution that comprises the agent and a solvent that will dissolve the implant.
11553. The method of item 11305 wherein the combining is performed by spraying the implant into a solution that comprises the agent, a polymer and an inert solvent for the implant.
11554. The method of item 11305 wherein the combining is performed by spraying the implant into a solution that comprises the agent, a polymer and a solvent that will swell the implant. 11555. The method of item 11305 wherein the combining is performed by spraying the implant into a solution that comprises the agent, a polymer and a solvent that will dissolve the implant.
11556. A method as in any one of items 11301 -11555, wherein the device is a catheter.
11557. A method as in any one of items 11301 -11555, wherein the device is a balloon catheter.
11558. A method as in any one of items 11301-11555, wherein the device is a balloon.
11559. A method as in any one of items 11301 -11555, wherein the device is a stent graft.
11560. A method as in any one of items 11301-11555, wherein the device is a guidewire.
11561. A method as in any one of items 11301-11555, wherein the device is a stent.
11562. A method as in any one of items 11301-11555, wherein the device is an intravascular stent.
11563. A method as in any one of items 11301-11555, wherein the device is a metallic stent.
11564. A method as in any one of items 11301 -11555, wherein the device is a polymeric stent. 11565. A method as in any one of items 11301-11555, wherein the device is a biodegradable stent.
11566. A method as in any one of items 11301-11555, wherein the device is a non-biodegradable stent.
11567. A method as in any one of items 11301 -11555, wherein the device is a self expandable stent.
11568. A method as in any one of items 11301-11555, wherein the device is a balloon expandable stent.
11569. A method as in any one of items 11301-11555, wherein the device is a covered stent.
11570. A method as in any one of items 11301 r11555, wherein the device is a drug eluting stent.
11571. A method as in any one of items 11301 -11555, wherein the device is a stent that comprises a radio-opaque material.
11572. A method as in any one of items 11301-11555, wherein the device is a stent that comprises an echogenic material.
11573. A method as in any one of items 11301-11555, wherein the device is a stent that comprises an MRI responsive material.
11574. A method as in any one of items 11301-11555, wherein the device is an anastomotic connector device. 11575. A method as in any one of items 11301-11555, wherein the device is an artery to artery anastomotic connector device.
11576. A method as in any one of items 11301-11555, wherein the device is a vein to artery anastomotic connector device.
11577. A method as in any one of items 11301 -11555, wherein the device is an artery to vein anastomotic connector device.
11578. A method as in any one of items 11301 -11555, wherein the device is a synthetic graft to artery anastomotic connector device.
11579. A method as in any one of items 11301-11555, wherein the device is a vein to synthetic graft anastomotic connector device.
11580. A method as in any one of items 11301-11555, wherein the device is a synthetic graft to vein anastomotic connector device.
11581. A method as in any one of items 11301-11555, wherein the device is a vascular clip.
11582. A method as in any one of items 11301 -11555, wherein the device is a vascular suture.
11583. A method as in any one of items 11301-11555, wherein the device is a vascular clamp.
11584. A method as in any one of items 11301-11555, wherein the device is a suturing device. 11585. A method as in any one of items 11301-11555, wherein the device is an anastomotic coupler.
11586. A method as in any one of items 11301-11555, wherein the device is an automated or modified suture device.
11587. A method as in any one of items 11301-11555, wherein the device is a micromechanical anastomotic connector device.
11588. A method as in any one of items 11301-11555, wherein the device is an anastomotic coupling device that facilitates automated attachment of a graft or vessel to an aperature or orifice in a target vessel without the use of sutures or staples.
11589. A method as in any one of items 11301-11555, wherein the device is an anastomotic coupling device that comprises a tubular graft conduit and may be placed in a side wall of a target vessel so that the tubular graft conduit may be extended from the target vessel.
11590. A method as in any one of items 11301 -11555, wherein the device is an anastomotic coupler in the form of a frame.
11591. A method as in any one of items 11301 -11555, wherein the device is an anastomotic coupler in a ringl-like form.
11592. A method as in any one of items 11301-11555, wherein the device is a resorbable anastomotic coupler.
11593. A method as in any one of items 11301-11555, wherein the device is an anastomotic coupler that comprises a bioabsorbable and elastomeric material. 11594. A method as in any one of items 11301 -11555, wherein the device is an anastomotic coupler adapted to connect a first blood vessel with a second blood vessel with a graft vessel.
11595. A method as in any one of items 11301 -11555, wherein the device is an anastomotic coupler adapted to connect a first blood vessel with a second blood vessel without a graft vessel.
11596. A method as in any one of items 11301-11555, wherein the device is an anastomotic coupler that is incorporated in the design of a vascular graft.
11597. A method as in any one of items 11301-11555, wherein the device is an anastomotic coupler that comprises a graft that incorporates a fixation mechanism.
11598. A method as in any one of items 11301-11555, wherein the device is an anastomotic coupler that comprises a compressible, expandable fitting for securing the ends of a bypass graft to two vessels.
11599. A method as in any one of items 11301 -11555, wherein the device is an anastomotic coupler that comprises a pair of coupling disc members for joining two vessels in an end to end or end to side fashion.
11600. A method as in any one of items 11301-11555, wherein the device is a proximal aortic connector.
11601. A method as in any one of items 11301-11555, wherein the device is a distal coronary connector. 11602. A method as in any one of items 11301-11555, wherein the device is a bypass device made of a biocompatible material.
11603. A method as in any one of items 11301-11555, wherein the device is a bypass device made of at least partially a metal or metal alloy.
11604. A method as in any one of items 11301-11555, wherein the device is a bypass device made of at least partially a synthetic polymer.
11605. A method as in any one of items 11301-11555, wherein the device is a bypass device made of at least partially naturally derived polymer.
11606. A method as in any one of items 1.1301-11555, ._ wherein the device is a tubular anastomotic coupler that comprises a tubular structure that may be attached directly to a proximal blood vessel.
11607. A method as in any one of items 11301 -11555, wherein the device is a tubular anastomotic coupler that comprises a tubular structure that may be attached directly to a distal blood vessel.
11608. A method as in any one of items 11301-11555, wherein the device is a tubular anastomotic coupler that has a proximal end attachable to a proximal vessel and a distal end attachable to a bypass graft.
11609. A method as in any one of items 11301 -11555, wherein the device is a tubular anastomotic coupler that has a proximal end attachable to a graft vessel that is secured to a proximal blood vessel and a distal end attachable to a distal blood vessel.
11610. A method as in any one of items 11301 -11555, wherein the device is an anastomotic connector device adapted for end to end anastomosis procedures.
11611. A method as in any one of items 11301-11555, wherein the device is an anastomotic stent.
11612. A method as in any one of items 11301-11555, wherein the device is an anastomotic sleeve.
11613. A method as in any one of items 11301-11555, wherein the device is an anastomotic connector device adapted for end to side anastomosis procedures.
11614. A method as in any one of items 11301-11555, wherein the device is a single lumen bypass device.
11615. A method as in any one of items 11301-11555, wherein the device is a multi-lumen bypass device.
11616. A method as in any one of items 11301-11555, wherein the device is an anastomotic coupling device that comprises a single tubular portion that may be used as a shunt to divert blood from a source vessel to a graft vessel.
11617. A method as in any one of items 11301-11555, wherein the device is an anastomotic coupling device that comprises more than one tubular portion, and wherein at least one tubular portion may be used as a shunt for diverting blood between a source vessel and a target vessel.
11618. A method as in any one of items 11301 -11555, wherein the device is an anastomotic connector device that comprises a tubular portion, and wherein one or more ends of the tubular portion may be inserted into the end or into the side of one or more blood vessels.
11619. A method as in any one of items 11301 -11555, wherein the device is a multi-lumen anastomotic connector device that at least one arm of the device may be attached to a graft vessel.
11620. A method as in any one of items 11301 -11555, wherein the device is an anastomotic connector device that includes three or more tubular arms that extend from a junction site.
11621. A method as in any one of items 11301-11555, wherein the device is a multi-lumen anastomotic connector device that is generally T-shaped.
11622. A method as in any one of items 11301 -11555, wherein the device is a multi-lumen anastomotic connector device that is generally Y-shaped.
11623. A method as in any one of items 11301-11555, wherein the device is an anastomotic connector device that comprises a tube for bypassing blood flow directly from a portion of the heart to a coronary artery. 11624. A method as in any one of items 11301-11555, wherein the device is an anastomotic connector device that comprises a network of interconnected tubular conduits.
11625. A method as in any one of items 11301-11555, wherein the device is an anastomotic connector device that is configured with two or more termini that provide a vessel interface without the need for sutures and a fluid communication through an intersecting lumen.
11626. A method of making a medical device comprising: combining a gastrointestinal stent (e.g., an implant) and an anti-scarring agent or a composition comprising an anti-scarring agent, wherein the agent inhibits scarring between the device and a host into which the device is implanted.
11627. The method of item 11626 wherein the agent is an adensosine A2A receptor antagonist.
11628. The method of item 11626 wherein the agent is an AKT inhibitor.
11629. The method of item 11626 wherein the agent is an alpha 2 integrin antagonist, wherein the alpha 2 integrin antagonist is Pharmaprojects No. 5754 (Merck KgaA).
11630. The method of item 11626 wherein the agent is an alpha 4 integrin antagonist.
11631. The method of item 11626 wherein the agent is an alpha 7 nicotinic receptor agonist. 11632. The method of item 11626 wherein the agent is an angiogenesis inhibitor selected from the group consisting of AG-12,958 (Pfizer), ATN-161 (Attenuon LLC), neovastat, an angiogenesis inhibitor from Jerina AG (Germany), NM-3 (Mercian), VGA-1155 (Taisho), FCE-26644 (Pfizer), FCE-26950 (Pfizer), FPMA (Meiji Daries), FR-111142 (Fujisawa), GGTI-298, GM-1306 (Ligand), GPA-1734 (Novartis), NNC-47-0011 (Novo Nordisk), herbamycin (Nippon Kayaku), lenalidomide (Celegene), IP-10 (NIH), ABT-828 (Abbott), KIN-841 (Tokushima University, Japan), SF-1126 (Semafore Pharmaceuticals), laminin technology (NIH), CHIR-258 (Chiron), NVP-AEW541 (Novartis), NVP-AEW541 (Novartis), Vt16907 (Alchemia), OXI-8007 (Oxigene), EG-3306 (Ark Therapeutics), Maspin (Arriva), ABT- 567 (Abbott), PPI-2458 (Praecis Pharmaceuticals), CC-5079, CC-4089 (Celgene), HIF-1alpha inhibitors (Xenova), S-247 (Pfizer), AP-23573 (Ariad), AZD-9935 (Astra Zeneca), mebendazole (Introgen Therapeutics), MetAP-2 inhibitors (GlaxoSmithKline), AG-615 (Angiogene Pharmaceuticals), Tie-2 antagonists (Hybrigenics), NC-381, CYC-381, NC-169, NC-219, NC-383, NC-384, NC-407 (Lorus Therapeutics), ATN-224 (Attenuon), ON-01370 (Onconova), Vitronectin antagonists (Amgen), SDX-103 (Salmedix), Vitronectin antagonists (Shire), CHP (Riemser), TEK (Amgen), Anecortave acetate (Alcon), T46.2 (Matrix Therapeutics), HG-2 (Heptagen), TEM antagonists (Genzyme), Oxi-4500 (Oxigene), ATN-161 (Attenuon), WX-293 (Wilex), M-2025 (Metris Therapeutics), Alphastatin (BioActa), YH-16 (Yantai Rongchang), BIBF-1120 (Boehringer Ingelheim), BAY-57-9352 (Bayer), AS- 1404 (Cancer Research Technology), SC-77964 (Pfizer), glycomimetics (BioTie Therapies), TIE-2 Inhibitors (Ontogen), DIMI, Octamer (Octamer), ABR-215050 (Active Biotech), ABT-518 (Abbott), KDR inhibitors (Abbott), BSF-466895 (Abbott), SCH-221153 (Schering-Plough), DAC:antiangiogenic (ConjuChem), TFPI (EntreMed), AZD-2171 (Astra-Zenaca), CDC-394 (Celgene), LY290293 (EIi Lilly), IDN-5390 (Indena), Kdr Kinase Inhibitors (Merck), CT-113020, CT-116433, CT-116563, CT-31890, CT-32228) (Cell Therapeutics), A-299620 (Abbott), TWEAK Inhibitor (Amgen), VEGF modulators (Johnson and Johnson), Tum-N53, tumstatin (Genzyme), Thios- 1 , Thios-2 (Thios Pharmaceuticals), MV-6401 (Miravant Medical Technologies), Spisulosine (PharmaMar), CEP-7055 (Cephalon), AUV-201 (Auvation), LM-609 (EIi Lilly), SKF-106615 (AnorMED), Oglufanide disodium (Cytran), BW-114 (Pharminox), Calreticulin (NIH)1 WX-678 (Wilex), SD-7784 (Pfizer), WX-UK1 (Wilex), SH-268 (Schering AG), 2-Me-PGA (Celgene), S- 137 (Pfizer), ZD-6126 (Angiogene Pharmaceuticals), SG-292 (SignalGen), Benefin (Lane Labs), A6, A36 (Angstrom), SB-2723005 (GlaxoSmithKline), SC-7 (Cell Therapeutics), ZEN-014 (AEterna Zentaris), 2-methoxyestradiol (EntreMed), NK-130119 (Nippon Kayaku), CC-10004 (Celgene), AVE- 8062A (Ajinomoto), Tacedinaline (Pfizer), Actinonin (Tokyo Metropolitan Institute of Medical Science), Lenalidomide (Celgene), VGA-1155, BTO-956 (SRI International), ER-68203-00 (Eisai), CT-6685 (UCB), JKC-362 (Phoenix Pharmaceuticals), DMI-3798 (DMI Biosciences, Angiomate (Ipsen), ZD-6474 (AstraZeneca), CEP-5214 (Cephalon), Canstatin (Genzyme), NM-3 (Mercian), Oridigm (MediQuest Therapeutics), Exherin (Adherex), BLS-0597 (Boston Life Sciences), PTC-299 (PTC Therapeutics), NPI-2358 (Nereus Pharmaceuticals), CGP-79787 (Novartis), AEE-788 (Novartis), CKD-732 (Chong Kun Dang), CP-564959 (OSI Pharmaceuticals), CM-101 (CarboMed), CT-2584, CT3501 (Cell Therapeutics), combretastatin and analogues and derivatives thereof (Oxigene), Rebimastat (Bristol-Meyers Squibb), Dextrin 2-sulfate (ML Laboratories), Cilengitide (Merk KGaA), NSC-706704 (Pharminox), KRN- 951 (Kirin Brewery), Ukrain, NSC-631570 (Nowicky Pharma), Tecogalan sodium (Daiichi Pharmaceutical), Tz-93 (Tsumura), TBC-1635 (Encysive Pharmaceuticals), TAN-1120 (Takeda), Semaxanib (Pfizer), BDI-7800 (Biopharmacopae), SD-186, SD-983 (Bristol-Meyers Squibb), SB-223245 (GlaxoSmithKline), SC-236 (Pfizer), RWJ-590973 (Johnson and Johnson), ILX-1850 (Genzyme), SC-68488, S-836 (Pfizer), CG-55069-11 (CuraGen), Ki-23057 (Kirin Brewery), CCX-700 (Chemoentryx), Pegaptanib octasodium (Giled Sciences), ANGIOCOL (available from Biostratum Inc.), or an analogue or derivative thereof.
11633. The method of item 11626 wherein the agent is an apoptosis antagonist.
11634. The method of item 11626 wherein the agent is an apoptosis activator.
11635. The method of item 11626 wherein the agent is a beta 1 integrin antagonist.
11636. The method of item 11626 wherein the agent is a beta tubulin inhibitor.
11637. The method of item 11626 wherein the.agentis a- blocker of enzyme production in Hepatitis C.
11638. The method of item 11626 wherein the agent is a Bruton's tyrosine kinase inhibitor.
11639. The method of item 11626 wherein the agent is a calcineurin inhibitor.
11640. The method of item 11626 wherein the agent is a caspase 3 inhibitor.
11641. The method of item 11626 wherein the agent is a CC chemokine receptor antagonist. 11642. The method of item 11626 wherein the agent is a cell cycle inhibitor selected from the group consisting of SNS-595 (Sunesis), synthadotin, KRX-0403, and an analogue or derivative thereof.
11643. The method of item 11626 wherein the agent is a cathepsin B inhibitor.
11644. The method of item 11626 wherein the agent is a cathepsin K inhibitor, wherein the cathepsin K inhibitor is 462795 (GlaxoSmithKline), INPL-022-D6 (Amura Therapeutics), or an analogue or derivative thereof.
11645. The method of item 11626 wherein the agent is a cathepsin L inhibitor.
11646. The method of item 11626 wherein the agent is a CD40 antagonist.
11647. The method of item 11626 wherein the agent is a chemokine receptor agonist.
11648. The method of item 11626 wherein the agent is a chymase inhibitor.
11649. The method of item 11626 wherein the agent is a collagenase antagonist.
11650. The method of item 11626 wherein the agent is a CXCR antagonist. 11651. The method of item 11626 wherein the agent is a cyclin dependent kinase inhibitor selected from the group consisting of a CDK-1 inhibitor, a CDK-2 inhibitor, a CDK- 4 inhibitor, a CDK-6 inhibitor, a CAK1 inhibitor from GPC Biotech and Bristol-Myers Squibb, RGB-286199 (GPC Biotech), an anticancer agent from Astex Technology, a CAK1 inhibitor from GPC Biotech, a CDK inhibitor from Sanofi-Aventis, a CDK1/CDK2 inhibitor from Amgen, a CDK2 inhibitor from SUGEN-2 (Pfizer), a hearing loss therapy agent (Sound Pharmaceuticals), PD-0332991 (Pfizer), RGB-286199 (GPC Biotech), Ro-0505124 (Hoffmann-La Roche), a Ser/Thr kinase inhibitor from Lilly (EIi Lilly), CVT-2584 (CAS No. 199986-75- 9) (CV Therapeutics), and an analogue or derivative thereof.
11652. The method of item 11626 wherein the agent is a cyclooxygenase 1 inhibitor.
11653. The method of item 11626 wherein the agent is a DHFR inhibitor.
11654. The method of item 11626 wherein the agent is a dual integrin inhibitor.
11655. The method of item 11626 wherein the agent is an elastase inhibitor.
11656. The method of item 11626 wherein the agent is an elongation factor-1 alpha inhibitor.
11657. The method of item 11626 wherein the agent is an endothelial growth factor antagonist. 11658. The method of item 11626 wherein the agent is an endothelial growth factor receptor kinase inhibitor selected from the group consisting of sorafenib tosylate (Bayer), AAL-993 (Novartis), ABP-309 (Novartis), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), E- 7080 (Eisai), EG-3306 (Ark Therapeutics), EXEL-2880 (Exelixis), GW- 654652 (GlaxoSmithKline), a KDR inhibitor from LG Life Sciences, CT-6685 and CT-6729 (UCB), KRN-633 and KRN-951 (Kirin Brewery), OSI-930 (OSI Pharmaceuticals), SP-5.2 (Supratek Pharma), SU-11657 (Pfizer), a Tie-2 antagonist (Hybrigenics), a VEGFR-2 kinase inhibitor (Bristol-Myers Squibb), XL-647 (Exelixis), a KDR inhibitor from Abbott Laboratories, sorafenib tosylate, and an analogue or derivative thereof.
11659. The method of item 11626 wherein the agent is an endotoxin antagonist.
11660. The method of item 11626 wherein the agent is an epothilone and tubulin binder.
11661. The method of item 11626 wherein the agent is an estrogen receptor antagonist.
11662. The method of item 11626 wherein the agent is an FGF inhibitor.
11663. The method of item 11626 wherein the agent is a famexyl transferase inhibitor.
11664. The method of item 11626 wherein the agent is famesyltransferase inhibitor selected from the group of A-197574 (Abbott), a farnesyltransferase inhibitor from Servier, FPTHI (Strathclyde Institute for Drug R), LB-42908 (LG Life Sciences), Pharmaprojects No. 5063 (Genzyme), Pharmaprojects No. 5597 (Ipsen), Yissum Project No. B-1055 (Yissum), and an analogue or derivative thereof.
11665. The method of item 11626 wherein the agent is an FLT-3 kinase inhibitor.
11666. The method of item 11626 wherein the agent is an FGF receptor kinase inhibitor.
11667. The method of item 11626 wherein the agent is a fibrinogen antagonist selected from the group consisting of AUV-201 (Auvation), MG-13926 (Sanofi-Aventis), plasminogen activator (CAS No. 105913-11-9) (from Sanofi-Aventis or UCB), plasminogen activator-2 (tPA- 2) (Sanofi-Aventis), pro-urokinase (CAS No. 82657-92-9) (Sanofi-Aventis), and an analogue or derivative thereof.
11668. The method of item 11626 wherein the agent is a heat shock protein 90 antagonist selected from the group consisting of SRN- 005 (Sirenade), geldanamycin, NSC-33050 (17-allylaminogeldanamycin; 17- AAG), 17-dimethylaminoethylamino-17-demethoxy-geldanamycin (17- DMAG), rifabutin (rifamycin XIV, 1',4-didehydro-1-deoxy-1 ,4-dihydro-5'-(2- methylpropyl)-1-oxo-), and an analogue or derivative thereof.
11669. The method of item 11626 wherein the agent is a histone deacetylase inhibitor.
11670. The method of item 11626 wherein the agent is an HMGCoA reductase inhibitor selected from the group consisting of an atherosclerosis therapeutic from Lipid Sciences, ATI-16000 (ARYx Therapeutics), KS-01-019 (Kos Pharmaceuticals), Pharmaprojects No. 2197 (Sanofi-Aventi), RP 61969 (Sanofi-Aventis), and an analogue or derivative thereof.
11671. The method of item 11626 wherein the agent is an ICAM inhibitor.
11672. The method of item 11626 wherein the agent is an IL, ICE and IRAK antagonist, wherein the antagonist is a CJ-14877, CP- 424174 (Pfizer), NF-61 (Negma-Lerads), and an analogue or derivative thereof.
11673. The method of item 11626 wherein the agent is an IL-2 inhibitor.
11674. The method of item 11626 wherein the agent is an immunosuppressant selected from the group consisting of teriflunomide (Sanofi Aventis), chlorsulfaquinoxalone (NSC-339004), chlorsulfaquinoxalone sulfate, CS-712 (Sankyo), ismomultin alfa (CAS No. 457913-93-8) (Akzo Nobel), antiallergics from GenPat77, antiinflammatories or AT-005 (Androclus Therapeutics), autoimmune disease therapeutics from EpiVax, BN-007 (Bone), budesonide (CAS No. 51333-22- 3) (MAP Pharmaceuticals), CO-14 (Genzyme), edratide (CAS No. 433922- 67-9) (Teva), EP-314 (Enanta), eprovafen (CAS No. 101335-99-3) (Sanofi- Aventis), HWA-131 (CAS No. 118788-41-3) (Sanofi-Aventis), immunomodulators from Merϋon Pharmaceuticals, immunosuppressives from Alchemia, IPL-12 (Inflazyme), MDL-9563 (CAS No. 27086-86-8) (Sanofi-Aventis), Pharmaprojects No. 2330 (Sanofi-Aventis), Pharmaprojects No. 6426 (Abgenix), PXS-25 (Pharmaxis), rosmarinic acid (CAS No. 20283-92-5) (Sanofi-Aventis), RP 42927 or RP 54745 (CAS No. 135330-08-4) (Sanofi-Aventis), SGN-35 (Seattle Genetics), ST-1959 (Sigma-Tau), type I diabetes therapy from SYNX Pharma, UNIL-88 (Debiopharm), VP-025 (Vasogen), VR-694 (Vectura), PRTX-001 (Protalex), and an analogue or derivative thereof.
11675. The method of item 11626 wherein the agent is an IMPDH (inosine monophosphate).
11676. The method of item 11626 wherein the agent is an integrin antagonist.
11677. The method of item 11626 wherein the agent is an interleukin antagonist.
11678. The method of item 11626 wherein the agent is an inhibitor of type III receptor tyrosine kinase.
11679. _ _. The method of item 11626 wherein the agent is . an irreversible inhibitor of enzyme methionine aminopeptidase type 2.
11680. The method of item 11626 wherein the agent is an isozyme selective delta protein kinase C inhibitor.
11681. The method of item 11626 wherein the agent a JAK3 enzyme inhibitor.
11682. The method of item 11626 wherein the agent is a JNK inhibitor.
11683. The method of item 11626 wherein the agent is a kinase inhibitor. 11684. The method of item 11626 wherein the agent is kinesin antagonist.
11685. The method of item 11626 wherein the agent is a kinesin antagonist.
11686. The method of item 11626 wherein the agent is a leukotriene inhibitor and antagonist selected from the group consisting of ambicromil (CAS No. 58805-38-2) (Sanofi-Aventis), amelubant (CAS No. 346735-24-8) (Boehringer Ingelheim), DW-1141 (Dong Wha), ebselen (Daiichi Pharmaceutical), ibudilast (Kyorin), leucotriene inhibitors from Sanofi-Aventis, lymphotoxin-beta receptor (LT-D) from Biogen Idee, Pharmaprojects No. 1535 and 2728 (CAS No. 119340-33-9) (Sanofi- Aventis), R-112 (Rigel), Rev-5367 (CAS No. 92532-05-3) (Sanofi-Aventis), RG-14893 (CAS No. 141835-49-6) (Sanofi-Aventis), RG-5901-A (CAS No. 101910-24-1), 92532-23-5, RP 66153 (CAS No. 142422-79-5), RP 66364 (CAS No.186916-92-5), RP 69698 (CAS No. 141748-00-7) (Sanofi-Aventis), SC-411930 (Pfizer), SC-41930 (CAS No. 120072-59-5) (Pfizer), SC-50605 (CAS No. 138828-39-4) (Pfizer), SC-51146 (CAS No. 141059-52-1), SC- 53228 (CAS No. 153633-01-3) (Pfizer), spaglumic acid (ZY-15106) (CAS No.3106-85-2), 80619-64-3 (Novartis), tipredane (CAS No. 85197-77-9) (Bristol-Myers Squibb), U-75302 (CAS No. 119477-85-9) (Pfizer), and analogue or derivative thereof.
11687. The method of item 11626 wherein the agent is an MAP kinase inhibitor.
11688. The method of item 11626 wherein the agent is a matrix metalloproteinase inhibitor. 11689. The method of item 11626 wherein the agent is an MCP-CCR2 inhibitor.
11690. The method of item 11626 wherein the agent is an mTOR inhibitor.
11691. The method of item 11626 wherein the agent is an mTOR kinase inhibitor.
11692. The method of item 11626 wherein the agent is a microtubule inhibitor selected from the group consisting of antibody- maytansinoid conjugates from Biogen Idee, colchicines (MantiCore Pharmaceuticals), anticancer immunoconjugates from Johnson & Johnson, DIME from Octamer, gni-1f (GNI), huC242-DM4, huMy9-6-DM1 (ImmunoGen), IDN-5404 (Indena), IMO-098, IMOderm (Imotep), mebendazole (Introgen Therapeutics), microtubule poisons from Cambridge Enterprise, paclitaxel such as LOTAX from Aphios (CAS No. 33069-62-4), Genexol-PM from Samyang, Pharmaprojects No. 6383 (Tapestry Pharmaceuticals), RPR-112378 (Sanofi-Aventis), SGN-75 (Seattle Genetics), SPL-7435 (Starpharma), SSR-250411 (Sanofi-Aventis), trastuzumab-DM1 (Genentech), vinorelbine, and an analogue or derivative thereof.
11693. The method of item 11626 wherein the agent is an MIF inhibitor.
11694. The method of item 11626 wherein the agent is an MMP inhibitor.
11695. The method of item 11626 wherein the agent is a neurokinin (NK) antagonist selected from the group consisting of anthrotainin (CAS No. 148084-40-6) (Sanofi-Aventis), an IBS thereapeutic from ArQuIe, MDL-105212A (CAS No. 167261-60-1) (Ssanofi-Aventis), Pharmaprojects No. 2744, 3258 (CAS No. 139167-47-8) 4006, 4201, or 5986 (Sanofi-Aventis), RP 67580 (CAS No. 135911-02-3), SR-144190 (CAS No. 201152-86-5), SSR-240600, SSR-241586 (Sanofi-Aventis), TKA-457 (Novartis), vestipitant mesylate (CAS No. 334476-64-1) (GlaxoSmithKline), Win-64821 (Sanofi-Aventis), PRX-96026 (Predix Pharmaceuticals), and an analogue or derivative thereof.
11696. The method of item 11626 wherein the agent is an NF kappa B inhibitor selected from the group consisting of AVE-0545 or AVE-0547 (Sanofi-Aventis), bortezomib (CAS No. 179324-69-7) (Millennium Pharmaceuticals), dexanabinol (CAS No. 112924-45-5) (Pharmos), dexlipotam (Viatris), Pharmaprojects No. 6283 (INDRA) (OXiGENE), IPL- 576092 (CAS No. 137571-30-3) (Inflazyme), NFKB decoy (Corgentech), NFKB decoy oligo (AnGes MG), NFKB's from Ariad, osteoporosis . . treatments or S5 (F005) from Fulcrum Pharmaceuticals, P61 (Phytopharm), R-flurbiprofen (CAS No. 5104-49-4) (Encore Pharmaceuticals), and an analogue or derivative thereof.
11697. The method of item 11626 wherein the agent is a nitric oxide agonist.
11698. The method of item 11626 wherein the agent is an ornithine decarboxylase inhibitor.
11699. The method of item 11626 wherein the agent is a p38 MAP kinase inhibitor selected from the group consisting of AZD-6703 (AstraZeneca), JX-401 (Jexys Pharmaceuticals), BMS-2 (Bristol-Myers Squibb), a p38 MAP kinase inhibitor from Novartis, a p38-alpha MAP kinase inhibitor from Amphora, Pharmaprojects No. 5704 (Pharmacopeia), RPR- 200765A (Sanofi-Aventis), SD-282 (Johnson & Johnson), TAK-715 (Takeda), and an analogue or derivative thereof.
11700. The method of item 11626 wherein the agent is a palmitoyl-protein thioesterase inhibitor.
11701. The method of item 11626 wherein the agent is a PDGF receptor kinase inhibitor selected from the group consisting of AAL- 993, AMN-107, or ABP-309 (Novartis), AMG-706 (Amgen), BAY-57-9352 (Bayer), CDP-860 (UCB), E-7080 (Eisai), imatinib (CAS No. 152459-95-5) (Novartis), OSI-930 (OSI Pharmaceuticals), RPR-127963E (Sanofi-Aventis), RWJ-540973 (Johnson & Johnson), sorafenib tosylate (Bayer), SU-11657 (Pfizer), tandutinib (CAS No. 387867-13-2) (Millennium Pharmaceuticals), vatalanib (Novartis), ZK-CDK (Schering AG), and an analogue or derivative thereof.
11702. The method of item 11626 wherein the agent is (-)-halofenate (Metabolex), AMG-131 (Amgen), antidiabetics from Japan Tobacco, AZD-4619, AZD-8450, AZD-8677 (AstraZeneca), DRF-10945, balaglitazone (Dr Reddy's), CS-00088, CS-00098 (Chipscreen Biosciences), E-3030 (Eisai), etalocib (CAS No. 161172-51-6) (EIi Lilly), GSK-641597 (Ligand), GSK-677954 (GlaxoSmithKline), GW-409544 (Ligand), GW- 590735 (GlaxoSmithKline), K-111 (Hoffmann-La Roche), LY-518674 (EIi Lilly), LY-674 (Ligand), LY-929 (Ligand), MC-3001 , MC-3002 (MaxoCore Pharmaceuticals), metformin HCI + pioglitazone (CAS No. 1115-70-4 and
112529-15-4), ACTOPLUS MET from Andrx), muraglitazar (CAS No. 331741-94-7) (Bristol-Myers Squibb), naveglitazar (Ligand), oleoylethanolamide (Kadmus Pharmaceuticals), ONO-5129, pioglitazone hydrochloride (CAS No. 111025-46-8 and 112529-15-4) (Takeda), PLX-204 (Plexxikon), PPAR agonists from Genfit, PPAR delta agonists from EIi Lilly, PPAR-alpha agonists from CrystalGenomics, PPAR-gamma modulators and PPAR-β modulators from CareX, rosiglitazone maleate (CAS No. 122324- 73-4 or 155141-29-0) (GlaxoSmithKline), rosiglitazone maleate/glimepir (CAS No. 155141-29-0 and 93479-97-1), AVANDARYL1 rosiglitazone maleate/metformin extend (CAS No. 155141-29-0 and 657-24-9), AVANDAMET, rosiglitazone maleate+metformin, AVANDAMET (GlaxoSmithKline), tesaglitazar (AstraZeneca), and an analogue or derivative thereof.
11703. The method of item 11626 wherein the agent is a phosphatase inhibitor.
11704. The method of item 11626 wherein the agent is a phosphodiesterase (PDE) inhibitor selected from the group consisting of avanafil (Tanabe Seiyaku), dasantafil (CAS No. 569351-91-3) (Schering- Plough), A-906119 (CAS No. 134072-58-5), DL-850 (Sanofi-Aventis), GRC- 3015, GRC-3566, GRC-3886 (Glenmark), HWA-153 (CAS No. 56395-66-5) (Sanofi-Aventis), hydroxypumafentrine (Altana), IBFB-130011 , IBFB-14-016, IBFB-140301 , IBFB-150007, IBFB-211913 (IBFB Pharma), L-826141 (Merck & Co), medorinone (CAS No. 88296-61-1) (Sanofi-Aventis), MEM- 1917 (Memory Pharmaceuticals), ND-1251 (Neuro3d), PDE inhibitors from ICOS, PDE IV inhibitors from Memory Pharmaceuticals and CrystalGenomics, Pharmaprojects No. 2742 and 6141 (Sanofi-Aventis), QAD-171 (Novartis), RHC-2963 (CAS No. 76993-12-9 and 76993-14-1), RPR-117658, RPR-122818 derivatives, SR-24870 , and RPR-132294 (Sanofi-Aventis), SK-350 (ln2Gen), stroke therapy agents from deCODE Genetics, TAS-203 (Taiho), tofimilast (CAS No. 185954-27-2) (Pfizer), UK- 371800 (Pfizer), WIN-65579 (CAS No. 158020-82-7) (Sanofi-Aventis), IBFB- 130020 (IBFB Pharma), OPC-6535 (CAS No. 145739-56-6) (Otsuka), a phosphodiesterase III inhibitor, enoximone, a phosphodiesterase IV inhibitor, fosfosal, Atopik (Barrier Therapeutics), triflusal, a phosphodiesterase V inhibitor, and an analogue or derivative thereof. 11705. The method of item 11626 wherein the agent is a PKC inhibitor.
11706. The method of item 11626 wherein the agent is a platelet activating factor antagonist.
11707. The method of item 11626 wherein the agent is a platelet-derived growth factor receptor kinase inhibitor.
11708. The method of item 11626 wherein the agent is a prolyl hydroxylase inhibitor.
11709. The method of item 11626 wherein the agent is a polymorphonuclear neutrophil inhibitor.
11710. The method of item 11626 wherein the agent is a protein kinase B inhibitor.
11711. The method of item 11626 wherein the agent is a protein kinase C stimulant.
11712. The method of item 11626 wherein the agent is a purine nucleoside analogue.
11713. The method of item 11626 wherein the agent is a purinoreceptor P2X antagonist.
11714. The method of item 11626 wherein the agent is a Raf kinase inhibitor. 11715. The method of item 11626 wherein the agent is a reversible inhibitor of ErbB1 and ErbB2.
11716. The method of item 11626 wherein the agent is a ribonucleoside triphosphate reductase inhibitor.
11717. The method of item 11626 wherein the agent is an SDF-1 antagonist.
11718. The method of item 11626 wherein the agent is a sheddase inhibitor.
11719. The method of item 11626 wherein the agent is an SRC inhibitor.
11720. The method of item 11626 wherein the agent is a stromelysin inhibitor.
11721. The method of item 11626 wherein the agent is an Syk kinase inhibitor.
11722. The method of item 11626 wherein the agent is a telomerase inhibitor.
11723. The method of item 11626 wherein the agent is a TGF beta inhibitor selected from the group consisting of pirfenidone (CAS No. 53179-13-8) (MARNAC), tranilast (CAS No. 53902-12-8) (Kisser), IN- 1130 (ln2Gen), mannose-6-phosphate (BTG), TGF-β antagonists from Inflazyme (Pharmaprojects No. 6075), TGF-β antagonists from Sydney, non-industrial source), TGF-β I receptor kinase inhibitors from EIi Lilly, TGF- β receptor inhibitors from Johnson & Johnson, and an analogue or derivative thereof.
11724. The method of item 11626 wherein the agent is a
TNFα antagonist or TACE inhibitor selected from the group consisting of adalimumab (CAS No. 331731-18-1) (Cambridge Antibody Technology), AGIX-4207 (AtheroGenics), AGT-1 (Advanced Biotherapy), an antiinflammatory from Borean Pharma, Cellzome, or Paradigm Therapeutics, anti-inflammatory vaccine (TNF-alpha kinoid) from Neovacs, humanized anti-TNF antibody or an anti-TNF MAb (CB0006) Celltech (UCB), apratastat (CAS No. 287445-51-0) (Wyeth), BMS-561392 (Bristol-Myers Squibb), BN- 006 (Bone), certolizumab pegol (CAS No. 428863-50-7 or CH-138 (UCB), cilomilast (CAS No. 153259-65-5) (GlaxoSmithKline), CR-1 (Nuada Pharmaceuticals), CRx-119 (CombinatoRx), D-5410 (UCB), dacopafant (CAS No. 125372-33-0) (Sanofi-Aventis), dersalazine (CAS No. 188913-57- 7/188913-58-8) (Uriach), etanercept (CAS No. 185243-69rO).(Amgen), -ethyl pyruvate (Critical (Critical Therapeutics), golimumab (CAS No. 476181-74-5) (Johnson & Johnson), hormono-immunotherapy from Ipsen, CDP571 (e.g., Humicade from UCB), IC-485 (ICOS), infliximab (CAS No. 170277-31-3) (Johnson & Johnson), IP-751 (Manhattan Pharmaceuticals), ISIS-104838 (CAS No. 250755-32-9) (ISIS Pharmaceuticals), lenalidomide (CAS No. 191732-72-6) (Celgene), lentinan (CAS No. 37339-90-5) (Ajinomoto), MDL- 201112 (CAS No. 142130-73-2) (Sanofi-Aventis), medroxyprogesterone (CAS No. 520-85-4) (InKine Pharmaceutical), N-acetylcysteine (CAS No. 616-91-1) (Zambon), NBE-P2 (DIREVO Biotech), nerelimomab (CAS No. 162774-06-3) (Chiron), OM-294DP (OM PHARMA), onercept (CAS No. 199685-57-9) (Yeda), PASSTNF-alpha (Verigen), pentoxifylline or oxypentifylline (Sanofi-Aventis), Pharmaprojects No. 4091 , 4241, 4295, or 4488 (Sanofi-Aventis), Pharmaprojects No. 5480 (Amgen), Pharmaprojects No. 6749 (Cengent), pirfenidone (CAS No. 53179-13-8) (MARNAC), RPR- 132294 (Sanofi-Aventis), S5 (F002) (Fulcrum Pharmaceuticals), simvastatin (CAS No. 79902-63-9) (Merck & Co), STA-6292 (Synta Pharmaceuticals), tacrolimus (CAS No. 104987-11-3) (Fujisawa LifeCycle Pharma), talactoferrin alfa (CAS No. 308240-58-6) (Agennix), thalidomide (CAS No. 50-35-1) (Celgene), TNF antagonists form ProStrakan, and Synergen, TNF inhibitors (Amgen), TNF-alpha antagonists from Dynavax Technologies and Jerina AG (Germany), TNF-alpha inhibitors from IBFB Pharma and Xencor (Xencor), torbafylline (CAS No. 105102-21-4) (Sanofi-Aventis), UR-1505 (Uriach), VT-346 (Viron Therapeutics), YSIL6 (Y's Therapeutics), YSTH2 (Y's Therapeutics), NPI-1302a-3 (Nereus Pharmaceuticals, a TNF antagonist from Jerina AG (Germany), dersalazine, and an analogue or derivative thereof.
11725. The method of item 11626 wherein the agent is a Toll receptor inhibitor.
11726. The method of item 11626 wherein the agent is a tubulin antagonist.
11727. The method of item 11626 wherein the agent is a tyrosine kinase inhibitor selected from the group consisting of SU-011248, SUTENT from Pfizer Inc. (New York, NY), BMS-354825, PN-355 (Paracelsian Pharmaceuticals), AGN-199659 (Allergan), AAL-993 or ABP- 309 (Novartis), adaphostin (NIH), AEE-788 (Novartis), AG-013736 (OSI Pharmaceuticals), AG-13736 (Pfizer), ALT-110 (Alteris Therapeutics), AMG- 706 (Amgen), anticancer MAbs from Xencor, anti-EGFrvlll MAbs from Abgenix, anti-HER2 MAb from Abiogen, AZD-2171 or A2D-9935 (AstraZeneca), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), CEP-5214 (Cephalon), CEP-7055 (Cephalon), cetuximab (ImClone Systems), CHIR-200131 and CHIR-258 (Chiron), CP-547632 (OSI Pharmaceuticals), CP-724714 (Pfizer), CT-301 (Creabilis Therapeutics), D- 69491 (Baxter International), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EGFR/ErbB2 inhibitors from Array BioPharma, erlotinib (CAS No. 183319- 69-9) (OSI Pharmaceuticals), EXEL-2880 (Exelixis), FK-778 (Sanofi- Aventis) , gefitinib (CAS No. 184475-35-2) (AstraZeneca), GW-2286 or GW- 654652 (GlaxoSmithKline), her2/neu antigen from AlphaVax, HER-2/neu inhibitor from Generex, Herzyme (Medipad) (Sirna Therapeutics), HKI-272 (Wyeth), HuMax-EGFr (Genmab), idronoxil (CAS No. 81267-65-4) (Novogen), IGF-1 inhibitors from Ontogen, IMC-11 F8 (ImCIone Systems), kahalalide F (CAS No. 149204-42-2) (PharmaMar), KDR inhibitor from LG Life Sciences, KDR inhibitors from Abbott Laboratories, KDR kinase inhibitors (UCB), Kdr kinase inhibitors from Merck & Co, KRN-633 and KRN- 951 (Kirin Brewery), KSB-102 (Xenova), lapatinib ditosylate (CAS No. 388082-78-8) (GlaxoSmithKline), matuzumab (Merck KGaA), MDX-214 (Medarex), ME-103 (Pharmexa), MED-A300 (Gerolymatos), MNAC-13 (Lay Line Genomics), nimotuzumab (Center of Molecular Immunology), NSC- 330507 or NSC-707545 (NIH), NV-50 (Novogen), OSI-930 (OSI Pharmaceuticals), panitumumab (Abgenix), pelitinib (CAS No. 287933-82-7) (Wyeth), pertuzumab (CAS No. 380610-27-5) (Genentech), Pharmaprojects No. 3985 (Sanofi-Aventis), prostate cancer therapeutics from Sequenom (SQPC35, SQPC36, SQPC90), removab and remoxab (Trion Pharma), RG- 13022 (CAS No. 136831-48-6), RG-13291 (CAS No. 138989-50-1), or RG- 14620 (CAS No. 136831-49-7) (Sanofi-Aventis), RM-6427 (Romark), RNAi breast cancer therapy from Benitec, RP 53801 (CAS No. 125882-88-4) (Sanofi-Aventis), sorafenib tosylate (Bayer), SU-11657 (Pfizer), Tie-2 antagonists from Semaia (Hybrigenics), Tie-2 inhibitors from Ontogen, trastuzumab (CAS No. 180288-69-1) (Genentech), tyrosine kinase inhibitors from Sanofi-Aventis, U3-1287, U3-1565, U3-1784, U3-1800 (U3 Pharma), vatalanib (Novartis), VEGFR-2 kinase inhibitor from Bristol-Myers Squibb, XL-647 (Exelixis), ZD-6474 (AstraZeneca), ZK-CDK (Schering AG), an EGFR tyrosine kinase inhibitor, EKB-569 (Wyeth), and an analogue or derivative thereof. 11728. The method of item 11626 wherein the agent is a VEGF inhibitor.
11729. The method of item 11626 wherein the agent is a vitamin D receptor agonist.
11730. The method of item 11626 wherein the agent is ZD-6474 (an angiogenesis inhibitor).
11731. The method of item 11626 wherein the agent is AP-23573 (an mTOR inhibitor).
11732. The method of item 11626 wherein the agent is synthadotin (a tubulin antagonist).
11733. The method of item 11626 wherein the agent is S-0885 (a collagenase inhibitor).
11734. The method of item 11626 wherein the agent is aplidine (an elongation factor-1 alpha inhibitor).
11735. The method of item 11626 wherein the agent is ixabepilone (an epithilone).
11736. The method of item 11626 wherein the agent is IDN-5390 (an angiogenesis inhibitor).
11737. The method of item 11626 wherein the agent is SB-2723005 (an angiogenesis inhibitor). 11738. The method of item 11626 wherein the agent is ABT-518 (an angiogenesis inhibitor).
11739. The method of item 11626 wherein the agent is combretastatin (an angiogenesis inhibitor).
11740. The method of item 11626 wherein the agent is anecortave acetate (an angiogenesis inhibitor).
11741. The method of item 11626 wherein the agent is SB-715992 (a kinesin antagonist).
11742. The method of item 11626 wherein the agent is temsirolimυs (an mTOR inhibitor).
11743. The method of item 11626 wherein the agent is adalimumab (a TNFα antagonist).
11744. The method of item 11626, wherein the composition comprises a polymer.
11745. The method of item 11626, wherein the composition comprises a polymeric carrier.
11746. The method of item 11626 wherein the anti- scarring agent inhibits adhesion between the device and a host into which the device is implanted.
11747. The method of item 11626 wherein the device delivers the anti-scarring agent locally to tissue proximate to the device. 11748. The method of item 11626 wherein the device has a coating that comprises the anti-scarring agent.
11749. The method of item 11626, wherein the device has a coating that comprises the agent and is disposed on a surface of the implant.
11750. The method of item 11626, wherein the device has a coating that comprises the agent and directly contacts the implant.
11751. The method of item 11626, wherein the device has a coating that comprises the agent and indirectly contacts the implant.
11752. The method of item 11626, wherein the device has a coating that comprises the agent and partially covers the implant.
11753. The method of item 11626, wherein the device has a coating that comprises the agent and completely covers the implant.
11754. The method of item 11626, wherein the device has a uniform coating.
11755. The method of item 11626, wherein the device has a non-uniform coating.
11756. The method of item 11626, wherein the device has a discontinuous coating.
11757. The method of item 11626, wherein the device has a patterned coating. 11758. The method of item 11626, wherein the device has a coating with a thickness of 100 Dm or less.
. 11759. The method of item 11626, wherein the device has a coating with a thickness of 10 Dm or less.
11760. The method of item 11626, wherein the device has a coating, and the coating adheres to the surface of the implant upon deployment of the implant.
11761. The method of item 11626, wherein the device has a coating, and wherein the coating is stable at room temperature for a period of 1 year.
11762. The method of item 11626, wherein the device has a coating, and wherein the anti-scarring agent is present in the coating in an amount ranging between about 0.0001% to about 1% by weight.
11763. The method of item 11626, wherein the device has a coating, and wherein the anti-scarring agent is present in the coating in an amount ranging between about 1% to about 10% by weight.
11764. The method of item 11626, wherein the device has a coating, and wherein the anti-scarring agent is present in the coating in an amount ranging between about 10% to about 25% by weight.
11765. The method of item 11626, wherein the device has a coating, and wherein the anti-scarring agent is present in the coating in an amount ranging between about 25% to about 70% by weight. 11766. The method of item 11626, wherein the device has a coating, and wherein the coating further comprises a polymer.
11767. The method of item 11626, wherein the device has a first coating having a first composition and a second coating having a second composition.
11768. The method of item 11626, wherein the device has a first coating having a first composition and a second coating having a second composition, wherein the first composition and the second composition are different.
11769. The method of item 11626, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a copolymer.
11770. The method of item 11626, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a block copolymer.
11771. The method of item 11626, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a random copolymer.
11772. The method of item 11626, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a biodegradable polymer.
11773. The method of item 11626, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a non-biodegradable polymer. 11774. The method of item 11626, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a hydrophilic polymer.
11775. The method of item 11626, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a hydrophobic polymer.
11776. The method of item 11626, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a polymer having hydrophilic domains.
11777. The method of item 11626, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a polymer having hydrophobic domains.
11778. The method of item 11626, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a non-conductive polymer.
11779. The method of item 11626, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises an elastomer.
11780. The method of item 11626, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a hydrogel.
11781. The method of item 11626, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a silicone polymer. 11782. The method of item 11626, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a hydrocarbon polymer.
11783. The method of item 11626, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a styrene-derived polymer.
11784. The method of item 11626, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a butadiene polymer.
11785. The method of item 11626, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a macromer.
11786. The method of item 11626, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a poly(ethylene glycol) polymer.
11787. The method of item 11626 wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises an amorphous polymer.
11788. The method of item 11626, wherein the device comprises a lubricious coating.
11789. The method of item 11626 wherein the anti- scarring agent is located within pores or holes of the device. 11790. The method of item 11626 wherein the anti- scarring agent is located within a channel, lumen, or divet of the device.
11791. The method of item 11626, wherein the device comprises a second pharmaceutically active agent.
11792. The method of item 11626 wherein the device comprises an anti-inflammatory agent.
11793. The method of item 11626 wherein the device comprises an agent that inhibits infection.
11794. The method of item 11626 wherein the device comprises an agent that inhibits infection, and wherein the agent is an anthracycline.
11795. The method of item 11626 wherein the device comprises an agent that inhibits infection, and wherein the agent is doxorubicin.
11796. The method of item 11626 wherein the device comprises an agent that inhibits infection, and wherein the agent is mitoxantrone.
11797. The method of item 11626 wherein the device comprises an agent that inhibits infection, and wherein the agent is a fluoropyrimidine.
11798. The method of item 11626 wherein the device comprises an agent that inhibits infection, and wherein the agent is 5- fluorouracil (5-FU). 11799. The method of item 11626 wherein the device comprises an agent that inhibits infection, and wherein the agent is a folic acid antagonist.
11800. The method of item 11626 wherein the device comprises an agent that inhibits infection, and wherein the agent is methotrexate.
11801. The method of item 11626 wherein the device comprises an agent that inhibits infection, and wherein the agent is a podophylotoxin.
11802. The method of item 11626 wherein the device comprises an agent that inhibits infection, and wherein the agent is etoposide.
11803. The method of item 11626 wherein the device comprises an agent that inhibits infection, and wherein the agent is a camptothecin.
11804. The method of item 11626 wherein the device comprises an agent that inhibits infection, and wherein the agent is a hydroxyurea.
11805. The method of item 11626 wherein the device comprises an agent that inhibits infection, and wherein the agent is a platinum complex.
11806. The method of item 11626 wherein the device comprises an agent that inhibits infection, and wherein the agent is cisplatin. 11807. The method of item 11626, further comprising an anti-thrombotic agent.
11808. The method of item 11626 wherein the device comprises a visualization agent.
11809. The method of item 11626 wherein the device comprises a visualization agent, wherein the visualization agent is a radiopaque material, and wherein the radiopaque material comprises a metal, a halogenated compound, or a barium containing compound.
11810. The method of item 11626 wherein the device comprises a visualization agent, wherein the visualization agent is a radiopaque material, and wherein the radiopaque material comprises barium, tantalum, or technetium.
11811. The method of item 11626 wherein the device comprises a visualization agent, and wherein the visualization agent is a MRI responsive material.
11812. The method of item 11626 wherein the device comprises a visualization agent, and wherein the visualization agent comprises a gadolinium chelate.
11813. The method of item 11626 wherein the device comprises a visualization agent, and wherein the visualization agent comprises iron, magnesium, manganese, copper, or chromium.
11814. The method of item 11626 wherein the device comprises a visualization agent, and wherein the visualization agent comprises an iron oxide compound. 11815. The method of item 11626 wherein the device comprises a visualization agent, and wherein the visualization agent comprises a dye, pigment, or colorant.
11816. The method of item 11626 wherein the device comprises an echogenic material.
11817. The method of item 11626 wherein the device comprises an echogenic material, and wherein the echogenic material is in the form of a coating.
11818. The method of item 11626 wherein the device is sterile.
11819. The method of item 11626 wherein the anti- scarring agent is released into tissue in the vicinity of the device after deployment of the device.
11820. The method of item 11626 wherein the anti- scarring agent is released into tissue in the vicinity of the device after deployment of the device, and wherein the tissue is connective tissue.
11821. The method of item 11626 wherein the anti- scarring agent is released into tissue in the vicinity of the device after deployment of the device, and wherein the tissue is muscle tissue.
11822. The method of item 11626 wherein the anti- scarring agent is released into tissue in the vicinity of the device after deployment of the device, and wherein the tissue is nerve tissue. 11823. The method of item 11626 wherein the anti- scarring agent is released into tissue in the vicinity of the device after deployment of the device, and wherein the tissue is epithelium tissue.
11824. The method of item 11626 wherein the anti- scarring agent is released in effective concentrations from the device over a period ranging from the time of deployment of the device to about 1 year.
11825. The method of item 11626 wherein the anti- scarring agent is released in effective concentrations from the device over a period ranging from about 1 month to 6 months.
11826. The method of item 11626 wherein the anti- scarring agent is released in effective concentrations from the device over a period ranging from about 1 - 90 days.
11827. The method of item 11626 wherein the anti- scarring agent is released in effective concentrations from the device at a constant rate.
11828. The method of item 11626 wherein the anti- scarring agent is released in effective concentrations from the device at an increasing rate.
11829. The method of item 11626 wherein the anti- scarring agent is released in effective concentrations from the device at a decreasing rate.
11830. The method of item 11626 wherein the anti- scarring agent is released in effective concentrations from the composition comprising the anti-scarring agent by diffusion over a period ranging from the time of deployment of the device to about 90 days.
11831. The method of item 11626 wherein the anti- scarring agent is released in effective concentrations from the composition comprising the anti-scarring agent by erosion of the composition over a period ranging from the time of deployment of the device to about 90 days.
11832. The method of item 11626 wherein the device comprises about 0.01 Dg to about 10 Dg of the anti-scarring agent.
11833. The method of item 11626 wherein the device comprises about 10 Dg to about 10 mg of the anti-scarring agent.
11834. The method of item 11626 wherein the device comprises about 10 mg to about 250 mg of the anti-scarring agent.
11835. The method of item 11626 wherein the device comprises about 250 mg to about 1000 mg of the anti-scarring agent.
11836. The method of item 11626 wherein the device comprises about 1000 mg to about 2500 mg of the anti-scarring agent.
11837. The method of item 11626 wherein a surface of the device comprises less than 0.01 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
11838. The method of item 11626 wherein a surface of the device comprises about 0.01 Dg to about 1 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied. 11839. The method of item 11626 wherein a surface of the device comprises about 1 Dg to about 10 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
11840. The method of item 11626 wherein a surface of the device comprises about 10 Dg to about 250 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
11841. The method of item 11626 wherein a surface of the device comprises about 250 Dg to about 1000 Dg of the anti-scarring agent of anti-scarring agent per mm2 of device surface to which the anti- scarring agent is applied.
11842. The method of item 11626 wherein a surface of the device comprises about 1000 Dg to about 2500 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
11843. The method of item 11626 wherein the combining is performed by direct affixing the agent or the composition to the implant.
11844. The method of item 11626 wherein the combining is performed by spraying the agent or the component onto the implant.
11845. The method of item 11626 wherein the combining is performed by electrospraying the agent or the composition onto the implant. 11846. The method of item 11626 wherein the combining is performed by dipping the implant into a solution comprising the agent or the composition.
11847. The method of item 11626 wherein the combining is performed by covalently attaching the agent or the composition to the implant.
11848. The method of item 11626 wherein the combining is performed by non-covalently attaching the agent or the composition to the implant.
11849. The method of item 11626 wherein the combining is performed by coating the implant with a substance that contains the agent or the composition.
11850. The method of item 11626 wherein the combining is performed by coating the implant with a substance that absorbs the agent.
11851. The method of item 11626 wherein the combining is performed by interweaving a thread composed of, or coated with, the agent or the composition.
11852. The method of item 11626 wherein the combining is performed by covering all the implant with a sleeve that contains the agent or the composition.
11853. The method of item 11626 wherein the combining is performed by covering a portion of the implant with a sleeve that contains the agent or the composition. 11854. The method of item 11626 wherein the combining is performed by covering all the implant with a cover that contains the agent or the composition.
11855. The method of item 11626 wherein the combining is performed by covering a portion of the implant with a cover that contains the agent or the composition.
11856. The method of item 11626 wherein the combining is performed by covering all the implant with an electrospun fabric that contains the agent or the composition.
11857. The method of item 11626 wherein the combining is performed by covering a portion of the implant with an electrospun fabric that contains the agent or the composition.
11858. The method of item 11626 wherein the combining is performed by covering all the implant with a mesh that contains the agent or the composition.
11859. The method of item 11626 wherein the combining is performed by covering a portion of the implant with a mesh that contains the agent or the composition.
11860. The method of item 11626 wherein the combining is performed by constructing all the implant with the agent or the composition.
11861. The method of item 11626 wherein the combining is performed by constructing a portion of the implant with the agent or the composition. 11862. The method of item 11626 wherein the combining is performed by impregnating the implant with the agent or the composition.
11863. The method of item 11626 wherein the combining is performed by constructing all of the implant from a degradable polymer that releases the agent.
11864. The method of item 11626 wherein the combining is performed by constructing a portion of the implant from a degradable polymer that releases the agent.
11865. The method of item 11626 wherein the combining is performed by dipping the implant into a solution that comprise the agent and an inert solvent for the implant.
11866. The method of item 11626 wherein the combining is performed by dipping the implant into a solution that comprises the agent and a solvent that will swell the implant.
11867. The method of item 11626 wherein the combining is performed by dipping the implant into a solution that comprises the agent and a solvent that will dissolve the implant.
11868. The method of item 11626 wherein the combining is performed by dipping the implant into a solution that comprises the agent, a polymer and an inert solvent for the implant.
11869. The method of item 11626 wherein the combining is performed by dipping the implant into a solution that comprises the agent, a polymer and a solvent that will swell the implant. 11870. The method of item 11626 wherein the combining is performed by dipping the implant into a solution that comprises the agent, a polymer and a solvent that will dissolve the implant.
11871. The method of item 11626 wherein the combining is performed by spraying the implant into a solution that comprises the agent and an inert solvent for the implant.
11872. The method of item 11626 wherein the combining is performed by spraying the implant into a solution that comprises the agent and a solvent that will swell the implant.
11873. The method of item 11626 wherein the combining is performed by spraying the implant into a solution that comprises the agent and a solvent that will dissolve the implant.
11874. The method of item 11626 wherein the combining is performed by spraying the implant into a solution that comprises the agent, a polymer and an inert solvent for the implant.
11875. The method of item 11626 wherein the combining is performed by spraying the implant into a solution that comprises the agent, a polymer and a solvent that will swell the implant.
11876. The method of item 11626 wherein the combining is performed by spraying the implant into a solution that comprises the agent, a polymer and a solvent that will dissolve the implant.
11877. A method as in any one of items 11626-11876, wherein the device is an esophageal stent. 11878. A method as in any one of items 11626-11876, wherein the device is a biliary stent.
11879. A method as in any one of items 11626-11876, wherein the device is a colonic stent.
11880. A method as in any one of items 11626-11876, wherein the device is a pancreatic stent.
11881. A method of making a medical device comprising: combining a tracheal and bronchial stent (e.g., an implant) and an anti-scarring agent or a composition comprising an anti-scarring agent, wherein the agent inhibits scarring between the device and a host into which the device is implanted.
11882. The method of item .11881 wherein the agent is an adensosine A2A receptor antagonist.
11883. The method of item 11881 wherein the agent is an AKT inhibitor.
11884. The method of item 11881 wherein the agent is an alpha 2 integrin antagonist, wherein the alpha 2 integrin antagonist is Pharmaprojects No. 5754 (Merck KgaA).
11885. The method of item 11881 wherein the agent is an alpha 4 integrin antagonist.
11886. The method of item 11881 wherein the agent is an alpha 7 nicotinic receptor agonist. 11887. The method of item 11881 wherein the agent is an angiogenesis inhibitor selected from the group consisting of AG-12,958 (Pfizer), ATN-161 (Attention LLC), neovastat, an angiogenesis inhibitor from Jerina AG (Germany), NM-3 (Mercian), VGA-1155 (Taisho), FCE-26644 (Pfizer), FCE-26950 (Pfizer), FPMA (Meiji Daries), FR-111142 (Fujisawa), GGTI-298, GM-1306 (Ligand), GPA-1734 (Novartis), NNC-47-0011 (Novo Nordisk), herbamycin (Nippon Kayaku), lenalidomide (Celegene), IP-10 (NIH), ABT-828 (Abbott), KIN-841 (Tokushima University, Japan), SF-1126 (Semafore Pharmaceuticals), laminin technology (NIH), CHIR-258 (Chiron), NVP-AEW541 (Novartis), NVP-AEW541 (Novartis), Vt16907 (Alchemia), OXI-8007 (Oxigene), EG-3306 (Ark Therapeutics), Maspin (Arriva), ABT- 567 (Abbott), PPI-2458 (Praecis Pharmaceuticals), CC-5079, CC-4089 (Celgene), HIF-1 alpha inhibitors (Xenova), S-247 (Pfizer), AP-23573 (Ariad), AZD-9935 (Astra Zeneca), mebendazole (Introgen Therapeutics), MetAP-2 inhibitors (GlaxoSmithKline), AG-615 (Angiogene Pharmaceuticals), Tie-2 antagonists (Hybrigenics), NC-381 , CYC-381, NC-169, NC-219, NC-383, NC-384, NC-407 (Lorus Therapeutics), ATN-224 (Attenuon), ON-01370 (Onconova), Vitronectin antagonists (Amgen), SDX-103 (Salmedix), Vitronectin antagonists (Shire), CHP (Riemser), TEK (Amgen), Anecortave acetate (Alcon), T46.2 (Matrix Therapeutics), HG-2 (Heptagen), TEM antagonists (Genzyme), Oxi-4500 (Oxigene), ATN-161 (Attenuon), WX-293 (Wilex), M-2025 (Metris Therapeutics), Alphastatin (BioActa), YH-16 (Yantai Rongchang), BIBF-1120 (Boehringer Ingelheim), BAY-57-9352 (Bayer), AS- 1404 (Cancer Research Technology), SC-77964 (Pfizer), glycomimetics (BioTie Therapies), TIE-2 Inhibitors (Ontogen), DIMI, Octamer (Octamer), ABR-215050 (Active Biotech), ABT-518 (Abbott), KDR inhibitors (Abbott), BSF-466895 (Abbott), SCH-221153 (Schering-Plough), DAC.antiangiogenic (ConjuChem), TFPI (EntreMed), AZD-2171 (Astra-Zenaca), CDC-394 (Celgene), LY290293 (EIi Lilly), IDN-5390 (Indena), Kdr Kinase Inhibitors (Merck), CT-113020, CT-116433, CT-116563, CT-31890, CT-32228) (Cell Therapeutics), A-299620 (Abbott), TWEAK Inhibitor (Amgen), VEGF modulators (Johnson and Johnson), Tum-N53, tumstatin (Genzyme), Thios- 1 , Thios-2 (Thios Pharmaceuticals), MV-6401 (Miravant Medical Technologies), Spisulosine (PharmaMar), CEP-7055 (Cephalon), AUV-201 (Auvation), LM-609 (EIi Lilly), SKF-106615 (AnorMED), Oglufanide disodium (Cytran), BW-114 (Pharminox), Calreticulin (NIH), WX-678 (Wilex), SD-7784 (Pfizer), WX-UK1 (Wilex), SH-268 (Schering AG), 2-Me-PGA (Celgene), S- 137 (Pfizer), ZD-6126 (Angiogene Pharmaceuticals), SG-292 (SignalGen), Benefin (Lane Labs), A6, A36 (Angstrom), SB-2723005 (GlaxoSmithKline), SC-7 (Cell Therapeutics), ZEN-014 (AEterna Zentaris), 2-methoxyestradiol (EntreMed), NK-130119 (Nippon Kayaku), CC-10004 (Celgene), AVE- 8062A (Ajinomoto), Tacedinaline (Pfizer), Actinonin (Tokyo Metropolitan Institute of Medical Science), Lenalidomide (Celgene), VGA-1155, BTO-956 (SRI International), ER-68203-00 (Eisai), CT-6685 (UCB), JKC-362 (Phoenix Pharmaceuticals), DMI-3798 (DMI Biosciences, Angiomate (Ipsen), ZD-6474 (AstraZeneca), CEP-5214 (Cephalon), Canstatin (Genzyme), NM-3 (Mercian), Oridigm (MediQuest Therapeutics), Exherin (Adherex), BLS-0597 (Boston Life Sciences), PTC-299 (PTC Therapeutics), NPI-2358 (Nereus Pharmaceuticals), CGP-79787 (Novartis), AEE-788 (Novartis), CKD-732 (Chong Kun Dang), CP-564959 (OSI Pharmaceuticals), CM-101 (CarboMed), CT-2584, CT3501 (Cell Therapeutics), combretastatin and analogues and derivatives thereof (Oxigene), Rebimastat (Bristol-Meyers Squibb), Dextrin 2-sulfate (ML Laboratories), Cilengitide (Merk KGaA), NSC-706704 (Pharminox), KRN- 951 (Kirin Brewery), Ukrain, NSC-631570 (Nowicky Pharma), Tecogalan sodium (Daiichi Pharmaceutical), Tz-93 (Tsumura), TBC-1635 (Encysive Pharmaceuticals), TAN-1120 (Takeda), Semaxanib (Pfizer), BDI-7800 (Biopharmacopae), SD-186, SD-983 (Bristol-Meyers Squibb), SB-223245 (GlaxoSmithKline), SC-236 (Pfizer), RWJ-590973 (Johnson and Johnson), ILX-1850 (Genzyme), SC-68488, S-836 (Pfizer), CG-55069-11 (CuraGen), Ki-23057 (Kirin Brewery), CCX-700 (Chemoentryx), Pegaptanib octasodium (Giled Sciences), ANGIOCOL (available from Biostratum Inc.), or an analogue or derivative thereof.
11888. The method of item 11881 wherein the agent is an apoptosis antagonist.
11889. The method of item 11881 wherein the agent is an apoptosis activator.
11890. The method of item 11881 wherein the agent is a beta 1 integrin antagonist.
11891. The method of item 11881 wherein the agent is a beta tubulin inhibitor.
11892. The method of item 11881 wherein the agent is a blocker of enzyme production in Hepatitis C.
11893. The method of item 11881 wherein the agent is a Bruton's tyrosine kinase inhibitor.
11894. The method of item 11881 wherein the agent is a calcineurin inhibitor.
11895. The method of item 11881 wherein the agent is a caspase 3 inhibitor.
11896. The method of item 11881 wherein the agent is a CC chemokine receptor antagonist. 11897. The method of item 11881 wherein the agent is a cell cycle inhibitor selected from the group consisting of SNS-595 (Sunesis), synthadotin, KRX-0403, and an analogue or derivative thereof.
11898. The method of item 11881 wherein the agent is a cathepsin B inhibitor.
11899. The method of item 11881 wherein the agent is a cathepsin K inhibitor, wherein the cathepsin K inhibitor is 462795 (GlaxoSmithKline), INPL-022-D6 (Amura Therapeutics), or an analogue or derivative thereof.
11900. The method of item 11881 wherein the agent is a cathepsin L inhibitor.
11901. The method of item 11881 wherein the agent is a CD40 antagonist.
11902. The method of item 11881 wherein the agent is a chemokine receptor agonist.
11903. The method of item 11881 wherein the agent is a chymase inhibitor.
11904. The method of item 11881 wherein the agent is a collagenase antagonist.
11905. The method of item 11881 wherein the agent is a CXCR antagonist. 11906. The method of item 11881 wherein the agent is a cyclin dependent kinase inhibitor selected from the group consisting of a CDK- 1 inhibitor, a CDK-2 inhibitor, a CDK- 4 inhibitor, a CDK-6 inhibitor, a CAK1 inhibitor from GPC Biotech and Bristol-Myers Squibb, RGB-286199 (GPC Biotech), an anticancer agent from Astex Technology, a CAK1 inhibitor from GPC Biotech, a CDK inhibitor from Sanofi-Aventis, a CDK1/CDK2 inhibitor from Amgen, a CDK2 inhibitor from SUGEN-2 (Pfizer), a hearing loss therapy agent (Sound Pharmaceuticals), PD-0332991 (Pfizer), RGB-286199 (GPC Biotech), Ro-0505124 (Hoffmann-La Roche), a Ser/Thr kinase inhibitor from Lilly (EIi Lilly), CVT-2584 (CAS No. 199986-75- 9) (CV Therapeutics), and an analogue or derivative thereof.
11907. The method of item 11881 wherein the agent is a cyclooxygenase 1 inhibitor.
11908. The method of item 11881 wherein the agent is a DHFR inhibitor.
11909. The method of item 11881 wherein the agent is a dual integrin inhibitor.
11910. The method of item 11881 wherein the agent is an elastase inhibitor.
11911. The method of item 11881 wherein the agent is an elongation factor-1 alpha inhibitor.
11912. The method of item 11881 wherein the agent is an endothelial growth factor antagonist. 11913. The method of item 11881 wherein the agent is an endothelial growth factor receptor kinase inhibitor selected from the group consisting of sorafenib tosylate (Bayer), AAL-993 (Novartis), ABP-309 (Novartis), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), E- 7080 (Eisai), EG-3306 (Ark Therapeutics), EXEL-2880 (Exelixis), GW- 654652 (GlaxoSmithKline), a KDR inhibitor from LG Life Sciences, CT-6685 and CT-6729 (UCB), KRN-633 and KRN-951 (Kirin Brewery), OSI-930 (OSI Pharmaceuticals), SP-5.2 (Supratek Pharma), SU-11657 (Pfizer), a Tie-2 antagonist (Hybrigenics), a VEGFR-2 kinase inhibitor (Bristol-Myers Squibb), XL-647 (Exelixis), a KDR inhibitor from Abbott Laboratories, sorafenib tosylate, and an analogue or derivative thereof.
11914. The method of item 11881 wherein the agent is an endotoxin antagonist.
11915. The method of item 11881 wherein the agent is an epothilone and tubulin binder.
11916. The method of item 11881 wherein the agent is an estrogen receptor antagonist.
11917. The method of item 11881 wherein the agent is an FGF inhibitor.
11918. The method of item 11881 wherein the agent is a famexyl transferase inhibitor.
11919. The method of item 11881 wherein the agent is famesyltransferase inhibitor selected from the group of A-197574 (Abbott), a farnesyltransferase inhibitor from Servier, FPTIII (Strathclyde Institute for Drug R), LB-42908 (LG Life Sciences), Pharmaprojects No. 5063 (Genzyme), Pharmaprojects No. 5597 (Ipsen), Yissum Project No. B-1055 (Yissum), and an analogue or derivative thereof.
11920. The method of item 11881 wherein the agent is an FLT-3 kinase inhibitor.
11921. The method of item 11881 wherein the agent is an FGF receptor kinase inhibitor.
11922. The method of item 11881 wherein the agent is a fibrinogen antagonist selected from the group consisting of AUV-201 (Auvation), MG-13926 (Sanofi-Aventis), plasminogen activator (CAS No. 105913-11-9) (from Sanofi-Aventis or UCB), plasminogen activator-2 (tPA- 2) (Sanofi-Aventis), pro-urokinase (CAS No. 82657-92-9) (Sanofi-Aventis), and an analogue or derivative thereof.
11923. The method of item 11881 wherein the agent is a heat shock protein 90 antagonist selected from the group consisting of SRN- 005 (Sirenade), geldanamycin, NSC-33050 (17-allylaminogeldanamycin; 17- AAG), 17-dimethylaminoethylamino-17-demethoxy-geldanamycin (17- DMAG), rifabutin (rifamycin XIV, r,4-didehydro-1-deoxy-1 ,4-dihydro-5'-(2- methylpropyl)-1-oxo-), and an analogue or derivative thereof.
11924. The method of item 11881 wherein the agent is a histone deacetylase inhibitor.
11925. The method of item 11881 wherein the agent is an HMGCoA reductase inhibitor selected from the group consisting of an atherosclerosis therapeutic from Lipid Sciences, ATI-16000 (ARYx Therapeutics), KS-01-019 (Kos Pharmaceuticals), Pharmaprojects No. 2197 (Sanofi-Aventi), RP 61969 (Sanofi-Aventis), and an analogue or derivative thereof.
11926. The method of item 11881 wherein the agent is an ICAM inhibitor.
11927. The method of item 11881 wherein the agent is an IL, ICE and IRAK antagonist, wherein the antagonist is a CJ-14877, CP- 424174 (Pfizer), NF-61 (Negma-Lerads), and an analogue or derivative thereof.
11928. The method of item 11881 wherein the agent is an IL-2 inhibitor.
11929. The method of item 11881 wherein the agent is an immunosuppressant selected from the group consisting of teriflunomide (Sanofi Aventis), chlorsulfaquinoxalone (NSC-339004), chlorsulfaquinoxalone sulfate, CS-712 (Sankyo), ismomultin alfa (CAS No. 457913-93-8) (Akzo Nobel), antiallergics from GenPat77, antiinflammatories or AT-005 (Androclus Therapeutics), autoimmune disease therapeutics from EpiVax, BN-007 (Bone), budesonide (CAS No. 51333-22- 3) (MAP Pharmaceuticals), CO-14 (Genzyme), edratide (CAS No. 433922- 67-9) (Teva), EP-314 (Enanta), eprovafen (CAS No. 101335-99-3) (Sanofi- Aventis), HWA-131 (CAS No. 118788-41-3) (Sanofi-Aventis), immunomodulators from MerLion Pharmaceuticals, immunosuppressives from Alchemia, IPL-12 (Inflazyme), MDL-9563 (CAS No. 27086-86-8) (Sanofi-Aventis), Pharmaprojects No. 2330 (Sanofi-Aventis), Pharmaprojects No. 6426 (Abgenix), PXS-25 (Pharmaxis), rosmarinic acid (CAS No. 20283-92-5) (Sanofi-Aventis), RP 42927 or RP 54745 (CAS No. 135330-08-4) (Sanofi-Aventis), SGN-35 (Seattle Genetics), ST-1959 (Sigma-Tau), type I diabetes therapy from SYNX Pharma, UNIL-88 (Debiopharm), VP-025 (Vasogen), VR-694 (Vectura), PRTX-001 (Protalex), and an analogue or derivative thereof.
11930. The method of item 11881 wherein the agent is an IMPDH (inosine monophosphate).
11931. The method of item 11881 wherein the agent is an integrin antagonist.
11932. The method of item 11881 wherein the agent is an interleukin antagonist.
11933. The method of item 11881 wherein the agent is an inhibitor of type III receptor tyrosine kinase.
11934. The method of item 11881 wherein the agent is an irreversible inhibitor of enzyme methionine aminopeptidase type 2.
11935. The method of item 11881 wherein the agent is an isozyme selective delta protein kinase C inhibitor.
11936. The method of item 11881 wherein the agent a JAK3 enzyme inhibitor.
11937. The method of item 11881 wherein the agent is a JNK inhibitor.
11938. The method of item 11881 wherein the agent is a kinase inhibitor. 11939. The method of item 11881 wherein the agent is kinesin antagonist.
11940. The method of item 11881 wherein the agent is a kinesin antagonist.
11941. The method of item 11881 wherein the agent is a leukotriene inhibitor and antagonist selected from the group consisting of ambicromii (CAS No. 58805-38-2) (Sanofi-Aventis), amelubant (CAS No. 346735-24-8) (Boehringer Ingelheim), DW-1141 (Dong Wha), ebselen (Daiichi Pharmaceutical), ibudilast (Kyorin), leucotriene inhibitors from Sanofi-Aventis, lymphotoxin-beta receptor (LT-D) from Biogen Idee, Pharmaprojects No. 1535 and 2728 (CAS No. 119340-33-9) (Sanofi- Aventis), R-112 (Rigel), Rev-5367 (CAS No. 92532-05-3) (Sanofi-Aventis), RG-14893 (CAS No. 141835-49-6) (Sanofi-Aventis), RG-5901-A (CAS No. 101910-24-1), 92532-23-5, RP 66153 (CAS No. 142422-79-5), RP 66364 (CAS No.186916-92-5), RP 69698 (CAS No. 141748-00-7) (Sanofi-Aventis), SC-411930 (Pfizer), SC-41930 (CAS No. 120072-59-5) (Pfizer), SC-50605 (CAS No. 138828-39-4) (Pfizer), SC-51146 (CAS No. 141059-52-1), SC- 53228 (CAS No. 153633-01-3) (Pfizer), spaglumic acid (ZY-15106) (CAS No.3106-85-2), 80619-64-3 (Novartis), tipredane (CAS No. 85197-77-9) (Bristol-Myers Squibb), U-75302 (CAS No. 119477-85-9) (Pfizer), and analogue or derivative thereof.
11942. The method of item 11881 wherein the agent is an MAP kinase inhibitor.
11943. The method of item 11881 wherein the agent is a matrix metalloproteinase inhibitor. 11944. The method of item 11881 wherein the agent is an MCP-CCR2 inhibitor.
11945. The method of item 11881 wherein the agent is an mTOR inhibitor.
11946. The method of item 11881 wherein the agent is an mTOR kinase inhibitor.
11947. The method of item 11881 wherein the agent is a microtubule inhibitor selected from the group consisting of antibody- maytansinoid conjugates from Biogen Idee, colchicines (MantiCore Pharmaceuticals), anticancer immunoconjugates from Johnson & Johnson, DIME from Octamer, gni-1f (GNI)1 huC242-DM4, huMy9-6-DM1 (ImmunoGen), IDN-5404 (Indena), IMO-098, IMOderm (Imotep), mebendazole (Introgen Therapeutics), microtubule poisons from Cambridge Enterprise, paclitaxel such as LOTAX from Aphios (CAS No. 33069-62-4), Genexol-PM from Samyang, Pharmaprojects No. 6383 (Tapestry Pharmaceuticals), RPR-112378 (Sanofi-Aventis), SGN-75 (Seattle Genetics), SPL-7435 (Starpharma), SSR-250411 (Sanofi-Aventis), trastuzumab-DM1 (Genentech), vinorelbine, and an analogue or derivative thereof.
11948. The method of item 11881 wherein the agent is an MIF inhibitor.
11949. The method of item 11881 wherein the agent is an MMP inhibitor.
11950. The method of item 11881 wherein the agent is a neurokinin (NK) antagonist selected from the group consisting of anthrotainin (CAS No. 148084-40-6) (Sanofi-Aventis), an IBS thereapeutic from ArQuIe, MDL-105212A (CAS No. 167261-60-1) (Ssanofi-Aventis), Pharmaprojects No. 2744, 3258 (CAS No. 139167-47-8) 4006, 4201 , or 5986 (Sanofi-Aventis), RP 67580 (CAS No. 135911-02-3), SR-144190 (CAS No. 201152-86-5), SSR-240600, SSR-241586 (Sanofi-Aventis), TKA-457 (Novartis), vestipitant mesylate (CAS No. 334476-64-1) (GlaxoSmithKline), Win-64821 (Sanofi-Aventis), PRX-96026 (Predix Pharmaceuticals), and an analogue or derivative thereof.
11951. The method of item 11881 wherein the agent is an NF kappa B inhibitor selected from the group consisting of AVE-0545 or AVE-0547 (Sanofi-Aventis), bortezomib (CAS No. 179324-69-7) (Millennium Pharmaceuticals), dexanabinol (CAS No. 112924-45-5) (Pharmos), dexlipotam (Viatris), Pharmaprojects No. 6283 (INDRA) (OXiGENE), IPL- 576092 (CAS No. 137571-30-3) (Inflazyme), NFKB decoy (Corgentech), NFKB decoy oligo (AnGes MG), NFKB's from Ariad, osteoporosis treatments or S5 (F005) from Fulcrum Pharmaceuticals, P61 (Phytopharm), R-flurbiprofen (CAS No. 5104-49-4) (Encore Pharmaceuticals), and an analogue or derivative thereof.
11952. The method of item 11881 wherein the agent is a nitric oxide agonist.
11953. The method of item 11881 wherein the agent is an ornithine decarboxylase inhibitor.
11954. The method of item 11881 wherein the agent is a p38 MAP kinase inhibitor selected from the group consisting of AZD-6703 (AstraZeneca), JX-401 (Jexys Pharmaceuticals), BMS-2 (Bristol-Myers Squibb), a p38 MAP kinase inhibitor from Novartis, a p38-alpha MAP kinase inhibitor from Amphora, Pharmaprojects No. 5704 (Pharmacopeia), RPR- 200765A (Sanofi-Aventis), SD-282 (Johnson & Johnson), TAK-715 (Takeda), and an analogue or derivative thereof.
11955. The method of item 11881 wherein the agent is a palmitoyl-protein thioesterase inhibitor.
11956. The method of item 11881 wherein the agent is a PDGF receptor kinase inhibitor selected from the group consisting of AAL- 993, AMN-107, or ABP-309 (Novartis), AMG-706 (Amgen), BAY-57-9352 (Bayer), CDP-860 (UCB), E-7080 (Eisai), imatinib (CAS No. 152459-95-5) (Novartis), OSI-930 (OSI Pharmaceuticals), RPR-127963E (Sanofi-Aventis), RWJ-540973 (Johnson & Johnson), sorafenib tosylate (Bayer), SU-11657 (Pfizer), tandutinib (CAS No. 387867-13-2) (Millennium Pharmaceuticals), vatalanib (Novartis), ZK-CDK (Schering AG), and an analogue or derivative thereof.
11957. The method of item 11881 wherein the agent is (-)-halofenate (Metabolex), AMG-131 (Amgen), antidiabetics from Japan Tobacco, AZD-4619, AZD-8450, AZD-8677 (AstraZeneca), DRF-10945, balaglitazone (Dr Reddy's), CS-00088, CS-00098 (Chipscreen Biosciences), E-3030 (Eisai), etalocib (CAS No. 161172-51-6) (EIi Lilly), GSK-641597 (Ligand), GSK-677954 (GlaxoSmithKline), GW-409544 (Ligand), GW- 590735 (GlaxoSmithKline), K-111 (Hoffmann-La Roche), LY-518674 (EIi Lilly), LY-674 (Ligand), LY-929 (Ligand), MC-3001 , MC-3002 (MaxoCore Pharmaceuticals), metformin HCI + pioglitazone (CAS No. 1115-70-4 and
112529-15-4), ACTOPLUS MET from Andrx), muraglitazar (CAS No. 331741-94-7) (Bristol-Myers Squibb), naveglitazar (Ligand), oleoylethanolamide (Kadmus Pharmaceuticals), ONO-5129, pioglitazone hydrochloride (CAS No. 111025-46-8 and 112529-15-4) (Takeda), PLX-204 (Plexxikon), PPAR agonists from Genfit, PPAR delta agonists from EIi Lilly, PPAR-alpha agonists from CrystalGenomics, PPAR-gamma modulators and PPAR-β modulators from CareX, rosiglitazone maleate (CAS No. 122324- 73-4 or 155141-29-0) (GlaxoSmithKline), rosiglitazone maleate/glimepir (CAS No. 155141-29-0 and 93479-97-1), AVANDARYL, rosiglitazone maleate/metformin extend (CAS No. 155141-29-0 and 657-24-9), AVANDAMET, rosiglitazone maleate+metformin, AVANDAMET (GlaxoSmithKline), tesaglitazar (AstraZeneca), and an analogue or derivative thereof.
11958. The method of item 11881 wherein the agent is a phosphatase inhibitor.
11959. The method of item 11881 wherein the agent is a phosphodiesterase (PDE) inhibitor selected from the group consisting of avanafil (Tanabe Seiyaku), dasantafil (CAS No. 569351-91-3) (Schering- Plough), A-906119 (CAS No. 134072-58-5), DL-850 (Sanofi-Aventis), GRC- 3015, GRC-3566, GRC-3886 (Glenmark), HWA-153 (CAS No. 56395-66-5) (Sanofi-Aventis), hydroxypumafentrine (Altana), IBFB-130011, IBFB-14-016, IBFB-140301 , IBFB-150007, IBFB-211913 (IBFB Pharma), L-826141 (Merck & Co), medorinone (CAS No. 88296-61-1) (Sanofi-Aventis), MEM- 1917 (Memory Pharmaceuticals), ND-1251 (Neuro3d), PDE inhibitors from ICOS, PDE IV inhibitors from Memory Pharmaceuticals and CrystalGenomics, Pharmaprojects No. 2742 and 6141 (Sanofi-Aventis), QAD-171 (Novartis), RHC-2963 (CAS No. 76993-12-9 and 76993-14-1), RPR-117658, RPR-122818 derivatives, SR-24870 , and RPR-132294 (Sanofi-Aventis), SK-350 (ln2Gen), stroke therapy agents from deCODE Genetics, TAS-203 (Taiho), tofimilast (CAS No. 185954-27-2) (Pfizer), UK- 371800 (Pfizer), WIN-65579 (CAS No. 158020-82-7) (Sanofi-Aventis), IBFB- 130020 (IBFB Pharma), OPC-6535 (CAS No. 145739-56-6) (Otsuka), a phosphodiesterase III inhibitor, enoximone, a phosphodiesterase IV inhibitor, fosfosal, Atopik (Barrier Therapeutics), triflusal, a phosphodiesterase V inhibitor, and an analogue or derivative thereof. 11960. The method of item 11881 wherein the agent is a PKC inhibitor.
11961. The method of item 11881 wherein the agent is a platelet activating factor antagonist.
11962. The method of item 11881 wherein the agent is a platelet-derived growth factor receptor kinase inhibitor.
11963. The method of item 11881 wherein the agent is a prolyl hydroxylase inhibitor.
11964. The method of item 11881 wherein the agent is a polymorphonuclear neutrophil inhibitor.
11965. The method of item 11881 wherein the agent is a protein kinase B inhibitor.
11966. The method of item 11881 wherein the agent is a protein kinase C stimulant.
11967. The method of item 11881 wherein the agent is a purine nucleoside analogue.
11968. The method of item 11881 wherein the agent is a purinoreceptor P2X antagonist.
11969. The method of item 11881 wherein the agent is a Raf kinase inhibitor. 11970. The method of item 11881 wherein the agent is a reversible inhibitor of ErbB1 and ErbB2.
11971. The method of item 11881 wherein the agent is a ribonucleoside triphosphate reductase inhibitor.
11972. The method of item 11881 wherein the agent is an SDF-1 antagonist.
11973. The method of item 11881 wherein the agent is a sheddase inhibitor.
11974. The method of item 11881 wherein the agent is an SRC inhibitor.
11975. The method of item 11881 wherein the agent is a stromelysin inhibitor.
11976. The method of item 11881 wherein the agent is an Syk kinase inhibitor.
11977. The method of item 11881 wherein the agent is a telomerase inhibitor.
11978. The method of item 11881 wherein the agent is a TGF beta inhibitor selected from the group consisting of pirfenidone (CAS No. 53179-13-8) (MARNAC), tranilast (CAS No. 53902-12-8) (Kissei), IN- 1130 (ln2Gen), mannose-6-phosphate (BTG), TGF-β antagonists from Inflazyme (Pharmaprojects No. 6075), TGF-β antagonists from Sydney, non-industrial source), TGF-β I receptor kinase inhibitors from EIi Lilly, TGF- β receptor inhibitors from Johnson & Johnson, and an analogue or derivative thereof.
11979. The method of item 11881 wherein the agent is a
TNFα antagonist or TACE inhibitor selected from the group consisting of adalimumab (CAS No. 331731-18-1) (Cambridge Antibody Technology), AGIX-4207 (AtheroGenics), AGT-1 (Advanced Biotherapy), an antiinflammatory from Borean Pharma, Cellzome, or Paradigm Therapeutics, anti-inflammatory vaccine (TNF-alpha kinoid) from Neovacs, humanized anti-TNF antibody or an anti-TNF MAb (CB0006) Celltech (UCB), apratastat (CAS No. 287445-51-0) (Wyeth), BMS-561392 (Bristol-Myers Squibb), BN- 006 (Bone), certolizumab pegol (CAS No. 428863-50-7 or CH-138 (UCB), cilomilast (CAS No. 153259-65-5) (GlaxoSmithKline), CR-1 (Nuada Pharmaceuticals), CRx-119 (CombinatoRx), D-5410 (UCB), dacopafant (CAS No. 125372-33-0) (Sanofi-Aventis), dersalazine (CAS No. 188913-57- 7/188913-58-8) (Uriach), etanercept (CAS No. 185243-69-0) (Amgen), ethyl pyruvate (Critical (Critical Therapeutics), golimumab (CAS No. 476181-74-5) (Johnson & Johnson), hormono-immunotherapy from Ipsen, CDP571 (e.g., Humicade from UCB), IC-485 (ICOS), infliximab (CAS No. 170277-31-3) (Johnson & Johnson), IP-751 (Manhattan Pharmaceuticals), ISIS-104838 (CAS No. 250755-32-9) (ISIS Pharmaceuticals), lenalidomide (CAS No. 191732-72-6) (Celgene), lentinan (CAS No. 37339-90-5) (Ajinomoto), MDL- 201112 (CAS No. 142130-73-2) (Sanofi-Aventis), medroxyprogesterone (CAS No. 520-85-4) (InKine Pharmaceutical), N-acetylcysteine (CAS No. 616-91-1) (Zambon), NBE-P2 (DIREVO Biotech), nerelimomab (CAS No. 162774-06-3) (Chiron), OM-294DP (OM PHARMA), onercept (CAS No. 199685-57-9) (Yeda), PASSTNF-alpha (Verigen), pentoxifylline or oxypentifylline (Sanofi-Aventis), Pharmaprojects No. 4091, 4241, 4295, or 4488 (Sanofi-Aventis), Pharmaprojects No. 5480 (Amgen), Pharmaprojects No. 6749 (Cengent), pirfenidone (CAS No. 53179-13-8) (MARNAC), RPR- 132294 (Sanofi-Aventis), S5 (F002) (Fulcrum Pharmaceuticals), simvastatin (CAS No. 79902-63-9) (Merck & Co), STA-6292 (Synta Pharmaceuticals), tacrolimus (CAS No. 104987-11-3) (Fujisawa LifeCycle Pharma), talactoferrin alfa (CAS No. 308240-58-6) (Agennix), thalidomide (CAS No. 50-35-1) (Celgene), TNF antagonists form ProStrakan, and Synergen, TNF inhibitors (Amgen), TNF-alpha antagonists from Dynavax Technologies and Jerina AG (Germany), TNF-alpha inhibitors from IBFB Pharma and Xencor (Xencor), torbafylline (CAS No. 105102-21-4) (Sanofi-Aventis), UR-1505 (Uriach), VT-346 (Viron Therapeutics), YSIL6 (Y1S Therapeutics), YSTH2 (Y's Therapeutics), NPI-1302a-3 (Nereus Pharmaceuticals, a TNF antagonist from Jerina AG (Germany), dersalazine, and an analogue or derivative thereof.
11980. The method of item 11881 wherein the agent is a Toll receptor inhibitor.
11981. The method of item 11881 wherein the agent is a tubulin antagonist.
11982. The method of item 11881 wherein the agent is a tyrosine kinase inhibitor selected from the group consisting of SU-011248, SUTENT from Pfizer Inc. (New York, NY), BMS-354825, PN-355 (Paracelsian Pharmaceuticals), AGN-199659 (Allergan), AAL-993 or ABP- 309 (Novartis), adaphostin (NIH), AEE-788 (Novartis), AG-013736 (OSI Pharmaceuticals), AG-13736 (Pfizer), ALT-110 (Alteris Therapeutics), AMG- 706 (Amgen), anticancer MAbs from Xencor, anti-EGFrvlll MAbs from Abgenix, anti-HER2 MAb from Abiogen, AZD-2171 or AZD-9935 (AstraZeneca), BAY-57-9352 (Bayer), B1BF-1120 (Boehringer Ingelheim), CEP-5214 (Cephalon), CEP-7055 (Cephalon), cetuximab (ImClone Systems), CHIR-200131 and CHIR-258 (Chiron), CP-547632 (OSI Pharmaceuticals), CP-724714 (Pfizer), CT-301 (Creabilis Therapeutics), D- 69491 (Baxter International), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EGFR/ErbB2 inhibitors from Array BioPharma, erlotinib (CAS No. 183319- 69-9) (OSI Pharmaceuticals), EXEL-2880 (Exelixis), FK-778 (Sanofi- Aventis) , gefitinib (CAS No. 184475-35-2) (AstraZeneca), GW-2286 or GW- 654652 (GlaxoSmithKline), her2/neu antigen from AlphaVax, HER-2/neu inhibitor from Generex, Herzyme (Medipad) (Sirna Therapeutics), HKI-272 (Wyeth), HuMax-EGFr (Genmab), idronoxil (CAS No. 81267-65-4) (Novogen), IGF-1 inhibitors from Ontogen, IMC-11 F8 (ImCIone Systems), kahalalide F (CAS No. 149204-42-2) (PharmaMar), KDR inhibitor from LG Life Sciences, KDR inhibitors from Abbott Laboratories, KDR kinase inhibitors (UCB), Kdr kinase inhibitors from Merck & Co, KRN-633 and KRN- 951 (Kirin Brewery), KSB-102 (Xenova), lapatinib ditosylate (CAS No. 388082-78-8) (GlaxoSmithKline), matuzumab (Merck KGaA), MDX-214 (Medarex), ME-103 (Pharmexa), MED-A300 (Gerolymatos), MNAC-13 (Lay Line Genomics), nimotuzumab (Center of Molecular Immunology), NSC- 330507 or NSC-707545 (NIH), NV-50 (Novogen), OSI-930 (OSI Pharmaceuticals), panitumumab (Abgenix), pelitinib (CAS No. 287933-82-7) (Wyeth), pertuzumab (CAS No. 380610-27-5)' (Genentech), Pharmaprojects No. 3985 (Sanofi-Aventis), prostate cancer therapeutics from Sequenom (SQPC35, SQPC36, SQPC90), removab and remoxab (Trion Pharma), RG- 13022 (CAS No. 136831-48-6), RG-13291 (CAS No. 138989-50-1), or RG- 14620 (CAS No. 136831-49-7) (Sanofi-Aventis), RM-6427 (Romark), RNAi breast cancer therapy from Benitec, RP 53801 (CAS No. 125882-88-4) (Sanofi-Aventis), sorafenib tosylate (Bayer), SU-11657 (Pfizer), Tie-2 antagonists from Semaia (Hybrigenics), Tie-2 inhibitors from Ontogen, trastuzumab (CAS No. 180288-69-1) (Genentech), tyrosine kinase inhibitors from Sanofi-Aventis, U3-1287, U3-1565, U3-1784, U3-1800 (U3 Pharma), vatalanib (Novartis), VEGFR-2 kinase inhibitor from Bristol-Myers Squibb, XL-647 (Exelixis), ZD-6474 (AstraZeneca), ZK-CDK (Schering AG), an EGFR tyrosine kinase inhibitor, EKB-569 (Wyeth), and an analogue or derivative thereof. 11983. The method of item 11881 wherein the agent is a VEGF inhibitor.
11984. The method of item 11881 wherein the agent is a vitamin D receptor agonist.
11985. The method of item 11881 wherein the agent is ZD-6474 (an angiogenesis inhibitor).
11986. The method of item 11881 wherein the agent is AP-23573 (an mTOR inhibitor).
11987. The method of item 11881 wherein the agent is synthadotin (a tubulin antagonist).
11988. The method of item 11881 wherein the agent is S-0885 (a collagenase inhibitor).
11989. The method of item 11881 wherein the agent is aplidine (an elongation factor-1 alpha inhibitor).
11990. The method of item 11881 wherein the agent is ixabepilone (an epithilone).
11991. The method of item 11881 wherein the agent is IDN-5390 (an angiogenesis inhibitor).
11992. The method of item 11881 wherein the agent is SB-2723005 (an angiogenesis inhibitor). 11993. The method of item 11881 wherein the agent is ABT-518 (an angiogenesis inhibitor).
11994. The method of item 11881 wherein the agent is combretastatin (an angiogenesis inhibitor).
11995. The method of item 11881 wherein the agent is anecortave acetate (an angiogenesis inhibitor).
11996. The method of item 11881 wherein the agent is SB-715992 (a kinesin antagonist).
11997. The method of item 11881 wherein the agent is temsirolimus (an mTOR inhibitor).
11998. The method of item 11881 wherein the agent is adalimumab (a TNFα antagonist).
11999. The method of item 11881 , wherein the composition comprises a polymer.
12000. The method of item 11881 , wherein the composition comprises a polymeric carrier.
12001. The method of item 11881 wherein the anti- scarring agent inhibits adhesion between the device and a host into which the device is implanted.
12002. The method of item 11881 wherein the device delivers the anti-scarring agent locally to tissue proximate to the device. 12003. The method of item 11881 wherein the device has a coating that comprises the anti-scarring agent.
12004. The method of item 11881 , wherein the device has a coating that comprises the agent and is disposed on a surface of the implant.
12005. The method of item 11881 , wherein the device has a coating that comprises the agent and directly contacts the implant.
12006. The method of item 11881 , wherein the device has a coating that comprises the agent and indirectly contacts the implant.
12007. The method of item 11881 , wherein the device has a coating that comprises the agent and partially covers the implant.
12008. The method of item 11881 , wherein the device has a coating that comprises the agent and completely covers the implant.
12009. The method of item 11881 , wherein the device has a uniform coating.
12010. The method of item 11881 , wherein the device has a non-uniform coating.
12011. The method of item 11881 , wherein the device has a discontinuous coating.
12012. The method of item 11881 , wherein the device has a patterned coating. 12013. The method of item 11881 , wherein the device has a coating with a thickness of 100 Dm or less.
12014. The method of item 11881 , wherein the device has a coating with a thickness of 10 Dm or less.
12015. The method of item 11881 , wherein the device has a coating, and the coating adheres to the surface of the implant upon deployment of the implant.
12016. The method of item 11881 , wherein the device has a coating, and wherein the coating is stable at room temperature for a period of 1 year.
12017. The method of item 11881 , wherein the device has a coating, and wherein the anti-scarring agent is present in the coating in an amount ranging between about 0.0001% to about 1% by weight.
12018. The method of item 11881 , wherein the device has a coating, and wherein the anti-scarring agent is present in the coating in an amount ranging between about 1% to about 10% by weight.
12019. The method of item 11881 , wherein the device has a coating, and wherein the anti-scarring agent is present in the coating in an amount ranging between about 10% to about 25% by weight.
12020. The method of item 11881 , wherein the device has a coating, and wherein the anti-scarring agent is present in the coating in an amount ranging between about 25% to about 70% by weight. 12021. The method of item 11881 , wherein the device has a coating, and wherein the coating further comprises a polymer.
12022. The method of item 11881 , wherein the device has a first coating having a first composition and a second coating having a second composition.
12023. The method of item 11881 , wherein the device has a first coating having a first composition and a second coating having a second composition, wherein the first composition and the second composition are different.
12024. The method of item 11881 , wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a copolymer.
12025. The method of item 11881 , wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a block copolymer.
12026. The method of item 11881, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a random copolymer.
12027. The method of item 11881 , wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a biodegradable polymer.
12028. The method of item 11881 , wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a non-biodegradable polymer. 12029. The method of item 11881 , wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a hydrophilic polymer.
12030. The method of item 11881 , wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a hydrophobic polymer.
12031. The method of item 11881 , wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a polymer having hydrophilic domains.
12032. The method of item 11881 , wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a polymer having hydrophobic domains.
12033. The method of item 11881 , wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a non-conductive polymer.
12034. The method of item 11881 , wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises an elastomer.
12035. The method of item 11881 , wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a hydrogel.
12036. The method of item 11881 , wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a silicone polymer. 12037. The method of item 11881 , wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a hydrocarbon polymer.
12038. The method of item 11881 , wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a styrene-derived polymer.
12039. The method of item 11881 , wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a butadiene polymer.
12040. The method of item 11881 , wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a macromer.
12041. The method of item 11881 , wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a poly(ethylene glycol) polymer.
12042. The method of item 11881 wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises an amorphous polymer.
12043. The method of item 11881 , wherein the device comprises a lubricious coating.
12044. The method of item 11881 wherein the anti- scarring agent is located within pores or holes of the device. 12045. The method of item 11881 wherein the anti- scarring agent is located within a channel, lumen, or divet of the device.
12046. The method of item 11881 , wherein the device comprises a second pharmaceutically active agent.
12047. The method of item 11881 wherein the device comprises an anti-inflammatory agent.
12048. The method of item 11881 wherein the device comprises an agent that inhibits infection.
12049. The method of item 11881 wherein the device comprises an agent that inhibits infection, and wherein the agent is an anthracycline.
12050. The method of item 11881 wherein the device comprises an agent that inhibits infection, and wherein the agent is doxorubicin.
12051. The method of item 11881 wherein the device comprises an agent that inhibits infection, and wherein the agent is mitoxantrone.
12052. The method of item 11881 wherein the device comprises an agent that inhibits infection, and wherein the agent is a fluoropyrimidine.
12053. The method of item 11881 wherein the device comprises an agent that inhibits infection, and wherein the agent is 5- fluorouracil (5-FU). 12054. The method of item 11881 wherein the device comprises an agent that inhibits infection, and wherein the agent is a folic acid antagonist.
12055. The method of item 11881 wherein the device comprises an agent that inhibits infection, and wherein the agent is methotrexate.
12056. The method of item 11881 wherein the device comprises an agent that inhibits infection, and wherein the agent is a podophylotoxin.
12057. The method of item 11881 wherein the device comprises an agent that inhibits infection, and wherein the agent is etoposide.
12058. The method of item 11881 wherein the device comprises an agent that inhibits infection, and wherein the agent is a camptothecin.
12059. The method of item 11881 wherein the device comprises an agent that inhibits infection, and wherein the agent is a hydroxyurea.
12060. The method of item 11881 wherein the device comprises an agent that inhibits infection, and wherein the agent is a platinum complex.
12061. The method of item 11881 wherein the device comprises an agent that inhibits infection, and wherein the agent is cisplatin. 12062. The method of item 11881 , further comprising an anti-thrombotic agent.
12063. The method of item 11881 wherein the device comprises a visualization agent.
12064. The method of item 11881 wherein the device comprises a visualization agent, wherein the visualization agent is a radiopaque material, and wherein the radiopaque material comprises a metal, a halogenated compound, or a barium containing compound.
12065. The method of item 11881 wherein the device comprises a visualization agent, wherein the visualization agent is a radiopaque material, and wherein the radiopaque material comprises barium, tantalum, or technetium.
12066. The method of item 11881 wherein the device comprises a visualization agent, and wherein the visualization agent is a MRI responsive material.
12067. The method of item 11881 wherein the device comprises a visualization agent, and wherein the visualization agent comprises a gadolinium chelate.
12068. The method of item 11881 wherein the device comprises a visualization agent, and wherein the visualization agent comprises iron, magnesium, manganese, copper, or chromium.
12069. The method of item 11881 wherein the device comprises a visualization agent, and wherein the visualization agent comprises an iron oxide compound. 12070. The method of item 11881 wherein the device comprises a visualization agent, and wherein the visualization agent comprises a dye, pigment, or colorant.
12071. The method of item 11881 wherein the device comprises an echogenic material.
12072. The method of item 11881 wherein the device comprises an echogenic material, and wherein the echogenic material is in the form of a coating.
12073. The method of item 11881 wherein the device is sterile.
12074. The method of item 11881 wherein the anti- scarring agent is released into tissue in the vicinity of the device after deployment of the device.
12075. The method of item 11881 wherein the anti- scarring agent is released into tissue in the vicinity of the device after deployment of the device, and wherein the tissue is connective tissue.
12076. The method of item 11881 wherein the anti- scarring agent is released into tissue in the vicinity of the device after deployment of the device, and wherein the tissue is muscle tissue.
12077. The method of item 11881 wherein the anti- scarring agent is released into tissue in the vicinity of the device after deployment of the device, and wherein the tissue is nerve tissue. 12078. The method of item 11881 wherein the anti- scarring agent is released into tissue in the vicinity of the device after deployment of the device, and wherein the tissue is epithelium tissue.
12079. The method of item 11881 wherein the anti- scarring agent is released in effective concentrations from the device over a period ranging from the time of deployment of the device to about 1 year.
12080. The method of item 11881 wherein the anti- scarring agent is released in effective concentrations from the device over a period ranging from about 1 month to 6 months.
12081. The method of item 11881 wherein the anti- scarring agent is released in effective concentrations from the device over a period ranging from about 1 - 90 days.
12082. The method of item 11881 wherein the anti- scarring agent is released in effective concentrations from the device at a constant rate.
12083. The method of item 11881 wherein the anti- scarring agent is released in effective concentrations from the device at an increasing rate.
12084. The method of item 11881 wherein the anti- scarring agent is released in effective concentrations from the device at a decreasing rate.
12085. The method of item 11881 wherein the anti- scarring agent is released in effective concentrations from the composition comprising the anti-scarring agent by diffusion over a period ranging from the time of deployment of the device to about 90 days.
12086. The method of item 11881 wherein the anti- scarring agent is released in effective concentrations from the composition comprising the anti-scarring agent by erosion of the composition over a period ranging from the time of deployment of the device to about 90 days.
12087. The method of item 11881 wherein the device comprises about 0.01 Dg to about 10 Dg of the anti-scarring agent.
12088. The method of item 11881 wherein the device comprises about 10 Dg to about 10 mg of the anti-scarring agent.
12089. The method of item 11881 wherein the device comprises about 10 mg to about 250 mg of the anti-scarring agent.
12090. The method of item 11881 wherein the device comprises about 250 mg to about 1000 mg of the anti-scarring agent.
12091. The method of item 11881 wherein the device comprises about 1000 mg to about 2500 mg of the anti-scarring agent.
12092. The method of item 11881 wherein a surface of the device comprises less than 0.01 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
12093. The method of item 11881 wherein a surface of the device comprises about 0.01 Dg to about 1 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied. 12094. The method of item 11881 wherein a surface of the device comprises about 1 Dg to about 10 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
12095. The method of item 11881 wherein a surface of the device comprises about 10 Dg to about 250 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
12096. The method of item 11881 wherein a surface of the device comprises about 250 Dg to about 1000 Dg of the anti-scarring agent of anti-scarring agent per mm2 of device surface to which the anti- scarring agent is applied.
12097. The method of item 11881 wherein a surface of the device comprises about 1000 Dg to about 2500 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied. _
12098. The method of item 11881 wherein the combining is performed by direct affixing the agent or the composition to the implant.
12099. The method of item 11881 wherein the combining is performed by spraying the agent or the component onto the implant.
12100. The method of item 11881 wherein the combining is performed by electrospraying the agent or the composition onto the implant. 12101. The method of item 11881 wherein the combining is performed by dipping the implant into a solution comprising the agent or the composition.
12102. The method of item 11881 wherein the combining is performed by covalently attaching the agent or the composition to the implant.
12103. The method of item 11881 wherein the combining is performed by non-covalently attaching the agent or the composition to the implant.
12104. The method of item 11881 wherein the combining is performed by coating the implant with a substance that contains the agent or the composition.
12105. The method of item 11881 wherein the combining is performed by coating the implant with a substance that absorbs the agent.
12106. The method of item 11881 wherein the combining is performed by interweaving a thread composed of, or coated with, the agent or the composition.
12107. The method of item 11881 wherein the combining is performed by covering all the implant with a sleeve that contains the agent or the composition.
12108. The method of item 11881 wherein the combining is performed by covering a portion of the implant with a sleeve that contains the agent or the composition. 12109. The method of item 11881 wherein the combining is performed by covering all the implant with a cover that contains the agent or the composition.
12110. The method of item 11881 wherein the combining is performed by covering a portion of the implant with a cover that contains the agent or the composition.
12111. The method of item 11881 wherein the combining is performed by covering all the implant with an electrospun fabric that contains the agent or the composition.
12112. The method of item 11881 wherein the combining is performed by covering a portion of the implant with an electrospun fabric that contains the agent or the composition.
12113. The method of item 11881 wherein the combining is performed by covering all the implant with a mesh that contains the agent or the composition.
12114. The method of item 11881 wherein the combining is performed by covering a portion of the implant with a mesh that contains the agent or the composition.
12115. The method of item 11881 wherein the combining is performed by constructing all the implant with the agent or the composition.
12116. The method of item 11881 wherein the combining is performed by constructing a portion of the implant with the agent or the composition. 12117. The method of item 11881 wherein the combining is performed by impregnating the implant with the agent or the composition.
12118. The method of item 11881 wherein the combining is performed by constructing all of the implant from a degradable polymer that releases the agent.
12119. The method of item 11881 wherein the combining is performed by constructing a portion of the implant from a degradable polymer that releases the agent.
12120. The method of item 11881 wherein the combining is performed by dipping the implant into a solution that comprise the agent and an inert solvent for the implant.
12121. The method of item 11881 wherein the combining is performed by dipping the implant into a solution that comprises the agent and a solvent that will swell the implant.
12122. The method of item 11881 wherein the combining is performed by dipping the implant into a solution that comprises the agent and a solvent that will dissolve the implant.
12123. The method of item 11881 wherein the combining is performed by dipping the implant into a solution that comprises the agent, a polymer and an inert solvent for the implant.
12124. The method of item 11881 wherein the combining is performed by dipping the implant into a solution that comprises the agent, a polymer and a solvent that will swell the implant. 12125. The method of item 11881 wherein the combining is performed by dipping the implant into a solution that comprises the agent, a polymer and a solvent that will dissolve the implant.
12126. The method of item 11881 wherein the combining is performed by spraying the implant into a solution that comprises the agent and an inert solvent for the implant.
12127. The method of item 11881 wherein the combining is performed by spraying the implant into a solution that comprises the agent and a solvent that will swell the implant.
12128. The method of item 11881 wherein the combining is performed by spraying the implant into a solution that comprises the agent and a solvent that will dissolve the implant.
12129. The method of item 11881 wherein the combining is performed by spraying the implant into a solution that comprises the agent, a polymer and an inert solvent for the implant.
12130. The method of item 11881 wherein the combining is performed by spraying the implant into a solution that comprises the agent, a polymer and a solvent that will swell the implant.
12131. The method of item 11881 wherein the combining is performed by spraying the implant into a solution that comprises the agent, a polymer and a solvent that will dissolve the implant.
12132. A method as in any one of items 11881-12131 , wherein the device is a tracheal stent. 12133. A method as in any one of items 11881-12131 , wherein the device is a bronchial stent.
12134. A method as in any one of items 11881-12131 , wherein the device is a metallic tracheal stent.
12135. A method as in any one of items 11881-12131 , wherein the device is a metallic bronchial stent.
12136. A method as in any one of items 11881-12131 , wherein the device is a polymeric tracheal stent.
12137. A method as in any one of items 11881-12131 , wherein the device is a polymeric bronchial stent.
12138. A method of making a medical device comprising: combining a genital urinary stent (e.g., an implant) and an anti- scarring agent or a composition comprising an anti-scarring agent, wherein the agent inhibits scarring between the device and a host into which the device is implanted.
12139. The method of item 12138 wherein the agent is an adensosine A2A receptor antagonist.
12140. The method of item 12138 wherein the agent is an AKT inhibitor.
12141. The method of item 12138 wherein the agent is an alpha 2 integrin antagonist, wherein the alpha 2 integrin antagonist is Pharmaprojects No. 5754 (Merck KgaA). 12142. The method of item 12138 wherein the agent is an alpha 4 integrin antagonist.
12143. The method of item 12138 wherein the agent is an alpha 7 nicotinic receptor agonist.
12144. The method of item 12138 wherein the agent is an angiogenesis inhibitor selected from the group consisting of AG-12,958 (Pfizer), ATN-161 (Attention LLC), neovastat, an angiogenesis inhibitor from Jerina AG (Germany), NM-3 (Mercian), VGA-1155 (Taisho), FCE-26644 (Pfizer), FCE-26950 (Pfizer), FPMA (Meiji Daries), FR-111142 (Fujisawa), GGTI-298, GM-1306 (Ligand), GPA-1734 (Novartis), NNC-47-0011 (Novo Nordisk), herbamycin (Nippon Kayaku), lenalidomide (Celegene), IP-10 (NIH), ABT-828 (Abbott), KIN-841 (Tokushima University, Japan), SF-1126 (Semafore Pharmaceuticals), laminin technology (NIH), CHIR-258 (Chiron), NVP-AEW541 (Novartis), NVP-AEW541 (Novartis), Vt16907 (Alchemia), OXI-8007 (Oxigene), EG-3306 (Ark Therapeutics), Maspin (Arriva), ABT- 567 (Abbott), PPI-2458 (Praecis Pharmaceuticals), CC-5079, CC-4089 (Celgene), HIF-1 alpha inhibitors (Xenova), S-247 (Pfizer), AP-23573 (Ariad), AZD-9935 (Astra Zeneca), mebendazole (Introgen Therapeutics), MetAP-2 inhibitors (GlaxoSmithKline), AG-615 (Angiogene Pharmaceuticals), Tie-2 antagonists (Hybrigenics), NC-381, CYC-381, NC-169, NC-219, NC-383, NC-384, NC-407 (Lorus Therapeutics), ATN-224 (Attenuon), ON-01370 (Onconova), Vitronectin antagonists (Amgen), SDX-103 (Salmedix), Vitronectin antagonists (Shire), CHP (Riemser), TEK (Amgen), Anecortave acetate (Alcon), T46.2 (Matrix Therapeutics), HG-2 (Heptagen), TEM antagonists (Genzyme), Oxi-4500 (Oxigene), ATN-161 (Attenuon), WX-293 (Wilex), M-2025 (Metris Therapeutics), Alphastatin (BioActa), YH-16 (Yantai Rongchang), BIBF-1120 (Boehringer Ingelheim), BAY-57-9352 (Bayer), AS- 1404 (Cancer Research Technology), SC-77964 (Pfizer), glycomimetics (BioTie Therapies), TIE-2 Inhibitors (Ontogen), DIMI, Octamer (Octamer), ABR-215050 (Active Biotech), ABT-518 (Abbott), KDR inhibitors (Abbott), BSF-466895 (Abbott), SCH-221153 (Schering-Plough), DAC:antiangiogenic (ConjuChem), TFPI (EntreMed), AZD-2171 (Astra-Zenaca), CDC-394 (Celgene), LY290293 (EIi Lilly), IDN-5390 (Indena), Kdr Kinase Inhibitors (Merck), CT-113020, CT-116433, CT-116563, CT-31890, CT-32228) (Cell Therapeutics), A-299620 (Abbott), TWEAK Inhibitor (Amgen), VEGF modulators (Johnson and Johnson), Tum-N53, tumstatin (Genzyme), Thios- 1 , Thios-2 (Thios Pharmaceuticals), MV-6401 (Miravant Medical Technologies), Spisulosine (PharmaMar), CEP-7055 (Cephalon), AUV-201 (Auvation), LM-609 (EIi Lilly), SKF-106615 (AnorMED), Oglufanide disodium (Cytran), BW-114 (Pharminox), Calreticulin (NIH), WX-678 (Wilex), SD-7784 (Pfizer), WX-UK1 (Wilex), SH-268 (Schering AG), 2-Me-PGA (Celgene), S- 137 (Pfizer), ZD-6126 (Angiogene Pharmaceuticals), SG-292 (SignalGen), Benefin (Lane Labs), A6, A36 (Angstrom), SB-2723005 (GlaxoSmithKline), SC-7 (Cell Therapeutics), ZEN-014 (AEterna Zentaris), 2-methoxyestradiol (EntreMed), NK-130119 (Nippon Kayaku), CC-10004 (Celgene), AVE- 8062A (Ajinomoto), Tacedinaline (Pfizer), Actinonin (Tokyo Metropolitan Institute of Medical Science), Lenalidomide (Celgene), VGA-1155, BTO-956 (SRI International), ER-68203-00 (Eisai), CT-6685 (UCB), JKC-362 (Phoenix Pharmaceuticals), DMI-3798 (DMI Biosciences, Angiomate (Ipsen), ZD-6474 (AstraZeneca), CEP-5214 (Cephalon), Canstatin (Genzyme), NM-3 (Mercian), Oridigm (MediQuest Therapeutics), Exherin (Adherex), BLS-0597 (Boston Life Sciences), PTC-299 (PTC Therapeutics), NPI-2358 (Nereus Pharmaceuticals), CGP-79787 (Novartis), AEE-788 (Novartis), CKD-732 (Chong Kun Dang), CP-564959 (OSI Pharmaceuticals), CM-101 (CarboMed), CT-2584, CT3501 (Cell Therapeutics), combretastatin and analogues and derivatives thereof (Oxigene), Rebimastat (Bristol-Meyers Squibb), Dextrin 2-sulfate (ML Laboratories), Cilengitide (Merk KGaA), NSC-706704 (Pharminox), KRN- 951 (Kirin Brewery), Ukrain, NSC-631570 (Nowicky Pharma), Tecogalan sodium (Daiichi Pharmaceutical), Tz-93 (Tsumura), TBC-1635 (Encysive Pharmaceuticals), TAN-1120 (Takeda), Semaxanib (Pfizer), BDI-7800 (Biopharmacopae), SD-186, SD-983 (Bristol-Meyers Squibb), SB-223245 (GlaxoSmithKline), SC-236 (Pfizer), RWJ-590973 (Johnson and Johnson), ILX-1850 (Genzyme), SC-68488, S-836 (Pfizer), CG-55069-11 (CuraGen), Ki-23057 (Kirin Brewery), CCX-700 (Chemoentryx), Pegaptanib octasodium (Giled Sciences), ANGIOCOL (available from Biostratum Inc.), or an analogue or derivative thereof.
12145. The method of item 12138 wherein the agent is an apoptosis antagonist.
12146. The method of item 12138 wherein the agent is an apoptosis activator.
12147. The method of item 12138 wherein the agent is a beta 1 integrin antagonist.
12148. The method of item 12138 wherein the agent is a beta tubulin inhibitor.
12149. The method of item 12138 wherein the agent is a blocker of enzyme production in Hepatitis C.
12150. The method of item 12138 wherein the agent is a Bruton's tyrosine kinase inhibitor.
12151. The method of item 12138 wherein the agent is a calcineurin inhibitor.
12152. The method of item 12138 wherein the agent is a caspase 3 inhibitor. 12153. The method of item 12138 wherein the agent is a CC chemokine receptor antagonist.
12154. The method of item 12138 wherein the agent is a cell cycle inhibitor selected from the group consisting of SNS-595 (Sunesis), synthadotin, KRX-0403, and an analogue or derivative thereof.
12155. The method of item 12138 wherein the agent is a cathepsin B inhibitor.
12156. The method of item 12138 wherein the agent is a cathepsin K inhibitor, wherein the cathepsin K inhibitor is 462795 (GlaxoSmithKline), INPL-022-D6 (Amura Therapeutics), or an analogue or derivative thereof.
12157. The method of item 12138 wherein the agent is a cathepsin L inhibitor.
12158. The method of item 12138 wherein the agent is a CD40 antagonist.
12159. The method of item 12138 wherein the agent is a chemokine receptor agonist.
12160. The method of item 12138 wherein the agent is a chymase inhibitor.
12161. The method of item 12138 wherein the agent is a collagenase antagonist. 12162. The method of item 12138 wherein the agent is a CXCR antagonist.
12163. The method of item 12138 wherein the agent is a cyclin dependent kinase inhibitor selected from the group consisting of a CDK-1 inhibitor, a CDK-2 inhibitor, a CDK- 4 inhibitor, a CDK-6 inhibitor, a CAK1 inhibitor from GPC Biotech and Bristol-Myers Squibb, RGB-286199 (GPC Biotech), an anticancer agent from Astex Technology, a CAK1 inhibitor from GPC Biotech, a CDK inhibitor from Sanofi-Aventis, a CDK1/CDK2 inhibitor from Amgen, a CDK2 inhibitor from SUGEN-2 (Pfizer), a hearing loss therapy agent (Sound Pharmaceuticals), PD-0332991 (Pfizer), RGB-286199 (GPC Biotech), Ro-0505124 (Hoffmann-La Roche), a Ser/Thr kinase inhibitor from Lilly (EIi Lilly), CVT-2584 (CAS No. 199986-75- 9) (CV Therapeutics), and an analogue or derivative thereof.
12164. The method of item 12138 wherein the agent is a cyclooxygenase 1 inhibitor.
12165. The method of item 12138 wherein the agent is a DHFR inhibitor.
12166. The method of item 12138 wherein the agent is a dual integrin inhibitor.
12167. The method of item 12138 wherein the agent is an elastase inhibitor.
12168. The method of item 12138 wherein the agent is an elongation factor-1 alpha inhibitor. 12169. The method of item 12138 wherein the agent is an endothelial growth factor antagonist.
12170. The method of item 12138 wherein the agent is an endothelial growth factor receptor kinase inhibitor selected from the group consisting of sorafenib tosylate (Bayer), AAL-993 (Novartis), ABP-309 (Novartis), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), E- 7080 (Eisai), EG-3306 (Ark Therapeutics), EXEL-2880 (Exelixis), GW- 654652 (GlaxoSmithKline), a KDR inhibitor from LG Life Sciences, CT-6685 and CT-6729 (UCB), KRN-633 and KRN-951 (Kirin Brewery), OSI-930 (OSI Pharmaceuticals), SP-5.2 (Supratek Pharma), SU-11657 (Pfizer), a Tie-2 antagonist (Hybrigenics), a VEGFR-2 kinase inhibitor (Bristol-Myers Squibb), XL-647 (Exelixis), a KDR inhibitor from Abbott Laboratories, sorafenib tosylate, and an analogue or derivative thereof.
12171. The method of item 12138 wherein the agent is an endotoxin antagonist.
12172. The method of item 12138 wherein the agent is an epothilone and tubulin binder.
12173. The method of item 12138 wherein the agent is an estrogen receptor antagonist.
12174. The method of item 12138 wherein the agent is an FGF inhibitor.
12175. The method of item 12138 wherein the agent is a farnexyl transferase inhibitor. 12176. The method of item 12138 wherein the agent is famesyltransferase inhibitor selected from the group of A-197574 (Abbott), a farnesyltransferase inhibitor from Servier, FPTIII (Strathclyde Institute for Drug R), LB-42908 (LG Life Sciences), Pharmaprojects No. 5063 (Genzyme), Pharmaprojects No. 5597 (Ipsen), Yissum Project No. B-1055 (Yissum), and an analogue or derivative thereof.
12177. The method of item 12138 wherein the agent is an FLT-3 kinase inhibitor.
12178. The method of item 12138 wherein the agent is an FGF receptor kinase inhibitor.
12179. The method of item 12138 wherein the agent is a fibrinogen antagonist selected from the group consisting of AUV-201 (Auvation), MG-13926 (Sanofi-Aventis), plasminogen activator (CAS No. 105913-11-9) (from Sanofi-Aventis or UCB), plasminogen activator-2 (tPA- 2) (Sanofi-Aventis), pro-urokinase (CAS No. 82657-92-9) (Sanofi-Aventis), and an analogue or derivative thereof.
12180. The method of item 12138 wherein the agent is a heat shock protein 90 antagonist selected from the group consisting of SRN- 005 (Sirenade), geldanamycin, NSC-33050 (17-allylaminogeldanamycin; 17- AAG), ^-dimethylaminoethylamino-^-demethoxy-geldanamycin (17- DMAG), rifabutin (rifamycin XIV, I'^-didehydro-i-deoxy-i ^-dihydro-δ1-^- methylpropyl)-1-oxo-), and an analogue or derivative thereof.
12181. The method of item 12138 wherein the agent is a histone deacetylase inhibitor. 12182. The method of item 12138 wherein the agent is an HMGCoA reductase inhibitor selected from the group consisting of an atherosclerosis therapeutic from Lipid Sciences, ATI-16000 (ARYx Therapeutics), KS-01-019 (Kos Pharmaceuticals), Pharmaprojects No. 2197 (Sanofi-Aventi), RP 61969 (Sanofi-Aventis), and an analogue or derivative thereof.
12183. The method of item 12138 wherein the agent is an ICAM inhibitor.
12184. The method of item 12138 wherein the agent is an IL, ICE and IRAK antagonist, wherein the antagonist is a CJ-14877, CP- 424174 (Pfizer), NF-61 (Negma-Lerads), and an analogue or derivative thereof.
12185. The method of item 12138 wherein the agent is an IL-2 inhibitor.
12186. The method of item 12138 wherein the agent is an immunosuppressant selected from the group consisting of teriflunomide (Sanofi Aventis), chlorsulfaquinoxalone (NSC-339004), chlorsulfaquinoxalone sulfate, CS-712 (Sankyo), ismomultin alfa (CAS No. 457913-93-8) (Akzo Nobel), antiallergics from GenPat77, antiinflammatories or AT-005 (Androclus Therapeutics), autoimmune disease therapeutics from EpiVax, BN-007 (Bone), budesonide (CAS No. 51333-22- 3) (MAP Pharmaceuticals), CO-14 (Genzyme), edratide (CAS No. 433922- 67-9) (Teva), EP-314 (Enanta), eprovafen (CAS No. 101335-99-3) (Sanofi- Aventis), HWA-131 (CAS No. 118788-41-3) (Sanofi-Aventis), immunomodulators from MerLion Pharmaceuticals, immunosuppressives from Alchemia, IPL-12 (Inflazyme), MDL-9563 (CAS No. 27086-86-8) (Sanofi-Aventis), Pharmaprojects No. 2330 (Sanofi-Aventis), Pharmaprojects No. 6426 (Abgenix), PXS-25 (Pharmaxis), rosmarinic acid (CAS No. 20283-92-5) (Sanofi-Aventis), RP 42927 or RP 54745 (CAS No. 135330-08-4) (Sanofi-Aventis), SGN-35 (Seattle Genetics), ST-1959 (Sigma-Tau), type I diabetes therapy from SYNX Pharma, UNIL-88 (Debiopharm), VP-025 (Vasogen), VR-694 (Vectura), PRTX-001 (Protalex), and an analogue or derivative thereof.
12187. The method of item 12138 wherein the agent is an IMPDH (inosine monophosphate).
12188. The method of item 12138 wherein the agent is an integrin antagonist.
12189. The method of item 12138 wherein the agent is an interleukin antagonist.
12190. The method of item 12138 wherein the agent is an inhibitor of type III receptor tyrosine kinase.
12191. The method of item 12138 wherein the agent is an irreversible inhibitor of enzyme methionine aminopeptidase type 2.
12192. The method of item 12138 wherein the agent is an isozyme selective delta protein kinase C inhibitor.
12193. The method of item 12138 wherein the agent a JAK3 enzyme inhibitor.
12194. The method of item 12138 wherein the agent is a JNK inhibitor. 12195. The method of item 12138 wherein the agent is a kinase inhibitor.
12196. The method of item 12138 wherein the agent is kinesin antagonist.
12197. The method of item 12138 wherein the agent is a kinesin antagonist.
12198. The method of item 12138 wherein the agent is a leukotriene inhibitor and antagonist selected from the group consisting of ambicromil (CAS No. 58805-38-2) (Sanofi-Aventis), amelubant (CAS No. 346735-24-8) (Boehringer Ingelheim), DW-1141 (Dong Wha), ebselen (Daiichi Pharmaceutical), ibudilast (Kyorin), leucotriene inhibitors from Sanofi-Aventis, lymphotoxin-beta receptor (LT-D) from Biogen Idee, Pharmaprojects No. 1535 and 2728 (CAS No. 119340-33-9) (Sanofi- Aventis), R-112 (Rigel), Rev-5367 (CAS No. 92532-05-3) (Sanofi-Aventis), RG-14893 (CAS No. 141835-49-6) (Sanofi-Aventis), RG-5901-A (CAS No. 101910-24-1), 92532-23-5, RP 66153 (CAS No. 142422-79-5), RP 66364 (CAS No.186916-92-5), RP 69698 (CAS No. 141748-00-7) (Sanofi-Aventis), SC-411930 (Pfizer), SC-41930 (CAS No. 120072-59-5) (Pfizer), SC-50605 (CAS No. 138828-39-4) (Pfizer), SC-51146 (CAS No. 141059-52-1), SC- 53228 (CAS No. 153633-01-3) (Pfizer), spaglumic acid (ZY-15106) (CAS No.3106-85-2), 80619-64-3 (Novartis), tipredane (CAS No. 85197-77-9) (Bristol-Myers Squibb), U-75302 (CAS No. 119477-85-9) (Pfizer), and analogue or derivative thereof.
12199. The method of item 12138 wherein the agent is an MAP kinase inhibitor. 12200. The method of item 12138 wherein the agent is a matrix metalloproteinase inhibitor.
12201. The method of item 12138 wherein the agent is an MCP-CCR2 inhibitor.
12202. The method of item 12138 wherein the agent is an mTOR inhibitor.
12203. The method of item 12138 wherein the agent is an mTOR kinase inhibitor.
12204. The method of item 12138 wherein the agent is a microtubule inhibitor selected from the group consisting of antibody- maytansinoid conjugates from Biogen Idee, colchicines (MantiCore Pharmaceuticals), anticancer immunoconjugates from Johnson & Johnson, DIME from Octamer, gni-1f (GNI)1 huC242-DM4, huMy9-6-DM1 (ImmunoGen), IDN-5404 (Indena), IMO-098, IMOderm (Imotep), mebendazole (Introgen Therapeutics), microtubule poisons from Cambridge Enterprise, paclitaxel such as LOTAX from Aphios (CAS No. 33069-62-4), Genexol-PM from Samyang, Pharmaprojects No. 6383 (Tapestry Pharmaceuticals), RPR-112378 (Sanofi-Aventis), SGN-75 (Seattle Genetics), SPL-7435 (Starpharma), SSR-250411 (Sanofi-Aventis), trastuzumab-DM1 (Genentech), vinorelbine, and an analogue or derivative thereof.
12205. The method of item 12138 wherein the agent is an MIF inhibitor.
12206. The method of item 12138 wherein the agent is an MMP inhibitor. 12207. The method of item 12138 wherein the agent is a neurokinin (NK) antagonist selected from the group consisting of anthrotainin (CAS No. 148084-40-6) (Sanofi-Aventis), an IBS thereapeutic from ArQuIe1 MDL-105212A (CAS No. 167261-60-1) (Ssanofi-Aventis), Pharmaprojects No. 2744, 3258 (CAS No. 139167-47-8) 4006, 4201, or 5986 (Sanofi-Aventis), RP 67580 (CAS No. 135911-02-3), SR-144190 (CAS No. 201152-86-5), SSR-240600, SSR-241586 (Sanofi-Aventis), TKA-457 (Novartis), vestipitant mesylate (CAS No. 334476-64-1) (GlaxoSmithKline), Win-64821 (Sanofi-Aventis), PRX-96026 (Predix Pharmaceuticals), and an analogue or derivative thereof.
12208. The method of item 12138 wherein the agent is an NF kappa B inhibitor selected from the group consisting of AVE-0545 or AVE-0547 (Sanofi-Aventis), bortezomib (CAS No. 179324-69-7) (Millennium Pharmaceuticals), dexanabinol (CAS No. 112924-45-5) (Pharmos), dexlipotam (Viatris), Pharmaprojects No. 6283 (INDRA) (OXiGENE), IPL- 576092 (CAS No. 137571-30-3) (Inflazyme), NFKB decoy (Corgentech), NFKB decoy oligo (AnGes MG), NFKB's from Ariad, osteoporosis treatments or S5 (F005) from Fulcrum Pharmaceuticals, P61 (Phytopharm), R-flurbiprofen (CAS No. 5104-49-4) (Encore Pharmaceuticals), and an analogue or derivative thereof.
12209. The method of item 12138 wherein the agent is a nitric oxide agonist.
12210. The method of item 12138 wherein the agent is an ornithine decarboxylase inhibitor.
12211. The method of item 12138 wherein the agent is a p38 MAP kinase inhibitor selected from the group consisting of AZD-6703 (AstraZeneca), JX-401 (Jexys Pharmaceuticals), BMS-2 (Bristol-Myers Squibb), a p38 MAP kinase inhibitor from Novartis, a p38-alpha MAP kinase inhibitor from Amphora, Pharmaprojects No. 5704 (Pharmacopeia), RPR- 200765A (Sanofi-Aventis), SD-282 (Johnson & Johnson), TAK-715 (Takeda), and an analogue or derivative thereof.
12212. The method of item 12138 wherein the agent is a palmitoyl-protein thioesterase inhibitor.
12213. The method of item 12138 wherein the agent is a PDGF receptor kinase inhibitor selected from the group consisting of AAL- 993, AMN-107, or ABP-309 (Novartis), AMG-706 (Amgen), BAY-57-9352 (Bayer), CDP-860 (UCB), E-7080 (Eisai), imatinib (CAS No. 152459-95-5) (Novartis), OSI-930 (OSI Pharmaceuticals), RPR-127963E (Sanofi-Aventis), RWJ-540973 (Johnson & Johnson), sorafenib tosylate (Bayer), SU-11657 (Pfizer), tandutinib (CAS No. 387867-13-2) (Millennium Pharmaceuticals), vatalanib (Novartis), ZK-CDK (Schering AG), and an analogue or derivative thereof.
12214. The method of item 12138 wherein the agent is (-)-halofenate (Metabolex), AMG-131 (Amgen), antidiabetics from Japan Tobacco, AZD-4619, AZD-8450, AZD-8677 (AstraZeneca), DRF-10945, balaglitazone (Dr Reddy's), CS-00088, CS-00098 (Chipscreen Biosciences), E-3030 (Eisai), etalocib (CAS No. 161172-51-6) (EIi Lilly), GSK-641597 (Ligand), GSK-677954 (GlaxoSmithKline), GW-409544 (Ligand), GW- 590735 (GlaxoSmithKline), K-111 (Hoffmann-La Roche), LY-518674 (EIi Lilly), LY-674 (Ligand), LY-929 (Ligand), MC-3001 , MC-3002 (MaxoCore Pharmaceuticals), metformin HCI + pioglitazone (CAS No. 1115-70-4 and
112529-15-4), ACTOPLUS MET from Andrx), muraglitazar (CAS No. 331741-94-7) (Bristol-Myers Squibb), naveglitazar (Ligand), oleoylethanolamide (Kadmus Pharmaceuticals), ONO-5129, pioglitazone hydrochloride (CAS No. 111025-46-8 and 112529-15-4) (Takeda), PLX-204 (Plexxikon), PPAR agonists from Genfit, PPAR delta agonists from EIi Lilly, PPAR-alpha agonists from CrystalGenomics, PPAR-gamma modulators and PPAR-β modulators from CareX, rosiglitazone maleate (CAS No. 122324- 73-4 or 155141-29-0) (GlaxoSmithKline), rosiglitazone maleate/glimepir (CAS No. 155141-29-0 and 93479-97-1), AVANDARYL, rosiglitazone maleate/metformin extend (CAS No. 155141-29-0 and 657-24-9), AVANDAMET, rosiglitazone maleate+metformin, AVANDAMET (GlaxoSmithKline), tesaglitazar (AstraZeneca), and an analogue or derivative thereof.
12215. The method of item 12138 wherein the agent is a phosphatase inhibitor.
12216. The method of item 12138 wherein the agent is a phosphodiesterase (PDE) inhibitor selected from the group consisting of avanafil (Tanabe Seiyaku), dasantafil (CAS No. 569351-91-3) (Schering- Plough), A-906119 (CAS No. 134072-58-5), DL-850 (Sanofi-Aventis), GRC- 3015, GRC-3566, GRC-3886 (Glenmark), HWA-153 (CAS No. 56395-66-5) (Sanofi-Aventis), hydroxypumafentrine (Altana), IBFB-130011, IBFB-14-016, IBFB-140301 , IBFB-150007, IBFB-211913 (IBFB Pharma), L-826141 (Merck & Co), medorinone (CAS No. 88296-61-1) (Sanofi-Aventis), MEM- 1917 (Memory Pharmaceuticals), ND-1251 (Neuro3d), PDE inhibitors from ICOS, PDE IV inhibitors from Memory Pharmaceuticals and CrystalGenomics, Pharmaprojects No. 2742 and 6141 (Sanofi-Aventis), QAD-171 (Novartis), RHC-2963 (CAS No. 76993-12-9 and 76993-14-1), RPR-117658, RPR-122818 derivatives, SR-24870 , and RPR-132294 (Sanofi-Aventis), SK-350 (ln2Gen), stroke therapy agents from deCODE Genetics, TAS-203 (Taiho), tofimilast (CAS No. 185954-27-2) (Pfizer), UK- 371800 (Pfizer), WIN-65579 (CAS No. 158020-82-7) (Sanofi-Aventis), IBFB- 130020 (IBFB Pharma), OPC-6535 (CAS No. 145739-56-6) (Otsuka), a phosphodiesterase III inhibitor, enoximone, a phosphodiesterase IV inhibitor, fosfosal, Atopik (Barrier Therapeutics), triflusal, a phosphodiesterase V inhibitor, and an analogue or derivative thereof.
12217. The method of item 12138 wherein the agent is a PKC inhibitor.
12218. The method of item 12138 wherein the agent is a platelet activating factor antagonist.
12219. The method of item 12138 wherein the agent is a platelet-derived growth factor receptor kinase inhibitor.
12220. The method of item 12138 wherein the agent is a prolyl hydroxylase inhibitor.
12221. The method of item 12138 wherein the agent is a polymorphonuclear neutrophil inhibitor.
12222. The method of item 12138 wherein the agent is a protein kinase B inhibitor.
12223. The method of item 12138 wherein the agent is a protein kinase C stimulant.
12224. The method of item 12138 wherein the agent is a purine nucleoside analogue.
12225. The method of item 12138 wherein the agent is a purinoreceptor P2X antagonist. 12226. The method of item 12138 wherein the agent is a Raf kinase inhibitor.
12227. The method of item 12138 wherein the agent is a reversible inhibitor of ErbB1 and ErbB2.
12228. The method of item 12138 wherein the agent is a ribonucleoside triphosphate reductase inhibitor.
12229. The method of item 12138 wherein the agent is an SDF-1 antagonist.
12230. The method of item 12138 wherein the agent is a sheddase inhibitor.
12231. The method of item 12138 wherein the agent is an SRC inhibitor.
12232. The method of item 12138 wherein the agent is a stromelysin inhibitor.
12233. The method of item 12138 wherein the agent is an Syk kinase inhibitor.
12234. The method of item 12138 wherein the agent is a telomerase inhibitor.
12235. The method of item 12138 wherein the agent is a TGF beta inhibitor selected from the group consisting of pirfenidone (CAS No. 53179-13-8) (MARNAC), tranilast (CAS No. 53902-12-8) (Kissei), IN- 1130 (ln2Gen), mannose-6-phosphate (BTG), TGF-β antagonists from Inflazyme (Pharmaprojects No. 6075), TGF-β antagonists from Sydney, non-industrial source), TGF-β I receptor kinase inhibitors from EIi Lilly, TGF- β receptor inhibitors from Johnson & Johnson, and an analogue or derivative thereof.
12236. The method of item 12138 wherein the agent is a
TNFα antagonist or TACE inhibitor selected from the group consisting of adalimumab (CAS No. 331731-18-1) (Cambridge Antibody Technology), AGIX-4207 (AtheroGenics), AGT-1 (Advanced Biotherapy), an antiinflammatory from Borean Pharma, Cellzome, or Paradigm Therapeutics, anti-inflammatory vaccine (TNF-alpha kinoid) from Neovacs, humanized anti-TNF antibody or an anti-TNF MAb (CB0006) Celltech (UCB), apratastat (CAS No. 287445-51-0) (Wyeth), BMS-561392 (Bristol-Myers Squibb), BN- 006 (Bone), certolizumab pegol (CAS No. 428863-50-7 or CH-138 (UCB), cilomilast (CAS No. 153259-65-5) (GlaxoSmithKline), CR-1 (Nuada Pharmaceuticals), CRx-119 (CombinatoRx), D-5410 (UCB), dacopafant (CAS No. 125372-33-0) (Sanofi-Aventis), dersalazine (CAS No. 188913-57- 7/188913-58-8) (Uriach), etanercept (CAS No. 185243-69-0) (Amgen), ethyl pyruvate (Critical (Critical Therapeutics), golimumab (CAS No. 476181-74-5) (Johnson & Johnson), hormono-immunotherapy from Ipsen, CDP571 (e.g., Humicade from UCB), IC-485 (ICOS), infliximab (CAS No. 170277-31-3) (Johnson & Johnson), IP-751 (Manhattan Pharmaceuticals), ISIS-104838 (CAS No. 250755-32-9) (ISIS Pharmaceuticals), ienalidomide (CAS No. 191732-72-6) (Celgene), lentinan (CAS No. 37339-90-5) (Ajinomoto), MDL- 201112 (CAS No. 142130-73-2) (Sanofi-Aventis), medroxyprogesterone (CAS No. 520-85-4) (InKine Pharmaceutical), N-acetylcysteine (CAS No. 616-91-1) (Zambon), NBE-P2 (DIREVO Biotech), nerelimomab (CAS No. 162774-06-3) (Chiron), OM-294DP (OM PHARMA), onercept (CAS No. 199685-57-9) (Yeda), PASSTNF-alpha (Verigen), pentoxifylline or oxypentifylline (Sanofi-Aventis), Pharmaprojects No. 4091 , 4241, 4295, or 4488 (Sanofi-Aventis), Pharmaprojects No. 5480 (Amgen), Pharmaprojects No. 6749 (Cengent), pirfenidone (CAS No. 53179-13-8) (MARNAC), RPR- 132294 (Sanofi-Aventis), S5 (F002) (Fulcrum Pharmaceuticals), simvastatin (CAS No. 79902-63-9) (Merck & Co), STA-6292 (Synta Pharmaceuticals), tacrolimus (CAS No. 104987-11-3) (Fujisawa LifeCycle Pharma), talactoferrin alfa (CAS No. 308240-58-6) (Agennix), thalidomide (CAS No. 50-35-1) (Celgene), TNF antagonists form ProStrakan, and Synergen, TNF inhibitors (Amgen), TNF-alpha antagonists from Dynavax Technologies and Jerina AG (Germany), TNF-alpha inhibitors from IBFB Pharma and Xencor (Xencor), torbafylline (CAS No. 105102-21-4) (Sanofi-Aventis), UR-1505 (Uriach), VT-346 (Viron Therapeutics), YSIL6 (Y's Therapeutics), YSTH2 (Y's Therapeutics), NPI-1302a-3 (Nereus Pharmaceuticals, a TNF antagonist from Jerina AG (Germany), dersalazine, and an analogue or derivative thereof.
12237. The method of item 12138 wherein the agent is a Toll receptor inhibitor.
12238. The method of item 12138 wherein the agent is a tubulin antagonist.
12239. The method of item 12138 wherein the agent is a tyrosine kinase inhibitor selected from the group consisting of SU-011248, SUTENT from Pfizer Inc. (New York, NY), BMS-354825, PN-355 (Paracelsian Pharmaceuticals), AGN-199659 (Allergan), AAL-993 or ABP- 309 (Novartis), adaphostin (NIH), AEE-788 (Novartis), AG-013736 (OSI Pharmaceuticals), AG-13736 (Pfizer), ALT-110 (Alteris Therapeutics), AMG- 706 (Amgen), anticancer MAbs from Xencor, anti-EGFrvlll MAbs from Abgenix, anti-HER2 MAb from Abiogen, AZD-2171 or AZD-9935 (AstraZeneca), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), CEP-5214 (Cephalon), CEP-7055 (Cephalon), cetuximab (ImClone Systems), CHIR-200131 and CHIR-258 (Chiron), CP-547632 (OSI Pharmaceuticals), CP-724714 (Pfizer), CT-301 (Creabilis Therapeutics), D- 69491 (Baxter International), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EGFR/ErbB2 inhibitors from Array BioPharma, erlotinib (CAS No. 183319- 69-9) (OSI Pharmaceuticals), EXEL-2880 (Exelixis), FK-778 (Sanofi- Aventis) , gefitinib (CAS No. 184475-35-2) (AstraZeneca), GW-2286 or GW- 654652 (GlaxoSmithKline), her2/neu antigen from AlphaVax, HER-2/neu inhibitor from Generex, Herzyme (Medipad) (Sirna Therapeutics), HKI-272 (Wyeth), HuMax-EGFr (Genmab), idronoxil (CAS No. 81267-65-4) (Novogen), IGF-1 inhibitors from Ontogen, IMC-11F8 (ImCIone Systems), kahalalide F (CAS No. 149204-42-2) (PharmaMar), KDR inhibitor from LG Life Sciences, KDR inhibitors from Abbott Laboratories, KDR kinase inhibitors (UCB), Kdr kinase inhibitors from Merck & Co, KRN-633 and KRN- 951 (Kirin Brewery), KSB-102 (Xenova), lapatinib ditosylate (CAS No. 388082-78-8) (GlaxoSmithKline), matuzumab (Merck KGaA), MDX-214 (Medarex), ME-103 (Pharmexa), MED-A300 (Gerolymatos), MNAC-13 (Lay Line Genomics), nimotuzumab (Center of Molecular Immunology), NSC- 330507 or NSC-707545 (NIH), NV-50 (Novogen), OSI-930 (OSI Pharmaceuticals), panitumumab (Abgenix), pelitinib (CAS No. 287933-82-7) (Wyeth), pertuzumab (CAS No. 380610-27-5) (Genentech), Pharmaprojects No. 3985 (Sanofi-Aventis), prostate cancer therapeutics from Sequenom (SQPC35, SQPC36, SQPC90), removab and remoxab (Trion Pharma), RG- 13022 (CAS No. 136831-48-6), RG-13291 (CAS No. 138989-50-1), or RG- 14620 (CAS No. 136831-49-7) (Sanofi-Aventis), RM-6427 (Romark), RNAi breast cancer therapy from Benitec, RP 53801 (CAS No. 125882-88-4) (Sanofi-Aventis), sorafenib tosylate (Bayer), SU-11657 (Pfizer), Tie-2 antagonists from Semaia (Hybrigenics), Tie-2 inhibitors from Ontogen, trastuzumab (CAS No. 180288-69-1) (Genentech), tyrosine kinase inhibitors from Sanofi-Aventis, U3-1287, U3-1565, U3-1784, U3-1800 (U3 Pharma), vatalanib (Novartis), VEGFR-2 kinase inhibitor from Bristol-Myers Squibb, XL-647 (Exelixis), ZD-6474 (AstraZeneca), ZK-CDK (Schering AG), an EGFR tyrosine kinase inhibitor, EKB-569 (Wyeth), and an analogue or derivative thereof.
12240. The method of item 12138 wherein the agent is a VEGF inhibitor.
12241. The method of item 12138 wherein the agent is a vitamin D receptor agonist.
12242. The method of item 12138 wherein the agent is ZD-6474 (an angiogenesis inhibitor).
12243. The method of item 12138 wherein the agent is AP-23573 (an mTOR inhibitor).
12244. The method of item 12138 wherein the agent is synthadotin (a tubulin antagonist).
12245. The method of item 12138 wherein the agent is S-0885 (a collagenase inhibitor).
12246. The method of item 12138 wherein the agent is aplidine (an elongation factor-1 alpha inhibitor).
12247. The method of item 12138 wherein the agent is ixabepilone (an epithilone).
12248. The method of item 12138 wherein the agent is IDN-5390 (an angiogenesis inhibitor). 12249. The method of item 12138 wherein the agent is SB-2723005 (an angiogenesis inhibitor).
12250. The method of item 12138 wherein the agent is ABT-518 (an angiogenesis inhibitor).
12251. The method of item 12138 wherein the agent is combretastatin (an angiogenesis inhibitor).
12252. The method of item 12138 wherein the agent is anecortave acetate (an angiogenesis inhibitor).
12253. The method of item 12138 wherein the agent is SB-715992 (a kinesin antagonist).
12254. The method of item 12138 wherein the agent is temsirolimus (an mTOR inhibitor).
12255. The method of item 12138 wherein the agent is adalimumab (a TNFα antagonist).
12256. The method of item 12138, wherein the composition comprises a polymer.
12257. The method of item 12138, wherein the composition comprises a polymeric carrier.
12258. The method of item 12138 wherein the anti- scarring agent inhibits adhesion between the device and a host into which the device is implanted. 12259. The method of item 12138 wherein the device delivers the anti-scarring agent locally to tissue proximate to the device.
12260. The method of item 12138 wherein the device has a coating that comprises the anti-scarring agent.
12261. The method of item 12138, wherein the device has a coating that comprises the agent and is disposed on a surface of the implant.
12262. The method of item 12138, wherein the device has a coating that comprises the agent and directly contacts the implant.
12263. The method of item 12138, wherein the device has a coating that comprises the agent and indirectly contacts the implant.
12264. The method of item 12138, wherein the device has a coating that comprises the agent and partially covers the implant.
12265. The method of item 12138, wherein the device has a coating that comprises the agent and completely covers the implant.
12266. The method of item 12138, wherein the device has a uniform coating.
12267. The method of item 12138, wherein the device has a non-uniform coating.
12268. The method of item 12138, wherein the device has a discontinuous coating. 12269. The method of item 12138, wherein the device has a patterned coating.
12270. The method of item 12138, wherein the device has a coating with a thickness of 100 Dm or less.
12271. The method of item 12138, wherein the device has a coating with a thickness of 10 Dm or less.
12272. The method of item 12138, wherein the device has a coating, and the coating adheres to the surface of the implant upon deployment of the implant.
12273. The method of item 12138, wherein the device has a coating, and wherein the coating is stable at room temperature for a period of 1 year.
12274. The method of item 12138, wherein the device has a coating, and wherein the anti-scarring agent is present in the coating in an amount ranging between about 0.0001% to about 1% by weight.
12275. The method of item 12138, wherein the device has a coating, and wherein the anti-scarring agent is present in the coating in an amount ranging between about 1% to about 10% by weight.
12276. The method of item 12138, wherein the device has a coating, and wherein the anti-scarring agent is present in the coating in an amount ranging between about 10% to about 25% by weight. 12277. The method of item 12138, wherein the device has a coating, and wherein the anti-scarring agent is present in the coating in an amount ranging between about 25% to about 70% by weight.
12278. The method of item 12138, wherein the device has a coating, and wherein the coating further comprises a polymer.
12279. The method of item 12138, wherein the device has a first coating having a first composition and a second coating having a second composition.
12280. The method of item 12138, wherein the device has a first coating having a first composition and a second coating having a second composition, wherein the first composition and the second composition are different.
12281. The method of item 12138, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a copolymer.
12282. The method of item 12138, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a block copolymer.
12283. The method of item 12138, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a random copolymer.
12284. The method of item 12138, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a biodegradable polymer. 12285. The method of item 12138, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a non-biodegradable polymer.
12286. The method of item 12138, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a hydrophilic polymer.
12287. The method of item 12138, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a hydrophobic polymer.
12288. The method of item 12138, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a polymer having hydrophilic domains.
12289. The method of item 12138, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a polymer having hydrophobic domains.
12290. The method of item 12138, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a non-conductive polymer.
12291. The method of item 12138, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises an elastomer.
12292. The method of item 12138, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a hydrogel. 12293. The method of item 12138, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a silicone polymer.
12294. The method of item 12138, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a hydrocarbon polymer.
12295. The method of item 12138, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a styrene-derived polymer.
12296. The method of item 12138, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a butadiene polymer.
12297. The method of item 12138, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a macromer.
12298. The method of item 12138, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a poly(ethylene glycol) polymer.
12299. The method of item 12138 wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises an amorphous polymer.
12300. The method of item 12138, wherein the device comprises a lubricious coating. 12301. The method of item 12138 wherein the anti- scarring agent is located within pores or holes of the device.
12302. The method of item 12138 wherein the anti- scarring agent is located within a channel, lumen, or divet of the device.
12303. The method of item 12138, wherein the device comprises a second pharmaceutically active agent.
12304. The method of item 12138 wherein the device comprises an anti-inflammatory agent.
12305. The method of item 12138 wherein the device comprises an agent that inhibits infection.
12306. The method of item 12138 wherein the_device comprises an agent that inhibits infection, and wherein the agent is an anthracycline.
12307. The method of item 12138 wherein the device comprises an agent that inhibits infection, and wherein the agent is doxorubicin.
12308. The method of item 12138 wherein the device comprises an agent that inhibits infection, and wherein the agent is mitoxantrone.
12309. The method of item 12138 wherein the device comprises an agent that inhibits infection, and wherein the agent is a fluoropyrimidine. 12310. The method of item 12138 wherein the device comprises an agent that inhibits infection, and wherein the agent is 5- fluorouracil (5-FU).
12311. The method of item 12138 wherein the device comprises an agent that inhibits infection, and wherein the agent is a folic acid antagonist.
12312. The method of item 12138 wherein the device comprises an agent that inhibits infection, and wherein the agent is methotrexate.
12313. The method of item 12138 wherein the device comprises an agent that inhibits infection, and wherein the agent is a podophylotoxin.
12314. The method of item 12138 wherein the device comprises an agent that inhibits infection, and wherein the agent is etoposide.
12315. The method of item 12138 wherein the device comprises an agent that inhibits infection, and wherein the agent is a camptothecin.
12316. The method of item 12138 wherein the device comprises an agent that inhibits infection, and wherein the agent is a hydroxyurea.
12317. The method of item 12138 wherein the device comprises an agent that inhibits infection, and wherein the agent is a platinum complex. 12318. The method of item 12138 wherein the device comprises an agent that inhibits infection, and wherein the agent is cisplatin.
12319. The method of item 12138, further comprising an anti-thrombotic agent.
12320. The method of item 12138 wherein the device comprises a visualization agent.
12321. The method of item 12138 wherein the device comprises a visualization agent, wherein the visualization agent is a radiopaque material, and wherein the radiopaque material comprises a metal, a halogenated compound, or a barium containing compound.
12322. The method of item 12138 wherein the device comprises a visualization agent, wherein the visualization agent is a radiopaque material, and wherein the radiopaque material comprises barium, tantalum, or technetium.
12323. The method of item 12138 wherein the device comprises a visualization agent, and wherein the visualization agent is a MRI responsive material.
12324. The method of item 12138 wherein the device comprises a visualization agent, and wherein the visualization agent comprises a gadolinium chelate.
12325. The method of item 12138 wherein the device comprises a visualization agent, and wherein the visualization agent comprises iron, magnesium, manganese, copper, or chromium. 12326. The method of item 12138 wherein the device comprises a visualization agent, and wherein the visualization agent comprises an iron oxide compound.
12327. The method of item 12138 wherein the device comprises a visualization agent, and wherein the visualization agent comprises a dye, pigment, or colorant.
12328. The method of item 12138 wherein the device comprises an echogenic material.
12329. The method of item 12138 wherein the device comprises an echogenic material, and wherein the echogenic material is in the form of a coating.
12330. The method of item 12138 wherein the device is sterile.
12331. The method of item 12138 wherein the anti- scarring agent is released into tissue in the vicinity of the device after deployment of the device.
12332. The method of item 12138 wherein the anti- scarring agent is released into tissue in the vicinity of the device after deployment of the device, and wherein the tissue is connective tissue.
12333. The method of item 12138 wherein the anti- scarring agent is released into tissue in the vicinity of the device after deployment of the device, and wherein the tissue is muscle tissue. 12334. The method of item 12138 wherein the anti- scarring agent is released into tissue in the vicinity of the device after deployment of the device, and wherein the tissue is nerve tissue.
12335. The method of item 12138 wherein the anti- scarring agent is released into tissue in the vicinity of the device after deployment of the device, and wherein the tissue is epithelium tissue.
12336. The method of item 12138 wherein the anti- scarring agent is released in effective concentrations from the device over a period ranging from the time of deployment of the device to about 1 year.
12337. The method of item 12138 wherein the anti- scarring agent is released in effective concentrations from the device over a period ranging from about 1 month to 6 months.
12338. The method of item 12138 wherein the anti- scarring agent is released in effective concentrations from the device over a period ranging from about 1 - 90 days.
12339. The method of item 12138 wherein the anti- scarring agent is released in effective concentrations from the device at a constant rate.
12340. The method of item 12138 wherein the anti- scarring agent is released in effective concentrations from the device at an increasing rate.
12341. The method of item 12138 wherein the anti- scarring agent is released in effective concentrations from the device at a decreasing rate. 12342. The method of item 12138 wherein the anti- scarring agent is released in effective concentrations from the composition comprising the anti-scarring agent by diffusion over a period ranging from the time of deployment of the device to about 90 days.
12343. The method of item 12138 wherein the anti- scarring agent is released in effective concentrations from the composition comprising the anti-scarring agent by erosion of the composition over a period ranging from the time of deployment of the device to about 90 days.
12344. The method of item 12138 wherein the device comprises about 0.01 Dg to about 10 Og of the anti-scarring agent.
12345. The method of item 12138 wherein the device comprises about 10 Dg to about 10 mg of the anti-scarring agent.
12346. The method of item 12138 wherein the device comprises about 10 mg to about 250 mg of the anti-scarring agent.
12347. The method of item 12138 wherein the device comprises about 250 mg to about 1000 mg of the anti-scarring agent.
12348. The method of item 12138 wherein the device comprises about 1000 mg to about 2500 mg of the anti-scarring agent.
12349. The method of item 12138 wherein a surface of the device comprises less than 0.01 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied. 12350. The method of item 12138 wherein a surface of the device comprises about 0.01 Dg to about 1 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
12351. The method of item 12138 wherein a surface of the device comprises about 1 Dg to about 10 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
12352. The method of item 12138 wherein a surface of the device comprises about 10 Dg to about 250 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
12353. The method of item 12138 wherein a surface of the device comprises about 250 Dg to about 1000 Dg of the anti-scarring agent of anti-scarring agent per mm2 of device surface to which the anti- scarring agent is applied.
12354. The method of item 12138 wherein a surface of the device comprises about 1000 Dg to about 2500 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
12355. The method of item 12138 wherein the combining is performed by direct affixing the agent or the composition to the implant.
12356. The method of item 12138 wherein the combining is performed by spraying the agent or the component onto the implant. 12357. The method of item 12138 wherein the combining is performed by electrospraying the agent or the composition onto the implant.
12358. The method of item 12138 wherein the combining is performed by dipping the implant into a solution comprising the agent or the composition.
12359. The method of item 12138 wherein the combining is performed by covalently attaching the agent or the composition to the implant.
12360. The method of item 12138 wherein the combining is performed by non-covalently attaching the agent or the composition to the implant.
12361. The method of item 12138 wherein the combining is performed by coating the implant with a substance that contains the agent or the composition.
12362. The method of item 12138 wherein the combining is performed by coating the implant with a substance that absorbs the agent.
12363. The method of item 12138 wherein the combining is performed by interweaving a thread composed of, or coated with, the agent or the composition.
12364. The method of item 12138 wherein the combining is performed by covering all the implant with a sleeve that contains the agent or the composition. 12365. The method of item 12138 wherein the combining is performed by covering a portion of the implant with a sleeve that contains the agent or the composition.
12366. The method of item 12138 wherein the combining is performed by covering all the implant with a cover that contains the agent or the composition.
12367. The method of item 12138 wherein the combining is performed by covering a portion of the implant with a cover that contains the agent or the composition.
12368. The method of item 12138 wherein the combining is performed by covering all the implant with an electrospun fabric that contains the agent or the composition.
12369. The method of item 12138 wherein the combining is performed by covering a portion of the implant with an electrospun fabric that contains the agent or the composition.
12370. The method of item 12138 wherein the combining is performed by covering all the implant with a mesh that contains the agent or the composition.
12371. The method of item 12138 wherein the combining is performed by covering a portion of the implant with a mesh that contains the agent or the composition.
12372. The method of item 12138 wherein the combining is performed by constructing all the implant with the agent or the composition. 12373. The method of item 12138 wherein the combining is performed by constructing a portion of the implant with the agent or the composition.
12374. The method of item 12138 wherein the combining is performed by impregnating the implant with the agent or the composition.
12375. The method of item 12138 wherein the combining is performed by constructing all of the implant from a degradable polymer that releases the agent.
12376. The method of item 12138 wherein the combining is performed by constructing a portion of the implant from a degradable polymer that releases the agent.
12377. The method of item 12138 wherein the combining is performed by dipping the implant into a solution that comprise the agent and an inert solvent for the implant.
12378. The method of item 12138 wherein the combining is performed by dipping the implant into a solution that comprises the agent and a solvent that will swell the implant.
12379. The method of item 12138 wherein the combining is performed by dipping the implant into a solution that comprises the agent and a solvent that will dissolve the implant.
12380. The method of item 12138 wherein the combining is performed by dipping the implant into a solution that comprises the agent, a polymer and an inert solvent for the implant. 12381. The method of item 12138 wherein the combining is performed by dipping the implant into a solution that comprises the agent, a polymer and a solvent that will swell the implant.
12382. The method of item 12138 wherein the combining is performed by dipping the implant into a solution that comprises the agent, a polymer and a solvent that will dissolve the implant.
12383. The method of item 12138 wherein the combining is performed by spraying the implant into a solution that comprises the agent and an inert solvent for the implant.
12384. The method of item 12138 wherein the combining is performed by spraying the implant into a solution that comprises the agent and a solvent that will swell the implant.
12385. The method of item 12138 wherein the combining is performed by spraying the implant into a solution that comprises the agent and a solvent that will dissolve the implant.
12386. The method of item 12138 wherein the combining is performed by spraying the implant into a solution that comprises the agent, a polymer and an inert solvent for the implant.
12387. The method of item 12138 wherein the combining is performed by spraying the implant into a solution that comprises the agent, a polymer and a solvent that will swell the implant.
12388. The method of item 12138 wherein the combining is performed by spraying the implant into a solution that comprises the agent, a polymer and a solvent that will dissolve the implant. 12389. A method as in any one of items 12138-12388, wherein the device is an ureteric stent.
12390. A method as in any one of items 12138-12388, wherein the device is an aurethral stent.
12391. A method as in any one of items 12138-12388, wherein the device is a fallopian tube stent.
12392. A method as in any one of items 12138-12388, wherein the device is a prostate stent.
12393. A method as in any one of items 12138-12388, wherein the device is a metallic genital urinary stent.
12394. A method as in any one of items 12138-12388, wherein the device is a polymeric genital urinary stent.
12395. A method of making a medical device comprising: combining an ear and nose stent (e.g., an implant) and an anti- scarring agent or a composition comprising an anti-scarring agent, wherein the agent inhibits scarring between the device and a host into which the device is implanted.
12396. The method of item 12395 wherein the agent is an adensosine A2A receptor antagonist.
12397. The method of item 12395 wherein the agent is an AKT inhibitor. 12398. The method of item 12395 wherein the agent is an alpha 2 integrin antagonist, wherein the alpha 2 integrin antagonist is Pharmaprojects No. 5754 (Merck KgaA).
12399. The method of item 12395 wherein the agent is an alpha 4 integrin antagonist.
12400. The method of item 12395 wherein the agent is an alpha 7 nicotinic receptor agonist.
12401. The method of item 12395 wherein the agent is an angiogenesis inhibitor selected from the group consisting of AG-12,958 (Pfizer), ATN-161 (Attention LLC), neovastat, an angiogenesis inhibitor from Jerina AG (Germany), NM-3 (Mercian), VGA-1155 (Taisho), FCE-26644 (Pfizer), FCE-26950 (Pfizer), FPMA (Meiji Daries), FR-111142 (Fujisawa), GGTI-298, GM-1306 (Ligand), GPA-1734 (Novartis), NNC-47-0011 (Novo Nordisk), herbamycin (Nippon Kayaku), lenalidomide (Celegene), IP-10 (NIH), ABT-828 (Abbott), KIN-841 (Tokushima University, Japan), SF-1126 (Semafore Pharmaceuticals), laminin technology (NIH), CHIR-258 (Chiron), NVP-AEW541 (Novartis), NVP-AEW541 (Novartis), Vt16907 (Alchemia), OXI-8007 (Oxigene), EG-3306 (Ark Therapeutics), Maspin (Arriva), ABT- 567 (Abbott), PPI-2458 (Praecis Pharmaceuticals), CC-5079, CC-4089 (Celgene), HIF-1 alpha inhibitors (Xenova), S-247 (Pfizer), AP-23573 (Ariad), AZD-9935 (Astra Zeneca), mebendazole (Introgen Therapeutics), MetAP-2 inhibitors (GlaxoSmithKline), AG-615 (Angiogene Pharmaceuticals), Tie-2 antagonists (Hybrigenics), NC-381 , CYC-381, NC-169, NC-219, NC-383, NC-384, NC-407 (Lorus Therapeutics), ATN-224 (Attenuon), ON-01370 (Onconova), Vitronectin antagonists (Amgen), SDX-103 (Salmedix), Vitronectin antagonists (Shire), CHP (Riemser), TEK (Amgen), Anecortave acetate (Alcon), T46.2 (Matrix Therapeutics), HG-2 (Heptagen), TEM antagonists (Genzyme), Oxi-4500 (Oxigene), ATN-161 (Attenuon), WX-293 (Wilex), M-2025 (Metris Therapeutics), Alphastatin (BioActa), YH-16 (Yantai Rongchang), BIBF-1120 (Boehringer Ingelheim), BAY-57-9352 (Bayer), AS- 1404 (Cancer Research Technology), SC-77964 (Pfizer), glycomimetics (BioTie Therapies), TIE-2 Inhibitors (Ontogen), DIMI, Octamer (Octamer), ABR-215050 (Active Biotech), ABT-518 (Abbott), KDR inhibitors (Abbott), BSF-466895 (Abbott), SCH-221153 (Schering-Plough), DAC.antiangiogenic (ConjuChem), TFPI (EntreMed), AZD-2171 (Astra-Zenaca), CDC-394 (Celgene), LY290293 (EIi Lilly), IDN-5390 (Indena), Kdr Kinase Inhibitors (Merck), CT-113020, CT-116433, CT-116563, CT-31890, CT-32228) (Cell Therapeutics), A-299620 (Abbott), TWEAK Inhibitor (Amgen), VEGF modulators (Johnson and Johnson), Tum-N53, tumstatin (Genzyme), Thios- 1 , Thios-2 (Thios Pharmaceuticals), MV-6401 (Miravant Medical Technologies), Spisulosine (PharmaMar), CEP-7055 (Cephalon), AUV-201 (Auvation), LM-609 (EIi Lilly), SKF-106615 (AnorMED), Oglufanide disodium (Cytran), BW-114 (Pharminox), Calreticulin (NIH), WX-678 (Wilex), SD-7784 (Pfizer), WX-UK1 (Wilex), SH-268 (Schering AG), 2-Me-PGA (Celgene), S- 137 (Pfizer), ZD-6126 (Angiogene Pharmaceuticals), SG-292 (SignalGen), Benefin (Lane Labs), A6, A36 (Angstrom), SB-2723005 (GlaxoSmithKline), SC-7 (Cell Therapeutics), ZEN-014 (AEterna Zentaris), 2-methoxyestradiol (EntreMed), NK-130119 (Nippon Kayaku), CC-10004 (Celgene), AVE- 8062A (Ajinomoto), Tacedinaline (Pfizer), Actinonin (Tokyo Metropolitan Institute of Medical Science), Lenalidomide (Celgene), VGA-1155, BTO-956 (SRl International), ER-68203-00 (Eisai), CT-6685 (UCB), JKC-362 (Phoenix Pharmaceuticals), DMI-3798 (DMI Biosciences, Angiomate (Ipsen), ZD-6474 (AstraZeneca), CEP-5214 (Cephalon), Canstatin (Genzyme), NM-3 (Mercian), Oridigm (MediQuest Therapeutics), Exherin (Adherex), BLS-0597 (Boston Life Sciences), PTC-299 (PTC Therapeutics), NPI-2358 (Nereus Pharmaceuticals), CGP-79787 (Novartis), AEE-788 (Novartis), CKD-732 (Chong Kun Dang), CP-564959 (OSI Pharmaceuticals), CM-101 (CarboMed), CT-2584, CT3501 (Cell Therapeutics), combretastatin and analogues and derivatives thereof (Oxigene), Rebimastat (Bristol-Meyers Squibb), Dextrin 2-sulfate (ML Laboratories), Cilengitide (Merk KGaA), NSC-706704 (Pharminox), KRN- 951 (Kirin Brewery), Ukrain, NSC-631570 (Nowicky Pharma), Tecogalan sodium (Daiichi Pharmaceutical), Tz-93 (Tsumura), TBC-1635 (Encysive Pharmaceuticals), TAN-1120 (Takeda), Semaxanib (Pfizer), BDI-7800 (Biopharmacopae), SD-186, SD-983 (Bristol-Meyers Squibb), SB-223245 (GlaxoSmithKline), SC-236 (Pfizer), RWJ-590973 (Johnson and Johnson), ILX-1850 (Genzyme), SC-68488, S-836 (Pfizer), CG-55069-11 (CuraGen), Ki-23057 (Kirin Brewery), CCX-700 (Chemoentryx), Pegaptanib octasodium (Giled Sciences), ANGIOCOL (available from Biostratum Inc.), or an analogue or derivative thereof.
12402. The method of item 12395 wherein the agent is an apoptosis antagonist.
12403. The method of item 12395 wherein the agent is an apoptosis activator.
12404. The method of item 12395 wherein the agent is a beta 1 integrin antagonist.
12405. The method of item 12395 wherein the agent is a beta tubulin inhibitor.
12406. The method of item 12395 wherein the agent is a blocker of enzyme production in Hepatitis C.
12407. The method of item 12395 wherein the agent is a Bruton's tyrosine kinase inhibitor. 12408. The method of item 12395 wherein the agent is a calcineurin inhibitor.
12409. The method of item 12395 wherein the agent is a caspase 3 inhibitor.
12410. The method of item 12395 wherein the agent is a CC chemokine receptor antagonist.
12411. The method of item 12395 wherein the agent is a cell cycle inhibitor selected from the group consisting of SNS-595 (Sunesis), synthadotin, KRX-0403, and an analogue or derivative thereof.
12412. The method of item 12395 wherein the agent is a cathepsin B inhibitor.
12413. The method of item 12395 wherein the agent is a cathepsin K inhibitor, wherein the cathepsin K inhibitor is 462795 (GlaxoSmithKline), INPL-022-D6 (Amura Therapeutics), or an analogue or derivative thereof.
12414. The method of item 12395 wherein the agent is a cathepsin L inhibitor.
12415. The method of item 12395 wherein the agent is a CD40 antagonist.
12416. The method of item 12395 wherein the agent is a chemokine receptor agonist. 12417. The method of item 12395 wherein the agent is a chymase inhibitor.
12418. The method of item 12395 wherein the agent is a collagenase antagonist.
12419. The method of item 12395 wherein the agent is a CXCR antagonist.
12420. The method of item 12395 wherein the agent is a cyclin dependent kinase inhibitor selected from the group consisting of a CDK-1 inhibitor, a CDK-2 inhibitor, a CDK- 4 inhibitor, a CDK-6 inhibitor, a CAK1 inhibitor from GPC Biotech and Bristol-Myers Squibb, RGB-286199 (GPC Biotech), an anticancer agent from Astex Technology, a CAK1 inhibitor from GPC Biotech, a CDK inhibitor from Sanofi-Aventis, a CDK1/CDK2 inhibitor from Amgen, a CDK2 inhibitor from SUGEN-2 (Pfizer), a hearing loss therapy agent (Sound Pharmaceuticals), PD-0332991 (Pfizer), RGB-286199 (GPC Biotech), Ro-0505124 (Hoffmann-La Roche), a Ser/Thr kinase inhibitor from Lilly (EIi Lilly), CVT-2584 (CAS No. 199986-75- 9) (CV Therapeutics), and an analogue or derivative thereof.
12421. ' The method of item 12395 wherein the agent is a cyclooxygenase 1 inhibitor.
12422. The method of item 12395 wherein the agent is a DHFR inhibitor.
12423. The method of item 12395 wherein the agent is a dual integrin inhibitor. 12424. The method of item 12395 wherein the agent is an elastase inhibitor.
12425. The method of item 12395 wherein the agent is an elongation factor-1 alpha inhibitor.
12426. The method of item 12395 wherein the agent is an endothelial growth factor antagonist.
12427. The method of item 12395 wherein the agent is an endothelial growth factor receptor kinase inhibitor selected from the group consisting of sorafenib tosylate (Bayer), AAL-993 (Novartis), ABP-309 (Novartis), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), E- 7080 (Eisai), EG-3306 (Ark Therapeutics), EXEL-2880 (Exelixis), GW- 654652 (GlaxoSmithKline), a KDR inhibitor from LG Life Sciences, CT-6685 and CT-6729 (UCB), KRN-633 and KRN-951 (Kirin Brewery), OSI-930 (OSI Pharmaceuticals), SP-5.2 (Supratek Pharma), SU-11657 (Pfizer), a Tie-2 antagonist (Hybrigenics), a VEGFR-2 kinase inhibitor (Bristol-Myers Squibb), XL-647 (Exelixis), a KDR inhibitor from Abbott Laboratories, sorafenib tosylate, and an analogue or derivative thereof.
12428. The method of item 12395 wherein the agent is an endotoxin antagonist.
12429. The method of item 12395 wherein the agent is an epothilone and tubulin binder.
12430. The method of item 12395 wherein the agent is an estrogen receptor antagonist. 12431. The method of item 12395 wherein the agent is an FGF inhibitor.
12432. The method of item 12395 wherein the agent is a famexyl transferase inhibitor.
12433. The method of item 12395 wherein the agent is famesyltransferase inhibitor selected from the group of A-197574 (Abbott), a farnesyltransferase inhibitor from Servier, FPTIII (Strathclyde Institute for Drug R), LB-42908 (LG Life Sciences), Pharmaprojects No. 5063 (Genzyme), Pharmaprojects No. 5597 (Ipsen), Yissum Project No. B-1055 (Yissum), and an analogue or derivative thereof.
12434. The method of item 12395 wherein the agent is an FLT-3 kinase inhibitor.
12435. The method of item 12395 wherein the agent is an FGF receptor kinase inhibitor.
12436. The method of item 12395 wherein the agent is a fibrinogen antagonist selected from the group consisting of AUV-201 (Auvation), MG-13926 (Sanofi-Aventis), plasminogen activator (CAS No. 105913-11-9) (from Sanofi-Aventis or UCB), plasminogen activator-2 (tPA- 2) (Sanofi-Aventis), pro-urokinase (CAS No. 82657-92-9) (Sanofi-Aventis), and an analogue or derivative thereof.
12437. The method of item 12395 wherein the agent is a heat shock protein 90 antagonist selected from the group consisting of SRN- 005 (Sirenade), geldanamycin, NSC-33050 (17-allylaminogeldanamycin; 17- AAG), 17-dimethylaminoethylamino-17-demethoxy-geldanamycin (17- DMAG), rifabutin (rifamycin XIV, 1',4-didehydro-1-deoxy-1 ,4-dihydro-5'-(2- methylpropyl)-1-oxo-), and an analogue or derivative thereof.
12438. The method of item 12395 wherein the agent is a histone deacetylase inhibitor.
12439. The method of item 12395 wherein the agent is an HMGCoA reductase inhibitor selected from the group consisting of an atherosclerosis therapeutic from Lipid Sciences, ATI-16000 (ARYx Therapeutics), KS-01-019 (Kos Pharmaceuticals), Pharmaprojects No. 2197 (Sanofi-Aventi), RP 61969 (Sanofi-Aventis), and an analogue or derivative thereof.
12440. The method of item 12395 wherein the agent is an ICAM inhibitor.
12441. The method of item 12395 wherein the agent is an IL, ICE and IRAK antagonist, wherein the antagonist is a CJ-14877, CP- 424174 (Pfizer), NF-61 (Negma-Lerads), and an analogue or derivative thereof.
12442. The method of item 12395 wherein the agent is an IL-2 inhibitor.
12443. The method of item 12395 wherein the agent is an immunosuppressant selected from the group consisting of teriflunomide (Sanofi Aventis), chlorsulfaquinoxalone (NSC-339004), chlorsulfaquinoxalone sulfate, CS-712 (Sankyo), ismomultin alfa (CAS No. 457913-93-8) (Akzo Nobel), antiallergics from GenPat77, antiinflammatories or AT-005 (Androclus Therapeutics), autoimmune disease therapeutics from EpiVax, BN-007 (Bone), budesonide (CAS No. 51333-22- 3) (MAP Pharmaceuticals), CO-14 (Genzyme), edratide (CAS No. 433922- 67-9) (Teva), EP-314 (Enanta), eprovafen (CAS No. 101335-99-3) (Sanofi- Aventis), HWA-131 (CAS No. 118788-41-3) (Sanofi-Aventis), immunomodulators from MerLion Pharmaceuticals, immunosuppressives from Alchemia, IPL-12 (Inflazyme), MDL-9563 (CAS No. 27086-86-8) (Sanofi-Aventis), Pharmaprojects No. 2330 (Sanofi-Aventis), Pharmaprojects No. 6426 (Abgenix), PXS-25 (Pharmaxis), rosmarinic acid (CAS No. 20283-92-5) (Sanofi-Aventis), RP 42927 or RP 54745 (CAS No. 135330-08-4) (Sanofi-Aventis), SGN-35 (Seattle Genetics), ST-1959 (Sigma-Tau), type I diabetes therapy from SYNX Pharma, UNIL-88 (Debiopharm), VP-025 (Vasogen), VR-694 (Vectura), PRTX-001 (Protalex), and an analogue or derivative thereof.
12444. The method of item 12395 wherein the agent is an IMPDH (inosine monophosphate).
12445. The method of item 12395 wherein the agent is an integrin antagonist.
12446. The method of item 12395 wherein the agent is an interleukin antagonist.
12447. The method of item 12395 wherein the agent is an inhibitor of type III receptor tyrosine kinase.
12448. The method of item 12395 wherein the agent is an irreversible inhibitor of enzyme methionine aminopeptidase type 2.
12449. The method of item 12395 wherein the agent is an isozyme selective delta protein kinase C inhibitor. 12450. The method of item 12395 wherein the agent a
JAK3 enzyme inhibitor.
12451. The method of item 12395 wherein the agent is a
JNK inhibitor.
12452. The method of item 12395 wherein the agent is a kinase inhibitor.
12453. The method of item 12395 wherein the agent is kinesin antagonist.
12454. The method of item 12395 wherein the agent is a kinesin antagonist.
12455. The method of item 12395 wherein the. agent is a leukotriene inhibitor and antagonist selected from the group consisting of ambicromil (CAS No. 58805-38-2) (Sanofi-Aventis), amelubant (CAS No. 346735-24-8) (Boehringer Ingelheim), DW-1141 (Dong Wha), ebselen (Daiichi Pharmaceutical), ibudilast (Kyorin), leucotriene inhibitors from Sanofi-Aventis, lymphotoxin-beta receptor (LT-D) from Biogen Idee, Pharmaprojects No. 1535 and 2728 (CAS No. 119340-33-9) (Sanofi- Aventis), R-112 (Rigel), Rev-5367 (CAS No. 92532-05-3) (Sanofi-Aventis), RG-14893 (CAS No. 141835-49-6) (Sanofi-Aventis), RG-5901-A (CAS No. 101910-24-1), 92532-23-5, RP 66153 (CAS No. 142422-79-5), RP 66364 (CAS No.186916-92-5), RP 69698 (CAS No. 141748-00-7) (Sanofi-Aventis), SC-411930 (Pfizer), SC-41930 (CAS No. 120072-59-5) (Pfizer), SC-50605 (CAS No. 138828-39-4) (Pfizer), SC-51146 (CAS No. 141059-52-1), SC- 53228 (CAS No. 153633-01-3) (Pfizer), spaglumic acid (ZY-15106) (CAS No.3106-85-2), 80619-64-3 (Novartis), tipredane (CAS No. 85197-77-9) (Bristol-Myers Squibb), U-75302 (CAS No. 119477-85-9) (Pfizer), and analogue or derivative thereof.
12456. The method of item 12395 wherein the agent is an MAP kinase inhibitor.
12457. The method of item 12395 wherein the agent is a matrix metalloproteinase inhibitor.
12458. The method of item 12395 wherein the agent is an MCP-CCR2 inhibitor.
12459. The method of item 12395 wherein the agent is an mTOR inhibitor.
12460. The method of item 12395 wherein the agent is an mTOR kinase inhibitor.
12461. The method of item 12395 wherein the agent is a microtubule inhibitor selected from the group consisting of antibody- maytansinoid conjugates from Biogen Idee, colchicines (MantiCore Pharmaceuticals), anticancer immunoconjugates from Johnson & Johnson, DIME from Octamer, gni-1f (GNI), huC242-DM4, huMy9-6-DM1 (ImmunoGen), IDN-5404 (Indena), IMO-098, IMOderm (Imotep), mebendazole (Introgen Therapeutics), microtubule poisons from Cambridge Enterprise, paclitaxel such as LOTAX from Aphios (CAS No. 33069-62-4), Genexol-PM from Samyang, Pharmaprojects No. 6383 (Tapestry Pharmaceuticals), RPR-112378 (Sanofi-Aventis), SGN-75 (Seattle Genetics), SPL-7435 (Starpharma), SSR-250411 (Sanofi-Aventis), trastuzumab-DM1 (Genentech), vinorelbine, and an analogue or derivative thereof. 12462. The method of item 12395 wherein the agent is an MIF inhibitor.
12463. The method of item 12395 wherein the agent is an MMP inhibitor.
12464. The method of item 12395 wherein the agent is a neurokinin (NK) antagonist selected from the group consisting of anthrotainin (CAS No. 148084-40-6) (Sanofi-Aventis), an IBS thereapeutic from ArQuIe, MDL-105212A (CAS No. 167261-60-1) (Ssanofi-Aventis), Pharmaprojects No. 2744, 3258 (CAS No. 139167-47-8) 4006, 4201, or 5986 (Sanofi-Aventis), RP 67580 (CAS No. 135911-02-3), SR-144190 (CAS No. 201152-86-5), SSR-240600, SSR-241586 (Sanofi-Aventis), TKA-457 (Novartis), vestipitant mesylate (CAS No. 334476-64-1) (GlaxoSmithKline), Win-64821 (Sanofi-Aventis), PRX-96026 (Predix Pharmaceuticals), and an analogue or derivative thereof.
12465. The method of item 12395 wherein the agent is an NF kappa B inhibitor selected from the group consisting of AVE-0545 or AVE-0547 (Sanofi-Aventis), bortezomib (CAS No. 179324-69-7) (Millennium Pharmaceuticals), dexanabinol (CAS No. 112924-45-5) (Pharmos), dexlipotam (Viatris), Pharmaprojects No. 6283 (INDRA) (OXiGENE), IPL- 576092 (CAS No. 137571-30-3) (Inflazyme), NFKB decoy (Corgentech), NFKB decoy oligo (AnGes MG), NFKB's from Ariad, osteoporosis treatments or S5 (F005) from Fulcrum Pharmaceuticals, P61 (Phytopharm), R-flurbiprofen (CAS No. 5104-49-4) (Encore Pharmaceuticals), and an analogue or derivative thereof.
12466. The method of item 12395 wherein the agent is a nitric oxide agonist. 12467. The method of item 12395 wherein the agent is an ornithine decarboxylase inhibitor.
12468. The method of item 12395 wherein the agent is a p38 MAP kinase inhibitor selected from the group consisting of AZD-6703 (AstraZeneca), JX-401 (Jexys Pharmaceuticals), BMS-2 (Bristol-Myers Squibb), a p38 MAP kinase inhibitor from Novartis, a p38-alpha MAP kinase inhibitor from Amphora, Pharmaprojects No. 5704 (Pharmacopeia), RPR- 200765A (Sanofi-Aventis), SD-282 (Johnson & Johnson), TAK-715 (Takeda), and an analogue or derivative thereof.
12469. The method of item 12395 wherein the agent is a palmitoyl-protein thioesterase inhibitor.
12470. The method of item 12395 wherein the agent is a PDGF receptor kinase inhibitor selected from the group consisting of AAL- 993, AMN-107, or ABP-309 (Novartis), AMG-706 (Amgen), BAY-57-9352 (Bayer), CDP-860 (UCB), E-7080 (Eisai), imatinib (CAS No. 152459-95-5) (Novartis), OSI-930 (OSI Pharmaceuticals), RPR-127963E (Sanofi-Aventis), RWJ-540973 (Johnson & Johnson), sorafenib tosylate (Bayer), SU-11657 (Pfizer), tandutinib (CAS No. 387867-13-2) (Millennium Pharmaceuticals), vatalanib (Novartis), ZK-CDK (Schering AG), and an analogue or derivative thereof.
12471. The method of item 12395 wherein the agent is (-)-halofenate (Metabolex), AMG-131 (Amgen), antidiabetics from Japan Tobacco, AZD-4619, AZD-8450, AZD-8677 (AstraZeneca), DRF-10945, balaglitazone (Dr Reddy's), CS-00088, CS-00098 (Chipscreen Biosciences), E-3030 (Eisai), etalocib (CAS No. 161172-51-6) (EIi Lilly), GSK-641597 (Ligand), GSK-677954 (GlaxoSmithKline), GW-409544 (Ligand), GW- 590735 (GlaxoSmithKline), K-111 (Hoffmann-La Roche), LY-518674 (EIi Lilly), LY-674 (Ligand), LY-929 (Ligand), MC-3001 , MC-3002 (MaxoCore Pharmaceuticals), metformin HCI + pioglitazone (CAS No. 1115-70-4 and 112529-15-4), ACTOPLUS MET from Andrx), muraglitazar (CAS No. 331741-94-7) (Bristol-Myers Squibb), naveglitazar (Ligand), oleoylethanolamide (Kadmus Pharmaceuticals), ONO-5129, pioglitazone hydrochloride (CAS No. 111025-46-8 and 112529-15-4) (Takeda), PLX-204 (Plexxikon), PPAR agonists from Genfit, PPAR delta agonists from EIi Lilly, PPAR-alpha agonists from CrystalGenomics, PPAR-gamma modulators and PPAR-β modulators from CareX, rosiglitazone maleate (CAS No. 122324- 73-4 or 155141-29-0) (GlaxoSmithKline), rosiglitazone maleate/glimepir (CAS No. 155141-29-0 and 93479-97-1), AVANDARYL, rosiglitazone maleate/metformin extend (CAS No. 155141-29-0 and 657-24-9), AVANDAMET, rosiglitazone maleate+metformin, AVANDAMET (GlaxoSmithKline), tesaglitazar (AstraZeneca), and an analogue or derivative thereof.
12472. The method of item 12395 wherein the agent is a phosphatase inhibitor.
12473. The method of item 12395 wherein the agent is a phosphodiesterase (PDE) inhibitor selected from the group consisting of avanafil (Tanabe Seiyaku), dasantafil (CAS No. 569351-91-3) (Schering- Plough), A-906119 (CAS No. 134072-58-5), DL-850 (Sanofi-Aventis), GRC- 3015, GRC-3566, GRC-3886 (Glenmark), HWA-153 (CAS No. 56395-66-5) (Sanofi-Aventis), hydroxypumafentrine (Altana), IBFB-130011 , IBFB-14-016, IBFB-140301 , IBFB-150007, IBFB-211913 (IBFB Pharma), L-826141 (Merck & Co), medorinone (CAS No. 88296-61-1) (Sanofi-Aventis), MEM- 1917 (Memory Pharmaceuticals), ND-1251 (Neuro3d), PDE inhibitors from ICOS, PDE IV inhibitors from Memory Pharmaceuticals and CrystalGenomics, Pharmaprojects No. 2742 and 6141 (Sanofi-Aventis), QAD-171 (Novartis), RHC-2963 (CAS No. 76993-12-9 and 76993-14-1), RPR-117658, RPR-122818 derivatives, SR-24870 , and RPR-132294 (Sanofi-Aventis), SK-350 (ln2Gen), stroke therapy agents from deCODE Genetics, TAS-203 (Taiho), tofimilast (CAS No. 185954-27-2) (Pfizer), UK- 371800 (Pfizer), WIN-65579 (CAS No. 158020-82-7) (Sanofi-Aventis), IBFB- 130020 (IBFB Pharma), OPC-6535 (CAS No. 145739-56-6) (Otsuka), a phosphodiesterase III inhibitor, enoximone, a phosphodiesterase IV inhibitor, fosfosal, Atopik (Barrier Therapeutics), triflusal, a phosphodiesterase V inhibitor, and an analogue or derivative thereof.
12474. The method of item 12395 wherein the agent is a PKC inhibitor.
12475. The method of item 12395 wherein the agent is a platelet activating factor antagonist.
12476. The method of item 12395 wherein the agent is a platelet-derived growth factor receptor kinase inhibitor.
12477. The method of item 12395 wherein the agent is a prolyl hydroxylase inhibitor.
12478. The method of item 12395 wherein the agent is a polymorphonuclear neutrophil inhibitor.
12479. The method of item 12395 wherein the agent is a protein kinase B inhibitor.
12480. The method of item 12395 wherein the agent is a protein kinase C stimulant. 12481. The method of item 12395 wherein the agent is a purine nucleoside analogue.
12482. The method of item 12395 wherein the agent is a purinoreceptor P2X antagonist.
12483. The method of item 12395 wherein the agent is a Raf kinase inhibitor.
12484. The method of item 12395 wherein the agent is a reversible inhibitor of ErbB1 and ErbB2.
12485. The method of item 12395 wherein the agent is a ribonucleoside triphosphate reductase inhibitor.
12486. The method of item 12395 wherein the agent is . an SDF-1 antagonist.
12487. The method of item 12395 wherein the agent is a sheddase inhibitor.
12488. The method of item 12395 wherein the agent is an SRC inhibitor.
12489. The method of item 12395 wherein the agent is a stromelysin inhibitor.
12490. The method of item 12395 wherein the agent is an Syk kinase inhibitor. 12491. The method of item 12395 wherein the agent is a telomerase inhibitor.
12492. The method of item 12395 wherein the agent is a TGF beta inhibitor selected from the group consisting of pirfenidone (CAS No. 53179-13-8) (MARNAC), tranilast (CAS No. 53902-12-8) (Kissei), IN- 1130 (ln2Gen), mannose-6-phosphate (BTG), TGF-β antagonists from Inflazyme (Pharmaprojects No. 6075), TGF-β antagonists from Sydney, non-industrial source), TGF-β I receptor kinase inhibitors from EIi Lilly, TGF- β receptor inhibitors from Johnson & Johnson, and an analogue or derivative thereof.
12493. The method of item 12395 wherein the agent is a TNFα antagonist or TACE inhibitor selected from the group consisting of adalimumab (CAS No. 331731-18-1) (Cambridge Antibody Technology), AGIX-4207 (AtheroGenics), AGT-1 (Advanced Biotherapy), an antiinflammatory from Borean Pharma, Cellzome, or Paradigm Therapeutics, anti-inflammatory vaccine (TNF-alpha kinoid) from Neovacs, humanized anti-TNF antibody or an anti-TNF MAb (CB0006) Celltech (UCB), apratastat (CAS No. 287445-51-0) (Wyeth), BMS-561392 (Bristol-Myers Squibb), BN- 006 (Bone), certolizumab pegol (CAS No. 428863-50-7 or CH-138 (UCB), cilomilast (CAS No. 153259-65-5) (GlaxoSmithKline), CR-1 (Nuada Pharmaceuticals), CRx-119 (CombinatoRx), D-5410 (UCB), dacopafant (CAS No. 125372-33-0) (Sanofi-Aventis), dersalazine (CAS No. 188913-57- 7/188913-58-8) (Uriach), etanercept (CAS No. 185243-69-0) (Amgen), ethyl pyruvate (Critical (Critical Therapeutics), golimumab (CAS No. 476181-74-5) (Johnson & Johnson), hormono-immunotherapy from Ipsen, CDP571 (e.g., Humicade from UCB), IC-485 (ICOS), infliximab (CAS No. 170277-31-3) (Johnson & Johnson), IP-751 (Manhattan Pharmaceuticals), ISIS-104838 (CAS No. 250755-32-9) (ISIS Pharmaceuticals), lenalidomide (CAS No. 191732-72-6) (Celgene), lentinan (CAS No. 37339-90-5) (Ajinomoto), MDL- 201112 (CAS No. 142130-73-2) (Sanofi-Aventis), medroxyprogesterone (CAS No. 520-85-4) (InKine Pharmaceutical), N-acetylcysteine (CAS No. 616-91-1) (Zambon), NBE-P2 (DIREVO Biotech), nerelimomab (CAS No. 162774-06-3) (Chiron), OM-294DP (OM PHARMA), onercept (CAS No. 199685-57-9) (Yeda), PASSTNF-alpha (Verigen), pentoxifylline or oxypentifylline (Sanofi-Aventis), Pharmaprojects No. 4091 , 4241 , 4295, or 4488 (Sanofi-Aventis), Pharmaprojects No. 5480 (Amgen), Pharmaprojects No. 6749 (Cengent), pirfenidone (CAS No. 53179-13-8) (MARNAC), RPR- 132294 (Sanofi-Aventis), S5 (F002) (Fulcrum Pharmaceuticals), simvastatin (CAS No. 79902-63-9) (Merck & Co), STA-6292 (Synta Pharmaceuticals), tacrolimus (CAS No. 104987-11-3) (Fujisawa LifeCycle Pharma), talactoferrin alfa (CAS No. 308240-58-6) (Agennix), thalidomide (CAS No. 50-35-1) (Celgene), TNF antagonists form ProStrakan, and Synergen, TNF inhibitors (Amgen), TNF-alpha antagonists from Dynavax Technologies and Jerina AG (Germany), TNF-alpha inhibitors from IBFB Pharma and Xencor (Xencor), torbafyliine (CAS No. 105102-21-4) (Sanofi-Aventis), UR-1505 (Uriach), VT-346 (Viron Therapeutics), YSIL6 (Y1S Therapeutics), YSTH2 (Y's Therapeutics), NPI-1302a-3 (Nereus Pharmaceuticals, a TNF antagonist from Jerina AG (Germany), dersalazine, and an analogue or derivative thereof.
12494. The method of item 12395 wherein the agent is a Toll receptor inhibitor.
12495. The method of item 12395 wherein the agent is a tubulin antagonist.
12496. The method of item 12395 wherein the agent is a tyrosine kinase inhibitor selected from the group consisting of SU-011248, SUTENT from Pfizer Inc. (New York, NY), BMS-354825, PN-355 (Paracelsian Pharmaceuticals), AGN-199659 (Allergan), AAL-993 or ABP- 309 (Novartis), adaphostin (NIH), AEE-788 (Novartis), AG-013736 (OSI Pharmaceuticals), AG-13736 (Pfizer), ALT-110 (Alteris Therapeutics), AMG- 706 (Amgen), anticancer MAbs from Xencor, anti-EGFrvlll MAbs from Abgenix, anti-HER2 MAb from Abiogen, AZD-2171 or AZD-9935 (AstraZeneca), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), CEP-5214 (Cephalon), CEP-7055 (Cephalon), cetuximab (ImClone Systems), CHIR-200131 and CHIR-258 (Chiron), CP-547632 (OSI Pharmaceuticals), CP-724714 (Pfizer), CT-301 (Creabilis Therapeutics), D- 69491 (Baxter International), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EGFR/ErbB2 inhibitors from Array BioPharma, erlotinib (CAS No. 183319- 69-9) (OSI Pharmaceuticals), EXEL-2880 (Exelixis), FK-778 (Sanofi- Aventis) , gefitinib (CAS No. 184475-35-2) (AstraZeneca), GW-2286 or GW- 654652 (GlaxoSmithKline), her2/neu antigen from AlphaVax, HER-2/neu inhibitor from Generex, Herzyme (Medipad) (Sirna Therapeutics), HKI-272 (Wyeth), HuMax-EGFr (Genmab), idronoxil (CAS No. 81267-65-4) (Novogen), IGF-1 inhibitors from Ontogen, IMC-11F8 (ImClone Systems), kahalalide F (CAS No. 149204-42-2) (PharmaMar), KDR inhibitor from LG Life Sciences, KDR inhibitors from Abbott Laboratories, KDR kinase inhibitors (UCB), Kdr kinase inhibitors from Merck & Co, KRN-633 and KRN- 951 (Kirin Brewery), KSB-102 (Xenova), lapatinib ditosylate (CAS No. 388082-78-8) (GlaxoSmithKline), matuzumab (Merck KGaA), MDX-214 (Medarex), ME-103 (Pharmexa), MED-A300 (Gerolymatos), MNAC-13 (Lay Line Genomics), nimotuzumab (Center of Molecular Immunology), NSC- 330507 or NSC-707545 (NIH), NV-50 (Novogen), OSI-930 (OSI Pharmaceuticals), panitumumab (Abgenix), pelitinib (CAS No. 287933-82-7) (Wyeth), pertuzumab (CAS No. 380610-27-5) (Genentech), Pharmaprojects No. 3985 (Sanofi-Aventis), prostate cancer therapeutics from Sequenom (SQPC35, SQPC36, SQPC90), removab and remoxab (Trion Pharma), RG- 13022 (CAS No. 136831-48-6), RG-13291 (CAS No. 138989-50-1), or RG- 14620 (CAS No. 136831-49-7) (Sanofi-Aventis), RM-6427 (Romark), RNAi breast cancer therapy from Benitec, RP 53801 (CAS No. 125882-88-4) (Sanofi-Aventis), sorafenib tosylate (Bayer), SU-11657 (Pfizer), Tie-2 antagonists from Semaia (Hybrigenics), Tie-2 inhibitors from Ontogen, trastuzumab (CAS No. 180288-69-1) (Genentech), tyrosine kinase inhibitors from Sanofi-Aventis, U3-1287, U3-1565, U3-1784, U3-1800 (U3 Pharma), vatalanib (Novartis), VEGFR-2 kinase inhibitor from Bristol-Myers Squibb, XL-647 (Exelixis), ZD-6474 (AstraZeneca), ZK-CDK (Schering AG), an EGFR tyrosine kinase inhibitor, EKB-569 (Wyeth), and an analogue or derivative thereof.
12497. The method of item 12395 wherein the agent is a VEGF inhibitor.
12498. The method of item 12395 wherein the agent is a vitamin D receptor agonist.
12499. The method of item 12395 wherein the agent is. ZD-6474 (an angiogenesis inhibitor).
12500. The method of item 12395 wherein the agent is AP-23573 (an mTOR inhibitor).
12501. The method of item 12395 wherein the agent is synthadotin (a tubulin antagonist).
12502. The method of item 12395 wherein the agent is S-0885 (a collagenase inhibitor).
12503. The method of item 12395 wherein the agent is aplidine (an elongation factor-1 alpha inhibitor). 12504. The method of item 12395 wherein the agent is ixabepilone (an epithilone).
12505. The method of item 12395 wherein the agent is IDN-5390 (an angiogenesis inhibitor).
12506. The method of item 12395 wherein the agent is SB-2723005 (an angiogenesis inhibitor).
12507. The method of item 12395 wherein the agent is ABT-518 (an angiogenesis inhibitor).
12508. The method of item 12395 wherein the agent is combretastatin (an angiogenesis inhibitor).
12509. The method of item 12395 wherein the agent is anecortave acetate (an angiogenesis inhibitor).
12510. The method of item 12395 wherein the agent is SB-715992 (a kinesin antagonist).
12511. The method of item 12395 wherein the agent is temsirolimus (an mTOR inhibitor).
12512. The method of item 12395 wherein the agent is adalimumab (a TNFα antagonist).
12513. The method of item 12395, wherein the composition comprises a polymer. 12514. The method of item 12395, wherein the composition comprises a polymeric carrier.
12515. The method of item 12395 wherein the anti- scarring agent inhibits adhesion between the device and a host into which the device is implanted.
12516. The method of item 12395 wherein the device delivers the anti-scarring agent locally to tissue proximate to the device.
12517. The method of item 12395 wherein the device has a coating that comprises the anti-scarring agent.
12518. The method of item 12395, wherein the device has a coating that comprises the agent and is disposed on a surface of the implant.
12519. The method of item 12395, wherein the device has a coating that comprises the agent and directly contacts the implant.
12520. The method of item 12395, wherein the device has a coating that comprises the agent and indirectly contacts the implant.
12521. The method of item 12395, wherein the device has a coating that comprises the agent and partially covers the implant.
12522. The method of item 12395, wherein the device has a coating that comprises the agent and completely covers the implant.
12523. The method of item 12395, wherein the device has a uniform coating. 12524. The method of item 12395, wherein the device has a non-uniform coating.
12525. The method of item 12395, wherein the device has a discontinuous coating.
12526. The method of item 12395, wherein the device has a patterned coating.
12527. The method of item 12395, wherein the device has a coating with a thickness of 100 Dm or less.
12528. The method of item 12395, wherein the device has a coating with a thickness of 10 Dm or less.
12529. The method of item 12395, wherein the device has a coating, and the coating adheres to the surface of the implant upon deployment of the implant.
12530. The method of item 12395, wherein the device has a coating, and wherein the coating is stable at room temperature for a period of 1 year.
12531. The method of item 12395, wherein the device has a coating, and wherein the anti-scarring agent is present in the coating in an amount ranging between about 0.0001% to about 1% by weight.
12532. The method of item 12395, wherein the device has a coating, and wherein the anti-scarring agent is present in the coating in an amount ranging between about 1% to about 10% by weight. 12533. The method of item 12395, wherein the device has a coating, and wherein the anti-scarring agent is present in the coating in an amount ranging between about 10% to about 25% by weight.
12534. The method of item 12395, wherein the device has a coating, and wherein the anti-scarring agent is present in the coating in an amount ranging between about 25% to about 70% by weight.
12535. The method of item 12395, wherein the device has a coating, and wherein the coating further comprises a polymer.
12536. The method of item 12395, wherein the device has a first coating having a first composition and a second coating having a second composition.
12537. The method of item 12395, wherein the device has a first coating having a first composition and a second coating having a second composition, wherein the first composition and the second composition are different.
12538. The method of item 12395, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a copolymer.
12539. The method of item 12395, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a block copolymer.
12540. The method of item 12395, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a random copolymer. 12541. The method of item 12395, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a biodegradable polymer.
12542. The method of item 12395, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a non-biodegradable polymer.
12543. The method of item 12395, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a hydrophilic polymer.
12544. The method of item 12395, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a hydrophobic polymer.
12545. The method of item 12395, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a polymer having hydrophilic domains.
12546. The method of item 12395, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a polymer having hydrophobic domains.
12547. The method of item 12395, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a non-conductive polymer.
12548. The method of item 12395, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises an elastomer. 12549. The method of item 12395, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a hydrogel.
12550. The method of item 12395, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a silicone polymer.
12551. The method of item 12395, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a hydrocarbon polymer.
12552. The method of item 12395, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a styrene-derived polymer.
12553. The method of item 12395, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a butadiene polymer.
12554. The method of item 12395, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a macromer.
12555. The method of item 12395, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a poly(ethylene glycol) polymer.
12556. The method of item 12395 wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises an amorphous polymer. 12557. The method of item 12395, wherein the device comprises a lubricious coating.
12558. The method of item 12395 wherein the anti- scarring agent is located within pores or holes of the device.
12559. The method of item 12395 wherein the anti- scarring agent is located within a channel, lumen, or divet of the device.
12560. The method of item 12395, wherein the device comprises a second pharmaceutically active agent.
12561. The method of item 12395 wherein the device comprises an anti-inflammatory agent.
12562. The method of item 12395 wherein the device comprises an agent that inhibits infection.
12563. The method of item 12395 wherein the device comprises an agent that inhibits infection, and wherein the agent is an anthracycline.
12564. The method of item 12395 wherein the device comprises an agent that inhibits infection, and wherein the agent is doxorubicin.
12565. The method of item 12395 wherein the device comprises an agent that inhibits infection, and wherein the agent is mitoxantrone. 12566. The method of item 12395 wherein the device comprises an agent that inhibits infection, and wherein the agent is a fluoropyrimidine.
12567. The method of item 12395 wherein the device comprises an agent that inhibits infection, and wherein the agent is 5- fluorouracil (5-FU).
12568. The method of item 12395 wherein the device comprises an agent that inhibits infection, and wherein the agent is a folic acid antagonist.
12569. The method of item 12395 wherein the device comprises an agent that inhibits infection, and wherein the agent is methotrexate.
12570. The method of item 12395 wherein the device comprises an agent that inhibits infection, and wherein the agent is a podophylotoxin.
12571. The method of item 12395 wherein the device comprises an agent that inhibits infection, and wherein the agent is etoposide.
12572. The method of item 12395 wherein the device comprises an agent that inhibits infection, and wherein the agent is a camptothecin.
12573. The method of item 12395 wherein the device comprises an agent that inhibits infection, and wherein the agent is a hydroxyurea. 12574. The method of item 12395 wherein the device comprises an agent that inhibits infection, and wherein the agent is a platinum complex.
12575. The method of item 12395 wherein the device comprises an agent that inhibits infection, and wherein the agent is cisplatin.
12576. The method of item 12395, further comprising an anti-thrombotic agent.
12577. The method of item 12395 wherein the device comprises a visualization agent.
12578. The method of item 12395 wherein the device comprises a visualization agent, wherein the visualization agent is a radiopaque material, and wherein the radiopaque material comprises a metal, a halogenated compound, or a barium containing compound.
12579. The method of item 12395 wherein the device comprises a visualization agent, wherein the visualization agent is a radiopaque material, and wherein the radiopaque material comprises barium, tantalum, or technetium.
12580. The method of item 12395 wherein the device comprises a visualization agent, and wherein the visualization agent is a MRI responsive material.
12581. The method of item 12395 wherein the device comprises a visualization agent, and wherein the visualization agent comprises a gadolinium chelate. 12582. The method of item 12395 wherein the device comprises a visualization agent, and wherein the visualization agent comprises iron, magnesium, manganese, copper, or chromium.
12583. The method of item 12395 wherein the device comprises a visualization agent, and wherein the visualization agent comprises an iron oxide compound.
12584. The method of item 12395 wherein the device comprises a visualization agent, and wherein the visualization agent comprises a dye, pigment, or colorant.
12585. The method of item 12395 wherein the device comprises an echogenic material.
12586. The method of item 12395 wherein the device comprises an echogenic material, and wherein the echogenic material is in the form of a coating.
12587. The method of item 12395 wherein the device is sterile.
12588. The method of item 12395 wherein the anti- scarring agent is released into tissue in the vicinity of the device after deployment of the device.
12589. The method of item 12395 wherein the anti- scarring agent is released into tissue in the vicinity of the device after deployment of the device, and wherein the tissue is connective tissue. 12590. The method of item 12395 wherein the anti- scarring agent is released into tissue in the vicinity of the device after deployment of the device, and wherein the tissue is muscle tissue.
12591. The method of item 12395 wherein the anti- scarring agent is released into tissue in the vicinity of the device after deployment of the device, and wherein the tissue is nerve tissue.
12592. The method of item 12395 wherein the anti- scarring agent is released into tissue in the vicinity of the device after deployment of the device, and wherein the tissue is epithelium tissue.
12593. The method of item 12395 wherein the anti- scarring agent is released in effective concentrations from the device over a period ranging from the time of deployment of the device to about 1 year.
12594. The method of item 12395 wherein the anti- scarring agent is released in effective concentrations from the device over a period ranging from about 1 month to 6 months.
12595. The method of item 12395 wherein the anti- scarring agent is released in effective concentrations from the device over a period ranging from about 1 - 90 days.
12596. The method of item 12395 wherein the anti- scarring agent is released in effective concentrations from the device at a constant rate.
12597. The method of item 12395 wherein the anti- scarring agent is released in effective concentrations from the device at an increasing rate. 12598. The method of item 12395 wherein the anti- scarring agent is released in effective concentrations from the device at a decreasing rate.
12599. The method of item 12395 wherein the anti- scarring agent is released in effective concentrations from the composition comprising the anti-scarring agent by diffusion over a period ranging from the time of deployment of the device to about 90 days.
12600. The method of item 12395 wherein the anti- scarring agent is released in effective concentrations from the composition comprising the anti-scarring agent by erosion of the composition over a period ranging from the time of deployment of the device to about 90 days.
12601. The method of item 12395 wherein the device comprises about 0.01 Dg to about 10 Dg of the anti-scarring agent.
12602. The method of item 12395 wherein the device comprises about 10 Dg to about 10 mg of the anti-scarring agent.
12603. The method of item 12395 wherein the device comprises about 10 mg to about 250 mg of the anti-scarring agent.
12604. The method of item 12395 wherein the device comprises about 250 mg to about 1000 mg of the anti-scarring agent.
12605. The method of item 12395 wherein the device comprises about 1000 mg to about 2500 mg of the anti-scarring agent. 12606. The method of item 12395 wherein a surface of the device comprises less than 0.01 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
12607. The method of item 12395 wherein a surface of the device comprises about 0.01 Dg to about 1 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
12608. The method of item 12395 wherein a surface of the device comprises about 1 Dg to about 10 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
12609. The method of item 12395 wherein a surface of the device comprises about 10 Dg to about 250 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
12610. The method of item 12395 wherein a surface of the device comprises about 250 Dg to about 1000 Dg of the anti-scarring agent of anti-scarring agent per mm2 of device surface to which the anti- scarring agent is applied.
12611. The method of item 12395 wherein a surface of the device comprises about 1000 Dg to about 2500 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
12612. The method of item 12395 wherein the combining is performed by direct affixing the agent or the composition to the implant. 12613. The method of item 12395 wherein the combining is performed by spraying the agent or the component onto the implant.
12614. The method of item 12395 wherein the combining is performed by electrospraying the agent or the composition onto the implant.
12615. The method of item 12395 wherein the combining is performed by dipping the implant into a solution comprising the agent or the composition.
12616. The method of item 12395 wherein the combining is performed by covalently attaching the agent or the composition to the implant.
12617. The method of item 12395 wherein the combining is performed by non-covalently attaching the agent or the composition to the implant.
12618. The method of item 12395 wherein the combining is performed by coating the implant with a substance that contains the agent or the composition.
12619. The method of item 12395 wherein the combining is performed by coating the implant with a substance that absorbs the agent.
12620. The method of item 12395 wherein the combining is performed by interweaving a thread composed of, or coated with, the agent or the composition. 12621. The method of item 12395 wherein the combining is performed by covering all the implant with a sleeve that contains the agent or the composition.
12622. The method of item 12395 wherein the combining is performed by covering a portion of the implant with a sleeve that contains the agent or the composition.
12623. The method of item 12395 wherein the combining is performed by covering all the implant with a cover that contains the agent or the composition.
12624. The method of item 12395 wherein the combining is performed by covering a portion of the implant with a cover that contains the agent or the composition.
12625. The method of item 12395 wherein the combining is performed by covering all the implant with an electrospun fabric that contains the agent or the composition.
12626. The method of item 12395 wherein the combining is performed by covering a portion of the implant with an electrospun fabric that contains the agent or the composition.
12627. The method of item 12395 wherein the combining is performed by covering all the implant with a mesh that contains the agent or the composition.
12628. The method of item 12395 wherein the combining is performed by covering a portion of the implant with a mesh that contains the agent or the composition. 12629. The method of item 12395 wherein the combining is performed by constructing all the implant with the agent or the composition.
12630. The method of item 12395 wherein the combining is performed by constructing a portion of the implant with the agent or the composition.
12631. The method of item 12395 wherein the combining is performed by impregnating the implant with the agent or the composition.
12632. The method of item 12395 wherein the combining is performed by constructing all of the implant from a degradable polymer that releases the agent.
12633. The method of item 12395 wherein the combining is performed by constructing a portion of the implant from a degradable polymer that releases the agent.
12634. The method of item 12395 wherein the combining is performed by dipping the implant into a solution that comprise the agent and an inert solvent for the implant.
12635. The method of item 12395 wherein the combining is performed by dipping the implant into a solution that comprises the agent and a solvent that will swell the implant.
12636. The method of item 12395 wherein the combining is performed by dipping the implant into a solution that comprises the agent and a solvent that will dissolve the implant. 12637. The method of item 12395 wherein the combining is performed by dipping the implant into a solution that comprises the agent, a polymer and an inert solvent for the implant.
12638. The method of item 12395 wherein the combining is performed by dipping the implant into a solution that comprises the agent, a polymer and a solvent that will swell the implant.
12639. The method of item 12395 wherein the combining is performed by dipping the implant into a solution that comprises the agent, a polymer and a solvent that will dissolve the implant.
12640. The method of item 12395 wherein the combining is performed by spraying the implant into a solution that comprises the agent and an inert solvent for the implant.
12641. The method of item 12395 wherein the combining is performed by spraying the implant into a solution that comprises the agent and a solvent that will swell the implant.
12642. The method of item 12395 wherein the combining is performed by spraying the implant into a solution that comprises the agent and a solvent that will dissolve the implant.
12643. The method of item 12395 wherein the combining is performed by spraying the implant into a solution that comprises the agent, a polymer and an inert solvent for the implant.
12644. The method of item 12395 wherein the combining is performed by spraying the implant into a solution that comprises the agent, a polymer and a solvent that will swell the implant. 12645. The method of item 12395 wherein the combining is performed by spraying the implant into a solution that comprises the agent, a polymer and a solvent that will dissolve the implant.
12646. A method as in any one of items 12395-12645, wherein the device is a lacrimal duct stent.
12647. A method as in any one of items 12395-12645, wherein the device is an Eustachian tube stent.
12648. A method as in any one of items 12395-12645, wherein the device is a nasal stent.
12649. A method as in any one of items 12395-12645, wherein the device is a sinus stent.
12650. A method of making a medical device comprising: combining an ear ventilation tube (e.g., an implant) and an anti- scarring agent or a composition comprising an anti-scarring agent, wherein the agent inhibits scarring between the device and a host into which the device is implanted.
12651. The method of item 12650 wherein the agent is an adensosine A2A receptor antagonist.
12652. The method of item 12650 wherein the agent is an AKT inhibitor.
12653. The method of item 12650 wherein the agent is an alpha 2 integrin antagonist, wherein the alpha 2 integrin antagonist is Pharmaprojects No. 5754 (Merck KgaA). 12654. The method of item 12650 wherein the agent is an alpha 4 integrin antagonist.
12655. The method of item 12650 wherein the agent is an alpha 7 nicotinic receptor agonist.
12656. The method of item 12650 wherein the agent is an angiogenesis inhibitor selected from the group consisting of AG-12,958 (Pfizer), ATN-161 (Attenuon LLC), neovastat, an angiogenesis inhibitor from Jerina AG (Germany), NM-3 (Mercian), VGA-1155 (Taisho), FCE-26644 (Pfizer), FCE-26950 (Pfizer), FPMA (Meiji Daries), FR-111142 (Fujisawa), GGTI-298, GM-1306 (Ligand), GPA-1734 (Novartis), NNC-47-0011 (Novo Nordisk), herbamycin (Nippon Kayaku), lenalidomide (Celegene), IP-10 (NIH), ABT-828 (Abbott), KIN-841 (Tokushima University, Japan), SF-1126 (Semafore Pharmaceuticals), laminin technology (NIH), CHIR-258 (Chiron), NVP-AEW541 (Novartis), NVP-AEW541 (Novartis), Vt16907 (Alchemia), OXI-8007 (Oxigene), EG-3306 (Ark Therapeutics), Maspin (Arriva), ABT- 567 (Abbott), PPI-2458 (Praecis Pharmaceuticals), CC-5079, CC-4089 (Celgene), HIF-1 alpha inhibitors (Xenova), S-247 (Pfizer), AP-23573 (Ariad), AZD-9935 (Astra Zeneca), mebendazole (Introgen Therapeutics), MetAP-2 inhibitors (GlaxoSmithKline), AG-615 (Angiogene Pharmaceuticals), Tie-2 antagonists (Hybrigenics), NC-381 , CYC-381, NC-169, NC-219, NC-383, NC-384, NC-407 (Lorus Therapeutics), ATN-224 (Attenuon), ON-01370 (Onconova), Vitronectin antagonists (Amgen), SDX-103 (Salmedix), Vitronectin antagonists (Shire), CHP (Riemser), TEK (Amgen), Anecortave acetate (Alcon), T46.2 (Matrix Therapeutics), HG-2 (Heptagen), TEM antagonists (Genzyme), Oxi-4500 (Oxigene), ATN-161 (Attenuon), WX-293 (Wilex), M-2025 (Metris Therapeutics), Alphastatin (BioActa), YH-16 (Yantai Rongchang), BIBF-1120 (Boehringer Ingelheim), BAY-57-9352 (Bayer), AS- 1404 (Cancer Research Technology), SC-77964 (Pfizer), glycomimetics (BioTie Therapies), TIE-2 Inhibitors (Ontogen), DIMI, Octamer (Octamer), ABR-215050 (Active Biotech), ABT-518 (Abbott), KDR inhibitors (Abbott), BSF-466895 (Abbott), SCH-221153 (Schering-Plough), DAC:antiangiogenic (ConjuChem), TFPI (EntreMed), AZD-2171 (Astra-Zenaca), CDC-394 (Celgene), LY290293 (EIi Lilly), IDN-5390 (Indena), Kdr Kinase Inhibitors (Merck), CT-113020, CT-116433, CT-116563, CT-31890, CT-32228) (Cell Therapeutics), A-299620 (Abbott), TWEAK Inhibitor (Amgen), VEGF modulators (Johnson and Johnson), Tum-N53, tumstatin (Genzyme), Thios- 1 , Thios-2 (Thios Pharmaceuticals), MV-6401 (Miravant Medical Technologies), Spisulosine (PharmaMar), CEP-7055 (Cephalon), AUV-201 (Auvation), LM-609 (EIi Lilly), SKF-106615 (AnorMED), Oglufanide disodium (Cytran), BW-114 (Pharminox), Calreticulin (NIH), WX-678 (Wilex), SD-7784 (Pfizer), WX-UK1 (Wilex), SH-268 (Schering AG), 2-Me-PGA (Celgene), S- 137 (Pfizer), ZD-6126 (Angiogene Pharmaceuticals), SG-292 (SignalGen), Benefin (Lane Labs), A6, A36 (Angstrom), SB-2723005 (GlaxoSmithKline), SC-7 (Cell Therapeutics), ZEN-014 (AEterna Zentaris), 2-methoxyestradiol (EntreMed), NK-130119 (Nippon Kayaku), CC-10004 (Celgene), AVE- 8062A (Ajinomoto), Tacedinaline (Pfizer), Actinonin (Tokyo Metropolitan Institute of Medical Science), Lenalidomide (Celgene), VGA-1155, BTO-956 (SRI International), ER-68203-00 (Eisai), CT-6685 (UCB), JKC-362 (Phoenix Pharmaceuticals), DMI-3798 (DMI Biosciences, Angiomate (Ipsen), ZD-6474 (AstraZeneca), CEP-5214 (Cephalon), Canstatin (Genzyme), NM-3 (Mercian), Oridigm (MediQuest Therapeutics), Exherin (Adherex), BLS-0597 (Boston Life Sciences), PTC-299 (PTC Therapeutics), NPI-2358 (Nereus Pharmaceuticals), CGP-79787 (Novartis), AEE-788 (Novartis), CKD-732 (Chong Kun Dang), CP-564959 (OSI Pharmaceuticals), CM-101 (CarboMed), CT-2584, CT3501 (Cell Therapeutics), combretastatin and analogues and derivatives thereof (Oxigene), Rebimastat (Bristol-Meyers Squibb), Dextrin 2-sulfate (ML Laboratories), Cilengitide (Merk KGaA), NSC-706704 (Pharminox), KRN- 951 (Kirin Brewery), Ukrain, NSC-631570 (Nowicky Pharma), Tecogalan sodium (Daiichi Pharmaceutical), Tz-93 (Tsumura), TBC-1635 (Encysive Pharmaceuticals), TAN-1120 (Takeda), Semaxanib (Pfizer), BDI-7800 (Biopharmacopae), SD-186, SD-983 (Bristol-Meyers Squibb), SB-223245 (GlaxoSmithKline), SC-236 (Pfizer), RWJ-590973 (Johnson and Johnson), ILX-1850 (Genzyme), SC-68488, S-836 (Pfizer), CG-55069-11 (CuraGen), Ki-23057 (Kirin Brewery), CCX-700 (Chemoentryx), Pegaptanib octasodium (Giled Sciences), ANGIOCOL (available from Biostratum Inc.), or an analogue or derivative thereof.
12657. The method of item 12650 wherein the agent is an apoptosis antagonist.
12658. The method of item 12650 wherein the agent is an apoptosis activator.
12659. The method of item 12650 wherein the agent is a beta 1 integrin antagonist.
12660. The method of item 12650 wherein the agent is a beta tubulin inhibitor.
12661. The method of item 12650 wherein the agent is a blocker of enzyme production in Hepatitis C.
12662. The method of item 12650 wherein the agent is a Bruton's tyrosine kinase inhibitor.
12663. The method of item 12650 wherein the agent is a calcineurin inhibitor.
12664. The method of item 12650 wherein the agent is a caspase 3 inhibitor. 12665. The method of item 12650 wherein the agent is a CC chemokine receptor antagonist.
12666. The method of item 12650 wherein the agent is a cell cycle inhibitor selected from the group consisting of SNS-595 (Sunesis), synthadotin, KRX-0403, and an analogue or derivative thereof.
12667. The method of item 12650 wherein the agent is a cathepsin B inhibitor.
12668. The method of item 12650 wherein the agent is a cathepsin K inhibitor, wherein the cathepsin K inhibitor is 462795 (GlaxoSmithKline), INPL-022-D6 (Amura Therapeutics), or an analogue or derivative thereof.
12669. The method of item 12650 wherein the agent is a cathepsin L inhibitor.
12670. The method of item 12650 wherein the agent is a CD40 antagonist.
12671. The method of item 12650 wherein the agent is a chemokine receptor agonist.
12672. The method of item 12650 wherein the agent is a chymase inhibitor.
12673. The method of item 12650 wherein the agent is a collagenase antagonist. 12674. The method of item 12650 wherein the agent is a CXCR antagonist.
12675. The method of item 12650 wherein the agent is a cyclin dependent kinase inhibitor selected from the group consisting of a CDK-1 inhibitor, a CDK-2 inhibitor, a CDK- 4 inhibitor, a CDK-6 inhibitor, a CAK1 inhibitor from GPC Biotech and Bristol-Myers Squibb, RGB-286199 (GPC Biotech), an anticancer agent from Astex Technology, a CAK1 inhibitor from GPC Biotech, a CDK inhibitor from Sanofi-Aventis, a CDK1/CDK2 inhibitor from Amgen, a CDK2 inhibitor from SUGEN-2 (Pfizer), a hearing loss therapy agent (Sound Pharmaceuticals), PD-0332991 (Pfizer), RGB-286199 (GPC Biotech), Ro-0505124 (Hoffmann-La Roche), a Ser/Thr kinase inhibitor from Lilly (EIi Lilly), CVT-2584 (CAS No. 199986-75- 9) (CV Therapeutics), and an analogue or derivative thereof.
12676. The method of item 12650 wherein the agent js a cyclooxygenase 1 inhibitor.
12677. The method of item 12650 wherein the agent is a DHFR inhibitor.
12678. The method of item 12650 wherein the agent is a dual integrin inhibitor.
12679. The method of item 12650 wherein the agent is an elastase inhibitor.
12680. The method of item 12650 wherein the agent is an elongation factor-1 alpha inhibitor. 12681. The method of item 12650 wherein the agent is an endothelial growth factor antagonist.
12682. The method of item 12650 wherein the agent is an endothelial growth factor receptor kinase inhibitor selected from the group consisting of sorafenib tosylate (Bayer), AAL-993 (Novartis), ABP-309 (Novartis), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), E- 7080 (Eisai), EG-3306 (Ark Therapeutics), EXEL-2880 (Exelixis), GW- 654652 (GlaxoSmithKline), a KDR inhibitor from LG Life Sciences, CT-6685 and CT-6729 (UCB), KRN-633 and KRN-951 (Kirin Brewery), OSI-930 (OSI Pharmaceuticals), SP-5.2 (Supratek Pharma), SU-11657 (Pfizer), a Tie-2 antagonist (Hybrigenics), a VEGFR-2 kinase inhibitor (Bristol-Myers Squibb), XL-647 (Exelixis), a KDR inhibitor from Abbott Laboratories, sorafenib tosylate, and an analogue or derivative thereof.
12683. The method of item 12650 wherein the agent is an endotoxin antagonist.
12684. The method of item 12650 wherein the agent is an epothilone and tubulin binder.
12685. The method of item 12650 wherein the agent is an estrogen receptor antagonist.
12686. The method of item 12650 wherein the agent is an FGF inhibitor.
12687. The method of item 12650 wherein the agent is a famexyl transferase inhibitor. 12688. The method of item 12650 wherein the agent is farnesyltransferase inhibitor selected from the group of A-197574 (Abbott), a farnesyltransferase inhibitor from Servier, FPTIII (Strathclyde Institute for Drug R), LB-42908 (LG Life Sciences), Pharmaprojects No. 5063 (Genzyme), Pharmaprojects No. 5597 (Ipsen), Yissum Project No. B-1055 (Yissum), and an analogue or derivative thereof.
12689. The method of item 12650 wherein the agent is an FLT-3 kinase inhibitor.
12690. The method of item 12650 wherein the agent is an FGF receptor kinase inhibitor.
12691. The method of item 12650 wherein the agent is a fibrinogen antagonist selected from the group consisting of AUV-201 (Auvation), MG-13926 (Sanofi-Aventis), plasminogen activator (CAS No. 105913-11-9) (from Sanofi-Aventis or UCB), plasminogen activator-2 (tPA- 2) (Sanofi-Aventis), pro-urokinase (CAS No. 82657-92-9) (Sanofi-Aventis), and an analogue or derivative thereof.
12692. The method of item 12650 wherein the agent is a heat shock protein 90 antagonist selected from the group consisting of SRN- 005 (Sirenade), geldanamycin, NSC-33050 (17-allylaminogeldanamycin; 17- AAG), 17-dimethylaminoethylamino-17-demethoxy-geldanamycin (17- DMAG), rifabutin (rifamycin XIV, 1',4-didehydro-1-deoxy-1 ,4-dihydro-5'-(2- methylpropyl)-1-oxo-), and an analogue or derivative thereof.
12693. The method of item 12650 wherein the agent is a histone deacetylase inhibitor. 12694. The method of item 12650 wherein the agent is an HMGCoA reductase inhibitor selected from the group consisting of an atherosclerosis therapeutic from Lipid Sciences, ATM 6000 (ARYx Therapeutics), KS-01-019 (Kos Pharmaceuticals), Pharmaprojects No. 2197 (Sanofi-Aventi), RP 61969 (Sanofi-Aventis), and an analogue or derivative thereof.
12695. The method of item 12650 wherein the agent is an ICAM inhibitor.
12696. The method of item 12650 wherein the agent is an IL, ICE and IRAK antagonist, wherein the antagonist is a CJ-14877, CP- 424174 (Pfizer), NF-61 (Negma-Lerads), and an analogue or derivative thereof.
12697. The method of item 12650 wherein the agent is an IL-2 inhibitor.
12698. The method of item 12650 wherein the agent is an immunosuppressant selected from the group consisting of teriflunomide (Sanofi Aventis), chlorsulfaquinoxalone (NSC-339004), chlorsulfaquinoxalone sulfate, CS-712 (Sankyo), ismomultin alfa (CAS No. 457913-93-8) (Akzo Nobel), antiallergics from GenPat77, antiinflammatories or AT-005 (Androclus Therapeutics), autoimmune disease therapeutics from EpiVax, BN-007 (Bone), budesonide (CAS No. 51333-22- 3) (MAP Pharmaceuticals), CO-14 (Genzyme), edratide (CAS No. 433922- 67-9) (Teva), EP-314 (Enanta), eprovafen (CAS No. 101335-99-3) (Sanofi- Aventis), HWA-131 (CAS No. 118788-41-3) (Sanofi-Aventis), immunomodulators from MerLion Pharmaceuticals, immunosuppressives from Alchemia, IPL-12 (Inflazyme), MDL-9563 (CAS No. 27086-86-8) (Sanofi-Aventis), Pharmaprojects No. 2330 (Sanofi-Aventis), Pharmaprojects No. 6426 (Abgenix), PXS-25 (Pharmaxis), rosmarinic acid (CAS No. 20283-92-5) (Sanofi-Aventis), RP 42927 or RP 54745 (CAS No. 135330-08-4) (Sanofi-Aventis), SGN-35 (Seattle Genetics), ST-1959 (Sigma-Tau), type I diabetes therapy from SYNX Pharma, UNIL-88 (Debiopharm), VP-025 (Vasogen), VR-694 (Vectura), PRTX-001 (Protalex), and an analogue or derivative thereof.
12699. The method of item 12650 wherein the agent is an IMPDH (inosine monophosphate).
12700. The method of item 12650 wherein the agent is an integrin antagonist.
12701. The method of item 12650 wherein the agent is an interleukin antagonist.
12702. The method of item 12650 wherein the agent is an inhibitor of type III receptor tyrosine kinase.
12703. The method of item 12650 wherein the agent is an irreversible inhibitor of enzyme methionine aminopeptidase type 2.
12704. The method of item 12650 wherein the agent is an isozyme selective delta protein kinase C inhibitor.
12705. The method of item 12650 wherein the agent a JAK3 enzyme inhibitor.
12706. The method of item 12650 wherein the agent is a JNK inhibitor. 12707. The method of item 12650 wherein the agent is a kinase inhibitor.
12708. The method of item 12650 wherein the agent is kinesin antagonist.
12709. The method of item 12650 wherein the agent is a kinesin antagonist.
12710. The method of item 12650 wherein the agent is a leukotriene inhibitor and antagonist selected from the group consisting of ambicromil (CAS No. 58805-38-2) (Sanofi-Aventis), amelubant (CAS No. 346735-24-8) (Boehringer Ingelheim), DW-1141 (Dong Wha), ebselen (Daiichi Pharmaceutical), ibudilast (Kyorin), leucotriene inhibitors from Sanofi-Aventis, lymphotoxin-beta receptor (LT-D) from Biogen Idee, Pharmaprojects No. 1535 and 2728 (CAS No. 119340-33-9) (Sanofi- Aventis), R-112 (Rigel), Rev-5367 (CAS No. 92532-05-3) (Sanofi-Aventis), RG-14893 (CAS No. 141835-49-6) (Sanofi-Aventis), RG-5901-A (CAS No. 101910-24-1), 92532-23-5, RP 66153 (CAS No. 142422-79-5), RP 66364 (CAS No.186916-92-5), RP 69698 (CAS No. 141748-00-7) (Sanofi-Aventis), SC-411930 (Pfizer), SC-41930 (CAS No. 120072-59-5) (Pfizer), SC-50605 (CAS No. 138828-39-4) (Pfizer), SC-51146 (CAS No. 141059-52-1), SC- 53228 (CAS No. 153633-01-3) (Pfizer), spaglumic acid (ZY-15106) (CAS No.3106-85-2), 80619-64-3 (Novartis), tipredane (CAS No. 85197-77-9) (Bristol-Myers Squibb), U-75302 (CAS No. 119477-85-9) (Pfizer), and analogue or derivative thereof.
12711. The method of item 12650 wherein the agent is an MAP kinase inhibitor. 12712. The method of item 12650 wherein the agent is a matrix metalloproteinase inhibitor.
12713. The method of item 12650 wherein the agent is an MCP-CCR2 inhibitor.
12714. The method of item 12650 wherein the agent is an mTOR inhibitor.
12715. The method of item 12650 wherein the agent is an mTOR kinase inhibitor.
12716. The method of item 12650 wherein the agent is a microtubule inhibitor selected from the group consisting of antibody- maytansinoid conjugates from Biogen Idee, colchicines (MantiCore Pharmaceuticals), anticancer immunoconjugates from Johnson & Johnson, DIME from Octamer, gni-1f (GNI)1 huC242-DM4, huMy9-6-DM1 (ImmunoGen), IDN-5404 (Indena), IMO-098, IMOderm (Imotep), mebendazole (Introgen Therapeutics), microtubule poisons from Cambridge Enterprise, paclitaxel such as LOTAX from Aphios (CAS No. 33069-62-4), Genexol-PM from Samyang, Pharmaprojects No. 6383 (Tapestry Pharmaceuticals), RPR-112378 (Sanofi-Aventis), SGN-75 (Seattle Genetics), SPL-7435 (Starpharma), SSR-250411 (Sanofi-Aventis), trastuzumab-DM1 (Genentech), vinorelbine, and an analogue or derivative thereof.
12717. The method of item 12650 wherein the agent is an MIF inhibitor.
12718. The method of item 12650 wherein the agent is an MMP inhibitor. 12719. The method of item 12650 wherein the agent is a neurokinin (NK) antagonist selected from the group consisting of anthrotainin (CAS No. 148084-40-6) (Sanofi-Aventis), an IBS thereapeutic from ArQuIe1 MDL-105212A (CAS No. 167261-60-1) (Ssanofi-Aventis), Pharmaprojects No. 2744, 3258 (CAS No. 139167-47-8) 4006, 4201, or 5986 (Sanofi-Aventis), RP 67580 (CAS No. 135911-02-3), SR-144190 (CAS No. 201152-86-5), SSR-240600, SSR-241586 (Sanofi-Aventis), TKA-457 (Novartis), vestipitant mesylate (CAS No. 334476-64-1) (GlaxoSmithKline), Win-64821 (Sanofi-Aventis), PRX-96026 (Predix Pharmaceuticals), and an analogue or derivative thereof.
12720. The method of item 12650 wherein the agent is an NF kappa B inhibitor selected from the group consisting of AVE-0545 or AVE-0547 (Sanofi-Aventis), bortezomib (CAS No. 179324-69-7) (Millennium Pharmaceuticals), dexanabinol (CAS No. 112924-45-5) (Pharmos), dexlipotam (Viatris), Pharmaprojects No. 6283 (INDRA) (OXiGENE), IPL- 576092 (CAS No. 137571-30-3) (Inflazyme), NFKB decoy (Corgentech), NFKB decoy oligo (AnGes MG), NFKB's from Ariad, osteoporosis treatments or S5 (F005) from Fulcrum Pharmaceuticals, P61 (Phytopharm), R-flurbiprofen (CAS No. 5104-49-4) (Encore Pharmaceuticals), and an analogue or derivative thereof.
12721. The method of item 12650 wherein the agent is a nitric oxide agonist.
12722. The method of item 12650 wherein the agent is an ornithine decarboxylase inhibitor.
12723. The method of item 12650 wherein the agent is a p38 MAP kinase inhibitor selected from the group consisting of AZD-6703 (AstraZeneca), JX-401 (Jexys Pharmaceuticals), BMS-2 (Bristol-Myers Squibb), a p38 MAP kinase inhibitor from Novartis, a p38-alpha MAP kinase inhibitor from Amphora, Pharmaprojects No. 5704 (Pharmacopeia), RPR- 200765A (Sanofi-Aventis), SD-282 (Johnson & Johnson), TAK-715 (Takeda), and an analogue or derivative thereof.
12724. The method of item 12650 wherein the agent is a palmitoyl-protein thioesterase inhibitor.
12725. The method of item 12650 wherein the agent is a PDGF receptor kinase inhibitor selected from the group consisting of AAL- 993, AMN-107, or ABP-309 (Novartis), AMG-706 (Amgen), BAY-57-9352 (Bayer), CDP-860 (UCB), E-7080 (Eisai), imatinib (CAS No. 152459-95-5) (Novartis), OSI-930 (OSI Pharmaceuticals), RPR-127963E (Sanofi-Aventis), RWJ-540973 (Johnson & Johnson), sorafenib tosylate (Bayer), SU-11657 (Pfizer), tandutinib (CAS No. 387867-13-2) (Millennium Pharmaceuticals), vatalanib (Novartis), ZK-CDK (Schering AG), and an analogue or derivative thereof.
12726. The method of item 12650 wherein the agent is (-)-halofenate (Metabolex), AMG-131 (Amgen), antidiabetics from Japan Tobacco, AZD-4619, AZD-8450, AZD-8677 (AstraZeneca), DRF-10945, balaglitazone (Dr Reddy's), CS-00088, CS-00098 (Chipscreen Biosciences), E-3030 (Eisai), etalocib (CAS No. 161172-51-6) (EIi Lilly), GSK-641597 (Ligand), GSK-677954 (GlaxoSmithKline), GW-409544 (Ligand), GW- 590735 (GlaxoSmithKline), K-111 (Hoffmann-La Roche), LY-518674 (EIi Lilly), LY-674 (Ligand), LY-929 (Ligand), MC-3001, MC-3002 (MaxoCore Pharmaceuticals), metformin HCI + pioglitazone (CAS No. 1115-70-4 and
112529-15-4), ACTOPLUS MET from Andrx), muraglitazar (CAS No. 331741-94-7) (Bristol-Myers Squibb), naveglitazar (Ligand), oleoylethanolamide (Kadmus Pharmaceuticals), ONO-5129, pioglitazone hydrochloride (CAS No. 111025-46-8 and 112529-15-4) (Takeda), PLX-204 (Plexxikon), PPAR agonists from Genfit, PPAR delta agonists from EIi Lilly, PPAR-alpha agonists from CrystalGenomics, PPAR-gamma modulators and PPAR-β modulators from CareX, rosiglitazone maleate (CAS No. 122324- 73-4 or 155141-29-0) (GlaxoSmithKline), rosiglitazone maleate/glimepir (CAS No. 155141-29-0 and 93479-97-1), AVANDARYL, rosiglitazone maleate/metformin extend (CAS No. 155141-29-0 and 657-24-9), AVANDAMET, rosiglitazone maleate+metformin, AVANDAMET (GlaxoSmithKline), tesaglitazar (AstraZeneca), and an analogue or derivative thereof.
12727. The method of item 12650 wherein the agent is a phosphatase inhibitor.
12728. The method of item 12650 wherein the agent is a phosphodiesterase (PDE) inhibitor selected from the group consisting of avanafil (Tanabe Seiyaku), dasantafil (CAS No. 569351-91-3) (Schering- Plough), A-906119 (CAS No. 134072-58-5), DL-850 (Sanofi-Aventis), GRC- 3015, GRC-3566, GRC-3886 (Glenmark), HWA-153 (CAS No. 56395-66-5) (Sanofi-Aventis), hydroxypumafentrine (Altana), IBFB-130011 , IBFB-14-016, IBFB-140301 , IBFB-150007, IBFB-211913 (IBFB Pharma), L-826141 (Merck & Co), medorinone (CAS No. 88296-61-1) (Sanofi-Aventis), MEM- 1917 (Memory Pharmaceuticals), ND-1251 (Neuro3d), PDE inhibitors from ICOS, PDE IV inhibitors from Memory Pharmaceuticals and CrystalGenomics, Pharmaprojects No. 2742 and 6141 (Sanofi-Aventis), QAD-171 (Novartis), RHC-2963 (CAS No. 76993-12-9 and 76993-14-1), RPR-117658, RPR-122818 derivatives, SR-24870 , and RPR-132294 (Sanofi-Aventis), SK-350 (ln2Gen), stroke therapy agents from deCODE Genetics, TAS-203 (Taiho), tofimilast (CAS No. 185954-27-2) (Pfizer), UK- 371800 (Pfizer), WIN-65579 (CAS No. 158020-82-7) (Sanofi-Aventis), IBFB- 130020 (IBFB Pharma), OPC-6535 (CAS No. 145739-56-6) (Otsuka), a phosphodiesterase III inhibitor, enoximone, a phosphodiesterase IV inhibitor, fosfosal, Atopik (Barrier Therapeutics), triflusal, a phosphodiesterase V inhibitor, and an analogue or derivative thereof.
12729. The method of item 12650 wherein the agent is a PKC inhibitor.
12730. The method of item 12650 wherein the agent is a platelet activating factor antagonist.
12731. The method of item 12650 wherein the agent is a platelet-derived growth factor receptor kinase inhibitor.
12732. The method of item 12650 wherein the agent is a prolyl hydroxylase inhibitor.
12733. The method of item 12650 wherein the agent is a polymorphonuclear neutrophil inhibitor.
12734. The method of item 12650 wherein the agent is a protein kinase B inhibitor.
12735. The method of item 12650 wherein the agent is a protein kinase C stimulant.
12736. The method of item 12650 wherein the agent is a purine nucleoside analogue.
12737. The method of item 12650 wherein the agent is a purinoreceptor P2X antagonist. 12738. The method of item 12650 wherein the agent is a Raf kinase inhibitor.
12739. The method of item 12650 wherein the agent is a reversible inhibitor of ErbB1 and ErbB2.
12740. The method of item 12650 wherein the agent is a ribonucleoside triphosphate reductase inhibitor.
12741. The method of item 12650 wherein the agent is an SDF-1 antagonist.
12742. The method of item 12650 wherein the agent is a sheddase inhibitor.
12743. The method of item 12650 wherein the agent is an SRC inhibitor.
12744. The method of item 12650 wherein the agent is a stromelysin inhibitor.
12745. The method of item 12650 wherein the agent is an Syk kinase inhibitor.
12746. The method of item 12650 wherein the agent is a telomerase inhibitor.
12747. The method of item 12650 wherein the agent is a TGF beta inhibitor selected from the group consisting of pirfenidone (CAS No. 53179-13-8) (MARNAC), tranilast (CAS No. 53902-12-8) (Kissei), IN- 1130 (ln2Gen), mannose-6-phosphate (BTG), TGF-β antagonists from Inflazyme (Pharmaprojects No. 6075), TGF-β antagonists from Sydney, non-industrial source), TGF-β I receptor kinase inhibitors from EIi Lilly, TGF- β> receptor inhibitors from Johnson & Johnson, and an analogue or derivative thereof.
12748. The method of item 12650 wherein the agent is a
TNFα antagonist or TACE inhibitor selected from the group consisting of adalimumab (CAS No. 331731-18-1) (Cambridge Antibody Technology), AGIX-4207 (AtheroGenics), AGT-1 (Advanced Biotherapy), an antiinflammatory from Borean Pharma, Cellzome, or Paradigm Therapeutics, anti-inflammatory vaccine (TNF-alpha kinoid) from Neovacs, humanized anti-TNF antibody or an anti-TNF MAb (CB0006) Celltech (UCB), apratastat (CAS No. 287445-51-0) (Wyeth), BMS-561392 (Bristol-Myers Squibb), BN- 006 (Bone), certolizumab pegol (CAS No. 428863-50-7 or CH-138 (UCB), cilomilast (CAS No. 153259-65-5) (GlaxoSmithKline), CR-1 (Nuada Pharmaceuticals), CRx-119 (CombinatoRx), D-5410 (UCB), dacopafant (CAS No. 125372-33-0) (Sanofi-Aventis), dersalazine (CAS No. 188913-57- 7/188913-58-8) (Uriach), etanercept (CAS No. 185243-69-0) (Amgen), ethyl pyruvate (Critical (Critical Therapeutics), golimumab (CAS No. 476181-74-5) (Johnson & Johnson), hormono-immunotherapy from Ipsen, CDP571 (e.g., Humicade from UCB), IC-485 (ICOS), infliximab (CAS No. 170277-31-3) (Johnson & Johnson), IP-751 (Manhattan Pharmaceuticals), ISIS-104838 (CAS No. 250755-32-9) (ISIS Pharmaceuticals), lenalidomide (CAS No. 191732-72-6) (Celgene), lentinan (CAS No. 37339-90-5) (Ajinomoto), MDL- 201112 (CAS No. 142130-73-2) (Sanofi-Aventis), medroxyprogesterone (CAS No. 520-85-4) (InKine Pharmaceutical), N-acetylcysteine (CAS No. 616-91-1) (Zambon), NBE-P2 (DIREVO Biotech), nerelimomab (CAS No. 162774-06-3) (Chiron), OM-294DP (OM PHARMA), onercept (CAS No. 199685-57-9) (Yeda), PASSTNF-alpha (Verigen), pentoxifylline or oxypentifylline (Sanofi-Aventis), Pharmaprojects No. 4091, 4241 , 4295, or 4488 (Sanofi-Aventis), Pharmaprojects No. 5480 (Amgen), Pharmaprojects No. 6749 (Cengent), pirfenidone (CAS No. 53179-13-8) (MARNAC), RPR- 132294 (Sanofi-Aventis), S5 (F002) (Fulcrum Pharmaceuticals), simvastatin (CAS No. 79902-63-9) (Merck & Co), STA-6292 (Synta Pharmaceuticals), tacrolimus (CAS No. 104987-11-3) (Fujisawa LifeCycle Pharma), talactoferrin alfa (CAS No. 308240-58-6) (Agennix), thalidomide (CAS No. 50-35-1) (Celgene), TNF antagonists form ProStrakan, and Synergen, TNF inhibitors (Amgen), TNF-alpha antagonists from Dynavax Technologies and Jerina AG (Germany), TNF-alpha inhibitors from IBFB Pharma and Xencor (Xencor), torbafylline (CAS No. 105102-21-4) (Sanofi-Aventis), UR-1505 (Uriach), VT-346 (Viron Therapeutics), YSIL6 (Y1S Therapeutics), YSTH2 (Y's Therapeutics), NPI-1302a-3 (Nereus Pharmaceuticals, a TNF antagonist from Jerina AG (Germany), dersalazine, and an analogue or derivative thereof.
12749. The method of item 12650 wherein the agent is a Toll receptor inhibitor.
12750. The method of item 12650 wherein the agent is a tubulin antagonist.
12751. The method of item 12650 wherein the agent is a tyrosine kinase inhibitor selected from the group consisting of SU-011248, SUTENT from Pfizer Inc. (New York, NY), BMS-354825, PN-355 (Paracelsian Pharmaceuticals), AGN-199659 (Allergan), AAL-993 or ABP- 309 (Novartis), adaphostin (NIH), AEE-788 (Novartis), AG-013736 (OSI Pharmaceuticals), AG-13736 (Pfizer), ALT-110 (Alteris Therapeutics), AMG- 706 (Amgen), anticancer MAbs from Xencor, anti-EGFrvlll MAbs from Abgenix, anti-HER2 MAb from Abiogen, AZD-2171 or AZD-9935 (AstraZeneca), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), CEP-5214 (Cephalon), CEP-7055 (Cephalon), cetuximab (ImClone Systems), CHIR-200131 and CHIR-258 (Chiron), CP-547632 (OSI Pharmaceuticals), CP-724714 (Pfizer), CT-301 (Creabilis Therapeutics), D- 69491 (Baxter International), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EGFR/ErbB2 inhibitors from Array BioPharma, erlotinib (CAS No. 183319- 69-9) (OSI Pharmaceuticals), EXEL-2880 (Exelixis), FK-778 (Sanofi- Aventis) , gefitinib (CAS No. 184475-35-2) (AstraZeneca), GW-2286 or GW- 654652 (GlaxoSmithKline), her2/neu antigen from AlphaVax, HER-2/neu inhibitor from Generex, Herzyme (Medipad) (Sirna Therapeutics), HKI-272 (Wyeth), HuMax-EGFr (Genmab), idronoxil (CAS No. 81267-65-4) (Novogen), IGF-1 inhibitors from Ontogen, IMC-11 F8 (ImCIone Systems), kahalalide F (CAS No. 149204-42-2) (PharmaMar), KDR inhibitor from LG Life Sciences, KDR inhibitors from Abbott Laboratories, KDR kinase inhibitors (UCB), Kdr kinase inhibitors from Merck & Co, KRN-633 and KRN- 951 (Kirin Brewery), KSB-102 (Xenova), lapatinib ditosylate (CAS No. 388082-78-8) (GlaxoSmithKline), matuzumab (Merck KGaA), MDX-214 (Medarex), ME-103 (Pharmexa), MED-A300 (Gerolymatos), MNAC-13 (Lay Line Genomics), nimotuzumab (Center of Molecular Immunology), NSC- 330507 or NSC-707545 (NIH), NV-50 (Novogen), OSI-930 (OSI Pharmaceuticals), panitumumab (Abgenix), pelitinib (CAS No. 287933-82-7) (Wyeth), pertuzumab (CAS No. 380610-27-5) (Genentech), Pharmaprojects No. 3985 (Sanofi-Aventis), prostate cancer therapeutics from Sequenom (SQPC35, SQPC36, SQPC90), removab and remoxab (Trion Pharma), RG- 13022 (CAS No. 136831-48-6), RG-13291 (CAS No. 138989-50-1), or RG- 14620 (CAS No. 136831-49-7) (Sanofi-Aventis), RM-6427 (Romark), RNAi breast cancer therapy from Benitec, RP 53801 (CAS No. 125882-88-4) (Sanofi-Aventis), sorafenib tosylate (Bayer), SU-11657 (Pfizer), Tie-2 antagonists from Semaia (Hybrigenics), Tie-2 inhibitors from Ontogen, trastuzumab (CAS No. 180288-69-1) (Genentech), tyrosine kinase inhibitors from Sanofi-Aventis, U3-1287, U3-1565, U3-1784, U3-1800 (U3 Pharma), vatalanib (Novartis), VEGFR-2 kinase inhibitor from Bristol-Myers Squibb, XL-647 (Exelixis), ZD-6474 (AstraZeneca), ZK-CDK (Schering AG), an EGFR tyrosine kinase inhibitor, EKB-569 (Wyeth), and an analogue or derivative thereof.
12752. The method of item 12650 wherein the agent is a VEGF inhibitor.
12753. The method of item 12650 wherein the agent is a vitamin D receptor agonist.
12754. The method of item 12650 wherein the agent is ZD-6474 (an angiogenesis inhibitor).
12755. The method of item 12650 wherein the agent is AP-23573 (an mTOR inhibitor).
12756. The method of item 12650 wherein the agent is synthadotin (a tubulin antagonist).
12757. The method of item 12650 wherein the agent is S-0885 (a collagenase inhibitor).
12758. The method of item 12650 wherein the agent is aplidine (an elongation factor-1 alpha inhibitor).
12759. The method of item 12650 wherein the agent is ixabepilone (an epithilone).
12760. The method of item 12650 wherein the agent is IDN-5390 (an angiogenesis inhibitor). 12761. The method of item 12650 wherein the agent is SB-2723005 (an angiogenesis inhibitor).
12762. The method of item 12650 wherein the agent is ABT-518 (an angiogenesis inhibitor).
12763. The method of item 12650 wherein the agent is combretastatin (an angiogenesis inhibitor).
12764. The method of item 12650 wherein the agent is anecortave acetate (an angiogenesis inhibitor).
12765. The method of item 12650 wherein the agent is SB-715992 (a kinesin antagonist).
12766. The method of item 12650 wherein the agent is temsirolimus (an mTOR inhibitor).
12767. The method of item 12650 wherein the agent is adalimumab (a TNFα antagonist).
12768. The method of item 12650, wherein the composition comprises a polymer.
12769. The method of item 12650, wherein the composition comprises a polymeric carrier.
12770. The method of item 12650 wherein the anti- scarring agent inhibits adhesion between the device and a host into which the device is implanted. 12771. The method of item 12650 wherein the device delivers the anti-scarring agent locally to tissue proximate to the device.
12772. The method of item 12650 wherein the device has a coating that comprises the anti-scarring agent.
12773. The method of item 12650, wherein the device has a coating that comprises the agent and is disposed on a surface of the implant.
12774. The method of item 12650, wherein the device has a coating that comprises the agent and directly contacts the implant.
12775. The method of item 12650, wherein the device has a coating that comprises the agent and indirectly contacts the implant.
12776. The method of item 12650, wherein the device has a coating that comprises the agent and partially covers the implant.
12777. The method of item 12650, wherein the device has a coating that comprises the agent and completely covers the implant.
12778. The method of item 12650, wherein the device has a uniform coating.
12779. The method of item 12650, wherein the device has a non-uniform coating.
12780. The method of item 12650, wherein the device has a discontinuous coating. 12781. The method of item 12650, wherein the device has a patterned coating.
12782. The method of item 12650, wherein the device has a coating with a thickness of 100 Dm or less.
12783. The method of item 12650, wherein the device has a coating with a thickness of 10 Dm or less.
12784. The method of item 12650, wherein the device has a coating, and the coating adheres to the surface of the implant upon deployment of the implant.
12785. The method of item 12650, wherein the device has a coating, and wherein the coating is stable at room temperature for a period of 1 year.
12786. The method of item 12650, wherein the device has a coating, and wherein the anti-scarring agent is present in the coating in an amount ranging between about 0.0001% to about 1% by weight.
12787. The method of item 12650, wherein the device has a coating, and wherein the anti-scarring agent is present in the coating in an amount ranging between about 1% to about 10% by weight.
12788. The method of item 12650, wherein the device has a coating, and wherein the anti-scarring agent is present in the coating in an amount ranging between about 10% to about 25% by weight. 12789. The method of item 12650, wherein the device has a coating, and wherein the anti-scarring agent is present in the coating in an amount ranging between about 25% to about 70% by weight.
12790. The method of item 12650, wherein the device has a coating, and wherein the coating further comprises a polymer.
12791. The method of item 12650, wherein the device has a first coating having a first composition and a second coating having a second composition.
12792. The method of item 12650, wherein the device has a first coating having a first composition and a second coating having a second composition, wherein the first composition and the second composition are different.
12793. The method of item 12650, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a copolymer.
12794. The method of item 12650, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a block copolymer.
12795. The method of item 12650, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a random copolymer.
12796. The method of item 12650, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a biodegradable polymer. 12797. The method of item 12650, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a non-biodegradable polymer.
12798. The method of item 12650, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a hydrophilic polymer.
12799. The method of item 12650, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a hydrophobic polymer.
12800. The method of item 12650, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a polymer having hydrophilic domains.
12801. The method of item 12650, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a polymer having hydrophobic domains.
12802. The method of item 12650, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a non-conductive polymer.
12803. The method of item 12650, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises an elastomer.
12804. The method of item 12650, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a hydrogel. 12805. The method of item 12650, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a silicone polymer.
12806. The method of item 12650, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a hydrocarbon polymer.
12807. The method of item 12650, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a styrene-derived polymer.
12808. The method of item 12650, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a butadiene polymer.
12809. The method of item 12650, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a macromer.
12810. The method of item 12650, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a poly(ethylene glycol) polymer.
12811. The method of item 12650 wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises an amorphous polymer.
12812. The method of item 12650, wherein the device comprises a lubricious coating. 12813. The method of item 12650 wherein the anti- scarring agent is located within pores or holes of the device.
12814. The method of item 12650 wherein the anti- scarring agent is located within a channel, lumen, or divet of the device.
12815. The method of item 12650, wherein the device comprises a second pharmaceutically active agent.
12816. The method of item 12650 wherein the device comprises an anti-inflammatory agent.
12817. The method of item 12650 wherein the device comprises an agent that inhibits infection.
12818. The method of item 12650 wherein the device comprises an agent that inhibits infection, and wherein the agent is an anthracycline.
12819. The method of item 12650 wherein the device comprises an agent that inhibits infection, and wherein the agent is doxorubicin.
12820. The method of item 12650 wherein the device comprises an agent that inhibits infection, and wherein the agent is mitoxantrone.
12821. The method of item 12650 wherein the device comprises an agent that inhibits infection, and wherein the agent is a fluoropyrimidine. 12822. The method of item 12650 wherein the device comprises an agent that inhibits infection, and wherein the agent is 5- fluorouracil (5-FU).
12823. The method of item 12650 wherein the device comprises an agent that inhibits infection, and wherein the agent is a folic acid antagonist.
12824. The method of item 12650 wherein the device comprises an agent that inhibits infection, and wherein the agent is methotrexate.
12825. The method of item 12650 wherein the device comprises an agent that inhibits infection, and wherein the agent is a podophylotoxin.
12826. The method of item 12650 wherein the device comprises an agent that inhibits infection, and wherein the agent is etoposide.
12827. The method of item 12650 wherein the device comprises an agent that inhibits infection, and wherein the agent is a camptothecin.
12828. The method of item 12650 wherein the device comprises an agent that inhibits infection, and wherein the agent is a hydroxyurea.
12829. The method of item 12650 wherein the device comprises an agent that inhibits infection, and wherein the agent is a platinum complex. 12830. The method of item 12650 wherein the device comprises an agent that inhibits infection, and wherein the agent is cisplatin.
12831. The method of item 12650, further comprising an anti-thrombotic agent.
12832. The method of item 12650 wherein the device comprises a visualization agent.
12833. The method of item 12650 wherein the device comprises a visualization agent, wherein the visualization agent is a radiopaque material, and wherein the radiopaque material comprises a metal, a halogenated compound, or a barium containing compound.
12834. The method of item 12650 wherein the device comprises a visualization agent, wherein the visualization agent is a radiopaque material, and wherein the radiopaque material comprises barium, tantalum, or technetium.
12835. The method of item 12650 wherein the device comprises a visualization agent, and wherein the visualization agent is a MRI responsive material.
12836. The method of item 12650 wherein the device comprises a visualization agent, and wherein the visualization agent comprises a gadolinium chelate.
12837. The method of item 12650 wherein the device comprises a visualization agent, and wherein the visualization agent comprises iron, magnesium, manganese, copper, or chromium. 12838. The method of item 12650 wherein the device comprises a visualization agent, and wherein the visualization agent comprises an iron oxide compound.
12839. The method of item 12650 wherein the device comprises a visualization agent, and wherein the visualization agent comprises a dye, pigment, or colorant.
12840. The method of item 12650 wherein the device comprises an echogenic material.
12841. The method of item 12650 wherein the device comprises an echogenic material, and wherein the echogenic material is in the form of a coating.
12842. The method of item 12650 wherein the device is sterile.
12843. The method of item 12650 wherein the anti- scarring agent is released into tissue in the vicinity of the device after deployment of the device.
12844. The method of item 12650 wherein the anti- scarring agent is released into tissue in the vicinity of the device after deployment of the device, and wherein the tissue is connective tissue.
12845. The method of item 12650 wherein the anti- scarring agent is released into tissue in the vicinity of the device after deployment of the device, and wherein the tissue is muscle tissue. 12846. The method of item 12650 wherein the anti- scarring agent is released into tissue in the vicinity of the device after deployment of the device, and wherein the tissue is nerve tissue.
12847. The method of item 12650 wherein the anti- scarring agent is released into tissue in the vicinity of the device after deployment of the device, and wherein the tissue is epithelium tissue.
12848. The method of item 12650 wherein the anti- scarring agent is released in effective concentrations from the device over a period ranging from the time of deployment of the device to about 1 year.
12849. The method of item 12650 wherein the anti- scarring agent is released in effective concentrations from the device over a period ranging from about 1 month to 6 months.
12850. The method of item 12650 wherein the anti- scarring agent is released in effective concentrations from the device over a period ranging from about 1 - 90 days.
12851. The method of item 12650 wherein the anti- scarring agent is released in effective concentrations from the device at a constant rate.
12852. The method of item 12650 wherein the anti- scarring agent is released in effective concentrations from the device at an increasing rate.
12853. The method of item 12650 wherein the anti- scarring agent is released in effective concentrations from the device at a decreasing rate. 12854. The method of item 12650 wherein the anti- scarring agent is released in effective concentrations from the composition comprising the anti-scarring agent by diffusion over a period ranging from the time of deployment of the device to about 90 days.
12855. The method of item 12650 wherein the anti- scarring agent is released in effective concentrations from the composition comprising the anti-scarring agent by erosion of the composition over a period ranging from the time of deployment of the device to about 90 days.
12856. The method of item 12650 wherein the device comprises about 0.01 Dg to about 10 Dg of the anti-scarring agent.
12857. The method of item 12650 wherein the device comprises about 10 Dg to about 10 mg of the anti-scarring agent.
12858. The method of item 12650 wherein the device comprises about 10 mg to about 250 mg of the anti-scarring agent.
12859. The method of item 12650 wherein the device comprises about 250 mg to about 1000 mg of the anti-scarring agent.
12860. The method of item 12650 wherein the device comprises about 1000 mg to about 2500 mg of the anti-scarring agent.
12861. The method of item 12650 wherein a surface of the device comprises less than 0.01 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied. 12862. The method of item 12650 wherein a surface of the device comprises about 0.01 Dg to about 1 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
12863. The method of item 12650 wherein a surface of the device comprises about 1 Dg to about 10 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
12864. The method of item 12650 wherein a surface of the device comprises about 10 Dg to about 250 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
12865. The method of item 12650 wherein a surface of the device comprises about 250 Dg to about 1000 Dg of the anti-scarring agent of anti-scarring agent per mm2 of device surface to which the anti- scarring agent is applied.
12866. The method of item 12650 wherein a surface of the device comprises about 1000 Dg to about 2500 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
12867. The method of item 12650 wherein the combining is performed by direct affixing the agent or the composition to the implant.
12868. The method of item 12650 wherein the combining is performed by spraying the agent or the component onto the implant. 12869. The method of item 12650 wherein the combining is performed by electrospraying the agent or the composition onto the implant.
12870. The method of item 12650 wherein the combining is performed by dipping the implant into a solution comprising the agent or the composition.
12871. The method of item 12650 wherein the combining is performed by covalently attaching the agent or the composition to the implant.
12872. The method of item 12650 wherein the combining is performed by non-covalently attaching the agent or the composition to the implant.
12873. The method of item 12650 wherein the combining is performed by coating the implant with a substance that contains the agent or the composition.
12874. The method of item 12650 wherein the combining is performed by coating the implant with a substance that absorbs the agent.
12875. The method of item 12650 wherein the combining is performed by interweaving a thread composed of, or coated with, the agent or the composition.
12876. The method of item 12650 wherein the combining is performed by covering all the implant with a sleeve that contains the agent or the composition. 12877. The method of item 12650 wherein the combining is performed by covering a portion of the implant with a sleeve that contains the agent or the composition.
12878. The method of item 12650 wherein the combining is performed by covering all the implant with a cover that contains the agent or the composition.
12879. The method of item 12650 wherein the combining is performed by covering a portion of the implant with a cover that contains the agent or the composition.
12880. The method of item 12650 wherein the combining is performed by covering all the implant with an electrospun fabric that contains the agent or the composition.
12881. The method of item 12650 wherein the combining is performed by covering a portion of the implant with an electrospun fabric that contains the agent or the composition.
12882. The method of item 12650 wherein the combining is performed by covering all the implant with a mesh that contains the agent or the composition.
12883. The method of item 12650 wherein the combining is performed by covering a portion of the implant with a mesh that contains the agent or the composition.
12884. The method of item 12650 wherein the combining is performed by constructing all the implant with the agent or the composition. 12885. The method of item 12650 wherein the combining is performed by constructing a portion of the implant with the agent or the composition.
12886. The method of item 12650 wherein the combining is performed by impregnating the implant with the agent or the composition.
12887. The method of item 12650 wherein the combining is performed by constructing all of the implant from a degradable polymer that releases the agent.
12888. The method of item 12650 wherein the combining is performed by constructing a portion of the implant from a degradable polymer that releases the agent.
12889. The method of item 12650 wherein the combining is performed by dipping the implant into a solution that comprise the agent and an inert solvent for the implant.
12890. The method of item 12650 wherein the combining is performed by dipping the implant into a solution that comprises the agent and a solvent that will swell the implant.
12891. The method of item 12650 wherein the combining is performed by dipping the implant into a solution that comprises the agent and a solvent that will dissolve the implant.
12892. The method of item 12650 wherein the combining is performed by dipping the implant into a solution that comprises the agent, a polymer and an inert solvent for the implant. 12893. The method of item 12650 wherein the combining is performed by dipping the implant into a solution that comprises the agent, a polymer and a solvent that will swell the implant.
12894. The method of item 12650 wherein the combining is performed by dipping the implant into a solution that comprises the agent, a polymer and a solvent that will dissolve the implant.
12895. The method of item 12650 wherein the combining is performed by spraying the implant into a solution that comprises the agent and an inert solvent for the implant.
12896. The method of item 12650 wherein the combining is performed by spraying the implant into a solution that comprises the agent and a solvent that will swell the implant.
12897. The method of item 12650 wherein the combining is performed by spraying the implant into a solution that comprises the agent and a solvent that will dissolve the implant.
12898. The method of item 12650 wherein the combining is performed by spraying the implant into a solution that comprises the agent, a polymer and an inert solvent for the implant.
12899. The method of item 12650 wherein the combining is performed by spraying the implant into a solution that comprises the agent, a polymer and a solvent that will swell the implant.
12900. The method of item 12650 wherein the combining is performed by spraying the implant into a solution that comprises the agent, a polymer and a solvent that will dissolve the implant. 12901. A method as in any one of items 12650-12900, wherein the device is a grommet shaped tube.
12902. A method as in any one of items 12650-12900, wherein the device is a T-tube.
12903. A method as in any one of items 12650-12900, wherein the device is a tympanostomy tube.
12904. A method as in any one of items 12650-12900, wherein the device is a drain tube.
12905. A method as in any one of items 12650-12900, wherein the device is a tympanic tube.
12906. A method as in any one of items 12650-12900, wherein the device is an otological tube.
12907. A method as in any one of items 12650-12900, wherein the device is a myringotomy tube.
12908. A method as in any one of items 12650-12900, wherein the device is an artificial Eustachian tube.
12909. A method as in any one of items 12650-12900, wherein the device is an Eustachian tube prosthesis.
12910. A method as in any one of items 12650-12900, wherein the device is an Eustachian stent. 12911. A method of making a medical device comprising: combining an intraocular implant and an anti-scarring agent or a composition comprising an anti-scarring agent, wherein the agent inhibits scarring between the device and a host into which the device is implanted.
12912. The method of item 12911 wherein the agent is an adensosine A2A receptor antagonist.
12913. The method of item 12911 wherein the agent is an AKT inhibitor.
12914. The method of item 12911 wherein the agent is an alpha 2 integrin antagonist, wherein the alpha 2 integrin antagonist is Pharmaprojects No. 5754 (Merck KgaA).
12915. The method of item 12911 wherein the agent js_ an alpha 4 integrin antagonist.
12916. The method of item 12911 wherein the agent is an alpha 7 nicotinic receptor agonist.
12917. The method of item 12911 wherein the agent is an angiogenesis inhibitor selected from the group consisting of AG-12,958 (Pfizer), ATN-161 (Attenuon LLC), neovastat, an angiogenesis inhibitor from Jerina AG (Germany), NM-3 (Mercian), VGA-1155 (Taisho), FCE-26644 (Pfizer), FCE-26950 (Pfizer), FPMA (Meiji Daries), FFM 11142 (Fujisawa), GGTI-298, GM-1306 (Ligand), GPA-1734 (Novartis), NNC-47-0011 (Novo Nordisk), herbamycin (Nippon Kayaku), lenalidomide (Celegene), IP-10 (NIH), ABT-828 (Abbott), KIN-841 (Tokushima University, Japan), SF-1126 (Semafore Pharmaceuticals), laminin technology (NIH), CHIR-258 (Chiron), NVP-AEW541 (Novartis), NVP-AEW541 (Novartis), Vt16907 (Alchemia), OXI-8007 (Oxigene), EG-3306 (Ark Therapeutics), Maspin (Arriva), ABT- 567 (Abbott), PPI-2458 (Praecis Pharmaceuticals), CC-5079, CC-4089 (Celgene), HIF-1 alpha inhibitors (Xenova), S-247 (Pfizer), AP-23573 (Ariad), AZD-9935 (Astra Zeneca), mebendazole (Introgen Therapeutics), MetAP-2 inhibitors (GlaxoSmithKline), AG-615 (Angiogene Pharmaceuticals), Tie-2 antagonists (Hybrigenics), NC-381, CYC-381, NC-169, NC-219, NC-383, NC-384, NC-407 (Lorus Therapeutics), ATN-224 (Attenuon), ON-01370 (Onconova), Vitronectin antagonists (Amgen), SDX-103 (Salmedix), Vitronectin antagonists (Shire), CHP (Riemser), TEK (Amgen), Anecortave acetate (Alcon), T46.2 (Matrix Therapeutics), HG-2 (Heptagen), TEM antagonists (Genzyme), Oxi-4500 (Oxigene), ATN-161 (Attenuon), WX-293 (Wilex), M-2025 (Metris Therapeutics), Alphastatin (BioActa), YH-16 (Yantai Rongchang), BIBF-1120 (Boehhnger Ingelheim), BAY-57-9352 (Bayer), AS- 1404 (Cancer Research Technology), SC-77964 (Pfizer), glycomimetics (BioTie Therapies), TIE-2 Inhibitors (Ontogen), DIMI, Octamer (Octamer), ABR-215050 (Active Biotech), ABT-518 (Abbott), KDR inhibitors (Abbott), BSF-466895 (Abbott), SCH-221153 (Schering-Plough), DAC.antiangiogenic (ConjuChem), TFPl (EntreMed), AZD-2171 (Astra-Zenaca), CDC-394 (Celgene), LY290293 (EIi Lilly), IDN-5390 (Indena), Kdr Kinase Inhibitors (Merck), CT-113020, CT-116433, CT-116563, CT-31890, CT-32228) (Cell Therapeutics), A-299620 (Abbott), TWEAK Inhibitor (Amgen), VEGF modulators (Johnson and Johnson), Tum-N53, tumstatin (Genzyme), Thios- 1 , Thios-2 (Thios Pharmaceuticals), MV-6401 (Miravant Medical Technologies), Spisulosine (PharmaMar), CEP-7055 (Cephalon), AUV-201 (Auvation), LM-609 (EIi Lilly), SKF-106615 (AnorMED), Oglufanide disodium (Cytran), BW-114 (Pharminox), Calreticulin (NIH), WX-678 (Wilex), SD-7784 (Pfizer), WX-UK1 (Wilex), SH-268 (Schering AG), 2-Me-PGA (Celgene), S- 137 (Pfizer), ZD-6126 (Angiogene Pharmaceuticals), SG-292 (SignalGen), Benefin (Lane Labs), A6, A36 (Angstrom), SB-2723005 (GlaxoSmithKline), SC-7 (Cell Therapeutics), ZEN-014 (AEterna Zentaris), 2-methoxyestradiol (EntreMed), NK-130119 (Nippon Kayaku), CC-10004 (Celgene), AVE- 8062A (Ajinomoto), Tacedinaline (Pfizer), Actinonin (Tokyo Metropolitan Institute of Medical Science), Lenalidomide (Celgene), VGA-1155, BTO-956 (SRI International), ER-68203-00 (Eisai), CT-6685 (UCB), JKC-362 (Phoenix Pharmaceuticals), DMI-3798 (DMI Biosciences, Angiomate (Ipsen), ZD-6474 (AstraZeneca), CEP-5214 (Cephalon), Canstatin (Genzyme), NM-3 (Mercian), Oridigm (MediQuest Therapeutics), Exherin (Adherex), BLS-0597 (Boston Life Sciences), PTC-299 (PTC Therapeutics), NPI-2358 (Nereus Pharmaceuticals), CGP-79787 (Novartis), AEE-788 (Novartis), CKD-732 (Chong Kun Dang), CP-564959 (OSI Pharmaceuticals), CM-101 (CarboMed), CT-2584, CT3501 (Cell Therapeutics), combretastatin and analogues and derivatives thereof (Oxigene), Rebimastat (Bristol-Meyers Squibb), Dextrin 2-sulfate (ML Laboratories), Cilengitide (Merk KGaA), NSC-706704 (Pharminox), KRN- 951 (Kirin Brewery), Ukrain, NSC-631570 (Nowicky Pharma), Tecogalan sodium (Daiichi Pharmaceutical), Tz-93 (Tsumura), TBC-1635 (Encysive Pharmaceuticals), TAN-1120 (Takeda), Semaxanib (Pfizer), BDI-7800 (Biopharmacopae), SD-186, SD-983 (Bristol-Meyers Squibb), SB-223245 (GlaxoSmithKline), SC-236 (Pfizer), RWJ-590973 (Johnson and Johnson), ILX-1850 (Genzyme), SC-68488, S-836 (Pfizer), CG-55069-11 (CuraGen), Ki-23057 (Kirin Brewery), CCX-700 (Chemoentryx), Pegaptanib octasodium (Giled Sciences), ANGIOCOL (available from Biostratum Inc.), or an analogue or derivative thereof.
12918. The method of item 12911 wherein the agent is an apoptosis antagonist.
12919. The method of item 12911 wherein the agent is an apoptosis activator.
12920. The method of item 12911 wherein the agent is a beta 1 integrin antagonist. 12921. The method of item 12911 wherein the agent is a beta tubulin inhibitor.
12922. The method of item 12911 wherein the agent is a blocker of enzyme production in Hepatitis C.
12923. The method of item 12911 wherein the agent is a Bruton's tyrosine kinase inhibitor.
12924. The method of item 12911 wherein the agent is a calcineurin inhibitor.
12925. The method of item 12911 wherein the agent is a caspase 3 inhibitor.
12926. The method of item 12911 wherein the agent is a CC chemokine receptor antagonist.
12927. The method of item 12911 wherein the agent is a cell cycle inhibitor selected from the group consisting of SNS-595 (Sunesis), synthadotin, KRX-0403, and an analogue or derivative thereof.
12928. The method of item 12911 wherein the agent is a cathepsin B inhibitor.
12929. The method of item 12911 wherein the agent is a cathepsin K inhibitor, wherein the cathepsin K inhibitor is 462795 (GlaxoSmithKline), INPL-022-D6 (Amura Therapeutics), or an analogue or derivative thereof. 12930. The method of item 12911 wherein the agent is a cathepsin L inhibitor.
12931. The method of item 12911 wherein the agent is a CD40 antagonist.
12932. The method of item 12911 wherein the agent is a chemokine receptor agonist.
12933. The method of item 12911 wherein the agent is a chymase inhibitor.
12934. The method of item 12911 wherein the agent is a collagenase antagonist.
12935. The method of item 12911 wherein the agent is a CXCR antagonist.
12936. The method of item 12911 wherein the agent is a cyclin dependent kinase inhibitor selected from the group consisting of a CDK-1 inhibitor, a CDK-2 inhibitor, a CDK- 4 inhibitor, a CDK-6 inhibitor, a CAK1 inhibitor from GPC Biotech and Bristol-Myers Squibb, RGB-286199 (GPC Biotech), an anticancer agent from Astex Technology, a CAK1 inhibitor from GPC Biotech, a CDK inhibitor from Sanofi-Aventis, a CDK1/CDK2 inhibitor from Amgen, a CDK2 inhibitor from SUGEN-2 (Pfizer), a hearing loss therapy agent (Sound Pharmaceuticals), PD-0332991 (Pfizer), RGB-286199 (GPC Biotech), Ro-0505124 (Hoffmann-La Roche), a Ser/Thr kinase inhibitor from Lilly (EIi Lilly), CVT-2584 (CAS No. 199986-75- 9) (CV Therapeutics), and an analogue or derivative thereof. 12937. The method of item 12911 wherein the agent is a cyclooxygenase 1 inhibitor.
12938. The method of item 12911 wherein the agent is a DHFR inhibitor.
12939. The method of item 12911 wherein the agent is a dual integrin inhibitor.
12940. The method of item 12911 wherein the agent is an elastase inhibitor.
12941. The method of item 12911 wherein the agent is an elongation factor-1 alpha inhibitor.
12942. The method of item 12911 wherein the agent is . an endothelial growth factor antagonist.
12943. The method of item 12911 wherein the agent is an endothelial growth factor receptor kinase inhibitor selected from the group consisting of sorafenib tosylate (Bayer), AAL-993 (Novartis), ABP-309 (Novartis), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), E- 7080 (Eisai), EG-3306 (Ark Therapeutics), EXEL-2880 (Exelixis), GW- 654652 (GlaxoSmithKline), a KDR inhibitor from LG Life Sciences, CT-6685 and CT-6729 (UCB), KRN-633 and KRN-951 (Kirin Brewery), OSI-930 (OSI Pharmaceuticals), SP-5.2 (Supratek Pharma), SU-11657 (Pfizer), a Tie-2 antagonist (Hybrigenics), a VEGFR-2 kinase inhibitor (Bristol-Myers Squibb), XL-647 (Exelixis), a KDR inhibitor from Abbott Laboratories, sorafenib tosylate, and an analogue or derivative thereof. 12944. The method of item 12911 wherein the agent is an endotoxin antagonist.
12945. The method of item 12911 wherein the agent is an epothilone and tubulin binder.
12946. The method of item 12911 wherein the agent is an estrogen receptor antagonist.
12947. The method of item 12911 wherein the agent is an FGF inhibitor.
12948. The method of item 12911 wherein the agent is a farnexyl transferase inhibitor.
12949. The method of item 12911 wherein the agent is farnesyltransferase inhibitor selected from the group of A-197574 (Abbott), a famesyltransferase inhibitor from Servier, FPTIII (Strathclyde Institute for Drug R)1 LB-42908 (LG Life Sciences), Pharmaprojects No. 5063 (Genzyme), Pharmaprojects No. 5597 (Ipsen), Yissum Project No. B-1055 (Yissum), and an analogue or derivative thereof.
12950. The method of item 12911 wherein the agent is an FLT-3 kinase inhibitor.
12951. The method of item 12911 wherein the agent is an FGF receptor kinase inhibitor.
12952. The method of item 12911 wherein the agent is a fibrinogen antagonist selected from the group consisting of AUV-201 (Auvation), MG-13926 (Sanofi-Aventis), plasminogen activator (CAS No. 105913-11-9) (from Sanofi-Aventis or UCB), plasminogen activator-2 (tPA- 2) (Sanofi-Aventis), pro-urokinase (CAS No. 82657-92-9) (Sanofi-Aventis), and an analogue or derivative thereof.
12953. The method of item 12911 wherein the agent is a heat shock protein 90 antagonist selected from the group consisting of SRN- 005 (Sirenade), geldanamycin, NSC-33050 (17-allylaminogeldanamycin; 17- AAG), 17-dimethylaminoethylamino-17-demethoxy-geldanamycin (17- DMAG), rifabutin (rifamycin XIV, 1',4-didehydro-1-deoxy-1,4-dihydro-5l-(2- methylpropyl)-1-oxo-), and an analogue or derivative thereof.
12954. The method of item 12911 wherein the agent is a histone deacetylase inhibitor.
12955. The method of item 12911 wherein the agent is an HMGCoA reductase inhibitor selected from the group consisting of an atherosclerosis therapeutic from Lipid Sciences, ATI-16000 (ARYx Therapeutics), KS-01-019 (Kos Pharmaceuticals), Pharmaprojects No. 2197 (Sanofi-Aventi), RP 61969 (Sanofi-Aventis), and an analogue or derivative thereof.
12956. The method of item 12911 wherein the agent is an ICAM inhibitor.
12957. The method of item 12911 wherein the agent is an IL, ICE and IRAK antagonist, wherein the antagonist is a CJ-14877, CP- 424174 (Pfizer), NF-61 (Negma-Lerads), and an analogue or derivative thereof.
12958. The method of item 12911 wherein the agent is an IL-2 inhibitor. 12959. The method of item 12911 wherein the agent is an immunosuppressant selected from the group consisting of teriflunomide (Sanofi Aventis), chlorsulfaquinoxaione (NSC-339004), chlorsulfaquinoxalone sulfate, CS-712 (Sankyo), ismomultin alfa (CAS No. 457913-93-8) (Akzo Nobel), antiallergics from GenPat77, antiinflammatories or AT-005 (Androclus Therapeutics), autoimmune disease therapeutics from EpiVax, BN-007 (Bone), budesonide (CAS No. 51333-22- 3) (MAP Pharmaceuticals), CO-14 (Genzyme), edratide (CAS No. 433922- 67-9) (Teva), EP-314 (Enanta), eprovafen (CAS No. 101335-99-3) (Sanofi- Aventis), HWA-131 (CAS No. 118788-41-3) (Sanofi-Aventis), immunomodulators from MerLion Pharmaceuticals, immunosuppressives from Alchemia, IPL-12 (Iπflazyme), MDL-9563 (CAS No. 27086-86-8) (Sanofi-Aventis), Pharmaprojects No. 2330 (Sanofi-Aventis), Pharmaprojects No. 6426 (Abgenix), PXS-25 (Pharmaxis), rosmarinic acid (CAS No. 20283-92-5) (Sanofi-Aventis), RP 42927 or RP 54745 (CAS No. 135330-08-4) (Sanofi-Aventis), SGN-35 (Seattle Genetics), ST-1959 (Sigma-Tau), type I diabetes therapy from SYNX Pharma, UNIL-88 (Debiopharm), VP-025 (Vasogen), VR-694 (Vectura), PRTX-001 (Protalex), and an analogue or derivative thereof.
12960. The method of item 12911 wherein the agent is an IMPDH (inosine monophosphate).
12961. The method of item 12911 wherein the agent is an integrin antagonist.
12962. The method of item 12911 wherein the agent is an interleukin antagonist.
12963. The method of item 12911 wherein the agent is an inhibitor of type III receptor tyrosine kinase. 12964. The method of item 12911 wherein the agent is an irreversible inhibitor of enzyme methionine aminopeptidase type 2.
12965. The method of item 12911 wherein the agent is an isozyme selective delta protein kinase C inhibitor.
12966. The method of item 12911 wherein the agent a JAK3 enzyme inhibitor.
12967. The method of item 12911 wherein the agent is a
JNK inhibitor.
12968. The method of item 12911 wherein the agent is a kinase inhibitor.
12969. The method of item 12911 wherein the agent is kinesin antagonist.
12970. The method of item 12911 wherein the agent is a kinesin antagonist.
12971. The method of item 12911 wherein the agent is a leukotriene inhibitor and antagonist selected from the group consisting of ambicromil (CAS No. 58805-38-2) (Sanofi-Aventis), amelubant (CAS No. 346735-24-8) (Boehringer Ingelheim), DW-1141 (Dong Wha), ebselen (Daiichi Pharmaceutical), ibudilast (Kyorin), leucotriene inhibitors from Sanofi-Aventis, lymphotoxin-beta receptor (LT-D) from Biogen Idee, Pharmaprojects No. 1535 and 2728 (CAS No. 119340-33-9) (Sanofi- Aventis), R-112 (Rigel), Rev-5367 (CAS No. 92532-05-3) (Sanofi-Aventis), RG-14893 (CAS No. 141835-49-6) (Sanofi-Aventis), RG-5901-A (CAS No. 101910-24-1), 92532-23-5, RP 66153 (CAS No. 142422-79-5), RP 66364 (CAS No.186916-92-5), RP 69698 (CAS No. 141748-00-7) (Sanofi-Aventis), SC-411930 (Pfizer), SC-41930 (CAS No. 120072-59-5) (Pfizer), SC-50605 (CAS No. 138828-39-4) (Pfizer), SC-51146 (CAS No. 141059-52-1), SC- 53228 (CAS No. 153633-01-3) (Pfizer), spaglumic acid (ZY-15106) (CAS No.3106-85-2), 80619-64-3 (Novartis), tipredane (CAS No. 85197-77-9) (Bristol-Myers Squibb), U-75302 (CAS No. 119477-85-9) (Pfizer), and analogue or derivative thereof.
12972. The method of item 12911 wherein the agent is an MAP kinase inhibitor.
12973. The method of item 12911 wherein the agent is a matrix metalloproteinase inhibitor.
12974. The method of item 12911 wherein the agent is an MCP-CCR2 inhibitor.
12975. The method of item 12911 wherein the agent is an mTOR inhibitor.
12976. The method of item 12911 wherein the agent is an mTOR kinase inhibitor.
12977. The method of item 12911 wherein the agent is a microtubule inhibitor selected from the group consisting of antibody- maytansinoid conjugates from Biogen Idee, colchicines (MantiCore Pharmaceuticals), anticancer immunoconjugates from Johnson & Johnson, DIME from Octamer, gni-1f (GNI), huC242-DM4, huMy9-6-DM1 (ImmunoGen), IDN-5404 (Indena), IMO-098, IMOderm (Imotep), mebendazole (Introgen Therapeutics), microtubule poisons from Cambridge Enterprise, paclitaxel such as LOTAX from Aphios (CAS No. 33069-62-4), Genexol-PM from Samyang, Pharmaprojects No. 6383 (Tapestry Pharmaceuticals), RPR-112378 (Sanofi-Aventis), SGN-75 (Seattle Genetics), SPL-7435 (Starpharma), SSR-250411 (Sanofi-Aventis), trastuzumab-DM1 (Genentech), vinorelbine, and an analogue or derivative thereof.
12978. The method of item 12911 wherein the agent is an MIF inhibitor.
12979. The method of item 12911 wherein the agent is an MMP inhibitor.
12980. The method of item 12911 wherein the agent is a neurokinin (NK) antagonist selected from the group consisting of anthrotainin (CAS No. 148084-40-6) (Sanofi-Aventis), an IBS thereapeutic from ArQuIe, MDL-105212A (CAS No. 167261-60-1) (Ssanofi-Aventis), Pharmaprojects No. 2744, 3258 (CAS No. 139167-47-8) 4006, 4201 , or 5986 (Sanofi-Aventis), RP 67580 (CAS No. 135911-02-3), SR-144190 (CAS No. 201152-86-5), SSR-240600, SSR-241586 (Sanofi-Aventis), TKA-457 (Novartis), vestipitant mesylate (CAS No. 334476-64-1) (GlaxoSmithKline), Win-64821 (Sanofi-Aventis), PRX-96026 (Predix Pharmaceuticals), and an analogue or derivative thereof.
12981. The method of item 12911 wherein the agent is an NF kappa B inhibitor selected from the group consisting of AVE-0545 or AVE-0547 (Sanofi-Aventis), bortezomib (CAS No. 179324-69-7) (Millennium Pharmaceuticals), dexanabinol (CAS No. 112924-45-5) (Pharmos), dexlipotam (Viatris), Pharmaprojects No. 6283 (INDRA) (OXiGENE), IPL- 576092 (CAS No. 137571-30-3) (Inflazyme), NFKB decoy (Corgentech), NFKB decoy oligo (AnGes MG), NFKB's from Ariad, osteoporosis treatments or S5 (F005) from Fulcrum Pharmaceuticals, P61 (Phytopharm), R-flurbiprofen (CAS No. 5104-49-4) (Encore Pharmaceuticals), and an analogue or derivative thereof.
12982. The method of item 12911 wherein the agent is a nitric oxide agonist.
12983. The method of item 12911 wherein the agent is an ornithine decarboxylase inhibitor.
12984. The method of item 12911 wherein the agent is a p38 MAP kinase inhibitor selected from the group consisting of AZD-6703 (AstraZeneca), JX-401 (Jexys Pharmaceuticals), BMS-2 (Bristol-Myers Squibb), a p38 MAP kinase inhibitor from Novartis, a p38-alpha MAP kinase inhibitor from Amphora, Pharmaprojects No. 5704 (Pharmacopeia), RPR- 200765A (Sanofi-Aventis), SD-282 (Johnson & Johnson), TAK-715 (Takeda), and an analogue or derivative thereof.
12985. The method of item 12911 wherein the agent is a palmitoyl-protein thioesterase inhibitor.
12986. The method of item 12911 wherein the agent is a PDGF receptor kinase inhibitor selected from the group consisting of AAL- 993, AMN-107, or ABP-309 (Novartis), AMG-706 (Amgen), BAY-57-9352 (Bayer), CDP-860 (UCB), E-7080 (Eisai), imatinib (CAS No. 152459-95-5) (Novartis), OSI-930 (OSI Pharmaceuticals), RPR-127963E (Sanofi-Aventis), RWJ-540973 (Johnson & Johnson), sorafenib tosylate (Bayer), SU-11657 (Pfizer), tandutinib (CAS No. 387867-13-2) (Millennium Pharmaceuticals), vatalanib (Novartis), ZK-CDK (Schering AG), and an analogue or derivative thereof. 12987. The method of item 12911 wherein the agent is (-)-halofenate (Metabolex), AMG-131 (Amgen), antidiabetics from Japan Tobacco, AZD-4619, AZD-8450, AZD-8677 (AstraZeneca), DRF-10945, balaglitazone (Dr Reddy's), CS-00088, CS-00098 (Chipscreen Biosciences), E-3030 (Eisai), etaiocib (CAS No. 161172-51-6) (EIi Lilly), GSK-641597 (Ligand), GSK-677954 (GlaxoSmithKline), GW-409544 (Ligand), GW- 590735 (GlaxoSmithKline), K-111 (Hoffmann-La Roche), LY-518674 (EIi Lilly), LY-674 (Ligand), LY-929 (Ligand), MC-3001 , MC-3002 (MaxoCore Pharmaceuticals), metformin HCI + pioglitazone (CAS No. 1115-70-4 and
112529-15-4), ACTOPLUS MET from Andrx), muraglitazar (CAS No. 331741-94-7) (Bristol-Myers Squibb), naveglitazar (Ligand), oleoylethanolamide (Kadmus Pharmaceuticals), ONO-5129, pioglitazone hydrochloride (CAS No. 111025-46-8 and 112529-15-4) (Takeda), PLX-204 (Plexxikon), PPAR agonists from Genfit, PPAR delta agonists from EIi Lilly, PPAR-alpha agonists from CrystalGenomics, PPAR-gamma modulators and PPAR-β modulators from CareX, rosiglitazone maleate (CAS No. 122324- 73-4 or 155141-29-0) (GlaxoSmithKline), rosiglitazone maleate/glimepir (CAS No. 155141-29-0 and 93479-97-1), AVANDARYL, rosiglitazone maleate/metformin extend (CAS No. 155141-29-0 and 657-24-9), AVANDAMET, rosiglitazone maleate+metformin, AVANDAMET (GlaxoSmithKline), tesaglitazar (AstraZeneca), and an analogue or derivative thereof.
12988. The method of item 12911 wherein the agent is a phosphatase inhibitor.
12989. The method of item 12911 wherein the agent is a phosphodiesterase (PDE) inhibitor selected from the group consisting of avanafi! (Tanabe Seiyaku), dasantafil (CAS No. 569351-91-3) (Schering- Plough), A-906119 (CAS No. 134072-58-5), DL-850 (Sanofi-Aventis), GRC- 3015, GRC-3566, GRC-3886 (Glenmark), HWA-153 (CAS No. 56395-66-5) (Sanofi-Aventis), hydroxypumafentrine (Altana), IBFB-130011 , IBFB-14-016, IBFB-140301 , IBFB-150007, IBFB-211913 (IBFB Pharma), L-826141 (Merck & Co), medorinone (CAS No. 88296-61-1) (Sanofi-Aventis), MEM- 1917 (Memory Pharmaceuticals), ND-1251 (Neuro3d), PDE inhibitors from ICOS, PDE IV inhibitors from Memory Pharmaceuticals and CrystalGenomics, Pharmaprojects No. 2742 and 6141 (Sanofi-Aventis), QAD-171 (Novartis), RHC-2963 (CAS No. 76993-12-9 and 76993-14-1), RPFM 17658, RPR-122818 derivatives, SR-24870 , and RPR-132294 (Sanofi-Aventis), SK-350 (ln2Gen), stroke therapy agents from deCODE Genetics, TAS-203 (Taiho), tofimilast (CAS No. 185954-27-2) (Pfizer), UK- 371800 (Pfizer), WIN-65579 (CAS No. 158020-82-7) (Sanofi-Aventis), IBFB- 130020 (IBFB Pharma), OPC-6535 (CAS No. 145739-56-6) (Otsuka), a phosphodiesterase III inhibitor, enoximone, a phosphodiesterase IV inhibitor, fosfosal, Atopik (Barrier Therapeutics), triflusal, a phosphodiesterase V inhibitor, and an analogue or derivative thereof.
12990. The method of item 12911 wherein the agent is a PKC inhibitor.
12991. The method of item 12911 wherein the agent is a platelet activating factor antagonist.
12992. The method of item 12911 wherein the agent is a platelet-derived growth factor receptor kinase inhibitor.
12993. The method of item 12911 wherein the agent is a prolyl hydroxylase inhibitor.
12994. The method of item 12911 wherein the agent is a polymorphonuclear neutrophil inhibitor. 12995. The method of item 12911 wherein the agent is a protein kinase B inhibitor.
12996. The method of item 12911 wherein the agent is a protein kinase C stimulant.
12997. The method of item 12911 wherein the agent is a purine nucleoside analogue.
12998. The method of item 12911 wherein the agent is a purinoreceptor P2X antagonist.
12999. The method of item 12911 wherein the agent is a Raf kinase inhibitor.
13000. The method of item 12911 wherein the agent Js a reversible inhibitor of ErbB1 and ErbB2.
13001. The method of item 12911 wherein the agent is a ribonucleoside triphosphate reductase inhibitor.
13002. The method of item 12911 wherein the agent is an SDF-1 antagonist.
13003. The method of item 12911 wherein the agent is a sheddase inhibitor.
13004. The method of item 12911 wherein the agent is an SRC inhibitor. 13005. The method of item 12911 wherein the agent is a stromelysin inhibitor.
13006. The method of item 12911 wherein the agent is an Syk kinase inhibitor.
13007. The method of item 12911 wherein the agent is a telomerase inhibitor.
13008. The method of item 12911 wherein the agent is a TGF beta inhibitor selected from the group consisting of pirfenidone (CAS No. 53179-13-8) (MARNAC), tranilast (CAS No. 53902-12-8) (Kissei), IN- 1130 (ln2Gen), mannose-6-phosphate (BTG), TGF-β antagonists from Inflazyme (Pharmaprojects No. 6075), TGF-β antagonists from Sydney, non-industrial source), TGF-β I receptor kinase inhibitors from EIi Lilly, TGF- β receptor inhibitors from Johnson & Johnson, and an analogue or derivative thereof.
13009. The method of item 12911 wherein the agent is a TNFα antagonist or TACE inhibitor selected from the group consisting of adalimumab (CAS No. 331731-18-1) (Cambridge Antibody Technology), AGIX-4207 (AtheroGenics), AGT-1 (Advanced Biotherapy), an antiinflammatory from Borean Pharma, Cellzome, or Paradigm Therapeutics, anti-inflammatory vaccine (TNF-alpha kinoid) from Neovacs, humanized anti-TNF antibody or an anti-TNF MAb (CB0006) Celltech (UCB), apratastat (CAS No. 287445-51-0) (Wyeth), BMS-561392 (Bristol-Myers Squibb), BN- 006 (Bone), certolizumab pegol (CAS No. 428863-50-7 or CH-138 (UCB), cilomilast (CAS No. 153259-65-5) (GlaxoSmithKline), CR-1 (Nuada Pharmaceuticals), CRx-119 (CombinatoRx), D-5410 (UCB), dacopafant (CAS No. 125372-33-0) (Sanofi-Aventis), dersalazine (CAS No. 188913-57- 7/188913-58-8) (Uriach), etanercept (CAS No. 185243-69-0) (Amgen), ethyl pyruvate (Critical (Critical Therapeutics), golimumab (CAS No. 476181-74-5) (Johnson & Johnson), hormono-immunotherapy from Ipsen, CDP571 (e.g., Humicade from UCB), IC-485 (ICOS), infliximab (CAS No. 170277-31-3) (Johnson & Johnson), IP-751 (Manhattan Pharmaceuticals), ISIS-104838 (CAS No. 250755-32-9) (ISIS Pharmaceuticals), lenalidomϊde (CAS No. 191732-72-6) (Celgene), lentinan (CAS No. 37339-90-5) (Ajinomoto), MDL- 201112 (CAS No. 142130-73-2) (Sanofi-Aventis), medroxyprogesterone (CAS No. 520-85-4) (InKine Pharmaceutical), N-acetylcysteine (CAS No. 616-91-1) (Zambon), NBE-P2 (DIREVO Biotech), nerelimomab (CAS No. 162774-06-3) (Chiron), OM-294DP (OM PHARMA), onercept (CAS No. 199685-57-9) (Yeda), PASSTNF-alpha (Verigen), pentoxifylline or oxypentifylline (Sanofi-Aventis), Pharmaprojects No. 4091, 4241, 4295, or 4488 (Sanofi-Aventis), Pharmaprojects No. 5480 (Amgen), Pharmaprojects No. 6749 (Cengent), pirfenidone (CAS No. 53179-13-8) (MARNAC), RPR- 132294 (Sanofi-Aventis), S5 (F002) (Fulcrum Pharmaceuticals), simvastatin (CAS No. 79902-63-9) (Merck & Co), STA-6292 (Synta Pharmaceuticals), tacrolimus (CAS No. 104987-11-3) (Fujisawa LifeCycle Pharma), talactoferrin alfa (CAS No. 308240-58-6) (Agennix), thalidomide (CAS No. 50-35-1) (Celgene), TNF antagonists form ProStrakan, and Synergen, TNF inhibitors (Amgen), TNF-alpha antagonists from Dynavax Technologies and Jerina AG (Germany), TNF-alpha inhibitors from IBFB Pharma and Xencor (Xencor), torbafylline (CAS No. 105102-21-4) (Sanofi-Aventis), UR-1505 (Uriach), VT-346 (Viron Therapeutics), YSIL6 (Y's Therapeutics), YSTH2 (Y's Therapeutics), NPI-1302a-3 (Nereus Pharmaceuticals, a TNF antagonist from Jerina AG (Germany), dersalazine, and an analogue or derivative thereof.
13010. The method of item 12911 wherein the agent is a
Toll receptor inhibitor. 13011. The method of item 12911 wherein the agent is a tubulin antagonist.
13012. The method of item 12911 wherein the agent is a tyrosine kinase inhibitor selected from the group consisting of SU-011248, SUTENT from Pfizer Inc. (New York, NY), BMS-354825, PN-355 (Paracelsian Pharmaceuticals), AGN-199659 (Allergan), AAL-993 or ABP- 309 (Novartis), adaphostin (NIH), AEE-788 (Novartis), AG-013736 (OSI Pharmaceuticals), AG-13736 (Pfizer), ALT-110 (Alteris Therapeutics), AMG- 706 (Amgen), anticancer MAbs from Xencor, anti-EGFrvlll MAbs from Abgenix, anti-HER2 MAb from Abiogen, AZD-2171 or AZD-9935 (AstraZeneca), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), CEP-5214 (Cephalon), CEP-7055 (Cephalon), cetuximab (ImClone Systems), CHIR-200131 and CHIR-258 (Chiron), CP-547632 (OSI Pharmaceuticals), CP-724714 (Pfizer), CT-301 (Creabilis Therapeutics), D- 69491 (Baxter International), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EGFR/ErbB2 inhibitors from Array BioPharma, erlotinib (CAS No. 183319- 69-9) (OSI Pharmaceuticals), EXEL-2880 (Exelixis), FK-778 (Sanofi- Aventis) , gefitinib (CAS No. 184475-35-2) (AstraZeneca), GW-2286 or GW- 654652 (GlaxoSmithKline), her2/neu antigen from AlphaVax, HER-2/neu inhibitor from Generex, Herzyme (Medipad) (Sima Therapeutics), HKI-272 (Wyeth), HuMax-EGFr (Genmab), idronoxil (CAS No. 81267-65-4) (Novogen), IGF-1 inhibitors from Ontogen, IMC-11F8 (ImClone Systems), kahalalide F (CAS No. 149204-42-2) (PharmaMar), KDR inhibitor from LG Life Sciences, KDR inhibitors from Abbott Laboratories, KDR kinase inhibitors (UCB), Kdr kinase inhibitors from Merck & Co, KRN-633 and KRN- 951 (Kirin Brewery), KSB-102 (Xenova), lapatinib ditosylate (CAS No. 388082-78-8) (GlaxoSmithKline), matuzumab (Merck KGaA), MDX-214 (Medarex), ME-103 (Pharmexa), MED-A300 (Gerolymatos), MNAC-13 (Lay Line Genomics), nimotuzumab (Center of Molecular Immunology), NSC- 330507 or NSC-707545 (NIH), NV-50 (Novogen), OSI-930 (OSI Pharmaceuticals), panitumumab (Abgenix), pelitinib (CAS No. 287933-82-7) (Wyeth), pertuzumab (CAS No. 380610-27-5) (Genentech), Pharmaprojects No. 3985 (Sanofi-Aventis), prostate cancer therapeutics from Sequenom (SQPC35, SQPC36, SQPC90), removab and remoxab (Trion Pharma), RG- 13022 (CAS No. 136831-48-6), RG-13291 (CAS No. 138989-50-1), or RG- 14620 (CAS No. 136831-49-7) (Sanofi-Aventis), RM-6427 (Romark), RNAi breast cancer therapy from Benitec, RP 53801 (CAS No. 125882-88-4) (Sanofi-Aventis), sorafenib tosylate (Bayer), SU-11657 (Pfizer), Tie-2 antagonists from Semaia (Hybrigenics), Tie-2 inhibitors from Ontogen, trastuzumab (CAS No. 180288-69-1) (Genentech), tyrosine kinase inhibitors from Sanofi-Aventis, U3-1287, U3-1565, U3-1784, U3-1800 (U3 Pharma), vatalanib (Novartis), VEGFR-2 kinase inhibitor from Bristol-Myers Squibb, XL-647 (Exelixis), ZD-6474 (AstraZeneca), ZK-CDK (Schering AG), an EGFR tyrosine kinase inhibitor, EKB-569 (Wyeth), and an analogue or derivative thereof.
13013. The method of item 12911 wherein the agent is a VEGF inhibitor.
13014. The method of item 12911 wherein the agent is a vitamin D receptor agonist.
13015. The method of item 12911 wherein the agent is ZD-6474 (an angiogenesis inhibitor).
13016. The method of item 12911 wherein the agent is AP-23573 (an mTOR inhibitor).
13017. The method of item 12911 wherein the agent is synthadotin (a tubulin antagonist). 13018. The method of item 12911 wherein the agent is S-0885 (a collagenase inhibitor).
13019. The method of item 12911 wherein the agent is aplidine (an elongation factor-1 alpha inhibitor).
13020. The method of item 12911 wherein the agent is ixabepilone (an epithilone).
13021. The method of item 12911 wherein the agent is lDN-5390 (an angiogenesis inhibitor).
13022. The method of item 12911 wherein the agent is SB-2723005 (an angiogenesis inhibitor).
13023. The method of item 12911 wherein the agent is ABT-518 (an angiogenesis inhibitor).
13024. The method of item 12911 wherein the agent is combretastatin (an angiogenesis inhibitor).
13025. The method of item 12911 wherein the agent is anecortave acetate (an angiogenesis inhibitor).
13026. The method of item 12911 wherein the agent is SB-715992 (a kinesin antagonist).
13027. The method of item 12911 wherein the agent is temsirolimus (an mTOR inhibitor). 13028. The method of item 12911 wherein the agent is adalimumab (a TNFα antagonist).
13029. The method of item 12911 , wherein the composition comprises a polymer.
13030. The method of item 12911 , wherein the composition comprises a polymeric carrier.
13031. The method of item 12911 wherein the anti- scarring agent inhibits adhesion between the device and a host into which the device is implanted.
13032. The method of item 12911 wherein the device delivers the anti-scarring agent locally to tissue proximate to the device.
13033. The method of item 12911 wherein the device has a coating that comprises the anti-scarring agent.
13034. The method of item 12911 , wherein the device has a coating that comprises the agent and is disposed on a surface of the implant.
13035. The method of item 12911 , wherein the device has a coating that comprises the agent and directly contacts the implant.
13036. The method of item 12911 , wherein the device has a coating that comprises the agent and indirectly contacts the implant.
13037. The method of item 12911 , wherein the device has a coating that comprises the agent and partially covers the implant. 13038. The method of item 12911 , wherein the device has a coating that comprises the agent and completely covers the implant.
13039. The method of item 12911 , wherein the device has a uniform coating.
13040. The method of item 12911 , wherein the device has a non-uniform coating.
13041. The method of item 12911 , wherein the device has a discontinuous coating.
13042. The method of item 12911 , wherein the device has a patterned coating.
13043. The method of item 12911 , wherejn the device has a coating with a thickness of 100 Dm or less.
13044. The method of item 12911 , wherein the device has a coating with a thickness of 10 Dm or less.
13045. The method of item 12911 , wherein the device has a coating, and the coating adheres to the surface of the implant upon deployment of the implant.
13046. The method of item 12911 , wherein the device has a coating, and wherein the coating is stable at room temperature for a period of 1 year. 13047. The method of item 12911 , wherein the device has a coating, and wherein the anti-scarring agent is present in the coating in an amount ranging between about 0.0001% to about 1% by weight.
13048. The method of item 12911 , wherein the device has a coating, and wherein the anti-scarring agent is present in the coating in an amount ranging between about 1 % to about 10% by weight.
13049. The method of item 12911 , wherein the device has a coating, and wherein the anti-scarring agent is present in the coating in an amount ranging between about 10% to about 25% by weight.
13050. The method of item 12911 , wherein the device has a coating, and wherein the anti-scarring agent is present in the coating in an amount ranging between about 25% to about 70% by weight.
13051. The method of item 12911 , wherein the device has a coating, and wherein the coating further comprises a polymer.
13052. The method of item 12911 , wherein the device has a first coating having a first composition and a second coating having a second composition.
13053. The method of item 12911 , wherein the device has a first coating having a first composition and a second coating having a second composition, wherein the first composition and the second composition are different.
13054. The method of item 12911, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a copolymer. 13055. The method of item 12911 , wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a block copolymer.
13056. The method of item 12911, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a random copolymer.
13057. The method of item 12911 , wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a biodegradable polymer.
13058. The method of item 12911, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a non-biodegradable polymer.
13059. The method of item 12911 , wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a hydrophilic polymer.
13060. The method of item 12911 , wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a hydrophobic polymer.
13061. The method of item 12911 , wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a polymer having hydrophilic domains.
13062. The method of item 12911 , wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a polymer having hydrophobic domains. 13063. The method of item 12911 , wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a non-conductive polymer.
13064. The method of item 12911 , wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises an elastomer.
13065. The method of item 12911 , wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a hydrogel.
13066. The method of item 12911 , wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a silicone polymer.
13067. The method of item 12911 , wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a hydrocarbon polymer.
13068. The method of item 12911 , wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a styrene-derived polymer.
13069. The method of item 12911 , wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a butadiene polymer.
13070. The method of item 12911 , wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a macromer. 13071. The method of item 12911 , wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a poly(ethylene glycol) polymer.
13072. The method of item 12911 wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises an amorphous polymer.
13073. The method of item 12911 , wherein the device comprises a lubricious coating.
13074. The method of item 12911 wherein the anti- scarring agent is located within pores or holes of the device.
13075. The method of item 12911 wherein the anti- scarring agent is located within a channel, lumen, or divet of the device.
13076. The method of item 12911, wherein the device comprises a second pharmaceutically active agent.
13077. The method of item 12911 wherein the device comprises an anti-inflammatory agent.
13078. The method of item 12911 wherein the device comprises an agent that inhibits infection.
13079. The method of item 12911 wherein the device comprises an agent that inhibits infection, and wherein the agent is an anthracycline. 13080. The method of item 12911 wherein the device comprises an agent that inhibits infection, and wherein the agent is doxorubicin.
13081. The method of item 12911 wherein the device comprises an agent that inhibits infection, and wherein the agent is mitoxantrone.
13082. The method of item 12911 wherein the device comprises an agent that inhibits infection, and wherein the agent is a fluoropyrimidine.
13083. The method of item 12911 wherein the device comprises an agent that inhibits infection, and wherein the agent is 5- fluorouracil (5-FU).
13084. The method of item 12911 wherein the device comprises an agent that inhibits infection, and wherein the agent is a folic acid antagonist.
13085. The method of item 12911 wherein the device comprises an agent that inhibits infection, and wherein the agent is methotrexate.
13086. The method of item 12911 wherein the device comprises an agent that inhibits infection, and wherein the agent is a podophylotoxin.
13087. The method of item 12911 wherein the device comprises an agent that inhibits infection, and wherein the agent is etoposide. 13088. The method of item 12911 wherein the device comprises an agent that inhibits infection, and wherein the agent is a camptothecin.
13089. The method of item 12911 wherein the device comprises an agent that inhibits infection, and wherein the agent is a hydroxyurea.
13090. The method of item 12911 wherein the device comprises an agent that inhibits infection, and wherein the agent is a platinum complex.
13091. The method of item 12911 wherein the device comprises an agent that inhibits infection, and wherein the agent is cisplatin.
13092. The method of item 12911, further comprising an anti-thrombotic agent.
13093. The method of item 12911 wherein the device comprises a visualization agent.
13094. The method of item 12911 wherein the device comprises a visualization agent, wherein the visualization agent is a radiopaque material, and wherein the radiopaque material comprises a metal, a halogenated compound, or a barium containing compound.
13095. The method of item 12911 wherein the device comprises a visualization agent, wherein the visualization agent is a radiopaque material, and wherein the radiopaque material comprises barium, tantalum, or technetium. 13096. The method of item 12911 wherein the device comprises a visualization agent, and wherein the visualization agent is a MRI responsive material.
13097. The method of item 12911 wherein the device comprises a visualization agent, and wherein the visualization agent comprises a gadolinium chelate.
13098. The method of item 12911 wherein the device comprises a visualization agent, and wherein the visualization agent comprises iron, magnesium, manganese, copper, or chromium.
13099. The method of item 12911 wherein the device comprises a visualization agent, and wherein the visualization agent comprises an iron oxide compound.
13100. The method of item 12911 wherein the device comprises a visualization agent, and wherein the visualization agent comprises a dye, pigment, or colorant.
13101. The method of item 12911 wherein the device comprises an echogenic material.
13102. The method of item 12911 wherein the device comprises an echogenic material, and wherein the echogenic material is in the form of a coating.
13103. The method of item 12911 wherein the device is sterile. 13104. The method of item 12911 wherein the anti- scarring agent is released into tissue in the vicinity of the device after deployment of the device.
13105. The method of item 12911 wherein the anti- scarring agent is released into tissue in the vicinity of the device after deployment of the device, and wherein the tissue is connective tissue.
13106. The method of item 12911 wherein the anti- scarring agent is released into tissue in the vicinity of the device after deployment of the device, and wherein the tissue is muscle tissue.
13107. The method of item 12911 wherein the anti- scarring agent is released into tissue in the vicinity of the device after deployment of the device, and wherein the tissue is nerve tissue.
13108. The method of item 12911 wherein the anti- scarring agent is released into tissue in the vicinity of the device after deployment of the device, and wherein the tissue is epithelium tissue.
13109. The method of item 12911 wherein the anti- scarring agent is released in effective concentrations from the device over a period ranging from the time of deployment of the device to about 1 year.
13110. The method of item 12911 wherein the anti- scarring agent is released in effective concentrations from the device over a period ranging from about 1 month to 6 months.
13111. The method of item 12911 wherein the anti- scarring agent is released in effective concentrations from the device over a period ranging from about 1 - 90 days. 13112. The method of item 12911 wherein the anti- scarring agent is released in effective concentrations from the device at a constant rate.
13113. The method of item 12911 wherein the anti- scarring agent is released in effective concentrations from the device at an increasing rate.
13114. The method of item 12911 wherein the anti- scarring agent is released in effective concentrations from the device at a decreasing rate.
13115. The method of item 12911 wherein the anti- scarring agent is released in effective concentrations from the composition comprising the anti-scarring agent by diffusion over a period ranging from the time of deployment of the device to about 90 days.
13116. The method of item 12911 wherein the anti- scarring agent is released in effective concentrations from the composition comprising the anti-scarring agent by erosion of the composition over a period ranging from the time of deployment of the device to about 90 days.
13117. The method of item 12911 wherein the device comprises about 0.01 Dg to about 10 Dg of the anti-scarring agent.
13118. The method of item 12911 wherein the device comprises about 10 Dg to about 10 mg of the anti-scarring agent.
13119. The method of item 12911 wherein the device comprises about 10 mg to about 250 mg of the anti-scarring agent. 13120. The method of item 12911 wherein the device comprises about 250 mg to about 1000 mg of the anti-scarring agent.
13121. The method of item 12911 wherein the device comprises about 1000 mg to about 2500 mg of the anti-scarring agent.
13122. The method of item 12911 wherein a surface of the device comprises less than 0.01 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
13123. The method of item 12911 wherein a surface of the device comprises about 0.01 Dg to about 1 Og of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
13124. The method of item 12911 wherein a surface of the device comprises about 1 Dg to about 10 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
13125. The method of item 12911 wherein a surface of the device comprises about 10 Dg to about 250 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
13126. The method of item 12911 wherein a surface of the device comprises about 250 Dg to about 1000 Dg of the anti-scarring agent of anti-scarring agent per mm2 of device surface to which the anti- scarring agent is applied.
13127. The method of item 12911 wherein a surface of the device comprises about 1000 Dg to about 2500 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied. 13128. The method of item 12911 wherein the combining is performed by direct affixing the agent or the composition to the implant.
13129. The method of item 12911 wherein the combining is performed by spraying the agent or the component onto the implant.
13130. The method of item 12911 wherein the combining is performed by electrospraying the agent or the composition onto the implant.
13131. The method of item 12911 wherein the combining is performed by dipping the implant into a solution comprising the agent or the composition.
13132. The method of item 12911 wherein the combining is performed by covalently attaching the agent or the composition to the implant.
13133. The method of item 12911 wherein the combining is performed by non-covalently attaching the agent or the composition to the implant.
13134. The method of item 12911 wherein the combining is performed by coating the implant with a substance that contains the agent or the composition.
13135. The method of item 12911 wherein the combining is performed by coating the implant with a substance that absorbs the agent. 13136. The method of item 12911 wherein the combining is performed by interweaving a thread composed of, or coated with, the agent or the composition.
13137. The method of item 12911 wherein the combining is performed by covering all the implant with a sleeve that contains the agent or the composition.
13138. The method of item 12911 wherein the combining is performed by covering a portion of the implant with a sleeve that contains the agent or the composition.
13139. The method of item 12911 wherein the combining is performed by covering all the implant with a cover that contains the agent or the composition.
13140. The method of item 12911 wherein the combining is performed by covering a portion of the implant with a cover that contains the agent or the composition.
13141. The method of item 12911 wherein the combining is performed by covering all the implant with an electrospun fabric that contains the agent or the composition.
13142. The method of item 12911 wherein the combining is performed by covering a portion of the implant with an electrospun fabric that contains the agent or the composition.
13143. The method of item 12911 wherein the combining is performed by covering all the implant with a mesh that contains the agent or the composition. 13144. The method of item 12911 wherein the combining is performed by covering a portion of the implant with a mesh that contains the agent or the composition.
13145. The method of item 12911 wherein the combining is performed by constructing a!! the implant with the agent or the composition.
13146. The method of item 12911 wherein the combining is performed by constructing a portion of the implant with the agent or the composition.
13147. The method of item 12911 wherein the combining is performed by impregnating the implant with the agent or the composition.
13148. The method of item 12911 wherein the combining is performed by constructing all of the implant from a degradable polymer that releases the agent.
13149. The method of item 12911 wherein the combining is performed by constructing a portion of the implant from a degradable polymer that releases the agent.
13150. The method of item 12911 wherein the combining is performed by dipping the implant into a solution that comprise the agent and an inert solvent for the implant.
13151. The method of item 12911 wherein the combining is performed by dipping the implant into a solution that comprises the agent and a solvent that will swell the implant. 13152. The method of item 12911 wherein the combining is performed by dipping the implant into a solution that comprises the agent and a solvent that will dissolve the implant.
13153. The method of item 12911 wherein the combining is performed by dipping the implant into a solution that comprises the agent, a polymer and an inert solvent for the implant.
13154. The method of item 12911 wherein the combining is performed by dipping the implant into a solution that comprises the agent, a polymer and a solvent that will swell the implant.
13155. The method of item 12911 wherein the combining is performed by dipping the implant into a solution that comprises the agent, a polymer and a solvent that will dissolve the implant.
13156. The method of item 12911 wherein the combining is performed by spraying the implant into a solution that comprises the agent and an inert solvent for the implant.
13157. The method of item 12911 wherein the combining is performed by spraying the implant into a solution that comprises the agent and a solvent that will swell the implant.
13158. The method of item 12911 wherein the combining is performed by spraying the implant into a solution that comprises the agent and a solvent that will dissolve the implant.
13159. The method of item 12911 wherein the combining is performed by spraying the implant into a solution that comprises the agent, a polymer and an inert solvent for the implant. 13160. The method of item 12911 wherein the combining is performed by spraying the implant into a solution that comprises the agent, a polymer and a solvent that will swell the implant.
13161. The method of item 12911 wherein the combining is performed by spraying the implant into a solution that comprises the agent, a polymer and a solvent that will dissolve the implant.
13162. A method as in any one of items 12911-13161 , wherein the device is an intraocular lens device for preventing lens opacification.
13163. A method as in any one of items 12911-13161 , wherein the device is a polymethylmethacrylate intraocular lens.
13164. A method as in any one of items 12911-13161, wherein the device is a silicone intraocular lens.
13165. A method as in any one of items 12911-13161, wherein the device is an achromatic lens.
13166. A method as in any one of items 12911-13161, wherein the device is a pseudophako.
13167. A method as in any one of items 12911-13161 , wherein the device is a phakic lens.
13168. A method as in any one of items 12911-13161, wherein the device is an aphakic lens. 13169. A method as in any one of items 12911-13161 , wherein the device is a multi-focal intraocular lens.
13170. A method as in any one of items 12911-13161 , wherein the device is a hydrophilic and hydrophobic acrylic intraocular lens.
13171. A method as in any one of items 12911-13161 , wherein the device is an intraocular implant.
13172. A method as in any one of items 12911-13161 , wherein the device is an optic lens.
13173. A method as in any one of items 12911-13161 , wherein the device is a rigid gas permeable lens.
13174. A method as in any one of items 12911-13161 , wherein the device is a foldable intraocular lens.
13175. A method as in any one of items 12911-13161 , wherein the device is a rigid intraocular lens.
13176. A method as in any one of items 12911-13161 , wherein the device is a corrective implant for vision impairment.
13177. A method as in any one of items 12911-13161 , wherein the device is an intraocular implant adapted for being used in conjunction with a transplant for the cornea.
13178. A method as in any one of items 12911-13161 , wherein the device is an intraocular implant adapted for being used in conjuction with treatment of secondary cataract after extracapsular cataract extraction.
13179. A method of making a medical device comprising: combining a medical device for treating hypertropic scar or keloid (e.g., an implant) and an anti-scarring agent or a composition comprising an anti-scarring agent, wherein the agent inhibits scarring between the device and a host into which the device is implanted.
13180. The method of item 13179 wherein the agent is an adensosine A2A receptor antagonist.
13181. The method of item 13179 wherein the agent is an AKT inhibitor.
13182. The method of item 13179 wherein the agent is an alpha 2 integrin antagonist, wherein the alpha 2 integrin antagonist is Pharmaprojects No. 5754 (Merck KgaA).
13183. The method of item 13179 wherein the agent is an alpha 4 integrin antagonist.
13184. The method of item 13179 wherein the agent is an alpha 7 nicotinic receptor agonist.
13185. The method of item 13179 wherein the agent is an angiogenesis inhibitor selected from the group consisting of AG-12,958 (Pfizer), ATN-161 (Attenuon LLC), neovastat, an angiogenesis inhibitor from Jerina AG (Germany), NM-3 (Mercian), VGA-1155 (Taisho), FCE-26644 (Pfizer), FCE-26950 (Pfizer), FPMA (Meiji Daries), FR-111142 (Fujisawa), GGTI-298, GM-1306 (Ligand), GPA-1734 (Novartis), NNC-47-0011 (Novo Nordisk), herbamycin (Nippon Kayaku), Ienalidomide (Celegene), IP-10 (NIH), ABT-828 (Abbott), KIN-841 (Tokushima University, Japan), SF-1126 (Semafore Pharmaceuticals), laminin technology (NIH), CHIR-258 (Chiron), NVP-AEW541 (Novartis), NVP-AEW541 (Novartis), Vt16907 (Alchemia), OXI-8007 (Oxigene), EG-3306 (Ark Therapeutics), Maspin (Arriva), ABT- 567 (Abbott), PPI-2458 (Praecis Pharmaceuticals), CC-5079, CC-4089 (Celgene), HIF-1 alpha inhibitors (Xenova), S-247 (Pfizer), AP-23573 (Ariad), AZD-9935 (Astra Zeneca), mebendazole (Introgen Therapeutics), MetAP-2 inhibitors (GlaxoSmithKline), AG-615 (Angiogene Pharmaceuticals), Tie-2 antagonists (Hybrigenics), NC-381 , CYC-381 , NC-169, NC-219, NC-383, NC-384, NC-407 (Lorus Therapeutics), ATN-224 (Attenuon), ON-01370 (Onconova), Vitronectin antagonists (Amgen), SDX-103 (Salmedix), Vitronectin antagonists (Shire), CHP (Riemser), TEK (Amgen), Anecortave acetate (Alcon), T46.2 (Matrix Therapeutics), HG-2 (Heptagen), TEM antagonists (Genzyme), Oxi-4500 (Oxigene), ATN-161 (Attenuon), WX-293 (Wilex), M-2025 (Metris Therapeutics), Alphastatin (BioActa), YH-16 (Yantai Rongchang), BIBF-1120 (Boehringer Ingelheim), BAY-57-9352 (Bayer), AS- 1404 (Cancer Research Technology), SC-77964 (Pfizer), glycomimetics (BioTie Therapies), TIE-2 Inhibitors (Ontogen), DIMI, Octamer (Octamer), ABR-215050 (Active Biotech), ABT-518 (Abbott), KDR inhibitors (Abbott), BSF-466895 (Abbott), SCH-221153 (Schering-Plough), DAC:antiangiogenic (ConjuChem), TFPI (EntreMed), AZD-2171 (Astra-Zenaca), CDC-394 (Celgene), LY290293 (EIi Lilly), IDN-5390 (Indena), Kdr Kinase Inhibitors (Merck), CT-113020, CT-116433, CT-116563, CT-31890, CT-32228) (Cell Therapeutics), A-299620 (Abbott), TWEAK Inhibitor (Amgen), VEGF modulators (Johnson and Johnson), Tum-N53, tumstatin (Genzyme), Thios- 1 , Thios-2 (Thios Pharmaceuticals), MV-6401 (Miravant Medical Technologies), Spisulosine (PharmaMar), CEP-7055 (Cephalon), AUV-201 (Auvation), LM-609 (EIi Lilly), SKF-106615 (AnorMED), Oglufanide disodium (Cytran), BW-114 (Pharminox), Calreticulin (NIH), WX-678 (Wilex), SD-7784 (Pfizer), WX-UK1 (Wilex), SH-268 (Schering AG), 2-Me-PGA (Celgene), S- 137 (Pfizer), ZD-6126 (Angiogene Pharmaceuticals), SG-292 (SignalGen), Benefin (Lane Labs), A6, A36 (Angstrom), SB-2723005 (GlaxoSmithKline), SC-7 (Cell Therapeutics), ZEN-014 (AEterna Zentaris), 2~methoxypestradiol (EntreMed), NK-130119 (Nippon Kayaku), CC-10004 (Celgene), AVE- 8062A (Ajinomoto), Tacedinaline (Pfizer), Actinonin (Tokyo Metropolitan Institute of Medical Science), Lenalidomide (Celgene), VGA-1155, BTO-956 (SRI International), ER-68203-00 (Eisai), CT-6685 (UCB)1 JKC-362 (Phoenix Pharmaceuticals), DMI-3798 (DMI Biosciences, Angiomate (Ipsen), ZD-6474 (AstraZeneca), CEP-5214 (Cephalon), Canstatin (Genzyme), NM-3 (Mercian), Oridigm (MediQuest Therapeutics), Exherin (Adherex), BLS-0597 (Boston Life Sciences), PTC-299 (PTC Therapeutics), NPI-2358 (Nereus Pharmaceuticals), CGP-79787 (Novartis), AEE-788 (Novartis), CKD-732 (Chong Kun Dang), CP-564959 (OSI Pharmaceuticals), CM-101 (CarboMed), CT-2584, CT3501 (Cell Therapeutics), combretastatin and analogues and derivatives thereof (Oxigene), Rebimastat (Bristol-Meyers Squibb), Dextrin 2-sulfate (ML Laboratories), Cilengitide (Merk KGaA), NSC-706704 (Pharminox), KRN- 951 (Kirin Brewery), Ukrain, NSC-631570 (Nowicky Pharma), Tecogalan sodium (Daiichi Pharmaceutical), Tz-93 (Tsumura), TBC-1635 (Encysive Pharmaceuticals), TAN-1120 (Takeda), Semaxanib (Pfizer), BDI-7800 (Biopharmacopae), SD-186, SD-983 (Bristol-Meyers Squibb), SB-223245 (GlaxoSmithKline), SC-236 (Pfizer), RWJ-590973 (Johnson and Johnson), ILX-1850 (Genzyme), SC-68488, S-836 (Pfizer), CG-55069-11 (CuraGen), Ki-23057 (Kirin Brewery), CCX-700 (Chemoentryx), Pegaptanib octasodium (Giled Sciences), ANGIOCOL (available from Biostratum Inc.), or an analogue or derivative thereof.
13186. The method of item 13179 wherein the agent is an apoptosis antagonist. 13187. The method of item 13179 wherein the agent is an apoptosis activator.
13188. The method of item 13179 wherein the agent is a beta 1 integrin antagonist.
13189. The method of item 13179 wherein the agent is a beta tubulin inhibitor.
13190. The method of item 13179 wherein the agent is a blocker of enzyme production in Hepatitis C.
13191. The method of item 13179 wherein the agent is a Bruton's tyrosine kinase inhibitor.
13192. The method of item 13179 wherein the agent is a calcineurin inhibitor.
13193. The method of item 13179 wherein the agent is a caspase 3 inhibitor.
13194. The method of item 13179 wherein the agent is a CC chemokine receptor antagonist.
13195. The method of item 13179 wherein the agent is a cell cycle inhibitor selected from the group consisting of SNS-595 (Sunesis), synthadotin, KRX-0403, and an analogue or derivative thereof.
13196. The method of item 13179 wherein the agent is a cathepsin B inhibitor. 13197. The method of item 13179 wherein the agent is a cathepsin K inhibitor, wherein the cathepsin K inhibitor is 462795 (GlaxoSmithKline), INPL-022-D6 (Amura Therapeutics), or an analogue or derivative thereof.
13198. The method of item 13179 wherein the agent is a cathepsin L inhibitor.
13199. The method of item 13179 wherein the agent is a CD40 antagonist.
13200. The method of item 13179 wherein the agent is a chemokine receptor agonist.
13201. The method of item 13179 wherein the agent is a chymase inhibitor.
13202. The method of item 13179 wherein the agent is a collagenase antagonist.
13203. The method of item 13179 wherein the agent is a CXCR antagonist.
13204. The method of item 13179 wherein the agent is a cyclin dependent kinase inhibitor selected from the group consisting of a CDK-1 inhibitor, a CDK-2 inhibitor, a CDK- 4 inhibitor, a CDK-6 inhibitor, a CAK1 inhibitor from GPC Biotech and Bristol-Myers Squibb, RGB-286199 (GPC Biotech), an anticancer agent from Astex Technology, a CAK1 inhibitor from GPC Biotech, a CDK inhibitor from Sanofi-Aventis, a CDK1/CDK2 inhibitor from Amgen, a CDK2 inhibitor from SUGEN-2 (Pfizer), a hearing loss therapy agent (Sound Pharmaceuticals), PD-0332991 (Pfizer), RGB-286199 (GPC Biotech), Ro-0505124 (Hoffmann-La Roche), a Ser/Thr kinase inhibitor from Lilly (EIi Lilly), CVT-2584 (CAS No. 199986-75- 9) (CV Therapeutics), and an analogue or derivative thereof.
13205. The method of item 13179 wherein the agent is a cyclooxygenase 1 inhibitor.
13206. The method of item 13179 wherein the agent is a DHFR inhibitor.
13207. The method of item 13179 wherein the agent is a dual integrin inhibitor.
13208. The method of item 13179 wherein the agent is an elastase inhibitor.
13209. The method of item 13179 wherein the agent is an elongation factor-1 alpha inhibitor.
13210. The method of item 13179 wherein the agent is an endothelial growth factor antagonist.
13211. The method of item 13179 wherein the agent is an endothelial growth factor receptor kinase inhibitor selected from the group consisting of sorafenib tosylate (Bayer), AAL-993 (Novartis), ABP-309 (Novartis), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), E- 7080 (Eisai), EG-3306 (Ark Therapeutics), EXEL-2880 (Exelixis), GW- 654652 (GlaxoSmithKline), a KDR inhibitor from LG Life Sciences, CT-6685 and CT-6729 (UCB), KRN-633 and KRN-951 (Kirin Brewery), OSI-930 (OSI Pharmaceuticals), SP-5.2 (Supratek Pharma), SLM 1657 (Pfizer), a Tie-2 antagonist (Hybrigenics), a VEGFR-2 kinase inhibitor (Bristol-Myers Squibb), XL-647 (Exelixis), a KDR inhibitor from Abbott Laboratories, sorafenib tosylate, and an analogue or derivative thereof.
13212. The method of item 13179 wherein the agent is an endotoxin antagonist.
13213. The method of item 13179 wherein the agent is an epothiione and tubulin binder.
13214. The method of item 13179 wherein the agent is an estrogen receptor antagonist.
13215. The method of item 13179 wherein the agent is an FGF inhibitor.
13216. The method of item 13179 wherein the agent is a farnexyl transferase inhibitor.
13217. The method of item 13179 wherein the agent is farnesyltransferase inhibitor selected from the group of A-197574 (Abbott), a farnesyltransferase inhibitor from Servier, FPTIH (Strathclyde Institute for Drug R), LB-42908 (LG Life Sciences), Pharmaprojects No. 5063 (Genzyme), Pharmaprojects No. 5597 (Ipsen), Yissum Project No. B-1055 (Yissum), and an analogue or derivative thereof.
13218. The method of item 13179 wherein the agent is an FLT-3 kinase inhibitor.
13219. The method of item 13179 wherein the agent is an FGF receptor kinase inhibitor. 13220. The method of item 13179 wherein the agent is a fibrinogen antagonist selected from the group consisting of AUV-201 (Auvation), MG-13926 (Sanofi-Aventis), plasminogen activator (CAS No. 105913-11-9) (from Sanofi-Aventis or UCB), plasminogen activator-2 (tPA- 2) (Sanofi-Aventis), pro-urokinase (CAS No. 82657-92-9) (Sanofi-Aventis), and an analogue or derivative thereof.
13221. The method of item 13179 wherein the agent is a heat shock protein 90 antagonist selected from the group consisting of SRN- 005 (Sirenade), geldanamycin, NSC-33050 (17-aliylaminogeldanamycin; 17- AAG), 17-dimethylaminoethylamino-17-demethoxy-geldanamycin (17- DMAG), rifabutin (rifamycin XIV, 1\4-didehydro-1-deoxy-1 ,4-dihydro-5'-(2- methylpropyl)-1-oxo-), and an analogue or derivative thereof.
13222. The method of item 13179 wherein the agent is a histone deacetylase inhibitor. _ .
13223. The method of item 13179 wherein the agent is an HMGCoA reductase inhibitor selected from the group consisting of an atherosclerosis therapeutic from Lipid Sciences, ATI-16000 (ARYx Therapeutics), KS-01-019 (Kos Pharmaceuticals), Pharmaprojects No. 2197 (Sanofi-Aventi), RP 61969 (Sanofi-Aventis), and an analogue or derivative thereof.
13224. The method of item 13179 wherein the agent is an ICAM inhibitor.
13225. The method of item 13179 wherein the agent is an IL, ICE and IRAK antagonist, wherein the antagonist is a CJ-14877, CP- 424174 (Pfizer), NF-61 (Negma-Lerads), and an analogue or derivative thereof. 13226. The method of item 13179 wherein the agent is an IL-2 inhibitor.
13227. The method of item 13179 wherein the agent is an immunosuppressant selected from the group consisting of teriflunomide (Sanofi Aventis), chlorsulfaquinoxalone (NSC-339004), chlorsulfaquinoxalone sulfate, CS-712 (Sankyo), ismomultin alfa (CAS No. 457913-93-8) (Akzo Nobel), antiallergics from GenPat77, antiinflammatories or AT-005 (Androclus Therapeutics), autoimmune disease therapeutics from EpiVax, BN-007 (Bone), budesonide (CAS No. 51333-22- 3) (MAP Pharmaceuticals), CO-14 (Genzyme), edratide (CAS No. 433922- 67-9) (Teva), EP-314 (Enanta), eprovafen (CAS No. 101335-99-3) (Sanofi- Aventis), HWA-131 (CAS No. 118788-41-3) (Sanofi-Aventis), immunomodulators from MerLion Pharmaceuticals, immunosuppressives from Alchemia, IPL-12 (Inflazyme), MDL-9563 (CAS No. 27086-86-8) (Sanofi-Aventis), Pharmaprojects No. 2330 (Sanofi-Aventis), .- Pharmaprojects No. 6426 (Abgenix), PXS-25 (Pharmaxis), rosmarinic acid (CAS No. 20283-92-5) (Sanofi-Aventis), RP 42927 or RP 54745 (CAS No. 135330-08-4) (Sanofi-Aventis), SGN-35 (Seattle Genetics), ST-1959 (Sigma-Tau), type I diabetes therapy from SYNX Pharma, UNIL-88 (Debiopharm), VP-025 (Vasogen), VR-694 (Vectura), PRTX-001 (Protalex), and an analogue or derivative thereof.
13228. The method of item 13179 wherein the agent is an IMPDH (inosine monophosphate).
13229. The method of item 13179 wherein the agent is an integrin antagonist.
13230. The method of item 13179 wherein the agent is an interleukin antagonist. 13231. The method of item 13179 wherein the agent is an inhibitor of type III receptor tyrosine kinase.
13232. The method of item 13179 wherein the agent is an irreversible inhibitor of enzyme methionine aminopeptidase type 2.
13233. The method of item 13179 wherein the agent is an isozyme selective delta protein kinase C inhibitor.
13234. The method of item 13179 wherein the agent a JAK3 enzyme inhibitor.
13235. The method of item 13179 wherein the agent is a JNK inhibitor.
13236. The method of item 13179 wherein the agent is a kinase inhibitor.
13237. The method of item 13179 wherein the agent is kinesin antagonist.
13238. The method of item 13179 wherein the agent is a kinesin antagonist.
13239. The method of item 13179 wherein the agent is a leukotriene inhibitor and antagonist selected from the group consisting of ambicromil (CAS No. 58805-38-2) (Sanofi-Aventis), amelubant (CAS No. 346735-24-8) (Boehringer Ingelheim), DW-1141 (Dong Wha), ebselen (Daiichi Pharmaceutical), ibudilast (Kyorin), leucotriene inhibitors from Sanofi-Aventis, lymphotoxin-beta receptor (LT-D) from Biogen Idee, Pharmaprojects No. 1535 and 2728 (CAS No. 119340-33-9) (Sanofi- Aventis), R-112 (Rigel), Rev-5367 (CAS No. 92532-05-3) (Sanofi-Aventis), RG-14893 (CAS No. 141835-49-6) (Sanofi-Aventis), RG-5901-A (CAS No. 101910-24-1), 92532-23-5, RP 66153 (CAS No. 142422-79-5), RP 66364 (CAS No.186916-92-5), RP 69698 (CAS No. 141748-00-7) (Sanofi-Aventis), SC-411930 (Pfizer), SC-41930 (CAS No. 120072-59-5) (Pfizer), SC-50605 (CAS No. 138828-39-4) (Pfizer), SC-51146 (CAS No. 141059-52-1), SC- 53228 (CAS No. 153633-01-3) (Pfizer), spaglumic acid (ZY-15106) (CAS No.3106-85-2), 80619-64-3 (Novartis), tipredane (CAS No. 85197-77-9) (Bristol-Myers Squibb), U-75302 (CAS No. 119477-85-9) (Pfizer), and analogue or derivative thereof.
13240. The method of item 13179 wherein the agent is an MAP kinase inhibitor.
13241. The method of item 13179 wherein the agent is a matrix metalloproteinase inhibitor.
13242. The method of item 13179 wherein the agent is an MCP-CCR2 inhibitor.
13243. The method of item 13179 wherein the agent is an mTOR inhibitor.
13244. The method of item 13179 wherein the agent is an mTOR kinase inhibitor.
13245. The method of item 13179 wherein the agent is a microtubule inhibitor selected from the group consisting of antibody- maytansinoid conjugates from Biogen Idee, colchicines (MantiCore Pharmaceuticals), anticancer immunoconjugates from Johnson & Johnson, DIME from Octamer, gni-1f (GNI), huC242-DM4, huMy9-6-DM1 (ImmunoGen), IDN-5404 (Indena), IMO-098, IMOderm (Imotep), mebendazole (Introgen Therapeutics), microtubule poisons from Cambridge Enterprise, paclitaxel such as LOTAX from Aphios (CAS No. 33069-62-4), Genexol-PM from Samyang, Pharmaprojects No. 6383 (Tapestry Pharmaceuticals), RPR-112378 (Sanofi-Aventis), SGN-75 (Seattle Genetics), SPL-7435 (Starpharma), SSR-250411 (Sanofi-Aventis), trastuzumab-DM1 (Genentech), vinorelbine, and an analogue or derivative thereof.
13246. The method of item 13179 wherein the agent is an MlF inhibitor.
13247. The method of item 13179 wherein the agent is an MMP inhibitor.
13248. The method of item 13179 wherein the agent is a neurokinin (NK) antagonist selected from the group consisting of anthrotainin (CAS No. 148084-40-6) (Sanofi-Aventis), an IBS thereapeutic from ArQuIe, MDL- 105212A (CAS No. 167261-60-1) (Ssanofi-Aventis), Pharmaprojects No. 2744, 3258 (CAS No. 139167-47-8) 4006, 4201, or 5986 (Sanofi-Aventis), RP 67580 (CAS No. 135911-02-3), SR-144190 (CAS No. 201152-86-5), SSR-240600, SSR-241586 (Sanofi-Aventis), TKA-457 (Novartis), vestipitant mesylate (CAS No. 334476-64-1) (GlaxoSmithKline), Win-64821 (Sanofi-Aventis), PRX-96026 (Predix Pharmaceuticals), and an analogue or derivative thereof.
13249. The method of item 13179 wherein the agent is an NF kappa B inhibitor selected from the group consisting of AVE-0545 or AVE-0547 (Sanofi-Aventis), bortezomib (CAS No. 179324-69-7) (Millennium Pharmaceuticals), dexanabinol (CAS No. 112924-45-5) (Pharmos), dexlipotam (Viatris), Pharmaprojects No. 6283 (INDRA) (OXiGENE), IPL- 576092 (CAS No. 137571-30-3) (Inflazyme), NFKB decoy (Corgentech), NFKB decoy oligo (AnGes MG)1 NFKB's from Ariad, osteoporosis treatments or S5 (F005) from Fulcrum Pharmaceuticals, P61 (Phytopharm), R-flurbiprofen (CAS No. 5104-49-4) (Encore Pharmaceuticals), and an analogue or derivative thereof.
13250. The method of item 13179 wherein the agent is a nitric oxide agonist.
13251. The method of item 13179 wherein the agent is an ornithine decarboxylase inhibitor.
13252. The method of item 13179 wherein the agent is a p38 MAP kinase inhibitor selected from the group consisting of AZD-6703 (AstraZeneca), JX-401 (Jexys Pharmaceuticals), BMS-2 (Bristol-Myers Squibb), a p38 MAP kinase inhibitor from Novartis, a p38-alpha MAP kinase inhibitor from Amphora, Pharmaprojects No. 5704 (Pharmacopeia), RPR- 200765A (Sanofi-Aventis), SD-282 (Johnson & Johnson), TAK-715 (Takeda), and an analogue or derivative thereof.
13253. The method of item 13179 wherein the agent is a palmitoyl-protein thioesterase inhibitor.
13254. The method of item 13179 wherein the agent is a PDGF receptor kinase inhibitor selected from the group consisting of AAL- 993, AMN-107, or ABP-309 (Novartis), AMG-706 (Amgen), BAY-57-9352 (Bayer), CDP-860 (UCB), E-7080 (Eisai), imatinib (CAS No. 152459-95-5) (Novartis), OSI-930 (OSI Pharmaceuticals), RPR-127963E (Sanofi-Aventis), RWJ-540973 (Johnson & Johnson), sorafenib tosylate (Bayer), SU-11657 (Pfizer), tandutinib (CAS No. 387867-13-2) (Millennium Pharmaceuticals), vatalanib (Novartis), ZK-CDK (Schering AG), and an analogue or derivative thereof.
13255. The method of item 13179 wherein the agent is (-)-halofenate (Metabolex), AMG-131 (Amgen), antidiabetics from Japan Tobacco, AZD-4619, AZD-8450, AZD-8677 (AstraZeneca), DRF-10945, balaglitazone (Dr Reddy's), CS-00088, CS-00098 (Chipscreen Biosciences), E-3030 (Eisai), etalocib (CAS No. 161172-51-6) (EIi Lilly), GSK-641597 (Ligand), GSK-677954 (GlaxoSmithKline), GW-409544 (Ligand), GW- 590735 (GlaxoSmithKline), K-111 (Hoffmann-La Roche), LY-518674 (EIi Lilly), LY-674 (Ligand), LY-929 (Ligand), MC-3001 , MC-3002 (MaxoCore Pharmaceuticals), metformin HCI + pioglitazone (CAS No. 1115-70-4 and
112529-15-4), ACTOPLUS MET from Andrx), muraglitazar (CAS No. 331741-94-7) (Bristol-Myers Squibb), naveglitazar (Ligand), oleoylethanolamide (Kadmus Pharmaceuticals), ONO-5129, pioglitazone hydrochloride (CAS No. 111025-46-8 and 112529-15-4) (Takeda),-PLX-204 (Plexxikon), PPAR agonists from Genfit, PPAR delta agonists from EIi Lilly, PPAR-alpha agonists from CrystalGenomics, PPAR-gamma modulators and PPAR-β modulators from CareX, rosiglitazone maleate (CAS No. 122324- 73-4 or 155141-29-0) (GlaxoSmithKline), rosiglitazone maleate/glimepir (CAS No. 155141-29-0 and 93479-97-1), AVANDARYL, rosiglitazone maleate/metformin extend (CAS No. 155141-29-0 and 657-24-9), AVANDAMET, rosiglitazone maleate+metformin, AVANDAMET (GlaxoSmithKline), tesaglitazar (AstraZeneca), and an analogue or derivative thereof.
13256. The method of item 13179 wherein the agent is a phosphatase inhibitor.
13257. The method of item 13179 wherein the agent is a phosphodiesterase (PDE) inhibitor selected from the group consisting of avanafil (Tanabe Seiyaku), dasantafil (CAS No. 569351-91-3) (Schering- Plough), A-906119 (CAS No. 134072-58-5), DL-850 (Sanofi-Aventis), GRC- 3015, GRC-3566, GRC-3886 (Glenmark), HWA-153 (CAS No. 56395-66-5) (Sanofi-Aventis), hydroxypumafentrine (Altana), IBFB-130011, IBFB-14-016, IBFB-140301 , IBFB-150007, IBFB-211913 (IBFB Pharma), L-826141 (Merck & Co), medorinone (CAS No. 88296-61-1) (Sanofi-Aventis), MEM- 1917 (Memory Pharmaceuticals), ND-1251 (Neuro3d), PDE inhibitors from ICOS, PDE IV inhibitors from Memory Pharmaceuticals and CrystalGenomics, Pharmaprojects No. 2742 and 6141 (Sanofi-Aventis), QAD-171 (Novartis), RHC-2963 (CAS No. 76993-12-9 and 76993-14-1), RPR-117658, RPR-122818 derivatives, SR-24870 , and RPR-132294 (Sanofi-Aventis), SK-350 (ln2Gen), stroke therapy agents from deCODE Genetics, TAS-203 (Taiho), tofimilast (CAS No. 185954-27-2) (Pfizer), UK- 371800 (Pfizer), WIN-65579 (CAS No. 158020-82-7) (Sanofi-Aventis), IBFB- 130020 (IBFB Pharma), OPC-6535 (CAS No. 145739-56-6) (Otsuka), a phosphodiesterase III inhibitor, enoximone, a phosphodiesterase IV inhibitor, fosfosal, Atopik (Barrier Therapeutics), triflusal, a phosphodiesterase V inhibitor, and an analogue or derivative thereof.
13258. The method of item 13179 wherein the agent is a PKC inhibitor.
13259. The method of item 13179 wherein the agent is a platelet activating factor antagonist.
13260. The method of item 13179 wherein the agent is a platelet-derived growth factor receptor kinase inhibitor.
13261. The method of item 13179 wherein the agent is a prolyl hydroxylase inhibitor. 13262. The method of item 13179 wherein the agent is a polymorphonuclear neutrophil inhibitor.
13263. The method of item 13179 wherein the agent is a protein kinase B inhibitor.
13264. The method of item 13179 wherein the agent is a protein kinase C stimulant.
13265. The method of item 13179 wherein the agent is a purine nucleoside analogue.
13266. The method of item 13179 wherein the agent is a purinoreceptor P2X antagonist.
13267. The method of item 13179 wherein the agent is a Raf kinase inhibitor.
13268. The method of item 13179 wherein the agent is a reversible inhibitor of ErbB1 and ErbB2.
13269. The method of item 13179 wherein the agent is a ribonucleoside triphosphate reductase inhibitor.
13270. The method of item 13179 wherein the agent is an SDF-1 antagonist.
13271. The method of item 13179 wherein the agent is a sheddase inhibitor. 13272. The method of item 13179 wherein the agent is an SRC inhibitor.
13273. The method of item 13179 wherein the agent is a stromelysin inhibitor.
13274. The method of item 13179 wherein the agent is an Syk kinase inhibitor.
13275. The method of item 13179 wherein the agent is a telomerase inhibitor.
13276. The method of item 13179 wherein the agent is a TGF beta inhibitor selected from the group consisting of pirfenidone (CAS No. 53179-13-8) (MARNAC)1 tranilast (CAS No. 53902-12-8) (Kissei), IN-
1130 (ln2Gen), mannose-6-phosphate (BTG), TGF-β antagonists from Inflazyme (Pharmaprojects No. 6075), TGF-β antagonists from Sydney, non-industrial source), TGF-β I receptor kinase inhibitors from EIi Lilly, TGF- β receptor inhibitors from Johnson & Johnson, and an analogue or derivative thereof.
13277. The method of item 13179 wherein the agent is a TNFα antagonist or TACE inhibitor selected from the group consisting of adalimumab (CAS No. 331731-18-1) (Cambridge Antibody Technology), AGIX-4207 (AtheroGenics), AGT-1 (Advanced Biotherapy), an antiinflammatory from Borean Pharma, Cellzome, or Paradigm Therapeutics, anti-inflammatory vaccine (TNF-alpha kinoid) from Neovacs, humanized anti-TNF antibody or an anti-TNF MAb (CB0006) Celltech (UCB), apratastat (CAS No. 287445^51-0) (Wyeth), BMS-561392 (Bristol-Myers Squibb), BN- 006 (Bone), certolizumab pegol (CAS No. 428863-50-7 or CH-138 (UCB), cilomilast (CAS No. 153259-65-5) (GlaxoSmithKline), CR-1 (Nuada Pharmaceuticals), CRx-119 (CombinatoRx), D-5410 (UCB), dacopafant (CAS No. 125372-33-0) (Sanofi-Aventis), dersalazine (CAS No. 188913-57- 7/188913-58-8) (Uriach), etanercept (CAS No. 185243-69-0) (Amgen), ethyl pyruvate (Critical (Critical Therapeutics), golimumab (CAS No. 476181-74-5) (Johnson & Johnson), hormono-immunotherapy from Ipsen, CDP571 (e.g., Humicade from UCB), IC-485 (ICOS), infliximab (CAS No. 170277-31-3) (Johnson & Johnson), IP-751 (Manhattan Pharmaceuticals), ISIS-104838 (CAS No. 250755-32-9) (ISIS Pharmaceuticals), lenalidomide (CAS No. 191732-72-6) (Celgene), lentinan (CAS No. 37339-90-5) (Ajinomoto), MDL- 201112 (CAS No. 142130-73-2) (Sanofi-Aventis), medroxyprogesterone (CAS No. 520-85-4) (InKine Pharmaceutical), N-acetylcysteine (CAS No. 616-91-1) (Zambon), NBE-P2 (DIREVO Biotech), nerelimomab (CAS No. 162774-06-3) (Chiron), OM-294DP (OM PHARMA), onercept (CAS No. 199685-57-9) (Yeda), PASSTNF-alpha (Verigen), pentoxifylline or oxypentifylline (Sanofi-Aventis), Pharmaprojects No. 4091 , 4241 , 4295, or 4488 (Sanofi-Aventis), Pharmaprojects No. 5480 (Amgen), Pharmaprojects No. 6749 (Cengent), pirfenidone (CAS No. 53179-13-8) (MARNAC), RPR- 132294 (Sanofi-Aventis), S5 (F002) (Fulcrum Pharmaceuticals), simvastatin (CAS No. 79902-63-9) (Merck & Co), STA-6292 (Synta Pharmaceuticals), tacrolimus (CAS No. 104987-11-3) (Fujisawa LifeCycle Pharma), talactoferrin alfa (CAS No. 308240-58-6) (Agennix), thalidomide (CAS No. 50-35-1) (Celgene), TNF antagonists form ProStrakan, and Synergen, TNF inhibitors (Amgen), TNF-alpha antagonists from Dynavax Technologies and Jerina AG (Germany), TNF-alpha inhibitors from IBFB Pharma and Xencor (Xencor), torbafylline (CAS No. 105102-21-4) (Sanofi-Aventis), UR-1505 (Uriach), VT-346 (Viron Therapeutics), YSIL6 (Y1S Therapeutics), YSTH2 (Y's Therapeutics), NPI-1302a-3 (Nereus Pharmaceuticals, a TNF antagonist from Jerina AG (Germany), dersalazine, and an analogue or derivative thereof. 13278. The method of item 13179 wherein the agent is a Toll receptor inhibitor.
13279. The method of item 13179 wherein the agent is a tubulin antagonist.
13280. The method of item 13179 wherein the agent is a tyrosine kinase inhibitor selected from the group consisting of SU-011248, SUTENT from Pfizer Inc. (New York, NY), BMS-354825, PN-355 (Paracelsian Pharmaceuticals), AGN-199659 (Allergan), AAL-993 or ABP- 309 (Novartis), adaphostin (NIH), AEE-788 (Novartis), AG-013736 (OSI Pharmaceuticals), AG-13736 (Pfizer), ALT-110 (Alteris Therapeutics), AMG- 706 (Amgen), anticancer MAbs from Xencor, anti-EGFrvlll MAbs from Abgenix, anti-HER2 MAb from Abiogen, AZD-2171 or AZD-9935 (AstraZeneca), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), CEP-5214 (Cephalon), CEP-7055 (Cephalon), cetuximab (ImClone Systems), CHIR-200131 and CHIR-258 (Chiron), CP-547632 (OSI Pharmaceuticals), CP-724714 (Pfizer), CT-301 (Creabilis Therapeutics), D- 69491 (Baxter International), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EGFR/ErbB2 inhibitors from Array BioPharma, erlotinib (CAS No. 183319- 69-9) (OSI Pharmaceuticals), EXEL-2880 (Exelixis), FK-778 (Sanofi- Aventis) , gefitinib (CAS No. 184475-35-2) (AstraZeneca), GW-2286 or GW- 654652 (GlaxoSmithKline), her2/neu antigen from AlphaVax, HER-2/neu inhibitor from Generex, Herzyme (Medipad) (Sirna Therapeutics), HKI-272 (Wyeth), HuMax-EGFr (Genmab), idronoxil (CAS No. 81267-65-4) (Novogen), IGF-1 inhibitors from Ontogen, IMC-11F8 (ImClone Systems), kahalalide F (CAS No. 149204-42-2) (PharmaMar), KDR inhibitor from LG Life Sciences, KDR inhibitors from Abbott Laboratories, KDR kinase inhibitors (UCB), Kdr kinase inhibitors from Merck & Co, KRN-633 and KRN- 951 (Kirin Brewery), KSB-102 (Xenova), lapatinib ditosylate (CAS No. 388082-78-8) (GlaxoSmithKline), matuzumab (Merck KGaA), MDX-214 (Medarex), ME-103 (Pharmexa), MED-A300 (Gerolymatos), MNAC-13 (Lay Line Genomics), nimotuzumab (Center of Molecular Immunology), NSC- 330507 or NSC-707545 (NIH), NV-50 (Novogen), OSI-930 (OSI Pharmaceuticals), panitumumab (Abgenix), pelitinib (CAS No. 287933-82-7) (Wyeth), pertuzumab (CAS No. 380610-27-5) (Genentech), Pharmaprojects No. 3985 (Sanofi-Aventis), prostate cancer therapeutics from Sequenom (SQPC35, SQPC36, SQPC90), removab and remoxab (Trion Pharma), RG- 13022 (CAS No. 136831-48-6), RG-13291 (CAS No. 138989-50-1), or RG- 14620 (CAS No. 136831-49-7) (Sanofi-Aventis), RM-6427 (Romark), RNAi breast cancer therapy from Benitec, RP 53801 (CAS No. 125882-88-4) (Sanofi-Aventis), sorafenib tosylate (Bayer), SU-11657 (Pfizer), Tie-2 antagonists from Semaia (Hybrigenics), Tie-2 inhibitors from Ontogen, trastuzumab (CAS No. 180288-69-1) (Genentech), tyrosine kinase inhibitors from Sanofi-Aventis, U3-1287, U3-1565, U3-1784, U3-1800 (U3 Pharma), vatalanib (Novartis), VEGFR-2 kinase inhibitor from Bristol-Myers Squibb, XL-647 (Exelixis), ZD-6474 (AstraZeneca), ZK-CDK (Schering AG), an EGFR tyrosine kinase inhibitor, EKB-569 (Wyeth), and an analogue or derivative thereof.
13281. The method of item 13179 wherein the agent is a VEGF inhibitor.
13282. The method of item 13179 wherein the agent is a vitamin D receptor agonist.
13283. The method of item 13179 wherein the agent is ZD-6474 (an angiogenesis inhibitor).
13284. The method of item 13179 wherein the agent is AP-23573 (an mTOR inhibitor). 13285. The method of item 13179 wherein the agent is synthadotin (a tubulin antagonist).
13286. The method of item 13179 wherein the agent is S-0885 (a collagenase inhibitor).
13287. The method of item 13179 wherein the agent is aplidine (an elongation factor-1 alpha inhibitor).
13288. The method of item 13179 wherein the agent is ixabepilone (an epithilone).
13289. The method of item 13179 wherein the agent is IDN-5390 (an angiogenesis inhibitor).
13290. The method of item 13179 wherein the agent is SB-2723005 (an angiogenesis inhibitor).
13291. The method of item 13179 wherein the agent is ABT-518 (an angiogenesis inhibitor).
13292. The method of item 13179 wherein the agent is combretastatin (an angiogenesis inhibitor).
13293. The method of item 13179 wherein the agent is anecortave acetate (an angiogenesis inhibitor).
13294. The method of item 13179 wherein the agent is SB-715992 (a kinesin antagonist). 13295. The method of item 13179 wherein the agent is temsirolimus (an mTOR inhibitor).
13296. The method of item 13179 wherein the agent is adalimumab (a TNFα antagonist).
13297. The method of item 13179, wherein the composition comprises a polymer.
13298. The method of item 13179, wherein the composition comprises a polymeric carrier.
13299. The method of item 13179 wherein the anti- scarring agent inhibits adhesion between the device and a host into which the device is implanted.
13300. The method of item 13179 wherein the device delivers the anti-scarring agent locally to tissue proximate to the device.
13301. The method of item 13179 wherein the device has a coating that comprises the anti-scarring agent.
13302. The method of item 13179, wherein the device has a coating that comprises the agent and is disposed on a surface of the implant.
13303. The method of item 13179, wherein the device has a coating that comprises the agent and directly contacts the implant.
13304. The method of item 13179, wherein the device has a coating that comprises the agent and indirectly contacts the implant. 13305. The method of item 13179, wherein the device has a coating that comprises the agent and partially covers the implant.
13306. The method of item 13179, wherein the device has a coating that comprises the agent and completely covers the implant.
13307. The method of item 13179, wherein the device has a uniform coating.
13308. The method of item 13179, wherein the device has a non-uniform coating.
13309. The method of item 13179, wherein the device has a discontinuous coating.
13310. The method of item 13179, wherein the device has a patterned coating.
13311. The method of item 13179, wherein the device has a coating with a thickness of 100 Dm or less.
13312. The method of item 13179, wherein the device has a coating with a thickness of 10 Dm or less.
13313. The method of item 13179, wherein the device has a coating, and the coating adheres to the surface of the implant upon deployment of the implant.
13314. The method of item 13179, wherein the device has a coating, and wherein the coating is stable at room temperature for a period of 1 year. 13315. The method of item 13179, wherein the device has a coating, and wherein the anti-scarring agent is present in the coating in an amount ranging between about 0.0001% to about 1% by weight.
13316. The method of item 13179, wherein the device has a coating, and wherein the anti-scarring agent is present in the coating in an amount ranging between about 1% to about 10% by weight.
13317. The method of item 13179, wherein the device has a coating, and wherein the anti-scarring agent is present in the coating in an amount ranging between about 10% to about 25% by weight.
13318. The method of item 13179, wherein the device has a coating, and wherein the anti-scarring agent is present in the coating in an amount ranging between about 25% to about 70% by weight.
13319. The method of item 13179, wherein the device has a coating, and wherein the coating further comprises a polymer.
13320. The method of item 13179, wherein the device has a first coating having a first composition and a second coating having a second composition.
13321. The method of item 13179, wherein the device has a first coating having a first composition and a second coating having a second composition, wherein the first composition and the second composition are different.
13322. The method of item 13179, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a copolymer. 13323. The method of item 13179, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a block copolymer.
13324. The method of item 13179, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a random copolymer.
13325. The method of item 13179, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a biodegradable polymer.
13326. The method of item 13179, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a non-biodegradable polymer.
13327. The method of item 13179, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a hydrophilic polymer.
13328. The method of item 13179, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a hydrophobic polymer.
13329. The method of item 13179, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a polymer having hydrophilic domains.
13330. The method of item 13179, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a polymer having hydrophobic domains. 13331. The method of item 13179, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a non-conductive polymer.
13332. The method of item 13179, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises an elastomer.
13333. The method of item 13179, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a hydrogel.
13334. The method of item 13179, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a silicone polymer.
13335. The method of item 13179, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a hydrocarbon polymer.
13336. The method of item 13179, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a styrene-derived polymer.
13337. The method of item 13179, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a butadiene polymer.
13338. The method of item 13179, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a macromer. 13339. The method of item 13179, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a poly(ethylene glycol) polymer.
13340. The method of item 13179 wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises an amorphous polymer.
13341. The method of item 13179, wherein the device comprises a lubricious coating.
13342. The method of item 13179 wherein the anti- scarring agent is located within pores or holes of the device.
13343. The method of item 13179 wherein the anti- scarring agent is located within a channel, lumen, or divet of the device.
13344. The method of item 13179, wherein the device comprises a second pharmaceutically active agent.
13345. The method of item 13179 wherein the device comprises an anti-inflammatory agent.
13346. The method of item 13179 wherein the device comprises an agent that inhibits infection.
13347. The method of item 13179 wherein the device comprises an agent that inhibits infection, and wherein the agent is an anthracycline. 13348. The method of item 13179 wherein the device comprises an agent that inhibits infection, and wherein the agent is doxorubicin.
13349. The method of item 13179 wherein the device comprises an agent that inhibits infection, and wherein the agent is mitoxantrone.
13350. The method of item 13179 wherein the device comprises an agent that inhibits infection, and wherein the agent is a fluoropyrimidine.
13351. The method of item 13179 wherein the device comprises an agent that inhibits infection, and wherein the agent is 5- fluorouracil (5-FU).
13352. The method of item 13179 wherein the device comprises an agent that inhibits infection, and wherein the agent is a folic acid antagonist.
13353. The method of item 13179 wherein the device comprises an agent that inhibits infection, and wherein the agent is methotrexate.
13354. The method of item 13179 wherein the device comprises an agent that inhibits infection, and wherein the agent is a podophylotoxin.
13355. The method of item 13179 wherein the device comprises an agent that inhibits infection, and wherein the agent is etoposide. 13356. The method of item 13179 wherein the device comprises an agent that inhibits infection, and wherein the agent is a camptothecin.
13357. The method of item 13179 wherein the device comprises an agent that inhibits infection, and wherein the agent is a hydroxyurea.
13358. The method of item 13179 wherein the device comprises an agent that inhibits infection, and wherein the agent is a platinum complex.
13359. The method of item 13179 wherein the device comprises an agent that inhibits infection, and wherein the agent is cisplatin.
13360. The method of item 13179, further comprising an anti-thrombotic agent.
13361. The method of item 13179 wherein the device comprises a visualization agent.
13362. The method of item 13179 wherein the device comprises a visualization agent, wherein the visualization agent is a radiopaque material, and wherein the radiopaque material comprises a metal, a halogenated compound, or a barium containing compound.
13363. The method of item 13179 wherein the device comprises a visualization agent, wherein the visualization agent is a radiopaque material, and wherein the radiopaque material comprises barium, tantalum, or technetium. 13364. The method of item 13179 wherein the device comprises a visualization agent, and wherein the visualization agent is a MRI responsive material.
13365. The method of item 13179 wherein the device comprises a visualization agent, and wherein the visualization agent comprises a gadolinium chelate.
13366. The method of item 13179 wherein the device comprises a visualization agent, and wherein the visualization agent comprises iron, magnesium, manganese, copper, or chromium.
13367. The method of item 13179 wherein the device comprises a visualization agent, and wherein the visualization agent comprises an iron oxide compound.
13368. The method of item 13179 wherein the device comprises a visualization agent, and wherein the visualization agent comprises a dye, pigment, or colorant.
13369. The method of item 13179 wherein the device comprises an echogenic material.
13370. The method of item 13179 wherein the device comprises an echogenic material, and wherein the echogenic material is in the form of a coating.
13371. The method of item 13179 wherein the device is sterile. 13372. The method of item 13179 wherein the anti- scarring agent is released into tissue in the vicinity of the device after deployment of the device.
13373. The method of item 13179 wherein the anti- scarring agent is released into tissue in the vicinity of the device after deployment of the device, and wherein the tissue is connective tissue.
13374. The method of item 13179 wherein the anti- scarring agent is released into tissue in the vicinity of the device after deployment of the device, and wherein the tissue is muscle tissue.
13375. The method of item 13179 wherein the anti- scarring agent is released into tissue in the vicinity of the device after deployment of the device, and wherein the tissue is nerve tissue.
13376. The method of item 13179 wherein the anti- scarring agent is released into tissue in the vicinity of the device after deployment of the device, and wherein the tissue is epithelium tissue.
13377. The method of item 13179 wherein the anti- scarring agent is released in effective concentrations from the device over a period ranging from the time of deployment of the device to about 1 year.
13378. The method of item 13179 wherein the anti- scarring agent is released in effective concentrations from the device over a period ranging from about 1 month to 6 months.
13379. The method of item 13179 wherein the anti- scarring agent is released in effective concentrations from the device over a period ranging from about 1 - 90 days. 13380. The method of item 13179 wherein the anti- scarring agent is released in effective concentrations from the device at a constant rate.
13381. The method of item 13179 wherein the anti- scarring agent is released in effective concentrations from the device at an increasing rate.
13382. The method of item 13179 wherein the anti- scarring agent is released in effective concentrations from the device at a decreasing rate.
13383. The method of item 13179 wherein the anti- scarring agent is released in effective concentrations from the composition comprising the anti-scarring agent by diffusion over a period ranging from the time of deployment of the device to about 90 days. -
13384. The method of item 13179 wherein the anti- scarring agent is released in effective concentrations from the composition comprising the anti-scarring agent by erosion of the composition over a period ranging from the time of deployment of the device to about 90 days.
13385. The method of item 13179 wherein the device comprises about 0.01 Dg to about 10 Dg of the anti-scarring agent.
13386. The method of item 13179 wherein the device comprises about 10 Dg to about 10 mg of the anti-scarring agent.
13387. The method of item 13179 wherein the device comprises about 10 mg to about 250 mg of the anti-scarring agent. 13388. The method of item 13179 wherein the device comprises about 250 mg to about 1000 mg of the anti-scarring agent.
13389. The method of item 13179 wherein the device comprises about 1000 mg to about 2500 mg of the anti-scarring agent.
13390. The method of item 13179 wherein a surface of the device comprises less than 0.01 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
13391. The method of item 13179 wherein a surface of the device comprises about 0.01 Og to about 1 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
13392. The method of item 13179 wherein a surface of the device comprises about 1 Dg to about 10 Dg of the anti-scarring agent . . per mm2 of device surface to which the anti-scarring agent is applied.
13393. The method of item 13179 wherein a surface of the device comprises about 10 Dg to about 250 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
13394. The method of item 13179 wherein a surface of the device comprises about 250 Dg to about 1000 Dg of the anti-scarring agent of anti-scarring agent per mm2 of device surface to which the anti- scarring agent is applied.
13395. The method of item 13179 wherein a surface of the device comprises about 1000 Dg to about 2500 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied. 13396. The method of item 13179 wherein the combining is performed by direct affixing the agent or the composition to the implant.
13397. The method of item 13179 wherein the combining is performed by spraying the agent or the component onto the implant.
13398. The method of item 13179 wherein the combining is performed by electrospraying the agent or the composition onto the implant.
13399. The method of item 13179 wherein the combining is performed by dipping the implant into a solution comprising the agent or the composition.
13400. The method of item 13179 wherein the combining is performed by covalently attaching the agent or the composition to the implant.
13401. The method of item 13179 wherein the combining is performed by non-covalently attaching the agent or the composition to the implant.
13402. The method of item 13179 wherein the combining is performed by coating the implant with a substance that contains the agent or the composition.
13403. The method of item 13179 wherein the combining is performed by coating the implant with a substance that absorbs the agent. 13404. The method of item 13179 wherein the combining is performed by interweaving a thread composed of, or coated with, the agent or the composition.
13405. The method of item 13179 wherein the combining is performed by covering all the implant with a sleeve that contains the agent or the composition.
13406. The method of item 13179 wherein the combining is performed by covering a portion of the implant with a sleeve that contains the agent or the composition.
13407. The method of item 13179 wherein the combining is performed by covering all the implant with a cover that contains the agent or the composition.
13408. The method of item 13179 wherein the combining is performed by covering a portion of the implant with a cover that contains the agent or the composition.
13409. The method of item 13179 wherein the combining is performed by covering all the implant with an electrospun fabric that contains the agent or the composition.
13410. The method of item 13179 wherein the combining is performed by covering a portion of the implant with an electrospun fabric that contains the agent or the composition.
13411. The method of item 13179 wherein the combining is performed by covering all the implant with a mesh that contains the agent or the composition. 13412. The method of item 13179 wherein the combining is performed by covering a portion of the implant with a mesh that contains the agent or the composition.
13413. The method of item 13179 wherein the combining is performed by constructing all the implant with the agent or the composition.
13414. The method of item 13179 wherein the combining is performed by constructing a portion of the implant with the agent or the composition.
13415. The method of item 13179 wherein the combining is performed by impregnating the implant with the agent or the composition.
13416. The method of item 13179 wherein the combining is performed by constructing all of the implant from a degradable polymer that releases the agent.
13417. The method of item 13179 wherein the combining is performed by constructing a portion of the implant from a degradable polymer that releases the agent.
13418. The method of item 13179 wherein the combining is performed by dipping the implant into a solution that comprise the agent and an inert solvent for the implant.
13419. The method of item 13179 wherein the combining is performed by dipping the implant into a solution that comprises the agent and a solvent that will swell the implant. 13420. The method of item 13179 wherein the combining is performed by dipping the implant into a solution that comprises the agent and a solvent that will dissolve the implant.
13421. The method of item 13179 wherein the combining is performed by dipping the implant into a solution that comprises the agent, a polymer and an inert solvent for the implant.
13422. The method of item 13179 wherein the combining is performed by dipping the implant into a solution that comprises the agent, a polymer and a solvent that will swell the implant.
13423. The method of item 13179 wherein the combining is performed by dipping the implant into a solution that comprises the agent, a polymer and a solvent that will dissolve the implant.
13424. The method of item 13179 wherein the combining is performed by spraying the implant into a solution that comprises the agent and an inert solvent for the implant.
13425. The method of item 13179 wherein the combining is performed by spraying the implant into a solution that comprises the agent and a solvent that will swell the implant.
13426. The method of item 13179 wherein the combining is performed by spraying the implant into a solution that comprises the agent and a solvent that will dissolve the implant.
13427. The method of item 13179 wherein the combining is performed by spraying the implant into a solution that comprises the agent, a polymer and an inert solvent for the implant. 13428. The method of item 13179 wherein the combining is performed by spraying the implant into a solution that comprises the agent, a polymer and a solvent that will swell the implant.
13429. The method of item 13179 wherein the combining is performed by spraying the implant into a solution that comprises the agent, a polymer and a solvent that will dissolve the implant.
13430. A method as in any one of items 13179-13429, wherein the device is a device for treating hypertropic scar or keloid that comprises an external tissue expansion device.
13431. A method as in any one of items 13179-13429, wherein the device is a device for treating hypertropic scar or keloid that comprises a masking element, and wherein the masking element may be pressed onto the scar tissue.
13432. A method as in any one of items 13179-13429, wherein the device is a device for treating hypertropic scar or keloid that comprises a locking element and a grasping structure so that the dermal and epidermal layers of a skin wound can be pushed together.
13433. A method of making a medical device comprising: combining a vascular graft (e.g., an implant) and an anti- scarring agent or a composition comprising an anti-scarring agent, wherein the agent inhibits scarring between the device and a host into which the device is implanted.
13434. The method of item 13433 wherein the agent is an adensosine A2A receptor antagonist. 13435. The method of item 13433 wherein the agent is an AKT inhibitor.
13436. The method of item 13433 wherein the agent is an alpha 2 integrin antagonist, wherein the alpha 2 integrin antagonist is Pharmaprojects No. 5754 (Merck KgaA).
13437. The method of item 13433 wherein the agent is an alpha 4 integrin antagonist.
13438. The method of item 13433 wherein the agent is an alpha 7 nicotinic receptor agonist.
13439. The method of item 13433 wherein the agent is an angiogenesis inhibitor selected from the group consisting of AG-12,958 (Pfizer), ATN-161 (Attention LLC), neovastat, an angiogenesis inhibitor from Jerina AG (Germany), NM-3 (Mercian), VGA-1155 (Taisho), FCE-26644 (Pfizer), FCE-26950 (Pfizer), FPMA (Meiji Daries), FR-111142 (Fυjisawa), GGTI-298, GM-1306 (Ligand), GPA-1734 (Novartis), NNC-47-0011 (Novo Nordisk), herbamycin (Nippon Kayaku), lenalidomide (Celegene), IP-10 (NIH), ABT-828 (Abbott), KIN-841 (Tokushima University, Japan), SF-1126 (Semafore Pharmaceuticals), laminin technology (NIH), CHIR-258 (Chiron), NVP-AEW541 (Novartis), NVP-AEW541 (Novartis), Vt16907 (Alchemia), OXI-8007 (Oxigene), EG-3306 (Ark Therapeutics), Maspin (Arriva), ABT- 567 (Abbott), PPI-2458 (Praecis Pharmaceuticals), CC-5079, CC-4089 (Celgene), HIF-1 alpha inhibitors (Xenova), S-247 (Pfizer), AP-23573 (Ariad), AZD-9935 (Astra Zeneca), mebendazole (Introgen Therapeutics), MetAP-2 inhibitors (GlaxoSmithKline), AG-615 (Angiogene Pharmaceuticals), Tie-2 antagonists (Hybrigenics), NC-381, CYC-381, NC-169, NC-219, NC-383, NC-384, NC-407 (Lorus Therapeutics), ATN-224 (Attenuon), ON-01370 (Onconova), Vitronectin antagonists (Amgen), SDX-103 (Salmedix), Vitronectin antagonists (Shire), CHP (Riemser), TEK (Amgen), Anecortave acetate (Alcon), T46.2 (Matrix Therapeutics), HG-2 (Heptagen), TEM antagonists (Genzyme), Oxi-4500 (Oxigene), ATN-161 (Attenuon), WX-293 (Wilex), M-2025 (Metris Therapeutics), Alphastatin (BioActa), YH-16 (Yantai Rongchang), BIBF-1120 (Boehringer Ingelheim), BAY-57-9352 (Bayer), AS- 1404 (Cancer Research Technology), SC-77964 (Pfizer), glycomimetics (BioTie Therapies), TIE-2 Inhibitors (Ontogen), DIMI, Octamer (Octamer), ABR-215050 (Active Biotech), ABT-518 (Abbott), KDR inhibitors (Abbott), BSF-466895 (Abbott), SCH-221153 (Schering-Plough), DAC:antiangiogenic (ConjuChem), TFPI (EntreMed), AZD-2171 (Astra-Zenaca), CDC-394 (Celgene), LY290293 (EIi Lilly), IDN-5390 (Indena), Kdr Kinase Inhibitors (Merck), CT-113020, CT-116433, CT-116563, CT-31890, CT-32228) (Cell Therapeutics), A-299620 (Abbott), TWEAK Inhibitor (Amgen), VEGF modulators (Johnson and Johnson), Tum-N53, tumstatin (Genzyme), Thios- 1 , Thios-2 (Thios Pharmaceuticals), MV-6401 (Miravant Medical Technologies), Spisulosine (PharmaMar), CEP-7055 (Cephalon), AUV-201 (Auvation), LM-609 (EIi Lilly), SKF-106615 (AnorMED), Oglufanide disodium (Cytran), BW-114 (Pharminox), Calreticulin (NIH), WX-678 (Wilex), SD-7784 (Pfizer), WX-UK1 (Wilex), SH-268 (Schering AG), 2-Me-PGA (Celgene), S- 137 (Pfizer), ZD-6126 (Angiogene Pharmaceuticals), SG-292 (SignalGen), Benefin (Lane Labs), A6, A36 (Angstrom), SB-2723005 (GlaxoSmithKline), SC-7 (Cell Therapeutics), ZEN-014 (AEterna Zentaris), 2-methoxyestradiol (EntreMed), NK-130119 (Nippon Kayaku), CC-10004 (Celgene), AVE- 8062A (Ajinomoto), Tacedinaline (Pfizer), Actinonin (Tokyo Metropolitan Institute of Medical Science), Lenalidomide (Celgene), VGA-1155, BTO-956 (SRI International), ER-68203-00 (Eisai), CT-6685 (UCB), JKC-362 (Phoenix Pharmaceuticals), DMI-3798 (DMI Biosciences, Angiomate (Ipsen), ZD-6474 (AstraZeneca), CEP-5214 (Cephalon), Canstatin (Genzyme), NM-3 (Mercian), Oridigm (MediQuest Therapeutics), Exherin (Adherex), BLS-0597 (Boston Life Sciences), PTC-299 (PTC Therapeutics), NPI-2358 (Nereus Pharmaceuticals), CGP-79787 (Novartis), AEE-788 (Novartis), CKD-732 (Chong Kun Dang), CP-564959 (OSI Pharmaceuticals), CM-101 (CarboMed), CT-2584, CT3501 (Cell Therapeutics), combretastatin and analogues and derivatives thereof (Oxigene), Rebimastat (Bristol-Meyers Squibb), Dextrin 2-sulfate (ML Laboratories), Cilengitide (Merk KGaA)1 NSC-706704 (Pharminox), KRN- 951 (Kirin Brewery), Ukrain, NSC-631570 (Nowicky Pharma), Tecogalan sodium (Daiichi Pharmaceutical), Tz-93 (Tsumura), TBC-1635 (Encysive Pharmaceuticals), TAN-1120 (Takeda), Semaxanib (Pfizer), BDI-7800 (Biopharmacopae), SD-186, SD-983 (Bristol-Meyers Squibb), SB-223245 (GlaxoSmithKline), SC-236 (Pfizer), RWJ-590973 (Johnson and Johnson), ILX-1850 (Genzyme), SC-68488, S-836 (Pfizer), CG-55069-11 (CuraGen), Ki-23057 (Kirin Brewery), CCX-700 (Chemoentryx), Pegaptanib octasodium (Giled Sciences), ANGIOCOL (available from Biostratum Inc.), or an analogue or derivative thereof.
13440. The method of item 13433 wherein the agent is an apoptosis antagonist.
13441. The method of item 13433 wherein the agent is an apoptosis activator.
13442. The method of item 13433 wherein the agent is a beta 1 integrin antagonist.
13443. The method of item 13433 wherein the agent is a beta tubulin inhibitor.
13444. The method of item 13433 wherein the agent is a blocker of enzyme production in Hepatitis C. 13445. The method of item 13433 wherein the agent is a Bruton's tyrosine kinase inhibitor.
13446. The method of item 13433 wherein the agent is a calcineurin inhibitor.
13447. The method of item 13433 wherein the agent is a caspase 3 inhibitor.
13448. The method of item 13433 wherein the agent is a CC chemokine receptor antagonist.
13449. The method of item 13433 wherein the agent is a cell cycle inhibitor selected from the group consisting of SNS-595 (Sunesis), synthadotin, KRX-0403, and an analogue or derivative thereof.
13450. The method of item 13433 wherein the agent is a cathepsin B inhibitor.
13451. The method of item 13433 wherein the agent is a cathepsin K inhibitor, wherein the cathepsin K inhibitor is 462795 (GlaxoSmithKline), INPL-022-D6 (Amura Therapeutics), or an analogue or derivative thereof.
13452. The method of item 13433 wherein the agent is a cathepsin L inhibitor.
13453. The method of item 13433 wherein the agent is a CD40 antagonist. 13454. The method of item 13433 wherein the agent is a chemokine receptor agonist.
13455. The method of item 13433 wherein the agent is a chymase inhibitor.
13456. The method of item 13433 wherein the agent is a collagenase antagonist.
13457. The method of item 13433 wherein the agent is a CXCR antagonist.
13458. The method of item 13433 wherein the agent is a cyclin dependent kinase inhibitor selected from the group consisting of a CDK-1 inhibitor, a CDK-2 inhibitor, a CDK- 4 inhibitor, a CDK-6 inhibitor, a CAK1 inhibitor from GPC Biotech and Bristol-Myers Squibb, RGB-286199 (GPC Biotech), an anticancer agent from Astex Technology, a CAK1 inhibitor from GPC Biotech, a CDK inhibitor from Sanofi-Aventis, a CDK1/CDK2 inhibitor from Amgen, a CDK2 inhibitor from SUGEN-2 (Pfizer), a hearing loss therapy agent (Sound Pharmaceuticals), PD-0332991 (Pfizer), RGB-286199 (GPC Biotech), Ro-0505124 (Hoffmann-La Roche), a Ser/Thr kinase inhibitor from Lilly (EIi Lilly), CVT-2584 (CAS No. 199986-75- 9) (CV Therapeutics), and an analogue or derivative thereof.
13459. The method of item 13433 wherein the agent is a cyclooxygenase 1 inhibitor.
13460. The method of item 13433 wherein the agent is a DHFR inhibitor. 13461. The method of item 13433 wherein the agent is a dual integrin inhibitor.
13462. The method of item 13433 wherein the agent is an elastase inhibitor.
13463. The method of item 13433 wherein the agent is an elongation factor-1 alpha inhibitor.
13464. The method of item 13433 wherein the agent is an endothelial growth factor antagonist.
13465. The method of item 13433 wherein the agent is an endothelial growth factor receptor kinase inhibitor selected from the group consisting of sorafenib tosylate (Bayer), AAL-993 (Novartis), ABP-309 (Novartis), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), E- 7080 (Eisai), EG-3306 (Ark Therapeutics), EXEL-2880 (Exelixis), GW- 654652 (GlaxoSmithKline), a KDR inhibitor from LG Life Sciences, CT-6685 and CT-6729 (UCB), KRN-633 and KRN-951 (Kirin Brewery), OSI-930 (OSI Pharmaceuticals), SP-5.2 (Supratek Pharma), SU-11657 (Pfizer), a Tie-2 antagonist (Hybrigenics), a VEGFR-2 kinase inhibitor (Bristol-Myers Squibb), XL-647 (Exelixis), a KDR inhibitor from Abbott Laboratories, sorafenib tosylate, and an analogue or derivative thereof.
13466. The method of item 13433 wherein the agent is an endotoxin antagonist.
13467. The method of item 13433 wherein the agent is an epothilone and tubulin binder. 13468. The method of item 13433 wherein the agent is an estrogen receptor antagonist.
13469. The method of item 13433 wherein the agent is an FGF inhibitor.
13470. The method of item 13433 wherein the agent is a famexyl transferase inhibitor.
13471. The method of item 13433 wherein the agent is famesyltransferase inhibitor selected from the group of A-197574 (Abbott), a farnesyltransferase inhibitor from Servier, FPTIII (Strathclyde Institute for Drug R), LB-42908 (LG Life Sciences), Pharmaprojects No. 5063 (Genzyme), Pharmaprojects No. 5597 (Ipsen), Yissum Project No. B-1055 (Yissum), and an analogue or derivative thereof.
13472. The method of item 13433 wherein the agent is an FLT-3 kinase inhibitor.
13473. The method of item 13433 wherein the agent is an FGF receptor kinase inhibitor.
13474. The method of item 13433 wherein the agent is a fibrinogen antagonist selected from the group consisting of AUV-201 (Auvation), MG-13926 (Sanofi-Aventis), plasminogen activator (CAS No. 105913-11-9) (from Sanofi-Aventis or UCB), plasminogen activator-2 (tPA- 2) (Sanofi-Aventis), pro-urokinase (CAS No. 82657-92-9) (Sanofi-Aventis), and an analogue or derivative thereof.
13475. The method of item 13433 wherein the agent is a heat shock protein 90 antagonist selected from the group consisting of SRN- 005 (Sirenade), geldanamycin, NSC-33050 (17-allylaminogeldanamycin; 17- AAG), ^-dimethylaminoethylamino-^-demethoxy-geldanamycin (17- DMAG), rifabutin (rifamycin XIV, 1',4-didehydro-1-deoxy-1 ,4-dihydro-5'-(2- methylpropyl)-1-oxo-), and an analogue or derivative thereof.
13476. The method of item 13433 wherein the agent is a histone deacetylase inhibitor.
13477. The method of item 13433 wherein the agent is an HMGCoA reductase inhibitor selected from the group consisting of an atherosclerosis therapeutic from Lipid Sciences, ATI-16000 (ARYx Therapeutics), KS-01-019 (Kos Pharmaceuticals), Pharmaprojects No. 2197 (Sanofi-Aventi), RP 61969 (Sanofi-Aventis), and an analogue or derivative thereof.
13478. The method of item 13433 wherein the agent is an ICAM inhibitor.
13479. The method of item 13433 wherein the agent is an IL, ICE and IRAK antagonist, wherein the antagonist is a CJ-14877, CP- 424174 (Pfizer), NF-61 (Negma-Lerads), and an analogue or derivative thereof.
13480. The method of item 13433 wherein the agent is an IL-2 inhibitor.
13481. The method of item 13433 wherein the agent is an immunosuppressant selected from the group consisting of teriflunomide (Sanofi Aventis), chlorsulfaquinoxalone (NSC-339004), chlorsulfaquinoxalone sulfate, CS-712 (Sankyo), ismomultin alfa (CAS No. 457913-93-8) (Akzo Nobel), antiallergics from GenPat77, anti- inflammatories or AT-005 (Androclus Therapeutics), autoimmune disease therapeutics from EpiVax, BN-007 (Bone), budesonide (CAS No. 51333-22- 3) (MAP Pharmaceuticals), CO-14 (Genzyme), edratide (CAS No. 433922- 67-9) (Teva), EP-314 (Enanta), eprovafen (CAS No. 101335-99-3) (Sanofi- Aventis), HWA-131 (CAS No. 118788-41-3) (Sanofi-Aventis), immunomodulators from MerLion Pharmaceuticals, immunosuppressives from Alchemia, IPL-12 (Inflazyme), MDL-9563 (CAS No. 27086-86-8) (Sanofi-Aventis), Pharmaprojects No. 2330 (Sanofi-Aventis), Pharmaprojects No. 6426 (Abgenix), PXS-25 (Pharmaxis), rosmarinic acid (CAS No. 20283-92-5) (Sanofi-Aventis), RP 42927 or RP 54745 (CAS No. 135330-08-4) (Sanofi-Aventis), SGN-35 (Seattle Genetics), ST-1959 (Sigma-Tau), type I diabetes therapy from SYNX Pharma, UNIL-88 (Debiopharm), VP-025 (Vasogen), VR-694 (Vectura), PRTX-001 (Protalex), and an analogue or derivative thereof.
13482. The method of item 13433 wherein the agent is an IMPDH (inosine monophosphate).
13483. The method of item 13433 wherein the agent is an integrin antagonist.
13484. The method of item 13433 wherein the agent is an interleukin antagonist.
13485. The method of item 13433 wherein the agent is an inhibitor of type III receptor tyrosine kinase.
13486. The method of item 13433 wherein the agent is an irreversible inhibitor of enzyme methionine aminopeptidase type 2. 13487. The method of item 13433 wherein the agent is an isozyme selective delta protein kinase C inhibitor.
13488. The method of item 13433 wherein the agent a JAK3 enzyme inhibitor.
13489. The method of item 13433 wherein the agent is a
JNK inhibitor.
13490. The method of item 13433 wherein the agent is a kinase inhibitor.
13491. The method of item 13433 wherein the agent is kinesin antagonist.
13492. The method of item 13433 wherein the agent is a kinesin antagonist.
13493. The method of item 13433 wherein the agent is a leukotriene inhibitor and antagonist selected from the group consisting of ambicromil (CAS No. 58805-38-2) (Sanofi-Aventis), amelubant (CAS No. 346735-24-8) (Boehringer Ingelheim), DW-1141 (Dong Wha), ebselen (Daiichi Pharmaceutical), ibudilast (Kyorin), leucotriene inhibitors from Sanofi-Aventis, lymphotoxin-beta receptor (LT-D) from Biogen Idee, Pharmaprojects No. 1535 and 2728 (CAS No. 119340-33-9) (Sanofi- Aventis), R-112 (Rigel), Rev-5367 (CAS No. 92532-05-3) (Sanofi-Aventis), RG-14893 (CAS No. 141835-49-6) (Sanofi-Aventis), RG-5901-A (CAS No. 101910-24-1), 92532-23-5, RP 66153 (CAS No. 142422-79-5), RP 66364 (CAS No.186916-92-5), RP 69698 (CAS No. 141748-00-7) (Sanofi-Aventis), SC-411930 (Pfizer), SC-41930 (CAS No. 120072-59-5) (Pfizer), SC-50605 (CAS No. 138828-39-4) (Pfizer), SC-51146 (CAS No. 141059-52-1), SC- 53228 (CAS No. 153633-01-3) (Pfizer), spaglumic acid (ZY-15106) (CAS No.3106-85-2), 80619-64-3 (Novartis), tipredane (CAS No. 85197-77-9) (Bristol-Myers Squibb), U-75302 (CAS No. 119477-85-9) (Pfizer), and analogue or derivative thereof.
13494. The method of item 13433 wherein the agent is an MAP kinase inhibitor.
13495. The method of item 13433 wherein the agent is a matrix metalloproteinase inhibitor.
13496. The method of item 13433 wherein the agent is an MCP-CCR2 inhibitor.
13497. The method of item 13433 wherein the agent is an mTOR inhibitor. . . . — - - -
13498. The method of item 13433 wherein the agent is an mTOR kinase inhibitor.
13499. The method of item 13433 wherein the agent is a microtubule inhibitor selected from the group consisting of antibody- maytansinoid conjugates from Biogen Idee, colchicines (MantiCore Pharmaceuticals), anticancer immunoconjugates from Johnson & Johnson, DIME from Octamer, gni-1f (GNI), huC242-DM4, huMy9-6-DM1 (ImmunoGen), IDN-5404 (Indena), IMO-098, IMOderm (Imotep), mebendazole (Introgen Therapeutics), microtubule poisons from Cambridge Enterprise, paclitaxel such as LOTAX from Aphios (CAS No. 33069-62-4), Genexol-PM from Samyang, Pharmaprojects No. 6383 (Tapestry Pharmaceuticals), RPR-112378 (Sanofi-Aventis), SGN-75 (Seattle Genetics), SPL-7435 (Starpharma), SSR-250411 (Sanofi-Aventis), trastuzumab-DM1 (Genentech), vinorelbine, and an analogue or derivative thereof.
13500. The method of item 13433 wherein the agent is an MIF inhibitor.
13501. The method of item 13433 wherein the agent is an MMP inhibitor.
13502. The method of item 13433 wherein the agent is a neurokinin (NK) antagonist selected from the group consisting of anthrotainin (CAS No. 148084-40-6) (Sanofi-Aventis), an IBS thereapeutic from ArQuIe, MDL-105212A (CAS No. 167261-60-1) (Ssanofi-Aventis), Pharmaprojects No. 2744, 3258 (CAS No. 139167-47-8) 4006, 4201, or 5986 (Sanofi-Aventis), RP 67580 (CAS No. 135911-02-3), SR-144190 (CAS No. 201152-86-5), SSR-240600, SSR-241586 (Sanofi-Aventis), TKA-457 (Novartis), vestipitant mesylate (CAS No. 334476-64-1) (GlaxoSmithKline), Win-64821 (Sanofi-Aventis), PRX-96026 (Predix Pharmaceuticals), and an analogue or derivative thereof.
13503. The method of item 13433 wherein the agent is an NF kappa B inhibitor selected from the group consisting of AVE-0545 or AVE-0547 (Sanofi-Aventis), bortezomib (CAS No. 179324-69-7) (Millennium Pharmaceuticals), dexanabinol (CAS No. 112924-45-5) (Pharmos), dexlipotam (Viatris), Pharmaprojects No. 6283 (INDRA) (OXiGENE), IPL- 576092 (CAS No. 137571-30-3) (Inflazyme), NFKB decoy (Corgentech), NFKB decoy oligo (AnGes MG), NFKB's from Ariad, osteoporosis treatments or S5 (F005) from Fulcrum Pharmaceuticals, P61 (Phytopharm), R-flurbiprofen (CAS No. 5104-49-4) (Encore Pharmaceuticals), and an analogue or derivative thereof. 13504. The method of item 13433 wherein the agent is a nitric oxide agonist.
13505. The method of item 13433 wherein the agent is an ornithine decarboxylase inhibitor.
13506. The method of item 13433 wherein the agent is a p38 MAP kinase inhibitor selected from the group consisting of AZD-6703 (AstraZeneca), JX-401 (Jexys Pharmaceuticals), BMS-2 (Bristol-Myers Squibb), a p38 MAP kinase inhibitor from Novartis, a p38-alpha MAP kinase inhibitor from Amphora, Pharmaprojects No. 5704 (Pharmacopeia), RPR- 200765A (Sanofi-Aventis), SD-282 (Johnson & Johnson), TAK-715 (Takeda), and an analogue or derivative thereof.
13507. The method of item 13433 wherein the agent is a palmitoyl-protein thioesterase inhibitor.
13508. The method of item 13433 wherein the agent is a PDGF receptor kinase inhibitor selected from the group consisting of AAL- 993, AMN-107, or ABP-309 (Novartis), AMG-706 (Amgen), BAY-57-9352 (Bayer), CDP-860 (UCB), E-7080 (Eisai), imatinib (CAS No. 152459-95-5) (Novartis), OSI-930 (OSI Pharmaceuticals), RPR-127963E (Sanofi-Aventis), RWJ-540973 (Johnson & Johnson), sorafenib tosylate (Bayer), SU-11657 (Pfizer), tandutinib (CAS No. 387867-13-2) (Millennium Pharmaceuticals), vatalanib (Novartis), ZK-CDK (Schering AG), and an analogue or derivative thereof.
13509. The method of item 13433 wherein the agent is (-)-halofenate (Metabolex), AMG-131 (Amgen), antidiabetics from Japan Tobacco, AZD-4619, AZD-8450, AZD-8677 (AstraZeneca), DRF-10945, balaglitazone (Dr Reddy's), CS-00088, CS-00098 (Chipscreen Biosciences), E-3030 (Eisai), etalocib (CAS No. 161172-51-6) (EIi Lilly), GSK-641597 (Ligand), GSK-677954 (GlaxoSmithKline), GW-409544 (Ligand), GW- 590735 (GlaxoSmithKline), K-111 (Hoffmann-La Roche), LY-518674 (EIi Lilly), LY-674 (Ligand), LY-929 (Ligand), MC-3001 , MC-3002 (MaxoCore Pharmaceuticals), metformin HCI + pioglitazone (CAS No. 1115-70-4 and 112529-15-4), ACTOPLUS MET from Andrx), muraglitazar (CAS No. 331741-94-7) (Bristol-Myers Squibb), naveglitazar (Ligand), oleoylethanolamide (Kadmus Pharmaceuticals), ONO-5129, pioglitazone hydrochloride (CAS No. 111025-46-8 and 112529-15-4) (Takeda), PLX-204 (Plexxikon), PPAR agonists from Genfit, PPAR delta agonists from EIi Lilly, PPAR-alpha agonists from CrystalGenomics, PPAR-gamma modulators and PPAR-β modulators from CareX, rosiglitazone maleate (CAS No. 122324- 73-4 or 155141-29-0) (GlaxoSmithKline), rosiglitazone maleate/glimepir (CAS No. 155141-29-0 and 93479-97-1), AVANDARYL, rosiglitazone maleate/metformin extend (CAS No. 155141-29-0 and 657-24-9), AVANDAMET, rosiglitazone maleate+metformin, AVANDAMET (GlaxoSmithKline), tesaglitazar (AstraZeneca), and an analogue or derivative thereof.
13510. The method of item 13433 wherein the agent is a phosphatase inhibitor.
13511. The method of item 13433 wherein the agent is a phosphodiesterase (PDE) inhibitor selected from the group consisting of avanafil (Tanabe Seiyaku), dasantafil (CAS No. 569351-91-3) (Schering- Plough), A-906119 (CAS No. 134072-58-5), DL-850 (Sanofi-Aventis), GRC- 3015, GRC-3566, GRC-3886 (Glenmark), HWA-153 (CAS No. 56395-66-5) (Sanofi-Aventis), hydroxypumafentrine (Altana), IBFB-130011 , IBFB-14-016, IBFB-140301, IBFB-150007, IBFB-211913 (IBFB Pharma), L-826141 (Merck & Co), medorinone (CAS No. 88296-61-1) (Sanofi-Aventis), MEM- 1917 (Memory Pharmaceuticals), ND-1251 (Neuro3d), PDE inhibitors from ICOS1 PDE IV inhibitors from Memory Pharmaceuticals and CrystalGenomics, Pharmaprojects No. 2742 and 6141 (Sanofi-Aventis), QAD-171 (Novartis), RHC-2963 (CAS No. 76993-12-9 and 76993-14-1), RPR-117658, RPR-122818 derivatives, SR-24870 , and RPR-132294 (Sanofi-Aventis), SK-350 (ln2Gen), stroke therapy agents from deCODE Genetics, TAS-203 (Taiho), tofimilast (CAS No. 185954-27-2) (Pfizer), UK- 371800 (Pfizer), WIN-65579 (CAS No! 158020-82-7) (Sanofi-Aventis), IBFB- 130020 (IBFB Pharma), OPC-6535 (CAS No. 145739-56-6) (Otsuka), a phosphodiesterase III inhibitor, enoximone, a phosphodiesterase IV inhibitor, fosfosal, Atopik (Barrier Therapeutics), triflusal, a phosphodiesterase V inhibitor, and an analogue or derivative thereof.
13512. The method of item 13433 wherein the agent is a PKC inhibitor.
13513. The method of item 13433 wherein the agent is a platelet activating factor antagonist.
13514. The method of item 13433 wherein the agent is a platelet-derived growth factor receptor kinase inhibitor.
13515. The method of item 13433 wherein the agent is a prolyl hydroxylase inhibitor.
13516. The method of item 13433 wherein the agent is a polymorphonuclear neutrophil inhibitor.
13517. The method of item 13433 wherein the agent is a protein kinase B inhibitor. 13518. The method of item 13433 wherein the agent is a protein kinase C stimulant.
13519. The method of item 13433 wherein the agent is a purine nucleoside analogue.
13520. The method of item 13433 wherein the agent is a purinoreceptor P2X antagonist.
13521. The method of item 13433 wherein the agent is a Raf kinase inhibitor.
13522. The method of item 13433 wherein the agent is a reversible inhibitor of ErbB1 and ErbB2.
13523. The method of item 13433 wherein the agent is a ribonucleoside triphosphate reductase inhibitor.
13524. The method of item 13433 wherein the agent is an SDF-1 antagonist.
13525. The method of item 13433 wherein the agent is a sheddase inhibitor.
13526. The method of item 13433 wherein the agent is an SRC inhibitor.
13527. The method of item 13433 wherein the agent is a stromelysin inhibitor. 13528. The method of item 13433 wherein the agent is an Syk kinase inhibitor.
13529. The method of item 13433 wherein the agent is a telomerase inhibitor.
13530. The method of item 13433 wherein the agent is a TGF beta inhibitor selected from the group consisting of pirfenidone (CAS No. 53179-13-8) (MARNAC), tranilast (CAS No. 53902-12-8) (Kissei), IN-
1130 (ln2Gen), mannose-6-phosphate (BTG), TGF-β antagonists from Inflazyme (Pharmaprojects No. 6075), TGF-β antagonists from Sydney, non-industrial source), TGF-β I receptor kinase inhibitors from EIi Lilly, TGF- β receptor inhibitors from Johnson & Johnson, and an analogue or derivative thereof.
13531. The method of item 13433 wherein the agent is a TNFα antagonist or TACE inhibitor selected from the group consisting of adalimumab (CAS No. 331731-18-1) (Cambridge Antibody Technology), AGIX-4207 (AtheroGenics), AGT-1 (Advanced Biotherapy), an antiinflammatory from Borean Pharma, Cellzome, or Paradigm Therapeutics, anti-inflammatory vaccine (TNF-alpha kinoid) from Neovacs, humanized anti-TNF antibody or an anti-TNF MAb (CB0006) Celltech (UCB), apratastat (CAS No. 287445-51-0) (Wyeth), BMS-561392 (Bristol-Myers Squibb), BN- 006 (Bone), certolizumab pegol (CAS No. 428863-50-7 or CH-138 (UCB), cilomilast (CAS No. 153259-65-5) (GlaxoSmithKline), CR-1 (Nuada Pharmaceuticals), CRx-119 (CombinatoRx), D-5410 (UCB), dacopafant (CAS No. 125372-33-0) (Sanofi-Aventis), dersalazine (CAS No. 188913-57- 7/188913-58-8) (Uriach), etanercept (CAS No. 185243-69-0) (Amgen), ethyl pyruvate (Critical (Critical Therapeutics), golimumab (CAS No. 476181-74-5) (Johnson & Johnson), hormono-immunotherapy from Ipsen, CDP571 {e.g., Humicade from UCB), IC-485 (ICOS), infliximab (CAS No. 170277-31-3) (Johnson & Johnson), IP-751 (Manhattan Pharmaceuticals), ISIS-104838 (CAS No. 250755-32-9) (ISIS Pharmaceuticals), lenalidomide (CAS No. 191732-72-6) (Celgene), lentinan (CAS No. 37339-90-5) (Ajinomoto), MDL- 201112 (CAS No. 142130-73-2) (Sanofi-Aventis), medroxyprogesterone (CAS No. 520-85-4) (InKine Pharmaceutical), N-acetylcysteine (CAS No. 616-91-1) (Zambon), NBE-P2 (DIREVO Biotech), nerelimomab (CAS No. 162774-06-3) (Chiron), OM-294DP (OM PHARMA), onercept (CAS No. 199685-57-9) (Yeda), PASSTNF-alpha (Verigen), pentoxifylline or oxypentifylline (Sanofi-Aventis), Pharmaprojects No. 4091 , 4241, 4295, or 4488 (Sanofi-Aventis), Pharmaprojects No. 5480 (Amgen), Pharmaprojects No. 6749 (Cengent), pirfenidone (CAS No. 53179-13-8) (MARNAC), RPR- 132294 (Sanofi-Aventis), S5 (F002) (Fulcrum Pharmaceuticals), simvastatin (CAS No. 79902-63-9) (Merck & Co), STA-6292 (Synta Pharmaceuticals), tacrolimus (CAS No. 104987-11-3) (Fujisawa LifeCycle Pharma), talactoferrin alfa (CAS No. 308240-58-6) (Agennix), thalidomide (CAS No. 50-35-1) (Celgene), TNF antagonists form ProStrakan, and Synergen, TNF inhibitors (Amgen), TNF-alpha antagonists from Dynavax Technologies and Jerina AG (Germany), TNF-alpha inhibitors from IBFB Pharma and Xencor (Xencor), torbafylline (CAS No. 105102-21-4) (Sanofi-Aventis), UR-1505 (Uriach), VT-346 (Won Therapeutics), YSIL6 (Y's Therapeutics), YSTH2 (Y's Therapeutics), NPI-1302a-3 (Nereus Pharmaceuticals, a TNF antagonist from Jerina AG (Germany), dersalazine, and an analogue or derivative thereof.
13532. The method of item 13433 wherein the agent is a Toll receptor inhibitor.
13533. The method of item 13433 wherein the agent is a tubulin antagonist. 13534. The method of item 13433 wherein the agent is a tyrosine kinase inhibitor selected from the group consisting of SU-011248, SUTENT from Pfizer Inc. (New York, NY)1 BMS-354825, PN-355 (Paracelsian Pharmaceuticals), AGN-199659 (Allergan), AAL-993 or ABP- 309 (Novartis), adaphostin (NIH), AEE-788 (Novartis), AG-013736 (OSI Pharmaceuticals), AG-13736 (Pfizer), ALT-110 (Alteris Therapeutics), AMG- 706 (Amgen), anticancer MAbs from Xencor, anti-EGFrvlll MAbs from Abgenix, anti-HER2 MAb from Abiogen, AZD-2171 or AZD-9935 (AstraZeneca), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), CEP-5214 (Cephalon), CEP-7055 (Cephalon), cetuximab (ImClone Systems), CHIR-200131 and CHIR-258 (Chiron), CP-547632 (OSI Pharmaceuticals), CP-724714 (Pfizer), CT-301 (Creabilis Therapeutics), D- 69491 (Baxter International), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EGFR/ErbB2 inhibitors from Array BioPharma, erlotinib (CAS No. 183319- 69-9) (OSI Pharmaceuticals), EXEL-2880 (Exelixis), FK-778 (Sanofi- Aventis) , gefitinib (CAS No. 184475-35-2) (AstraZeneca), GW-2286 or GW- 654652 (GlaxoSmithKline), her2/neu antigen from AlphaVax, HER-2/neu inhibitor from Generex, Herzyme (Medipad) (Sirna Therapeutics), HKI-272 (Wyeth), HuMax-EGFr (Genmab), idronoxil (CAS No. 81267-65-4) (Novogen), IGF-1 inhibitors from Ontogen, IMC-11F8 (ImClone Systems), kahalalide F (CAS No. 149204-42-2) (PharmaMar), KDR inhibitor from LG Life Sciences, KDR inhibitors from Abbott Laboratories, KDR kinase inhibitors (UCB), Kdr kinase inhibitors from Merck & Co, KRN-633 and KRN- 951 (Kirin Brewery), KSB-102 (Xenova), lapatinib ditosylate (CAS No. 388082-78-8) (GlaxoSmithKline), matuzumab (Merck KGaA), MDX-214 (Medarex), ME-103 (Pharmexa), MED-A300 (Gerolymatos), MNAC-13 (Lay Line Genomics), nimotuzumab (Center of Molecular Immunology), NSC- 330507 or NSC-707545 (NIH)1 NV-50 (Novogen), OSI-930 (OSI Pharmaceuticals), panitumumab (Abgenix), pelitinib (CAS No. 287933-82-7) (Wyeth), pertuzumab (CAS No. 380610-27-5) (Genentech), Pharmaprojects No. 3985 (Sanofi-Aventis), prostate cancer therapeutics from Sequenom (SQPC35, SQPC36, SQPC90), removab and remoxab (Trion Pharma), RG- 13022 (CAS No. 136831-48-6), RG-13291 (CAS No. 138989-50-1), or RG- 14620 (CAS No. 136831-49-7) (Sanofi-Aventis), RM-6427 (Romark), RNAi breast cancer therapy from Benitec, RP 53801 (CAS No. 125882-88-4) (Sanofi-Aventis), sorafenib tosylate (Bayer), SU-11657 (Pfizer), Tie-2 antagonists from Semaia (Hybrigenics), Tie-2 inhibitors from Ontogen, trastuzumab (CAS No. 180288-69-1) (Genentech), tyrosine kinase inhibitors from Sanofi-Aventis, U3-1287, U3-1565, U3-1784, U3-1800 (U3 Pharma), vatalanib (Novartis), VEGFR-2 kinase inhibitor from Bristol-Myers Squibb, XL-647 (Exelixis), ZD-6474 (AstraZeneca), ZK-CDK (Schering AG), an EGFR tyrosine kinase inhibitor, EKB-569 (Wyeth), and an analogue or derivative thereof.
13535. The method of item 13433 wherein the agent is a VEGF inhibitor.
13536. The method of item 13433 wherein the agent is a vitamin D receptor agonist.
13537. The method of item 13433 wherein the agent is ZD-6474 (an angiogenesis inhibitor).
13538. The method of item 13433 wherein the agent is AP-23573 (an mTOR inhibitor).
13539. The method of item 13433 wherein the agent is synthadotin (a tubulin antagonist).
13540. The method of item 13433 wherein the agent is S-0885 (a collagenase inhibitor). 13541. The method of item 13433 wherein the agent is aplidine (an elongation factor-1 alpha inhibitor).
13542. The method of item 13433 wherein the agent is ixabepilone (an epithilone).
13543. The method of item 13433 wherein the agent is IDN-5390 (an angiogenesis inhibitor).
13544. The method of item 13433 wherein the agent is SB-2723005 (an angiogenesis inhibitor).
13545. The method of item 13433 wherein the agent is ABT-518 (an angiogenesis inhibitor).
13546. The method of item 13433 wherein the agent is combretastatin (an angiogenesis inhibitor).
13547. The method of item 13433 wherein the agent is anecortave acetate (an angiogenesis inhibitor).
13548. The method of item 13433 wherein the agent is SB-715992 (a kinesin antagonist).
13549. The method of item 13433 wherein the agent is temsirolimus (an mTOR inhibitor).
13550. The method of item 13433 wherein the agent is adalimumab (a TNFα antagonist). 13551. The method of item 13433, wherein the composition comprises a polymer.
13552. The method of item 13433, wherein the composition comprises a polymeric carrier.
13553. The method of item 13433 wherein the anti- scarring agent inhibits adhesion between the device and a host into which the device is implanted.
13554. The method of item 13433 wherein the device delivers the anti-scarring agent locally to tissue proximate to the device.
13555. The method of item 13433 wherein the device has a coating that comprises the anti-scarring agent.
13556. The method of item 13433, wherein the device has a coating that comprises the agent and is disposed on a surface of the implant.
13557. The method of item 13433, wherein the device has a coating that comprises the agent and directly contacts the implant.
13558. The method of item 13433, wherein the device has a coating that comprises the agent and indirectly contacts the implant.
13559. The method of item 13433, wherein the device has a coating that comprises the agent and partially covers the implant.
13560. The method of item 13433, wherein the device has a coating that comprises the agent and completely covers the implant. 13561. The method of item 13433, wherein the device has a uniform coating.
13562. The method of item 13433, wherein the device has a non-uniform coating.
13563. The method of item 13433, wherein the device has a discontinuous coating.
13564. The method of item 13433, wherein the device has a patterned coating.
13565. The method of item 13433, wherein the device has a coating with a thickness of 100 Dm or less.
13566. The method of item 13433, wherein the device has a coating with a thickness of 10 Dm or less.
13567. The method of item 13433, wherein the device has a coating, and the coating adheres to the surface of the implant upon deployment of the implant.
13568. The method of item 13433, wherein the device has a coating, and wherein the coating is stable at room temperature for a period of 1 year.
13569. The method of item 13433, wherein the device has a coating, and wherein the anti-scarring agent is present in the coating in an amount ranging between about 0.0001% to about 1% by weight. 13570. The method of item 13433, wherein the device has a coating, and wherein the anti-scarring agent is present in the coating in an amount ranging between about 1% to about 10% by weight.
13571. The method of item 13433, wherein the device has a coating, and wherein the anti-scarring agent is present in the coating in an amount ranging between about 10% to about 25% by weight.
13572. The method of item 13433, wherein the device has a coating, and wherein the anti-scarring agent is present in the coating in an amount ranging between about 25% to about 70% by weight.
13573. The method of item 13433, wherein the device has a coating, and wherein the coating further comprises a polymer.
13574. The method of item 13433, wherein the device has a first coating having a first composition and a second coating having a second composition.
13575. The method of item 13433, wherein the device has a first coating having a first composition and a second coating having a second composition, wherein the first composition and the second composition are different.
13576. The method of item 13433, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a copolymer.
13577. The method of item 13433, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a block copolymer. 13578. The method of item 13433, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a random copolymer.
13579. The method of item 13433, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a biodegradable polymer.
13580. The method of item 13433, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a non-biodegradable polymer.
13581. The method of item 13433, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a hydrophilic polymer.
13582. The method of item 13433, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a hydrophobic polymer.
13583. The method of item 13433, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a polymer having hydrophilic domains.
13584. The method of item 13433, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a polymer having hydrophobic domains.
13585. The method of item 13433, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a non-conductive polymer. 13586. The method of item 13433, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises an elastomer.
13587. The method of item 13433, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a hydrogel.
13588. The method of item 13433, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a silicone polymer.
13589. The method of item 13433, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a hydrocarbon polymer.
13590. The method of item 13433, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a styrene-derived polymer.
13591. The method of item 13433, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a butadiene polymer.
13592. The method of item 13433, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a macromer.
13593. The method of item 13433, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a poly(ethylene glycol) polymer. 13594. The method of item 13433 wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises an amorphous polymer.
13595. The method of item 13433, wherein the device comprises a lubricious coating.
13596. The method of item 13433 wherein the anti- scarring agent is located within pores or holes of the device.
13597. The method of item 13433 wherein the anti- scarring agent is located within a channel, lumen, or divet of the device.
13598. The method of item 13433, wherein the device comprises a second pharmaceutically active agent.
13599. The method of item 13433 wherein the device comprises an anti-inflammatory agent.
13600. The method of item 13433 wherein the device comprises an agent that inhibits infection.
13601. The method of item 13433 wherein the device comprises an agent that inhibits infection, and wherein the agent is an anthracycline.
13602. The method of item 13433 wherein the device comprises an agent that inhibits infection, and wherein the agent is doxorubicin. 13603. The method of item 13433 wherein the device comprises an agent that inhibits infection, and wherein the agent is mitoxantrone.
13604. The method of item 13433 wherein the device comprises an agent that inhibits infection, and wherein the agent is a fluoropyrimidine.
13605. The method of item 13433 wherein the device comprises an agent that inhibits infection, and wherein the agent is 5- fluorouracil (5-FU).
13606. The method of item 13433 wherein the device comprises an agent that inhibits infection, and wherein the agent is a folic acid antagonist.
13607. The method of item 13433 wherein the device comprises an agent that inhibits infection, and wherein the agent is methotrexate.
13608. The method of item 13433 wherein the device comprises an agent that inhibits infection, and wherein the agent is a podophylotoxin.
13609. The method of item 13433 wherein the device comprises an agent that inhibits infection, and wherein the agent is etoposide.
13610. The method of item 13433 wherein the device comprises an agent that inhibits infection, and wherein the agent is a camptothecin. 13611. The method of item 13433 wherein the device comprises an agent that inhibits infection, and wherein the agent is a hydroxyurea.
13612. The method of item 13433 wherein the device comprises an agent that inhibits infection, and wherein the agent is a platinum complex.
13613. The method of item 13433 wherein the device comprises an agent that inhibits infection, and wherein the agent is cisplatin.
13614. The method of item 13433, further comprising an anti-thrombotic agent.
13615. The method of item 13433 wherein the device comprises a visualization agent.
13616. The method of item 13433 wherein the device comprises a visualization agent, wherein the visualization agent is a radiopaque material, and wherein the radiopaque material comprises a metal, a halogenated compound, or a barium containing compound.
13617. The method of item 13433 wherein the device comprises a visualization agent, wherein the visualization agent is a radiopaque material, and wherein the radiopaque material comprises barium, tantalum, or technetium.
13618. The method of item 13433 wherein the device comprises a visualization agent, and wherein the visualization agent is a MRI responsive material. 13619. The method of item 13433 wherein the device comprises a visualization agent, and wherein the visualization agent comprises a gadolinium chelate.
13620. The method of item 13433 wherein the device comprises a visualization agent, and wherein the visualization agent comprises iron, magnesium, manganese, copper, or chromium.
13621. The method of item 13433 wherein the device comprises a visualization agent, and wherein the visualization agent comprises an iron oxide compound.
13622. The method of item 13433 wherein the device comprises a visualization agent, and wherein the visualization agent comprises a dye, pigment, or colorant.
13623. The method of item 13433 wherein the device comprises an echogenic material.
13624. The method of item 13433 wherein the device comprises an echogenic material, and wherein the echogenic material is in the form of a coating.
13625. The method of item 13433 wherein the device is sterile.
13626. The method of item 13433 wherein the anti- scarring agent is released into tissue in the vicinity of the device after deployment of the device. 13627. The method of item 13433 wherein the anti- scarring agent is released into tissue in the vicinity of the device after deployment of the device, and wherein the tissue is connective tissue.
13628. The method of item 13433 wherein the anti- scarring agent is released into tissue in the vicinity of the device after deployment of the device, and wherein the tissue is muscle tissue.
13629. The method of item 13433 wherein the anti- scarring agent is released into tissue in the vicinity of the device after deployment of the device, and wherein the tissue is nerve tissue.
13630. The method of item 13433 wherein the anti- scarring agent is released into tissue in the vicinity of the device after deployment of the device, and wherein the tissue is epithelium tissue.
13631. The method of item 13433 wherein the anti- scarring agent is released in effective concentrations from the device over a period ranging from the time of deployment of the device to about 1 year.
13632. The method of item 13433 wherein the anti- scarring agent is released in effective concentrations from the device over a period ranging from about 1 month to 6 months.
13633. The method of item 13433 wherein the anti- scarring agent is released in effective concentrations from the device over a period ranging from about 1 - 90 days.
13634. The method of item 13433 wherein the anti- scarring agent is released in effective concentrations from the device at a constant rate. 13635. The method of item 13433 wherein the anti- scarring agent is released in effective concentrations from the device at an increasing rate.
13636. The method of item 13433 wherein the anti- scarring agent is released in effective concentrations from the device at a decreasing rate.
13637. The method of item 13433 wherein the anti- scarring agent is released in effective concentrations from the composition comprising the anti-scarring agent by diffusion over a period ranging from the time of deployment of the device to about 90 days.
13638. The method of item 13433 wherein the anti- scarring agent is released in effective concentrations from the composition comprising the anti-scarring agent by erosion of the composition over a period ranging from the time of deployment of the device to about 90 days.
13639. The method of item 13433 wherein the device comprises about 0.01 Dg to about 10 Dg of the anti-scarring agent.
13640. The method of item 13433 wherein the device comprises about 10 Dg to about 10 mg of the anti-scarring agent.
13641. The method of item 13433 wherein the device comprises about 10 mg to about 250 mg of the anti-scarring agent.
13642. The method of item 13433 wherein the device comprises about 250 mg to about 1000 mg of the anti-scarring agent. 13643. The method of item 13433 wherein the device comprises about 1000 mg to about 2500 mg of the anti-scarring agent.
13644. The method of item 13433 wherein a surface of the device comprises less than 0.01 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
13645. The method of item 13433 wherein a surface of the device comprises about 0.01 Dg to about 1 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
13646. The method of item 13433 wherein a surface of the device comprises about 1 Dg to about 10 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
13647. The method of item 13433 wherein a surface of the device comprises about 10 Dg to about 250 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
13648. The method of item 13433 wherein a surface of the device comprises about 250 Dg to about 1000 Dg of the anti-scarring agent of anti-scarring agent per mm2 of device surface to which the anti- scarring agent is applied.
13649. The method of item 13433 wherein a surface of the device comprises about 1000 Dg to about 2500 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
13650. The method of item 13433 wherein the combining is performed by direct affixing the agent or the composition to the implant. 13651. The method of item 13433 wherein the combining is performed by spraying the agent or the component onto the implant.
13652. The method of item 13433 wherein the combining is performed by electrospraying the agent or the composition onto the implant.
13653. The method of item 13433 wherein the combining is performed by dipping the implant into a solution comprising the agent or the composition.
13654. The method of item 13433 wherein the combining is performed by covalently attaching the agent or the composition to the implant.
13655. The method of item 13433 wherein the combining is performed by non-covalently attaching the agent or the composition to the implant.
13656. The method of item 13433 wherein the combining is performed by coating the implant with a substance that contains the agent or the composition.
13657. The method of item 13433 wherein the combining is performed by coating the implant with a substance that absorbs the agent.
13658. The method of item 13433 wherein the combining is performed by interweaving a thread composed of, or coated with, the agent or the composition. 13659. The method of item 13433 wherein the combining is performed by covering all the implant with a sleeve that contains the agent or the composition.
13660. The method of item 13433 wherein the combining is performed by covering a portion of the implant with a sleeve that contains the agent or the composition.
13661. The method of item 13433 wherein the combining is performed by covering all the implant with a cover that contains the agent or the composition.
13662. The method of item 13433 wherein the combining is performed by covering a portion of the implant with a cover that contains the agent or the composition.
13663. The method of item 13433 wherein the combining is performed by covering all the implant with an electrospun fabric that contains the agent or the composition.
13664. The method of item 13433 wherein the combining is performed by covering a portion of the implant with an electrospun fabric that contains the agent or the composition.
13665. The method of item 13433 wherein the combining is performed by covering all the implant with a mesh that contains the agent or the composition.
13666. The method of item 13433 wherein the combining is performed by covering a portion of the implant with a mesh that contains the agent or the composition. 13667. The method of item 13433 wherein the combining is performed by constructing all the implant with the agent or the composition.
13668. The method of item 13433 wherein the combining is performed by constructing a portion of the implant with the agent or the composition.
13669. The method of item 13433 wherein the combining is performed by impregnating the implant with the agent or the composition.
13670. The method of item 13433 wherein the combining is performed by constructing all of the implant from a degradable polymer that releases the agent.
13671. The method of item 13433 wherein the combining is performed by constructing a portion of the implant from a degradable polymer that releases the agent.
13672. The method of item 13433 wherein the combining is performed by dipping the implant into a solution that comprise the agent and an inert solvent for the implant.
13673. The method of item 13433 wherein the combining is performed by dipping the implant into a solution that comprises the agent and a solvent that will swell the implant.
13674. The method of item 13433 wherein the combining is performed by dipping the implant into a solution that comprises the agent and a solvent that will dissolve the implant. 13675. The method of item 13433 wherein the combining is performed by dipping the implant into a solution that comprises the agent, a polymer and an inert solvent for the implant.
13676. The method of item 13433 wherein the combining is performed by dipping the implant into a solution that comprises the agent, a polymer and a solvent that will swell the implant.
13677. The method of item 13433 wherein the combining is performed by dipping the implant into a solution that comprises the agent, a polymer and a solvent that will dissolve the implant.
13678. The method of item 13433 wherein the combining is performed by spraying the implant into a solution that comprises the agent and an inert solvent for the implant.
13679. The method of item 13433 wherein the combining is performed by spraying the implant into a solution that comprises the agent and a solvent that will swell the implant.
13680. The method of item 13433 wherein the combining is performed by spraying the implant into a solution that comprises the agent and a solvent that will dissolve the implant.
13681. The method of item 13433 wherein the combining is performed by spraying the implant into a solution that comprises the agent, a polymer and an inert solvent for the implant.
13682. The method of item 13433 wherein the combining is performed by spraying the implant into a solution that comprises the agent, a polymer and a solvent that will swell the implant. 13683. The method of item 13433 wherein the combining is performed by spraying the implant into a solution that comprises the agent, a polymer and a solvent that will dissolve the implant.
13684. A method as in any one of items 13433-13683, wherein the device is an extravascυlar graft.
13685. A method as in any one of items 13433-13683, wherein the device is an intravascular graft.
13686. A method as in any one of items 13433-13683, wherein the device is a vascular graft adapted for replacing a blood vessel damaged by aneurysm.
13687. A method as in any one of items 13433-13683, wherein the device is a vascular graft adapted for replacing a blood vessel damaged by intimal hyperplasia.
13688. A method as in any one of items 13433-13683, wherein the device is a vascular graft adapted for replacing a blood vessel damaged by thrombosis.
13689. A method as in any one of items 13433-13683, wherein the device is a vascular graft adapted for providing access to blood vessel.
13690. A method as in any one of items 13433-13683, wherein the device is a vascular graft adapted for providing an alternative conduit for blood flow through a damaged or diseased area in a vein. 13691. A method as in any one of items 13433-13683, wherein the device is a vascular graft adapted for providing an alternative conduit for blood flow through a damaged or diseased area in an artery.
13692. A method as in any one of items 13433-13683, wherein the device is a synthetic bypass graft.
13693. A method as in any one of items 13433-13683, wherein the device is afemoral-popliteal bypass graft.
13694. A method as in any one of items 13433-13683, wherein the device is a femoral-femoral bypass graft.
13695. A method as in any one of items 13433-13683, wherein the device is an axillary-femoral bypass graft.
13696. A method as in any one of items 13433-13683, wherein the device is a vein graft.
13697. A method as in any one of items 13433-13683, wherein the device is a peripheral vein graft.
13698. A method as in any one of items 13433-13683, wherein the device is a coronary vein graft.
13699. A method as in any one of items 13433-13683, wherein the device is an internal mammary graft.
13700. A method as in any one of items 13433-13683, wherein the device is a bifurcated vascular graft. 13701. A method as in any one of items 13433-13683, wherein the device is an intraluminal graft.
13702. A method as in any one of items 13433-13683, wherein the device is a prosthetic vascular graft.
13703. A method of making a medical device comprising: combining a hemodialysis access device (e.g., an implant) and an anti-scarring agent or a composition comprising an anti-scarring agent, wherein the agent inhibits scarring between the device and a host into which the device is implanted.
13704. The method of item 13703 wherein the agent is an adensosine A2A receptor antagonist.
13705. The method of item 13703 wherein the agent is an AKT inhibitor.
13706. The method of item 13703 wherein the agent is an alpha 2 integrin antagonist, wherein the alpha 2 integrin antagonist is Pharmaprojects No. 5754 (Merck KgaA).
13707. The method of item 13703 wherein the agent is an alpha 4 integrin antagonist.
13708. The method of item 13703 wherein the agent is an alpha 7 nicotinic receptor agonist.
13709. The method of item 13703 wherein the agent is an angiogenesis inhibitor selected from the group consisting of AG-12,958 (Pfizer), ATN-161 (Attenuon LLC), neovastat, an angiogenesis inhibitor from Jerina AG (Germany), NM-3 (Mercian), VGA-1155 (Taisho), FCE-26644 (Pfizer), FCE-26950 (Pfizer), FPMA (Meiji Daries), FR-111142 (Fujisawa), GGTI-298, GM-1306 (Ligand), GPA-1734 (Novartis), NNC-47-0011 (Novo Nordisk), herbamycin (Nippon Kayaku), lenalidomide (Celegene), IP-10 (NIH), ABT-828 (Abbott), KIN-841 (Tokushima University, Japan), SF-1126 (Semafore Pharmaceuticals), laminin technology (NIH), CHIR-258 (Chiron), NVP-AEW541 (Novartis), NVP-AEW541 (Novartis), Vt16907 (Alchemia), OXI-8007 (Oxigene), EG-3306 (Ark Therapeutics), Maspin (Arriva), ABT- 567 (Abbott), PPI-2458 (Praecis Pharmaceuticals), CC-5079, CC-4089 (Celgene), HIF-1 alpha inhibitors (Xenova), S-247 (Pfizer), AP-23573 (Ariad), AZD-9935 (Astra Zeneca), mebendazole (Introgen Therapeutics), MetAP-2 inhibitors (GlaxoSmithKline), AG-615 (Angiogene Pharmaceuticals), Tie-2 antagonists (Hybrigenics), NC-381 , CYC-381, NC-169, NC-219, NC-383, NC-384, NC-407 (Lorus Therapeutics), ATN-224 (Attenuon), ON-01370 (Onconova), Vitronectin antagonists (Amgen), SDX-103 (Salmedix), Vitronectin antagonists (Shire), CHP (Riemser), TEK (Amgen), Anecortave acetate (Alcon), T46.2 (Matrix Therapeutics), HG-2 (Heptagen), TEM antagonists (Genzyme), Oxi-4500 (Oxigene), ATN-161 (Attenuon), WX-293 (Wilex), M-2025 (Metris Therapeutics), Alphastatin (BioActa), YH-16 (Yantai Rongchang), BIBF-1120 (Boehringer Ingelheim), BAY-57-9352 (Bayer), AS- 1404 (Cancer Research Technology), SC-77964 (Pfizer), glycomimetics (BioTie Therapies), TIE-2 Inhibitors (Ontogen), DIMI, Octamer (Octamer), ABR-215050 (Active Biotech), ABT-518 (Abbott), KDR inhibitors (Abbott), BSF-466895 (Abbott), SCH-221153 (Schering-Plough), DAC:antiangiogenic (ConjuChem), TFPI (EntreMed), AZD-2171 (Astra-Zenaca), CDC-394 (Celgene), LY290293 (EIi Lilly), IDN-5390 (Indena), Kdr Kinase Inhibitors (Merck), CT-113020, CT-116433, CT-116563, CT-31890, CT-32228) (Cell Therapeutics), A-299620 (Abbott), TWEAK Inhibitor (Amgen), VEGF modulators (Johnson and Johnson), Tum-N53, tumstatin (Genzyme), Thios- 1 , Thios-2 (Thios Pharmaceuticals), MV-6401 (Miravant Medical Technologies), Spisulosine (PharmaMar), CEP-7055 (Cephalon), AUV-201 (Auvation), LM-609 (EIi Lilly), SKF-106615 (AnorMED), Oglufanide disodium (Cytran), BW-114 (Pharminox), Calreticulin (NIH), WX-678 (Wilex), SD-7784 (Pfizer), WX-UK1 (Wilex), SH-268 (Schering AG), 2-Me-PGA (Celgeπe), S- 137 (Pfizer), ZD-6126 (Angiogene Pharmaceuticals), SG-292 (SignalGen), Benefin (Lane Labs), A6, A36 (Angstrom), SB-2723005 (GlaxoSmithKline), SC-7 (Cell Therapeutics), ZEN-014 (AEterna Zentaris), 2-methoxyestradiol (EntreMed), NK-130119 (Nippon Kayaku), CC-10004 (Celgene), AVE- 8062A (Ajinomoto), Tacedinaline (Pfizer), Actinonin (Tokyo Metropolitan Institute of Medical Science), Lenalidomide (Celgene), VGA-1155, BTO-956 (SRI International), ER-68203-00 (Eisai), CT-6685 (UCB), JKC-362 (Phoenix Pharmaceuticals), DMI-3798 (DMI Biosciences, Angiomate (Ipsen), ZD-6474 (AstraZeneca), CEP-5214 (Cephalon), Canstatin (Genzyme), NM-3 (Mercian), Oridigm (MediQuest Therapeutics), Exherin (Adherex), BLS-0597 (Boston Life Sciences), PTC-299 (PTC Therapeutics), NPI-2358 (Nereus Pharmaceuticals), CGP-79787 (Novartis), AEE-788 (Novartis), CKD-732 (Chong Kun Dang), CP-564959 (OSI Pharmaceuticals), CM-101 (CarboMed), CT-2584, CT3501 (Cell Therapeutics), combretastatin and analogues and derivatives thereof (Oxigene), Rebimastat (Bristol-Meyers Squibb), Dextrin 2-sulfate (ML Laboratories), Cilengitide (Merk KGaA), NSC-706704 (Pharminox), KRN- 951 (Kirin Brewery), Ukrain, NSC-631570 (Nowicky Pharma), Tecogalan sodium (Daiichi Pharmaceutical), Tz-93 (Tsumura), TBC-1635 (Encysive Pharmaceuticals), TAN-1120 (Takeda), Semaxanib (Pfizer), BDI-7800 (Biopharmacopae), SD-186, SD-983 (Bristol-Meyers Squibb), SB-223245 (GlaxoSmithKline), SC-236 (Pfizer), RWJ-590973 (Johnson and Johnson), ILX-1850 (Genzyme), SC-68488, S-836 (Pfizer), CG-55069-11 (CuraGen), Ki-23057 (Kirin Brewery), CCX-700 (Chemoentryx), Pegaptanib octasodium (Giled Sciences), ANGIOCOL (available from Biostratum Inc.), or an analogue or derivative thereof. 13710. The method of item 13703 wherein the agent is an apoptosis antagonist.
13711. The method of item 13703 wherein the agent is an apoptosis activator.
13712. The method of item 13703 wherein the agent is a beta 1 integrin antagonist.
13713. The method of item 13703 wherein the agent is a beta tubulin inhibitor.
13714. The method of item 13703 wherein the agent is a blocker of enzyme production in Hepatitis C.
13715. The method of item 13703 wherein the agent is a Bruton's tyrosine kinase inhibitor.
13716. The method of item 13703 wherein the agent is a calcineurin inhibitor.
13717. The method of item 13703 wherein the agent is a caspase 3 inhibitor.
13718. The method of item 13703 wherein the agent is a CC chemokine receptor antagonist.
13719. The method of item 13703 wherein the agent is a cell cycle inhibitor selected from the group consisting of SNS-595 (Sunesis), synthadotin, KRX-0403, and an analogue or derivative thereof. 13720. The method of item 13703 wherein the agent is a cathepsin B inhibitor.
13721. The method of item 13703 wherein the agent is a cathepsin K inhibitor, wherein the cathepsin K inhibitor is 462795 (GlaxoSmithKline), INPL-022-D6 (Amura Therapeutics), or an analogue or derivative thereof.
13722. The method of item 13703 wherein the agent is a cathepsin L inhibitor.
13723. The method of item 13703 wherein the agent is a CD40 antagonist.
13724. The method of item 13703 wherein the agent is a chemokine receptor agonist.
13725. The method of item 13703 wherein the agent is a chymase inhibitor.
13726. The method of item 13703 wherein the agent is a collagenase antagonist.
13727. The method of item 13703 wherein the agent is a CXCR antagonist.
13728. The method of item 13703 wherein the agent is a cyclin dependent kinase inhibitor selected from the group consisting of a CDK-1 inhibitor, a CDK-2 inhibitor, a CDK- 4 inhibitor, a CDK-6 inhibitor, a CAK1 inhibitor from GPC Biotech and Bristol-Myers Squibb, RGB-286199 (GPC Biotech), an anticancer agent from Astex Technology, a CAK1 inhibitor from GPC Biotech, a CDK inhibitor from Sanofi-Aventis, a CDK1/CDK2 inhibitor from Amgen, a CDK2 inhibitor from SUGEN-2 (Pfizer), a hearing loss therapy agent (Sound Pharmaceuticals), PD-0332991 (Pfizer), RGB-286199 (GPC Biotech), Ro-0505124 (Hoffmann-La Roche), a Ser/Thr kinase inhibitor from Lilly (EIi Lilly), CVT-2584 (CAS No. 199986-75- 9) (CV Therapeutics), and an analogue or derivative thereof.
13729. The method of item 13703 wherein the agent is a cyclooxygenase 1 inhibitor.
13730. The method of item 13703 wherein the agent is a DHFR inhibitor.
13731. The method of item 13703 wherein the agent is a dual integrin inhibitor.
13732. The method of item 13703 wherein the agent is an elastase inhibitor.
13733. The method of item 13703 wherein the agent is an elongation factor-1 alpha inhibitor.
13734. The method of item 13703 wherein the agent is an endothelial growth factor antagonist.
13735. The method of item 13703 wherein the agent is an endothelial growth factor receptor kinase inhibitor selected from the group consisting of sorafenib tosylate (Bayer), AAL-993 (Novartis), ABP-309 (Novartis), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), E- 7080 (Eisai), EG-3306 (Ark Therapeutics), EXEL-2880 (Exelixis), GW- 654652 (GlaxoSmithKline), a KDR inhibitor from LG Life Sciences, CT-6685 and CT-6729 (UCB), KRN-633 and KRN-951 (Kirin Brewery), OSI-930 (OSI Pharmaceuticals), SP-5.2 (Supratek Pharma), SU-11657 (Pfizer), a Tie-2 antagonist (Hybrigenics), a VEGFR-2 kinase inhibitor (Bristol-Myers Squibb), XL-647 (Exelixis), a KDR inhibitor from Abbott Laboratories, sorafenib tosylate, and an analogue or derivative thereof.
13736. The method of item 13703 wherein the agent is an endotoxin antagonist.
13737. The method of item 13703 wherein the agent is an epothilone and tubulin binder.
13738. The method of item 13703 wherein the agent is an estrogen receptor antagonist.
13739. The method of item 13703 wherein the agent is an FGF inhibitor.
13740. The method of item 13703 wherein the agent is a farnexyl transferase inhibitor.
13741. The method of item 13703 wherein the agent is farnesyltransferase inhibitor selected from the group of A-197574 (Abbott), a farnesyltransferase inhibitor from Servier, FPTIH (Strathclyde Institute for Drug R), LB-42908 (LG Life Sciences), Pharmaprojects No. 5063 (Genzyme), Pharmaprojects No. 5597 (Ipsen), Yissum Project No. B-1055 (Yissum), and an analogue or derivative thereof.
13742. The method of item 13703 wherein the agent is an FLT-3 kinase inhibitor. 13743. The method of item 13703 wherein the agent is an FGF receptor kinase inhibitor.
13744. The method of item 13703 wherein the agent is a fibrinogen antagonist selected from the group consisting of AUV-201 (Auvation), MG-13926 (Sanofi-Aventis), plasminogen activator (CAS No. 105913-11-9) (from Sanofi-Aventis or UCB), plasminogen activator-2 (tPA- 2) (Sanofi-Aventis), pro-urokinase (CAS No. 82657-92-9) (Sanofi-Aventis), and an analogue or derivative thereof.
13745. The method of item 13703 wherein the agent is a heat shock protein 90 antagonist selected from the group consisting of SRN- 005 (Sirenade), geldanamycin, NSC-33050 (17-allylaminσgeldanamycin; 17- AAG), 17-dimethylaminoethylamino-17-demethoxy-geldanamycin (17- DMAG), rifabutin (rifamycin XIV, 1',4-didehydro-1-deoxy-1 ,4-dihydro-5'-(2- methylpropyl)-1 -OXO-), and an analogue or derivative thereof. - - -
13746. The method of item 13703 wherein the agent is a histone deacetylase inhibitor.
13747. The method of item 13703 wherein the agent is an HMGCoA reductase inhibitor selected from the group consisting of an atherosclerosis therapeutic from Lipid Sciences, ATI-16000 (ARYx Therapeutics), KS-01-019 (Kos Pharmaceuticals), Pharmaprojects No. 2197 (Sanofi-Aventi), RP 61969 (Sanofi-Aventis), and an analogue or derivative thereof.
13748. The method of item 13703 wherein the agent is an ICAM inhibitor. 13749. The method of item 13703 wherein the agent is an IL, ICE and IRAK antagonist, wherein the antagonist is a CJ-14877, CP- 424174 (Pfizer), NF-61 (Negma-Lerads), and an analogue or derivative thereof.
13750. The method of item 13703 wherein the agent is an IL-2 inhibitor.
13751. The method of item 13703 wherein the agent is an immunosuppressant selected from the group consisting of teriflunomide (Sanofi Aventis), chlorsulfaquinoxalone (NSC-339004), chlorsulfaquinoxalone sulfate, CS-712 (Sankyo), ismomultin alfa (CAS No. 457913-93-8) (Akzo Nobel), antiallergics from GenPat77, antiinflammatories or AT-005 (Androclus Therapeutics), autoimmune disease therapeutics from EpiVax, BN-007 (Bone), budesonide (CAS No. 51333-22- 3) (MAP Pharmaceuticals), CO-14 (Genzyme), edratide (CAS No.433922-- 67-9) (Teva), EP-314 (Enanta), eprovafen (CAS No. 101335-99-3) (Sanofi- Aventis), HWA-131 (CAS No. 118788-41-3) (Sanofi-Aventis), immunomodulators from MerLion Pharmaceuticals, immunosuppressives from Alchemia, IPL-12 (Inflazyme), MDL-9563 (CAS No. 27086-86-8) (Sanofi-Aventis), Pharmaprojects No. 2330 (Sanofi-Aventis), Pharmaprojects No. 6426 (Abgenix), PXS-25 (Pharmaxis), rosmarinic acid (CAS No. 20283-92-5) (Sanofi-Aventis), RP 42927 or RP 54745 (CAS No. 135330-08-4) (Sanofi-Aventis), SGN-35 (Seattle Genetics), ST-1959 (Sigma-Tau), type I diabetes therapy from SYNX Pharma, UNIL-88 (Debiopharm), VP-025 (Vasogen), VR-694 (Vectura), PRTX-001 (Protalex), and an analogue or derivative thereof.
13752. The method of item 13703 wherein the agent is an IMPDH (inosine monophosphate). 13753. The method of item 13703 wherein the agent is an integrin antagonist.
13754. The method of item 13703 wherein the agent is an interleukin antagonist.
13755. The method of item 13703 wherein the agent is an inhibitor of type III receptor tyrosine kinase.
13756. The method of item 13703 wherein the agent is an irreversible inhibitor of enzyme methionine aminopeptidase type 2.
13757. The method of item 13703 wherein the agent is an isozyme selective delta protein kinase C inhibitor.
13758. The method of item 13703 wherein the agent a JAK3 enzyme inhibitor.
13759. The method of item 13703 wherein the agent is a JNK inhibitor.
13760. The method of item 13703 wherein the agent is a kinase inhibitor.
13761. The method of item 13703 wherein the agent is kinesin antagonist.
13762. The method of item 13703 wherein the agent is a kinesin antagonist. 13763. The method of item 13703 wherein the agent is a leukotriene inhibitor and antagonist selected from the group consisting of ambicromil (CAS No. 58805-38-2) (Sanofi-Aventis), amelubant (CAS No. 346735-24-8) (Boehringer Ingelheim), DW-1141 (Dong Wha), ebselen (Daiichi Pharmaceutical), ibudilast (Kyorin), leucotriene inhibitors from Sanofi-Aventis, lymphotoxin-beta receptor (LT-D) from Biogen Idee, Pharmaprojects No. 1535 and 2728 (CAS No. 119340-33-9) (Sanofi- Aventis), R-112 (Rigel), Rev-5367 (CAS No. 92532-05-3) (Sanofi-Aventis), RG-14893 (CAS No. 141835-49-6) (Sanofi-Aventis), RG-5901-A (CAS No. 101910-24-1), 92532-23-5, RP 66153 (CAS No. 142422-79-5), RP 66364 (CAS No.186916-92-5), RP 69698 (CAS No. 141748-00-7) (Sanofi-Aventis), SC-411930 (Pfizer), SC-41930 (CAS No. 120072-59-5) (Pfizer), SC-50605 (CAS No. 138828-39-4) (Pfizer), SC-51146 (CAS No. 141059-52-1), SC- 53228 (CAS No. 153633-01-3) (Pfizer), spaglumic acid (ZY-15106) (CAS No.3106-85-2), 80619-64-3 (Novartis), tipredane (CAS No. 85197-77-9) (Bristol-Myers Squibb), U-75302 (CAS No. 119477-85-9) (Pfizer), and. analogue or derivative thereof.
13764. The method of item 13703 wherein the agent is an MAP kinase inhibitor.
13765. The method of item 13703 wherein the agent is a matrix metalloproteinase inhibitor.
13766. The method of item 13703 wherein the agent is an MCP-CCR2 inhibitor.
13767. The method of item 13703 wherein the agent is an mTOR inhibitor. 13768. The method of item 13703 wherein the agent is an mTOR kinase inhibitor.
13769. The method of item 13703 wherein the agent is a microtubule inhibitor selected from the group consisting of antibody- maytansinoid conjugates from Biogen Idee, colchicines (MantiCore Pharmaceuticals), anticancer immunoconjugates from Johnson & Johnson, DIME from Octamer, gnMf (GNI)1 huC242-DM4, huMy9-6-DM1 (ImmunoGen), IDN-5404 (Indena), IMO-098, IMOderm (Imotep), mebendazole (Introgen Therapeutics), microtubule poisons from Cambridge Enterprise, paclitaxel such as LOTAX from Aphios (CAS No. 33069-62-4), Genexol-PM from Samyang, Pharmaprojects No. 6383 (Tapestry Pharmaceuticals), RPR-112378 (Sanofi-Aventis), SGN-75 (Seattle Genetics), SPL-7435 (Starpharma), SSR-250411 (Sanofi-Aventis), trastuzumab-DM1 (Genentech), vinorelbine, and an analogue or derivative thereof. . ._ .. _ - - ■
13770. The method of item 13703 wherein the agent is an MIF inhibitor.
13771. The method of item 13703 wherein the agent is an MMP inhibitor.
13772. The method of item 13703 wherein the agent is a neurokinin (NK) antagonist selected from the group consisting of anthrotainin (CAS No. 148084-40-6) (Sanofi-Aventis), an IBS thereapeutic from ArQuIe, MDL-105212A (CAS No. 167261-60-1) (Ssanofi-Aventis), Pharmaprojects No. 2744, 3258 (CAS No. 139167-47-8) 4006, 4201, or 5986 (Sanofi-Aventis), RP 67580 (CAS No. 135911-02-3), SR-144190 (CAS No. 201152-86-5), SSR-240600, SSR-241586 (Sanofi-Aventis), TKA-457 (Novartis), vestipitant mesylate (CAS No. 334476-64-1) (GlaxoSmithKline), Win-64821 (Sanofi-Aventis), PRX-96026 (Predix Pharmaceuticals), and an analogue or derivative thereof.
13773. The method of item 13703 wherein the agent is an NF kappa B inhibitor selected from the group consisting of AVE-0545 or AVE-0547 (Sanofi-Aventis), bortezomib (CAS No. 179324-69-7) (Millennium Pharmaceuticals), dexanabinol (CAS No. 112924-45-5) (Pharmos), dexlipotam (Viatris), Pharmaprojects No. 6283 (INDRA) (OXiGENE), IPL- 576092 (CAS No. 137571-30-3) (Inflazyme), NFKB decoy (Corgentech), NFKB decoy oligo (AnGes MG), NFKB's from Ariad, osteoporosis treatments or S5 (F005) from Fulcrum Pharmaceuticals, P61 (Phytopharm), R-flurbiprofen (CAS No. 5104-49-4) (Encore Pharmaceuticals), and an analogue or derivative thereof.
13774. The method of item 13703 wherein the agent is a nitric oxide agonist.
13775. The method of item 13703 wherein the agent is an ornithine decarboxylase inhibitor.
13776. The method of item 13703 wherein the agent is a p38 MAP kinase inhibitor selected from the group consisting of AZD-6703 (AstraZeneca), JX-401 (Jexys Pharmaceuticals), BMS-2 (Bristol-Myers Squibb), a p38 MAP kinase inhibitor from Novartis, a p38-alpha MAP kinase inhibitor from Amphora, Pharmaprojects No. 5704 (Pharmacopeia), RPR- 200765A (Sanofi-Aventis), SD-282 (Johnson & Johnson), TAK-715 (Takeda), and an analogue or derivative thereof.
13777. The method of item 13703 wherein the agent is a palmitoyl-protein thioesterase inhibitor. 13778. The method of item 13703 wherein the agent is a PDGF receptor kinase inhibitor selected from the group consisting of AAL- 993, AMN-107, or ABP-309 (Novartis), AMG-706 (Amgen), BAY-57-9352 (Bayer), CDP-860 (UCB), E-7080 (Eisai), imatinib (CAS No. 152459-95-5) (Novartis), OSI-930 (OS! Pharmaceuticals), RPR-127963E (Sanofi-Aventis), RWJ-540973 (Johnson & Johnson), sorafenib tosylate (Bayer), SU-11657 (Pfizer), tandutinib (CAS No. 387867-13-2) (Millennium Pharmaceuticals), vatalanib (Novartis), ZK-CDK (Schering AG), and an analogue or derivative thereof.
13779. The method of item 13703 wherein the agent is (-)-halofenate (Metabolex), AMG-131 (Amgen), antidiabetics from Japan Tobacco, AZD-4619, AZD-8450, AZD-8677 (AstraZeneca), DRF-10945, balaglitazone (Dr Reddy's), CS-00088, CS-00098 (Chipscreen Biosciences), E-3030 (Eisai), etalocib (CAS No. 161172-51-6) (EIi Lilly), GSK-641597 (Ligand), GSK-677954 (GlaxoSmithKline), GW-409544 (Ligand), GW- 590735 (GlaxoSmithKline), K-111 (Hoffmann-La Roche), LY-518674 (EIi Lilly), LY-674 (Ligand), LY-929 (Ligand), MC-3001 , MC-3002 (MaxoCore Pharmaceuticals), metformin HCI + pioglitazone (CAS No. 1115-70-4 and 112529-15-4), ACTOPLUS MET from Andrx), muraglitazar (CAS No. 331741-94-7) (Bristol-Myers Squibb), naveglitazar (Ligand), oleoylethanolamide (Kadmus Pharmaceuticals), ONO-5129, pioglitazone hydrochloride (CAS No. 111025-46-8 and 112529-15-4) (Takeda), PLX-204 (Plexxikon), PPAR agonists from Genfit, PPAR delta agonists from Eli Lilly, PPAR-alpha agonists from CrystalGenomics, PPAR-gamma modulators and PPAR-β modulators from CareX, rosiglitazone maleate (CAS No. 122324- 73-4 or 155141-29-0) (GlaxoSmithKline), rosiglitazone maleate/glimepir (CAS No. 155141-29-0 and 93479-97-1), AVANDARYL, rosiglitazone maleate/metformin extend (CAS No. 155141-29-0 and 657-24-9), AVANDAMET, rosiglitazone maleate+metformin, AVANDAMET (GlaxoSmithKline), tesaglitazar (AstraZeneca), and an analogue or derivative thereof.
13780. The method of item 13703 wherein the agent is a phosphatase inhibitor.
13781. The method of item 13703 wherein the agent is a phosphodiesterase (PDE) inhibitor selected from the group consisting of avanafil (Tanabe Seiyaku), dasantafil (CAS No. 569351-91-3) (Schering- Plough), A-906119 (CAS No. 134072-58-5), DL-850 (Sanofi-Aventis), GRC- 3015, GRC-3566, GRC-3886 (Glenmark), HWA-153 (CAS No. 56395-66-5) (Sanofi-Aventis), hydroxypumafentrine (Altana), IBFB-130011 , IBFB-14-016, IBFB-140301 , IBFB-150007, IBFB-211913 (IBFB Pharma), L-826141 (Merck & Co), medorinone (CAS No. 88296-61-1) (Sanofi-Aventis), MEM- 1917 (Memory Pharmaceuticals), ND-1251 (Neuro3d), PDE inhibitors from ICOS, PDE IV inhibitors from Memory Pharmaceuticals and CrystalGenomics, Pharmaprojects No. 2742 and 6141 (Sanofi-Aventis), QAD-171 (Novartis), RHC-2963 (CAS No. 76993-12-9 and 76993-14-1), RPR-117658, RPR-122818 derivatives, SR-24870 , and RPR-132294 (Sanofi-Aventis), SK-350 (ln2Gen), stroke therapy agents from deCODE Genetics, TAS-203 (Taiho), tofimilast (CAS No. 185954-27-2) (Pfizer), UK- 371800 (Pfizer), WIN-65579 (CAS No. 158020-82-7) (Sanofi-Aventis), IBFB- 130020 (IBFB Pharma), OPC-6535 (CAS No. 145739-56-6) (Otsuka), a phosphodiesterase III inhibitor, enoximone, a phosphodiesterase IV inhibitor, fosfosal, Atopik (Barrier Therapeutics), triflusal, a phosphodiesterase V inhibitor, and an analogue or derivative thereof.
13782. The method of item 13703 wherein the agent is a PKC inhibitor. 13783. The method of item 13703 wherein the agent is a platelet activating factor antagonist.
13784. The method of item 13703 wherein the agent is a platelet-derived growth factor receptor kinase inhibitor.
13785. The method of item 13703 wherein the agent is a prolyl hydroxylase inhibitor.
13786. The method of item 13703 wherein the agent is a polymorphonuclear neutrophil inhibitor.
13787. The method of item 13703 wherein the agent is a protein kinase B inhibitor.
13788. The method of item 13703 wherein the agent is a protein kinase C stimulant.
13789. The method of item 13703 wherein the agent is a purine nucleoside analogue.
13790. The method of item 13703 wherein the agent is a purinoreceptor P2X antagonist.
13791. The method of item 13703 wherein the agent is a Raf kinase inhibitor.
13792. The method of item 13703 wherein the agent is a reversible inhibitor of ErbB1 and ErbB2. 13793. The method of item 13703 wherein the agent is a ribonucleoside triphosphate reductase inhibitor.
13794. The method of item 13703 wherein the agent is an SDF-1 antagonist.
13795. The method of item 13703 wherein the agent is a sheddase inhibitor.
13796. The method of item 13703 wherein the agent is an SRC inhibitor.
13797. The method of item 13703 wherein the agent is a stromelysin inhibitor.
13798. The method of item 13703 wherein the agent is an Syk kinase inhibitor.
13799. The method of item 13703 wherein the agent is a telomerase inhibitor.
13800. The method of item 13703 wherein the agent is a TGF beta inhibitor selected from the group consisting of pirfenidone (CAS No. 53179-13-8) (MARNAC), tranilast (CAS No. 53902-12-8) (Kissei), IN- 1130 (ln2Gen), mannose-6-phosphate (BTG), TGF-β antagonists from Inflazyme (Pharmaprojects No. 6075), TGF-β antagonists from Sydney, non-industrial source), TGF-β I receptor kinase inhibitors from EIi Lilly, TGF- β receptor inhibitors from Johnson & Johnson, and an analogue or derivative thereof. 13801. The method of item 13703 wherein the agent is a
TNFα antagonist or TACE inhibitor selected from the group consisting of adalimumab (CAS No. 331731-18-1) (Cambridge Antibody Technology), AGIX-4207 (AtheroGenics), AGT-1 (Advanced Biotherapy), an antiinflammatory from Borean Pharma, Cellzome, or Paradigm Therapeutics, anti-inflammatory vaccine (TNF-alpha kinoid) from Neovacs, humanized anti-TNF antibody or an anti-TNF MAb (CB0006) Celltech (UCB), apratastat (CAS No. 287445-51-0) (Wyeth), BMS-561392 (Bristol-Myers Squibb), BN- 006 (Bone), certolizumab pegol (CAS No. 428863-50-7 or CH-138 (UCB), cilomilast (CAS No. 153259-65-5) (GlaxoSmithKline), CR-1 (Nuada Pharmaceuticals), CRx-119 (CombinatoRx), D-5410 (UCB), dacopafant (CAS No. 125372-33-0) (Sanofi-Aventis), dersalazine (CAS No. 188913-57- 7/188913-58-8) (Uriach), etanercept (CAS No. 185243-69-0) (Amgen), ethyl pyruvate (Critical (Critical Therapeutics), golimumab (CAS No. 476181-74-5) (Johnson & Johnson), hormono-immunotherapy from Ipsen, CDP571 (e.g., Humicade from UCB), IC-485 (ICOS), infliximab (CAS No. 170277-31-3) (Johnson & Johnson), IP-751 (Manhattan Pharmaceuticals), ISIS-104838 (CAS No. 250755-32-9) (ISIS Pharmaceuticals), lenalidomide (CAS No. 191732-72-6) (Celgene), lentinan (CAS No. 37339-90-5) (Ajinomoto), MDL- 201112 (CAS No. 142130-73-2) (Sanofi-Aventis), medroxyprogesterone (CAS No. 520-85-4) (InKine Pharmaceutical), N-acetylcysteine (CAS No. 616-91-1) (Zambon), NBE-P2 (DIREVO Biotech), nerelimomab (CAS No. 162774-06-3) (Chiron), OM-294DP (OM PHARMA), onercept (CAS No. 199685-57-9) (Yeda), PASSTNF-alpha (Verigen), pentoxifylline or oxypentifylline (Sanofi-Aventis), Pharmaprojects No. 4091, 4241 , 4295, or 4488 (Sanofi-Aventis), Pharmaprojects No. 5480 (Amgen), Pharmaprojects No. 6749 (Cengent), pirfenidone (CAS No. 53179-13-8) (MARNAC), RPR- 132294 (Sanofi-Aventis), S5 (F002) (Fulcrum Pharmaceuticals), simvastatin (CAS No. 79902-63-9) (Merck & Co), STA-6292 (Synta Pharmaceuticals), tacrolimus (CAS No. 104987-11-3) (Fujisawa LifeCycle Pharma), talactoferrin alfa (CAS No. 308240-58-6) (Agennix), thalidomide (CAS No. 50-35-1) (Celgene), TNF antagonists form ProStrakan, and Synergen, TNF inhibitors (Amgen), TNF-alpha antagonists from Dynavax Technologies and Jerina AG (Germany), TNF-alpha inhibitors from IBFB Pharma and Xencor (Xencor), torbafylline (CAS No. 105102-21-4) (Sanofi-Aventis), UR-1505 (Uriach), VT-346 (Viron Therapeutics), YSIL6 (Y's Therapeutics), YSTH2 (Y's Therapeutics), NPI-1302a-3 (Nereus Pharmaceuticals, a TNF antagonist from Jerina AG (Germany), dersalazine, and an analogue or derivative thereof.
13802. The method of item 13703 wherein the agent is a Toll receptor inhibitor.
13803. The method of item 13703 wherein the agent is a tubulin antagonist.
13804. The method of item 13703 wherein the agent is a tyrosine kinase inhibitor selected from the group consisting of SU-011248, SUTENT from Pfizer Inc. (New York, NY), BMS-354825, PN-355 (Paracelsian Pharmaceuticals), AGN-199659 (Allergan), AAL-993 or ABP- 309 (Novartis), adaphostin (NIH)1 AEE-788 (Novartis), AG-013736 (OSI Pharmaceuticals), AG-13736 (Pfizer), ALT-110 (Alteris Therapeutics), AMG- 706 (Amgen), anticancer MAbs from Xencor, anti-EGFrvlll MAbs from Abgenix, anti-HER2 MAb from Abiogen, AZD-2171 or AZD-9935 (AstraZeneca), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), CEP-5214 (Cephalon), CEP-7055 (Cephalon), cetuximab (ImClone Systems), CHIR-200131 and CHIR-258 (Chiron), CP-547632 (OSI Pharmaceuticals), CP-724714 (Pfizer), CT-301 (Creabilis Therapeutics), D- 69491 (Baxter International), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EGFR/ErbB2 inhibitors from Array BioPharma, erlotinib (CAS No. 183319- 69-9) (OSI Pharmaceuticals), EXEL-2880 (Exelixis), FK-778 (Sanofi- Aventis) , gefitinib (CAS No. 184475-35-2) (AstraZeneca), GW-2286 or GW- 654652 (GlaxoSmithKline), her2/neu antigen from AlphaVax, HER-2/neu inhibitor from Generex, Herzyme (Medipad) (Sirna Therapeutics), HKI-272 (Wyeth), HuMax-EGFr (Genmab), idronoxil (CAS No. 81267-65-4) (Novogen), IGF-1 inhibitors from Ontogen, IMC-11F8 (ImCIone Systems), kahalalide F (CAS No. 149204-42-2) (PharmaMar), KDR inhibitor from LG Life Sciences, KDR inhibitors from Abbott Laboratories, KDR kinase inhibitors (UCB), Kdr kinase inhibitors from Merck & Co, KRN-633 and KRN- 951 (Kirin Brewery), KSB-102 (Xenova), lapatinib ditosylate (CAS No. 388082-78-8) (GlaxoSmithKline), matuzumab (Merck KGaA), MDX-214 (Medarex), ME-103 (Pharmexa), MED-A300 (Gerolymatos), MNAC-13 (Lay Line Genomics), nimotuzumab (Center of Molecular Immunology), NSC- 330507 or NSC-707545 (NIH), NV-50 (Novogen), OSI-930 (OSI Pharmaceuticals), panitumumab (Abgenix), pelitinib (CAS No. 287933-82-7) (Wyeth), pertuzumab (CAS No. 380610-27-5) (Genentech), Pharmaprojects No. 3985 (Sanofi-Aventis), prostate cancer therapeutics from Sequenom (SQPC35, SQPC36, SQPC90), removab and remoxab (Trion Pharma), RG- 13022 (CAS No. 136831-48-6), RG-13291 (CAS No. 138989-50-1), or RG- 14620 (CAS No. 136831-49-7) (Sanofi-Aventis), RM-6427 (Romark), RNAi breast cancer therapy from Benitec, RP 53801 (CAS No. 125882-88-4) (Sanofi-Aventis), sorafenib tosylate (Bayer), SU-11657 (Pfizer), Tie-2 antagonists from Semaia (Hybrigenics), Tie-2 inhibitors from Ontogen, trastuzumab (CAS No. 180288-69-1) (Genentech), tyrosine kinase inhibitors from Sanofi-Aventis, U3-1287, U3-1565, U3-1784, U3-1800 (U3 Pharma), vatalanib (Novartis), VEGFR-2 kinase inhibitor from Bristol-Myers Squibb, XL-647 (Exelixis), ZD-6474 (AstraZeneca), ZK-CDK (Schering AG), an EGFR tyrosine kinase inhibitor, EKB-569 (Wyeth), and an analogue or derivative thereof.
13805. The method of item 13703 wherein the agent is a
VEGF inhibitor. 13806. The method of item 13703 wherein the agent is a vitamin D receptor agonist.
13807. The method of item 13703 wherein the agent is ZD-6474 (an angiogenesis inhibitor).
13808. The method of item 13703 wherein the agent is AP-23573 (an mTOR inhibitor).
13809. The method of item 13703 wherein the agent is synthadotin (a tubulin antagonist).
13810. The method of item 13703 wherein the agent is S-0885 (a collagenase inhibitor).
13811. The method of item 13703 wherein the agent is aplidine (an elongation factor-1 alpha inhibitor).
13812. The method of item 13703 wherein the agent is ixabepilone (an epithilone).
13813. The method of item 13703 wherein the agent is IDN-5390 (an angiogenesis inhibitor).
13814. The method of item 13703 wherein the agent is SB-2723005 (an angiogenesis inhibitor).
13815. The method of item 13703 wherein the agent is ABT-518 (an angiogenesis inhibitor). 13816. The method of item 13703 wherein the agent is combretastatin (an angiogenesis inhibitor).
13817. The method of item 13703 wherein the agent is anecortave acetate (an angiogenesis inhibitor).
13818. The method of item 13703 wherein the agent is SB-715992 (a kinesin antagonist).
13819. The method of item 13703 wherein the agent is temsirolimus (an mTOR inhibitor).
13820. The method of item 13703 wherein the agent is adalimumab (a TNFα antagonist).
13821. The method of item 13703, wherein the composition comprises a polymer.
13822. The method of item 13703, wherein the composition comprises a polymeric carrier.
13823. The method of item 13703 wherein the anti- scarring agent inhibits adhesion between the device and a host into which the device is implanted.
13824. The method of item 13703 wherein the device delivers the anti-scarring agent locally to tissue proximate to the device.
13825. The method of item 13703 wherein the device has a coating that comprises the anti-scarring agent. 13826. The method of item 13703, wherein the device has a coating that comprises the agent and is disposed on a surface of the implant.
13827. The method of item 13703, wherein the device has a coating that comprises the agent and directly contacts the implant.
13828. The method of item 13703, wherein the device has a coating that comprises the agent and indirectly contacts the implant.
13829. The method of item 13703, wherein the device has a coating that comprises the agent and partially covers the implant.
13830. The method of item 13703, wherein the device has a coating that comprises the agent and completely covers the implant.
13831. The method of item 13703, wherein the device has a uniform coating.
13832. The method of item 13703, wherein the device has a non-uniform coating.
13833. The method of item 13703, wherein the device has a discontinuous coating.
13834. The method of item 13703, wherein the device has a patterned coating.
13835. The method of item 13703, wherein the device has a coating with a thickness of 100 Dm or less. 13836. The method of item 13703, wherein the device has a coating with a thickness of 10 Dm or less.
13837. The method of item 13703, wherein the device has a coating, and the coating adheres to the surface of the implant upon deployment of the implant.
13838. The method of item 13703, wherein the device has a coating, and wherein the coating is stable at room temperature for a period of 1 year.
13839. The method of item 13703, wherein the device has a coating, and wherein the anti-scarring agent is present in the coating in an amount ranging between about 0.0001% to about 1% by weight.
13840. The method of item 13703, wherein the device has a coating, and wherein the anti-scarring agent is present in the coating in an amount ranging between about 1% to about 10% by weight.
13841. The method of item 13703, wherein the device has a coating, and wherein the anti-scarring agent is present in the coating in an amount ranging between about 10% to about 25% by weight.
13842. The method of item 13703, wherein the device has a coating, and wherein the anti-scarring agent is present in the coating in an amount ranging between about 25% to about 70% by weight.
13843. The method of item 13703, wherein the device has a coating, and wherein the coating further comprises a polymer. 13844. The method of item 13703, wherein the device has a first coating having a first composition and a second coating having a second composition.
13845. The method of item 13703, wherein the device has a first coating having a first composition and a second coating having a second composition, wherein the first composition and the second composition are different.
13846. The method of item 13703, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a copolymer.
13847. The method of item 13703, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a block copolymer. . . . .. .
13848. The method of item 13703, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a random copolymer.
13849. The method of item 13703, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a biodegradable polymer.
13850. The method of item 13703, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a non-biodegradable polymer. 13851. The method of item 13703, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a hydrophilic polymer.
13852. The method of item 13703, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a hydrophobic polymer.
13853. The method of item 13703, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a polymer having hydrophilic domains.
13854. The method of item 13703, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a polymer having hydrophobic domains.
13855. The method of item 13703, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a non-conductive polymer.
13856. The method of item 13703, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises an elastomer.
13857. The method of item 13703, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a hydrogel.
13858. The method of item 13703, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a silicone polymer. 13859. The method of item 13703, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a hydrocarbon polymer.
13860. The method of item 13703, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a styrene-derived polymer.
13861. The method of item 13703, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a butadiene polymer.
13862. The method of item 13703, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a macromer.
13863. The method of item 13703, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a poly(ethylene glycol) polymer.
13864. The method of item 13703 wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises an amorphous polymer.
13865. The method of item 13703, wherein the device comprises a lubricious coating.
13866. The method of item 13703 wherein the anti- scarring agent is located within pores or holes of the device. 13867. The method of item 13703 wherein the anti- scarring agent is located within a channel, lumen, or divet of the device.
13868. The method of item 13703, wherein the device comprises a second pharmaceutically active agent.
13869. The method of item 13703 wherein the device comprises an anti-inflammatory agent.
13870. The method of item 13703 wherein the device comprises an agent that inhibits infection.
13871. The method of item 13703 wherein the device comprises an agent that inhibits infection, and wherein the agent is an anthracycline.
13872. The method of item 13703 wherein the device comprises an agent that inhibits infection, and wherein the agent is doxorubicin.
13873. The method of item 13703 wherein the device comprises an agent that inhibits infection, and wherein the agent is mitoxantrone.
13874. The method of item 13703 wherein the device comprises an agent that inhibits infection, and wherein the agent is a fluoropyrimidine.
13875. The method of item 13703 wherein the device comprises an agent that inhibits infection, and wherein the agent is 5- fluorouracil (5-FU). 13876. The method of item 13703 wherein the device comprises an agent that inhibits infection, and wherein the agent is a folic acid antagonist.
13877. The method of item 13703 wherein the device comprises an agent that inhibits infection, and wherein the agent is methotrexate.
13878. The method of item 13703 wherein the device comprises an agent that inhibits infection, and wherein the agent is a podophyiotoxin.
13879. The method of item 13703 wherein the device comprises an agent that inhibits infection, and wherein the agent is etoposide.
13880. The method of item 13703 wherein the device comprises an agent that inhibits infection, and wherein the agent is a camptothecin.
13881. The method of item 13703 wherein the device comprises an agent that inhibits infection, and wherein the agent is a hydroxyurea.
13882. The method of item 13703 wherein the device comprises an agent that inhibits infection, and wherein the agent is a platinum complex.
13883. The method of item 13703 wherein the device comprises an agent that inhibits infection, and wherein the agent is cisplatin. 13884. The method of item 13703, further comprising an anti-thrombotic agent.
13885. The method of item 13703 wherein the device comprises a visualization agent.
13886. The method of item 13703 wherein the device comprises a visualization agent, wherein the visualization agent is a radiopaque material, and wherein the radiopaque material comprises a metal, a halogenated compound, or a barium containing compound.
13887. The method of item 13703 wherein the device comprises a visualization agent, wherein the visualization agent is a radiopaque material, and wherein the radiopaque material comprises barium, tantalum, or technetium.
13888. The method of item 13703 wherein the device comprises a visualization agent, and wherein the visualization agent is a MRI responsive material.
13889. The method of item 13703 wherein the device comprises a visualization agent, and wherein the visualization agent comprises a gadolinium chelate.
13890. The method of item 13703 wherein the device comprises a visualization agent, and wherein the visualization agent comprises iron, magnesium, manganese, copper, or chromium.
13891. The method of item 13703 wherein the device comprises a visualization agent, and wherein the visualization agent comprises an iron oxide compound. 13892. The method of item 13703 wherein the device comprises a visualization agent, and wherein the visualization agent comprises a dye, pigment, or colorant.
13893. The method of item 13703 wherein the device comprises an echogenic material.
13894. The method of item 13703 wherein the device comprises an echogenic material, and wherein the echogenic material is in the form of a coating.
13895. The method of item 13703 wherein the device is sterile.
13896. The method of item 13703 wherein the anti- scarring agent is released into tissue in the vicinity of the device after deployment of the device.
13897. The method of item 13703 wherein the anti- scarring agent is released into tissue in the vicinity of the device after deployment of the device, and wherein the tissue is connective tissue.
13898. The method of item 13703 wherein the anti- scarring agent is released into tissue in the vicinity of the device after deployment of the device, and wherein the tissue is muscle tissue.
13899. The method of item 13703 wherein the anti- scarring agent is released into tissue in the vicinity of the device after deployment of the device, and wherein the tissue is nerve tissue. 13900. The method of item 13703 wherein the anti- scarring agent is released into tissue in the vicinity of the device after deployment of the device, and wherein the tissue is epithelium tissue.
13901. The method of item 13703 wherein the anti- scarring agent is released in effective concentrations from the device over a period ranging from the time of deployment of the device to about 1 year.
13902. The method of item 13703 wherein the anti- scarring agent is released in effective concentrations from the device over a period ranging from about 1 month to 6 months.
13903. The method of item 13703 wherein the anti- scarring agent is released in effective concentrations from the device over a period ranging from about 1 - 90 days.
13904. The method of item 13703 wherein the anti- scarring agent is released in effective concentrations from the device at a constant rate.
13905. The method of item 13703 wherein the anti- scarring agent is released in effective concentrations from the device at an increasing rate.
13906. The method of item 13703 wherein the anti- scarring agent is released in effective concentrations from the device at a decreasing rate.
13907. The method of item 13703 wherein the anti- scarring agent is released in effective concentrations from the composition comprising the anti-scarring agent by diffusion over a period ranging from the time of deployment of the device to about 90 days.
13908. The method of item 13703 wherein the anti- scarring agent is released in effective concentrations from the composition comprising the anti-scarring agent by erosion of the composition over a period ranging from the time of deployment of the device to about 90 days.
13909. The method of item 13703 wherein the device comprises about 0.01 Og to about 10 Dg of the anti-scarring agent.
13910. The method of item 13703 wherein the device comprises about 10 Dg to about 10 mg of the anti-scarring agent.
13911. The method of item 13703 wherein the device comprises about 10 mg to about 250 mg of the anti-scarring agent
13912. The method of item 13703 wherein the device comprises about 250 mg to about 1000 mg of the anti-scarring agent.
13913. The method of item 13703 wherein the device comprises about 1000 mg to about 2500 mg of the anti-scarring agent.
13914. The method of item 13703 wherein a surface of the device comprises less than 0.01 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
13915. The method of item 13703 wherein a surface of the device comprises about 0.01 Dg to about 1 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied. 13916. The method of item 13703 wherein a surface of the device comprises about 1 Dg to about 10 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
13917. The method of item 13703 wherein a surface of the device comprises about 10 Dg to about 250 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
13918. The method of item 13703 wherein a surface of the device comprises about 250 Dg to about 1000 Dg of the anti-scarring agent of anti-scarring agent per mm2 of device surface to which the anti- scarring agent is applied.
13919. The method of item 13703 wherein a surface of the device comprises about 1000 Dg to about 2500 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
13920. The method of item 13703 wherein the combining is performed by direct affixing the agent or the composition to the implant.
13921. The method of item 13703 wherein the combining is performed by spraying the agent or the component onto the implant.
13922. The method of item 13703 wherein the combining is performed by electrospraying the agent or the composition onto the implant. 13923. The method of item 13703 wherein the combining is performed by dipping the implant into a solution comprising the agent or the composition.
13924. The method of item 13703 wherein the combining is performed by covalently attaching the agent or the composition to the implant.
13925. The method of item 13703 wherein the combining is performed by non-covalently attaching the agent or the composition to the implant.
13926. The method of item 13703 wherein the combining is performed by coating the implant with a substance that contains the agent or the composition.
13927. The method of item 13703 wherein the combining is performed by coating the implant with a substance that absorbs the agent.
13928. The method of item 13703 wherein the combining is performed by interweaving a thread composed of, or coated with, the agent or the composition.
13929. The method of item 13703 wherein the combining is performed by covering all the implant with a sleeve that contains the agent or the composition.
13930. The method of item 13703 wherein the combining is performed by covering a portion of the implant with a sleeve that contains the agent or the composition. 13931. The method of item 13703 wherein the combining is performed by covering all the implant with a cover that contains the agent or the composition.
13932. The method of item 13703 wherein the combining is performed by covering a portion of the implant with a cover that contains the agent or the composition.
13933. The method of item 13703 wherein the combining is performed by covering all the implant with an electrospun fabric that contains the agent or the composition.
13934. The method of item 13703 wherein the combining is performed by covering a portion of the implant with an electrospun fabric that contains the agent or the composition.
13935. The method of item 13703 wherein the combining is performed by covering all the implant with a mesh that contains the agent or the composition.
13936. The method of item 13703 wherein the combining is performed by covering a portion of the implant with a mesh that contains the agent or the composition.
13937. The method of item 13703 wherein the combining is performed by constructing all the implant with the agent or the composition.
13938. The method of item 13703 wherein the combining is performed by constructing a portion of the implant with the agent or the composition. 13939. The method of item 13703 wherein the combining is performed by impregnating the implant with the agent or the composition.
13940. The method of item 13703 wherein the combining is performed by constructing all of the implant from a degradable polymer that releases the agent.
13941. The method of item 13703 wherein the combining is performed by constructing a portion of the implant from a degradable polymer that releases the agent.
13942. The method of item 13703 wherein the combining is performed by dipping the implant into a solution that comprise the agent and an inert solvent for the implant.
13943. The method of item 13703 wherein the combining is performed by dipping the implant into a solution that comprises the agent and a solvent that will swell the implant.
13944. The method of item 13703 wherein the combining is performed by dipping the implant into a solution that comprises the agent and a solvent that will dissolve the implant.
13945. The method of item 13703 wherein the combining is performed by dipping the implant into a solution that comprises the agent, a polymer and an inert solvent for the implant.
13946. The method of item 13703 wherein the combining is performed by dipping the implant into a solution that comprises the agent, a polymer and a solvent that will swell the implant. 13947. The method of item 13703 wherein the combining is performed by dipping the implant into a solution that comprises the agent, a polymer and a solvent that will dissolve the implant.
13948. The method of item 13703 wherein the combining is performed by spraying the implant into a solution that comprises the agent and an inert solvent for the implant.
13949. The method of item 13703 wherein the combining is performed by spraying the implant into a solution that comprises the agent and a solvent that will swell the implant.
13950. The method of item 13703 wherein the combining is performed by spraying the implant into a solution that comprises the agent and a solvent that will dissolve the implant.
13951. The method of item 13703 wherein the combining is performed by spraying the implant into a solution that comprises the agent, a polymer and an inert solvent for the implant.
13952. The method of item 13703 wherein the combining is performed by spraying the implant into a solution that comprises the agent, a polymer and a solvent that will swell the implant.
13953. The method of item 13703 wherein the combining is performed by spraying the implant into a solution that comprises the agent, a polymer and a solvent that will dissolve the implant.
13954. A method as in any one of items 13703-13953, wherein the device is an AV fistula graft. 13955. A method as in any one of items 13703-13953, wherein the device is an AV access graft.
13956. A method as in any one of items 13703-13953, wherein the device is a venous catheter.
13957. A method as in any one of items 13703-13953, wherein the device is a vascular graft.
13958. A method as in any one of items 13703-13953, wherein the device is an implantable port.
13959. A method as in any one of items 13703-13953, wherein the device is an AV shunt.
13960. A method of making a medical device comprising: combining a medical device comprising a film or a mesh (e.g., an implant) and an anti-scarring agent or a composition comprising an anti- scarring agent, wherein the agent inhibits scarring between the device and a host into which the device is implanted.
13961. The method of item 13960 wherein the agent is an adensosine A2A receptor antagonist.
13962. The method of item 13960 wherein the agent is an AKT inhibitor.
13963. The method of item 13960 wherein the agent is an alpha 2 integrin antagonist, wherein the alpha 2 integrin antagonist is Pharmaprojects No. 5754 (Merck KgaA). 13964. The method of item 13960 wherein the agent is an alpha 4 integrin antagonist.
13965. The method of item 13960 wherein the agent is an alpha 7 nicotinic receptor agonist.
13966. The method of item 13960 wherein the agent is an angiogenesis inhibitor selected from the group consisting of AG-12,958 (Pfizer), ATN-161 (Attenuon LLC), neovastat, an angiogenesis inhibitor from Jerina AG (Germany), NM-3 (Mercian), VGA-1155 (Taisho), FCE-26644 (Pfizer), FCE-26950 (Pfizer), FPMA (Meiji Daries), FR-111142 (Fujisawa), GGTI-298, GM-1306 (Ligand), GPA-1734 (Novartis), NNC-47-0011 (Novo Nordisk), herbamycin (Nippon Kayaku), lenalidomide (Celegene), IP-10 (NIH), ABT-828 (Abbott), KIN-841 (Tokushima University, Japan), SF-1126 (Semafore Pharmaceuticals), laminin technology (NIH), CHIR-258 (Chiron), NVP-AEW541 (Novartis), NVP-AEW541 (Novartis), Vt16907 (Alchemia), OXI-8007 (Oxigene), EG-3306 (Ark Therapeutics), Maspin (Arriva), ABT- 567 (Abbott), PPI-2458 (Praecis Pharmaceuticals), CC-5079, CC-4089 (Celgene), HIF-1 alpha inhibitors (Xenova), S-247 (Pfizer), AP-23573 (Ariad), AZD-9935 (Astra Zeneca), mebendazole (Introgen Therapeutics), MetAP-2 inhibitors (GlaxoSmithKline), AG-615 (Angiogene Pharmaceuticals), Tie-2 antagonists (Hybrigenics), NC-381 , CYC-381 , NC-169, NC-219, NC-383, NC-384, NC-407 (Lorus Therapeutics), ATN-224 (Attenuon), ON-01370 (Onconova), Vitronectin antagonists (Amgen), SDX-103 (Salmedix), Vitronectin antagonists (Shire), CHP (Riemser), TEK (Amgen), Anecortave acetate (Alcon), T46.2 (Matrix Therapeutics), HG-2 (Heptagen), TEM antagonists (Genzyme), Oxi-4500 (Oxigene), ATN-161 (Attenuon), WX-293 (Wilex), M-2025 (Metris Therapeutics), Alphastatin (BioActa), YH-16 (Yantai Rongchang), BIBF-1120 (Boehringer Ingelheim), BAY-57-9352 (Bayer), AS- 1404 (Cancer Research Technology), SC-77964 (Pfizer), glycomimetics (BioTie Therapies), TIE-2 Inhibitors (Ontogen), DIMI, Octamer (Octamer), ABR-215050 (Active Biotech), ABT-518 (Abbott), KDR inhibitors (Abbott), BSF-466895 (Abbott), SCH-221153 (Schering-Plough), DAC:antiangiogenic (ConjuChem), TFPI (EntreMed), AZD-2171 (Astra-Zenaca), CDC-394 (Celgene), LY290293 (EIi Lilly), IDN-5390 (Indena), Kdr Kinase Inhibitors (Merck), CT-113020, CT-116433, CT-116563, CT-31890, CT-32228) (Cell Therapeutics), A-299620 (Abbott), TWEAK Inhibitor (Amgen), VEGF modulators (Johnson and Johnson), Tum-N53, tumstatin (Genzyme), Thios- 1, Thios-2 (Thios Pharmaceuticals), MV-6401 (Miravant Medical Technologies), Spisulosine (PharmaMar), CEP-7055 (Cephalon), AUV-201 (Auvation), LM-609 (EIi Lilly), SKF-106615 (AnorMED), Oglufanide disodium (Cytran), BW-114 (Pharminox), Calreticulin (NIH), WX-678 (Wilex), SD-7784 (Pfizer), WX-UK1 (Wilex), SH-268 (Schering AG), 2-Me-PGA (Celgene), S- 137 (Pfizer), ZD-6126 (Angiogene Pharmaceuticals), SG-292 (SignalGen), Benefin (Lane Labs), A6, A36 (Angstrom), SB-2723005 (GlaxoSmithKline), SC-7 (Cell Therapeutics), ZEN-014 (AEterna Zentaris), 2-methoxyestradiol (EntreMed), NK-130119 (Nippon Kayaku), CC-10004 (Celgene), AVE- 8062A (Ajinomoto), Tacedinaline (Pfizer), Actinonin (Tokyo Metropolitan Institute of Medical Science), Lenalidomide (Celgene), VGA-1155, BTO-956 (SRI International), ER-68203-00 (Eisai), CT-6685 (UCB), JKC-362 (Phoenix Pharmaceuticals), DMI-3798 (DMI Biosciences, Angiomate (Ipsen), ZD-6474 (AstraZeneca), CEP-5214 (Cephalon), Canstatin (Genzyme), NM-3 (Mercian), Oridigm (MediQuest Therapeutics), Exherin (Adherex), BLS-0597 (Boston Life Sciences), PTC-299 (PTC Therapeutics), NPI-2358 (Nereus Pharmaceuticals), CGP-79787 (Novartis), AEE-788 (Novartis), CKD-732 (Chong Kun Dang), CP-564959 (OSI Pharmaceuticals), CM-101 (CarboMed), CT-2584, CT3501 (Cell Therapeutics), combretastatin and analogues and derivatives thereof (Oxigene), Rebimastat (Bristol-Meyers Squibb), Dextrin 2-sulfate (ML Laboratories), Cilengitide (Merk KGaA), NSC-706704 (Pharminox), KRN- 951 (Kirin Brewery), Ukrain, NSC-631570 (Nowicky Pharma), Tecogalan sodium (Daiichi Pharmaceutical), Tz-93 (Tsumura), TBC-1635 (Encysive Pharmaceuticals), TAN-1120 (Takeda), Semaxanib (Pfizer), BDl-7800 (Biopharmacopae), SD-186, SD-983 (Bristol-Meyers Squibb), SB-223245 (GlaxoSmithKline), SC-236 (Pfizer), RWJ-590973 (Johnson and Johnson), ILX-1850 (Genzyme), SC-68488, S-836 (Pfizer), CG-55069-11 (CuraGen), Ki-23057 (Kirin Brewery), CCX-700 (Chemoentryx), Pegaptanib octasodium (Giled Sciences), ANGIOCOL (available from Biostratum Inc.), or an analogue or derivative thereof.
13967. The method of item 13960 wherein the agent is an apoptosis antagonist.
13968. The method of item 13960 wherein the agent is an apoptosis activator.
13969. The method of item 13960 wherein the agent is a beta 1 integrin antagonist.
13970. The method of item 13960 wherein the agent is a beta tubulin inhibitor.
13971. The method of item 13960 wherein the agent is a blocker of enzyme production in Hepatitis C.
13972. The method of item 13960 wherein the agent is a Bruton's tyrosine kinase inhibitor.
13973. The method of item 13960 wherein the agent is a calcineurin inhibitor.
13974. The method of item 13960 wherein the agent is a caspase 3 inhibitor. 13975. The method of item 13960 wherein the agent is a CC chemokine receptor antagonist.
13976. The method of item 13960 wherein the agent is a cell cycle inhibitor selected from the group consisting of SNS-595 (Sunesis), synthadotin, KRX-0403, and an analogue or derivative thereof.
13977. The method of item 13960 wherein the agent is a cathepsin B inhibitor.
13978. The method of item 13960 wherein the agent is a cathepsin K inhibitor, wherein the cathepsin K inhibitor is 462795 (GlaxoSmithKline), INPL-022-D6 (Amura Therapeutics), or an analogue or derivative thereof.
13979. The method of item 13960 wherein the agent is a cathepsin L inhibitor.
13980. The method of item 13960 wherein the agent is a CD40 antagonist.
13981. The method of item 13960 wherein the agent is a chemokine receptor agonist.
13982. The method of item 13960 wherein the agent is a chymase inhibitor.
13983. The method of item 13960 wherein the agent is a collagenase antagonist. 13984. The method of item 13960 wherein the agent is a CXCR antagonist.
13985. The method of item 13960 wherein the agent is a cyclin dependent kinase inhibitor selected from the group consisting of a CDK-1 inhibitor, a CDK-2 inhibitor, a CDK- 4 inhibitor, a CDK-6 inhibitor, a CAK1 inhibitor from GPC Biotech and Bristol-Myers Squibb, RGB-286199 (GPC Biotech), an anticancer agent from Astex Technology, a CAK1 inhibitor from GPC Biotech, a CDK inhibitor from Sanofi-Aventis, a CDK1/CDK2 inhibitor from Amgen, a CDK2 inhibitor from SUGEN-2 (Pfizer), a hearing loss therapy agent (Sound Pharmaceuticals), PD-0332991 (Pfizer), RGB-286199 (GPC Biotech), Ro-0505124 (Hoffmann-La Roche), a Ser/Thr kinase inhibitor from Lilly (EIi Lilly), CVT-2584 (CAS No. 199986-75- 9) (CV Therapeutics), and an analogue or derivative thereof.
13986. The method of item 13960 wherein the agent is a cyclooxygenase 1 inhibitor.
13987. The method of item 13960 wherein the agent is a DHFR inhibitor.
13988. The method of item 13960 wherein the agent is a dual integrin inhibitor.
13989. The method of item 13960 wherein the agent is an elastase inhibitor.
13990. The method of item 13960 wherein the agent is an elongation factor-1 alpha inhibitor. 13991. The method of item 13960 wherein the agent is an endothelial growth factor antagonist.
13992. The method of item 13960 wherein the agent is an endothelial growth factor receptor kinase inhibitor selected from the group consisting of sorafenib tosylate (Bayer), AAL-993 (Novartis), ABP-309 (Novartis), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), E- 7080 (Eisai), EG-3306 (Ark Therapeutics), EXEL-2880 (Exelixis), GW- 654652 (GlaxoSmithKline), a KDR inhibitor from LG Life Sciences, CT-6685 and CT-6729 (UCB), KRN-633 and KRN-951 (Kirin Brewery), OSI-930 (OSI Pharmaceuticals), SP-5.2 (Supratek Pharma), SU-11657 (Pfizer), a Tie-2 antagonist (Hybrigenics), a VEGFR-2 kinase inhibitor (Bristol-Myers Squibb), XL-647 (Exelixis), a KDR inhibitor from Abbott Laboratories, sorafenib tosylate, and an analogue or derivative thereof.
13993. The method of item 13960 wherein the agent is . an endotoxin antagonist.
13994. The method of item 13960 wherein the agent is an epothilone and tubulin binder.
13995. The method of item 13960 wherein the agent is an estrogen receptor antagonist.
13996. The method of item 13960 wherein the agent is an FGF inhibitor.
13997. The method of item 13960 wherein the agent is a farnexyl transferase inhibitor. 13998. The method of item 13960 wherein the agent is farnesyitransferase inhibitor selected from the group of A-197574 (Abbott), a famesyltransferase inhibitor from Servier, FPTIII (Strathclyde Institute for Drug R)1 LB-42908 (LG Life Sciences), Pharmaprojects No. 5063 (Genzyme), Pharmaprojects No. 5597 (Ipsen), Yissum Project No. B-1055 (Yissum), and an analogue or derivative thereof.
13999. The method of item 13960 wherein the agent is an FLT-3 kinase inhibitor.
14000. The method of item 13960 wherein the agent is an FGF receptor kinase inhibitor.
14001. The method of item 13960 wherein the agent is a fibrinogen antagonist selected from the group consisting of AUV-201 (Auvation), MG-13926 (Sanofi-Aventis), plasminogen activator (CAS No. 105913-11-9) (from Sanofi-Aventis or UCB), plasminogen activator-2 (tPA- 2) (Sanofi-Aventis), pro-urokinase (CAS No. 82657-92-9) (Sanofi-Aventis), and an analogue or derivative thereof.
14002. The method of item 13960 wherein the agent is a heat shock protein 90 antagonist selected from the group consisting of SRN- 005 (Sirenade), geldanamycin, NSC-33050 (17-allylaminogeldanamycin; 17- AAG), 17-dimethylaminoethylamino-17-demethoxy-geldanamycin (17- DMAG), rifabutin (rifamycin XIV, 1',4-didehydro-1-deoxy-1,4-dihydro-5'-(2- methylpropyl)-1-oxo-), and an analogue or derivative thereof.
14003. The method of item 13960 wherein the agent is a histone deacetylase inhibitor. 14004. The method of item 13960 wherein the agent is an HMGCoA reductase inhibitor selected from the group consisting of an atherosclerosis therapeutic from Lipid Sciences, ATI-16000 (ARYx Therapeutics), KS-01-019 (Kos Pharmaceuticals), Pharmaprojects No. 2197 (Sanofi-Aventi), RP 61969 (Sanofi-Aventis), and an analogue or derivative thereof.
14005. The method of item 13960 wherein the agent is an ICAM inhibitor.
14006. The method of item 13960 wherein the agent is an IL, ICE and IRAK antagonist, wherein the antagonist is a CJ-14877, CP- 424174 (Pfizer), NF-61 (Negma-Lerads), and an analogue or derivative thereof.
14007. The method of item 13960 wherein the agent is an IL-2 inhibitor.
14008. The method of item 13960 wherein the agent is an immunosuppressant selected from the group consisting of teriflunomide (Sanofi Aventis), chlorsulfaquinoxalone (NSC-339004), chlorsulfaqυinoxalone sulfate, CS-712 (Sankyo), ismomultin alfa (CAS No. 457913-93-8) (Akzo Nobel), antiailergics from GenPat77, antiinflammatories or AT-005 (Androclus Therapeutics), autoimmune disease therapeutics from EpiVax, BN-007 (Bone), budesonide (CAS No. 51333-22- 3) (MAP Pharmaceuticals), CO-14 (Genzyme), edratide (CAS No. 433922- 67-9) (Teva), EP-314 (Enanta), eprovafen (CAS No. 101335-99-3) (Sanofi- Aventis), HWA-131 (CAS No. 118788-41-3) (Sanofi-Aventis), immunomodulators from MerLion Pharmaceuticals, immunosuppressives from Alchemia, IPL-12 (Inflazyme), MDL-9563 (CAS No. 27086-86-8) (Sanofi-Aventis), Pharmaprojects No. 2330 (Sanofi-Aventis), Pharmaprojects No. 6426 (Abgenix), PXS-25 (Pharmaxis), rosmarinic acid (CAS No. 20283-92-5) (Sanofi-Aventis), RP 42927 or RP 54745 (CAS No. 135330-08-4) (Sanofi-Aventis), SGN-35 (Seattle Genetics), ST-1959 (Sigma-Tau), type I diabetes therapy from SYNX Pharma, UNIL-88 (Debiopharm), VP-025 (Vasogen), VR-694 (Vectura), PRTX-001 (Protalex), and an analogue or derivative thereof.
14009. The method of item 13960 wherein the agent is an IMPDH (inosine monophosphate).
14010. The method of item 13960 wherein the agent is an integrin antagonist.
14011. The method of item 13960 wherein the agent is an interleukin antagonist.
14012. The method of item 13960 wherein the agent is an inhibitor of type III receptor tyrosine kinase.
14013. The method of item 13960 wherein the agent is an irreversible inhibitor of enzyme methionine aminopeptidase type 2.
14014. The method of item 13960 wherein the agent is an isozyme selective delta protein kinase C inhibitor.
14015. The method of item 13960 wherein the agent a JAK3 enzyme inhibitor.
14016. The method of item 13960 wherein the agent is a JNK inhibitor. 14017. The method of item 13960 wherein the agent is a kinase inhibitor.
14018. The method of item 13960 wherein the agent is kinesin antagonist.
14019. The method of item 13960 wherein the agent is a kinesin antagonist.
14020. The method of item 13960 wherein the agent is a leukotriene inhibitor and antagonist selected from the group consisting of ambicromil (CAS No. 58805-38-2) (Sanofi-Aventis), amelubant (CAS No. 346735-24-8) (Boehringer Ingelheim), DW-1141 (Dong Wha), ebselen (Daiichi Pharmaceutical), ibudilast (Kyorin), leucotriene inhibitors from Sanofi-Aventis, lymphotoxin-beta receptor (LT-D) from Biogen Idee, Pharmaprojects No. 1535 and 2728 (CAS No. 119340-33-9) (Sanofi- Aventis), R-112 (Rigel), Rev-5367 (CAS No. 92532-05-3) (Sanofi-Aventis), RG-14893 (CAS No. 141835-49-6) (Sanofi-Aventis), RG-5901-A (CAS No. 101910-24-1), 92532-23-5, RP 66153 (CAS No. 142422-79-5), RP 66364 (CAS No.186916-92-5), RP 69698 (CAS No. 141748-00-7) (Sanofi-Aventis), SC-411930 (Pfizer), SC-41930 (CAS No. 120072-59-5) (Pfizer), SC-50605 (CAS No. 138828-39-4) (Pfizer), SC-51146 (CAS No. 141059-52-1), SC- 53228 (CAS No. 153633-01-3) (Pfizer), spaglumic acid (ZY-15106) (CAS No.3106-85-2), 80619-64-3 (Novartis), tipredane (CAS No. 85197-77-9) (Bristol-Myers Squibb), U-75302 (CAS No. 119477-85-9) (Pfizer), and analogue or derivative thereof.
14021. The method of item 13960 wherein the agent is an MAP kinase inhibitor. 14022. The method of item 13960 wherein the agent is a matrix metalloproteinase inhibitor.
14023. The method of item 13960 wherein the agent is an MCP-CCR2 inhibitor.
14024. The method of item 13960 wherein the agent is an mTOR inhibitor.
14025. The method of item 13960 wherein the agent is an mTOR kinase inhibitor.
14026. The method of item 13960 wherein the agent is a microtubule inhibitor selected from the group consisting of antibody- maytansinoid conjugates from Biogen Idee, colchicines (MantiCore Pharmaceuticals), anticancer immunoconjugates from Johnson & Johnson, DIME from Octamer, gni-1f (GNI), huC242-DM4, huMy9-6-DM1 (ImmunoGen), IDN-5404 (Indena), IMO-098, IMOderm (Imotep), mebendazole (Introgen Therapeutics), microtubule poisons from Cambridge Enterprise, paclitaxel such as LOTAX from Aphios (CAS No. 33069-62-4), Genexol-PM from Samyang, Pharmaprojects No. 6383 (Tapestry Pharmaceuticals), RPR-112378 (Sanofi-Aventis), SGN-75 (Seattle Genetics), SPL-7435 (Starpharma), SSR-250411 (Sanofi-Aventis), trastuzumab-DM1 (Genentech), vinorelbine, and an analogue or derivative thereof.
14027. The method of item 13960 wherein the agent is an MIF inhibitor.
14028. The method of item 13960 wherein the agent is an MMP inhibitor. 14029. The method of item 13960 wherein the agent is a neurokinin (NK) antagonist selected from the group consisting of anthrotainin (CAS No. 148084-40-6) (Sanofi-Aventis), an IBS thereapeutic from ArQuIe, MDL-105212A (CAS No. 167261-60-1) (Ssanofi-Aventis), Pharmaprojects No. 2744, 3258 (CAS No. 139167-47-8) 4006, 4201, or 5986 (Sanofi-Aventis), RP 67580 (CAS No. 135911-02-3), SR-144190 (CAS No. 201152-86-5), SSR-240600, SSR-241586 (Sanofi-Aventis), TKA-457 (Novartis), vestipitant mesylate (CAS No. 334476-64-1) (GlaxoSmithKline), Win-64821 (Sanofi-Aventis), PRX-96026 (Predix Pharmaceuticals), and an analogue or derivative thereof.
14030. The method of item 13960 wherein the agent is an NF kappa B inhibitor selected from the group consisting of AVE-0545 or AVE-0547 (Sanofi-Aventis), bortezomib (CAS No. 179324-69-7) (Millennium Pharmaceuticals), dexanabinol (CAS No. 112924-45-5) (Pharmos), dexlipotam (Viatris), Pharmaprojects No. 6283 (INDRA) (OXiGENE), IPL- 576092 (CAS No. 137571-30-3) (Inflazyme), NFKB decoy (Corgentech), NFKB decoy oligo (AnGes MG), NFKB's from Ariad, osteoporosis treatments or S5 (F005) from Fulcrum Pharmaceuticals, P61 (Phytopharm), R-flurbiprofen (CAS No. 5104-49-4) (Encore Pharmaceuticals), and an analogue or derivative thereof.
14031. The method of item 13960 wherein the agent is a nitric oxide agonist.
14032. The method of item 13960 wherein the agent is an ornithine decarboxylase inhibitor.
14033. The method of item 13960 wherein the agent is a p38 MAP kinase inhibitor selected from the group consisting of AZD-6703 (AstraZeneca), JX-401 (Jexys Pharmaceuticals), BMS-2 (Bristol-Myers Squibb), a p38 MAP kinase inhibitor from Novartis, a p38-alpha MAP kinase inhibitor from Amphora, Pharmaprojects No. 5704 (Pharmacopeia), RPR- 200765A (Sanofi-Aventis), SD-282 (Johnson & Johnson), TAK-715 (Takeda), and an analogue or derivative thereof.
14034. The method of item 13960 wherein the agent is a palmitoyl-protein thioesterase inhibitor.
14035. The method of item 13960 wherein the agent is a PDGF receptor kinase inhibitor selected from the group consisting of AAL- 993, AMN-107, or ABP-309 (Novartis), AMG-706 (Amgen), BAY-57-9352 (Bayer), CDP-860 (UCB), E-7080 (Eisai), imatinib (CAS No. 152459-95-5) (Novartis), OSI-930 (OSI Pharmaceuticals), RPR-127963E (Sanofi-Aventis), RWJ-540973 (Johnson & Johnson), sorafenib tosylate (Bayer), SU-11657 (Pfizer), tandutinib (CAS No. 387867-13-2) (Millennium Pharmaceuticals), vatalanib (Novartis), ZK-CDK (Schering AG), and an analogue or derivative thereof.
14036. The method of item 13960 wherein the agent is (-)-halofenate (Metabolex), AMG-131 (Amgen), antidiabetics from Japan Tobacco, AZD-4619, AZD-8450, AZD-8677 (AstraZeneca), DRF-10945, balaglitazone (Dr Reddy's), CS-00088, CS-00098 (Chipscreen Biosciences), E-3030 (Eisai), etalocib (CAS No. 161172-51-6) (EIi Lilly), GSK-641597 (Ligand), GSK-677954 (GlaxoSmithKline), GW-409544 (Ligand), GW- 590735 (GlaxoSmithKline), K-111 (Hoffmann-La Roche), LY-518674 (EIi Lilly), LY-674 (Ligand), LY-929 (Ligand), MC-3001, MC-3002 (MaxoCore Pharmaceuticals), metformin HCI + pioglitazone (CAS No. 1115-70-4 and 112529-15-4), ACTOPLUS MET from Andrx), muraglitazar (CAS No. 331741-94-7) (Bristol-Myers Squibb), naveglitazar (Ligand), oleoylethanolamide (Kadmus Pharmaceuticals), ONO-5129, pioglitazone hydrochloride (CAS No. 111025-46-8 and 112529-15-4) (Takeda), PLX-204 (Plexxikon), PPAR agonists from Genfit, PPAR delta agonists from EIi Lilly, PPAR-alpha agonists from CrystalGenomics, PPAR-gamma modulators and PPAR-β modulators from CareX, rosiglitazone maleate (CAS No. 122324- 73-4 or 155141-29-0) (GlaxoSmithKline), rosiglitazone maleate/glimepir (CAS No. 155141-29-0 and 93479-97-1), AVANDARYL1 rosiglitazone maleate/metformin extend (CAS No. 155141-29-0 and 657-24-9), AVANDAMET, rosiglitazone maleate+metformin, AVANDAMET (GlaxoSmithKline), tesaglitazar (AstraZeneca), and an analogue or derivative thereof.
14037. The method of item 13960 wherein the agent is a phosphatase inhibitor.
14038. The method of item 13960 wherein the agent is a phosphodiesterase (PDE) inhibitor selected from the group consisting of avanafil (Tanabe Seiyaku), dasantafil (CAS No. 569351-91-3) (Schering- Plough), A-906119 (CAS No. 134072-58-5), DL-850 (Sanofi-Aventis), GRC- 3015, GRC-3566, GRC-3886 (Glenmark), HWA-153 (CAS No. 56395-66-5) (Sanofi-Aventis), hydroxypumafentrine (Altana), IBFB-130011, IBFB-14-016, IBFB-140301 , IBFB-150007, IBFB-211913 (IBFB Pharma), L-826141 (Merck & Co), medorinone (CAS No. 88296-61-1) (Sanofi-Aventis), MEM- 1917 (Memory Pharmaceuticals), ND-1251 (Neuro3d), PDE inhibitors from ICOS, PDE IV inhibitors from Memory Pharmaceuticals and CrystalGenomics, Pharmaprojects No. 2742 and 6141 (Sanofi-Aventis), QAD-171 (Novartis), RHC-2963 (CAS No. 76993-12-9 and 76993-14-1), RPR-117658, RPR-122818 derivatives, SR-24870 , and RPR-132294 (Sanofi-Aventis), SK-350 (ln2Gen), stroke therapy agents from deCODE Genetics, TAS-203 (Taiho), tofimilast (CAS No. 185954-27-2) (Pfizer), UK- 371800 (Pfizer), WIN-65579 (CAS No. 158020-82-7) (Sanofi-Aventis), IBFB- 130020 (IBFB Pharma), OPC-6535 (CAS No. 145739-56-6) (Otsuka), a phosphodiesterase III inhibitor, enoximone, a phosphodiesterase IV inhibitor, fosfosal, Atopik (Barrier Therapeutics), triflusal, a phosphodiesterase V inhibitor, and an analogue or derivative thereof.
14039. The method of item 13960 wherein the agent is a PKC inhibitor.
14040. The method of item 13960 wherein the agent is a platelet activating factor antagonist.
14041. The method of item 13960 wherein the agent is a platelet-derived growth factor receptor kinase inhibitor.
14042. The method of item 13960 wherein the agent is a prolyl hydroxylase inhibitor.
14043. The method of item 13960 wherein the agent is a polymorphonuclear neutrophil inhibitor.
14044. The method of item 13960 wherein the agent is a protein kinase B inhibitor.
14045. The method of item 13960 wherein the agent is a protein kinase C stimulant.
14046. The method of item 13960 wherein the agent is a purine nucleoside analogue.
14047. The method of item 13960 wherein the agent is a purinoreceptor P2X antagonist. 14048. The method of item 13960 wherein the agent is a Raf kinase inhibitor.
14049. The method of item 13960 wherein the agent is a reversible inhibitor of ErbB1 and ErbB2.
14050. The method of item 13960 wherein the agent is a ribonucleoside triphosphate reductase inhibitor.
14051. The method of item 13960 wherein the agent is an SDF-1 antagonist.
14052. The method of item 13960 wherein the agent is a sheddase inhibitor.
14053. The method of item 13960 wherein the agent is an SRC inhibitor.
14054. The method of item 13960 wherein the agent is a stromelysin inhibitor.
14055. The method of item 13960 wherein the agent is an Syk kinase inhibitor.
14056. The method of item 13960 wherein the agent is a telomerase inhibitor.
14057. The method of item 13960 wherein the agent is a TGF beta inhibitor selected from the group consisting of pirfenidone (CAS No. 53179-13-8) (MARNAC), tranilast (CAS No. 53902-12-8) (Kissei), IN- 1130 (ln2Gen), mannose-6-phosphate (BTG), TGF-β antagonists from Inflazyme (Pharmaprojects No. 6075), TGF-β antagonists from Sydney, non-industrial source), TGF-β I receptor kinase inhibitors from EIi Lilly, TGF- β receptor inhibitors from Johnson & Johnson, and an analogue or derivative thereof.
14058. The method of item 13960 wherein the agent is a
TNFα antagonist or TACE inhibitor selected from the group consisting of adalimumab (CAS No. 331731-18-1) (Cambridge Antibody Technology), AGIX-4207 (AtheroGenics), AGT-1 (Advanced Biotherapy), an antiinflammatory from Borean Pharma, Cellzome, or Paradigm Therapeutics, anti-inflammatory vaccine (TNF-alpha kinoid) from Neovacs, humanized anti-TNF antibody or an anti-TNF MAb (CB0006) Celltech (UCB), apratastat (CAS No. 287445-51-0) (Wyeth), BMS-561392 (Bristol-Myers Squibb), BN- 006 (Bone), certolizumab pegol (CAS No. 428863-50-7 or CH-138 (UCB), cilomilast (CAS No. 153259-65-5) (GlaxoSmithKline), CR-1 (Nuada Pharmaceuticals), CRx-119 (CombinatoRx), D-5410 (UCB), dacopafant (CAS No. 125372-33-0) (Sanofi-Aventis), dersalazine (CAS No. 188913-57- 7/188913-58-8) (Uriach), etanercept (CAS No. 185243-69-0) (Amgen), ethyl pyruvate (Critical (Critical Therapeutics), golimumab (CAS No. 476181-74-5) (Johnson & Johnson), hormono-immunotherapy from Ipsen, CDP571 (e.g., Humicade from UCB), IC-485 (ICOS), infliximab (CAS No. 170277-31-3) (Johnson & Johnson), IP-751 (Manhattan Pharmaceuticals), ISIS-104838 (CAS No. 250755-32-9) (ISIS Pharmaceuticals), lenalidomide (CAS No. 191732-72-6) (Celgene), lentinan (CAS No. 37339-90-5) (Ajinomoto), MDL- 201112 (CAS No. 142130-73-2) (Sanofi-Aventis), medroxyprogesterone (CAS No. 520-85-4) (InKine Pharmaceutical), N-acetylcysteine (CAS No. 616-91-1) (Zambon), NBE-P2 (DIREVO Biotech), nerelimomab (CAS No. 162774-06-3) (Chiron), OM-294DP (OM PHARMA), onercept (CAS No. 199685-57-9) (Yeda), PASSTNF-alpha (Verigen), pentoxifylline or oxypentifylline (Sanofi-Aventis), Pharmaprojects No. 4091 , 4241, 4295, or 4488 (Sanofi-Aventis), Pharmaprojects No. 5480 (Amgen), Pharmaprojects No. 6749 (Cengent), pirfenidone (CAS No. 53179-13-8) (MARNAC), RPR- 132294 (Sanofi-Aventis), S5 (F002) (Fulcrum Pharmaceuticals), simvastatin (CAS No. 79902-63-9) (Merck & Co), STA-6292 (Synta Pharmaceuticals), tacrolimus (CAS No. 104987-11-3) (Fujisawa LifeCycle Pharma), talactoferrin alfa (CAS No. 308240-58-6) (Agennix), thalidomide (CAS No. 50-35-1) (Celgene), TNF antagonists form ProStrakan, and Synergen, TNF inhibitors (Amgen), TNF-alpha antagonists from Dynavax Technologies and Jerina AG (Germany), TNF-alpha inhibitors from IBFB Pharma and Xencor (Xencor), torbafylline (CAS No. 105102-21-4) (Sanofi-Aventis), UR-1505 (Uriach), VT-346 (Viron Therapeutics), YSIL6 (Y1S Therapeutics), YSTH2 (Y's Therapeutics), NPI-1302a-3 (Nereus Pharmaceuticals, a TNF antagonist from Jerina AG (Germany), dersalazine, and an analogue or derivative thereof.
14059. The method of item 13960 wherein the agent is a Toll receptor inhibitor.
14060. The method of item 13960 wherein the agent is a tubulin antagonist.
14061. The method of item 13960 wherein the agent is a tyrosine kinase inhibitor selected from the group consisting of SU-011248, SUTENT from Pfizer Inc. (New York, NY), BMS-354825, PN-355 (Paracelsian Pharmaceuticals), AGN-199659 (Allergan), AAL-993 or ABP- 309 (Novartis), adaphostin (NIH), AEE-788 (Novartis), AG-013736 (OSI Pharmaceuticals), AG-13736 (Pfizer), ALT-110 (Alteris Therapeutics), AMG- 706 (Amgen), anticancer MAbs from Xencor, anti-EGFrvlll MAbs from Abgenix, anti-HER2 MAb from Abiogen, AZD-2171 or AZD-9935 (AstraZeneca), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), CEP-5214 (Cephalon), CEP-7055 (Cephalon), cetuximab (ImClone Systems), CHIR-200131 and CHIR-258 (Chiron), CP-547632 (OSI Pharmaceuticals), CP-724714 (Pfizer), CT-301 (Creabilis Therapeutics), D- 69491 (Baxter International), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EGFR/ErbB2 inhibitors from Array BioPharma, erlotinib (CAS No. 183319- 69-9) (OSI Pharmaceuticals), EXEL-2880 (Exelixis), FK-778 (Sanofi- Aventis) , gefitinib (CAS No. 184475-35-2) (AstraZeneca), GW-2286 or GW- 654652 (GlaxoSmithKline), her2/neu antigen from AlphaVax, HER-2/neu inhibitor from Generex, Herzyme (Medipad) (Sirna Therapeutics), HKI-272 (Wyeth), HuMax-EGFr (Genmab), idronoxil (CAS No. 81267-65-4) (Novogen), IGF-1 inhibitors from Ontogen, IMC-11F8 (ImCIone Systems), kahalalide F (CAS No. 149204-42-2) (PharmaMar), KDR inhibitor from LG Life Sciences, KDR inhibitors from Abbott Laboratories, KDR kinase inhibitors (UCB), Kdr kinase inhibitors from Merck & Co, KRN-633 and KRN- 951 (Kirin Brewery), KSB-102 (Xenova), lapatinib ditosylate (CAS No. 388082-78-8) (GlaxoSmithKline), matuzumab (Merck KGaA), MDX-214 (Medarex), ME-103 (Pharmexa), MED-A300 (Gerolymatos), MNAC-13 (Lay Line Genomics), nimotuzumab (Center of Molecular Immunology), NSC- . 330507 or NSC-707545 (NIH), NV-50 (Novogen), OSI-930 (OSI Pharmaceuticals), panitumumab (Abgenix), pelitinib (CAS No. 287933-82-7) (Wyeth), pertuzumab (CAS No. 380610-27-5) (Genentech), Pharmaprojects No. 3985 (Sanofi-Aventis), prostate cancer therapeutics from Sequenom (SQPC35, SQPC36, SQPC90), removab and remoxab (Trion Pharma), RG- 13022 (CAS No. 136831-48-6), RG-13291 (CAS No. 138989-50-1), or RG- 14620 (CAS No. 136831-49-7) (Sanofi-Aventis), RM-6427 (Romark), RNAi breast cancer therapy from Benitec, RP 53801 (CAS No. 125882-88-4) (Sanofi-Aventis), sorafenib tosylate (Bayer), SU-11657 (Pfizer), Tie-2 antagonists from Semaia (Hybrigenics), Tie-2 inhibitors from Ontogen, trastuzumab (CAS No. 180288-69-1) (Genentech), tyrosine kinase inhibitors from Sanofi-Aventis, U3-1287, U3-1565, U3-1784, U3-1800 (U3 Pharma), vatalanib (Novartis), VEGFR-2 kinase inhibitor from Bristol-Myers Squibb, XL-647 (Exelixis), ZD-6474 (AstraZeneca), ZK-CDK (Schering AG), an EGFR tyrosine kinase inhibitor, EKB-569 (Wyeth), and an analogue or derivative thereof.
14062. The method of item 13960 wherein the agent is a VEGF inhibitor.
14063. The method of item 13960 wherein the agent is a vitamin D receptor agonist.
14064. The method of item 13960 wherein the agent is ZD-6474 (an angiogenesis inhibitor).
14065. The method of item 13960 wherein the agent is AP-23573 (an mTOR inhibitor).
14066. The method of item 13960 wherein the agent is synthadotin (a tubulin antagonist).
14067. The method of item 13960 wherein the agent is S-0885 (a collagenase inhibitor).
14068. The method of item 13960 wherein the agent is aplidine (an elongation factor-1 alpha inhibitor).
14069. The method of item 13960 wherein the agent is ixabepilone (an epithilone).
14070. The method of item 13960 wherein the agent is IDN-5390 (an angiogenesis inhibitor). 14071. The method of item 13960 wherein the agent is SB-2723005 (an angiogenesis inhibitor).
14072. The method of item 13960 wherein the agent is ABT-518 (an angiogenesis inhibitor).
14073. The method of item 13960 wherein the agent is combretastatin (an angiogenesis inhibitor).
14074. The method of item 13960 wherein the agent is anecortave acetate (an angiogenesis inhibitor).
14075. The method of item 13960 wherein the agent is SB-715992 (a kinesin antagonist).
14076. The method of item 13960 wherein the agent is temsirolimus (an mTOR inhibitor).
14077. The method of item 13960 wherein the agent is adalimumab (a TNFα antagonist).
14078. The method of item 13960, wherein the composition comprises a polymer.
14079. The method of item 13960, wherein the composition comprises a polymeric carrier.
14080. The method of item 13960 wherein the anti- scarring agent inhibits adhesion between the device and a host into which the device is implanted. 14081. The method of item 13960 wherein the device delivers the anti-scarring agent locally to tissue proximate to the device.
14082. The method of item 13960 wherein the device has a coating that comprises the anti-scarring agent.
14083. The method of item 13960, wherein the device has a coating that comprises the agent and is disposed on a surface of the implant.
14084. The method of item 13960, wherein the device has a coating that comprises the agent and directly contacts the implant.
14085. The method of item 13960, wherein the device has a coating that comprises the agent and indirectly contacts the implant.
14086. The method of item 13960, wherein the device has a coating that comprises the agent and partially covers the implant.
14087. The method of item 13960, wherein the device has a coating that comprises the agent and completely covers the implant.
14088. The method of item 13960, wherein the device has a uniform coating.
14089. The method of item 13960, wherein the device has a non-uniform coating.
14090. The method of item 13960, wherein the device has a discontinuous coating. 14091. The method of item 13960, wherein the device has a patterned coating.
14092. The method of item 13960, wherein the device has a coating with a thickness of 100 Dm or less.
14093. The method of item 13960, wherein the device has a coating with a thickness of 10 Dm or less.
14094. The method of item 13960, wherein the device has a coating, and the coating adheres to the surface of the implant upon deployment of the implant.
14095. The method of item 13960, wherein the device has a coating, and wherein the coating is stable at room temperature for a period of 1 year.
14096. The method of item 13960, wherein the device has a coating, and wherein the anti-scarring agent is present in the coating in an amount ranging between about 0.0001% to about 1% by weight.
14097. The method of item 13960, wherein the device has a coating, and wherein the anti-scarring agent is present in the coating in an amount ranging between about 1% to about 10% by weight.
14098. The method of item 13960, wherein the device has a coating, and wherein the anti-scarring agent is present in the coating in an amount ranging between about 10% to about 25% by weight. 14099. The method of item 13960, wherein the device has a coating, and wherein the anti-scarring agent is present in the coating in an amount ranging between about 25% to about 70% by weight.
14100. The method of item 13960, wherein the device has a coating, and wherein the coating further comprises a polymer.
14101. The method of item 13960, wherein the device has a first coating having a first composition and a second coating having a second composition.
14102. The method of item 13960, wherein the device has a first coating having a first composition and a second coating having a second composition, wherein the first composition and the second composition are different.
14103. The method of item 13960, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a copolymer.
14104. The method of item 13960, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a block copolymer.
14105. The method of item 13960, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a random copolymer.
14106. The method of item 13960, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a biodegradable polymer. 14107. The method of item 13960, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a non-biodegradable polymer.
14108. The method of item 13960, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a hydrophilic polymer.
14109. The method of item 13960, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a hydrophobic polymer.
14110. The method of item 13960, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a polymer having hydrophilic domains.
14111. The method of item 13960, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a polymer having hydrophobic domains.
14112. The method of item 13960, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a non-conductive polymer.
14113. The method of item 13960, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises an elastomer.
14114. The method of item 13960, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a hydrogel. 14115. The method of item 13960, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a silicone polymer.
14116. The method of item 13960, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a hydrocarbon polymer.
14117. The method of item 13960, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a styrene-derived polymer.
14118. The method of item 13960, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a butadiene polymer.
14119. The method of item 13960, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a macromer.
14120. The method of item 13960, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a poly(ethylene glycol) polymer.
14121. The method of item 13960 wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises an amorphous polymer.
14122. The method of item 13960, wherein the device comprises a lubricious coating. 14123. The method of item 13960 wherein the anti- scarring agent is located within pores or holes of the device.
14124. The method of item 13960 wherein the anti- scarring agent is located within a channel, lumen, or divet of the device.
14125. The method of item 13960, wherein the device comprises a second pharmaceutically active agent.
14126. The method of item 13960 wherein the device comprises an anti-inflammatory agent.
14127. The method of item 13960 wherein the device comprises an agent that inhibits infection.
14128. The method of item 13960 wherein the device . comprises an agent that inhibits infection, and wherein the agent is an anthracycline.
14129. The method of item 13960 wherein the device comprises an agent that inhibits infection, and wherein the agent is doxorubicin.
14130. The method of item 13960 wherein the device comprises an agent that inhibits infection, and wherein the agent is mitoxantrone.
14131. The method of item 13960 wherein the device comprises an agent that inhibits infection, and wherein the agent is a fluoropyrimidine. 14132. The method of item 13960 wherein the device comprises an agent that inhibits infection, and wherein the agent is 5- fluorouracil (5-FU).
14133. The method of item 13960 wherein the device comprises an agent that inhibits infection, and wherein the agent is a folic acid antagonist.
14134. The method of item 13960 wherein the device comprises an agent that inhibits infection, and wherein the agent is methotrexate.
14135. The method of item 13960 wherein the device comprises an agent that inhibits infection, and wherein the agent is a podophylotoxin.
14136. The method of item 13960 wherein the device comprises an agent that inhibits infection, and wherein the agent is etoposide.
14137. The method of item 13960 wherein the device comprises an agent that inhibits infection, and wherein the agent is a camptothecin.
14138. The method of item 13960 wherein the device comprises an agent that inhibits infection, and wherein the agent is a hydroxyurea.
14139. The method of item 13960 wherein the device comprises an agent that inhibits infection, and wherein the agent is a platinum complex. 14140. The method of item 13960 wherein the device comprises an agent that inhibits infection, and wherein the agent is cisplatin.
14141. The method of item 13960, further comprising an antithrombotic agent.
14142. The method of item 13960 wherein the device comprises a visualization agent.
14143. The method of item 13960 wherein the device comprises a visualization agent, wherein the visualization agent is a radiopaque material, and wherein the radiopaque material comprises a metal, a halogenated compound, or a barium containing compound.
14144. The method of item 13960 wherein the device comprises a visualization agent, wherein the visualization agent is a radiopaque material, and wherein the radiopaque material comprises barium, tantalum, or technetium.
14145. The method of item 13960 wherein the device comprises a visualization agent, and wherein the visualization agent is a MRI responsive material.
14146. The method of item 13960 wherein the device comprises a visualization agent, and wherein the visualization agent comprises a gadolinium chelate.
14147. The method of item 13960 wherein the device comprises a visualization agent, and wherein the visualization agent comprises iron, magnesium, manganese, copper, or chromium. 14148. The method of item 13960 wherein the device comprises a visualization agent, and wherein the visualization agent comprises an iron oxide compound.
14149. The method of item 13960 wherein the device comprises a visualization agent, and wherein the visualization agent comprises a dye, pigment, or colorant.
14150. The method of item 13960 wherein the device comprises an echogenic material.
14151. The method of item 13960 wherein the device comprises an echogenic material, and wherein the echogenic material is in the form of a coating.
14152. The method of item 13960 wherein the device is sterile.
14153. The method of item 13960 wherein the anti- scarring agent is released into tissue in the vicinity of the device after deployment of the device.
14154. The method of item 13960 wherein the anti- scarring agent is released into tissue in the vicinity of the device after deployment of the device, and wherein the tissue is connective tissue.
14155. The method of item 13960 wherein the anti- scarring agent is released into tissue in the vicinity of the device after deployment of the device, and wherein the tissue is muscle tissue. 14156. The method of item 13960 wherein the anti- scarring agent is released into tissue in the vicinity of the device after deployment of the device, and wherein the tissue is nerve tissue.
14157. The method of item 13960 wherein the anti- scarring agent is released into tissue in the vicinity of the device after deployment of the device, and wherein the tissue is epithelium tissue.
14158. The method of item 13960 wherein the anti- scarring agent is released in effective concentrations from the device over a period ranging from the time of deployment of the device to about 1 year.
14159. The method of item 13960 wherein the anti- scarring agent is released in effective concentrations from the device over a period ranging from about 1 month to 6 months.
14160. The method of item 13960 wherein the anti- scarring agent is released in effective concentrations from the device over a period ranging from about 1 - 90 days.
14161. The method of item 13960 wherein the anti- scarring agent is released in effective concentrations from the device at a constant rate.
14162. The method of item 13960 wherein the anti- scarring agent is released in effective concentrations from the device at an increasing rate.
14163. The method of item 13960 wherein the anti- scarring agent is released in effective concentrations from the device at a decreasing rate. 14164. The method of item 13960 wherein the anti- scarring agent is released in effective concentrations from the composition comprising the anti-scarring agent by diffusion over a period ranging from the time of deployment of the device to about 90 days.
14165. The method of item 13960 wherein the anti- scarring agent is released in effective concentrations from the composition comprising the anti-scarring agent by erosion of the composition over a period ranging from the time of deployment of the device to about 90 days.
14166. The method of item 13960 wherein the device comprises about 0.01 Dg to about 10 Dg of the anti-scarring agent.
14167. The method of item 13960 wherein the device comprises about 10 Dg to about 10 mg of the anti-scarring agent.
14168. The method of item 13960 wherein the device comprises about 10 mg to about 250 mg of the anti-scarring agent.
14169. The method of item 13960 wherein the device comprises about 250 mg to about 1000 mg of the anti-scarring agent.
14170. The method of item 13960 wherein the device comprises about 1000 mg to about 2500 mg of the anti-scarring agent.
14171. The method of item 13960 wherein a surface of the device comprises less than 0.01 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied. 14172. The method of item 13960 wherein a surface of the device comprises about 0.01 Dg to about 1 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
14173. The method of item 13960 wherein a surface of the device comprises about 1 Dg to about 10 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
14174. The method of item 13960 wherein a surface of the device comprises about 10 Dg to about 250 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
14175. The method of item 13960 wherein a surface of the device comprises about 250 Dg to about 1000 Dg of the anti-scarring agent of anti-scarring agent per mm2 of device surface to which the anti- scarring agent is applied.
14176. The method of item 13960 wherein a surface of the device comprises about 1000 Dg to about 2500 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
14177. The method of item 13960 wherein the combining is performed by direct affixing the agent or the composition to the implant.
14178. The method of item 13960 wherein the combining is performed by spraying the agent or the component onto the implant. 14179. The method of item 13960 wherein the combining is performed by electrospraying the agent or the composition onto the implant.
14180. The method of item 13960 wherein the combining is performed by dipping the implant into a solution comprising the agent or the composition.
14181. The method of item 13960 wherein the combining is performed by covalently attaching the agent or the composition to the implant.
14182. The method of item 13960 wherein the combining is performed by non-covalently attaching the agent or the composition to the implant.
14183. The method of item 13960 wherein the combining is performed by coating the implant with a substance that contains the agent or the composition.
14184. The method of item 13960 wherein the combining is performed by coating the implant with a substance that absorbs the agent.
14185. The method of item 13960 wherein the combining is performed by interweaving a thread composed of, or coated with, the agent or the composition.
14186. The method of item 13960 wherein the combining is performed by covering all the implant with a sleeve that contains the agent or the composition. 14187. The method of item 13960 wherein the combining is performed by covering a portion of the implant with a sleeve that contains the agent or the composition.
14188. The method of item 13960 wherein the combining is performed by covering all the implant with a cover that contains the agent or the composition.
14189. The method of item 13960 wherein the combining is performed by covering a portion of the implant with a cover that contains the agent or the composition.
14190. The method of item 13960 wherein the combining is performed by covering all the implant with an electrospun fabric that contains the agent or the composition.
14191. The method of item 13960 wherein the combining is performed by covering a portion of the implant with an electrospun fabric that contains the agent or the composition.
14192. The method of item 13960 wherein the combining is performed by covering all the implant with a mesh that contains the agent or the composition.
14193. The method of item 13960 wherein the combining is performed by covering a portion of the implant with a mesh that contains the agent or the composition.
14194. The method of item 13960 wherein the combining is performed by constructing all the implant with the agent or the composition. 14195. The method of item 13960 wherein the combining is performed by constructing a portion of the implant with the agent or the composition.
14196. The method of item 13960 wherein the combining is performed by impregnating the implant with the agent or the composition.
14197. The method of item 13960 wherein the combining is performed by constructing all of the implant from a degradable polymer that releases the agent.
14198. The method of item 13960 wherein the combining is performed by constructing a portion of the implant from a degradable polymer that releases the agent.
14199. The method of item 13960 wherein the combining is performed by dipping the implant into a solution that comprise the agent and an inert solvent for the implant.
14200. The method of item 13960 wherein the combining is performed by dipping the implant into a solution that comprises the agent and a solvent that will swell the implant.
14201. The method of item 13960 wherein the combining is performed by dipping the implant into a solution that comprises the agent and a solvent that will dissolve the implant.
14202. The method of item 13960 wherein the combining is performed by dipping the implant into a solution that comprises the agent, a polymer and an inert solvent for the implant. 14203. The method of item 13960 wherein the combining is performed by dipping the implant into a solution that comprises the agent, a polymer and a solvent that will swell the implant.
14204. The method of item 13960 wherein the combining is performed by dipping the implant into a solution that comprises the agent, a polymer and a solvent that will dissolve the implant.
14205. The method of item 13960 wherein the combining is performed by spraying the implant into a solution that comprises the agent and an inert solvent for the implant.
14206. The method of item 13960 wherein the combining is performed by spraying the implant into a solution that comprises the agent and a solvent that will swell the implant.
14207. The method of item 13960 wherein the combining is performed by spraying the implant into a solution that comprises the agent and a solvent that will dissolve the implant.
14208. The method of item 13960 wherein the combining is performed by spraying the implant into a solution that comprises the agent, a polymer and an inert solvent for the implant.
14209. The method of item 13960 wherein the combining is performed by spraying the implant into a solution that comprises the agent, a polymer and a solvent that will swell the implant.
14210. The method of item 13960 wherein the combining is performed by spraying the implant into a solution that comprises the agent, a polymer and a solvent that will dissolve the implant. 14211. A method as in any one of items 13960-14210, wherein the device is a surgical barrier.
14212. A method as in any one of items 13960-14210, wherein the device is a surgical adhesion barrier.
14213. A method as in any one of items 13960-14210, wherein the device is a surgical sheet.
14214. A method as in any one of items 13960-14210, wherein the device is a surgical patch.
14215. A method as in any one of items 13960-14210, wherein the device is a surgical wrap.
14216. A method as in any one of items 13960-14210, wherein the device is a vascular wrap.
14217. A method as in any one of items 13960-14210, wherein the device is a perivascular wrap.
14218. A method as in any one of items 13960-14210, wherein the device is an adventitial wrap.
14219. A method as in any one of items 13960-14210, wherein the device is a periadventitial wrap.
14220. A method as in any one of items 13960-14210, wherein the device is an adventitial sheet. 14221. A method as in any one of items 13960-14210, wherein the device is a perivascular mesh.
14222. A method as in any one of items 13960-14210, wherein the device is a bandage.
14223. A method as in any one of items 13960-14210, wherein the device is a liquid bandage.
14224. A method as in any one of items 13960-14210, wherein the device is a surgical dressing.
14225. A method as in any one of items 13960-14210, wherein the device is a gauze.
14226. A method as in any one of items 13960-14210, wherein the device is a fabric.
14227. A method as in any one of items 13960-14210, wherein the device is a tape.
14228. A method as in any one of items 13960-14210, wherein the device is a surgical membrane.
14229. A method as in any one of items 13960-14210, wherein the device is a polymer matrix.
14230. A method as in any one of items 13960-14210, wherein the device is a tissue covering. 14231. A method as in any one of items 13960-14210, wherein the device is a surgical matrix.
14232. A method as in any one of items 13960-14210, wherein the device is an envelope.
14233. A method as in any one of items 13960-14210, wherein the device is a tissue covering.
14234. A method of making a medical device comprising: combining a glaucoma drainage device (e.g., an implant) and an anti-scarring agent or a composition comprising an anti-scarring agent, wherein the agent inhibits scarring between the device and a host into which the device is implanted.
14235. The method of item 14234 wherein the agent is an adensosine A2A receptor antagonist.
14236. The method of item 14234 wherein the agent is an AKT inhibitor.
14237. The method of item 14234 wherein the agent is an alpha 2 integrin antagonist, wherein the alpha 2 integrin antagonist is Pharmaprojects No. 5754 (Merck KgaA).
14238. The method of item 14234 wherein the agent is an alpha 4 integrin antagonist.
14239. The method of item 14234 wherein the agent is an alpha 7 nicotinic receptor agonist. 14240. The method of item 14234 wherein the agent is an angiogenesis inhibitor selected from the group consisting of AG-12,958 (Pfizer), ATN-161 (Attenuon LLC), neovastat, an angiogenesis inhibitor from Jerina AG (Germany), NM-3 (Mercian), VGA-1155 (Taisho), FCE-26644 (Pfizer), FCE-26950 (Pfizer), FPMA (Meiji Daries), FR-111142 (Fujisawa), GGTI-298, GM-1306 (Ligand), GPA-1734 (Novartis), NNC-47-0011 (Novo Nordisk), herbamycin (Nippon Kayaku), lenalidomide (Celegene), IP-10 (NIH), ABT-828 (Abbott), KIN-841 (Tokushima University, Japan), SF-1126 (Semafore Pharmaceuticals), laminin technology (NIH), CHIR-258 (Chiron), NVP-AEW541 (Novartis), NVP-AEW541 (Novartis), Vt16907 (Alchemia), OXI-8007 (Oxigene), EG-3306 (Ark Therapeutics), Maspin (Arriva), ABT- 567 (Abbott), PPI-2458 (Praecis Pharmaceuticals), CC-5079, CC-4089 (Celgene), HIF-1 alpha inhibitors (Xenova), S-247 (Pfizer), AP-23573 (Ariad), AZD-9935 (Astra Zeneca), mebendazole (Introgen Therapeutics), MetAP-2 inhibitors (GlaxoSmithKline), AG-615 (Angiogene Pharmaceuticals), Tie-2 antagonists (Hybrigenics), NC-381 , CYC-381 , NC-169, NC-219, NC-383, NC-384, NC-407 (Lorus Therapeutics), ATN-224 (Attenuon), ON-01370 (Onconova), Vitronectin antagonists (Amgen), SDX-103 (Salmedix), Vitronectin antagonists (Shire), CHP (Riemser), TEK (Amgen), Anecortave acetate (Alcon), T46.2 (Matrix Therapeutics), HG-2 (Heptagen), TEM antagonists (Genzyme), Oxi-4500 (Oxigene), ATN-161 (Attenuon), WX-293 (Wilex), M-2025 (Metris Therapeutics), Alphastatin (BioActa), YH-16 (Yantai Rongchang), BIBF-1120 (Boehringer Ingelheim), BAY-57-9352 (Bayer), AS- 1404 (Cancer Research Technology), SC-77964 (Pfizer), glycomimetics (BioTie Therapies), TIE-2 Inhibitors (Ontogen), DIMI, Octamer (Octamer), ABR-215050 (Active Biotech), ABT-518 (Abbott), KDR inhibitors (Abbott), BSF-466895 (Abbott), SCH-221153 (Schering-Plough), DAC:antiangiogenic (ConjuChem), TFPI (EntreMed), AZD-2171 (Astra-Zenaca), CDC-394 (Celgene), LY290293 (EIi Lilly), IDN-5390 (Indena), Kdr Kinase Inhibitors (Merck), CT-113020, CT-116433, CT-116563, CT-31890, CT-32228) (Cell Therapeutics), A-299620 (Abbott), TWEAK Inhibitor (Amgen), VEGF modulators (Johnson and Johnson), Tum-N53, tumstatin (Genzyme), Thios- 1 , Thios-2 (Thios Pharmaceuticals), MV-6401 (Miravant Medical Technologies), Spisulosine (PharmaMar), CEP-7055 (Cephalon), AUV-201 (Auvation), LM-609 (EIi Lilly), SKF-106615 (AnorMED), Oglufanide disodium (Cytran), BW-114 (Pharminox), Calreticulin (NIH), WX-678 (Wilex), SD-7784 (Pfizer), WX-UK1 (Wilex), SH-268 (Schering AG), 2-Me-PGA (Celgene), S- 137 (Pfizer), ZD-6126 (Angiogene Pharmaceuticals), SG-292 (SignalGen), Benefin (Lane Labs), A6, A36 (Angstrom), SB-2723005 (GlaxoSmithKline), SC-7 (Cell Therapeutics), ZEN-014 (AEterna Zentaris), 2-methoxyestradiol (EntreMed), NK-130119 (Nippon Kayaku), CC-10004 (Celgene), AVE- 8062A (Ajinomoto), Tacedinaline (Pfizer), Actinonin (Tokyo Metropolitan Institute of Medical Science), Lenalidomide (Celgene), VGA-1155, BTO-956 (SRI International), ER-68203-00 (Eisai), CT-6685 (UCB), JKC-362 (Phoenix Pharmaceuticals), DMI-3798 (DMI Biosciences, Angiomate (Ipsen), ZD-6474 (AstraZeneca), CEP-5214 (Cephalon), Canstatin (Genzyme), NM-3 (Mercian), Oridigm (MediQuest Therapeutics), Exherin (Adherex), BLS-0597 (Boston Life Sciences), PTC-299 (PTC Therapeutics), NPI-2358 (Nereus Pharmaceuticals), CGP-79787 (Novartis), AEE-788 (Novartis), CKD-732 (Chong Kun Dang), CP-564959 (OSI Pharmaceuticals), CM-101 (CarboMed), CT-2584, CT3501 (Cell Therapeutics), combretastatin and analogues and derivatives thereof (Oxigene), Rebimastat (Bristol-Meyers Squibb), Dextrin 2-sulfate (ML Laboratories), Cilengitide (Merk KGaA)1 NSC-706704 (Pharminox), KRN- 951 (Kirin Brewery), Ukrain, NSC-631570 (Nowicky Pharma), Tecogalan sodium (Daiichi Pharmaceutical), Tz-93 (Tsumura), TBC-1635 (Encysive Pharmaceuticals), TAN-1120 (Takeda), Semaxanib (Pfizer), BDI-7800 (Biopharmacopae), SD-186, SD-983 (Bristol-Meyers Squibb), SB-223245 (GlaxoSmithKline), SC-236 (Pfizer), RWJ-590973 (Johnson and Johnson), ILX-1850 (Genzyme), SC-68488, S-836 (Pfizer), CG-55069-11 (CuraGen), Ki-23057 (Kirin Brewery), CCX-700 (Chemoentryx), Pegaptanib octasodium (Giled Sciences), ANGIOCOL (available from Biostratum Inc.), or an analogue or derivative thereof.
14241. The method of item 14234 wherein the agent is an apoptosis antagonist.
14242. The method of item 14234 wherein the agent is an apoptosis activator.
14243. The method of item 14234 wherein the agent is a beta 1 integrin antagonist.
14244. The method of item 14234 wherein the agent is a beta tubulin inhibitor.
14245. The method of item 14234 wherein the agent is a blocker of enzyme production in Hepatitis C.
14246. The method of item 14234 wherein the agent is a Bruton's tyrosine kinase inhibitor.
14247. The method of item 14234 wherein the agent is a calcineurin inhibitor.
14248. The method of item 14234 wherein the agent is a caspase 3 inhibitor.
14249. The method of item 14234 wherein the agent is a CC chemokine receptor antagonist. 14250. The method of item 14234 wherein the agent is a cell cycle inhibitor selected from the group consisting of SNS-595 (Sunesis), synthadotin, KRX-0403, and an analogue or derivative thereof.
14251. The method of item 14234 wherein the agent is a cathepsin B inhibitor.
14252. The method of item 14234 wherein the agent is a cathepsin K inhibitor, wherein the cathepsin K inhibitor is 462795 (GlaxoSmithKline), INPL-022-D6 (Amura Therapeutics), or an analogue or derivative thereof.
14253. The method of item 14234 wherein the agent is a cathepsin L inhibitor.
14254. The method of item 14234 wherein the agent is a CD40 antagonist.
14255. The method of item 14234 wherein the agent is a chemokine receptor agonist.
14256. The method of item 14234 wherein the agent is a chymase inhibitor.
14257. The method of item 14234 wherein the agent is a collagenase antagonist.
14258. The method of item 14234 wherein the agent is a CXCR antagonist. 14259. The method of item 14234 wherein the agent is a cyclin dependent kinase inhibitor selected from the group consisting of a CDK-1 inhibitor, a CDK-2 inhibitor, a CDK- 4 inhibitor, a CDK-6 inhibitor, a CAK1 inhibitor from GPC Biotech and Bristol-Myers Squibb, RGB-286199 (GPC Biotech), an anticancer agent from Astex Technology, a CAK1 inhibitor from GPC Biotech, a CDK inhibitor from Sanofi-Aventis, a CDK1/CDK2 inhibitor from Amgen, a CDK2 inhibitor from SUGEN-2 (Pfizer), a hearing loss therapy agent (Sound Pharmaceuticals), PD-0332991 (Pfizer), RGB-286199 (GPC Biotech), Ro-0505124 (Hoffmann-La Roche), a Ser/Thr kinase inhibitor from Lilly (EIi Lilly), CVT-2584 (CAS No. 199986-75- 9) (CV Therapeutics), and an analogue or derivative thereof.
14260. The method of item 14234 wherein the agent is a cyclooxygenase 1 inhibitor.
14261. The method of item 14234 wherein the_ agent is a DHFR inhibitor.
14262. The method of item 14234 wherein the agent is a dual integrin inhibitor.
14263. The method of item 14234 wherein the agent is an elastase inhibitor.
14264. The method of item 14234 wherein the agent is an elongation factor-1 alpha inhibitor.
14265. The method of item 14234 wherein the agent is an endothelial growth factor antagonist. 14266. The method of item 14234 wherein the agent is an endothelial growth factor receptor kinase inhibitor selected from the group consisting of sorafenib tosylate (Bayer), AAL-993 (Novartis), ABP-309 (Novartis), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), E- 7080 (Eisai), EG-3306 (Ark Therapeutics), EXEL-2880 (Exelixis), GW- 654652 (GlaxoSmithKiine), a KDR inhibitor from LG Life Sciences, CT-6685 and CT-6729 (UCB), KRN-633 and KRN-951 (Kirin Brewery), OSI-930 (OSI Pharmaceuticals), SP-5.2 (Supratek Pharma), SU-11657 (Pfizer), a Tie-2 antagonist (Hybrigenics), a VEGFR-2 kinase inhibitor (Bristol-Myers Squibb), XL-647 (Exelixis), a KDR inhibitor from Abbott Laboratories, sorafenib tosylate, and an analogue or derivative thereof.
14267. The method of item 14234 wherein the agent is an endotoxin antagonist.
14268. The method of item 14234 wherein the agent is an epothilone and tubulin binder.
14269. The method of item 14234 wherein the agent is an estrogen receptor antagonist.
14270. The method of item 14234 wherein the agent is an FGF inhibitor.
14271. The method of item 14234 wherein the agent is a famexyl transferase inhibitor.
14272. The method of item 14234 wherein the agent is famesyltransferase inhibitor selected from the group of A-197574 (Abbott), a farnesyltransferase inhibitor from Servier, FPTIlI (Strathclyde Institute for Drug R), LB-42908 (LG Life Sciences), Pharmaprojects No. 5063 (Genzyme), Pharmaprojects No. 5597 (Ipsen), Yissum Project No. B-1055 (Yissum), and an analogue or derivative thereof.
14273. The method of item 14234 wherein the agent is an FLT-3 kinase inhibitor.
14274. The method of item 14234 wherein the agent is an FGF receptor kinase inhibitor.
14275. The method of item 14234 wherein the agent is a fibrinogen antagonist selected from the group consisting of AUV-201 (Auvation), MG-13926 (Sanofi-Aventis), plasminogen activator (CAS No. 105913-11-9) (from Sanofi-Aventis or UCB), plasminogen activator-2 (tPA- 2) (Sanofi-Aventis), pro-urokinase (CAS No. 82657-92-9) (Sanofi-Aventis), and an analogue or derivative thereof.
14276. The method of item 14234 wherein the agent is a heat shock protein 90 antagonist selected from the group consisting of SRN- 005 (Sirenade), geldanamycin, NSC-33050 (17-allylaminogeldanamycin; 17- AAG), 17-dimethylaminoethylamino-17-demethoxy-geldanamycin (17- DMAG), rifabutin (rifamycin XIV, 1',4-didehydro-1-deoxy-1 ,4-dihydro-5'-(2- methylpropyl)-1-oxo-), and an analogue or derivative thereof.
14277. The method of item 14234 wherein the agent is a histone deacetylase inhibitor.
14278. The method of item 14234 wherein the agent is an HMGCoA reductase inhibitor selected from the group consisting of an atherosclerosis therapeutic from Lipid Sciences, ATI-16000 (ARYx Therapeutics), KS-01-019 (Kos Pharmaceuticals), Pharmaprojects No. 2197 (Sanofi-Aventi), RP 61969 (Sanofi-Aventis), and an analogue or derivative thereof.
14279. The method of item 14234 wherein the agent is an ICAM inhibitor.
14280. The method of item 14234 wherein the agent is an IL, ICE and IRAK antagonist, wherein the antagonist is a CJ-14877, CP- 424174 (Pfizer), NF-61 (Negma-Lerads), and an analogue or derivative thereof.
14281. The method of item 14234 wherein the agent is an IL-2 inhibitor.
14282. The method of item 14234 wherein the agent is an immunosuppressant selected from the group consisting of teriflunomide (Sanofi Aventis), chlorsulfaquinoxalone (NSC-339004), chlorsulfaquinoxalone sulfate, CS-712 (Sankyo), ismomultin alfa (CAS No. 457913-93-8) (Akzo Nobel), antiallergics from GenPat77, antiinflammatories or AT-005 (Androclus Therapeutics), autoimmune disease therapeutics from EpiVax, BN-007 (Bone), budesonide (CAS No. 51333-22- 3) (MAP Pharmaceuticals), CO-14 (Genzyme), edratide (CAS No. 433922- 67-9) (Teva), EP-314 (Enanta), eprovafen (CAS No. 101335-99-3) (Sanofi- Aventis), HWA-131 (CAS No. 118788-41-3) (Sanofi-Aventis), immunomodulators from MerLion Pharmaceuticals, immunosuppressives from Alchemia, IPL-12 (Inflazyme), MDL-9563 (CAS No. 27086-86-8) (Sanofi-Aventis), Pharmaprojects No. 2330 (Sanofi-Aventis), Pharmaprojects No. 6426 (Abgenix), PXS-25 (Pharmaxis), rosmarinic acid (CAS No. 20283-92-5) (Sanofi-Aventis), RP 42927 or RP 54745 (CAS No. 135330-08-4) (Sanofi-Aventis), SGN-35 (Seattle Genetics), ST-1959 (Sigma-Tau), type I diabetes therapy from SYNX Pharma, UNIL-88 (Debiopharm), VP-025 (Vasogen), VR-694 (Vectura), PRTX-001 (Protalex), and an analogue or derivative thereof.
14283. The method of item 14234 wherein the agent is an IMPDH (inosine monophosphate).
14284. The method of item 14234 wherein the agent is an integrin antagonist.
14285. The method of item 14234 wherein the agent is an interleukin antagonist.
14286. The method of item 14234 wherein the agent is an inhibitor of type III receptor tyrosine kinase.
14287. The method of item 14234 wherein the agent is an irreversible inhibitor of enzyme methionine aminopeptidase type 2.
14288. The method of item 14234 wherein the agent is an isozyme selective delta protein kinase C inhibitor.
14289. The method of item 14234 wherein the agent a JAK3 enzyme inhibitor.
14290. The method of item 14234 wherein the agent is a JNK inhibitor.
14291. The method of item 14234 wherein the agent is a kinase inhibitor. 14292. The method of item 14234 wherein the agent is kinesin antagonist.
14293. The method of item 14234 wherein the agent is a kinesin antagonist.
14294. The method of item 14234 wherein the agent is a leukotriene inhibitor and antagonist selected from the group consisting of ambicromil (CAS No. 58805-38-2) (Sanofi-Aventis), amelubant (CAS No. 346735-24-8) (Boehringer Ingelheim), DW-1141 (Dong Wha), ebselen (Daiichi Pharmaceutical), ibudilast (Kyorin), leucotriene inhibitors from Sanofi-Aventis, lymphotoxin-beta receptor (LT-D) from Biogen Idee, Pharmaprojects No. 1535 and 2728 (CAS No. 119340-33-9) (Sanofi- Aventis), R-112 (Rigel), Rev-5367 (CAS No. 92532-05-3) (Sanofi-Aventis), RG-14893 (CAS No. 141835-49-6) (Sanofi-Aventis), RG-5901-A (CAS No. 101910-24-1), 92532-23-5, RP 66153 (CAS No. 142422-79-5), RP 66364 . (CAS No.186916-92-5), RP 69698 (CAS No. 141748-00-7) (Sanofi-Aventis), SC-411930 (Pfizer), SC-41930 (CAS No. 120072-59-5) (Pfizer), SC-50605 (CAS No. 138828-39-4) (Pfizer), SC-51146 (CAS No. 141059-52-1), SC- 53228 (CAS No. 153633-01-3) (Pfizer), spaglumic acid (ZY-15106) (CAS No.3106-85-2), 80619-64-3 (Novartis), tipredane (CAS No. 85197-77-9) (Bristol-Myers Squibb), U-75302 (CAS No. 119477-85-9) (Pfizer), and analogue or derivative thereof.
14295. The method of item 14234 wherein the agent is an MAP kinase inhibitor.
14296. The method of item 14234 wherein the agent is a matrix metalloproteinase inhibitor. 14297. The method of item 14234 wherein the agent is an MCP-CCR2 inhibitor.
14298. The method of item 14234 wherein the agent is an mTOR inhibitor.
14299. The method of item 14234 wherein the agent is an mTOR kinase inhibitor.
14300. The method of item 14234 wherein the agent is a microtubule inhibitor selected from the group consisting of antibody- maytansinoid conjugates from Biogen Idee, colchicines (MantiCore Pharmaceuticals), anticancer immunoconjugates from Johnson & Johnson, DIME from Octamer, gni-1f (GNI), huC242-DM4, huMy9-6-DM1 (ImmunoGen), IDN-5404 (Indena), IMO-098, IMOderm (Imotep), mebendazole (Introgen Therapeutics), microtubule poisons from Cambridge - Enterprise, paclitaxel such as LOTAX from Aphios (CAS No. 33069-62-4), Genexol-PM from Samyang, Pharmaprojects No. 6383 (Tapestry Pharmaceuticals), RPR-112378 (Sanofi-Aventis), SGN-75 (Seattle Genetics), SPL-7435 (Starpharma), SSR-250411 (Sanofi-Aventis), trastuzumab-DM1 (Genentech), vinorelbine, and an analogue or derivative thereof.
14301. The method of item 14234 wherein the agent is an MIF inhibitor.
14302. The method of item 14234 wherein the agent is an MMP inhibitor.
14303. The method of item 14234 wherein the agent is a neurokinin (NK) antagonist selected from the group consisting of anthrotainin (CAS No. 148084-40-6) (Sanofi-Aventis), an IBS thereapeutic from ArQuIe, MDL-105212A (CAS No. 167261-60-1) (Ssanofi-Aventis), Pharmaprojects No. 2744, 3258 (CAS No. 139167-47-8) 4006, 4201 , or 5986 (Sanofi-Aventis), RP 67580 (CAS No. 135911-02-3), SR-144190 (CAS No. 201152-86-5), SSR-240600, SSR-241586 (Sanofi-Aventis), TKA-457 (Novartis), vestipitant mesylate (CAS No. 334476-64-1) (GiaxoSmithKline), Win-64821 (Sanofi-Aventis), PRX-96026 (Predix Pharmaceuticals), and an analogue or derivative thereof.
14304. The method of item 14234 wherein the agent is an NF kappa B inhibitor selected from the group consisting of AVE-0545 or AVE-0547 (Sanofi-Aventis), bortezomib (CAS No. 179324-69-7) (Millennium Pharmaceuticals), dexanabinol (CAS No. 112924-45-5) (Pharmos), dexlipotam (Viatris), Pharmaprojects No. 6283 (INDRA) (OXiGENE), IPL- 576092 (CAS No. 137571-30-3) (Inflazyme), NFKB decoy (Corgentech), NFKB decoy oligo (AnGes MG), NFKB's from Ariad, osteoporosis _ treatments or S5 (F005) from Fulcrum Pharmaceuticals, P61 (Phytopharm), R-flurbiprofen (CAS No. 5104-49-4) (Encore Pharmaceuticals), and an analogue or derivative thereof.
14305. The method of item 14234 wherein the agent is a nitric oxide agonist.
14306. The method of item 14234 wherein the agent is an ornithine decarboxylase inhibitor.
14307. The method of item 14234 wherein the agent is a p38 MAP kinase inhibitor selected from the group consisting of AZD-6703 (AstraZeneca), JX-401 (Jexys Pharmaceuticals), BMS-2 (Bristol-Myers Squibb), a p38 MAP kinase inhibitor from Novartis, a p38-alpha MAP kinase inhibitor from Amphora, Pharmaprojects No. 5704 (Pharmacopeia), RPR- 200765A (Sanofi-Aventis), SD-282 (Johnson & Johnson), TAK-715 (Takeda), and an analogue or derivative thereof.
14308. The method of item 14234 wherein the agent is a palmitoyl-protein thioesterase inhibitor.
14309. The method of item 14234 wherein the agent is a PDGF receptor kinase inhibitor selected from the group consisting of AAL- 993, AMN-107, or ABP-309 (Novartis), AMG-706 (Amgen), BAY-57-9352 (Bayer), CDP-860 (UCB), E-7080 (Eisai), imatinib (CAS No. 152459-95-5) (Novartis), OSI-930 (OSI Pharmaceuticals), RPR-127963E (Sanofi-Aventis), RWJ-540973 (Johnson & Johnson), sorafenib tosylate (Bayer), SU-11657 (Pfizer), tandutinib (CAS No. 387867-13-2) (Millennium Pharmaceuticals), vatalanib (Novartis), ZK-CDK (Schering AG), and an analogue or derivative thereof.
14310. The method of item 14234 wherein the agent is (-)-halofenate (Metabolex), AMG-131 (Amgen), antidiabetics from Japan Tobacco, AZD-4619, AZD-8450, AZD-8677 (AstraZeneca), DRF-10945, balaglitazone (Dr Reddy's), CS-00088, CS-00098 (Chipscreen Biosciences), E-3030 (Eisai), etalocib (CAS No. 161172-51-6) (EIi Lilly), GSK-641597 (Ligand), GSK-677954 (GlaxoSmithKline), GW-409544 (Ligand), GW- 590735 (GlaxoSmithKline), K-111 (Hoffmann-La Roche), LY-518674 (EIi Lilly), LY-674 (Ligand), LY-929 (Ligand), MC-3001 , MC-3002 (MaxoCore Pharmaceuticals), metformin HCI + pioglitazone (CAS No. 1115-70-4 and
112529-15-4), ACTOPLUS MET from Andrx), muraglitazar (CAS No. 331741-94-7) (Bristol-Myers Squibb), naveglitazar (Ligand), oleoylethanolamide (Kadmus Pharmaceuticals), ONO-5129, pioglitazone hydrochloride (CAS No. 111025-46-8 and 112529-15-4) (Takeda), PLX-204 (Plexxikon), PPAR agonists from Genfit, PPAR delta agonists from EIi Lilly, PPAR-alpha agonists from CrystalGenomics, PPAR-gamma modulators and PPAR-β modulators from CareX, rosigϋtazone maleate (CAS No. 122324- 73-4 or 155141-29-0) (GlaxoSmithKline), rosiglitazone maleate/glimepir (CAS No. 155141-29-0 and 93479-97-1), AVANDARYL, rosiglitazone maleate/metformin extend (CAS No. 155141-29-0 and 657-24-9), AVANDAMET, rosiglitazone maleate+metformin, AVANDAMET (GlaxoSmithKline), tesaglitazar (AstraZeneca), and an analogue or derivative thereof.
14311. The method of item 14234 wherein the agent is a phosphatase inhibitor.
14312. The method of item 14234 wherein the agent is a phosphodiesterase (PDE) inhibitor selected from the group consisting of avanafil (Tanabe Seiyaku), dasantafil (CAS No. 569351-91-3) (Schering- Plough), A-906119 (CAS No. 134072-58-5), DL-850 (Sanofi-Aventis), GRC- 3015, GRC-3566, GRC-3886 (Glenmark), HWA-153 (CAS No. 56395-66-5) (Sanofi-Aventis), hydroxypumafentrine (Altana), IBFB-130011 , IBFB-14-016, IBFB-140301 , IBFB-150007, IBFB-211913 (IBFB Pharma), L-826141 (Merck & Co), medorinone (CAS No. 88296-61-1) (Sanofi-Aventis), MEM- 1917 (Memory Pharmaceuticals), ND-1251 (Neuro3d), PDE inhibitors from ICOS, PDE IV inhibitors from Memory Pharmaceuticals and CrystalGenomics, Pharmaprojects No. 2742 and 6141 (Sanofi-Aventis), QAD-171 (Novartis), RHC-2963 (CAS No. 76993-12-9 and 76993-14-1), RPR-117658, RPR-122818 derivatives, SR-24870 , and RPR-132294 (Sanofi-Aventis), SK-350 (ln2Gen), stroke therapy agents from deCODE Genetics, TAS-203 (Taiho), tofimilast (CAS No. 185954-27-2) (Pfizer), UK- 371800 (Pfizer), WIN-65579 (CAS No. 158020-82-7) (Sanofi-Aventis), IBFB- 130020 (IBFB Pharma), OPC-6535 (CAS No. 145739-56-6) (Otsuka), a phosphodiesterase III inhibitor, enoximone, a phosphodiesterase IV inhibitor, fosfosai, Atopik (Barrier Therapeutics), triflusal, a phosphodiesterase V inhibitor, and an analogue or derivative thereof. 14313. The method of item 14234 wherein the agent is a PKC inhibitor.
14314. The method of item 14234 wherein the agent is a platelet activating factor antagonist.
14315. The method of item 14234 wherein the agent is a platelet-derived growth factor receptor kinase inhibitor.
14316. The method of item 14234 wherein the agent is a prolyl hydroxylase inhibitor.
14317. The method of item 14234 wherein the agent is a polymorphonuclear neutrophil inhibitor.
14318. The method of item 14234 wherein the agent is a protein kinase B inhibitor.
14319. The method of item 14234 wherein the agent is a protein kinase C stimulant.
14320. The method of item 14234 wherein the agent is a purine nucleoside analogue.
14321. The method of item 14234 wherein the agent is a purinoreceptor P2X antagonist.
14322. The method of item 14234 wherein the agent is a Raf kinase inhibitor. 14323. The method of item 14234 wherein the agent is a reversible inhibitor of ErbB1 and ErbB2.
14324. The method of item 14234 wherein the agent is a ribonucleoside triphosphate reductase inhibitor.
14325. The method of item 14234 wherein the agent is an SDF-1 antagonist.
14326. The method of item 14234 wherein the agent is a sheddase inhibitor.
14327. The method of item 14234 wherein the agent is an SRC inhibitor.
14328. The method of item 14234 wherein the agent is a stromelysin inhibitor.
14329. The method of item 14234 wherein the agent is an Syk kinase inhibitor.
14330. The method of item 14234 wherein the agent is a telomerase inhibitor.
14331. The method of item 14234 wherein the agent is a TGF beta inhibitor selected from the group consisting of pirfenidone (CAS No. 53179-13-8) (MARNAC)1 tranilast (CAS No. 53902-12-8) (Kissei), IN- 1130 (ln2Gen), mannose-6-phosphate (BTG), TGF-β antagonists from Inflazyme (Pharmaprojects No. 6075), TGF-β antagonists from Sydney, non-industrial source), TGF-β I receptor kinase inhibitors from EIi Lilly, TGF- β receptor inhibitors from Johnson & Johnson, and an analogue or derivative thereof.
14332. The method of item 14234 wherein the agent is a
TNFα antagonist or TACE inhibitor selected from the group consisting of adalimumab (CAS No. 331731-18-1) (Cambridge Antibody Technology), AGIX-4207 (AtheroGenics), AGT-1 (Advanced Biotherapy), an antiinflammatory from Borean Pharma, Cellzome, or Paradigm Therapeutics, anti-inflammatory vaccine (TNF-alpha kinoid) from Neovacs, humanized anti-TNF antibody or an anti-TNF MAb (CB0006) Celltech (UCB), apratastat (CAS No. 287445-51-0) (Wyeth), BMS-561392 (Bristol-Myers Squibb), BN- 006 (Bone), certolizumab pegol (CAS No. 428863-50-7 or CH-138 (UCB), cilomilast (CAS No. 153259-65-5) (GlaxoSmithKline), CR-1 (Nuada Pharmaceuticals), CRx-119 (CombinatoRx), D-5410 (UCB), dacopafant (CAS No. 125372-33-0) (Sanofi-Aventis), dersalazine (CAS No. 188913-57- 7/188913-58-8) (Uriach), etanercept (CAS No. 185243-69-0) (Amgen), ethyl pyruvate (Critical (Critical Therapeutics), golimumab (CAS No. 476181-74-5) (Johnson & Johnson), hormono-immunotherapy from Ipsen, CDP571 (e.g., Humicade from UCB), IC-485 (ICOS), infliximab (CAS No. 170277-31-3) (Johnson & Johnson), IP-751 (Manhattan Pharmaceuticals), ISIS-104838 (CAS No. 250755-32-9) (ISIS Pharmaceuticals), lenalidomide (CAS No. 191732-72-6) (Celgene), lentinan (CAS No. 37339-90-5) (Ajinomoto), MDL- 201112 (CAS No. 142130-73-2) (Sanofi-Aventis), medroxyprogesterone (CAS No. 520-85-4) (InKine Pharmaceutical), N-acetylcysteine (CAS No. 616-91-1) (Zambon), NBE-P2 (DIREVO Biotech), nerelimomab (CAS No. 162774-06-3) (Chiron), OM-294DP (OM PHARMA), onercept (CAS No. 199685-57-9) (Yeda), PASSTNF-alpha (Verigen), pentoxifylline. or oxypentifylline (Sanofi-Aventis), Pharmaprojects No. 4091 , 4241, 4295, or 4488 (Sanofi-Aventis), Pharmaprojects No. 5480 (Amgen), Pharmaprojects No. 6749 (Cengent), pirfenidone (CAS No. 53179-13-8) (MARNAC), RPR- 132294 (Sanofi-Aventis), S5 (F002) (Fulcrum Pharmaceuticals), simvastatin (CAS No. 79902-63-9) (Merck & Co), STA-6292 (Synta Pharmaceuticals), tacrolimus (CAS No. 104987-11-3) (Fujisawa LifeCycle Pharma), talactoferrin alfa (CAS No. 308240-58-6) (Agennix), thalidomide (CAS No. 50-35-1) (Celgene), TNF antagonists form ProStrakan, and Synergen, TNF inhibitors (Amgen), TNF-alpha antagonists from Dynavax Technologies and Jerina AG (Germany), TNF-alpha inhibitors from IBFB Pharma and Xencor (Xencor), torbafylline (CAS No. 105102-21-4) (Sanofi-Aventis), UR-1505 (Uriach), VT-346 (Viron Therapeutics), YSIL6 (Y's Therapeutics), YSTH2 (Y's Therapeutics), NPI-1302a-3 (Nereus Pharmaceuticals, a TNF antagonist from Jerina AG (Germany), dersalazine, and an analogue or derivative thereof.
14333. The method of item 14234 wherein the agent is a Toll receptor inhibitor.
14334. The method of item 14234 wherein the agent is a tubulin antagonist.
14335. The method of item 14234 wherein the agent is a tyrosine kinase inhibitor selected from the group consisting of SU-011248, SUTENT from Pfizer Inc. (New York, NY), BMS-354825, PN-355 (Paracelsian Pharmaceuticals), AGN-199659 (Allergan), AAL-993 or ABP- 309 (Novartis), adaphostin (NIH), AEE-788 (Novartis), AG-013736 (OSI Pharmaceuticals), AG-13736 (Pfizer), ALT-110 (Alteris Therapeutics), AMG- 706 (Amgen), anticancer MAbs from Xencor, anti-EGFrvlll MAbs from Abgenix, anti-HER2 MAb from Abiogen, AZD-2171 or AZD-9935 (AstraZeneca), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), CEP-5214 (Cephalon), CEP-7055 (Cephalon), cetuximab (ImClone Systems), CHIR-200131 and CHIR-258 (Chiron), CP-547632 (OSI Pharmaceuticals), CP-724714 (Pfizer), CT-301 (Creabilis Therapeutics), D- 69491 (Baxter International), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EGFR/ErbB2 inhibitors from Array BioPharma, erlotinib (CAS No. 183319- 69-9) (OSI Pharmaceuticals), EXEL-2880 (Exelixis), FK-778 (Sanofi- Aventis) , gefitinib (CAS No. 184475-35-2) (AstraZeneca), GW-2286 or GW- 654652 (GlaxoSmithKline), her2/neu antigen from AlphaVax, HER-2/neu inhibitor from Generex, Herzyme (Medipad) (Sirna Therapeutics), HKI-272 (Wyeth), HuMax-EGFr (Genmab), idronoxil (CAS No. 81267-65-4) (Novogen), IGF-1 inhibitors from Ontogen, IMC-11 F8 (ImCIone Systems), kahalalide F (CAS No. 149204-42-2) (PharmaMar), KDR inhibitor from LG Life Sciences, KDR inhibitors from Abbott Laboratories, KDR kinase inhibitors (UCB), Kdr kinase inhibitors from Merck & Co, KRN-633 and KRN- 951 (Kirin Brewery), KSB-102 (Xenova), lapatinib ditosylate (CAS No. 388082-78-8) (GlaxoSmithKline), matuzumab (Merck KGaA), MDX-214 (Medarex), ME-103 (Pharmexa), MED-A300 (Gerolymatos), MNAC-13 (Lay Line Genomics), nimotuzumab (Center of Molecular Immunology), NSC- 330507 or NSC-707545 (NIH), NV-50 (Novogen), OSI-930 (OSI Pharmaceuticals), panitumumab (Abgenix), pelitinib (CAS No. 287933-82-7) (Wyeth), pertuzumab (CAS No. 380610-27-5) (Genentech), Pharmaprojects No. 3985 (Sanofi-Aventis), prostate cancer therapeutics from Sequenom (SQPC35, SQPC36, SQPC90), removab and remoxab (Trion Pharma), RG- 13022 (CAS No. 136831-48-6), RG-13291 (CAS No. 138989-50-1), or RG- 14620 (CAS No. 136831-49-7) (Sanofi-Aventis), RM-6427 (Romark), RNAi breast cancer therapy from Benitec, RP 53801 (CAS No. 125882-88-4) (Sanofi-Aventis), sorafenib tosylate (Bayer), SU-11657 (Pfizer), Tie-2 antagonists from Semaia (Hybrigenics), Tie-2 inhibitors from Ontogen, trastuzumab (CAS No. 180288-69-1) (Genentech), tyrosine kinase inhibitors from Sanofi-Aventis, U3-1287, U3-1565, U3-1784, U3-1800 (U3 Pharma), vatalanib (Novartis), VEGFR-2 kinase inhibitor from Bristol-Myers Squibb, XL-647 (Exelixis), ZD-6474 (AstraZeneca), ZK-CDK (Schering AG), an EGFR tyrosine kinase inhibitor, EKB-569 (Wyeth), and an analogue or derivative thereof. 14336. The method of item 14234 wherein the agent is a VEGF inhibitor.
14337. The method of item 14234 wherein the agent is a vitamin D receptor agonist.
14338. The method of item 14234 wherein the agent is ZD-6474 (an angiogenesis inhibitor).
14339. The method of item 14234 wherein the agent is AP-23573 (an mTOR inhibitor).
14340. The method of item 14234 wherein the agent is synthadotin (a tubulin antagonist).
14341. The method of item 14234 wherein the agenϋs S-0885 (a collagenase inhibitor).
14342. The method of item 14234 wherein the agent is aplidine (an elongation factor-1 alpha inhibitor).
14343. The method of item 14234 wherein the agent is ixabepilone (an epithilone).
14344. The method of item 14234 wherein the agent is IDN-5390 (an angiogenesis inhibitor).
14345. The method of item 14234 wherein the agent is SB-2723005 (an angiogenesis inhibitor). 14346. The method of item 14234 wherein the agent is ABT-518 (an angiogenesis inhibitor).
14347. The method of item 14234 wherein the agent is combretastatin (an angiogenesis inhibitor).
14348. The method of item 14234 wherein the agent is anecortave acetate (an angiogenesis inhibitor).
14349. The method of item 14234 wherein the agent is SB-715992 (a kinesin antagonist).
14350. The method of item 14234 wherein the agent is temsirolimus (an mTOR inhibitor).
14351. _ The method of item 14234 wherein the agent is adalimumab (a TNFα antagonist).
14352. The method of item 14234, wherein the composition comprises a polymer.
14353. The method of item 14234, wherein the composition comprises a polymeric carrier.
14354. The method of item 14234 wherein the anti- scarring agent inhibits adhesion between the device and a host into which the device is implanted.
14355. The method of item 14234 wherein the device delivers the anti-scarring agent locally to tissue proximate to the device. 14356. The method of item 14234 wherein the device has a coating that comprises the anti-scarring agent.
14357. The method of item 14234, wherein the device has a coating that comprises the agent and is disposed on a surface of the implant.
14358. The method of item 14234, wherein the device has a coating that comprises the agent and directly contacts the implant.
14359. The method of item 14234, wherein the device has a coating that comprises the agent and indirectly contacts the implant.
14360. The method of item 14234, wherein the device has a coating that comprises the agent and partially covers the implant.
14361. The method of item 14234, wherein the device has a coating that comprises the agent and completely covers the implant.
14362. The method of item 14234, wherein the device has a uniform coating.
14363. The method of item 14234, wherein the device has a non-uniform coating.
14364. The method of item 14234, wherein the device has a discontinuous coating.
14365. The method of item 14234, wherein the device has a patterned coating. 14366. The method of item 14234, wherein the device has a coating with a thickness of 100 Dm or less.
14367. The method of item 14234, wherein the device has a coating with a thickness of 10 Dm or less.
14368. The method of item 14234, wherein the device has a coating, and the coating adheres to the surface of the implant upon deployment of the implant.
14369. The method of item 14234, wherein the device has a coating, and wherein the coating is stable at room temperature for a period of 1 year.
14370. The method of item 14234, wherein the device has a coating, and wherein the anti-scarring agent is present in the coating in an amount ranging between about 0.0001% to about 1% by weight.
14371. The method of item 14234, wherein the device has a coating, and wherein the anti-scarring agent is present in the coating in an amount ranging between about 1% to about 10% by weight.
14372. The method of item 14234, wherein the device has a coating, and wherein the anti-scarring agent is present in the coating in an amount ranging between about 10% to about 25% by weight.
14373. The method of item 14234, wherein the device has a coating, and wherein the anti-scarring agent is present in the coating in an amount ranging between about 25% to about 70% by weight. 14374. The method of item 14234, wherein the device has a coating, and wherein the coating further comprises a polymer.
14375. The method of item 14234, wherein the device has a first coating having a first composition and a second coating having a second composition.
14376. The method of item 14234, wherein the device has a first coating having a first composition and a second coating having a second composition, wherein the first composition and the second composition are different.
14377. The method of item 14234, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a copolymer.
14378. The method of item 14234, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a block copolymer.
14379. The method of item 14234, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a random copolymer.
14380. The method of item 14234, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a biodegradable polymer.
14381. The method of item 14234, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a non-biodegradable polymer. 14382. The method of item 14234, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a hydrophilic polymer.
14383. The method of item 14234, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a hydrophobic polymer.
14384. The method of item 14234, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a polymer having hydrophilic domains.
14385. The method of item 14234, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a polymer having hydrophobic domains.
14386. The method of item 14234, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a non-conductive polymer.
14387. The method of item 14234, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises an elastomer.
14388. The method of item 14234, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a hydrogel.
14389. The method of item 14234, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a silicone polymer. 14390. The method of item 14234, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a hydrocarbon polymer.
14391. The method of item 14234, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a styrene-derived polymer.
14392. The method of item 14234, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a butadiene polymer.
14393. The method of item 14234, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a macromer.
14394. The method of item 14234, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a poly(ethylene glycol) polymer.
14395. The method of item 14234 wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises an amorphous polymer.
14396. The method of item 14234, wherein the device comprises a lubricious coating.
14397. The method of item 14234 wherein the anti- scarring agent is located within pores or holes of the device. 14398. The method of item 14234 wherein the anti- scarring agent is located within a channel, lumen, or divet of the device.
14399. The method of item 14234, wherein the device comprises a second pharmaceutically active agent.
14400. The method of item 14234 wherein the device comprises an anti-inflammatory agent.
14401. The method of item 14234 wherein the device comprises an agent that inhibits infection.
14402. The method of item 14234 wherein the device comprises an agent that inhibits infection, and wherein the agent is an anthracycline.
14403. The method of item 14234 wherein the device comprises an agent that inhibits infection, and wherein the agent is doxorubicin.
14404. The method of item 14234 wherein the device comprises an agent that inhibits infection, and wherein the agent is mitoxantrone.
14405. The method of item 14234 wherein the device comprises an agent that inhibits infection, and wherein the agent is a fluoropyrimidine.
14406. The method of item 14234 wherein the device comprises an agent that inhibits infection, and wherein the agent is 5- fluorouracil (5-FU). 14407. The method of item 14234 wherein the device comprises an agent that inhibits infection, and wherein the agent is a folic acid antagonist.
14408. The method of item 14234 wherein the device comprises an agent that inhibits infection, and wherein the agent is methotrexate.
14409. The method of item 14234 wherein the device comprises an agent that inhibits infection, and wherein the agent is a podophylotoxin.
14410. The method of item 14234 wherein the device comprises an agent that inhibits infection, and wherein the agent is etoposide.
14411. The method of item 14234 wherein the device comprises an agent that inhibits infection, and wherein the agent is a camptothecin.
14412. The method of item 14234 wherein the device comprises an agent that inhibits infection, and wherein the agent is a hydroxyurea.
14413. The method of item 14234 wherein the device comprises an agent that inhibits infection, and wherein the agent is a platinum complex.
14414. The method of item 14234 wherein the device comprises an agent that inhibits infection, and wherein the agent is cisplatin. 14415. The method of item 14234, further comprising an anti-thrombotic agent.
14416. The method of item 14234 wherein the device comprises a visualization agent.
14417. The method of item 14234 wherein the device comprises a visualization agent, wherein the visualization agent is a radiopaque material, and wherein the radiopaque material comprises a metal, a halogenated compound, or a barium containing compound.
14418. The method of item 14234 wherein the device comprises a visualization agent, wherein the visualization agent is a radiopaque material, and wherein the radiopaque material comprises barium, tantalum, or technetium.
14419. The method of item 14234 wherein the device comprises a visualization agent, and wherein the visualization agent is a MRI responsive material.
14420. The method of item 14234 wherein the device comprises a visualization agent, and wherein the visualization agent comprises a gadolinium chelate.
14421. The method of item 14234 wherein the device comprises a visualization agent, and wherein the visualization agent comprises iron, magnesium, manganese, copper, or chromium.
14422. The method of item 14234 wherein the device comprises a visualization agent, and wherein the visualization agent comprises an iron oxide compound. 14423. The method of item 14234 wherein the device comprises a visualization agent, and wherein the visualization agent comprises a dye, pigment, or colorant.
14424. The method of item 14234 wherein the device comprises an echogenic material.
14425. The method of item 14234 wherein the device comprises an echogenic material, and wherein the echogenic material is in the form of a coating.
14426. The method of item 14234 wherein the device is sterile.
14427. The method of item 14234 wherein the anti- scarring agent is released into tissue in the vicinity of the device after deployment of the device.
14428. The method of item 14234 wherein the anti- scarring agent is released into tissue in the vicinity of the device after deployment of the device, and wherein the tissue is connective tissue.
14429. The method of item 14234 wherein the anti- scarring agent is released into tissue in the vicinity of the device after deployment of the device, and wherein the tissue is muscle tissue.
14430. The method of item 14234 wherein the anti- scarring agent is released into tissue in the vicinity of the device after deployment of the device, and wherein the tissue is nerve tissue. 14431. The method of item 14234 wherein the anti- scarring agent is released into tissue in the vicinity of the device after deployment of the device, and wherein the tissue is epithelium tissue.
14432. The method of item 14234 wherein the anti- scarring agent is released in effective concentrations from the device over a period ranging from the time of deployment of the device to about 1 year.
14433. The method of item 14234 wherein the anti- scarring agent is released in effective concentrations from the device over a period ranging from about 1 month to 6 months.
14434. The method of item 14234 wherein the anti- scarring agent is released in effective concentrations from the device over a period ranging from about 1 - 90 days.
14435. The method of item 14234 wherein the anti- scarring agent is released in effective concentrations from the device at a constant rate.
14436. The method of item 14234 wherein the anti- scarring agent is released in effective concentrations from the device at an increasing rate.
14437. The method of item 14234 wherein the anti- scarring agent is released in effective concentrations from the device at a decreasing rate.
14438. The method of item 14234 wherein the anti- scarring agent is released in effective concentrations from the composition comprising the anti-scarring agent by diffusion over a period ranging from the time of deployment of the device to about 90 days.
14439. The method of item 14234 wherein the anti- scarring agent is released in effective concentrations from the composition comprising the anti-scarring agent by erosion of the composition over a period ranging from the time of deployment of the device to about 90 days.
14440. The method of item 14234 wherein the device comprises about 0.01 Dg to about 10 Dg of the anti-scarring agent.
14441. The method of item 14234 wherein the device comprises about 10 Dg to about 10 mg of the anti-scarring agent.
14442. The method of item 14234 wherein the device comprises about 10 mg to about 250 mg of the anti-scarring agent. . . .
14443. The method of item 14234 wherein the device comprises about 250 mg to about 1000 mg of the anti-scarring agent.
14444. The method of item 14234 wherein the device comprises about 1000 mg to about 2500 mg of the anti-scarring agent.
14445. The method of item 14234 wherein a surface of the device comprises less than 0.01 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
14446. The method of item 14234 wherein a surface of the device comprises about 0.01 Dg to about 1 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied. 14447. The method of item 14234 wherein a surface of the device comprises about 1 Dg to about 10 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
14448. The method of item 14234 wherein a surface of the device comprises about 10 Dg to about 250 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
14449. The method of item 14234 wherein a surface of the device comprises about 250 Dg to about 1000 Dg of the anti-scarring agent of anti-scarring agent per mm2 of device surface to which the anti- scarring agent is applied.
14450. The method of item 14234 wherein a surface of the device comprises about 1000 Dg to about 2500 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
14451. The method of item 14234 wherein the combining is performed by direct affixing the agent or the composition to the implant.
14452. The method of item 14234 wherein the combining is performed by spraying the agent or the component onto the implant.
14453. The method of item 14234 wherein the combining is performed by electrospraying the agent or the composition onto the implant. -14454. The method of item 14234 wherein the combining is performed by dipping the implant into a solution comprising the agent or the composition.
14455. The method of item 14234 wherein the combining is performed by covalently attaching the agent or the composition to the implant.
14456. The method of item 14234 wherein the combining is performed by non-covalently attaching the agent or the composition to the implant.
14457. The method of item 14234 wherein the combining is performed by coating the implant with a substance that contains the agent or the composition.
14458. The method of item 14234 wherein the combining is performed by coating the implant with a substance that absorbs the agent.
14459. The method of item 14234 wherein the combining is performed by interweaving a thread composed of, or coated with, the agent or the composition.
14460. The method of item 14234 wherein the combining is performed by covering all the implant with a sleeve that contains the agent or the composition.
14461. The method of item 14234 wherein the combining is performed by covering a portion of the implant with a sleeve that contains the agent or the composition. ' 14462. The method of item 14234 wherein the combining is performed by covering all the implant with a cover that contains the agent or the composition.
14463. The method of item 14234 wherein the combining is performed by covering a portion of the implant with a cover that contains the agent or the composition.
14464. The method of item 14234 wherein the combining is performed by covering all the implant with an electrospun fabric that contains the agent or the composition.
14465. The method of item 14234 wherein the combining is performed by covering a portion of the implant with an electrospun fabric that contains the agent or the composition.
14466. The method of item 14234 wherein the combining is performed by covering all the implant with a mesh that contains the agent or the composition.
14467. The method of item 14234 wherein the combining is performed by covering a portion of the implant with a mesh that contains the agent or the composition.
14468. The method of item 14234 wherein the combining is performed by constructing all the implant with the agent or the composition.
14469. The method of item 14234 wherein the combining is performed by constructing a portion of the implant with the agent or the composition. 14470. The method of item 14234 wherein the combining is performed by impregnating the implant with the agent or the composition.
14471. The method of item 14234 wherein the combining is performed by constructing all of the implant from a degradable polymer that releases the agent.
14472. The method of item 14234 wherein the combining is performed by constructing a portion of the implant from a degradable polymer that releases the agent.
14473. The method of item 14234 wherein the combining is performed by dipping the implant into a solution that comprise the agent and an inert solvent for the implant.
14474. The method of item 14234 wherein the combining is performed by dipping the implant into a solution that comprises the agent and a solvent that will swell the implant.
14475. The method of item 14234 wherein the combining is performed by dipping the implant into a solution that comprises the agent and a solvent that will dissolve the implant.
14476. The method of item 14234 wherein the combining is performed by dipping the implant into a solution that comprises the agent, a polymer and an inert solvent for the implant.
14477. The method of item 14234 wherein the combining is performed by dipping the implant into a solution that comprises the agent, a polymer and a solvent that will swell the implant. 14478. The method of item 14234 wherein the combining is performed by dipping the implant into a solution that comprises the agent, a polymer and a solvent that will dissolve the implant.
14479. The method of item 14234 wherein the combining is performed by spraying the implant into a solution that comprises the agent and an inert solvent for the implant.
14480. The method of item 14234 wherein the combining is performed by spraying the implant into a solution that comprises the agent and a solvent that will swell the implant.
14481. The method of item 14234 wherein the combining is performed by spraying the implant into a solution that comprises the agent and a solvent that will dissolve the implant.
14482. The method of item 14234 wherein the combining is performed by spraying the implant into a solution that comprises the agent, a polymer and an inert solvent for the implant.
14483. The method of item 14234 wherein the combining is performed by spraying the implant into a solution that comprises the agent, a polymer and a solvent that will swell the implant.
14484. The method of item 14234 wherein the combining is performed by spraying the implant into a solution that comprises the agent, a polymer and a solvent that will dissolve the implant.
14485. A method as in any one of items 14234-14484, wherein the device is a glaucoma drainage device comprising a plate and a tube. 14486. A method of making a medical device comprising: combining a prosthetic heart valve or component thereof (e.g., an implant) and an anti-scarring agent or a composition comprising an anti- scarring agent, wherein the agent inhibits scarring between the device and a host into which the device is implanted.
14487. The method of item 14486 wherein the agent is an adensosine A2A receptor antagonist.
14488. The method of item 14486 wherein the agent is an AKT inhibitor.
14489. The method of item 14486 wherein the agent is an alpha 2 integrin antagonist, wherein the alpha 2 integrin antagonist is Pharmaprojects No. 5754 (Merck KgaA).
14490. The method of item 14486 wherein the agent is an alpha 4 integrin antagonist.
14491. The method of item 14486 wherein the agent is an alpha 7 nicotinic receptor agonist.
14492. The method of item 14486 wherein the agent is an angiogenesis inhibitor selected from the group consisting of AG-12,958 (Pfizer), ATN-161 (Attention LLC), neovastat, an angiogenesis inhibitor from Jerina AG (Germany), NM-3 (Mercian), VGA-1155 (Taisho), FCE-26644 (Pfizer), FCE-26950 (Pfizer), FPMA (Meiji Daries), FR-111142 (Fujisawa), GGTI-298, GM-1306 (Ligand), GPA-1734 (Novartis), NNC-47-0011 (Novo Nordisk), herbamycin (Nippon Kayaku), lenalidomide (Celegene), IP-10 (NIH), ABT-828 (Abbott), KIN-841 (Tokushima University, Japan), SF-1126 (Semafore Pharmaceuticals), laminin technology (NIH), CHIR-258 (Chiron), NVP-AEW541 (Novartis), NVP-AEW541 (Novartis), Vt16907 (Alchemia), OXI-8007 (Oxigene), EG-3306 (Ark Therapeutics), Maspin (Arriva), ABT- 567 (Abbott), PPI-2458 (Praecis Pharmaceuticals), CC-5079, CC-4089 (Celgene), HIF-1 alpha inhibitors (Xenova), S-247 (Pfizer), AP-23573 (Ariad), AZD-9935 (Astra Zeneca), mebendazole (Introgen Therapeutics), MetAP-2 inhibitors (GlaxoSmithKline), AG-615 (Angiogene Pharmaceuticals), Tie-2 antagonists (Hybrigenics), NC-381 , CYC-381 , NC-169, NC-219, NC-383, NC-384, NC-407 (Lorus Therapeutics), ATN-224 (Attenuon), ON-01370 (Onconova), Vitronectin antagonists (Amgen), SDX-103 (Salmedix), Vitronectin antagonists (Shire), CHP (Riemser), TEK (Amgen), Anecortave acetate (Alcon), T46.2 (Matrix Therapeutics), HG-2 (Heptagen), TEM antagonists (Genzyme), Oxi-4500 (Oxigene), ATN-161 (Attenuon), WX-293 (Wilex), M-2025 (Metris Therapeutics), Alphastatin (BioActa), YH-16 (Yantai Rongchang), BIBF-1120 (Boehringer Ingelheim), BAY-57-9352 (Bayer), AS- 1404 (Cancer Research Technology), SC-77964 (Pfizer), glycomimetics (BioTie Therapies), TIE-2 Inhibitors (Ontogen), DIMI, Octamer (Octamer), ABR-215050 (Active Biotech), ABT-518 (Abbott), KDR inhibitors (Abbott), BSF-466895 (Abbott), SCH-221153 (Schering-Plough), DAC:antiangiogenic (ConjuChem), TFPI (EntreMed), AZD-2171 (Astra-Zenaca), CDC-394 (Celgene), LY290293 (EIi Lilly), IDN-5390 (Indena), Kdr Kinase Inhibitors (Merck), CT-113020, CT-116433, CT-116563, CT-31890, CT-32228) (Cell Therapeutics), A-299620 (Abbott), TWEAK Inhibitor (Amgen), VEGF modulators (Johnson and Johnson), Tum-N53, tumstatin (Genzyme), Thios- 1 , Thios-2 (Thios Pharmaceuticals), MV-6401 (Miravant Medical Technologies), Spisulosine (PharmaMar), CEP-7055 (Cephalon), AUV-201 (Auvation), LM-609 (EIi Lilly), SKF-106615 (AnorMED), Oglufanide disodium (Cytran), BW-114 (Pharminox), Calreticulin (NIH), WX-678 (Wilex), SD-7784 (Pfizer), WX-UK1 (Wilex), SH-268 (Schering AG), 2-Me-PGA (Celgene), S- 137 (Pfizer), ZD-6126 (Angiogene Pharmaceuticals), SG-292 (SignalGen), Benefin (Lane Labs), A6, A36 (Angstrom), SB-2723005 (GlaxoSmithKline), SC-7 (Cell Therapeutics), ZEN-014 (AEterna Zentaris), 2-methoxyestradiol (EntreMed), NK-130119 (Nippon Kayaku), CC-10004 (Celgene), AVE- 8062A (Ajinomoto), Tacedinaline (Pfizer), Actinonin (Tokyo Metropolitan Institute of Medical Science), Lenalidomide (Celgene), VGA-1155, BTO-956 (SRI International), ER-68203-00 (Eisai), CT-6685 (UCB), JKC-362 (Phoenix Pharmaceuticals), DMI-3798 (DMI Biosciences, Angiomate (Ipsen), ZD-6474 (AstraZeneca), CEP-5214 (Cephalon), Canstatin (Genzyme), NM-3 (Mercian), Oridigm (MediQuest Therapeutics), Exherin (Adherex), BLS-0597 (Boston Life Sciences), PTC-299 (PTC Therapeutics), NPI-2358 (Nereus Pharmaceuticals), CGP-79787 (Novartis), AEE-788 (Novartis), CKD-732 (Chong Kun Dang), CP-564959 (OSI Pharmaceuticals), CM-101 (CarboMed), CT-2584, CT3501 (Cell Therapeutics), combretastatin and analogues and derivatives thereof (Oxigene), Rebimastat (Bristol-Meyers Squibb), Dextrin 2-sulfate (ML Laboratories), Cilengitide (Merk KGaA), NSC-706704 (Pharminox), KRN- 951 (Kirin Brewery), Ukrain, NSC-631570 (Nowicky Pharma), Tecogalan sodium (Daiichi Pharmaceutical), Tz-93 (Tsumura), TBC-1635 (Encysive Pharmaceuticals), TAN-1120 (Takeda), Semaxanib (Pfizer), BDI-7800 (Biopharmacopae), SD-186, SD-983 (Bristol-Meyers Squibb), SB-223245 (GlaxoSmithKline), SC-236 (Pfizer), RWJ-590973 (Johnson and Johnson), ILX-1850 (Genzyme), SC-68488, S-836 (Pfizer), CG-55069-11 (CuraGen), Ki-23057 (Kirin Brewery), CCX-700 (Chemoentryx), Pegaptanib octasodium (Giled Sciences), ANGIOCOL (available from Biostratum Inc.), or an analogue or derivative thereof.
14493. The method of item 14486 wherein the agent is an apoptosis antagonist.
14494. The method of item 14486 wherein the agent is an apoptosis activator. 14495. The method of item 14486 wherein the agent is a beta 1 integrin antagonist.
14496. The method of item 14486 wherein the agent is a beta tubulin inhibitor.
14497. The method of item 14486 wherein the agent is a blocker of enzyme production in Hepatitis C.
14498. The method of item 14486 wherein the agent is a Bruton's tyrosine kinase inhibitor.
14499. The method of item 14486 wherein the agent is a calcineurin inhibitor.
14500. _ The method of item 14486 wherein the agent is a caspase 3 inhibitor.
14501. The method of item 14486 wherein the agent is a CC chemokine receptor antagonist.
14502. The method of item 14486 wherein the agent is a cell cycle inhibitor selected from the group consisting of SNS-595 (Sunesis), synthadotin, KRX-0403, and an analogue or derivative thereof.
14503. The method of item 14486 wherein the agent is a cathepsin B inhibitor.
14504. The method of item 14486 wherein the agent is a cathepsin K inhibitor, wherein the cathepsin K inhibitor is 462795 (GlaxoSmithKline), INPL-022-D6 (Amura Therapeutics), or an analogue or derivative thereof.
14505. The method of item 14486 wherein the agent is a cathepsin L inhibitor.
14506. The method of item 14486 wherein the agent is a CD40 antagonist.
14507. The method of item 14486 wherein the agent is a chemokine receptor agonist.
14508. The method of item 14486 wherein the agent is a chymase inhibitor.
14509. The method of item 14486 wherein the agent is a collagenase antagonist.
14510. The method of item 14486 wherein the agent is a CXCR antagonist.
14511. The method of item 14486 wherein the agent is a cyclin dependent kinase inhibitor selected from the group consisting of a CDK-1 inhibitor, a CDK-2 inhibitor, a CDK- 4 inhibitor, a CDK-6 inhibitor, a CAK1 inhibitor from GPC Biotech and Bristol-Myers Squibb, RGB-286199 (GPC Biotech), an anticancer agent from Astex Technology, a CAK1 inhibitor from GPC Biotech, a CDK inhibitor from Sanofi-Aventis, a CDK1/CDK2 inhibitor from Amgen, a CDK2 inhibitor from SUGEN-2 (Pfizer), a hearing loss therapy agent (Sound Pharmaceuticals), PD-0332991 (Pfizer), RGB-286199 (GPC Biotech), Ro-0505124 (Hoffmann-La Roche), a Ser/Thr kinase inhibitor from Lilly (EIi Lilly), CVT-2584 (CAS No. 199986-75- 9) (CV Therapeutics), and an analogue or derivative thereof.
14512. The method of item 14486 wherein the agent is a cyclooxygenase 1 inhibitor.
14513. The method of item 14486 wherein the agent is a DHFR inhibitor.
14514. The method of item 14486 wherein the agent is a dual integrin inhibitor.
14515. The method of item 14486 wherein the agent is an elastase inhibitor.
14516. The method of item 14486 wherein the agent is an elongation factor-1 alpha inhibitor.
14517. The method of item 14486 wherein the agent is an endothelial growth factor antagonist.
14518. The method of item 14486 wherein the agent is an endothelial growth factor receptor kinase inhibitor selected from the group consisting of sorafenib tosylate (Bayer), AAL-993 (Novartis), ABP-309 (Novartis), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), E- 7080 (Eisai), EG-3306 (Ark Therapeutics), EXEL-2880 (Exelixis), GW- 654652 (GlaxoSmithKline), a KDR inhibitor from LG Life Sciences, CT-6685 and CT-6729 (UCB), KRN-633 and KRN-951 (Kirin Brewery), OSI-930 (OSI Pharmaceuticals), SP-5.2 (Supratek Pharma), SU-11657 (Pfizer), a Tie-2 antagonist (Hybrigenics), a VEGFR-2 kinase inhibitor (Bristol-Myers Squibb), XL-647 (Exelixis), a KDR inhibitor from Abbott Laboratories, sorafenib tosylate, and an analogue or derivative thereof.
14519. The method of item 14486 wherein the agent is an endotoxin antagonist.
14520. The method of item 14486 wherein the agent is an epothilone and tubulin binder.
14521. The method of item 14486 wherein the agent is an estrogen receptor antagonist.
14522. The method of item 14486 wherein the agent is an FGF inhibitor.
14523. The method of item 14486 wherein the agent is a farnexyl transferase inhibitor.
14524. The method of item 14486 wherein the agent is famesyltransferase inhibitor selected from the group of A-197574 (Abbott), a famesyltransferase inhibitor from Servier, FPTIII (Strathclyde Institute for Drug R), LB-42908 (LG Life Sciences), Pharmaprojects No. 5063 (Genzyme), Pharmaprojects No. 5597 (Ipsen), Yissum Project No. B-1055 (Yissum), and an analogue or derivative thereof.
14525. The method of item 14486 wherein the agent is an FLT-3 kinase inhibitor.
14526. The method of item 14486 wherein the agent is an FGF receptor kinase inhibitor. 14527. The method of item 14486 wherein the agent is a fibrinogen antagonist selected from the group consisting of AUV-201 (Auvation), MG-13926 (Sanofi-Aventis), plasminogen activator (CAS No. 105913-11-9) (from Sanofi-Aventis or UCB)1 plasminogen activator-2 (tPA- 2) (Sanofi-Aventis), pro-urokinase (CAS No. 82657-92-9) (Sanofi-Aventis), and an analogue or derivative thereof.
14528. The method of item 14486 wherein the agent is a heat shock protein 90 antagonist selected from the group consisting of SRN- 005 (Sirenade), geldanamycin, NSC-33050 (17-allylaminogeldanamycin; 17- AAG), 17-dimethylaminoethylamino-17-demethoxy-geldanamycin (17- DMAG), rifabutin (rifamycin XIV, 1',4-didehydro-1-deoxy-1 )4-dihydro-5'-(2- methylpropyl)-1-oxo-), and an analogue or derivative thereof.
14529. The method of item 14486 wherein the agent is a histone deacetylase inhibitor. .
14530. The method of item 14486 wherein the agent is an HMGCoA reductase inhibitor selected from the group consisting of an atherosclerosis therapeutic from Lipid Sciences, ATI-16000 (ARYx Therapeutics), KS-01-019 (Kos Pharmaceuticals), Pharmaprojects No. 2197 (Sanofi-Aventi), RP 61969 (Sanofi-Aventis), and an analogue or derivative thereof.
14531. The method of item 14486 wherein the agent is an ICAM inhibitor.
14532. The method of item 14486 wherein the agent is an IL, ICE and IRAK antagonist, wherein the antagonist is a CJ-14877, CP- 424174 (Pfizer), NF-61 (Negma-Lerads), and an analogue or derivative thereof. 14533. The method of item 14486 wherein the agent is an IL-2 inhibitor.
14534. The method of item 14486 wherein the agent is an immunosuppressant selected from the group consisting of teriflunomide (Sanofi Aventis), chlorsulfaquinoxalone (NSC-339004), chlorsulfaquinoxalone sulfate, CS-712 (Sankyo), ismomultin alfa (CAS No. 457913-93-8) (Akzo Nobel), antiallergics from GenPat77, antiinflammatories or AT-005 (Androclus Therapeutics), autoimmune disease therapeutics from EpiVax, BN-007 (Bone), budesonide (CAS No. 51333-22- 3) (MAP Pharmaceuticals), CO-14 (Genzyme), edratide (CAS No. 433922- 67-9) (Teva), EP-314 (Enanta), eprovafen (CAS No. 101335-99-3) (Sanofi- Aventis), HWA-131 (CAS No. 118788-41-3) (Sanofi-Aventis), immunomodulators from MerLion Pharmaceuticals, immunosuppressives from Alchemia, IPL-12 (Inflazyme), MDL-9563 (CAS No. 27086-86-8) (Sanofi-Aventis), Pharmaprojects No. 2330 (Sanofi-Aventis), Pharmaprojects No. 6426 (Abgenix), PXS-25 (Pharmaxis), rosmarinic acid (CAS No. 20283-92-5) (Sanofi-Aventis), RP 42927 or RP 54745 (CAS No. 135330-08-4) (Sanofi-Aventis), SGN-35 (Seattle Genetics), ST-1959 (Sigma-Tau), type I diabetes therapy from SYNX Pharma, UNIL-88 (Debiopharm), VP-025 (Vasogen), VR-694 (Vectura), PRTX-001 (Protalex), and an analogue or derivative thereof.
14535. The method of item 14486 wherein the agent is an IMPDH (inosine monophosphate).
14536. The method of item 14486 wherein the agent is an integrin antagonist.
14537. The method of item 14486 wherein the agent is an interleukin antagonist. 14538. The method of item 14486 wherein the agent is an inhibitor of type III receptor tyrosine kinase.
14539. The method of item 14486 wherein the agent is an irreversible inhibitor of enzyme methionine aminopeptidase type 2.
14540. The method of item 14486 wherein the agent is an isozyme selective delta protein kinase C inhibitor.
14541. The method of item 14486 wherein the agent a JAK3 enzyme inhibitor.
14542. The method of item 14486 wherein the agent is a JNK inhibitor.
14543. The method of item 14486 wherein the agent is a kinase inhibitor.
14544. The method of item 14486 wherein the agent is kinesin antagonist.
14545. The method of item 14486 wherein the agent is a kinesin antagonist.
14546. The method of item 14486 wherein the agent is a leukotriene inhibitor and antagonist selected from the group consisting of ambicromil (CAS No. 58805-38-2) (Sanofi-Aventis), amelubant (CAS No. 346735-24-8) (Boehringer Ingelheim), DW-1141 (Dong Wha), ebselen (Daiichi Pharmaceutical), ibudilast (Kyorin), leucotriene inhibitors from Sanofi-Aventis, lymphotoxin-beta receptor (LT-D) from Biogen Idee, Pharmaprojects No. 1535 and 2728 (CAS No. 119340-33-9) (Sanofi- Aventis), R-112 (Rigel), Rev-5367 (CAS No. 92532-05-3) (Sanofi-Aventis), RG-14893 (CAS No. 141835-49-6) (Sanofi-Aventis), RG-5901-A (CAS No. 101910-24-1), 92532-23-5, RP 66153 (CAS No. 142422-79-5), RP 66364 (CAS No.186916-92-5), RP 69698 (CAS No. 141748-00-7) (Sanofi-Aventis), SC-411930 (Pfizer), SC-41930 (CAS No. 120072-59-5) (Pfizer), SC-50605 (CAS No. 138828-39-4) (Pfizer), SC-51146 (CAS No. 141059-52-1), SC- 53228 (CAS No. 153633-01-3) (Pfizer), spaglumic acid (ZY-15106) (CAS No.3106-85-2), 80619-64-3 (Novartis), tipredane (CAS No. 85197-77-9) (Bristol-Myers Squibb), U-75302 (CAS No. 119477-85-9) (Pfizer), and analogue or derivative thereof.
14547. The method of item 14486 wherein the agent is an MAP kinase inhibitor.
14548. The method of item 14486 wherein the agent is a matrix metalloproteinase inhibitor.
14549. The method of item 14486 wherein the agent is an MCP-CCR2 inhibitor.
14550. The method of item 14486 wherein the agent is an mTOR inhibitor.
14551. The method of item 14486 wherein the agent is an mTOR kinase inhibitor.
14552. The method of item 14486 wherein the agent is a microtubule inhibitor selected from the group consisting of antibody- maytansinoid conjugates from Biogen Idee, colchicines (MantiCore Pharmaceuticals), anticancer immunoconjugates from Johnson & Johnson, DIME from Octamer, gni-1f (GNI), huC242-DM4, huMy9-6-DM1 (ImmunoGen), IDN-5404 (Indena), IMO-098, IMOderm (Imotep), mebendazole (Introgen Therapeutics), microtubule poisons from Cambridge Enterprise, paclitaxel such as LOTAX from Aphios (CAS No. 33069-62-4), Genexol-PM from Samyang, Pharmaprojects No. 6383 (Tapestry Pharmaceuticals), RPR-112378 (Sanofi-Aventis), SGN-75 (Seattle Genetics), SPL-7435 (Starpharma), SSR-250411 (Sanofi-Aventis), trastuzumab-DM1 (Genentech), vinorelbine, and an analogue or derivative thereof.
14553. The method of item 14486 wherein the agent is an MIF inhibitor.
14554. The method of item 14486 wherein the agent is an MMP inhibitor.
14555. The method of item 14486 wherein the agent is a neurokinin (NK) antagonist selected from the group consisting of anthrotainin (CAS No. 148084-40-6) (Sanofi-Aventis), an IBS thereapeutic from ArQuIe, MDL-105212A (CAS No. 167261-60-1) (Ssanofi-Aventis), Pharmaprojects No. 2744, 3258 (CAS No. 139167-47-8) 4006, 4201, or 5986 (Sanofi-Aventis), RP 67580 (CAS No. 135911-02-3), SR-144190 (CAS No. 201152-86-5), SSR-240600, SSR-241586 (Sanofi-Aventis), TKA-457 (Novartis), vestipitant mesylate (CAS No. 334476-64-1) (GlaxoSmith Kline), Win-64821 (Sanofi-Aventis), PRX-96026 (Predix Pharmaceuticals), and an analogue or derivative thereof.
14556. The method of item 14486 wherein the agent is an NF kappa B inhibitor selected from the group consisting of AVE-0545 or AVE-0547 (Sanofi-Aventis), bortezomib (CAS No. 179324-69-7) (Millennium Pharmaceuticals), dexanabinol (CAS No. 112924-45-5) (Pharmos), dexlipotam (Viatris), Pharmaprojects No. 6283 (INDRA) (OXiGENE), IPL- 576092 (CAS No. 137571-30-3) (Inflazyme), NFKB decoy (Corgentech), NFKB decoy oligo (AnGes MG), NFKB's from Ariad, osteoporosis treatments or S5 (F005) from Fulcrum Pharmaceuticals, P61 (Phytopharm), R-flurbiprofen (CAS No. 5104-49-4) (Encore Pharmaceuticals), and an analogue or derivative thereof.
14557. The method of item 14486 wherein the agent is a nitric oxide agonist.
14558. The method of item 14486 wherein the agent is an ornithine decarboxylase inhibitor.
14559. The method of item 14486 wherein the agent is a p38 MAP kinase inhibitor selected from the group consisting of AZD-6703 (AstraZeneca), JX-401 (Jexys Pharmaceuticals), BMS-2 (Bristol-Myers Squibb), a p38 MAP kinase inhibitor from Novartis, a p38-alpha MAP kinase inhibitor from Amphora, Pharmaprojects No. 5704 (Pharmacopeia), RPR- 200765A (Sanofi-Aventis), SD-282 (Johnson & Johnson), TAK-715 (Takeda), and an analogue or derivative thereof.
14560. The method of item 14486 wherein the agent is a palmitoyl-protein thioesterase inhibitor.
14561. The method of item 14486 wherein the agent is a PDGF receptor kinase inhibitor selected from the group consisting of AAL- 993, AMN-107, or ABP-309 (Novartis), AMG-706 (Amgen), BAY-57-9352 (Bayer), CDP-860 (UCB), E-7080 (Eisai), imatinib (CAS No. 152459-95-5) (Novartis), OSI-930 (OSI Pharmaceuticals), RPR-127963E (Sanofi-Aventis), RWJ-540973 (Johnson & Johnson), sorafenib tosylate (Bayer), SU-11657 (Pfizer), tandutinib (CAS No. 387867-13-2) (Millennium Pharmaceuticals), vatalanib (Novartis), ZK-CDK (Schering AG), and an analogue or derivative thereof.
14562. The method of item 14486 wherein the agent is (-)-halofenate (Metabolex), AMG-131 (Amgen), antidiabetics from Japan Tobacco, AZD-4619, AZD-8450, AZD-8677 (AstraZeneca), DRF-10945, balaglitazone (Dr Reddy's), CS-00088, CS-00098 (Chipscreen Biosciences), E-3030 (Eisai), etalocib (CAS No. 161172-51-6) (EIi Lilly), GSK-641597 (Ligand), GSK-677954 (GlaxoSmithKline), GW-409544 (Ligand), GW- 590735 (GlaxoSmithKline), K-111 (Hoffmann-La Roche), LY-518674 (EIi Lilly), LY-674 (Ligand), LY-929 (Ligand), MC-3001 , MC-3002 (MaxoCore Pharmaceuticals), metformin HCI + pioglitazone (CAS No. 1115-70-4 and 112529-15-4), ACTOPLUS MET from Andrx), muraglitazar (CAS No. 331741-94-7) (Bristol-Myers Squibb), naveglitazar (Ligand), oleoylethanolamide (Kadmus Pharmaceuticals), ONO-5129, pioglitazone hydrochloride (CAS No. 111025-46-8 and 112529-15-4) (Takeda), PLX-204 (Plexxikon), PPAR agonists from Genfit, PPAR delta agonists from EIi Lilly, PPAR-alpha agonists from CrystalGenomics, PPAR-gamma modulators and PPAR-β modulators from CareX, rosiglitazone maleate (CAS No. 122324- 73-4 or 155141-29-0) (GlaxoSmithKline), rosiglitazone maleate/glimepir (CAS No. 155141-29-0 and 93479-97-1), AVANDARYL, rosiglitazone maleate/metformin extend (CAS No. 155141-29-0 and 657-24-9), AVANDAMET, rosiglitazone maleate+metformin, AVANDAMET (GlaxoSmithKline), tesaglitazar (AstraZeneca), and an analogue or derivative thereof.
14563. The method of item 14486 wherein the agent is a phosphatase inhibitor.
14564. The method of item 14486 wherein the agent is a phosphodiesterase (PDE) inhibitor selected from the group consisting of avanafil (Tanabe Seiyaku), dasantafil (CAS No. 569351-91-3) (Schering- Plough), A-906119 (CAS No. 134072-58-5), DL-850 (Sanofi-Aventis), GRC- 3015, GRC-3566, GRC-3886 (Glenmark), HWA-153 (CAS No. 56395-66-5) (Sanofi-Aventis), hydroxypumafentrine (Altana), IBFB-130011 , IBFB-14-016, IBFB-140301, 1BFB-150007, IBFB-211913 (IBFB Pharma), L-826141 (Merck & Co), medorinone (CAS No. 88296-61-1) (Sanofi-Aventis), MEM- 1917 (Memory Pharmaceuticals), ND-1251 (Neuro3d), PDE inhibitors from ICOS, PDE IV inhibitors from Memory Pharmaceuticals and CrystalGenomics, Pharmaprojects No. 2742 and 6141 (Sanofi-Aventis), QAD-171 (Novartis), RHC-2963 (CAS No. 76993-12-9 and 76993-14-1), RPR-117658, RPR-122818 derivatives, SR-24870 , and RPR-132294 (Sanofi-Aventis), SK-350 (ln2Gen), stroke therapy agents from deCODE Genetics, TAS-203 (Taiho), tofimilast (CAS No. 185954-27-2) (Pfizer), UK- 371800 (Pfizer), WIN-65579 (CAS No. 158020-82-7) (Sanofi-Aventis), IBFB- 130020 (IBFB Pharma), OPC-6535 (CAS No. 145739-56-6) (Otsuka), a phosphodiesterase III inhibitor, enoximone, a phosphodiesterase IV inhibitor, fosfosal, Atopik (Barrier Therapeutics), triflusal, a phosphodiesterase V inhibitor, and an analogue or derivative thereof.
14565. The method of item 14486 wherein the agent is a PKC inhibitor.
14566. The method of item 14486 wherein the agent is a platelet activating factor antagonist.
14567. The method of item 14486 wherein the agent is a platelet-derived growth factor receptor kinase inhibitor.
14568. The method of item 14486 wherein the agent is a prolyl hydroxylase inhibitor. 14569. The method of item 14486 wherein the agent is a polymorphonuclear neutrophil inhibitor.
14570. The method of item 14486 wherein the agent is a protein kinase B inhibitor.
14571. The method of item 14486 wherein the agent is a protein kinase C stimulant.
14572. The method of item 14486 wherein the agent is a purine nucleoside analogue.
14573. The method of item 14486 wherein the agent is a purinoreceptor P2X antagonist.
14574. The method of item 14486 wherein the agent is a. Raf kinase inhibitor.
14575. The method of item 14486 wherein the agent is a reversible inhibitor of ErbB1 and ErbB2.
14576. The method of item 14486 wherein the agent is a ribonucleoside triphosphate reductase inhibitor.
14577. The method of item 14486 wherein the agent is an SDF-1 antagonist.
14578. The method of item 14486 wherein the agent is a sheddase inhibitor. 14579. The method of item 14486 wherein the agent is an SRC inhibitor.
14580. The method of item 14486 wherein the agent is a stromelysin inhibitor.
14581. The method of item 14486 wherein the agent is an Syk kinase inhibitor.
14582. The method of item 14486 wherein the agent is a telomerase inhibitor.
14583. The method of item 14486 wherein the agent is a TGF beta inhibitor selected from the group consisting of pirfenidone (CAS No. 53179-13-8) (MARNAC), tranilast (CAS No. 53902-12-8) (Kissei), IN-
1130 (ln2Gen), mannose-6-phosphate (BTG), TGF-β antagonists from Inflazyme (Pharmaprojects No. 6075), TGF-β antagonists from Sydney, non-industrial source), TGF-β I receptor kinase inhibitors from EIi Lilly, TGF- β receptor inhibitors from Johnson & Johnson, and an analogue or derivative thereof.
14584. The method of item 14486 wherein the agent is a TNFα antagonist or TACE inhibitor selected from the group consisting of adalimumab (CAS No. 331731-18-1) (Cambridge Antibody Technology), AGIX-4207 (AtheroGenics), AGT-1 (Advanced Biotherapy), an antiinflammatory from Borean Pharma, Cellzome, or Paradigm Therapeutics, anti-inflammatory vaccine (TNF-alpha kinoid) from Neovacs, humanized anti-TNF antibody or an anti-TNF MAb (CB0006) Celltech (UCB), apratastat (CAS No. 287445-51-0) (Wyeth), BMS-561392 (Bristol-Myers Squibb), BN- 006 (Bone), certolizumab pegol (CAS No. 428863-50-7 or CH-138 (UCB), cilomilast (CAS No. 153259-65-5) (GlaxoSmithKline), CR-1 (Nuada Pharmaceuticals), CRx-119 (CombinatoRx), D-5410 (UCB), dacopafant (CAS No. 125372-33-0) (Sanofi-Aventis), dersalazine (CAS No. 188913-57- 7/188913-58-8) (Uriach), etanercept (CAS No. 185243-69-0) (Amgen), ethyl pyruvate (Critical (Critical Therapeutics), golimumab (CAS No. 476181-74-5) (Johnson & Johnson), hormono-immunotherapy from Ipsen, CDP571 (e.g., Humicade from UCB), IC-485 (ICOS), infliximab (CAS No. 170277-31-3) (Johnson & Johnson), IP-751 (Manhattan Pharmaceuticals), ISIS-104838 (CAS No. 250755-32-9) (ISIS Pharmaceuticals), lenalidomide (CAS No. 191732-72-6) (Celgene), lentinan (CAS No. 37339-90-5) (Ajinomoto), MDL- 201112 (CAS No. 142130-73-2) (Sanofi-Aventis), medroxyprogesterone (CAS No. 520-85-4) (InKine Pharmaceutical), N-acetylcysteine (CAS No. 616-91-1) (Zambon), NBE-P2 (DIREVO Biotech), nerelimomab (CAS No. 162774-06-3) (Chiron), OM-294DP (OM PHARMA), onercept (CAS No. 199685-57-9) (Yeda), PASSTNF-alpha (Verigen), pentoxifylline or oxypentifylline (Sanofi-Aventis), Pharmaprojects No. 4091 , 4241, 4295, or 4488 (Sanofi-Aventis), Pharmaprojects No. 5480 (Amgen), Pharmaprojects No. 6749 (Cengent), pirfenidone (CAS No. 53179-13-8) (MARNAC), RPR- 132294 (Sanofi-Aventis), S5 (F002) (Fulcrum Pharmaceuticals), simvastatin (CAS No. 79902-63-9) (Merck & Co), STA-6292 (Synta Pharmaceuticals), tacrolimus (CAS No. 104987-11-3) (Fujisawa LifeCycle Pharma), talactoferrin alfa (CAS No. 308240-58-6) (Agennix), thalidomide (CAS No. 50-35-1) (Celgene), TNF antagonists form ProStrakan, and Synergen, TNF inhibitors (Amgen), TNF-alpha antagonists from Dynavax Technologies and Jerina AG (Germany), TNF-alpha inhibitors from IBFB Pharma and Xencor (Xencor), torbafylline (CAS No. 105102-21-4) (Sanofi-Aventis), UR-1505 (Uriach), VT-346 (Viron Therapeutics), YSIL6 (Y's Therapeutics), YSTH2 (Y's Therapeutics), NPI-1302a-3 (Nereus Pharmaceuticals, a TNF antagonist from Jerina AG (Germany), dersalazine, and an analogue or derivative thereof. 14585. The method of item 14486 wherein the agent is a Toll receptor inhibitor.
14586. The method of item 14486 wherein the agent is a tubulin antagonist.
14587. The method of item 14486 wherein the agent is a tyrosine kinase inhibitor selected from the group consisting of SU-011248, SUTENT from Pfizer Inc. (New York, NY), BMS-354825, PN-355 (Paracelsian Pharmaceuticals), AGN-199659 (Allergan), AAL-993 or ABP- 309 (Novartis), adaphostin (NIH), AEE-788 (Novartis), AG-013736 (OSI Pharmaceuticals), AG-13736 (Pfizer), ALT-110 (Alteris Therapeutics), AMG- 706 (Amgen), anticancer MAbs from Xencor, anti-EGFrvlll MAbs from Abgenix, anti-HER2 MAb from Abiogen, AZD-2171 or AZD-9935 (AstraZeneca), BAY-57-9352 (Bayer), BIBF-1120 (Boehhnger Ingelheim), CEP-5214 (Cephalon), CEP-7055 (Cephalon), cetuximab (ImClone Systems), CHIR-200131 and CHIR-258 (Chiron), CP-547632 (OSI Pharmaceuticals), CP-724714 (Pfizer), CT-301 (Creabilis Therapeutics), D- 69491 (Baxter International), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EGFR/ErbB2 inhibitors from Array BioPharma, erlotinib (CAS No. 183319- 69-9) (OSI Pharmaceuticals), EXEL-2880 (Exelixis), FK-778 (Sanofi- Aventis) , gefitinib (CAS No. 184475-35-2) (AstraZeneca), GW-2286 or GW- 654652 (GlaxoSmithKline), her2/neu antigen from AlphaVax, HER-2/neu inhibitor from Generex, Herzyme (Medipad) (Sirna Therapeutics), HKI-272 (Wyeth), HuMax-EGFr (Genmab), idronoxil (CAS No. 81267-65-4) (Novogen), 1GF-1 inhibitors from Ontogen, IMC-11F8 (ImClone Systems), kahalalide F (CAS No. 149204-42-2) (PharmaMar), KDR inhibitor from LG Life Sciences, KDR inhibitors from Abbott Laboratories, KDR kinase inhibitors (UCB), Kdr kinase inhibitors from Merck & Co, KRN-633 and KRN- 951 (Kirin Brewery), KSB-102 (Xenova), lapatinib ditosylate (CAS No. 388082-78-8) (GlaxoSmithKline), matuzumab (Merck KGaA), MDX-214 (Medarex), ME-103 (Pharmexa), MED-A300 (Gerolymatos), MNAC-13 (Lay Line Genomics), nimotuzumab (Center of Molecular Immunology), NSC- 330507 or NSC-707545 (NIH), NV-50 (Novogen), OSI-930 (OSI Pharmaceuticals), panitumumab (Abgenix), pelitinib (CAS No. 287933-82-7) (Wyeth), pertuzumab (CAS No. 380610-27-5) (Genentech), Pharmaprojects No. 3985 (Sanofi-Aventis), prostate cancer therapeutics from Sequenom (SQPC35, SQPC36, SQPC90), removab and remoxab (Trion Pharma), RG- 13022 (CAS No. 136831-48-6), RG-13291 (CAS No. 138989-50-1), or RG- 14620 (CAS No. 136831-49-7) (Sanofi-Aventis), RM-6427 (Romark), RNAi breast cancer therapy from Benitec, RP 53801 (CAS No. 125882-88-4) (Sanofi-Aventis), sorafenib tosylate (Bayer), SU-11657 (Pfizer), Tie-2 antagonists from Semaia (Hybrigenics), Tie-2 inhibitors from Ontogen, trastuzumab (CAS No. 180288-69-1) (Genentech), tyrosine kinase inhibitors from Sanofi-Aventis, U3-1287, U3-1565, U3-1784, U3-1800 (U3 Pharma), vatalanib (Novartis), VEGFR-2 kinase inhibitor from Bristol-Myers Squibb, XL-647 (Exelixis), ZD-6474 (AstraZeneca), ZK-CDK (Schering AG), an EGFR tyrosine kinase inhibitor, EKB-569 (Wyeth), and an analogue or derivative thereof.
14588. The method of item 14486 wherein the agent is a VEGF inhibitor.
14589. The method of item 14486 wherein the agent is a vitamin D receptor agonist.
14590. The method of item 14486 wherein the agent is ZD-6474 (an angiogenesis inhibitor).
14591. The method of item 14486 wherein the agent is AP-23573 (an mTOR inhibitor). 14592. The method of item 14486 wherein the agent is synthadotin (a tubulin antagonist).
14593. The method of item 14486 wherein the agent is S-0885 (a collagenase inhibitor).
14594. The method of item 14486 wherein the agent is aplidine (an elongation factor-1 alpha inhibitor).
14595. The method of item 14486 wherein the agent is ixabepilone (an epithilone).
14596. The method of item 14486 wherein the agent is IDN-5390 (an angiogenesis inhibitor).
14597. The method of item 14486 wherein the agent is SB-2723005 (an angiogenesis inhibitor).
14598. The method of item 14486 wherein the agent is ABT-518 (an angiogenesis inhibitor).
14599. The method of item 14486 wherein the agent is combretastatin (an angiogenesis inhibitor).
14600. The method of item 14486 wherein the agent is anecortave acetate (an angiogenesis inhibitor).
14601. The method of item 14486 wherein the agent is SB-715992 (a kinesin antagonist). 14602. The method of item 14486 wherein the agent is temsirolimus (an mTOR inhibitor).
14603. The method of item 14486 wherein the agent is adalimumab (a TNFα antagonist).
14604. The method of item 14486, wherein the composition comprises a polymer.
14605. The method of item 14486, wherein the composition comprises a polymeric carrier.
14606. The method of item 14486 wherein the anti- scarring agent inhibits adhesion between the device and a host into which the device is implanted.
14607. The method of item 14486 wherein the device delivers the anti-scarring agent locally to tissue proximate to the device.
14608. The method of item 14486 wherein the device has a coating that comprises the anti-scarring agent.
14609. The method of item 14486, wherein the device has a coating that comprises the agent and is disposed on a surface of the implant.
14610. The method of item 14486, wherein the device has a coating that comprises the agent and directly contacts the implant.
14611. The method of item 14486, wherein the device has a coating that comprises the agent and indirectly contacts the implant. 14612. The method of item 14486, wherein the device has a coating that comprises the agent and partially covers the implant.
14613. The method of item 14486, wherein the device has a coating that comprises the agent and completely covers the implant.
14614. The method of item 14486, wherein the device has a uniform coating.
14615. The method of item 14486, wherein the device has a non-uniform coating.
14616. The method of item 14486, wherein the device has a discontinuous coating.
14617. The method of item 14486, wherein the device has a patterned coating.
14618. The method of item 14486, wherein the device has a coating with a thickness of 100 Dm or less.
14619. The method of item 14486, wherein the device has a coating with a thickness of 10 Dm or less.
14620. The method of item 14486, wherein the device has a coating, and the coating adheres to the surface of the implant upon deployment of the implant.
14621. The method of item 14486, wherein the device has a coating, and wherein the coating is stable at room temperature for a period of 1 year. 14622. The method of item 14486, wherein the device has a coating, and wherein the anti-scarring agent is present in the coating in an amount ranging between about 0.0001% to about 1 % by weight.
14623. The method of item 14486, wherein the device has a coating, and wherein the anti-scarring agent is present in the coating in an amount ranging between about 1% to about 10% by weight.
14624. The method of item 14486, wherein the device has a coating, and wherein the anti-scarring agent is present in the coating in an amount ranging between about 10% to about 25% by weight.
14625. The method of item 14486, wherein the device has a coating, and wherein the anti-scarring agent is present in the coating in an amount ranging between about 25% to about 70% by weight.
14626. The method of item 14486, wherein the device has a coating, and wherein the coating further comprises a polymer.
14627. The method of item 14486, wherein the device has a first coating having a first composition and a second coating having a second composition.
14628. The method of item 14486, wherein the device has a first coating having a first composition and a second coating having a second composition, wherein the first composition and the second composition are different.
14629. The method of item 14486, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a copolymer. 14630. The method of item 14486, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a block copolymer.
14631. The method of item 14486, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a random copolymer.
14632. The method of item 14486, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a biodegradable polymer.
14633. The method of item 14486, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a non-biodegradable polymer.
14634. The method of item 14486, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a hydrophilic polymer.
14635. The method of item 14486, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a hydrophobic polymer.
14636. The method of item 14486, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a polymer having hydrophilic domains.
14637. The method of item 14486, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a polymer having hydrophobic domains. 14638. The method of item 14486, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a non-conductive polymer.
14639. The method of item 14486, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises an elastomer.
14640. The method of item 14486, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a hydrogel.
14641. The method of item 14486, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a silicone polymer.
14642. The method of item 14486, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a hydrocarbon polymer.
14643. The method of item 14486, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a styrene-derived polymer.
14644. The method of item 14486, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a butadiene polymer.
14645. The method of item 14486, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a macromer. 14646. The method of item 14486, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a poly(ethylene glycol) polymer.
14647. The method of item 14486 wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises an amorphous polymer.
14648. The method of item 14486, wherein the device comprises a lubricious coating.
14649. The method of item 14486 wherein the anti- scarring agent is located within pores or holes of the device.
14650. The method of item 14486 wherein the anti- scarring agent is located within a channel, lumen, or divet of the device.
14651. The method of item 14486, wherein the device comprises a second pharmaceutically active agent.
14652. The method of item 14486 wherein the device comprises an anti-inflammatory agent.
14653. The method of item 14486 wherein the device comprises an agent that inhibits infection.
14654. The method of item 14486 wherein the device comprises an agent that inhibits infection, and wherein the agent is an anthracycline. 14655. The method of item 14486 wherein the device comprises an agent that inhibits infection, and wherein the agent is doxorubicin.
14656. The method of item 14486 wherein the device comprises an agent that inhibits infection, and wherein the agent is mitoxantrone.
14657. The method of item 14486 wherein the device comprises an agent that inhibits infection, and wherein the agent is a fluoropyrimidine.
14658. The method of item 14486 wherein the device comprises an agent that inhibits infection, and wherein the agent is 5- fluorouracil (5-FU).
14659. The method of item 14486 wherein the device comprises an agent that inhibits infection, and wherein the agent is a folic acid antagonist.
14660. The method of item 14486 wherein the device comprises an agent that inhibits infection, and wherein the agent is methotrexate.
14661. The method of item 14486 wherein the device comprises an agent that inhibits infection, and wherein the agent is a podophylotoxin.
14662. The method of item 14486 wherein the device comprises an agent that inhibits infection, and wherein the agent is etoposide. 14663. The method of item 14486 wherein the device comprises an agent that inhibits infection, and wherein the agent is a camptothecin.
14664. The method of item 14486 wherein the device comprises an agent that inhibits infection, and wherein the agent is a hydroxyurea.
14665. The method of item 14486 wherein the device comprises an agent that inhibits infection, and wherein the agent is a platinum complex.
14666. The method of item 14486 wherein the device comprises an agent that inhibits infection, and wherein the agent is cisplatin.
14667. The method of item 14486, further comprising an anti-thrombotic agent.
14668. The method of item 14486 wherein the device comprises a visualization agent.
14669. The method of item 14486 wherein the device comprises a visualization agent, wherein the visualization agent is a radiopaque material, and wherein the radiopaque material comprises a metal, a halogenated compound, or a barium containing compound.
14670. The method of item 14486 wherein the device comprises a visualization agent, wherein the visualization agent is a radiopaque material, and wherein the radiopaque material comprises barium, tantalum, or technetium. 14671. The method of item 14486 wherein the device comprises a visualization agent, and wherein the visualization agent is a MRI responsive material.
14672. The method of item 14486 wherein the device comprises a visualization agent, and wherein the visualization agent comprises a gadolinium chelate.
14673. The method of item 14486 wherein the device comprises a visualization agent, and wherein the visualization agent comprises iron, magnesium, manganese, copper, or chromium.
14674. The method of item 14486 wherein the device comprises a visualization agent, and wherein the visualization agent comprises an iron oxide compound.
14675. The method of item 14486 wherein the device comprises a visualization agent, and wherein the visualization agent comprises a dye, pigment, or colorant.
14676. The method of item 14486 wherein the device comprises an echogenic material.
14677. The method of item 14486 wherein the device comprises an echogenic material, and wherein the echogenic material is in the form of a coating.
14678. The method of item 14486 wherein the device is sterile. 14679. The method of item 14486 wherein the anti- scarring agent is released into tissue in the vicinity of the device after deployment of the device.
14680. The method of item 14486 wherein the anti- scarring agent is released into tissue in the vicinity of the device after deployment of the device, and wherein the tissue is connective tissue.
14681. The method of item 14486 wherein the anti- scarring agent is released into tissue in the vicinity of the device after deployment of the device, and wherein the tissue is muscle tissue.
14682. The method of item 14486 wherein the anti- scarring agent is released into tissue in the vicinity of the device after deployment of the device, and wherein the tissue is nerve tissue.
14683. The method of item 14486 wherein the anti- scarring agent is released into tissue in the vicinity of the device after deployment of the device, and wherein the tissue is epithelium tissue.
14684. The method of item 14486 wherein the anti- scarring agent is released in effective concentrations from the device over a period ranging from the time of deployment of the device to about 1 year.
14685. The method of item 14486 wherein the anti- scarring agent is released in effective concentrations from the device over a period ranging from about 1 month to 6 months.
14686. The method of item 14486 wherein the anti- scarring agent is released in effective concentrations from the device over a period ranging from about 1 - 90 days. 14687. The method of item 14486 wherein the anti- scarring agent is released in effective concentrations from the device at a constant rate.
14688. The method of item 14486 wherein the anti- scarring agent is released in effective concentrations from the device at an increasing rate.
14689. The method of item 14486 wherein the anti- scarring agent is released in effective concentrations from the device at a decreasing rate.
14690. The method of item 14486 wherein the anti- scarring agent is released in effective concentrations from the composition comprising the anti-scarring agent by diffusion over a period ranging from the time of deployment of the device to about 90 days.
14691. The method of item 14486 wherein the anti- scarring agent is released in effective concentrations from the composition comprising the anti-scarring agent by erosion of the composition over a period ranging from the time of deployment of the device to about 90 days.
14692. The method of item 14486 wherein the device comprises about 0.01 Dg to about 10 Dg of the anti-scarring agent.
14693. The method of item 14486 wherein the device comprises about 10 Dg to about 10 mg of the anti-scarring agent.
14694. The method of item 14486 wherein the device comprises about 10 mg to about 250 mg of the anti-scarring agent. 14695. The method of item 14486 wherein the device comprises about 250 mg to about 1000 mg of the anti-scarring agent.
14696. The method of item 14486 wherein the device comprises about 1000 mg to about 2500 mg of the anti-scarring agent.
14697. The method of item 14486 wherein a surface of the device comprises less than 0.01 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
14698. The method of item 14486 wherein a surface of the device comprises about 0.01 Dg to about 1 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
14699. The method of item 14486 wherein a surface of the device comprises about 1 Dg to about 10 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
14700. The method of item 14486 wherein a surface of the device comprises about 10 Dg to about 250 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
14701. The method of item 14486 wherein a surface of the device comprises about 250 Dg to about 1000 Dg of the anti-scarring agent of anti-scarring agent per mm2 of device surface to which the anti- scarring agent is applied.
14702. The method of item 14486 wherein a surface of the device comprises about 1000 Dg to about 2500 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied. 14703. The method of item 14486 wherein the combining is performed by direct affixing the agent or the composition to the implant.
14704. The method of item 14486 wherein the combining is performed by spraying the agent or the component onto the implant.
14705. The method of item 14486 wherein the combining is performed by electrospraying the agent or the composition onto the implant.
14706. The method of item 14486 wherein the combining is performed by dipping the implant into a solution comprising the agent or the composition.
14707. The method of item 14486 wherein the combining is performed by covalently attaching the agent or the composition to the implant.
14708. The method of item 14486 wherein the combining is performed by non-covalently attaching the agent or the composition to the implant.
14709. The method of item 14486 wherein the combining is performed by coating the implant with a substance that contains the agent or the composition.
14710. The method of item 14486 wherein the combining is performed by coating the implant with a substance that absorbs the agent. 14711. The method of item 14486 wherein the combining is performed by interweaving a thread composed of, or coated with, the agent or the composition.
14712. The method of item 14486 wherein the combining is performed by covering all the implant with a sleeve that contains the agent or the composition.
14713. The method of item 14486 wherein the combining is performed by covering a portion of the implant with a sleeve that contains the agent or the composition.
14714. The method of item 14486 wherein the combining is performed by covering all the implant with a cover that contains the agent or the composition.
14715. The method of item 14486 wherein the combining is performed by covering a portion of the implant with a cover that contains the agent or the composition.
14716. The method of item 14486 wherein the combining is performed by covering all the implant with an electrospun fabric that contains the agent or the composition.
14717. The method of item 14486 wherein the combining is performed by covering a portion of the implant with an electrospun fabric that contains the agent or the composition.
14718. The method of item 14486 wherein the combining is performed by covering all the implant with a mesh that contains the agent or the composition. 14719. The method of item 14486 wherein the combining is performed by covering a portion of the implant with a mesh that contains the agent or the composition.
14720. The method of item 14486 wherein the combining is performed by constructing all the implant with the agent or the composition.
14721. The method of item 14486 wherein the combining is performed by constructing a portion of the implant with the agent or the composition.
14722. The method of item 14486 wherein the combining is performed by impregnating the implant with the agent or the composition.
14723. The method of item 14486 wherein the combining is performed by constructing all of the implant from a degradable polymer that releases the agent.
14724. The method of item 14486 wherein the combining is performed by constructing a portion of the implant from a degradable polymer that releases the agent.
14725. The method of item 14486 wherein the combining is performed by dipping the implant into a solution that comprise the agent and an inert solvent for the implant.
14726. The method of item 14486 wherein the combining is performed by dipping the implant into a solution that comprises the agent and a solvent that will swell the implant. 14727. The method of item 14486 wherein the combining is performed by dipping the implant into a solution that comprises the agent and a solvent that will dissolve the implant.
14728. The method of item 14486 wherein the combining is performed by dipping the implant into a solution that comprises the agent, a polymer and an inert solvent for the implant.
14729. The method of item 14486 wherein the combining is performed by dipping the implant into a solution that comprises the agent, a polymer and a solvent that will swell the implant.
14730. The method of item 14486 wherein the combining is performed by dipping the implant into a solution that comprises the agent, a polymer and a solvent that will dissolve the implant.
14731. The method of item 14486 wherein the combining is performed by spraying the implant into a solution that comprises the agent and an inert solvent for the implant.
14732. The method of item 14486 wherein the combining is performed by spraying the implant into a solution that comprises the agent and a solvent that will swell the implant.
14733. The method of item 14486 wherein the combining is performed by spraying the implant into a solution that comprises the agent and a solvent that will dissolve the implant.
14734. The method of item 14486 wherein the combining is performed by spraying the implant into a solution that comprises the agent, a polymer and an inert solvent for the implant. 14735. The method of item 14486 wherein the combining is performed by spraying the implant into a solution that comprises the agent, a polymer and a solvent that will swell the implant.
14736. The method of item 14486 wherein the combining is performed by spraying the implant into a solution that comprises the agent, a polymer and a solvent that will dissolve the implant.
14737. A method as in any one of items 14486-14736, wherein the device is a mechanical prosthetic heart valve.
14738. A method as in any one of items 14486-14736, wherein the device is a bioprosthetic heart valve.
14739. A method as in any one of items 14486-14736, wherein the device is an implantable annular ring for receiving a prosthetic heart valve.
14740. A method as in any one of items 14486-14736, wherein the device is a suture ring having an outer peripheral tapered thread for attaching a heart valve prosthesis.
14741. A method as in any one of items 14486-14736, wherein the device is a suture ring for a mechanical heart valve.
14742. A method of making a medical device comprising: combining a penile implant and an anti-scarring agent or a composition comprising an anti-scarring agent, wherein the agent inhibits scarring between the device and a host into which the device is implanted. 14743. The method of item 14742 wherein the agent is an adensosine A2A receptor antagonist.
14744. The method of item 14742 wherein the agent is an AKT inhibitor.
14745. The method of item 14742 wherein the agent is an alpha 2 integrin antagonist, wherein the alpha 2 integrin antagonist is Pharmaprojects No. 5754 (Merck KgaA).
14746. The method of item 14742 wherein the agent is an alpha 4 integrin antagonist.
14747. The method of item 14742 wherein the agent is an alpha 7 nicotinic receptor agonist.
14748. The method of item 14742 wherein the agent is an angiogenesis inhibitor selected from the group consisting of AG-12,958 (Pfizer), ATN-161 (Attention LLC), neovastat, an angiogenesis inhibitor from Jerina AG (Germany), NM-3 (Mercian), VGA-1155 (Taisho), FCE-26644 (Pfizer), FCE-26950 (Pfizer), FPMA (Meiji Daries), FR-111142 (Fujisawa), GGTI-298, GM-1306 (Ligand), GPA-1734 (Novartis), NNC-47-0011 (Novo Nordisk), herbamycin (Nippon Kayaku), lenalidomide (Celegene), 1P-10 (NIH), ABT-828 (Abbott), KIN-841 (Tokushima University, Japan), SF-1126 (Semafore Pharmaceuticals), laminin technology (NIH), CHIR-258 (Chiron), NVP-AEW541 (Novartis), NVP-AEW541 (Novartis), Vt16907 (Alchemia), OXI-8007 (Oxigene), EG-3306 (Ark Therapeutics), Maspin (Arriva), ABT- 567 (Abbott), PPI-2458 (Praecis Pharmaceuticals), CC-5079, CC-4089 (Celgene), HIF-1 alpha inhibitors (Xenova), S-247 (Pfizer), AP-23573 (Ariad), AZD-9935 (Astra Zeneca), mebendazole (Introgen Therapeutics), MetAP-2 inhibitors (GlaxoSmithKline), AG-615 (Angiogene Pharmaceuticals), Tie-2 antagonists (Hybrigenics), NC-381 , CYC-381 , NC-169, NC-219, NC-383, NC-384, NC-407 (Lorus Therapeutics), ATN-224 (Attenuon), ON-01370 (Onconova), Vitronectin antagonists (Amgen), SDX-103 (Salmedix), Vitronectin antagonists (Shire), CHP (Riemser), TEK (Amgen), Anecortave acetate (Alcon), T46.2 (Matrix Therapeutics), HG-2 (Heptagen), TEM antagonists (Genzyme), Oxi-4500 (Oxigene), ATN-161 (Attenuon), WX-293 (Wilex), M-2025 (Metris Therapeutics), Alphastatin (BioActa), YH-16 (Yantai Rongchang), BIBF-1120 (Boehringer Ingelheim), BAY-57-9352 (Bayer), AS- 1404 (Cancer Research Technology), SC-77964 (Pfizer), glycomimetics (BioTie Therapies), TIE-2 Inhibitors (Ontogen), DIMI, Octamer (Octamer), ABR-215050 (Active Biotech), ABT-518 (Abbott), KDR inhibitors (Abbott), BSF-466895 (Abbott), SCH-221153 (Schering-Plough), DAC:antiangiogenic (ConjuChem), TFPI (EntreMed), AZD-2171 (Astra-Zenaca), CDC-394 (Celgene), LY290293 (EIi Lilly), IDN-5390 (Indena), Kdr Kinase Inhibitors (Merck), CT-113020, CT-116433, CT-116563, CT-31890, CT-32228) (Cell Therapeutics), A-299620 (Abbott), TWEAK Inhibitor (Amgen), VEGF modulators (Johnson and Johnson), Tum-N53, tumstatin (Genzyme), Thios- 1 , Thios-2 (Thios Pharmaceuticals), MV-6401 (Miravant Medical Technologies), Spisulosine (PharmaMar), CEP-7055 (Cephalon), AUV-201 (Auvation), LM-609 (EIi Lilly), SKF-106615 (AnorMED), Oglufanide disodium (Cytran), BW-114 (Pharminox), Calreticulin (NIH), WX-678 (Wilex), SD-7784 (Pfizer), WX-UK1 (Wilex), SH-268 (Schering AG), 2-Me-PGA (Celgene), S- 137 (Pfizer), ZD-6126 (Angiogene Pharmaceuticals), SG-292 (SignalGen), Benefin (Lane Labs), A6, A36 (Angstrom), SB-2723005 (GlaxoSmithKline), SC-7 (Cell Therapeutics), ZEN-014 (AEterna Zentaris), 2-methoxyestradiol (EntreMed), NK-130119 (Nippon Kayaku), CC-10004 (Celgene), AVE- 8062A (Ajinomoto), Tacedinaline (Pfizer), Actinonin (Tokyo Metropolitan Institute of Medical Science), Lenalidomide (Celgene), VGA-1155, BTO-956 (SRI International), ER-68203-00 (Eisai), CT-6685 (UCB), JKC-362 (Phoenix Pharmaceuticals), DMl-3798 (DMI Biosciences, Angiomate (Ipsen), ZD-6474 (AstraZeneca), CEP-5214 (Cephalon), Canstatin (Genzyme), NM-3 (Mercian), Oridigm (MediQuest Therapeutics), Exherin (Adherex), BLS-0597 (Boston Life Sciences), PTC-299 (PTC Therapeutics), NPI-2358 (Nereus Pharmaceuticals), CGP-79787 (Novartis), AEE-788 (Novartis), CKD-732 (Chong Kun Dang), CP-564959 (OSI Pharmaceuticals), CM-101 (CarboMed), CT-2584, CT3501 (Cell Therapeutics), combretastatin and analogues and derivatives thereof (Oxigene), Rebimastat (Bristol-Meyers Squibb), Dextrin 2-sulfate (ML Laboratories), Cilengitide (Merk KGaA), NSC-706704 (Pharminox), KRN- 951 (Kirin Brewery), Ukrain, NSC-631570 (Nowicky Pharma), Tecogalan sodium (Daiichi Pharmaceutical), Tz-93 (Tsumura), TBC-1635 (Encysive Pharmaceuticals), TAN-1120 (Takeda), Semaxanib (Pfizer), BDI-7800 (Biopharmacopae), SD-186, SD-983 (Bristol-Meyers Squibb), SB-223245 (GlaxoSmithKline), SC-236 (Pfizer), RWJ-590973 (Johnson and Johnson), ILX-1850 (Genzyme), SC-68488, S-836 (Pfizer), CG-55069-11 (CuraGen), Ki-23057 (Kirin Brewery), CCX-700 (Chemoentryx), Pegaptanib octasodium (Giled Sciences), ANGIOCOL (available from Biostratum Inc.), or an analogue or derivative thereof.
14749. The method of item 14742 wherein the agent is an apoptosis antagonist.
14750. The method of item 14742 wherein the agent is an apoptosis activator.
14751. The method of item 14742 wherein the agent is a beta 1 integrin antagonist.
14752. The method of item 14742 wherein the agent is a beta tubulin inhibitor. 14753. The method of item 14742 wherein the agent is a blocker of enzyme production in Hepatitis C.
14754. The method of item 14742 wherein the agent is a Bruton's tyrosine kinase inhibitor.
14755. The method of item 14742 wherein the agent is a calcineurin inhibitor.
14756. The method of item 14742 wherein the agent is a caspase 3 inhibitor.
14757. The method of item 14742 wherein the agent is a CC chemokine receptor antagonist.
14758. The method of item 14742 wherein the agent is a cell cycle inhibitor selected from the group consisting of SNS-595 (Sunesis), synthadotin, KRX-0403, and an analogue or derivative thereof.
14759. The method of item 14742 wherein the agent is a cathepsin B inhibitor.
14760. The method of item 14742 wherein the agent is a cathepsin K inhibitor, wherein the cathepsin K inhibitor is 462795 (GlaxoSmithKline), INPL-022-D6 (Amura Therapeutics), or an analogue or derivative thereof.
14761. The method of item 14742 wherein the agent is a cathepsin L inhibitor. 14762. The method of item 14742 wherein the agent is a CD40 antagonist.
14763. The method of item 14742 wherein the agent is a chemokine receptor agonist.
14764. The method of item 14742 wherein the agent is a chymase inhibitor.
14765. The method of item 14742 wherein the agent is a collagenase antagonist.
14766. The method of item 14742 wherein the agent is a CXCR antagonist.
14767. The method of item 14742 wherein the agent is a cyclin dependent kinase inhibitor selected from the group consisting of a CDK-1 inhibitor, a CDK-2 inhibitor, a CDK- 4 inhibitor, a CDK-6 inhibitor, a CAK1 inhibitor from GPC Biotech and Bristol-Myers Squibb, RGB-286199 (GPC Biotech), an anticancer agent from Astex Technology, a CAK1 inhibitor from GPC Biotech, a CDK inhibitor from Sanofi-Aventis, a CDK1/CDK2 inhibitor from Amgen, a CDK2 inhibitor from SUGEN-2 (Pfizer), a hearing loss therapy agent (Sound Pharmaceuticals), PD-0332991 (Pfizer), RGB-286199 (GPC Biotech), Ro-0505124 (Hoffmann-La Roche), a Ser/Thr kinase inhibitor from Lilly (EIi Lilly), CVT-2584 (CAS No. 199986-75- 9) (CV Therapeutics), and an analogue or derivative thereof.
14768. The method of item 14742 wherein the agent is a cyclooxygenase 1 inhibitor. 14769. The method of item 14742 wherein the agent is a DHFR inhibitor.
14770. The method of item 14742 wherein the agent is a dual integrin inhibitor.
14771. The method of item 14742 wherein the agent is an elastase inhibitor.
14772. The method of item 14742 wherein the agent is an elongation factor-1 alpha inhibitor.
14773. The method of item 14742 wherein the agent is an endothelial growth factor antagonist.
14774. The method of item 14742 wherein the agent is an endothelial growth factor receptor kinase inhibitor selected from the group consisting of sorafenib tosylate (Bayer), AAL-993 (Novartis), ABP-309 (Novartis), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), E- 7080 (Eisai), EG-3306 (Ark Therapeutics), EXEL-2880 (Exelixis), GW- 654652 (GlaxoSmithKline), a KDR inhibitor from LG Life Sciences, CT-6685 and CT-6729 (UCB), KRN-633 and KRN-951 (Kirin Brewery), OSI-930 (OSI Pharmaceuticals), SP-5.2 (Supratek Pharma), SU-11657 (Pfizer), a Tie-2 antagonist (Hybrigenics), a VEGFR-2 kinase inhibitor (Bristol-Myers Squibb), XL-647 (Exelixis), a KDR inhibitor from Abbott Laboratories, sorafenib tosylate, and an analogue or derivative thereof.
14775. The method of item 14742 wherein the agent is an endotoxin antagonist. 14776. The method of item 14742 wherein the agent is an epothilone and tubulin binder.
14777. The method of item 14742 wherein the agent is an estrogen receptor antagonist.
14778. The method of item 14742 wherein the agent is an FGF inhibitor.
14779. The method of item 14742 wherein the agent is a famexyl transferase inhibitor.
14780. The method of item 14742 wherein the agent is farnesyltransferase inhibitor selected from the group of A-197574 (Abbott), a farnesyltransferase inhibitor from Servier, FPTIII (Strathclyde Institute for Drug R), LB-42908 (LG Life Sciences), Pharmaprojects No. 5063 (Genzyme), Pharmaprojects No. 5597 (Ipsen), Yissum Project No. B-1055 (Yissum), and an analogue or derivative thereof.
14781. The method of item 14742 wherein the agent is an FLT-3 kinase inhibitor.
14782. The method of item 14742 wherein the agent is an FGF receptor kinase inhibitor.
14783. The method of item 14742 wherein the agent is a fibrinogen antagonist selected from the group consisting of AUV-201 (Auvation), MG-13926 (Sanofi-Aventis), plasminogen activator (CAS No. 105913-11-9) (from Sanofi-Aventis or UCB), plasminogen activator-2 (tPA- 2) (Sanofi-Aventis), pro-urokinase (CAS No. 82657-92-9) (Sanofi-Aventis), and an analogue or derivative thereof. 14784. The method of item 14742 wherein the agent is a heat shock protein 90 antagonist selected from the group consisting of SRN- 005 (Sirenade), geldanamycin, NSC-33050 (17-allylaminogeldanamycin; 17- AAG), ^-dimethylaminoethylamino-^-demethoxy-geldanamycin (17- DMAG), rifabutin (rifamycin XIV, 1',4-didehydro-1-deoxy-1 ,4-dihydro-5'-(2- methylp ropy I)-I-OXo-), and an analogue or derivative thereof.
14785. The method of item 14742 wherein the agent is a histone deacetylase inhibitor.
14786. The method of item 14742 wherein the agent is an HMGCoA reductase inhibitor selected from the group consisting of an atherosclerosis therapeutic from Lipid Sciences, ATI-16000 (ARYx Therapeutics), KS-01-019 (Kos Pharmaceuticals), Pharmaprojects No. 2197 (Sanofi-Aventi), RP 61969 (Sanofi-Aventis), and an analogue or derivative thereof,
14787. The method of item 14742 wherein the agent is an ICAM inhibitor.
14788. The method of item 14742 wherein the agent is an IL, ICE and IRAK antagonist, wherein the antagonist is a CJ-14877, CP- 424174 (Pfizer), NF-61 (Negma-Lerads), and an analogue or derivative thereof.
14789. The method of item 14742 wherein the agent is an IL-2 inhibitor.
14790. The method of item 14742 wherein the agent is an immunosuppressant selected from the group consisting of teriflunomide (Sanofi Aventis), chlorsulfaquinoxalone (NSC-339004), chlorsulfaquinoxalone sulfate, CS-712 (Sankyo), ismomultin alfa (CAS No. 457913-93-8) (Akzo Nobel), antiallergics from GenPat77, antiinflammatories or AT-005 (Androclus Therapeutics), autoimmune disease therapeutics from EpiVax, BN-007 (Bone), budesonide (CAS No. 51333-22- 3) (MAP Pharmaceuticals), CO-14 (Genzyme), edratide (CAS No. 433922- 67-9) (Teva), EP-314 (Enanta), eprovafen (CAS No. 101335-99-3) (Sanofi- Aventis), HWA-131 (CAS No. 118788-41-3) (Sanofi-Aventis), immunomodulators from MerLion Pharmaceuticals, immunosuppressives from Alchemia, IPL-12 (Inflazyme), MDL-9563 (CAS No. 27086-86-8) (Sanofi-Aventis), Pharmaprojects No. 2330 (Sanofi-Aventis), Pharmaprojects No. 6426 (Abgenix), PXS-25 (Pharmaxis), rosmarinic acid (CAS No. 20283-92-5) (Sanofi-Aventis), RP 42927 or RP 54745 (CAS No. 135330-08-4) (Sanofi-Aventis), SGN-35 (Seattle Genetics), ST-1959 (Sigma-Tau), type I diabetes therapy from SYNX Pharma, UNIL-88 (Debiopharm), VP-025 (Vasogen), VR-694 (Vectura), PRTX-001 (Protalex), and an analogue or derivative thereof.
14791. The method of item 14742 wherein the agent is an IMPDH (inosine monophosphate).
14792. The method of item 14742 wherein the agent is an integrin antagonist.
14793. The method of item 14742 wherein the agent is an interleukin antagonist.
14794. The method of item 14742 wherein the agent is an inhibitor of type III receptor tyrosine kinase.
14795. The method of item 14742 wherein the agent is an irreversible inhibitor of enzyme methionine aminopeptidase type 2. 14796. The method of item 14742 wherein the agent is an isozyme selective delta protein kinase C inhibitor.
14797. The method of item 14742 wherein the agent a JAK3 enzyme inhibitor.
14798. The method of item 14742 wherein the agent is a
JNK inhibitor.
14799. The method of item 14742 wherein the agent is a kinase inhibitor.
14800. The method of item 14742 wherein the agent is kinesin antagonist.
14801. The method of item 14742 wherein the agent is a kinesin antagonist.
14802. The method of item 14742 wherein the agent is a leukotriene inhibitor and antagonist selected from the group consisting of ambicromil (CAS No. 58805-38-2) (Sanofi-Aventis), amelubant (CAS No. 346735-24-8) (Boehringer Ingelheim), DW-1141 (Dong Wha), ebselen (Daiichi Pharmaceutical), ibudilast (Kyorin), leucotriene inhibitors from Sanofi-Aventis, lymphotoxin-beta receptor (LT-D) from Biogen Idee, Pharmaprojects No. 1535 and 2728 (CAS No. 119340-33-9) (Sanofi- Aventis), R-112 (Rigel), Rev-5367 (CAS No. 92532-05-3) (Sanofi-Aventis), RG-14893 (CAS No. 141835-49-6) (Sanofi-Aventis), RG-5901-A (CAS No. 101910-24-1), 92532-23-5, RP 66153 (CAS No. 142422-79-5), RP 66364 (CAS No.186916-92-5), RP 69698 (CAS No. 141748-00-7) (Sanofi-Aventis), SC-411930 (Pfizer), SC-41930 (CAS No. 120072-59-5) (Pfizer), SC-50605 (CAS No. 138828-39-4) (Pfizer), SC-51146 (CAS No. 141059-52-1), SC- 53228 (CAS No. 153633-01-3) (Pfizer), spaglumic acid (ZY-15106) (CAS No.3106-85-2), 80619-64-3 (Novartis), tipredane (CAS No. 85197-77-9) (Bristol-Myers Squibb), U-75302 (CAS No. 119477-85-9) (Pfizer), and analogue or derivative thereof.
14803. The method of item 14742 wherein the agent is an MAP kinase inhibitor.
14804. The method of item 14742 wherein the agent is a matrix metalloproteinase inhibitor.
14805. The method of item 14742 wherein the agent is an MCP-CCR2 inhibitor.
14806. The method of item 14742 wherein the agent is an mTOR inhibitor.
14807. The method of item 14742 wherein the agent is an mTOR kinase inhibitor.
14808. The method of item 14742 wherein the agent is a microtubule inhibitor selected from the group consisting of antibody- maytansinoid conjugates from Biogen Idee, colchicines (MantiCore Pharmaceuticals), anticancer immunoconjugates from Johnson & Johnson, DIME from Octamer, gni-1f (GNI), huC242-DM4, huMy9-6-DM1 (ImmunoGen), IDN-5404 (Indena), IMO-098, IMOderm (Imotep), mebendazole (Introgen Therapeutics), microtubule poisons from Cambridge Enterprise, paclitaxel such as LOTAX from Aphios (CAS No. 33069-62-4), Genexol-PM from Samyang, Pharmaprojects No. 6383 (Tapestry Pharmaceuticals), RPR-112378 (Sanofi-Aventis), SGN-75 (Seattle Genetics), SPL-7435 (Starpharma), SSR-250411 (Sanofi-Aventis), trastuzumab-DM1 (Genentech), vinorelbine, and an analogue or derivative thereof.
14809. The method of item 14742 wherein the agent is an MIF inhibitor.
14810. The method of item 14742 wherein the agent is an MMP inhibitor.
14811. The method of item 14742 wherein the agent is a neurokinin (NK) antagonist selected from the group consisting of anthrotainin (CAS No. 148084-40-6) (Sanofi-Aventis), an IBS thereapeutic from ArQuIe, MDL-105212A (CAS No. 167261-60-1) (Ssanofi-Aventis), Pharmaprojects No. 2744, 3258 (CAS No. 139167-47-8) 4006, 4201, or 5986 (Sanofi-Aventis), RP 67580 (CAS No. 135911-02-3), SR-144190 (CAS No. 201152-86-5), SSR-240600, SSR-241586 (Sanofi-Aventis), TKA-457 (Novartis), vestipitant mesylate (CAS No. 334476-64-1) (GlaxoSmithKline), Win-64821 (Sanofi-Aventis), PRX-96026 (Predix Pharmaceuticals), and an analogue or derivative thereof.
14812. The method of item 14742 wherein the agent is an NF kappa B inhibitor selected from the group consisting of AVE-0545 or AVE-0547 (Sanofi-Aventis), bortezomib (CAS No. 179324-69-7) (Millennium Pharmaceuticals), dexanabinol (CAS No. 112924-45-5) (Pharmos), dexlipotam (Viatris), Pharmaprojects No. 6283 (INDRA) (OXiGENE), IPL- 576092 (CAS No. 137571-30-3) (Inflazyme), NFKB decoy (Corgentech), NFKB decoy oligo (AnGes MG), NFKB's from Ariad, osteoporosis treatments or S5 (F005) from Fulcrum Pharmaceuticals, P61 (Phytopharm), R-flurbiprofen (CAS No. 5104-49-4) (Encore Pharmaceuticals), and an analogue or derivative thereof. 14813. The method of item 14742 wherein the agent is a nitric oxide agonist.
14814. The method of item 14742 wherein the agent is an ornithine decarboxylase inhibitor.
14815. The method of item 14742 wherein the agent is a p38 MAP kinase inhibitor selected from the group consisting of AZD-6703 (AstraZeneca), JX-401 (Jexys Pharmaceuticals), BMS-2 (Bristol-Myers Squibb), a p38 MAP kinase inhibitor from Novartis, a p38-alpha MAP kinase inhibitor from Amphora, Pharmaprojects No. 5704 (Pharmacopeia), RPR- 200765A (Sanofi-Aventis), SD-282 (Johnson & Johnson), TAK-715 (Takeda), and an analogue or derivative thereof.
14816. The method of item 14742 wherein the agent is a palmitoyl-protein thioesterase inhibitor.
14817. The method of item 14742 wherein the agent is a PDGF receptor kinase inhibitor selected from the group consisting of AAL- 993, AMN-107, or ABP-309 (Novartis), AMG-706 (Amgen), BAY-57-9352 (Bayer), CDP-860 (UCB), E-7080 (Eisai), imatinib (CAS No. 152459-95-5) (Novartis), OSI-930 (OSI Pharmaceuticals), RPR-127963E (Sanofi-Aventis), RWJ-540973 (Johnson & Johnson), sorafenib tosylate (Bayer), SU-11657 (Pfizer), tandutinib (CAS No. 387867-13-2) (Millennium Pharmaceuticals), vatalanib (Novartis), ZK-CDK (Schering AG), and an analogue or derivative thereof.
14818. The method of item 14742 wherein the agent is (-)-halofenate (Metabolex), AMG-131 (Amgen), antidiabetics from Japan Tobacco, AZD-4619, AZD-8450, AZD-8677 (AstraZeneca), DRF-10945, balaglitazone (Dr Reddy's), CS-00088, CS-00098 (Chipscreen Biosciences), E-3030 (Eisai), etalocib (CAS No. 161172-51-6) (EIi Lilly), GSK-641597 (Ligand), GSK-677954 (GlaxoSmithKline), GW-409544 (Ligand), GW- 590735 (GlaxoSmithKline), K-111 (Hoffmann-La Roche), LY-518674 (EIi Lilly), LY-674 (Ligand), LY-929 (Ligand), MC-3001, MC-3002 (MaxoCore Pharmaceuticals), metformin HCI + pioglitazone (CAS No. 1115-70-4 and 112529-15-4), ACTOPLUS MET from Andrx), muraglitazar (CAS No. 331741-94-7) (Bristol-Myers Squibb), naveglitazar (Ligand), oleoylethanolamide (Kadmus Pharmaceuticals), ONO-5129, pioglitazone hydrochloride (CAS No. 111025-46-8 and 112529-15-4) (Takeda), PLX-204 (Plexxikon), PPAR agonists from Genfit, PPAR delta agonists from EIi Lilly, PPAR-alpha agonists from CrystalGenomics, PPAR-gamma modulators and PPAR-β modulators from CareX, rosiglitazone maleate (CAS No. 122324- 73-4 or 155141-29-0) (GlaxoSmithKline), rosiglitazone maleate/glimepir (CAS No. 155141-29-0 and 93479-97-1), AVANDARYL, rosiglitazone maleate/metformin extend (CAS No. 155141-29-0 and 657-24-9), AVANDAMET, rosiglitazone maleate+metformin, AVANDAMET (GlaxoSmithKline), tesaglitazar (AstraZeneca), and an analogue or derivative thereof.
14819. The method of item 14742 wherein the agent is a phosphatase inhibitor.
14820. The method of item 14742 wherein the agent is a phosphodiesterase (PDE) inhibitor selected from the group consisting of avanafil (Tanabe Seiyaku), dasantafil (CAS No. 569351-91-3) (Schering- Plough), A-906119 (CAS No. 134072-58-5), DL-850 (Sanofi-Aventis), GRC- 3015, GRC-3566, GRC-3886 (Glenmark), HWA-153 (CAS No. 56395-66-5) (Sanofi-Aventis), hydroxypumafentrine (Altana), IBFB-130011 , IBFB-14-016, 1BFB-140301 , IBFB-150007, IBFB-211913 (IBFB Pharma), L-826141 (Merck & Co), medorinone (CAS No. 88296-61-1) (Sanofi-Aventis), MEM- 1917 (Memory Pharmaceuticals), ND-1251 (Neuro3d), PDE inhibitors from ICOS, PDE IV inhibitors from Memory Pharmaceuticals and CrystalGenomics, Pharmaprojects No. 2742 and 6141 (Sanofi-Aventis), QAD-171 (Novartis), RHC-2963 (CAS No. 76993-12-9 and 76993-14-1), RPR-117658, RPR-122818 derivatives, SR-24870 , and RPR-132294 (Sanofi-Aventis), SK-350 (ln2Gen), stroke therapy agents from deCODE Genetics, TAS-203 (Taiho), tofimilast (CAS No. 185954-27-2) (Pfizer), UK- 371800 (Pfizer), WIN-65579 (CAS No. 158020-82-7) (Sanofi-Aventis), IBFB- 130020 (IBFB Pharma), OPC-6535 (CAS No. 145739-56-6) (Otsuka), a phosphodiesterase III inhibitor, enoximone, a phosphodiesterase IV inhibitor, fosfosal, Atopik (Barrier Therapeutics), triflusal, a phosphodiesterase V inhibitor, and an analogue or derivative thereof.
14821. The method of item 14742 wherein the agent is a PKC inhibitor.
14822. The method of item 14742 wherein the agent is a platelet activating factor antagonist.
14823. The method of item 14742 wherein the agent is a platelet-derived growth factor receptor kinase inhibitor.
14824. The method of item 14742 wherein the agent is a prolyl hydroxylase inhibitor.
14825. The method of item 14742 wherein the agent is a polymorphonuclear neutrophil inhibitor.
14826. The method of item 14742 wherein the agent is a protein kinase B inhibitor. 14827. The method of item 14742 wherein the agent is a protein kinase C stimulant.
14828. The method of item 14742 wherein the agent is a purine nucleoside analogue.
14829. The method of item 14742 wherein the agent is a purinoreceptor P2X antagonist.
14830. The method of item 14742 wherein the agent is a Raf kinase inhibitor.
14831. The method of item 14742 wherein the agent is a reversible inhibitor of ErbB1 and ErbB2.
14832. The method of item 14742 wherein, the agent is a ribonucleoside triphosphate reductase inhibitor.
14833. The method of item 14742 wherein the agent is an SDF-1 antagonist.
14834. The method of item 14742 wherein the agent is a sheddase inhibitor.
14835. The method of item 14742 wherein the agent is an SRC inhibitor.
14836. The method of item 14742 wherein the agent is a stromelysin inhibitor. 14837. The method of item 14742 wherein the agent is an Syk kinase inhibitor.
14838. The method of item 14742 wherein the agent is a telomerase inhibitor.
14839. The method of item 14742 wherein the agent is a TGF beta inhibitor selected from the group consisting of pirfenidone (CAS No. 53179-13-8) (MARNAC), tranilast (CAS No. 53902-12-8) (Kissei), IN-
1130 (ln2Gen), mannose-6-phosphate (BTG), TGF-β antagonists from Inflazyme (Pharmaprojects No. 6075), TGF-β antagonists from Sydney, non-industrial source), TGF-β I receptor kinase inhibitors from EIi Lilly, TGF- β receptor inhibitors from Johnson & Johnson, and an analogue or derivative thereof.
14840. The method of item 14742 wherein the agent is a TNFα antagonist or TACE inhibitor selected from the group consisting of adalimumab (CAS No. 331731-18-1) (Cambridge Antibody Technology), AGIX-4207 (AtheroGenics), AGT-1 (Advanced Biotherapy), an antiinflammatory from Borean Pharma, Cellzome, or Paradigm Therapeutics, anti-inflammatory vaccine (TNF-alpha kinoid) from Neovacs, humanized anti-TNF antibody or an anti-TNF MAb (CB0006) Celltech (UCB), apratastat (CAS No. 287445-51-0) (Wyeth), BMS-561392 (Bristol-Myers Squibb), BN- 006 (Bone), certolizumab pegol (CAS No. 428863-50-7 or CH-138 (UCB), ciiomilast (CAS No. 153259-65-5) (GlaxoSmithKline), CR-1 (Nuada Pharmaceuticals), CRx-119 (CombinatoRx), D-5410 (UCB), dacopafant (CAS No. 125372-33-0) (Sanofi-Aventis), dersalazine (CAS No. 188913-57- 7/188913-58-8) (Uriach), etanercept (CAS No. 185243-69-0) (Amgen), ethyl pyruvate (Critical (Critical Therapeutics), golimumab (CAS No. 476181-74-5) (Johnson & Johnson), hormono-immunotherapy from Ipsen, CDP571 (e.g., Humicade from UCB), IC-485 (ICOS), infliximab (CAS No. 170277-31-3) (Johnson & Johnson), IP-751 (Manhattan Pharmaceuticals), ISIS-104838 (CAS No. 250755-32-9) (ISIS Pharmaceuticals), lenalidomide (CAS No. 191732-72-6) (Celgene), lentinan (CAS No. 37339-90-5) (Ajinomoto), MDL- 201112 (CAS No. 142130-73-2) (Sanofi-Aventis), medroxyprogesterone (CAS No. 520-85-4) (InKine Pharmaceutical), N-acetylcysteine (CAS No. 616-91-1) (Zambon), NBE-P2 (DIREVO Biotech), nerelimomab (CAS No. 162774-06-3) (Chiron), OM-294DP (OM PHARMA), onercept (CAS No. 199685-57-9) (Yeda), PASSTNF-alpha (Verigen), pentoxifylline or oxypentifylline (Sanofi-Aventis), Pharmaprojects No. 4091 , 4241, 4295, or 4488 (Sanofi-Aventis), Pharmaprojects No. 5480 (Amgen), Pharmaprojects No. 6749 (Cengent), pirfenidone (CAS No. 53179-13-8) (MARNAC), RPR- 132294 (Sanofi-Aventis), S5 (F002) (Fulcrum Pharmaceuticals), simvastatin (CAS No. 79902-63-9) (Merck & Co), STA-6292 (Synta Pharmaceuticals), tacrolimus (CAS No. 104987-11-3) (Fujisawa LifeCycle Pharma), talactoferrin alfa (CAS No. 308240-58-6) (Agennix), thalidomide (CAS No. 50-35-1) (Celgene), TNF antagonists form ProStrakan, and Synergen, TNF inhibitors (Amgen), TNF-alpha antagonists from Dynavax Technologies and Jerina AG (Germany), TNF-alpha inhibitors from IBFB Pharma and Xencor (Xencor), torbafylline (CAS No. 105102-21-4) (Sanofi-Aventis), UR-1505 (Uriach), VT-346 (Viron Therapeutics), YSIL6 (Y1S Therapeutics), YSTH2 (Y's Therapeutics), NPI-1302a-3 (Nereus Pharmaceuticals, a TNF antagonist from Jerina AG (Germany), dersalazine, and an analogue or derivative thereof.
14841. The method of item 14742 wherein the agent is a Toll receptor inhibitor.
14842. The method of item 14742 wherein the agent is a tubulin antagonist. 14843. The method of item 14742 wherein the agent is a tyrosine kinase inhibitor selected from the group consisting of SU-011248, SUTENT from Pfizer Inc. (New York, NY), BMS-354825, PN-355 (Paracelsian Pharmaceuticals), AGN-199659 (Allergan), AAL-993 or ABP- 309 (Novartis), adaphostin (NIH), AEE-788 (Novartis), AG-013736 (OSI Pharmaceuticals), AG-13736 (Pfizer), ALT-110 (Alteris Therapeutics), AMG- 706 (Amgen), anticancer MAbs from Xencor, anti-EGFrvlll MAbs from Abgenix, anti-HER2 MAb from Abiogen, AZD-2171 or AZD-9935 (AstraZeneca), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), CEP-5214 (Cephalon), CEP-7055 (Cephalon), cetuximab (ImClone Systems), CHIR-200131 and CHIR-258 (Chiron), CP-547632 (OSI Pharmaceuticals), CP-724714 (Pfizer), CT-301 (Creabilis Therapeutics), D- 69491 (Baxter International), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EGFR/ErbB2 inhibitors from Array BioPharma, erlotinib (CAS No. 183319- 69-9) (OSI Pharmaceuticals), EXEL-2880 (Exelixis), FK-778 (Sanofi- Aventis) , gefitinib (CAS No. 184475-35-2) (AstraZeneca), GW-2286 or GW- 654652 (GlaxoSmithKline), her2/neu antigen from AlphaVax, HER-2/neu inhibitor from Generex, Herzyme (Medipad) (Sirna Therapeutics), HKI-272 (Wyeth), HuMax-EGFr (Genmab), idronoxil (CAS No. 81267-65-4) (Novogen), IGF-1 inhibitors from Ontogen, IMC-11 F8 (ImClone Systems), kahalalide F (CAS No. 149204-42-2) (PharmaMar), KDR inhibitor from LG Life Sciences, KDR inhibitors from Abbott Laboratories, KDR kinase inhibitors (UCB), Kdr kinase inhibitors from Merck & Co, KRN-633 and KRN- 951 (Kirin Brewery), KSB-102 (Xenova), lapatinib ditosylate (CAS No. 388082-78-8) (GlaxoSmithKline), matuzumab (Merck KGaA), MDX-214 (Medarex), ME-103 (Pharmexa), MED-A300 (Gerolymatos), MNAC-13 (Lay Line Genomics), nimotuzumab (Center of Molecular Immunology), NSC- 330507 or NSC-707545 (NIH), NV-50 (Novogen), OSI-930 (OSI Pharmaceuticals), panitumumab (Abgenix), pelitinib (CAS No. 287933-82-7) (Wyeth), pertuzumab (CAS No. 380610-27-5) (Genentech), Pharmaprojects No. 3985 (Sanofi-Aventis), prostate cancer therapeutics from Sequenom (SQPC35, SQPC36, SQPC90), removab and remoxab (Trion Pharma), RG- 13022 (CAS No. 136831-48-6), RG-13291 (CAS No. 138989-50-1), or RG- 14620 (CAS No. 136831-49-7) (Sanofi-Aventis), RM-6427 (Romark), RNAi breast cancer therapy from Benitec, RP 53801 (CAS No. 125882-88-4) (Sanofi-Aventis), sorafenib tosylate (Bayer), SU-11657 (Pfizer), Tie-2 antagonists from Semaia (Hybrigenics), Tie-2 inhibitors from Ontogen, trastuzumab (CAS No. 180288-69-1) (Genentech), tyrosine kinase inhibitors from Sanofi-Aventis, U3-1287, U3-1565, U3-1784, U3-1800 (U3 Pharma), vatalanib (Novartis), VEGFR-2 kinase inhibitor from Bristol-Myers Squibb, XL-647 (Exelixis), ZD-6474 (AstraZeneca), ZK-CDK (Schering AG), an EGFR tyrosine kinase inhibitor, EKB-569 (Wyeth), and an analogue or derivative thereof.
14844. The method of item 14742 wherein the agent is a VEGF inhibitor.
14845. The method of item 14742 wherein the agent is a vitamin D receptor agonist.
14846. The method of item 14742 wherein the agent is ZD-6474 (an angiogenesis inhibitor).
14847. The method of item 14742 wherein the agent is AP-23573 (an mTOR inhibitor).
14848. The method of item 14742 wherein the agent is synthadotin (a tubulin antagonist).
14849. The method of item 14742 wherein the agent is S-0885 (a collagenase inhibitor). 14850. The method of item 14742 wherein the agent is aplidine (an elongation factor-1 alpha inhibitor).
14851. The method of item 14742 wherein the agent is ixabepilone (an epithilone).
14852. The method of item 14742 wherein the agent is IDN-5390 (an angiogenesis inhibitor).
14853. The method of item 14742 wherein the agent is SB-2723005 (an angiogenesis inhibitor).
14854. The method of item 14742 wherein the agent is ABT-518 (an angiogenesis inhibitor).
14855. The method of item 14742 wherein the agent is combretastatin (an angiogenesis inhibitor).
14856. The method of item 14742 wherein the agent is anecortave acetate (an angiogenesis inhibitor).
14857. The method of item 14742 wherein the agent is SB-715992 (a kinesin antagonist).
14858. The method of item 14742 wherein the agent is temsirolimus (an mTOR inhibitor).
14859. The method of item 14742 wherein the agent is adalimumab (a TNFα antagonist). 14860. The method of item 14742, wherein the composition comprises a polymer.
14861. The method of item 14742, wherein the composition comprises a polymeric carrier.
14862. The method of item 14742 wherein the anti- scarring agent inhibits adhesion between the device and a host into which the device is implanted.
14863. The method of item 14742 wherein the device delivers the anti-scarring agent locally to tissue proximate to the device.
14864. The method of item 14742 wherein the device has a coating that comprises the anti-scarring agent.
14865. The method of item 14742, wherein the device has a coating that comprises the agent and is disposed on a surface of the implant.
14866. The method of item 14742, wherein the device has a coating that comprises the agent and directly contacts the implant.
14867. The method of item 14742, wherein the device has a coating that comprises the agent and indirectly contacts the implant.
14868. The method of item 14742, wherein the device has a coating that comprises the agent and partially covers the implant.
14869. The method of item 14742, wherein the device has a coating that comprises the agent and completely covers the implant. 14870. The method of item 14742, wherein the device has a uniform coating.
14871. The method of item 14742, wherein the device has a non-uniform coating.
14872. The method of item 14742, wherein the device has a discontinuous coating.
14873. The method of item 14742, wherein the device has a patterned coating.
14874. The method of item 14742, wherein the device has a coating with a thickness of 100 Dm or less.
14875. The method of item 14742, wherein the device has a coating with a thickness of 10 Dm or less.
14876. The method of item 14742, wherein the device has a coating, and the coating adheres to the surface of the implant upon deployment of the implant.
14877. The method of item 14742, wherein the device has a coating, and wherein the coating is stable at room temperature for a period of 1 year.
14878. The method of item 14742, wherein the device has a coating, and wherein the anti-scarring agent is present in the coating in an amount ranging between about 0.0001% to about 1% by weight. 14879. The method of item 14742, wherein the device has a coating, and wherein the anti-scarring agent is present in the coating in an amount ranging between about 1% to about 10% by weight.
14880. The method of item 14742, wherein the device has a coating, and wherein the anti-scarring agent is present in the coating in an amount ranging between about 10% to about 25% by weight.
14881. The method of item 14742, wherein the device has a coating, and wherein the anti-scarring agent is present in the coating in an amount ranging between about 25% to about 70% by weight.
14882. The method of item 14742, wherein the device has a coating, and wherein the coating further comprises a polymer.
14883. The method of item 14742, wherein the device .- has a first coating having a first composition and a second coating having a second composition.
14884. The method of item 14742, wherein the device has a first coating having a first composition and a second coating having a second composition, wherein the first composition and the second composition are different.
14885. The method of item 14742, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a copolymer.
14886. The method of item 14742, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a block copolymer. 14887. The method of item 14742, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a random copolymer.
14888. The method of item 14742, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a biodegradable polymer.
14889. The method of item 14742, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a non-biodegradable polymer.
14890. The method of item 14742, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a hydrophilic polymer.
14891. The method of item 14742, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a hydrophobic polymer.
14892. The method of item 14742, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a polymer having hydrophilic domains.
14893. The method of item 14742, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a polymer having hydrophobic domains.
14894. The method of item 14742, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a non-conductive polymer. 14895. The method of item 14742, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises an elastomer.
14896. The method of item 14742, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a hydrogel.
14897. The method of item 14742, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a silicone polymer.
14898. The method of item 14742, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a hydrocarbon polymer.
14899. The method of item 14742, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a styrene-derived polymer.
14900. The method of item 14742, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a butadiene polymer.
14901. The method of item 14742, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a macromer.
14902. The method of item 14742, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a poly(ethylene glycol) polymer. 14903. The method of item 14742 wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises an amorphous polymer.
14904. The method of item 14742, wherein the device comprises a lubricious coating.
14905. The method of item 14742 wherein the anti- scarring agent is located within pores or holes of the device.
14906. The method of item 14742 wherein the anti- scarring agent is located within a channel, lumen, or divet of the device.
14907. The method of item 14742, wherein the device comprises a second pharmaceutically active agent.
14908. The method of item 14742 wherein the device comprises an anti-inflammatory agent.
14909. The method of item 14742 wherein the device comprises an agent that inhibits infection.
14910. The method of item 14742 wherein the device comprises an agent that inhibits infection, and wherein the agent is an anthracycline.
14911. The method of item 14742 wherein the device comprises an agent that inhibits infection, and wherein the agent is doxorubicin. 14912. The method of item 14742 wherein the device comprises an agent that inhibits infection, and wherein the agent is mitoxantrone.
14913. The method of item 14742 wherein the device comprises an agent that inhibits infection, and wherein the agent is a fluoropyrimidine.
14914. The method of item 14742 wherein the device comprises an agent that inhibits infection, and wherein the agent is 5- fluorouracil (5-FU).
14915. The method of item 14742 wherein the device comprises an agent that inhibits infection, and wherein the agent is a folic acid antagonist.
14916. The method of item 14742 wherein the device comprises an agent that inhibits infection, and wherein the agent is methotrexate.
14917. The method of item 14742 wherein the device comprises an agent that inhibits infection, and wherein the agent is a podophylotoxin.
14918. The method of item 14742 wherein the device comprises an agent that inhibits infection, and wherein the agent is etoposide.
14919. The method of item 14742 wherein the device comprises an agent that inhibits infection, and wherein the agent is a camptothecin. 14920. The method of item 14742 wherein the device comprises an agent that inhibits infection, and wherein the agent is a hydroxyurea.
14921. The method of item 14742 wherein the device comprises an agent that inhibits infection, and wherein the agent is a platinum complex.
14922. The method of item 14742 wherein the device comprises an agent that inhibits infection, and wherein the agent is cisplatin.
14923. The method of item 14742, further comprising an anti-thrombotic agent.
14924. The method of item 14742 wherein the device comprises a visualization agent.
14925. The method of item 14742 wherein the device comprises a visualization agent, wherein the visualization agent is a radiopaque material, and wherein the radiopaque material comprises a metal, a halogenated compound, or a barium containing compound.
14926. The method of item 14742 wherein the device comprises a visualization agent, wherein the visualization agent is a radiopaque material, and wherein the radiopaque material comprises barium, tantalum, or technetium.
14927. The method of item 14742 wherein the device comprises a visualization agent, and wherein the visualization agent is a MRI responsive material. 14928. The method of item 14742 wherein the device comprises a visualization agent, and wherein the visualization agent comprises a gadolinium chelate.
14929. The method of item 14742 wherein the device comprises a visualization agent, and wherein the visualization agent comprises iron, magnesium, manganese, copper, or chromium.
14930. The method of item 14742 wherein the device comprises a visualization agent, and wherein the visualization agent comprises an iron oxide compound.
14931. The method of item 14742 wherein the device comprises a visualization agent, and wherein the visualization agent comprises a dye, pigment, or colorant.
14932. The method of item 14742 wherein the device comprises an echogenic material.
14933. The method of item 14742 wherein the device comprises an echogenic material, and wherein the echogenic material is in the form of a coating.
14934. The method of item 14742 wherein the device is sterile.
14935. The method of item 14742 wherein the anti- scarring agent is released into tissue in the vicinity of the device after deployment of the device. 14936. The method of item 14742 wherein the anti- scarring agent is released into tissue in the vicinity of the device after deployment of the device, and wherein the tissue is connective tissue.
14937. The method of item 14742 wherein the anti- scarring agent is released into tissue in the vicinity of the device after deployment of the device, and wherein the tissue is muscle tissue.
14938. The method of item 14742 wherein the anti- scarring agent is released into tissue in the vicinity of the device after deployment of the device, and wherein the tissue is nerve tissue.
14939. The method of item 14742 wherein the anti- scarring agent is released into tissue in the vicinity of the device after deployment of the device, and wherein the tissue is epithelium tissue.
14940. The method of item 14742 wherein the anti- scarring agent is released in effective concentrations from the device over a period ranging from the time of deployment of the device to about 1 year.
14941. The method of item 14742 wherein the anti- scarring agent is released in effective concentrations from the device over a period ranging from about 1 month to 6 months.
14942. The method of item 14742 wherein the anti- scarring agent is released in effective concentrations from the device over a period ranging from about 1 - 90 days.
14943. The method of item 14742 wherein the anti- scarring agent is released in effective concentrations from the device at a constant rate. 14944. The method of item 14742 wherein the anti- scarring agent is released in effective concentrations from the device at an increasing rate.
14945. The method of item 14742 wherein the anti- scarring agent is released in effective concentrations from the device at a decreasing rate.
14946. The method of item 14742 wherein the anti- scarring agent is released in effective concentrations from the composition comprising the anti-scarring agent by diffusion over a period ranging from the time of deployment of the device to about 90 days.
14947. The method of item 14742 wherein the anti- scarring agent is released in effective concentrations from the composition comprising the anti-scarring agent by erosion of the composition over a period ranging from the time of deployment of the device to about 90 days.
14948. The method of item 14742 wherein the device comprises about 0.01 Dg to about 10 Dg of the anti-scarring agent.
14949. The method of item 14742 wherein the device comprises about 10 Dg to about 10 mg of the anti-scarring agent.
14950. The method of item 14742 wherein the device comprises about 10 mg to about 250 mg of the anti-scarring agent.
14951. The method of item 14742 wherein the device comprises about 250 mg to about 1000 mg of the anti-scarring agent. 14952. The method of item 14742 wherein the device comprises about 1000 mg to about 2500 mg of the anti-scarring agent.
14953. The method of item 14742 wherein a surface of the device comprises less than 0.01 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
14954. The method of item 14742 wherein a surface of the device comprises about 0.01 Dg to about 1 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
14955. The method of item 14742 wherein a surface of the device comprises about 1 Dg to about 10 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
14956. . The method of item 14742 wherein a surface of the device comprises about 10 Dg to about 250 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
14957. The method of item 14742 wherein a surface of the device comprises about 250 Dg to about 1000 Dg of the anti-scarring agent of anti-scarring agent per mm2 of device surface to which the anti- scarring agent is applied.
14958. The method of item 14742 wherein a surface of the device comprises about 1000 Dg to about 2500 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
14959. The method of item 14742 wherein the combining is performed by direct affixing the agent or the composition to the implant. 14960. The method of [tern 14742 wherein the combining is performed by spraying the agent or the component onto the implant.
14961. The method of item 14742 wherein the combining is performed by electrospraying the agent or the composition onto the implant.
14962. The method of item 14742 wherein the combining is performed by dipping the implant into a solution comprising the agent or the composition.
14963. The method of item 14742 wherein the combining is performed by covalently attaching the agent or the composition to the implant.
14964. The method of item 14742 wherein the combining is performed by non-covalently attaching the agent or the composition to the implant.
14965. The method of item 14742 wherein the combining is performed by coating the implant with a substance that contains the agent or the composition.
14966. The method of item 14742 wherein the combining is performed by coating the implant with a substance that absorbs the agent.
14967. The method of item 14742 wherein the combining is performed by interweaving a thread composed of, or coated with, the agent or the composition. 14968. The method of item 14742 wherein the combining is performed by covering all the implant with a sleeve that contains the agent or the composition.
14969. The method of item 14742 wherein the combining is performed by covering a portion of the implant with a sleeve that contains the agent or the composition.
14970. The method of item 14742 wherein the combining is performed by covering all the implant with a cover that contains the agent or the composition.
14971. The method of item 14742 wherein the combining is performed by covering a portion of the implant with a cover that contains the agent or the composition.
14972. The method of item 14742 wherein the combining is performed by covering all the implant with an electrospun fabric that contains the agent or the composition.
14973. The method of item 14742 wherein the combining is performed by covering a portion of the implant with an electrospun fabric that contains the agent or the composition.
14974. The method of item 14742 wherein the combining is performed by covering all the implant with a mesh that contains the agent or the composition.
14975. The method of item 14742 wherein the combining is performed by covering a portion of the implant with a mesh that contains the agent or the composition. 14976. The method of item 14742 wherein the combining is performed by constructing all the implant with the agent or the composition.
14977. The method of item 14742 wherein the combining is performed by constructing a portion of the implant with the agent or the composition.
14978. The method of item 14742 wherein the combining is performed by impregnating the implant with the agent or the composition.
14979. The method of item 14742 wherein the combining is performed by constructing all of the implant from a degradable polymer that releases the agent.
14980. The method of item 14742 wherein the combining is performed by constructing a portion of the implant from a degradable polymer that releases the agent.
14981. The method of item 14742 wherein the combining is performed by dipping the implant into a solution that comprise the agent and an inert solvent for the implant.
14982. The method of item 14742 wherein the combining is performed by dipping the implant into a solution that comprises the agent and a solvent that will swell the implant.
14983. The method of item 14742 wherein the combining is performed by dipping the implant into a solution that comprises the agent and a solvent that will dissolve the implant. 14984. The method of item 14742 wherein the combining is performed by dipping the implant into a solution that comprises the agent, a polymer and an inert solvent for the implant.
14985. The method of item 14742 wherein the combining is performed by dipping the implant into a solution that comprises the agent, a polymer and a solvent that will swell the implant.
14986. The method of item 14742 wherein the combining is performed by dipping the implant into a solution that comprises the agent, a polymer and a solvent that will dissolve the implant.
14987. The method of item 14742 wherein the combining is performed by spraying the implant into a solution that comprises the agent and an inert solvent for the implant.
14988. The method of item 14742 wherein the combining is performed by spraying the implant into a solution that comprises the agent and a solvent that will swell the implant.
14989. The method of item 14742 wherein the combining is performed by spraying the implant into a solution that comprises the agent and a solvent that will dissolve the implant.
14990. The method of item 14742 wherein the combining is performed by spraying the implant into a solution that comprises the agent, a polymer and an inert solvent for the implant.
14991. The method of item 14742 wherein the combining is performed by spraying the implant into a solution that comprises the agent, a polymer and a solvent that will swell the implant. 14992. The method of item 14742 wherein the combining is performed by spraying the implant into a solution that comprises the agent, a polymer and a solvent that will dissolve the implant.
14993. A method as in any one of items 14742-14992, wherein the device is a penile implant that is a flexible rod.
14994. A method as in any one of items 14742-14992, wherein the device is a penile implant that is a hinged rod.
14995. A method as in any one of items 14742-14992, wherein the device is a penile implant that is an inflatable device with a pump.
14996. A method of making a medical device comprising: combining an endotracheal or tracheostomy tube (e.g., an implant) and an anti-scarring agent or a composition comprising an anti- scarring agent, wherein the agent inhibits scarring between the device and a host into which the device is implanted.
14997. The method of item 14996 wherein the agent is an adensosine A2A receptor antagonist.
14998. The method of item 14996 wherein the agent is an AKT inhibitor.
14999. The method of item 14996 wherein the agent is an alpha 2 integrin antagonist, wherein the alpha 2 integrin antagonist is Pharmaprojects No. 5754 (Merck KgaA). 15000. The method of item 14996 wherein the agent is an alpha 4 integrin antagonist.
15001. The method of item 14996 wherein the agent is an alpha 7 nicotinic receptor agonist.
15002. The method of item 14996 wherein the agent is an angiogenesis inhibitor selected from the group consisting of AG-12,958 (Pfizer), ATN-161 (Attention LLC), neovastat, an angiogenesis inhibitor from Jerina AG (Germany), NM-3 (Mercian), VGA-1155 (Taisho), FCE-26644 (Pfizer), FCE-26950 (Pfizer), FPMA (Meiji Daries), FR-111142 (Fujisawa), GGTI-298, GM-1306 (Ligand), GPA-1734 (Novartis), NNC-47-0011 (Novo Nordisk), herbamycin (Nippon Kayaku), ienalidomide (Celegene), IP-10 (NIH), ABT-828 (Abbott), KIN-841 (Tokushima University, Japan), SF-1126 (Semafore Pharmaceuticals), laminin technology (NIH), CHIR-258 (Chiron), NVP-AEW541 (Novartis), NVP-AEW541 (Novartis), Vt16907 (Alchemia), OXI-8007 (Oxigene), EG-3306 (Ark Therapeutics), Maspin (Arriva), ABT- 567 (Abbott), PPI-2458 (Praecis Pharmaceuticals), CC-5079, CC-4089 (Celgene), HIF-1 alpha inhibitors (Xenova), S-247 (Pfizer), AP-23573 (Ariad), AZD-9935 (Astra Zeneca), mebendazole (Introgen Therapeutics), MetAP-2 inhibitors (GlaxoSmithKline), AG-615 (Angiogene Pharmaceuticals), Tie-2 antagonists (Hybrigenics), NC-381, CYC-381, NC-169, NC-219, NC-383, NC-384, NC-407 (Lorus Therapeutics), ATN-224 (Attenuon), ON-01370 (Onconova), Vitronectin antagonists (Amgen), SDX-103 (Salmedix), Vitronectin antagonists (Shire), CHP (Riemser), TEK (Amgen), Anecortave acetate (Alcon), T46.2 (Matrix Therapeutics), HG-2 (Heptagen), TEM antagonists (Genzyme), Oxi-4500 (Oxigene), ATN-161 (Attenuon), WX-293 (Wilex), M-2025 (Metris Therapeutics), Alphastatin (BioActa), YH-16 (Yantai Rongchang), BIBF-1120 (Boehringer Ingelheim), BAY-57-9352 (Bayer), AS- 1404 (Cancer Research Technology), SC-77964 (Pfizer), glycomimetics (BioTie Therapies), TIE-2 Inhibitors (Ontogen), DIMI, Octamer (Octamer), ABR-215050 (Active Biotech), ABT-518 (Abbott), KDR inhibitors (Abbott), BSF-466895 (Abbott), SCH-221153 (Schering-Plough), DAC:antiangiogenic (ConjuChem), TFPI (EntreMed), AZD-2171 (Astra-Zenaca), CDC-394 (Celgene), LY290293 (EIi Lilly), IDN-5390 (Indena), Kdr Kinase Inhibitors (Merck), CT-113020, CT-116433, CT-116563, CT-31890, CT-32228) (Cell Therapeutics), A-299620 (Abbott), TWEAK Inhibitor (Amgen), VEGF modulators (Johnson and Johnson), Tum-N53, tumstatin (Genzyme), Thios- 1 , Thios-2 (Thios Pharmaceuticals), MV-6401 (Miravant Medical Technologies), Spisulosine (PharmaMar), CEP-7055 (Cephalon), AUV-201 (Auvation), LM-609 (EIi Lilly), SKF-106615 (AnorMED), Oglufanide disodium (Cytran), BW-114 (Pharminox), Calreticulin (NIH), WX-678 (Wilex), SD-7784 (Pfizer), WX-U K1 (Wilex), SH-268 (Schering AG), 2-Me-PGA (Celgene), S- 137 (Pfizer), ZD-6126 (Angiogene Pharmaceuticals), SG-292 (SignalGen), Benefin (Lane Labs), A6, A36 (Angstrom), SB-2723005 (GlaxoSmithKline), SC-7 (Cell Therapeutics), ZEN-014 (AEterna Zentaris), 2-methoxyestradiol (EntreMed), NK-130119 (Nippon Kayaku), CC-10004 (Celgene), AVE- 8062A (Ajinomoto), Tacedinaline (Pfizer), Actinonin (Tokyo Metropolitan Institute of Medical Science), Lenalidomide (Celgene), VGA-1155, BTO-956 (SRI International), ER-68203-00 (Eisai), CT-6685 (UCB), JKC-362 (Phoenix Pharmaceuticals), DMI-3798 (DMI Biosciences, Angiomate (Ipsen), ZD-6474 (AstraZeneca), CEP-5214 (Cephalon), Canstatin (Genzyme), NM-3 (Mercian), Oridigm (MediQuest Therapeutics), Exherin (Adherex), BLS-0597 (Boston Life Sciences), PTC-299 (PTC Therapeutics), NPI-2358 (Nereus Pharmaceuticals), CGP-79787 (Novartis), AEE-788 (Novartis), CKD-732 (Chong Kun Dang), CP-564959 (OSI Pharmaceuticals), CM-101 (CarboMed), CT-2584, CT3501 (Cell Therapeutics), combretastatin and analogues and derivatives thereof (Oxigene), Rebimastat (Bristol-Meyers Squibb), Dextrin 2-sulfate (ML Laboratories), Cilengitide (Merk KGaA), NSC-706704 (Pharminox), KRN- 951 (Kirin Brewery), Ukrain, NSC-631570 (Nowicky Pharma), Tecogalan sodium (Daiichi Pharmaceutical), Tz-93 (Tsumura), TBC-1635 (Encysive Pharmaceuticals), TAN-1120 (Takeda), Semaxanib (Pfizer), BDI-7800 (Biopharmacopae), SD-186, SD-983 (Bristol-Meyers Squibb), SB-223245 (GlaxoSmith Kline), SC-236 (Pfizer), RWJ-590973 (Johnson and Johnson), ILX-1850 (Genzyme), SC-68488, S-836 (Pfizer), CG-55069-11 (CuraGen), Ki-23057 (Kirin Brewery), CCX-700 (Chemoentryx), Pegaptanib octasodium (Giled Sciences), ANGIOCOL (available from Biostratum Inc.), or an analogue or derivative thereof.
15003. The method of item 14996 wherein the agent is an apoptosis antagonist.
15004. The method of item 14996 wherein the agent is an apoptosis activator.
15005. The method of item 14996 wherein the agent is a beta 1 integrin antagonist.
15006. The method of item 14996 wherein the agent is a beta tubulin inhibitor.
15007. The method of item 14996 wherein the agent is a blocker of enzyme production in Hepatitis C.
15008. The method of item 14996 wherein the agent is a Bruton's tyrosine kinase inhibitor.
15009. The method of item 14996 wherein the agent is a calcineurin inhibitor.
15010. The method of item 14996 wherein the agent is a caspase 3 inhibitor. 15011. The method of item 14996 wherein the agent is a CC chemokine receptor antagonist.
15012. The method of item 14996 wherein the agent is a cell cycle inhibitor selected from the group consisting of SNS-595 (Sunesis), synthadotin, KRX-0403, and an analogue or derivative thereof.
15013. The method of item 14996 wherein the agent is a cathepsin B inhibitor.
15014. The method of item 14996 wherein the agent is a cathepsin K inhibitor, wherein the cathepsin K inhibitor is 462795 (GlaxoSmithKline), INPL-022-D6 (Amura Therapeutics), or an analogue or derivative thereof.
15015. The method of item 14996 wherein the agent is a cathepsin L inhibitor.
15016. The method of item 14996 wherein the agent is a CD40 antagonist.
15017. The method of item 14996 wherein the agent is a chemokine receptor agonist.
15018. The method of item 14996 wherein the agent is a chymase inhibitor.
15019. The method of item 14996 wherein the agent is a collagenase antagonist. 15020. The method of item 14996 wherein the agent is a CXCR antagonist.
15021. The method of item 14996 wherein the agent is a cyclin dependent kinase inhibitor selected from the group consisting of a CDK- 1 inhibitor, a CDK-2 inhibitor, a CDK- 4 inhibitor, a CDK-6 inhibitor, a CAK1 inhibitor from GPC Biotech and Bristol-Myers Squibb, RGB-286199 (GPC Biotech), an anticancer agent from Astex Technology, a CAK1 inhibitor from GPC Biotech, a CDK inhibitor from Sanofi-Aventis, a CDK1/CDK2 inhibitor from Amgen, a CDK2 inhibitor from SUGEN-2 (Pfizer), a hearing loss therapy agent (Sound Pharmaceuticals), PD-0332991 (Pfizer), RGB-286199 (GPC Biotech), Ro-0505124 (Hoffmann-La Roche), a Ser/Thr kinase inhibitor from Lilly (EIi Lilly), CVT-2584 (CAS No. 199986-75- 9) (CV Therapeutics), and an analogue or derivative thereof.
15022. The method of item 14996 wherein the agent is a cyclooxygenase 1 inhibitor.
15023. The method of item 14996 wherein the agent is a DHFR inhibitor.
15024. The method of item 14996 wherein the agent is a dual integrin inhibitor.
15025. The method of item 14996 wherein the agent is an elastase inhibitor.
15026. The method of item 14996 wherein the agent is an elongation factor-1 alpha inhibitor. 15027. The method of item 14996 wherein the agent is an endothelial growth factor antagonist.
15028. The method of item 14996 wherein the agent is an endothelial growth factor receptor kinase inhibitor selected from the group consisting of sorafenib tosylate (Bayer), AAL-993 (Novartis), ABP-309 (Novartis), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), E- 7080 (Eisai), EG-3306 (Ark Therapeutics), EXEL-2880 (Exelixis), GW- 654652 (GlaxoSmithKline), a KDR inhibitor from LG Life Sciences, CT-6685 and CT-6729 (UCB), KRN-633 and KRN-951 (Kirin Brewery), OSI-930 (OSI Pharmaceuticals), SP-5.2 (Supratek Pharma), SLM 1657 (Pfizer), a Tie-2 antagonist (Hybrigenics), a VEGFR-2 kinase inhibitor (Bristol-Myers Squibb), XL-647 (Exelixis), a KDR inhibitor from Abbott Laboratories, sorafenib tosylate, and an analogue or derivative thereof.
15029. The method of item 14996 wherein the agent is an endotoxin antagonist.
15030. The method of item 14996 wherein the agent is an epothilone and tubulin binder.
15031. The method of item 14996 wherein the agent is an estrogen receptor antagonist.
15032. The method of item 14996 wherein the agent is an FGF inhibitor.
15033. The method of item 14996 wherein the agent is a farnexyl transferase inhibitor. 15034. The method of item 14996 wherein the agent is famesyltransferase inhibitor selected from the group of A-197574 (Abbott), a famesyltransferase inhibitor from Servier, FPTIII (Strathclyde Institute for Drug R), LB-42908 (LG Life Sciences), Pharmaprojects No. 5063 (Genzyme), Pharmaprojects No. 5597 (Ipsen), Yissum Project No. B-1055 (Yissum), and an analogue or derivative thereof.
15035. The method of item 14996 wherein the agent is an FLT-3 kinase inhibitor.
15036. The method of item 14996 wherein the agent is an FGF receptor kinase inhibitor.
15037. The method of item 14996 wherein the agent is a fibrinogen antagonist selected from the group consisting of AUV-201 (Auvation), MG-13926 (Sanofi-Aventis), plasminogen activator (CAS No. 105913-11-9) (from Sanofi-Aventis or UCB), plasminogen activator-2 (tPA- 2) (Sanofi-Aventis), pro-urokinase (CAS No. 82657-92-9) (Sanofi-Aventis), and an analogue or derivative thereof.
15038. The method of item 14996 wherein the agent is a heat shock protein 90 antagonist selected from the group consisting of SRN- 005 (Sirenade), geldanamycin, NSC-33050 (17-allylaminogeldanamycin; 17- AAG), 17-dimethylaminoethylamino-17-demethoxy-geldanamycin (17- DMAG), rifabutin (rifamycin XIV, 1',4-didehydro-1-deoxy-1 ,4-dihydro-5'-(2- methylpropyl)-1-oxo-), and an analogue or derivative thereof.
15039. The method of item 14996 wherein the agent is a histone deacetylase inhibitor. 15040. The method of item 14996 wherein the agent is an HMGCoA reductase inhibitor selected from the group consisting of an atherosclerosis therapeutic from Lipid Sciences, ATI-16000 (ARYx Therapeutics), KS-01-019 (Kos Pharmaceuticals), Pharmaprojects No. 2197 (Sanofi-Aventi), RP 61969 (Sanofi-Aventis), and an analogue or derivative thereof.
15041. The method of item 14996 wherein the agent is an ICAM inhibitor.
15042. The method of item 14996 wherein the agent is an IL, ICE and IRAK antagonist, wherein the antagonist is a CJ-14877, CP- 424174 (Pfizer), NF-61 (Negma-Lerads), and an analogue or derivative thereof.
15043. The method of item 14996 wherein the agent, is __ an IL-2 inhibitor.
15044. The method of item 14996 wherein the agent is an immunosuppressant selected from the group consisting of teriflunomide (Sanofi Aventis), chlorsulfaquinoxalone (NSC-339004), chlorsulfaquinoxalone sulfate, CS-712 (Sankyo), ismomultin alfa (CAS No. 457913-93-8) (Akzo Nobel), antiallergics from GenPat77, antiinflammatories or AT-005 (Androclus Therapeutics), autoimmune disease therapeutics from EpiVax, BN-007 (Bone), budesonide (CAS No. 51333-22- 3) (MAP Pharmaceuticals), CO-14 (Genzyme), edratide (CAS No. 433922- 67-9) (Teva), EP-314 (Enanta), eprovafen (CAS No. 101335-99-3) (Sanofi- Aventis), HWA-131 (CAS No. 118788-41-3) (Sanofi-Aventis), immunomodulators from MerLion Pharmaceuticals, immunosuppressives from Alchemia, IPL-12 (Inflazyme), MDL-9563 (CAS No. 27086-86-8) (Sanofi-Aventis), Pharmaprojects No. 2330 (Sanofi-Aventis), Pharmaprojects No. 6426 (Abgenix), PXS-25 (Pharmaxis), rosmarinic acid (CAS No. 20283-92-5) (Sanofi-Aventis), RP 42927 or RP 54745 (CAS No. 135330-08-4) (Sanofi-Aventis), SGN-35 (Seattle Genetics), ST-1959 (Sigma-Tau), type I diabetes therapy from SYNX Pharma, UNIL-88 (Debiopharm), VP-025 (Vasogen), VR-694 (Vectura), PRTX-001 (Protalex), and an analogue or derivative thereof.
15045. The method of item 14996 wherein the agent is an IMPDH (inosine monophosphate).
15046. The method of item 14996 wherein the agent is an integrin antagonist.
15047. The method of item 14996 wherein the agent is an interleukin antagonist.
15048. The method of item 14996 wherein the agent is an inhibitor of type III receptor tyrosine kinase.
15049. The method of item 14996 wherein the agent is an irreversible inhibitor of enzyme methionine aminopeptidase type 2.
15050. The method of item 14996 wherein the agent is an isozyme selective delta protein kinase C inhibitor.
15051. The method of item 14996 wherein the agent a JAK3 enzyme inhibitor.
15052. The method of item 14996 wherein the agent is a JNK inhibitor. 15053. The method of item 14996 wherein the agent is a kinase inhibitor.
15054. The method of item 14996 wherein the agent is kinesin antagonist.
15055. The method of item 14996 wherein the agent is a kinesin antagonist.
15056. The method of item 14996 wherein the agent is a leukotriene inhibitor and antagonist selected from the group consisting of ambicromil (CAS No. 58805-38-2) (Sanofi-Aventis), amelubant (CAS No. 346735-24-8) (Boehringer Ingelheim), DW-1141 (Dong Wha), ebselen (Daiichi Pharmaceutical), ibudilast (Kyorin), leucotriene inhibitors from Sanofi-Aventis, lymphotoxin-beta receptor (LT-D) from Biogen Idee, Pharmaprojects No. 1535 and 2728 (CAS No. 119340-33-9) (Sanofi- Aventis), R-112 (Rigel), Rev-5367 (CAS No. 92532-05-3) (Sanofi-Aventis), RG-14893 (CAS No. 141835-49-6) (Sanofi-Aventis), RG-5901-A (CAS No. 101910-24-1), 92532-23-5, RP 66153 (CAS No. 142422-79-5), RP 66364 (CAS No.186916-92-5), RP 69698 (CAS No. 141748-00-7) (Sanofi-Aventis), SC-411930 (Pfizer), SC-41930 (CAS No. 120072-59-5) (Pfizer), SC-50605 (CAS No. 138828-39-4) (Pfizer), SC-51146 (CAS No. 141059-52-1), SC- 53228 (CAS No. 153633-01-3) (Pfizer), spaglumic acid (ZY-15106) (CAS No.3106-85-2), 80619-64-3 (Novartis), tipredane (CAS No. 85197-77-9) (Bristol-Myers Squibb), U-75302 (CAS No. 119477-85-9) (Pfizer), and analogue or derivative thereof.
15057. The method of item 14996 wherein the agent is an MAP kinase inhibitor. 15058. The method of item 14996 wherein the agent is a matrix metalloproteinase inhibitor.
15059. The method of item 14996 wherein the agent is an MCP-CCR2 inhibitor.
15060. The method of item 14996 wherein the agent is an mTOR inhibitor.
15061. The method of item 14996 wherein the agent is an mTOR kinase inhibitor.
15062. The method of item 14996 wherein the agent is a microtubule inhibitor selected from the group consisting of antibody- maytansinoid conjugates from Biogen Idee, colchicines (MantiCore Pharmaceuticals), anticancer immunoconjugates from Johnson & Johnson, DIME from Octamer, gni-1f (GNI), huC242-DM4, huMy9-6-DM1 (ImmunoGen), IDN-5404 (Indena), IMO-098, IMOderm (Imotep), mebendazole (Introgen Therapeutics), microtubule poisons from Cambridge Enterprise, paclitaxel such as LOTAX from Aphios (CAS No. 33069-62-4), Genexol-PM from Samyang, Pharmaprojects No. 6383 (Tapestry Pharmaceuticals), RPR-112378 (Sanofi-Aventis), SGN-75 (Seattle Genetics), SPL-7435 (Starpharma), SSR-250411 (Sanofi-Aventis), trastuzumab~DM1 (Genentech), vinorelbine, and an analogue or derivative thereof.
15063. The method of item 14996 wherein the agent is an MIF inhibitor.
15064. The method of item 14996 wherein the agent is an MMP inhibitor. 15065. The method of item 14996 wherein the agent is a neurokinin (NK) antagonist selected from the group consisting of anthrotainin (CAS No. 148084-40-6) (Sanofi-Aventis), an IBS thereapeutic from ArQuIe, MDL-105212A (CAS No. 167261-60-1) (Ssanofi-Aventis), Pharmaprojects No. 2744, 3258 (CAS No. 139167-47-8) 4006, 4201 , or 5986 (Sanofi-Aventis), RP 67580 (CAS No. 135911-02-3), SR-144190 (CAS No. 201152-86-5), SSR-240600, SSR-241586 (Sanofi-Aventis), TKA-457 (Novartis), vestipitant mesylate (CAS No. 334476-64-1) (GlaxoSmithKline), Win-64821 (Sanofi-Aventis), PRX-96026 (Predix Pharmaceuticals), and an analogue or derivative thereof.
15066. The method of item 14996 wherein the agent is an NF kappa B inhibitor selected from the group consisting of AVE-0545 or AVE-0547 (Sanofi-Aventis), bortezomib (CAS No. 179324-69-7) (Millennium Pharmaceuticals), dexanabinol (CAS No. 112924-45-5) (Pharmos), dexlipotam (Viatris), Pharmaprojects No. 6283 (INDRA) (OXiGENE), IPL- 576092 (CAS No. 137571-30-3) (Inflazyme), NFKB decoy (Corgentech), NFKB decoy oligo (AnGes MG), NFKB's from Ariad, osteoporosis treatments or S5 (F005) from Fulcrum Pharmaceuticals, P61 (Phytopharm), R-flurbiprofen (CAS No. 5104-49-4) (Encore Pharmaceuticals), and an analogue or derivative thereof.
15067. The method of item 14996 wherein the agent is a nitric oxide agonist.
15068. The method of item 14996 wherein the agent is an ornithine decarboxylase inhibitor.
15069. The method of item 14996 wherein the agent is a p38 MAP kinase inhibitor selected from the group consisting of AZD-6703 (AstraZeneca), JX-401 (Jexys Pharmaceuticals), BMS-2 (Bristol-Myers Squibb), a p38 MAP kinase inhibitor from Novartis, a p38-alpha MAP kinase inhibitor from Amphora, Pharmaprojects No. 5704 (Pharmacopeia), RPR- 200765A (Sanofi-Aventis), SD-282 (Johnson & Johnson), TAK-715 (Takeda), and an analogue or derivative thereof.
15070. The method of item 14996 wherein the agent is a palmitoyl-protein thioesterase inhibitor.
15071. The method of item 14996 wherein the agent is a PDGF receptor kinase inhibitor selected from the group consisting of AAL- 993, AMN-107, or ABP-309 (Novartis), AMG-706 (Amgen), BAY-57-9352 (Bayer), CDP-860 (UCB), E-7080 (Eisai), imatinib (CAS No. 152459-95-5) (Novartis), OSI-930 (OSI Pharmaceuticals), RPR-127963E (Sanofi-Aventis), RWJ-540973 (Johnson & Johnson), sorafenib tosylate (Bayer), SU-11657 (Pfizer), tandutinib (CAS No. 387867-13-2) (Millennium Pharmaceuticals), vatalanib (Novartis), ZK-CDK (Schering AG), and an analogue or derivative thereof.
15072. The method of item 14996 wherein the agent is (-)-halofenate (Metabolex), AMG-131 (Amgen), antidiabetics from Japan Tobacco, AZD-4619, AZD-8450, AZD-8677 (AstraZeneca), DRF-10945, balaglitazone (Dr Reddy's), CS-00088, CS-00098 (Chipscreen Biosciences), E-3030 (Eisai), etalocib (CAS No. 161172-51-6) (EIi Lilly), GSK-641597 (Ligand), GSK-677954 (GlaxoSmithKline), GW-409544 (Ligand), GW- 590735 (GlaxoSmithKline), K-111 (Hoffmann-La Roche), LY-518674 (EIi Lilly), LY-674 (Ligand), LY-929 (Ligand), MC-3001, MC-3002 (MaxoCore Pharmaceuticals), metformin HCI + pioglitazone (CAS No. 1115-70-4 and
112529-15-4), ACTOPLUS MET from Andrx), muraglitazar (CAS No. 331741-94-7) (Bristol-Myers Squibb), naveglitazar (Ligand), oleoylethanolamide (Kadmus Pharmaceuticals), ONO-5129, pioglitazone hydrochloride (CAS No. 111025-46-8 and 112529-15-4) (Takeda), PLX-204 (Plexxikon), PPAR agonists from Genfit, PPAR delta agonists from EIi Lilly, PPAR-alpha agonists from CrystalGenomics, PPAR-gamma modulators and PPAR-β modulators from CareX, rosiglitazone maleate (CAS No. 122324- 73-4 or 155141-29-0) (GlaxoSmithKline), rosiglitazone maleate/glimepir (CAS No. 155141-29-0 and 93479-97-1), AVANDARYL, rosiglitazone maleate/metformin extend (CAS No. 155141-29-0 and 657-24-9), AVANDAMET, rosiglitazone maleate+metformin, AVANDAMET (GlaxoSmithKline), tesaglitazar (AstraZeneca), and an analogue or derivative thereof.
15073. The method of item 14996 wherein the agent is a phosphatase inhibitor.
15074. The method of item 14996 wherein the agent is a phosphodiesterase (PDE) inhibitor selected from the group consisting of avanafil (Tanabe Seiyaku), dasantafil (CAS No. 569351-91-3) (Schering- . Plough), A-906119 (CAS No. 134072-58-5), DL-850 (Sanofi-Aventis), GRC- 3015, GRC-3566, GRC-3886 (Glenmark), HWA-153 (CAS No. 56395-66-5) (Sanofi-Aventis), hydroxypumafentrine (Altana), IBFB-130011 , IBFB-14-016, IBFB-140301 , IBFB-150007, IBFB-211913 (IBFB Pharma), L-826141 (Merck & Co), medorinone (CAS No. 88296-61-1) (Sanofi-Aventis), MEM- 1917 (Memory Pharmaceuticals), ND-1251 (Neuro3d), PDE inhibitors from ICOS, PDE IV inhibitors from Memory Pharmaceuticals and CrystalGenomics, Pharmaprojects No. 2742 and 6141 (Sanofi-Aventis), QAD-171 (Novartis), RHC-2963 (CAS No. 76993-12-9 and 76993-14-1), RPR-117658, RPR-122818 derivatives, SR-24870 , and RPR-132294 (Sanofi-Aventis), SK-350 (ln2Gen), stroke therapy agents from deCODE Genetics, TAS-203 (Taiho), tofimilast (CAS No. 185954-27-2) (Pfizer), UK- 371800 (Pfizer), WIN-65579 (CAS No. 158020-82-7) (Sanofi-Aventis), IBFB- 130020 (IBFB Pharma), OPC-6535 (CAS No. 145739-56-6) (Otsuka), a phosphodiesterase III inhibitor, enoximone, a phosphodiesterase IV inhibitor, fosfosal, Atopik (Barrier Therapeutics), triflusal, a phosphodiesterase V inhibitor, and an analogue or derivative thereof.
15075. The method of item 14996 wherein the agent is a PKC inhibitor.
15076. The method of item 14996 wherein the agent is a platelet activating factor antagonist.
15077. The method of item 14996 wherein the agent is a platelet-derived growth factor receptor kinase inhibitor.
15078. The method of item 14996 wherein the agent is a prolyl hydroxylase inhibitor.
15079. The method of item 14996 wherein the agent is a polymorphonuclear neutrophil inhibitor.
15080. The method of item 14996 wherein the agent is a protein kinase B inhibitor.
15081. The method of item 14996 wherein the agent is a protein kinase C stimulant.
15082. The method of item 14996 wherein the agent is a purine nucleoside analogue.
15083. The method of item 14996 wherein the agent is a purinoreceptor P2X antagonist. 15084. The method of item 14996 wherein the agent is a Raf kinase inhibitor.
15085. The method of item 14996 wherein the agent is a reversible inhibitor of ErbB1 and ErbB2.
15086. The method of item 14996 wherein the agent is a ribonucleoside triphosphate reductase inhibitor.
15087. The method of item 14996 wherein the agent is an SDF-1 antagonist.
15088. The method of item 14996 wherein the agent is a sheddase inhibitor.
15089. The method of item 14996 wherein the agent is an SRC inhibitor.
15090. The method of item 14996 wherein the agent is a stromelysin inhibitor.
15091. The method of item 14996 wherein the agent is an Syk kinase inhibitor.
15092. The method of item 14996 wherein the agent is a telomerase inhibitor.
15093. The method of item 14996 wherein the agent is a TGF beta inhibitor selected from the group consisting of pirfenidone (CAS No. 53179-13-8) (MARNAC), tranilast (CAS No. 53902-12-8) (Kissei), IN- 1130 (ln2Gen), mannose-6-phosphate (BTG), TGF-β antagonists from Inflazyme (Pharmaprojects No. 6075), TGF-β antagonists from Sydney, non-industrial source), TGF-β I receptor kinase inhibitors from EIi Lilly, TGF- β receptor inhibitors from Johnson & Johnson, and an analogue or derivative thereof.
15094. The method of item 14996 wherein the agent is a
TNFα antagonist or TACE inhibitor selected from the group consisting of adalimumab (CAS No. 331731-18-1) (Cambridge Antibody Technology), AGIX-4207 (AtheroGenics), AGT-1 (Advanced Biotherapy), an antiinflammatory from Borean Pharma, Cellzome, or Paradigm Therapeutics, anti-inflammatory vaccine (TNF-alpha kinoid) from Neovacs, humanized anti-TNF antibody or an anti-TNF MAb (CB0006) Celltech (UCB), apratastat (CAS No. 287445-51-0) (Wyeth), BMS-561392 (Bristol-Myers Squibb), BN- 006 (Bone), certolizumab pegol (CAS No. 428863-50-7 or CH-138 (UCB), cilomilast (CAS No. 153259-65-5) (GlaxoSmithKline), CR-1 (Nuada Pharmaceuticals), CRx-119 (CombinatoRx), D-5410 (UCB), dacopafant (CAS No. 125372-33-0) (Sanofi-Aventis), dersalazine (CAS No. 188913-57- 7/188913-58-8) (Uriach), etanercept (CAS No. 185243-69-0) (Amgen), ethyl pyruvate (Critical (Critical Therapeutics), golimumab (CAS No. 476181-74-5) (Johnson & Johnson), hormono-immunotherapy from Ipsen, CDP571 (e.g., Humicade from UCB), IC-485 (ICOS), infliximab (CAS No. 170277-31-3) (Johnson & Johnson), IP-751 (Manhattan Pharmaceuticals), ISIS-104838 (CAS No. 250755-32-9) (ISIS Pharmaceuticals), lenalidomide (CAS No. 191732-72-6) (Celgene), lentinan (CAS No. 37339-90-5) (Ajinomoto), MDL- 201112 (CAS No. 142130-73-2) (Sanofi-Aventis), medroxyprogesterone (CAS No. 520-85-4) (InKine Pharmaceutical), N-acetylcysteine (CAS No. 616-91-1) (Zambon), NBE-P2 (DIREVO Biotech), nerelimomab (CAS No. 162774-06-3) (Chiron), OM-294DP (OM PHARMA), onercept (CAS No. 199685-57-9) (Yeda), PASSTNF-alpha (Verigen), pentoxifylline or oxypentifylline (Sanofi-Aventis), Pharmaprojects No. 4091, 4241 , 4295, or 4488 (Sanofi-Aventis), Pharmaprojects No. 5480 (Amgen), Pharmaprojects No. 6749 (Cengent), pirfenidone (CAS No. 53179-13-8) (MARNAC)1 RPR- 132294 (Sanofi-Aventis), S5 (F002) (Fulcrum Pharmaceuticals), simvastatin (CAS No. 79902-63-9) (Merck & Co), STA-6292 (Synta Pharmaceuticals), tacrolimus (CAS No. 104987-11-3) (Fujisawa LifeCycle Pharma), talactoferrin alfa (CAS No. 308240-58-6) (Agennix), thalidomide (CAS No. 50-35-1) (Celgene), TNF antagonists form ProStrakan, and Synergen, TNF inhibitors (Amgen), TNF-alpha antagonists from Dynavax Technologies and Jerina AG (Germany), TNF-alpha inhibitors from IBFB Pharma and Xencor (Xencor), torbafylline (CAS No. 105102-21-4) (Sanofi-Aventis), UR-1505 (Uriach), VT-346 (Viron Therapeutics), YSIL6 (Y1S Therapeutics), YSTH2 (Y's Therapeutics), NPI-1302a-3 (Nereus Pharmaceuticals, a TNF antagonist from Jerina AG (Germany), dersalazine, and an analogue or derivative thereof.
15095. The method of item 14996 wherein the agent is a Toll receptor inhibitor.
15096. The method of item 14996 wherein the agent is a tubulin antagonist.
15097. The method of item 14996 wherein the agent is a tyrosine kinase inhibitor selected from the group consisting of SU-011248, SUTENT from Pfizer Inc. (New York, NY), BMS-354825, PN-355 (Paracelsian Pharmaceuticals), AGN-199659 (Allergan), AAL-993 or ABP- 309 (Novartis), adaphostin (NIH), AEE-788 (Novartis), AG-013736 (OSI Pharmaceuticals), AG-13736 (Pfizer), ALT-110 (Alteris Therapeutics), AMG- 706 (Amgen), anticancer MAbs from Xencor, anti-EGFrvlll MAbs from Abgenix, anti-HER2 MAb from Abiogen, AZD-2171 or AZD-9935 (AstraZeneca), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), CEP-5214 (Cephalon), CEP-7055 (Cephalon), cetuximab (ImClone Systems), CHIR-200131 and CHIR-258 (Chiron), CP-547632 (OSI Pharmaceuticals), CP-724714 (Pfizer), CT-301 (Creabilis Therapeutics), D- 69491 (Baxter International), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EGFR/ErbB2 inhibitors from Array BioPharma, erlotinib (CAS No. 183319- 69-9) (OSI Pharmaceuticals), EXEL-2880 (Exelixis), FK-778 (Sanofi- Aventis) , gefitinib (CAS No. 184475-35-2) (AstraZeneca), GW-2286 or GW- 654652 (GlaxoSmithKline), her2/neu antigen from AlphaVax, HER-2/neu inhibitor from Generex, Herzyme (Medipad) (Sirna Therapeutics), HKI-272 (Wyeth), HuMax-EGFr (Genmab), idronoxil (CAS No. 81267-65-4) (Novogen), IGF-1 inhibitors from Ontogen, IMC-11 F8 (ImCIone Systems), kahalalide F (CAS No. 149204-42-2) (PharmaMar), KDR inhibitor from LG Life Sciences, KDR inhibitors from Abbott Laboratories, KDR kinase inhibitors (UCB), Kdr kinase inhibitors from Merck & Co, KRN-633 and KRN- 951 (Kirin Brewery), KSB-102 (Xenova), lapatinib ditosylate (CAS No. 388082-78-8) (GlaxoSmithKline), matuzumab (Merck KGaA), MDX-214 (Medarex), ME-103 (Pharmexa), MED-A300 (Gerolymatos), MNAC-13 (Lay Line Genomics), nimotuzumab (Center of Molecular Immunology), NSC- 330507 or NSC-707545 (NIH)1 NV-50 (Novogen), OSI-930 (OSI Pharmaceuticals), panitumumab (Abgenix), pelitinib (CAS No. 287933-82-7) (Wyeth), pertuzumab (CAS No. 380610-27-5) (Genentech), Pharmaprojects No. 3985 (Sanofi-Aventis), prostate cancer therapeutics from Sequenom (SQPC35, SQPC36, SQPC90), removab and remoxab (Trion Pharma), RG- 13022 (CAS No. 136831-48-6), RG-13291 (CAS No. 138989-50-1), or RG- 14620 (CAS No. 136831-49-7) (Sanofi-Aventis), RM-6427 (Romark), RNAi breast cancer therapy from Benitec, RP 53801 (CAS No. 125882-88-4) (Sanofi-Aventis), sorafenib tosylate (Bayer), SU-11657 (Pfizer), Tie-2 antagonists from Semaia (Hybrigenics), Tie-2 inhibitors from Ontogen, trastuzumab (CAS No. 180288-69-1) (Genentech), tyrosine kinase inhibitors from Sanofi-Aventis, U3-1287, U3-1565, U3-1784, U3-1800 (U3 Pharma), vatalanib (Novartis), VEGFR-2 kinase inhibitor from Bristol-Myers Squibb, XL-647 (Exelixis), ZD-6474 (AstraZeneca), ZK-CDK (Schering AG), an EGFR tyrosine kinase inhibitor, EKB-569 (Wyeth), and an analogue or derivative thereof.
15098. The method of item 14996 wherein the agent is a VEGF inhibitor.
15099. The method of item 14996 wherein the agent is a vitamin D receptor agonist.
15100. The method of item 14996 wherein the agent is ZD-6474 (an angiogenesis inhibitor).
15101. The method of item 14996 wherein the agent is AP-23573 (an mTOR inhibitor).
15102. The method of item 14996 wherein the agent is synthadotin (a tubulin antagonist).
15103. The method of item 14996 wherein the agent is S-0885 (a collagenase inhibitor).
15104. The method of item 14996 wherein the agent is aptidine (an elongation factor-1 alpha inhibitor).
15105. The method of item 14996 wherein the agent is ixabepilone (an epithilone).
15106. The method of item 14996 wherein the agent is IDN-5390 (an angiogenesis inhibitor). 15107. The method of item 14996 wherein the agent is SB-2723005 (an angiogenesis inhibitor).
15108. The method of item 14996 wherein the agent is ABT-518 (an angiogenesis inhibitor).
15109. The method of item 14996 wherein the agent is combretastatin (an angiogenesis inhibitor).
15110. The method of item 14996 wherein the agent is anecortave acetate (an angiogenesis inhibitor).
15111. The method of item 14996 wherein the agent is SB-715992 (a kinesin antagonist).
15112. The method of item 14996 wherein. the agent is temsirolimus (an mTOR inhibitor).
15113. The method of item 14996 wherein the agent is adalimumab (a TNFα antagonist).
15114. The method of item 14996, wherein the composition comprises a polymer.
15115. The method of item 14996, wherein the composition comprises a polymeric carrier.
15116. The method of item 14996 wherein the anti- scarring agent inhibits adhesion between the device and a host into which the device is implanted. 15117. The method of item 14996 wherein the device delivers the anti-scarring agent locally to tissue proximate to the device.
15118. The method of item 14996 wherein the device has a coating that comprises the anti-scarring agent.
15119. The method of item 14996, wherein the device has a coating that comprises the agent and is disposed on a surface of the implant.
15120. The method of item 14996, wherein the device has a coating that comprises the agent and directly contacts the implant.
15121. The method of item 14996, wherein the device has a coating that comprises the agent and indirectly contacts the implant.
15122. The method of item 14996, wherein the device has a coating that comprises the agent and partially covers the implant.
15123. The method of item 14996, wherein the device has a coating that comprises the agent and completely covers the implant.
15124. The method of item 14996, wherein the device has a uniform coating.
15125. The method of item 14996, wherein the device has a non-uniform coating.
15126. The method of item 14996, wherein the device has a discontinuous coating. 15127. The method of item 14996, wherein the device has a patterned coating.
15128. The method of item 14996, wherein the device has a coating with a thickness of 100 Dm or less.
15129. The method of item 14996, wherein the device has a coating with a thickness of 10 Dm or less.
15130. The method of item 14996, wherein the device has a coating, and the coating adheres to the surface of the implant upon deployment of the implant.
15131. The method of item 14996, wherein the device has a coating, and wherein the coating is stable at room temperature for a period of 1 year.
15132. The method of item 14996, wherein the device has a coating, and wherein the anti-scarring agent is present in the coating in an amount ranging between about 0.0001% to about 1% by weight.
15133. The method of item 14996, wherein the device has a coating, and wherein the anti-scarring agent is present in the coating in an amount ranging between about 1% to about 10% by weight.
15134. The method of item 14996, wherein the device has a coating, and wherein the anti-scarring agent is present in the coating in an amount ranging between about 10% to about 25% by weight. 15135. The method of item 14996, wherein the device has a coating, and wherein the anti-scarring agent is present in the coating in an amount ranging between about 25% to about 70% by weight.
15136. The method of item 14996, wherein the device has a coating, and wherein the coating further comprises a polymer.
15137. The method of item 14996, wherein the device has a first coating having a first composition and a second coating having a second composition.
15138. The method of item 14996, wherein the device has a first coating having a first composition and a second coating having a second composition, wherein the first composition and the second composition are different.
15139. The method of item 14996, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a copolymer.
15140. The method of item 14996, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a block copolymer.
15141. The method of item 14996, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a random copolymer.
15142. The method of item 14996, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a biodegradable polymer. 15143. The method of item 14996, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a non-biodegradable polymer.
15144. The method of item 14996, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a hydrophilic polymer.
15145. The method of item 14996, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a hydrophobic polymer.
15146. The method of item 14996, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a polymer having hydrophilic domains.
15147. The method of item 14996, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a polymer having hydrophobic domains.
15148. The method of item 14996, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a non-conductive polymer.
15149. The method of item 14996, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises an elastomer.
15150. The method of item 14996, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a hydrogel. 15151. The method of item 14996, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a silicone polymer.
15152. The method of item 14996, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a hydrocarbon polymer.
15153. The method of item 14996, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a styrene-derived polymer.
15154. The method of item 14996, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a butadiene polymer.
15155. The method of item 14996, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a macromer.
15156. The method of item 14996, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a poly(ethylene glycol) polymer.
15157. The method of item 14996 wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises an amorphous polymer.
15158. The method of item 14996, wherein the device comprises a lubricious coating. 15159. The method of item 14996 wherein the anti- scarring agent is located within pores or holes of the device.
15160. The method of item 14996 wherein the anti- scarring agent is located within a channel, lumen, or divet of the device.
15161. The method of item 14996, wherein the device comprises a second pharmaceutically active agent.
15162. The method of item 14996 wherein the device comprises an anti-inflammatory agent.
15163. The method of item 14996 wherein the device comprises an agent that inhibits infection.
15164. The method of item 14996 wherein the device comprises an agent that inhibits infection, and wherein the agent is an anthracycline.
15165. The method of item 14996 wherein the device comprises an agent that inhibits infection, and wherein the agent is doxorubicin.
15166. The method of item 14996 wherein the device comprises an agent that inhibits infection, and wherein the agent is mitoxantrone.
15167. The method of item 14996 wherein the device comprises an agent that inhibits infection, and wherein the agent is a fluoropyrimidine. 15168. The method of item 14996 wherein the device comprises an agent that inhibits infection, and wherein the agent is 5- fluorouracil (5-FU).
15169. The method of item 14996 wherein the device comprises an agent that inhibits infection, and wherein the agent is a folic acid antagonist.
15170. The method of item 14996 wherein the device comprises an agent that inhibits infection, and wherein the agent is methotrexate.
15171. The method of item 14996 wherein the device comprises an agent that inhibits infection, and wherein the agent is a podophylotoxin.
15172. The method of item 14996 wherein the device comprises an agent that inhibits infection, and wherein the agent is etoposide.
15173. The method of item 14996 wherein the device comprises an agent that inhibits infection, and wherein the agent is a camptothecin.
15174. The method of item 14996 wherein the device comprises an agent that inhibits infection, and wherein the agent is a hydroxyurea.
15175. The method of item 14996 wherein the device comprises an agent that inhibits infection, and wherein the agent is a platinum complex. 15176. The method of item 14996 wherein the device comprises an agent that inhibits infection, and wherein the agent is cisplatin.
15177. The method of item 14996, further comprising an antithrombotic agent.
15178. The method of item 14996 wherein the device comprises a visualization agent.
15179. The method of item 14996 wherein the device comprises a visualization agent, wherein the visualization agent is a radiopaque material, and wherein the radiopaque material comprises a metal, a halogenated compound, or a barium containing compound.
15180. The method of item 14996 wherein the device comprises a visualization agent, wherein the visualization agent is a radiopaque material, and wherein the radiopaque material comprises barium, tantalum, or technetium.
15181. The method of item 14996 wherein the device comprises a visualization agent, and wherein the visualization agent is a MRI responsive material.
15182. The method of item 14996 wherein the device comprises a visualization agent, and wherein the visualization agent comprises a gadolinium chelate.
15183. The method of item 14996 wherein the device comprises a visualization agent, and wherein the visualization agent comprises iron, magnesium, manganese, copper, or chromium. 15184. The method of item 14996 wherein the device comprises a visualization agent, and wherein the visualization agent comprises an iron oxide compound.
15185. The method of item 14996 wherein the device comprises a visualization agent, and wherein the visualization agent comprises a dye, pigment, or colorant.
15186. The method of item 14996 wherein the device comprises an echogenic material.
15187. The method of item 14996 wherein the device comprises an echogenic material, and wherein the echogenic material is in the form of a coating.
15188. The method of item 14996 wherein the device is sterile.
15189. The method of item 14996 wherein the anti- scarring agent is released into tissue in the vicinity of the device after deployment of the device.
15190. The method of item 14996 wherein the anti- scarring agent is released into tissue in the vicinity of the device after deployment of the device, and wherein the tissue is connective tissue.
15191. The method of item 14996 wherein the anti- scarring agent is released into tissue in the vicinity of the device after deployment of the device, and wherein the tissue is muscle tissue. 15192. The method of item 14996 wherein the anti- scarring agent is released into tissue in the vicinity of the device after deployment of the device, and wherein the tissue is nerve tissue.
15193. The method of item 14996 wherein the anti- scarring agent is released into tissue in the vicinity of the device after deployment of the device, and wherein the tissue is epithelium tissue.
15194. The method of item 14996 wherein the anti- scarring agent is released in effective concentrations from the device over a period ranging from the time of deployment of the device to about 1 year.
15195. The method of item 14996 wherein the anti- scarring agent is released in effective concentrations from the device over a period ranging from about 1 month to 6 months.
15196. The method of item 14996 wherein the anti- scarring agent is released in effective concentrations from the device over a period ranging from about 1 - 90 days.
15197. The method of item 14996 wherein the anti- scarring agent is released in effective concentrations from the device at a constant rate.
15198. The method of item 14996 wherein the anti- scarring agent is released in effective concentrations from the device at an increasing rate.
15199. The method of item 14996 wherein the anti- scarring agent is released in effective concentrations from the device at a decreasing rate. 15200. The method of item 14996 wherein the anti- scarring agent is released in effective concentrations from the composition comprising the anti-scarring agent by diffusion over a period ranging from the time of deployment of the device to about 90 days.
15201. The method of item 14996 wherein the anti- scarring agent is released in effective concentrations from the composition comprising the anti-scarring agent by erosion of the composition over a period ranging from the time of deployment of the device to about 90 days.
15202. The method of item 14996 wherein the device comprises about 0.01 Dg to about 10 Dg of the anti-scarring agent.
15203. The method of item 14996 wherein the device comprises about 10 Dg to about 10 mg of the anti-scarring agent.
15204. The method of item 14996 wherein the device comprises about 10 mg to about 250 mg of the anti-scarring agent.
15205. The method of item 14996 wherein the device comprises about 250 mg to about 1000 mg of the anti-scarring agent.
15206. The method of item 14996 wherein the device comprises about 1000 mg to about 2500 mg of the anti-scarring agent.
15207. The method of item 14996 wherein a surface of the device comprises less than 0.01 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied. 15208. The method of item 14996 wherein a surface of the device comprises about 0.01 Dg to about 1 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
15209. The method of item 14996 wherein a surface of the device comprises about 1 Dg to about 10 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
15210. The method of item 14996 wherein a surface of the device comprises about 10 Dg to about 250 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
15211. The method of item 14996 wherein a surface of the device comprises about 250 Dg to about 1000 Dg of the anti-scarring agent of anti-scarring agent per mm2 of device surface to which the anti- scarring agent is applied.
15212. The method of item 14996 wherein a surface of the device comprises about 1000 Dg to about 2500 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
15213. The method of item 14996 wherein the combining is performed by direct affixing the agent or the composition to the implant.
15214. The method of item 14996 wherein the combining is performed by spraying the agent or the component onto the implant. 15215. The method of item 14996 wherein the combining is performed by electrospraying the agent or the composition onto the implant.
15216. The method of item 14996 wherein the combining is performed by dipping the implant into a solution comprising the agent or the composition.
15217. The method of item 14996 wherein the combining is performed by covalently attaching the agent or the composition to the implant.
15218. The method of item 14996 wherein the combining is performed by non-covalently attaching the agent or the composition to the implant.
15219. The method of item 14996 wherein the combining is performed by coating the implant with a substance that contains the agent or the composition.
15220. The method of item 14996 wherein the combining is performed by coating the implant with a substance that absorbs the agent.
15221. The method of item 14996 wherein the combining is performed by interweaving a thread composed of, or coated with, the agent or the composition.
15222. The method of item 14996 wherein the combining is performed by covering all the implant with a sleeve that contains the agent or the composition. 15223. The method of item 14996 wherein the combining is performed by covering a portion of the implant with a sleeve that contains the agent or the composition.
15224. The method of item 14996 wherein the combining is performed by covering all the implant with a cover that contains the agent or the composition.
15225. The method of item 14996 wherein the combining is performed by covering a portion of the implant with a cover that contains the agent or the composition.
15226. The method of item 14996 wherein the combining is performed by covering all the implant with an electrospun fabric that contains the agent or the composition.
15227. The method of item 14996 wherein the combining is performed by covering a portion of the implant with an electrospun fabric that contains the agent or the composition.
15228. The method of item 14996 wherein the combining is performed by covering all the implant with a mesh that contains the agent or the composition.
15229. The method of item 14996 wherein the combining is performed by covering a portion of the implant with a mesh that contains the agent or the composition.
15230. The method of item 14996 wherein the combining is performed by constructing all the implant with the agent or the composition. 15231. The method of item 14996 wherein the combining is performed by constructing a portion of the implant with the agent or the composition.
15232. The method of item 14996 wherein the combining is performed by impregnating the implant with the agent or the composition.
15233. The method of item 14996 wherein the combining is performed by constructing all of the implant from a degradable polymer that releases the agent.
15234. The method of item 14996 wherein the combining is performed by constructing a portion of the implant from a degradable polymer that releases the agent.
15235. The method of item 14996 wherein the combining is performed by dipping the implant into a solution that comprise the agent and an inert solvent for the implant.
15236. The method of item 14996 wherein the combining is performed by dipping the implant into a solution that comprises the agent and a solvent that will swell the implant.
15237. The method of item 14996 wherein the combining is performed by dipping the implant into a solution that comprises the agent and a solvent that will dissolve the implant.
15238. The method of item 14996 wherein the combining is performed by dipping the implant into a solution that comprises the agent, a polymer and an inert solvent for the implant. 15239. The method of item 14996 wherein the combining is performed by dipping the implant into a solution that comprises the agent, a polymer and a solvent that will swell the implant.
15240. The method of item 14996 wherein the combining is performed by dipping the implant into a solution that comprises the agent, a polymer and a solvent that will dissolve the implant.
15241. The method of item 14996 wherein the combining is performed by spraying the implant into a solution that comprises the agent and an inert solvent for the implant.
15242. The method of item 14996 wherein the combining is performed by spraying the implant into a solution that comprises the agent and a solvent that will swell the implant.
15243. The method of item 14996 wherein the combining is performed by spraying the implant into a solution that comprises the agent and a solvent that will dissolve the implant.
15244. The method of item 14996 wherein the combining is performed by spraying the implant into a solution that comprises the agent, a polymer and an inert solvent for the implant.
15245. The method of item 14996 wherein the combining is performed by spraying the implant into a solution that comprises the agent, a polymer and a solvent that will swell the implant.
15246. The method of item 14996 wherein the combining is performed by spraying the implant into a solution that comprises the agent, a polymer and a solvent that will dissolve the implant. 15247. A method as in any one of items 14996-15246, wherein the device is an endotracheal tube.
15248. A method as in any one of items 14996-15246, wherein the device is an endotracheal tube with a single lumen.
15249. A method as in any one of items 14996-15246, wherein the device is an endotracheal tube with double lumens.
15250. A method as in any one of items 14996-15246, wherein the device is a tracheostomy tube.
15251. A method of making a medical device comprising: combining a peritoneal dialysis catheter (e.g., an implant) and an anti-scarring agent or a composition comprising an anti-scarring agent, wherein the agent inhibits scarring between the device and a host into which the device is implanted.
15252. The method of item 15251 wherein the agent is an adensosine A2A receptor antagonist.
15253. The method of item 15251 wherein the agent is an AKT inhibitor.
15254. The method of item 15251 wherein the agent is an alpha 2 integrin antagonist, wherein the alpha 2 integrin antagonist is Pharmaprojects No. 5754 (Merck KgaA).
15255. The method of item 15251 wherein the agent is an alpha 4 integrin antagonist. 15256. The method of item 15251 wherein the agent is an alpha 7 nicotinic receptor agonist.
15257. The method of item 15251 wherein the agent is an angiogenesis inhibitor selected from the group consisting of AG-12,958 (Pfizer), ATN-161 (Attenuon LLC), neovastat, an angiogenesis inhibitor from Jerina AG (Germany), NM-3 (Mercian), VGA-1155 (Taisho), FCE-26644 (Pfizer), FCE-26950 (Pfizer), FPMA (Meiji Daries), FR-111142 (Fujisawa), GGTI-298, GM-1306 (Ligand), GPA-1734 (Novartis), NNC-47-0011 (Novo Nordisk), herbamycin (Nippon Kayaku), lenalidomide (Celegene), IP-10 (NIH), ABT-828 (Abbott), KIN-841 (Tokushima University, Japan), SF-1126 (Semafore Pharmaceuticals), laminin technology (NIH), CHIR-258 (Chiron), NVP-AEW541 (Novartis), NVP-AEW541 (Novartis), Vt16907 (Alchemia), OXI-8007 (Oxigene), EG-3306 (Ark Therapeutics), Maspin (Arriva), ABT- 567 (Abbott), PPI-2458 (Praecis Pharmaceuticals), CC-5079, CC-4089 (Celgene), HIF-1 alpha inhibitors (Xenova), S-247 (Pfizer), AP-23573 (Ariad), AZD-9935 (Astra Zeneca), mebendazole (Introgen Therapeutics), MetAP-2 inhibitors (GlaxoSmithKline), AG-615 (Angiogene Pharmaceuticals), Tie-2 antagonists (Hybrigenics), NC-381, CYC-381, NC-169, NC-219, NC-383, NC-384, NC-407 (Lorus Therapeutics), ATN-224 (Attenuon), ON-01370 (Onconova), Vitronectin antagonists (Amgen), SDX-103 (Salmedix), Vitronectin antagonists (Shire), CHP (Riemser), TEK (Amgen), Anecortave acetate (Alcon), T46.2 (Matrix Therapeutics), HG-2 (Heptagen), TEM antagonists (Genzyme), Oxi-4500 (Oxigene), ATN-161 (Attenuon), WX-293 (Wilex), M-2025 (Metris Therapeutics), Alphastatin (BioActa), YH-16 (Yantai Rongchang), BIBF-1120 (Boehringer Ingelheim), BAY-57-9352 (Bayer), AS- 1404 (Cancer Research Technology), SC-77964 (Pfizer), glycomimetics (BioTie Therapies), TIE-2 Inhibitors (Ontogen), DIMI, Octamer (Octamer), ABR-215050 (Active Biotech), ABT-518 (Abbott), KDR inhibitors (Abbott), BSF-466895 (Abbott), SCH-221153 (Schering-Plough), DAC.antiangiogenic (ConjuChem), TFPI (EntreMed), AZD-2171 (Astra-Zenaca), CDC-394 (Celgene), LY290293 (EIi Lilly), IDN-5390 (Indena), Kdr Kinase Inhibitors (Merck), CT-113020, CT-116433, CT-116563, CT-31890, CT-32228) (Cell Therapeutics), A-299620 (Abbott), TWEAK Inhibitor (Amgen), VEGF modulators (Johnson and Johnson), Tum-N53, tumstatin (Genzyme), Thios- 1 , Thios-2 (Thios Pharmaceuticals), MV-6401 (Miravant Medical Technologies), Spisulosine (PharmaMar), CEP-7055 (Cephalon), AUV-201 (Auvation), LM-609 (EIi Lilly), SKF-106615 (AnorMED), Oglufanide disodium (Cytran), BW-114 (Pharminox), Calreticulin (NIH), WX-678 (Wilex), SD-7784 (Pfizer), WX-UK1 (Wilex), SH-268 (Schering AG), 2-Me-PGA (Celgene), S- 137 (Pfizer), ZD-6126 (Angiogene Pharmaceuticals), SG-292 (SignalGen), Benefin (Lane Labs), A6, A36 (Angstrom), SB-2723005 (GlaxoSmithKline), SC-7 (Cell Therapeutics), ZEN-014 (AEterna Zentaris), 2-methoxyestradioi (EntreMed), NK-130119 (Nippon Kayaku), CC-10004 (Celgene), AVE- 8062A (Ajinomoto), Tacedinaline (Pfizer), Actinonin (Tokyo Metropolitan Institute of Medical Science), Lenalidomide (Celgene), VGA-1155, BTO-956 (SRI International), ER-68203-00 (Eisai), CT-6685 (UCB), JKC-362 (Phoenix Pharmaceuticals), DMI-3798 (DMI Biosciences, Angiomate (Ipsen), ZD-6474 (AstraZeneca), CEP-5214 (Cephalon), Canstatin (Genzyme), NM-3 (Mercian), Oridigm (MediQuest Therapeutics), Exherin (Adherex), BLS-0597 (Boston Life Sciences), PTC-299 (PTC Therapeutics), NPI-2358 (Nereus Pharmaceuticals), CGP-79787 (Novartis), AEE-788 (Novartis), CKD-732 (Chong Kun Dang), CP-564959 (OSI Pharmaceuticals), CM-101 (CarboMed), CT-2584, CT3501 (Cell Therapeutics), combretastatin and analogues and derivatives thereof (Oxigene), Rebimastat (Bristol-Meyers Squibb), Dextrin 2-sulfate (ML Laboratories), Cilengitide (Merk KGaA), NSC-706704 (Pharminox), KRN- 951 (Kirin Brewery), Ukrain, NSC-631570 (Nowicky Pharma), Tecogalan sodium (Daiichi Pharmaceutical), Tz-93 (Tsumura), TBC-1635 (Encysive Pharmaceuticals), TAN-1120 (Takeda), Semaxanib (Pfizer), BDI-7800 (Biopharmacopae), SD-186, SD-983 (Bristol-Meyers Squibb), SB-223245 (GlaxoSmithKline), SC-236 (Pfizer), RWJ-590973 (Johnson and Johnson), ILX-1850 (Genzyme), SC-68488, S-836 (Pfizer), CG-55069-11 (CuraGen), Ki-23057 (Kirin Brewery), CCX-700 (Chemoentryx), Pegaptanib octasodium (Giled Sciences), ANGIOCOL (available from Biostratum Inc.), or an analogue or derivative thereof.
15258. The method of item 15251 wherein the agent is an apoptosis antagonist.
15259. The method of item 15251 wherein the agent is an apoptosis activator.
15260. The method of item 15251 wherein the agent is a beta 1 integrin antagonist.
15261. The method of item 15251 wherein the agent is a beta tubulin inhibitor.
15262. The method of item 15251 wherein the agent is a blocker of enzyme production in Hepatitis C.
15263. The method of item 15251 wherein the agent is a Bruton's tyrosine kinase inhibitor.
15264. The method of item 15251 wherein the agent is a calcineurin inhibitor.
15265. The method of item 15251 wherein the agent is a caspase 3 inhibitor.
15266. The method of item 15251 wherein the agent is a CC chemokine receptor antagonist. 15267. The method of item 15251 wherein the agent is a cell cycle inhibitor selected from the group consisting of SNS-595 (Sunesis), synthadotin, KRX-0403, and an analogue or derivative thereof.
15268. The method of item 15251 wherein the agent is a cathepsin B inhibitor.
15269. The method of item 15251 wherein the agent is a cathepsin K inhibitor, wherein the cathepsin K inhibitor is 462795 (GlaxoSmithKline), INPL-022-D6 (Amura Therapeutics), or an analogue or derivative thereof.
15270. The method of item 15251 wherein the agent is a cathepsin L inhibitor.
15271. The method of item 15251 wherein the agent is a CD40 antagonist.
15272. The method of item 15251 wherein the agent is a chemokine receptor agonist.
15273. The method of item 15251 wherein the agent is a chymase inhibitor.
15274. The method of item 15251 wherein the agent is a collagenase antagonist.
15275. The method of item 15251 wherein the agent is a CXCR antagonist. 15276. The method of item 15251 wherein the agent is a cyclin dependent kinase inhibitor selected from the group consisting of a CDK-1 inhibitor, a CDK-2 inhibitor, a CDK- 4 inhibitor, a CDK-6 inhibitor, a CAK1 inhibitor from GPC Biotech and Bristol-Myers Squibb, RGB-286199 (GPC Biotech), an anticancer agent from Astex Technology, a CAK1 inhibitor from GPC Biotech, a CDK inhibitor from Sanofi-Aventis, a CDK1/CDK2 inhibitor from Amgen, a CDK2 inhibitor from SUGEN-2 (Pfizer), a hearing loss therapy agent (Sound Pharmaceuticals), PD-0332991 (Pfizer), RGB-286199 (GPC Biotech), Ro-0505124 (Hoffmann-La Roche), a Ser/Thr kinase inhibitor from Lilly (EIi Lilly), CVT-2584 (CAS No. 199986-75- 9) (CV Therapeutics), and an analogue or derivative thereof.
15277. The method of item 15251 wherein the agent is a cyclooxygenase 1 inhibitor.
15278. The method of item 15251 wherein the agent is a DHFR inhibitor.
15279. The method of item 15251 wherein the agent is a dual integrin inhibitor.
15280. The method of item 15251 wherein the agent is an elastase inhibitor.
15281. The method of item 15251 wherein the agent is an elongation factor-1 alpha inhibitor.
15282. The method of item 15251 wherein the agent is an endothelial growth factor antagonist. 15283. The method of item 15251 wherein the agent is an endothelial growth factor receptor kinase inhibitor selected from the group consisting of sorafenib tosylate (Bayer), AAL-993 (Novartis), ABP-309 (Novartis), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), E- 7080 (Eisai), EG-3306 (Ark Therapeutics), EXEL-2880 (Exelixis), GW- 654652 (GlaxoSmithKline), a KDR inhibitor from LG Life Sciences, CT-6685 and CT-6729 (UCB), KRN-633 and KRN-951 (Kirin Brewery), OSI-930 (OSI Pharmaceuticals), SP-5.2 (Supratek Pharma), SU-11657 (Pfizer), a Tie-2 antagonist (Hybrigenics), a VEGFR-2 kinase inhibitor (Bristol-Myers Squibb), XL-647 (Exelixis), a KDR inhibitor from Abbott Laboratories, sorafenib tosylate, and an analogue or derivative thereof.
15284. The method of item 15251 wherein the agent is an endotoxin antagonist.
15285. The method of [tern 15251 wherein the agent is an epothilone and tubulin binder.
15286. The method of item 15251 wherein the agent is an estrogen receptor antagonist.
15287. The method of item 15251 wherein the agent is an FGF inhibitor.
15288. The method of item 15251 wherein the agent is a famexyl transferase inhibitor.
15289. The method of item 15251 wherein the agent is farnesyltransferase inhibitor selected from the group of A-197574 (Abbott), a famesyltransferase inhibitor from Servier, FPTIII (Strathclyde Institute for Drug R), LB-42908 (LG Life Sciences), Pharmaprojects No. 5063 (Genzyme), Pharmaprojects No. 5597 (Ipsen), Yissum Project No. B-1055 (Yissum), and an analogue or derivative thereof.
15290. The method of item 15251 wherein the agent is an FLT-3 kinase inhibitor.
15291. The method of item 15251 wherein the agent is an FGF receptor kinase inhibitor.
15292. The method of item 15251 wherein the agent is a fibrinogen antagonist selected from the group consisting of AUV-201 (Auvation), MG-13926 (Sanofi-Aventis), plasminogen activator (CAS No. 105913-11-9) (from Sanofi-Aventis or L)CB), plasminogen activator-2 (tPA- 2) (Sanofi-Aventis), pro-urokinase (CAS No. 82657-92-9) (Sanofi-Aventis), and an analogue or derivative thereof.
15293. The method of item 15251 wherein the agent is a heat shock protein 90 antagonist selected from the group consisting of SRN- 005 (Sirenade), geldanamycin, NSC-33050 (17-allylaminogeldanamycin; 17- AAG), 17-dimethylaminoethylamino-17-demethoxy-geldanamycin (17- DMAG), rifabutin (rifamycin XIV, 1',4-didehydro-1-deoxy-1 ,4-dihydro-5'-(2- methylpropyl)-1-oxo-), and an analogue or derivative thereof.
15294. The method of item 15251 wherein the agent is a histone deacetylase inhibitor.
15295. The method of item 15251 wherein the agent is an HMGCoA reductase inhibitor selected from the group consisting of an atherosclerosis therapeutic from Lipid Sciences, ATM 6000 (ARYx Therapeutics), KS-01-019 (Kos Pharmaceuticals), Pharmaprojects No. 2197 (Sanofi-Aventi), RP 61969 (Sanofi-Aventis), and an analogue or derivative thereof.
15296. The method of item 15251 wherein the agent is an ICAM inhibitor.
15297. The method of item 15251 wherein the agent is an IL1 ICE and IRAK antagonist, wherein the antagonist is a CJ-14877, CP- 424174 (Pfizer), NF-61 (Negma-Lerads), and an analogue or derivative thereof.
15298. The method of item 15251 wherein the agent is an IL-2 inhibitor.
15299. The method of item 15251 wherein the agent is an immunosuppressant selected from the group consisting of teriflunomide (Sanofi Aventis), chlorsulfaquinoxalone (NSC-339004), chlorsulfaquinoxalone sulfate, CS-712 (Sankyo), ismomultin alfa (CAS No. 457913-93-8) (Akzo Nobel), antiallergics from GenPat77, antiinflammatories or AT-005 (Androclus Therapeutics), autoimmune disease therapeutics from EpiVax, BN-007 (Bone), budesonide (CAS No. 51333-22- 3) (MAP Pharmaceuticals), CO-14 (Genzyme), edratide (CAS No. 433922- 67-9) (Teva), EP-314 (Enanta), eprovafen (CAS No. 101335-99-3) (Sanofi- Aventis), HWA-131 (CAS No. 118788-41-3) (Sanofi-Aventis), immunomodulators from MerLion Pharmaceuticals, immunosuppressives from Alchemia, IPL-12 (Inflazyme), MDL-9563 (CAS No. 27086-86-8) (Sanofi-Aventis), Pharmaprojects No. 2330 (Sanofi-Aventis), Pharmaprojects No. 6426 (Abgenix), PXS-25 (Pharmaxis), rosmarinic acid (CAS No. 20283-92-5) (Sanofi-Aventis), RP 42927 or RP 54745 (CAS No. 135330-08-4) (Sanofi-Aventis), SGN-35 (Seattle Genetics), ST-1959 (Sigma-Tau), type I diabetes therapy from SYNX Pharma, UNIL-88 (Debiopharm), VP-025 (Vasogen), VR-694 (Vectura), PRTX-001 (Protalex), and an analogue or derivative thereof.
15300. The method of item 15251 wherein the agent is an IMPDH (inosine monophosphate).
15301. The method of item 15251 wherein the agent is an integrin antagonist.
15302. The method of item 15251 wherein the agent is an interleukin antagonist.
15303. The method of item 15251 wherein the agent is an inhibitor of type III receptor tyrosine kinase.
15304. The method of item 15251 wherein the agent is an irreversible inhibitor of enzyme methionine aminopeptidase type 2.
15305. The method of item 15251 wherein the agent is an isozyme selective delta protein kinase C inhibitor.
15306. The method of item 15251 wherein the agent a JAK3 enzyme inhibitor.
15307. The method of item 15251 wherein the agent is a JNK inhibitor.
15308. The method of item 15251 wherein the agent is a kinase inhibitor. 15309. The method of item 15251 wherein the agent is kinesin antagonist.
15310. The method of item 15251 wherein the agent is a kinesin antagonist.
15311. The method of item 15251 wherein the agent is a leukotriene inhibitor and antagonist selected from the group consisting of ambicromil (CAS No. 58805-38-2) (Sanofi-Aventis), amelubant (CAS No. 346735-24-8) (Boehringer Ingelheim), DW-1141 (Dong Wha), ebselen (Daiichi Pharmaceutical), ibudilast (Kyorin), leucotriene inhibitors from Sanofi-Aventis, lymphotoxin-beta receptor (LT-D) from Biogen Idee, Pharmaprojects No. 1535 and 2728 (CAS No. 119340-33-9) (Sanofi- Aventis), R-112 (Rigel), Rev-5367 (CAS No. 92532-05-3) (Sanofi-Aventis), RG-14893 (CAS No. 141835-49-6) (Sanofi-Aventis), RG-5901-A (CAS No. 101910-24-1), 92532-23-5, RP 66153 (CAS No. 142422-79-5), RP 66364 (CAS No.186916-92-5), RP 69698 (CAS No. 141748-00-7) (Sanofi-Aventis), SC-411930 (Pfizer), SC-41930 (CAS No. 120072-59-5) (Pfizer), SC-50605 (CAS No. 138828-39-4) (Pfizer), SC-51146 (CAS No. 141059-52-1), SC- 53228 (CAS No. 153633-01-3) (Pfizer), spaglumic acid (ZY-15106) (CAS No.3106-85-2), 80619-64-3 (Novartis), tipredane (CAS No. 85197-77-9) (Bristol-Myers Squibb), U-75302 (CAS No. 119477-85-9) (Pfizer), and analogue or derivative thereof.
15312. The method of item 15251 wherein the agent is an MAP kinase inhibitor.
15313. The method of item 15251 wherein the agent is a matrix metalloproteinase inhibitor. 15314. The method of item 15251 wherein the agent is an MCP-CCR2 inhibitor.
15315. The method of item 15251 wherein the agent is an mTOR inhibitor.
15316. The method of item 15251 wherein the agent is an mTOR kinase inhibitor.
15317. The method of item 15251 wherein the agent is a microtubule inhibitor selected from the group consisting of antibody- maytansinoid conjugates from Biogen Idee, colchicines (MantiCore Pharmaceuticals), anticancer immunoconjugates from Johnson & Johnson, DIME from Octamer, gni-1f (GNI), huC242-DM4, huMy9-6-DM1 (ImmunoGen), IDN-5404 (Indena), IMO-098, IMOderm (Imotep), mebendazole (Introgen Therapeutics), microtubule poisons from Cambridge Enterprise, paclitaxel such as LOTAX from Aphios (CAS No. 33069-62-4), Genexol-PM from Samyang, Pharmaprojects No. 6383 (Tapestry Pharmaceuticals), RPR-112378 (Sanofi-Aventis), SGN-75 (Seattle Genetics), SPL-7435 (Starpharma), SSR-250411 (Sanofi-Aventis), trastuzumab-DM1 (Genentech), vinorelbine, and an analogue or derivative thereof.
15318. The method of item 15251 wherein the agent is an MIF inhibitor.
15319. The method of item 15251 wherein the agent is an MMP inhibitor.
15320. The method of item 15251 wherein the agent is a neurokinin (NK) antagonist selected from the group consisting of anthrotainin (CAS No. 148084-40-6) (Sanofi-Aventis), an IBS thereapeutic from ArQuIe, MDL-105212A (CAS No. 167261-60-1) (Ssanofi-Aventis), Pharmaprojects No. 2744, 3258 (CAS No. 139167-47-8) 4006, 4201, or 5986 (Sanofi-Aventis), RP 67580 (CAS No. 135911-02-3), SR-144190 (CAS No. 201152-86-5), SSR-240600, SSR-241586 (Sanofi-Aventis), TKA-457 (Novartis), vestipitant mesylate (CAS No. 334476-64-1) (GlaxoSmithKline), Win-64821 (Sanofi-Aventis), PRX-96026 (Predix Pharmaceuticals), and an analogue or derivative thereof.
15321. The method of item 15251 wherein the agent is an NF kappa B inhibitor selected from the group consisting of AVE-0545 or AVE-0547 (Sanofi-Aventis), bortezomib (CAS No. 179324-69-7) (Millennium Pharmaceuticals), dexanabinol (CAS No. 112924-45-5) (Pharmos), dexlipotam (Viatris), Pharmaprojects No. 6283 (INDRA) (OXiGENE), IPL- 576092 (CAS No. 137571-30-3) (Inflazyme), NFKB decoy (Corgentech), NFKB decoy oligo (AnGes MG), NFKB's from Ariad, osteoporosis, treatments or S5 (F005) from Fulcrum Pharmaceuticals, P61 (Phytopharm), R-flurbiprofen (CAS No. 5104-49-4) (Encore Pharmaceuticals), and an analogue or derivative thereof.
15322. The method of item 15251 wherein the agent is a nitric oxide agonist.
15323. The method of item 15251 wherein the agent is an ornithine decarboxylase inhibitor.
15324. The method of item 15251 wherein the agent is a p38 MAP kinase inhibitor selected from the group consisting of AZD-6703 (AstraZeneca), JX-401 (Jexys Pharmaceuticals), BMS-2 (Bristol-Myers Squibb), a p38 MAP kinase inhibitor from Novartis, a p38-alpha MAP kinase inhibitor from Amphora, Pharmaprojects No. 5704 (Pharmacopeia), RPR- 200765A (Sanofi-Aventis), SD-282 (Johnson & Johnson), TAK-715 (Takeda), and an analogue or derivative thereof.
15325. The method of item 15251 wherein the agent is a palmitoyl-protein thioesterase inhibitor.
15326. The method of item 15251 wherein the agent is a PDGF receptor kinase inhibitor selected from the group consisting of AAL- 993, AMN-107, or ABP-309 (Novartis), AMG-706 (Amgen), BAY-57-9352 (Bayer), CDP-860 (UCB), E-7080 (Eisai), imatinib (CAS No. 152459-95-5) (Novartis), OSI-930 (OSI Pharmaceuticals), RPR-127963E (Sanofi-Aventis), RWJ-540973 (Johnson & Johnson), sorafenib tosylate (Bayer), SU-11657 (Pfizer), tandutinib (CAS No. 387867-13-2) (Millennium Pharmaceuticals), vatalanib (Novartis), ZK-CDK (Schering AG), and an analogue or derivative thereof.
15327. The method of item 15251 wherein the agent is (-)-halofenate (Metabolex), AMG-131 (Amgen), antidiabetics from Japan Tobacco, AZD-4619, AZD-8450, AZD-8677 (AstraZeneca), DRF-10945, balaglitazone (Dr Reddy's), CS-00088, CS-00098 (Chipscreen Biosciences), E-3030 (Eisai), etalocib (CAS No. 161172-51-6) (EIi Lilly), GSK-641597 (Ligand), GSK-677954 (GlaxoSmithKline), GW-409544 (Ligand), GW- 590735 (GlaxoSmithKline), K-111 (Hoffmann-La Roche), LY-518674 (EIi Lilly), LY-674 (Ligand), LY-929 (Ligand), MC-3001 , MC-3002 (MaxoCore Pharmaceuticals), metformin HCI + pioglitazone (CAS No. 1115-70-4 and
112529-15-4), ACTOPLUS MET from Andrx), muraglitazar (CAS No. 331741-94-7) (Bristol-Myers Squibb), naveglitazar (Ligand), oleoylethanolamide (Kadmus Pharmaceuticals), ONO-5129, pioglitazone hydrochloride (CAS No. 111025-46-8 and 112529-15-4) (Takeda), PLX-204 (Plexxikon), PPAR agonists from Genfit, PPAR delta agonists from EIi Lilly, PPAR-alpha agonists from CrystalGenomics, PPAR-gamma modulators and PPAR-β modulators from CareX, rosiglitazone maleate (CAS No. 122324- 73-4 or 155141-29-0) (GlaxoSmithKline), rosiglitazone maleate/glimepir (CAS No. 155141-29-0 and 93479-97-1), AVANDARYL, rosiglitazone maleate/metformin extend (CAS No. 155141-29-0 and 657-24-9), AVANDAMET, rosiglitazone maleate+metformin, AVANDAMET (GlaxoSmithKline), tesaglitazar (AstraZeneca), and an analogue or derivative thereof.
15328. The method of item 15251 wherein the agent is a phosphatase inhibitor.
15329. The method of item 15251 wherein the agent is a phosphodiesterase (PDE) inhibitor selected from the group consisting of avanafil (Tanabe Seiyaku), dasantafil (CAS No. 569351-91-3) (Schering- Plough), A-906119 (CAS No. 134072-58-5), DL-850 (Sanofi-Aventis), GRC- 3015, GRC-3566, GRC-3886 (Glenmark), HWA-153 (CAS No. 56395-66-5) (Sanofi-Aventis), hydroxypumafentrine (Altana), IBFB-130011 , IBFB-14-016, IBFB-140301 , IBFB-150007, IBFB-211913 (IBFB Pharma), L-826141 (Merck & Co), medorinone (CAS No. 88296-61-1) (Sanofi-Aventis), MEM- 1917 (Memory Pharmaceuticals), ND-1251 (Neuro3d), PDE inhibitors from ICOS, PDE IV inhibitors from Memory Pharmaceuticals and CrystalGenomics, Pharmaprojects No. 2742 and 6141 (Sanofi-Aventis), QAD-171 (Novartis), RHC-2963 (CAS No. 76993-12-9 and 76993-14-1), RPR-117658, RPR-122818 derivatives, SR-24870 , and RPR-132294 (Sanofi-Aventis), SK-350 (ln2Gen), stroke therapy agents from deCODE Genetics, TAS-203 (Taiho), tofimilast (CAS No. 185954-27-2) (Pfizer), UK- 371800 (Pfizer), WIN-65579 (CAS No. 158020-82-7) (Sanofi-Aventis), IBFB- 130020 (IBFB Pharma), OPC-6535 (CAS No. 145739-56-6) (Otsuka), a phosphodiesterase III inhibitor, enoximone, a phosphodiesterase IV inhibitor, fosfosal, Atopik (Barrier Therapeutics), triflusal, a phosphodiesterase V inhibitor, and an analogue or derivative thereof. 15330. The method of item 15251 wherein the agent is a PKC inhibitor.
15331. The method of item 15251 wherein the agent is a platelet activating factor antagonist.
15332. The method of item 15251 wherein the agent is a platelet-derived growth factor receptor kinase inhibitor.
15333. The method of item 15251 wherein the agent is a prolyl hydroxylase inhibitor.
15334. The method of item 15251 wherein the agent is a polymorphonuclear neutrophil inhibitor.
15335. The method of item 15251 wherein the agent is a protein kinase B inhibitor.
15336. The method of item 15251 wherein the agent is a protein kinase C stimulant.
15337. The method of item 15251 wherein the agent is a purine nucleoside analogue.
15338. The method of item 15251 wherein the agent is a purinoreceptor P2X antagonist.
15339. The method of item 15251 wherein the agent is a Raf kinase inhibitor. 15340. The method of item 15251 wherein the agent is a reversible inhibitor of ErbB1 and ErbB2.
15341. The method of item 15251 wherein the agent is a ribonucleoside triphosphate reductase inhibitor.
15342. The method of item 15251 wherein the agent is an SDF-1 antagonist.
15343. The method of item 15251 wherein the agent is a sheddase inhibitor.
15344. The method of item 15251 wherein the agent is an SRC inhibitor.
15345. The method of item 15251 wherein the agent is a stromelysin inhibitor.
15346. The method of item 15251 wherein the agent is an Syk kinase inhibitor.
15347. The method of item 15251 wherein the agent is a telomerase inhibitor.
15348. The method of item 15251 wherein the agent is a TGF beta inhibitor selected from the group consisting of pirfenidone (CAS No. 53179-13-8) (MARNAC), tranilast (CAS No. 53902-12-8) (Kissei), IN- 1130 (ln2Gen), mannose-6-phosphate (BTG), TGF-β antagonists from Inflazyme (Pharmaprojects No. 6075), TGF-β antagonists from Sydney, non-industrial source), TGF-β I receptor kinase inhibitors from EIi Lilly, TGF- β receptor inhibitors from Johnson & Johnson, and an analogue or derivative thereof.
15349. The method of item 15251 wherein the agent is a
TNFα antagonist or TACE inhibitor selected from the group consisting of adalimumab (CAS No. 331731-18-1) (Cambridge Antibody Technology), AGIX-4207 (AtheroGenics), AGT-1 (Advanced Biotherapy), an antiinflammatory from Borean Pharma, Cellzome, or Paradigm Therapeutics, anti-inflammatory vaccine (TNF-alpha kinoid) from Neovacs, humanized anti-TNF antibody or an anti-TNF MAb (CB0006) Celltech (UCB), apratastat (CAS No. 287445-51-0) (Wyeth), BMS-561392 (Bristol-Myers Squibb), BN- 006 (Bone), certolizumab pegol (CAS No. 428863-50-7 or CH-138 (UCB), cilomilast (CAS No. 153259-65-5) (GlaxoSmithKline), CR-1 (Nuada Pharmaceuticals), CRx-119 (CombinatoRx), D-5410 (UCB), dacopafant (CAS No. 125372-33-0) (Sanofi-Aventis), dersalazine (CAS No. 188913-57- 7/188913-58-8) (Uriach), etanercept (CAS No. 185243-69-0) (Amgen), ethyl pyruvate (Critical (Critical Therapeutics), golimumab (CAS No. 476181-74-5) (Johnson & Johnson), hormono-immunotherapy from Ipsen, CDP571 (e.g., Humicade from UCB), IC-485 (ICOS), infliximab (CAS No. 170277-31-3) (Johnson & Johnson), IP-751 (Manhattan Pharmaceuticals), ISIS-104838 (CAS No. 250755-32-9) (ISIS Pharmaceuticals), lenalidomide (CAS No. 191732-72-6) (Celgene), lentinan (CAS No. 37339-90-5) (Ajinomoto), MDL- 201112 (CAS No. 142130-73-2) (Sanofi-Aventis), medroxyprogesterone (CAS No. 520-85-4) (InKine Pharmaceutical), N-acetylcysteine (CAS No. 616-91-1) (Zambon), NBE-P2 (DIREVO Biotech), nerelimomab (CAS No. 162774-06-3) (Chiron), OM-294DP (OM PHARMA), onercept (CAS No. 199685-57-9) (Yeda), PASSTNF-alpha (Verigen), pentoxifylline or oxypentifylline (Sanofi-Aventis), Pharmaprojects No. 4091 , 4241 , 4295, or 4488 (Sanofi-Aventis), Pharmaprojects No. 5480 (Amgen), Pharmaprojects No. 6749 (Cengent), pirfenidone (CAS No. 53179-13-8) (MARNAC), RPR- 132294 (Sanofi-Aventis), S5 (F002) (Fulcrum Pharmaceuticals), simvastatin (CAS No. 79902-63-9) (Merck & Co), STA-6292 (Synta Pharmaceuticals), tacrolimus (CAS No. 104987-11-3) (Fujisawa LifeCycle Pharma), talactoferrin alfa (CAS No. 308240-58-6) (Agennix), thalidomide (CAS No. 50-35-1) (Celgene), TNF antagonists form ProStrakan, and Synergen, TNF inhibitors (Amgen), TNF-alpha antagonists from Dynavax Technologies and Jerina AG (Germany), TNF-alpha inhibitors from IBFB Pharma and Xencor (Xencor), torbafylline (CAS No. 105102-21-4) (Sanofi-Aventis), UR-1505 (Uriach), VT-346 (Viron Therapeutics), YSIL6 (Y1S Therapeutics), YSTH2 (Y's Therapeutics), NPI-1302a-3 (Nereus Pharmaceuticals, a TNF antagonist from Jerina AG (Germany), dersalazine, and an analogue or derivative thereof.
15350. The method of item 15251 wherein the agent is a Toll receptor inhibitor.
15351. The method of item 15251 wherein the agent is a tubulin antagonist.
15352. The method of item 15251 wherein the agent is a tyrosine kinase inhibitor selected from the group consisting of SU-011248, SUTENT from Pfizer Inc. (New York, NY), BMS-354825, PN-355 (Paracelsian Pharmaceuticals), AGN-199659 (Allergan), AAL-993 or ABP- 309 (Novartis), adaphostin (NIH), AEE-788 (Novartis), AG-013736 (OSI Pharmaceuticals), AG-13736 (Pfizer), ALT-110 (Alteris Therapeutics), AMG- 706 (Amgen), anticancer MAbs from Xencor, anti-EGFrvlll MAbs from Abgenix, anti-HER2 MAb from Abiogen, AZD-2171 or AZD-9935 (AstraZeneca), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), CEP-5214 (Cephalon), CEP-7055 (Cephalon), cetuximab (ImClone Systems), CHIR-200131 and CHIR-258 (Chiron), CP-547632 (OSI Pharmaceuticals), CP-724714 (Pfizer), CT-301 (Creabilis Therapeutics), D- 69491 (Baxter International), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EGFR/ErbB2 inhibitors from Array BioPharma, erlotinib (CAS No. 183319- 69-9) (OSI Pharmaceuticals), EXEL-2880 (Exelixis), FK-778 (Sanofi- Aventis) , gefitinib (CAS No. 184475-35-2) (AstraZeneca), GW-2286 or GW- 654652 (GlaxoSmithKline), her2/neu antigen from AlphaVax, HER-2/neu inhibitor from Generex, Herzyme (Medipad) (Sima Therapeutics), HKI-272 (Wyeth), HuMax-EGFr (Genmab), idronoxil (CAS No. 81267-65-4) (Novogen), IGF-1 inhibitors from Ontogen, IMC-11F8 (ImCIone Systems), kahalalide F (CAS No. 149204-42-2) (PharmaMar), KDR inhibitor from LG Life Sciences, KDR inhibitors from Abbott Laboratories, KDR kinase inhibitors (UCB), Kdr kinase inhibitors from Merck & Co, KRN-633 and KRN- 951 (Kirin Brewery), KSB-102 (Xenova), lapatinib ditosylate (CAS No. 388082-78-8) (GlaxoSmithKline), matuzumab (Merck KGaA), MDX-214 (Medarex), ME-103 (Pharmexa), MED-A300 (Gerolymatos), MNAC-13 (Lay Line Genomics), nimotuzumab (Center of Molecular Immunology), NSC- 330507 or NSC-707545 (NIH)1 NV-50 (Novogen), OSI-930 (OSI Pharmaceuticals), panitumumab (Abgenix), pelitinib (CAS No. 287933-82-7) (Wyeth), pertuzumab (CAS No. 380610-27-5) (Genentech), Pharmaprojects No. 3985 (Sanofi-Aventis), prostate cancer therapeutics from Sequenom (SQPC35, SQPC36, SQPC90), removab and remoxab (Trion Pharma), RG- 13022 (CAS No. 136831-48-6), RG-13291 (CAS No. 138989-50-1), or RG- 14620 (CAS No. 136831-49-7) (Sanofi-Aventis), RM-6427 (Romark), RNAi breast cancer therapy from Benitec, RP 53801 (CAS No. 125882-88-4) (Sanofi-Aventis), sorafenib tosylate (Bayer), SU-11657 (Pfizer), Tie-2 antagonists from Semaia (Hybrϊgenics), Tie-2 inhibitors from Ontogen, trastuzumab (CAS No. 180288-69-1) (Genentech), tyrosine kinase inhibitors from Sanofi-Aventis, U3-1287, U3-1565, U3-1784, U3-1800 (U3 Pharma), vatalanib (Novartis), VEGFR-2 kinase inhibitor from Bristol-Myers Squibb, XL-647 (Exelixis), ZD-6474 (AstraZeneca), ZK-CDK (Schering AG), an EGFR tyrosine kinase inhibitor, EKB-569 (Wyeth), and an analogue or derivative thereof. 15353. The method of item 15251 wherein the agent is a VEGF inhibitor.
15354. The method of item 15251 wherein the agent is a vitamin D receptor agonist.
15355. The method of item 15251 wherein the agent is ZD-6474 (an angiogenesis inhibitor).
15356. The method of item 15251 wherein the agent is AP-23573 (an mTOR inhibitor).
15357. The method of item 15251 wherein the agent is synthadotin (a tubulin antagonist).
15358. The method of item 15251 wherein the agent is S-0885 (a collagenase inhibitor).
15359. The method of item 15251 wherein the agent is aplidine (an elongation factor-1 alpha inhibitor).
15360. The method of item 15251 wherein the agent is ixabepilone (an epithilone).
15361. The method of item 15251 wherein the agent is IDN-5390 (an angiogenesis inhibitor).
15362. The method of item 15251 wherein the agent is SB-2723005 (an angiogenesis inhibitor). 15363. The method of item 15251 wherein the agent is ABT-518 (an angiogenesis inhibitor).
15364. The method of item 15251 wherein the agent is combretastatin (an angiogenesis inhibitor).
15365. The method of item 15251 wherein the agent is anecortave acetate (an angiogenesis inhibitor).
15366. The method of item 15251 wherein the agent is SB-715992 (a kinesin antagonist).
15367. The method of item 15251 wherein the agent is temsirolimus (an mTOR inhibitor).
15368. The method of item 15251 wherein the agent is adalimumab (a TNFα antagonist).
15369. The method of item 15251 , wherein the composition comprises a polymer.
15370. The method of item 15251 , wherein the composition comprises a polymeric carrier.
15371. The method of item 15251 wherein the anti- scarring agent inhibits adhesion between the device and a host into which the device is implanted.
15372. The method of item 15251 wherein the device delivers the anti-scarring agent locally to tissue proximate to the device. 15373. The method of item 15251 wherein the device has a coating that comprises the anti-scarring agent.
15374. The method of item 15251 , wherein the device has a coating that comprises the agent and is disposed on a surface of the implant.
15375. The method of item 15251 , wherein the device has a coating that comprises the agent and directly contacts the implant.
15376. The method of item 15251 , wherein the device has a coating that comprises the agent and indirectly contacts the implant.
15377. The method of item 15251 , wherein the device has a coating that comprises the agent and partially covers the implant.
15378. The method of item 15251 , wherein the device has a coating that comprises the agent and completely covers the implant.
15379. The method of item 15251 , wherein the device has a uniform coating.
15380. The method of item 15251, wherein the device has a non-uniform coating.
15381. The method of item 15251 , wherein the device has a discontinuous coating.
15382. The method of item 15251 , wherein the device has a patterned coating. 15383. The method of item 15251 , wherein the device has a coating with a thickness of 100 Dm or less.
15384. The method of item 15251 , wherein the device has a coating with a thickness of 10 Dm or less.
15385. The method of item 15251 , wherein the device has a coating, and the coating adheres to the surface of the implant upon deployment of the implant.
15386. The method of item 15251 , wherein the device has a coating, and wherein the coating is stable at room temperature for a period of 1 year.
15387. The method of item 15251 , wherein the device has a coating, and wherein the anti-scarring agent is present in the coating in an amount ranging between about 0.0001% to about 1% by weight.
15388. The method of item 15251 , wherein the device has a coating, and wherein the anti-scarring agent is present in the coating in an amount ranging between about 1% to about 10% by weight.
15389. The method of item 15251 , wherein the device has a coating, and wherein the anti-scarring agent is present in the coating in an amount ranging between about 10% to about 25% by weight.
15390. The method of item 15251 , wherein the device has a coating, and wherein the anti-scarring agent is present in the coating in an amount ranging between about 25% to about 70% by weight. 15391. The method of item 15251 , wherein the device has a coating, and wherein the coating further comprises a polymer.
15392. The method of item 15251 , wherein the device has a first coating having a first composition and a second coating having a second composition.
15393. The method of item 15251 , wherein the device has a first coating having a first composition and a second coating having a second composition, wherein the first composition and the second composition are different.
15394. The method of item 15251 , wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a copolymer.
15395. The method of item 15251 , wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a block copolymer.
15396. The method of item 15251 , wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a random copolymer.
15397. The method of item 15251 , wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a biodegradable polymer.
15398. The method of item 15251 , wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a non-biodegradable polymer. 15399. The method of item 15251 , wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a hydrophilic polymer.
15400. The method of item 15251 , wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a hydrophobic polymer.
15401. The method of item 15251 , wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a polymer having hydrophilic domains.
15402. The method of item 15251 , wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a polymer having hydrophobic domains.
15403. The method of item 15251 , wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a non-conductive polymer.
15404. The method of item 15251 , wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises an elastomer.
15405. The method of item 15251, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a hydrogel.
15406. The method of item 15251, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a silicone polymer. 15407. The method of item 15251 , wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a hydrocarbon polymer.
15408. The method of item 15251 , wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a styrene-derived polymer.
15409. The method of item 15251 , wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a butadiene polymer.
15410. The method of item 15251 , wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a macromer.
15411. The method of item 15251 , wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a poly(ethylene glycol) polymer.
15412. The method of item 15251 wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises an amorphous polymer.
15413. The method of item 15251 , wherein the device comprises a lubricious coating.
15414. The method of item 15251 wherein the anti- scarring agent is located within pores or holes of the device. 15415. The method of item 15251 wherein the anti- scarring agent is located within a channel, lumen, or divet of the device.
15416. The method of item 15251 , wherein the device comprises a second pharmaceutically active agent.
15417. The method of item 15251 wherein the device comprises an anti-inflammatory agent.
15418. The method of item 15251 wherein the device comprises an agent that inhibits infection.
15419. The method of item 15251 wherein the device comprises an agent that inhibits infection, and wherein the agent is an anthracycline.
15420. The method of item 15251 wherein the device comprises an agent that inhibits infection, and wherein the agent is doxorubicin.
15421. The method of item 15251 wherein the device comprises an agent that inhibits infection, and wherein the agent is mitoxantrone.
15422. The method of item 15251 wherein the device comprises an agent that inhibits infection, and wherein the agent is a fluoropyrimidine.
15423. The method of item 15251 wherein the device comprises an agent that inhibits infection, and wherein the agent is 5- fluorouracil (5-FU). 15424. The method of item 15251 wherein the device comprises an agent that inhibits infection, and wherein the agent is a folic acid antagonist.
15425. The method of item 15251 wherein the device comprises an agent that inhibits infection, and wherein the agent is methotrexate.
15426. The method of item 15251 wherein the device comprises an agent that inhibits infection, and wherein the agent is a podophylotoxin.
15427. The method of item 15251 wherein the device comprises an agent that inhibits infection, and wherein the agent is etoposide.
15428. The method of item 15251 wherein the device comprises an agent that inhibits infection, and wherein the agent is a camptothecin.
15429. The method of item 15251 wherein the device comprises an agent that inhibits infection, and wherein the agent is a hydroxyurea.
15430. The method of item 15251 wherein the device comprises an agent that inhibits infection, and wherein the agent is a platinum complex.
15431. The method of item 15251 wherein the device comprises an agent that inhibits infection, and wherein the agent is cisplatin. 15432. The method of item 15251 , further comprising an antithrombotic agent.
15433. The method of item 15251 wherein the device comprises a visualization agent.
15434. The method of item 15251 wherein the device comprises a visualization agent, wherein the visualization agent is a radiopaque material, and wherein the radiopaque material comprises a metal, a halogenated compound, or a barium containing compound.
15435. The method of item 15251 wherein the device comprises a visualization agent, wherein the visualization agent is a radiopaque material, and wherein the radiopaque material comprises barium, tantalum, or technetium.
15436. The method of item 15251 wherein the device comprises a visualization agent, and wherein the visualization agent is a MRI responsive material.
15437. The method of item 15251 wherein the device comprises a visualization agent, and wherein the visualization agent comprises a gadolinium chelate.
15438. The method of item 15251 wherein the device comprises a visualization agent, and wherein the visualization agent comprises iron, magnesium, manganese, copper, or chromium.
15439. The method of item 15251 wherein the device comprises a visualization agent, and wherein the visualization agent comprises an iron oxide compound. 15440. The method of item 15251 wherein the device comprises a visualization agent, and wherein the visualization agent comprises a dye, pigment, or colorant.
15441. The method of item 15251 wherein the device comprises an echogenic material.
15442. The method of item 15251 wherein the device comprises an echogenic material, and wherein the echogenic material is in the form of a coating.
15443. The method of item 15251 wherein the device is sterile.
15444. The method of item 15251 wherein the anti- scarring agent is released into tissue in the vicinity of the device after deployment of the device.
15445. The method of item 15251 wherein the anti- scarring agent is released into tissue in the vicinity of the device after deployment of the device, and wherein the tissue is connective tissue.
15446. The method of item 15251 wherein the anti- scarring agent is released into tissue in the vicinity of the device after deployment of the device, and wherein the tissue is muscle tissue.
15447. The method of item 15251 wherein the anti- scarring agent is released into tissue in the vicinity of the device after deployment of the device, and wherein the tissue is nerve tissue. 15448. The method of item 15251 wherein the anti- scarring agent is released into tissue in the vicinity of the device after deployment of the device, and wherein the tissue is epithelium tissue.
15449. The method of item 15251 wherein the anti- scarring agent is released in effective concentrations from the device over a period ranging from the time of deployment of the device to about 1 year.
15450. The method of item 15251 wherein the anti- scarring agent is released in effective concentrations from the device over a period ranging from about 1 month to 6 months.
15451. The method of item 15251 wherein the anti- scarring agent is released in effective concentrations from the device over a period ranging from about 1 - 90 days.
15452. The method of item 15251 wherein the anti- scarring agent is released in effective concentrations from the device at a constant rate.
15453. The method of item 15251 wherein the anti- scarring agent is released in effective concentrations from the device at an increasing rate.
15454. The method of item 15251 wherein the anti- scarring agent is released in effective concentrations from the device at a decreasing rate.
15455. The method of item 15251 wherein the anti- scarring agent is released in effective concentrations from the composition comprising the anti-scarring agent by diffusion over a period ranging from the time of deployment of the device to about 90 days.
15456. The method of item 15251 wherein the anti- scarring agent is released in effective concentrations from the composition comprising the anti-scarring agent by erosion of the composition over a period ranging from the time of deployment of the device to about 90 days.
15457. The method of item 15251 wherein the device comprises about 0.01 Dg to about 10 Dg of the anti-scarring agent.
15458. The method of item 15251 wherein the device comprises about 10 Dg to about 10 mg of the anti-scarring agent.
15459. The method of item 15251 wherein the device comprises about 10 mg to about 250 mg of the anti-scarring agent.
15460. The method of item 15251 wherein the device comprises about 250 mg to about 1000 mg of the anti-scarring agent.
15461. The method of item 15251 wherein the device comprises about 1000 mg to about 2500 mg of the anti-scarring agent.
15462. The method of item 15251 wherein a surface of the device comprises less than 0.01 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
15463. The method of item 15251 wherein a surface of the device comprises about 0.01 Dg to about 1 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied. 15464. The method of item 15251 wherein a surface of the device comprises about 1 Dg to about 10 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
15465. The method of item 15251 wherein a surface of the device comprises about 10 Dg to about 250 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
15466. The method of item 15251 wherein a surface of the device comprises about 250 Dg to about 1000 Dg of the anti-scarring agent of anti-scarring agent per mm2 of device surface to which the anti- scarring agent is applied.
15467. The method of item 15251 wherein a surface of the device comprises about 1000 Dg to about 2500 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
15468. The method of item 15251 wherein the combining is performed by direct affixing the agent or the composition to the implant.
15469. The method of item 15251 wherein the combining is performed by spraying the agent or the component onto the implant.
15470. The method of item 15251 wherein the combining is performed by electrospraying the agent or the composition onto the implant. 15471. The method of item 15251 wherein the combining is performed by dipping the implant into a solution comprising the agent or the composition.
15472. The method of item 15251 wherein the combining is performed by covalently attaching the agent or the composition to the implant.
15473. The method of item 15251 wherein the combining is performed by non-covalently attaching the agent or the composition to the implant.
15474. The method of item 15251 wherein the combining is performed by coating the implant with a substance that contains the agent or the composition.
15475. The method of item 15251 wherein the combining is performed by coating the implant with a substance that absorbs the agent.
15476. The method of item 15251 wherein the combining is performed by interweaving a thread composed of, or coated with, the agent or the composition.
15477. The method of item 15251 wherein the combining is performed by covering all the implant with a sleeve that contains the agent or the composition.
15478. The method of item 15251 wherein the combining is performed by covering a portion of the implant with a sleeve that contains the agent or the composition. 15479. The method of item 15251 wherein the combining is performed by covering all the implant with a cover that contains the agent or the composition.
15480. The method of item 15251 wherein the combining is performed by covering a portion of the implant with a cover that contains the agent or the composition.
15481. The method of item 15251 wherein the combining is performed by covering all the implant with an electrospun fabric that contains the agent or the composition.
15482. The method of item 15251 wherein the combining is performed by covering a portion of the implant with an electrospun fabric that contains the agent or the composition.
15483. The method of item 15251 wherein the combining is performed by covering all the implant with a mesh that contains the agent or the composition.
15484. The method of item 15251 wherein the combining is performed by covering a portion of the implant with a mesh that contains the agent or the composition.
15485. The method of item 15251 wherein the combining is performed by constructing all the implant with the agent or the composition.
15486. The method of item 15251 wherein the combining is performed by constructing a portion of the implant with the agent or the composition. 15487. The method of item 15251 wherein the combining is performed by impregnating the implant with the agent or the composition.
15488. The method of item 15251 wherein the combining is performed by constructing all of the implant from a degradable polymer that releases the agent.
15489. The method of item 15251 wherein the combining is performed by constructing a portion of the implant from a degradable polymer that releases the agent.
15490. The method of item 15251 wherein the combining is performed by dipping the implant into a solution that comprise the agent and an inert solvent for the implant.
15491. The method of item 15251 wherein the combining is performed by dipping the implant into a solution that comprises the agent and a solvent that will swell the implant.
15492. The method of item 15251 wherein the combining is performed by dipping the implant into a solution that comprises the agent and a solvent that will dissolve the implant.
15493. The method of item 15251 wherein the combining is performed by dipping the implant into a solution that comprises the agent, a polymer and an inert solvent for the implant.
15494. The method of item 15251 wherein the combining is performed by dipping the implant into a solution that comprises the agent, a polymer and a solvent that will swell the implant. 15495. The method of item 15251 wherein the combining is performed by dipping the implant into a solution that comprises the agent, a polymer and a solvent that will dissolve the implant.
15496. The method of item 15251 wherein the combining is performed by spraying the implant into a solution that comprises the agent and an inert solvent for the implant.
15497. The method of item 15251 wherein the combining is performed by spraying the implant into a solution that comprises the agent and a solvent that will swell the implant.
15498. The method of item 15251 wherein the combining is performed by spraying the implant into a solution that comprises the agent and a solvent that will dissolve the implant.
15499. The method of item 15251 wherein the combining is performed by spraying the implant into a solution that comprises the agent, a polymer and an inert solvent for the implant.
15500. The method of item 15251 wherein the combining is performed by spraying the implant into a solution that comprises the agent, a polymer and a solvent that will swell the implant.
15501. The method of item 15251 wherein the combining is performed by spraying the implant into a solution that comprises the agent, a polymer and a solvent that will dissolve the implant.
15502. A method as in any one of items 15251-15501, wherein the device is a peritoneal dialysis catheter adapted for delivering a drug to the peritoneum. 15503. A method of making a medical device comprising: combining a central nervous system shunt or pressure monitor device (e.g., an implant) and an anti-scarring agent or a composition comprising an anti-scarring agent, wherein the agent inhibits scarring between the device and a host into which the device is implanted.
15504. The method of item 15503 wherein the agent is an adensosine A2A receptor antagonist.
15505. The method of item 15503 wherein the agent is an AKT inhibitor.
15506. The method of item 15503 wherein the agent is an alpha 2 integrin antagonist, wherein the alpha 2 integrin antagonist is Pharmaprojects No. 5754 (Merck KgaA).
15507. The method of item 15503 wherein the agent is an alpha 4 integrin antagonist.
15508. The method of item 15503 wherein the agent is an alpha 7 nicotinic receptor agonist.
15509. The method of item 15503 wherein the agent is an angiogenesis inhibitor selected from the group consisting of AG-12,958 (Pfizer), ATN-161 (Attenuon LLC), neovastat, an angiogenesis inhibitor from Jerina AG (Germany), NM-3 (Mercian), VGA-1155 (Taisho), FCE-26644 (Pfizer), FCE-26950 (Pfizer), FPMA (Meiji Daries), FR-111142 (Fujisawa), GGTI-298, GM-1306 (Ligand), GPA-1734 (Novartis), NNC-47-0011 (Novo Nordisk), herbamycin (Nippon Kayaku), lenalidomide (Celegene), IP-10 (NIH)1 ABT-828 (Abbott), KIN-841 (Tokushima University, Japan), SF-1126 (Semafore Pharmaceuticals), laminin technology (NIH), CHIR-258 (Chiron), NVP-AEW541 (Novartis), NVP-AEW541 (Novartis), Vt16907 (Alchemia), OXI-8007 (Oxigene), EG-3306 (Ark Therapeutics), Maspin (AmVa), ABT- 567 (Abbott), PPI-2458 (Praecis Pharmaceuticals), CC-5079, CC-4089 (Celgene), HIF-1 alpha inhibitors (Xenova), S-247 (Pfizer), AP-23573 (Ariad), AZD-9935 (Astra Zeneca), mebendazole (Introgen Therapeutics), MetAP-2 inhibitors (GlaxoSmithKline), AG-615 (Angiogene Pharmaceuticals), Tie-2 antagonists (Hybrigenics), NC-381 , CYC-381 , NC-169, NC-219, NC-383, NC-384, NC-407 (Lorus Therapeutics), ATN-224 (Attenuon), ON-01370 (Onconova), Vitronectin antagonists (Amgen), SDX-103 (Salmedix), Vitronectin antagonists (Shire), CHP (Riemser), TEK (Amgen), Anecortave acetate (Alcon), T46.2 (Matrix Therapeutics), HG-2 (Heptagen), TEM antagonists (Genzyme), Oxi-4500 (Oxigene), ATN-161 (Attenuon), WX-293 (Wilex), M-2025 (Metris Therapeutics), Alphastatin (BioActa), YH-16 (Yantai Rongchang), BIBF-1120 (Boehringer Ingelheim), BAY-57-9352 (Bayer), AS- 1404 (Cancer Research Technology), SC-77964 (Pfizer), glycomimetics (BioTie Therapies), TIE-2 Inhibitors (Ontogen), DIMI, Octamer (Octamer), ABR-215050 (Active Biotech), ABT-518 (Abbott), KDR inhibitors (Abbott), BSF-466895 (Abbott), SCH-221153 (Schering-Plough), DAC:antiangiogenic (ConjuChem), TFPI (EntreMed), AZD-2171 (Astra-Zenaca), CDC-394 (Celgene), LY290293 (EIi Lilly), IDN-5390 (Indena), Kdr Kinase Inhibitors (Merck), CT-113020, CT-116433, CT-116563, CT-31890, CT-32228) (Cell Therapeutics), A-299620 (Abbott), TWEAK Inhibitor (Amgen), VEGF modulators (Johnson and Johnson), Tum-N53, tumstatin (Genzyme), Thios- 1 , Thios-2 (Thios Pharmaceuticals), MV-6401 (Miravant Medical Technologies), Spisulosine (PharmaMar), CEP-7055 (Cephalon), AUV-201 (Auvation), LM-609 (EIi Lilly), SKF-106615 (AnorMED), Oglufanide disodium (Cytran), BW-114 (Pharminox), Calreticulin (NIH), WX-678 (Wilex), SD-7784 (Pfizer), WX-UK1 (Wilex), SH-268 (Schering AG), 2-Me-PGA (Celgene), S- 137 (Pfizer), ZD-6126 (Angiogene Pharmaceuticals), SG-292 (SignalGen), Benefin (Lane Labs), A6, A36 (Angstrom), SB-2723005 (GlaxoSmithKline), SC-7 (Cell Therapeutics), ZEN-014 (AEterna Zentaris), 2-methoxyestradiol (EntreMed), NK-130119 (Nippon Kayaku), CC-10004 (Celgene), AVE- 8062A (Ajinomoto), Tacedinaline (Pfizer), Actinonin (Tokyo Metropolitan Institute of Medical Science), Lenalidomide (Celgene), VGA-1155, BTO-956 (SRI International), ER-68203-00 (Eisai), CT-6685 (UCB), JKC-362 (Phoenix Pharmaceuticals), DMI-3798 (DMI Biosciences, Angiomate (Ipsen), ZD-6474 (AstraZeneca), CEP-5214 (Cephalon), Canstatin (Genzyme), NM-3 (Mercian), Oridigm (MediQuest Therapeutics), Exherin (Adherex), BLS-0597 (Boston Life Sciences), PTC-299 (PTC Therapeutics), NPI-2358 (Nereus Pharmaceuticals), CGP-79787 (Novartis), AEE-788 (Novartis), CKD-732 (Chong Kun Dang), CP-564959 (OSI Pharmaceuticals), CM-101 (CarboMed), CT-2584, CT3501 (Cell Therapeutics), combretastatin and analogues and derivatives thereof (Oxigene), Rebimastat (Bristol-Meyers Squibb), Dextrin 2-sulfate (ML Laboratories), Cilengitide (Merk KGaA), NSC-706704 (Pharminox), KRN- 951 (Kirin Brewery), Ukrain, NSC-631570 (Nowicky Pharma), Tecogalan sodium (Daiichi Pharmaceutical), Tz-93 (Tsumura), TBC-1635 (Encysive- Pharmaceuticals), TAN-1120 (Takeda), Semaxanib (Pfizer), BDI-7800 (Biopharmacopae), SD-186, SD-983 (Bristol-Meyers Squibb), SB-223245 (GlaxoSmithKline), SC-236 (Pfizer), RWJ-590973 (Johnson and Johnson), ILX-1850 (Genzyme), SC-68488, S-836 (Pfizer), CG-55069-11 (CuraGen), Ki-23057 (Kirin Brewery), CCX-700 (Chemoentryx), Pegaptanib octasodium (Giled Sciences), ANGIOCOL (available from Biostratum Inc.), or an analogue or derivative thereof.
15510. The method of item 15503 wherein the agent is an apoptosis antagonist.
15511. The method of item 15503 wherein the agent is an apoptosis activator. 15512. The method of item 15503 wherein the agent is a beta 1 integrin antagonist.
15513. The method of item 15503 wherein the agent is a beta tubulin inhibitor.
15514. The method of item 15503 wherein the agent is a blocker of enzyme production in Hepatitis C.
15515. The method of item 15503 wherein the agent is a Bruton's tyrosine kinase inhibitor.
15516. The method of item 15503 wherein the agent is a calcineurin inhibitor.
15517. The method of item 15503 wherein the agent is a caspase 3 inhibitor.
15518. The method of item 15503 wherein the agent is a CC chemokine receptor antagonist.
15519. The method of item 15503 wherein the agent is a cell cycle inhibitor selected from the group consisting of SNS-595 (Sunesis), synthadotin, KRX-0403, and an analogue or derivative thereof.
15520. The method of item 15503 wherein the agent is a cathepsin B inhibitor.
15521. The method of item 15503 wherein the agent is a cathepsin K inhibitor, wherein the cathepsin K inhibitor is 462795 (GlaxoSmithKline), INPL-022-D6 (Amura Therapeutics), or an analogue or derivative thereof.
15522. The method of item 15503 wherein the agent is a cathepsin L inhibitor.
15523. The method of item 15503 wherein the agent is a CD40 antagonist.
15524. The method of item 15503 wherein the agent is a chemokine receptor agonist.
15525. The method of item 15503 wherein the agent is a chymase inhibitor.
15526. The method of item 15503 wherein the agent is a collagenase antagonist.
15527. The method of item 15503 wherein the agent is a CXCR antagonist.
15528. The method of item 15503 wherein the agent is a cyclin dependent kinase inhibitor selected from the group consisting of a CDK-1 inhibitor, a CDK-2 inhibitor, a CDK- 4 inhibitor, a CDK-6 inhibitor, a CAK1 inhibitor from GPC Biotech and Bristol-Myers Squibb, RGB-286199 (GPC Biotech), an anticancer agent from Astex Technology, a CAK1 inhibitor from GPC Biotech, a CDK inhibitor from Sanofi-Aventis, a CDK1/CDK2 inhibitor from Amgen, a CDK2 inhibitor from SUGEN-2 (Pfizer), a hearing loss therapy agent (Sound Pharmaceuticals), PD-0332991 (Pfizer), RGB-286199 (GPC Biotech), Ro-0505124 (Hoffmann-La Roche), a Ser/Thr kinase inhibitor from Lilly (EIi Lilly), CVT-2584 (CAS No. 199986-75- 9) (CV Therapeutics), and an analogue or derivative thereof.
15529. The method of item 15503 wherein the agent is a cyclooxygenase 1 inhibitor.
15530. The method of item 15503 wherein the agent is a DHFR inhibitor.
15531. The method of item 15503 wherein the agent is a dual integrin inhibitor.
15532. The method of item 15503 wherein the agent is an elastase inhibitor.
15533. The method of item 15503 wherein the agent is an elongation factor-1 alpha inhibitor.
15534. The method of item 15503 wherein the agent is an endothelial growth factor antagonist.
15535. The method of item 15503 wherein the agent is an endothelial growth factor receptor kinase inhibitor selected from the group consisting of sorafenib tosylate (Bayer), AAL-993 (Novartis), ABP-309 (Novartis), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), E- 7080 (Eisai), EG-3306 (Ark Therapeutics), EXEL-2880 (Exelixis), GW- 654652 (GlaxoSmithKline), a KDR inhibitor from LG Life Sciences, CT-6685 and CT-6729 (UCB), KRN-633 and KRN-951 (Kirin Brewery), OSI-930 (OSI Pharmaceuticals), SP-5.2 (Supratek Pharma), SU-11657 (Pfizer), a Tie-2 antagonist (Hybrigenics), a VEGFR-2 kinase inhibitor (Bristol-Myers Squibb), XL-647 (Exelixis), a KDR inhibitor from Abbott Laboratories, sorafenib tosylate, and an analogue or derivative thereof.
15536. The method of item 15503 wherein the agent is an endotoxin antagonist.
15537. The method of item 15503 wherein the agent is an epothilone and tubulin binder.
15538. The method of item 15503 wherein the agent is an estrogen receptor antagonist.
15539. The method of item 15503 wherein the agent is an FGF inhibitor.
15540. The method of item 15503 wherein the agent is a farnexyl transferase inhibitor.
15541. The method of item 15503 wherein the agent is farnesyltransferase inhibitor selected from the group of A-197574 (Abbott), a famesyltransferase inhibitor from Servier, FPTIII (Strathclyde Institute for Drug R), LB-42908 (LG Life Sciences), Pharmaprojects No. 5063 (Genzyme), Pharmaprojects No. 5597 (Ipsen), Yissum Project No. B-1055 (Yissum), and an analogue or derivative thereof.
15542. The method of item 15503 wherein the agent is an FLT-3 kinase inhibitor.
15543. The method of item 15503 wherein the agent is an FGF receptor kinase inhibitor. 15544. The method of item 15503 wherein the agent is a fibrinogen antagonist selected from the group consisting of AUV-201 (Auvation), MG-13926 (Sanofi-Aventis), plasminogen activator (CAS No. 105913-11-9) (from Sanofi-Aventis or UCB), plasminogen activator-2 (tPA- 2) (Sanofi-Aventis), pro-urokinase (CAS No. 82657-92-9) (Sanofi-Aventis), and an analogue or derivative thereof.
15545. The method of item 15503 wherein the agent is a heat shock protein 90 antagonist selected from the group consisting of SRN- 005 (Sirenade), geldanamycin, NSC-33050 (17-aliylaminogeldanamycin; 17- AAG), 17-dimethylaminoethylamino-17-demethoxy-geldanamycin (17- DMAG), rifabutin (rifamycin XIV, 1',4-didehydro-1-deoxy-1 ,4-dihydro-5'-(2- methylpropyl)-1-oxo-), and an analogue or derivative thereof.
15546. The method of item 15503 wherein the agent is a histone deacetylase inhibitor.
15547. The method of item 15503 wherein the agent is an HMGCoA reductase inhibitor selected from the group consisting of an atherosclerosis therapeutic from Lipid Sciences, ATI-16000 (ARYx Therapeutics), KS-01-019 (Kos Pharmaceuticals), Pharmaprojects No. 2197 (Sanofi-Aventi), RP 61969 (Sanofi-Aventis), and an analogue or derivative thereof.
15548. The method of item 15503 wherein the agent is an ICAM inhibitor.
15549. The method of item 15503 wherein the agent is an IL, ICE and IRAK antagonist, wherein the antagonist is a CJ-14877, CP- 424174 (Pfizer), NF-61 (Negma-Lerads), and an analogue or derivative thereof. 15550. The method of item 15503 wherein the agent is an IL-2 inhibitor.
15551. The method of item 15503 wherein the agent is an immunosuppressant selected from the group consisting of teriflunomide (Sanofi Aventis), chlorsulfaquinoxalone (NSC-339004), chlorsulfaquinoxalone sulfate, CS-712 (Sankyo), ismomultin alfa (CAS No. 457913-93-8) (Akzo Nobel), antiallergics from GenPat77, antiinflammatories or AT-005 (Androclus Therapeutics), autoimmune disease therapeutics from EpiVax, BN-007 (Bone), budesonide (CAS No. 51333-22- 3) (MAP Pharmaceuticals), CO-14 (Genzyme), edratide (CAS No. 433922- 67-9) (Teva), EP-314 (Enanta), eprovafen (CAS No. 101335-99-3) (Sanofi- Aventis), H WA- 131 (CAS No. 118788-41-3) (Sanofi-Aventis), immunomodulators from MerLion Pharmaceuticals, immunosuppressives from Alchemia, IPL-12 (Inflazyme), MDL-9563 (CAS No. 27086-86-8) (Sanofi-Aventis), Pharmaprojects No. 2330 (Sanofi-Aventis), Pharmaprojects No. 6426 (Abgenix), PXS-25 (Pharmaxis), rosmarinic acid (CAS No. 20283-92-5) (Sanofi-Aventis), RP 42927 or RP 54745 (CAS No. 135330-08-4) (Sanofi-Aventis), SGN-35 (Seattle Genetics), ST-1959 (Sigma-Tau), type I diabetes therapy from SYNX Pharma, UNIL-88 (Debiopharm), VP-025 (Vasogen), VR-694 (Vectura), PRTX-001 (Protalex), and an analogue or derivative thereof.
15552. The method of item 15503 wherein the agent is an IMPDH (inosine monophosphate).
15553. The method of item 15503 wherein the agent is an integrin antagonist.
15554. The method of item 15503 wherein the agent is an interleukin antagonist. 15555. The method of item 15503 wherein the agent is an inhibitor of type III receptor tyrosine kinase.
15556. The method of item 15503 wherein the agent is an irreversible inhibitor of enzyme methionine aminopeptidase type 2.
15557. The method of item 15503 wherein the agent is an isozyme selective delta protein kinase C inhibitor.
15558. The method of item 15503 wherein the agent a JAK3 enzyme inhibitor.
15559. The method of item 15503 wherein the agent is a
JNK inhibitor.
15560. The method of item 15503 wherein the agent is a kinase inhibitor.
15561. The method of item 15503 wherein the agent is kinesin antagonist.
15562. The method of item 15503 wherein the agent is a kinesin antagonist.
15563. The method of item 15503 wherein the agent is a leukotriene inhibitor and antagonist selected from the group consisting of ambicromil (CAS No. 58805-38-2) (Sanofi-Aventis), amelubant (CAS No. 346735-24-8) (Boehringer Ingelheim), DW-1141 (Dong Wha), ebselen (Daiichi Pharmaceutical), ibudilast (Kyorin), leucotriene inhibitors from Sanofi-Aventis, lymphotoxin-beta receptor (LT-D) from Biogen Idee, Pharmaprojects No. 1535 and 2728 (CAS No. 119340-33-9) (Sanofi- Aventis), R-112 (Rigel), Rev-5367 (CAS No. 92532-05-3) (Sanofi-Aventis), RG-14893 (CAS No. 141835-49-6) (Sanofi-Aventis), RG-5901-A (CAS No. 101910-24-1), 92532-23-5, RP 66153 (CAS No. 142422-79-5), RP 66364 (CAS No.186916-92-5), RP 69698 (CAS No. 141748-00-7) (Sanofi-Aventis), SC-411930 (Pfizer), SC-41930 (CAS No. 120072-59-5) (Pfizer), SC-50605 (CAS No. 138828-39-4) (Pfizer), SC-51146 (CAS No. 141059-52-1), SC- 53228 (CAS No. 153633-01-3) (Pfizer), spaglumic acid (ZY-15106) (CAS No.3106-85-2), 80619-64-3 (Novartis), tipredane (CAS No. 85197-77-9) (Bristol-Myers Squibb), U-75302 (CAS No. 119477-85-9) (Pfizer), and analogue or derivative thereof.
15564. The method of item 15503 wherein the agent is an MAP kinase inhibitor.
15565. The method of item 15503 wherein the agent is a matrix metalloproteinase inhibitor.
15566. The method of item 15503 wherein the agent is an MCP-CCR2 inhibitor.
15567. The method of item 15503 wherein the agent is an mTOR inhibitor.
15568. The method of item 15503 wherein the agent is an mTOR kinase inhibitor.
15569. The method of item 15503 wherein the agent is a microtubule inhibitor selected from the group consisting of antibody- maytansinoid conjugates from Biogen Idee, colchicines (MantiCore Pharmaceuticals), anticancer immunoconjugates from Johnson & Johnson, DIME from Octamer, gni-1f (GNI), huC242-DM4, huMy9-6-DM1 (ImmunoGen), IDN-5404 (Indena), IMO-098, IMOderm (Imotep), mebendazole (Introgen Therapeutics), microtubule poisons from Cambridge Enterprise, paclitaxel such as LOTAX from Aphios (CAS No. 33069-62-4), Genexol-PM from Samyang, Pharmaprojects No. 6383 (Tapestry Pharmaceuticals), RPR-112378 (Sanofi-Aventis), SGN-75 (Seattle Genetics), SPL-7435 (Starpharma), SSR-250411 (Sanofi-Aventis), trastuzumab-DM1 (Genentech), vinorelbine, and an analogue or derivative thereof.
15570. The method of item 15503 wherein the agent is an MIF inhibitor.
15571. The method of item 15503 wherein the agent is an MMP inhibitor.
15572. The method of item 15503 wherein the agent is a neurokinin (NK) antagonist selected from the group consisting of anthrotainin (CAS No. 148084-40-6) (Sanofi-Aventis), an IBS thereapeutic from ArQuIe, MDL-105212A (CAS No. 167261-60-1) (Ssanofi-Aventis), Pharmaprojects No. 2744, 3258 (CAS No. 139167-47-8) 4006, 4201, or 5986 (Sanofi-Aventis), RP 67580 (CAS No. 135911-02-3), SR-144190 (CAS No. 201152-86-5), SSR-240600, SSR-241586 (Sanofi-Aventis), TKA-457 (Novartis), vestipitant mesylate (CAS No. 334476-64-1) (GlaxoSmithKline), Win-64821 (Sanofi-Aventis), PRX-96026 (Predix Pharmaceuticals), and an analogue or derivative thereof.
15573. The method of item 15503 wherein the agent is an NF kappa B inhibitor selected from the group consisting of AVE-0545 or AVE-0547 (Sanofi-Aventis), bortezomib (CAS No. 179324-69-7) (Millennium Pharmaceuticals), dexanabinol (CAS No. 112924-45-5) (Pharmos), dexlipotam (Viatris), Pharmaprojects No. 6283 (INDRA) (OXiGENE), IPL- 576092 (CAS No. 137571-30-3) (Inflazyme), NFKB decoy (Corgentech), NFKB decoy oligo (AnGes MG)1 NFKB's from Ariad, osteoporosis treatments or S5 (F005) from Fulcrum Pharmaceuticals, P61 (Phytopharm), R-flurbiprofen (CAS No. 5104-49-4) (Encore Pharmaceuticals), and an analogue or derivative thereof.
15574. The method of item 15503 wherein the agent is a nitric oxide agonist.
15575. The method of item 15503 wherein the agent is an ornithine decarboxylase inhibitor.
15576. The method of item 15503 wherein the agent is a p38 MAP kinase inhibitor selected from the group consisting of AZD-6703 (AstraZeneca), JX-401 (Jexys Pharmaceuticals), BMS-2 (Bristol-Myers Squibb), a p38 MAP kinase inhibitor from Novartis, a p38-alpha MAP kinase inhibitor from Amphora, Pharmaprojects No. 5704 (Pharmacopeia), RPR- 200765A (Sanofi-Aventis), SD-282 (Johnson & Johnson), TAK-715 (Takeda), and an analogue or derivative thereof.
15577. The method of item 15503 wherein the agent is a palmitoyl-protein thioesterase inhibitor.
15578. The method of item 15503 wherein the agent is a PDGF receptor kinase inhibitor selected from the group consisting of AAL- 993, AMN-107, or ABP-309 (Novartis), AMG-706 (Amgen), BAY-57-9352 (Bayer), CDP-860 (UCB), E-7080 (Eisai), imatinib (CAS No. 152459-95-5) (Novartis), OSI-930 (OSI Pharmaceuticals), RPR-127963E (Sanofi-Aventis), RWJ-540973 (Johnson & Johnson), sorafenib tosylate (Bayer), SU-11657 (Pfizer), tandutinib (CAS No. 387867-13-2) (Millennium Pharmaceuticals), vatalanib (Novartis), ZK-CDK (Schering AG)1 and an analogue or derivative thereof.
15579. The method of item 15503 wherein the agent is (-)-halofenate (Metabolex), AMG-131 (Amgen), antidiabetics from Japan Tobacco, AZD-4619, AZD-8450, AZD-8677 (AstraZeneca), DRF-10945, balaglitazone (Dr Reddy's), CS-00088, CS-00098 (Chipscreen Biosciences), E-3030 (Eisai), etalocib (CAS No. 161172-51-6) (EIi Lilly), GSK-641597 (Ligand), GSK-677954 (GlaxoSmithKline), GW-409544 (Ligand), GW- 590735 (GlaxoSmithKline), K-111 (Hoffmann-La Roche), LY-518674 (EIi Lilly), LY-674 (Ligand), LY-929 (Ligand), MC-3001 , MC-3002 (MaxoCore Pharmaceuticals), metformin HCI + pioglitazone (CAS No. 1115-70-4 and 112529-15-4), ACTOPLUS MET from Andrx), muraglitazar (CAS No. 331741-94-7) (Bristol-Myers Squibb), naveglitazar (Ligand), oleoylethanolamide (Kadmus Pharmaceuticals), ONO-5129, pioglitazone hydrochloride (CAS No. 111025-46-8 and 112529-15-4) (Takeda), PLX-204 (Plexxikon), PPAR agonists from Genfit, PPAR delta agonists from EIi Lilly, PPAR-alpha agonists from CrystaiGenomics, PPAR-gamma modulators and PPAR-β modulators from CareX, rosiglitazone maleate (CAS No. 122324- 73-4 or 155141-29-0) (GlaxoSmithKline), rosiglitazone maleate/glimepir (CAS No. 155141-29-0 and 93479-97-1), AVANDARYL, rosiglitazone maleate/metformin extend (CAS No. 155141-29-0 and 657-24-9), AVANDAMET, rosiglitazone maleate+metformin, AVANDAMET (GlaxoSmithKline), tesaglitazar (AstraZeneca), and an analogue or derivative thereof.
15580. The method of item 15503 wherein the agent is a phosphatase inhibitor.
15581. The method of item 15503 wherein the agent is a phosphodiesterase (PDE) inhibitor selected from the group consisting of avanafil (Tanabe Seiyaku), dasantafil (CAS No. 569351-91-3) (Schering- Plough), A-906119 (CAS No. 134072-58-5), DL-850 (Sanofi-Aventis), GRC- 3015, GRC-3566, GRC-3886 (Glenmark), HWA-153 (CAS No. 56395-66-5) (Sanofi-Aventis), hydroxypumafentrine (Altana), IBFB-130011 , IBFB-14-016, IBFB-140301 , IBFB-150007, IBFB-211913 (IBFB Pharma), L-826141 (Merck & Co), medorinone (CAS No. 88296-61-1) (Sanofi-Aventis), MEM- 1917 (Memory Pharmaceuticals), ND-1251 (Neuro3d), PDE inhibitors from ICOS, PDE IV inhibitors from Memory Pharmaceuticals and CrystalGenomics, Pharmaprojects No. 2742 and 6141 (Sanofi-Aventis), QAD-171 (Novartis), RHC-2963 (CAS No. 76993-12-9 and 76993-14-1), RPR-117658, RPR-122818 derivatives, SR-24870 , and RPR-132294 (Sanofi-Aventis), SK-350 (ln2Gen), stroke therapy agents from deCODE Genetics, TAS-203 (Taiho), tofimilast (CAS No. 185954-27-2) (Pfizer), UK- 371800 (Pfizer), WIN-65579 (CAS No. 158020-82-7) (Sanofi-Aventis), IBFB- 130020 (IBFB Pharma), OPC-6535 (CAS No. 145739-56-6) (Otsuka), a phosphodiesterase III inhibitor, enoximone, a phosphodiesterase IV inhibitor, fosfosal, Atopik (Barrier Therapeutics), triflusal, a phosphodiesterase V inhibitor, and an analogue or derivative thereof.
15582. The method of item 15503 wherein the agent is a PKC inhibitor.
15583. The method of item 15503 wherein the agent is a platelet activating factor antagonist.
15584. The method of item 15503 wherein the agent is a platelet-derived growth factor receptor kinase inhibitor.
15585. The method of item 15503 wherein the agent is a prolyl hydroxylase inhibitor. 15586. The method of item 15503 wherein the agent is a polymorphonuclear neutrophil inhibitor.
15587. The method of item 15503 wherein the agent is a protein kinase B inhibitor.
15588. The method of item 15503 wherein the agent is a protein kinase C stimulant.
15589. The method of item 15503 wherein the agent is a purine nucleoside analogue.
15590. The method of item 15503 wherein the agent is a purinoreceptor P2X antagonist.
15591. The method of item 15503 wherein the agent is a Raf kinase inhibitor.
15592. The method of item 15503 wherein the agent is a reversible inhibitor of ErbB1 and ErbB2.
15593. The method of item 15503 wherein the agent is a ribonucleoside triphosphate reductase inhibitor.
15594. The method of item 15503 wherein the agent is an SDF-1 antagonist.
15595. The method of item 15503 wherein the agent is a sheddase inhibitor. 15596. The method of item 15503 wherein the agent is an SRC inhibitor.
15597. The method of item 15503 wherein the agent is a stromelysin inhibitor.
15598. The method of item 15503 wherein the agent is an Syk kinase inhibitor.
15599. The method of item 15503 wherein the agent is a telomerase inhibitor.
15600. The method of item 15503 wherein the agent is a TGF beta inhibitor selected from the group consisting of pirfenidone (CAS No. 53179-13-8) (MARNAC), tranilast (CAS No. 53902-12-8) (Kissei), IN- 1130 (ln2Gen), mannose-6-phosphate (BTG), TGF-β antagonists from Inflazyme (Pharmaprojects No. 6075), TGF-β antagonists from Sydney, non-industrial source), TGF-β I receptor kinase inhibitors from EIi Lilly, TGF- β receptor inhibitors from Johnson & Johnson, and an analogue or derivative thereof.
15601. The method of item 15503 wherein the agent is a TNFα antagonist or TACE inhibitor selected from the group consisting of adalimumab (CAS No. 331731-18-1) (Cambridge Antibody Technology), AGIX-4207 (AtheroGenics), AGT-1 (Advanced Biotherapy), an antiinflammatory from Borean Pharma, Cellzome, or Paradigm Therapeutics, anti-inflammatory vaccine (TNF-alpha kinoid) from Neovacs, humanized anti-TNF antibody or an anti-TNF MAb (CB0006) Celltech (UCB), apratastat (CAS No. 287445-51-0) (Wyeth), BMS-561392 (Bristol-Myers Squibb), BN- 006 (Bone), certolizumab pegol (CAS No. 428863-50-7 or CH-138 (UCB), cilomilast (CAS No. 153259-65-5) (GlaxoSmithKline), CR-1 (Nuada Pharmaceuticals), CRx-119 (CombinatoRx), D-5410 (UCB), dacopafant (CAS No. 125372-33-0) (Sanofi-Aventis), dersalazine (CAS No. 188913-57- 7/188913-58-8) (Uriach), etanercept (CAS No. 185243-69-0) (Amgen), ethyl pyruvate (Critical (Critical Therapeutics), golimumab (CAS No. 476181-74-5) (Johnson & Johnson), hormono-immunotherapy from Ipsen, CDP571 (e.g., Humicade from UCB), IC-485 (ICOS)1 infliximab (CAS No. 170277-31-3) (Johnson & Johnson), IP-751 (Manhattan Pharmaceuticals), ISIS-104838 (CAS No. 250755-32-9) (ISIS Pharmaceuticals), lenalidomide (CAS No. 191732-72-6) (Celgene), lentinan (CAS No. 37339-90-5) (Ajinomoto), MDL- 201112 (CAS No. 142130-73-2) (Sanofi-Aventis), medroxyprogesterone (CAS No. 520-85-4) (InKine Pharmaceutical), N-acetylcysteine (CAS No. 616-91-1) (Zambon), NBE-P2 (DIREVO Biotech), nerelimomab (CAS No. 162774-06-3) (Chiron), OM-294DP (OM PHARMA), onercept (CAS No. 199685-57-9) (Yeda), PASSTNF-alpha (Verigen), pentoxifylline or oxypentifylline (Sanofi-Aventis), Pharmaprojects No. 4091 , 4241 , 4295, or 4488 (Sanofi-Aventis), Pharmaprojects No. 5480 (Amgen), Pharmaprojects No. 6749 (Cengent), pirfenidone (CAS No. 53179-13-8) (MARNAC), RPR- 132294 (Sanofi-Aventis), S5 (F002) (Fulcrum Pharmaceuticals), simvastatin (CAS No. 79902-63-9) (Merck & Co), STA-6292 (Synta Pharmaceuticals), tacrolimus (CAS No. 104987-11-3) (Fujisawa LifeCycle Pharma), talactoferrin alfa (CAS No. 308240-58-6) (Agennix), thalidomide (CAS No. 50-35-1) (Celgene), TNF antagonists form ProStrakan, and Synergen, TNF inhibitors (Amgen), TNF-alpha antagonists from Dynavax Technologies and Jerina AG (Germany), TNF-alpha inhibitors from IBFB Pharma and Xencor (Xencor), torbafylline (CAS No. 105102-21-4) (Sanofi-Aventis), UR-1505 (Uriach), VT-346 (Viron Therapeutics), YSIL6 (Y's Therapeutics), YSTH2 (Y's Therapeutics), NPI-1302a-3 (Nereus Pharmaceuticals, a TNF antagonist from Jerina AG (Germany), dersalazine, and an analogue or derivative thereof. 15602. The method of item 15503 wherein the agent is a Toll receptor inhibitor.
15603. The method of item 15503 wherein the agent is a tubulin antagonist.
15604. The method of item 15503 wherein the agent is a tyrosine kinase inhibitor selected from the group consisting of SU-011248, SUTENT from Pfizer Inc. (New York, NY), BMS-354825, PN-355 (Paracelsian Pharmaceuticals), AGN-199659 (Allergan), AAL-993 or ABP- 309 (Novartis), adaphostin (NIH), AEE-788 (Novartis), AG-013736 (OSI Pharmaceuticals), AG-13736 (Pfizer), ALT-110 (Alteris Therapeutics), AMG- 706 (Amgen), anticancer MAbs from Xencor, anti-EGFrvlll MAbs from Abgenix, anti-HER2 MAb from Abiogen, AZD-2171 or AZD-9935 (AstraZeneca), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), CEP-5214 (Cephalon), CEP-7055 (Cephalon), cetuximab (ImClone Systems), CHIR-200131 and CHIR-258 (Chiron), CP-547632 (OSI Pharmaceuticals), CP-724714 (Pfizer), CT-301 (Creabilis Therapeutics), D- 69491 (Baxter International), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EGFR/ErbB2 inhibitors from Array BioPharma, erlotinib (CAS No. 183319- 69-9) (OSI Pharmaceuticals), EXEL-2880 (Exelixis), FK-778 (Sanofi- Aventis) , gefitinib (CAS No. 184475-35-2) (AstraZeneca), GW-2286 or GW- 654652 (GlaxoSmithKline), her2/neu antigen from AlphaVax, HER-2/neu inhibitor from Generex, Herzyme (Medipad) (Sirna Therapeutics), HKI-272 (Wyeth), HuMax-EGFr (Genmab), idronoxil (CAS No. 81267-65-4) (Novogen), IGF-1 inhibitors from Ontogen, IMC-11 F8 (ImClone Systems), kahalalide F (CAS No. 149204-42-2) (PharmaMar), KDR inhibitor from LG Life Sciences, KDR inhibitors from Abbott Laboratories, KDR kinase inhibitors (UCB), Kdr kinase inhibitors from Merck & Co, KRN-633 and KRN- 951 (Kirin Brewery), KSB-102 (Xenova), lapatinib ditosylate (CAS No. 388082-78-8) (GlaxoSmithKline), matuzumab (Merck KGaA), MDX-214 (Medarex), ME-103 (Pharmexa), MED-A300 (Gerolymatos), MNAC-13 (Lay Line Genomics), nimotuzumab (Center of Molecular Immunology), NSC- 330507 or NSC-707545 (NIH), NV-50 (Novogen), OSI-930 (OSI Pharmaceuticals), panitumumab (Abgenix), pelitinib (CAS No. 287933-82-7) (Wyeth), pertuzumab (CAS No. 380610-27-5) (Genentech), Pharmaprojects No. 3985 (Sanofi-Aventis), prostate cancer therapeutics from Sequenom (SQPC35, SQPC36, SQPC90), removab and remoxab (Trion Pharma), RG- 13022 (CAS No. 136831-48-6), RG-13291 (CAS No. 138989-50-1), or RG- 14620 (CAS No. 136831-49-7) (Sanofi-Aventis), RM-6427 (Romark), RNAi breast cancer therapy from Benitec, RP 53801 (CAS No. 125882-88-4) (Sanofi-Aventis), sorafenib tosylate (Bayer), SU-11657 (Pfizer), Tie-2 antagonists from Semaia (Hybrigenics), Tie-2 inhibitors from Ontogen, trastuzumab (CAS No. 180288-69-1) (Genentech), tyrosine kinase inhibitors from Sanofi-Aventis, U3-1287, U3-1565, U3-1784, U3-1800 (U3 Pharma), vatalanib (Novartis), VEGFR-2 kinase inhibitor from Bristol-Myers Squibb, XL-647 (Exelixis), ZD-6474 (AstraZeneca), ZK-CDK (Schering AG), an EGFR tyrosine kinase inhibitor, EKB-569 (Wyeth), and an analogue or derivative thereof.
15605. The method of item 15503 wherein the agent is a VEGF inhibitor.
15606. The method of item 15503 wherein the agent is a vitamin D receptor agonist.
15607. The method of item 15503 wherein the agent is ZD-6474 (an angiogenesis inhibitor).
15608. The method of item 15503 wherein the agent is AP-23573 (an mTOR inhibitor). 15609. The method of item 15503 wherein the agent is synthadotin (a tubulin antagonist).
15610. The method of item 15503 wherein the agent is S-0885 (a collagenase inhibitor).
15611. The method of item 15503 wherein the agent is aplidine (an elongation factor-1 alpha inhibitor).
15612. The method of item 15503 wherein the agent is ixabepilone (an epithilone).
15613. The method of item 15503 wherein the agent is IDN-5390 (an angiogenesis inhibitor).
15614. The method of item 15503 wherein the agent is SB-2723005 (an angiogenesis inhibitor).
15615. The method of item 15503 wherein the agent is ABT-518 (an angiogenesis inhibitor).
15616. The method of item 15503 wherein the agent is combretastatin (an angiogenesis inhibitor).
15617. The method of item 15503 wherein the agent is anecortave acetate (an angiogenesis inhibitor).
15618. The method of item 15503 wherein the agent is SB-715992 (a kinesin antagonist). 15619. The method of item 15503 wherein the agent is temsirolimus (an mTOR inhibitor).
15620. The method of item 15503 wherein the agent is adalimumab (a TNFα antagonist).
15621. The method of item 15503, wherein the composition comprises a polymer.
15622. The method of item 15503, wherein the composition comprises a polymeric carrier.
15623. The method of item 15503 wherein the anti- scarring agent inhibits adhesion between the device and a host into which the device is implanted.
15624. The method of item 15503 wherein the device delivers the anti-scarring agent locally to tissue proximate to the device.
15625. The method of item 15503 wherein the device has a coating that comprises the anti-scarring agent.
15626. The method of item 15503, wherein the device has a coating that comprises the agent and is disposed on a surface of the implant.
15627. The method of item 15503, wherein the device has a coating that comprises the agent and directly contacts the implant.
15628. The method of item 15503, wherein the device has a coating that comprises the agent and indirectly contacts the implant. 15629. The method of item 15503, wherein the device has a coating that comprises the agent and partially covers the implant.
15630. The method of item 15503, wherein the device has a coating that comprises the agent and completely covers the implant.
15631. The method of item 15503, wherein the device has a uniform coating.
15632. The method of item 15503, wherein the device has a non-uniform coating.
15633. The method of item 15503, wherein the device has a discontinuous coating.
15634. The method of item 15503, wherein the device has a patterned coating.
15635. The method of item 15503, wherein the device has a coating with a thickness of 100 Dm or less.
15636. The method of item 15503, wherein the device has a coating with a thickness of 10 Dm or less.
15637. The method of item 15503, wherein the device has a coating, and the coating adheres to the surface of the implant upon deployment of the implant.
15638. The method of item 15503, wherein the device has a coating, and wherein the coating is stable at room temperature for a period of 1 year. 15639. The method of item 15503, wherein the device has a coating, and wherein the anti-scarring agent is present in the coating in an amount ranging between about 0.0001% to about 1% by weight.
15640. The method of item 15503, wherein the device has a coating, and wherein the anti-scarring agent is present in the coating in an amount ranging between about 1 % to about 10% by weight.
15641. The method of item 15503, wherein the device has a coating, and wherein the anti-scarring agent is present in the coating in an amount ranging between about 10% to about 25% by weight.
15642. The method of item 15503, wherein the device has a coating, and wherein the anti-scarring agent is present in the coating in an amount ranging between about 25% to about 70% by weight.
15643. The method of item 15503, wherein the device has a coating, and wherein the coating further comprises a polymer.
15644. The method of item 15503, wherein the device has a first coating having a first composition and a second coating having a second composition.
15645. The method of item 15503, wherein the device has a first coating having a first composition and a second coating having a second composition, wherein the first composition and the second composition are different.
15646. The method of item 15503, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a copolymer. 15647. The method of item 15503, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a block copolymer.
15648. The method of item 15503, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a random copolymer.
15649. The method of item 15503, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a biodegradable polymer.
15650. The method of item 15503, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a non-biodegradable polymer.
15651. The method of item 15503, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a hydrophilic polymer.
15652. The method of item 15503, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a hydrophobic polymer.
15653. The method of item 15503, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a polymer having hydrophilic domains.
15654. The method of item 15503, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a polymer having hydrophobic domains. 15655. The method of item 15503, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a non-conductive polymer.
15656. The method of item 15503, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises an elastomer.
15657. The method of item 15503, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a hydrogel.
15658. The method of item 15503, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a silicone polymer.
15659. The method of item 15503, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a hydrocarbon polymer.
15660. The method of item 15503, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a styrene-derived polymer.
15661. The method of item 15503, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a butadiene polymer.
15662. The method of item 15503, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a macromer. 15663. The method of item 15503, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a poly(ethylene glycol) polymer.
15664. The method of item 15503 wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises an amorphous polymer.
15665. The method of item 15503, wherein the device comprises a lubricious coating.
15666. The method of item 15503 wherein the anti- scarring agent is located within pores or holes of the device.
15667. The method of item 15503 wherein the anti- scarring agent is located within a channel, lumen, or divet of the device,
15668. The method of item 15503, wherein the device comprises a second pharmaceutically active agent.
15669. The method of item 15503 wherein the device comprises an anti-inflammatory agent.
15670. The method of item 15503 wherein the device comprises an agent that inhibits infection.
15671. The method of item 15503 wherein the device comprises an agent that inhibits infection, and wherein the agent is an anthracycline. 15672. The method of item 15503 wherein the device comprises an agent that inhibits infection, and wherein the agent is doxorubicin.
15673. The method of item 15503 wherein the device comprises an agent that inhibits infection, and wherein the agent is mitoxantrone.
15674. The method of item 15503 wherein the device comprises an agent that inhibits infection, and wherein the agent is a fluoropyrimidine.
15675. The method of item 15503 wherein the device comprises an agent that inhibits infection, and wherein the agent is 5- fluorouracil (5-FU).
15676. The method of item 15503 wherein the device comprises an agent that inhibits infection, and wherein the agent is a folic acid antagonist.
15677. The method of item 15503 wherein the device comprises an agent that inhibits infection, and wherein the agent is methotrexate.
15678. The method of item 15503 wherein the device comprises an agent that inhibits infection, and wherein the agent is a podophyiotoxin.
15679. The method of item 15503 wherein the device comprises an agent that inhibits infection, and wherein the agent is etoposide. 15680. The method of item 15503 wherein the device comprises an agent that inhibits infection, and wherein the agent is a camptothecin.
15681. The method of item 15503 wherein the device comprises an agent that inhibits infection, and wherein the agent is a hydroxyurea.
15682. The method of item 15503 wherein the device comprises an agent that inhibits infection, and wherein the agent is a platinum complex.
15683. The method of item 15503 wherein the device comprises an agent that inhibits infection, and wherein the agent is cisplatin.
15684. The method of item 15503, further comprising an anti-thrombotic agent.
15685. The method of item 15503 wherein the device comprises a visualization agent.
15686. The method of item 15503 wherein the device comprises a visualization agent, wherein the visualization agent is a radiopaque material, and wherein the radiopaque material comprises a metal, a halogenated compound, or a barium containing compound.
15687. The method of item 15503 wherein the device comprises a visualization agent, wherein the visualization agent is a radiopaque material, and wherein the radiopaque material comprises barium, tantalum, or technetium. 15688. The method of item 15503 wherein the device comprises a visualization agent, and wherein the visualization agent is a MRI responsive material.
15689. The method of item 15503 wherein the device comprises a visualization agent, and wherein the visualization agent comprises a gadolinium chelate.
15690. The method of item 15503 wherein the device comprises a visualization agent, and wherein the visualization agent comprises iron, magnesium, manganese, copper, or chromium.
15691. The method of item 15503 wherein the device comprises a visualization agent, and wherein the visualization agent comprises an iron oxide compound.
15692. The method of item 15503 wherein the device comprises a visualization agent, and wherein the visualization agent comprises a dye, pigment, or colorant.
15693. The method of item 15503 wherein the device comprises an echogenic material.
15694. The method of item 15503 wherein the device comprises an echogenic material, and wherein the echogenic material is in the form of a coating.
15695. The method of item 15503 wherein the device is sterile. 15696. The method of item 15503 wherein the anti- scarring agent is released into tissue in the vicinity of the device after deployment of the device.
15697. The method of item 15503 wherein the anti- scarring agent is released into tissue in the vicinity of the device after deployment of the device, and wherein the tissue is connective tissue.
15698. The method of item 15503 wherein the anti- scarring agent is released into tissue in the vicinity of the device after deployment of the device, and wherein the tissue is muscle tissue.
15699. The method of item 15503 wherein the anti- scarring agent is released into tissue in the vicinity of the device after deployment of the device, and wherein the tissue is nerve tissue.
15700. The method of item 15503 wherein the anti- scarring agent is released into tissue in the vicinity of the device after deployment of the device, and wherein the tissue is epithelium tissue.
15701. The method of item 15503 wherein the anti- scarring agent is released in effective concentrations from the device over a period ranging from the time of deployment of the device to about 1 year.
15702. The method of item 15503 wherein the anti- scarring agent is released in effective concentrations from the device over a period ranging from about 1 month to 6 months.
15703. The method of item 15503 wherein the anti- scarring agent is released in effective concentrations from the device over a period ranging from about 1 - 90 days. 15704. The method of item 15503 wherein the anti- scarring agent is released in effective concentrations from the device at a constant rate.
15705. The method of item 15503 wherein the anti- scarring agent is released in effective concentrations from the device at an increasing rate.
15706. The method of item 15503 wherein the anti- scarring agent is released in effective concentrations from the device at a decreasing rate.
15707. The method of item 15503 wherein the anti- scarring agent is released in effective concentrations from the composition comprising the anti-scarring agent by diffusion over a period ranging from the time of deployment of the device to about 90 days.
15708. The method of item 15503 wherein the anti- scarring agent is released in effective concentrations from the composition comprising the anti-scarring agent by erosion of the composition over a period ranging from the time of deployment of the device to about 90 days.
15709. The method of item 15503 wherein the device comprises about 0.01 Dg to about 10 Dg of the anti-scarring agent.
15710. The method of item 15503 wherein the device comprises about 10 Dg to about 10 mg of the anti-scarring agent.
15711. The method of item 15503 wherein the device comprises about 10 mg to about 250 mg of the anti-scarring agent. 15712. The method of item 15503 wherein the device comprises about 250 mg to about 1000 mg of the anti-scarring agent.
15713. The method of item 15503 wherein the device comprises about 1000 mg to about 2500 mg of the anti-scarring agent.
15714. The method of item 15503 wherein a surface of the device comprises less than 0.01 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
15715. The method of item 15503 wherein a surface of the device comprises about 0.01 Dg to about 1 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
15716. The method of item 15503 wherein a surface of the device comprises about 1 Dg to about 10 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
15717. The method of item 15503 wherein a surface of the device comprises about 10 Dg to about 250 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
15718. The method of item 15503 wherein a surface of the device comprises about 250 Dg to about 1000 Dg of the anti-scarring agent of anti-scarring agent per mm2 of device surface to which the anti- scarring agent is applied.
15719. The method of item 15503 wherein a surface of the device comprises about 1000 Dg to about 2500 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied. 15720. The method of item 15503 wherein the combining is performed by direct affixing the agent or the composition to the implant.
15721. The method of item 15503 wherein the combining is performed by spraying the agent or the component onto the implant.
15722. The method of item 15503 wherein the combining is performed by electrospraying the agent or the composition onto the implant.
15723. The method of item 15503 wherein the combining is performed by dipping the implant into a solution comprising the agent or the composition.
15724. The method of item 15503 wherein the combining is performed by covalentiy attaching the agent or the composition to the implant.
15725. The method of item 15503 wherein the combining is performed by non-covalently attaching the agent or the composition to the implant.
15726. The method of item 15503 wherein the combining is performed by coating the implant with a substance that contains the agent or the composition.
15727. The method of item 15503 wherein the combining is performed by coating the implant with a substance that absorbs the agent. 15728. The method of item 15503 wherein the combining is performed by interweaving a thread composed of, or coated with, the agent or the composition.
15729. The method of item 15503 wherein the combining is performed by covering all the implant with a sleeve that contains the agent or the composition.
15730. The method of item 15503 wherein the combining is performed by covering a portion of the implant with a sleeve that contains the agent or the composition.
15731. The method of item 15503 wherein the combining is performed by covering all the implant with a cover that contains the agent or the composition.
15732. The method of item 15503 wherein the combining is performed by covering a portion of the implant with a cover that contains the agent or the composition.
15733. The method of item 15503 wherein the combining is performed by covering all the implant with an electrospun fabric that contains the agent or the composition.
15734. The method of item 15503 wherein the combining is performed by covering a portion of the implant with an electrospun fabric that contains the agent or the composition.
15735. The method of item 15503 wherein the combining is performed by covering all the implant with a mesh that contains the agent or the composition. 15736. The method of item 15503 wherein the combining is performed by covering a portion of the implant with a mesh that contains the agent or the composition.
15737. The method of item 15503 wherein the combining is performed by constructing all the implant with the agent or the composition.
15738. The method of item 15503 wherein the combining is performed by constructing a portion of the implant with the agent or the composition.
15739. The method of item 15503 wherein the combining is performed by impregnating the implant with the agent or the composition.
15740. The method of item 15503 wherein the combining is performed by constructing all of the implant from a degradable polymer that releases the agent.
15741. The method of item 15503 wherein the combining is performed by constructing a portion of the implant from a degradable polymer that releases the agent.
15742. The method of item 15503 wherein the combining is performed by dipping the implant into a solution that comprise the agent and an inert solvent for the implant.
15743. The method of item 15503 wherein the combining is performed by dipping the implant into a solution that comprises the agent and a solvent that will swell the implant. 15744. The method of item 15503 wherein the combining is performed by dipping the implant into a solution that comprises the agent and a solvent that will dissolve the implant.
15745. The method of item 15503 wherein the combining is performed by dipping the implant into a solution that comprises the agent, a polymer and an inert solvent for the implant.
15746. The method of item 15503 wherein the combining is performed by dipping the implant into a solution that comprises the agent, a polymer and a solvent that will swell the implant.
15747. The method of item 15503 wherein the combining is performed by dipping the implant into a solution that comprises the agent, a polymer and a solvent that will dissolve the implant.
15748. The method of item 15503 wherein the combining is performed by spraying the implant into a solution that comprises the agent and an inert solvent for the implant.
15749. The method of item 15503 wherein the combining is performed by spraying the implant into a solution that comprises the agent and a solvent that will swell the implant.
15750. The method of item 15503 wherein the combining is performed by spraying the implant into a solution that comprises the agent and a solvent that will dissolve the implant.
15751. The method of item 15503 wherein the combining is performed by spraying the implant into a solution that comprises the agent, a polymer and an inert solvent for the implant. 15752. The method of item 15503 wherein the combining is performed by spraying the implant into a solution that comprises the agent, a polymer and a solvent that will swell the implant.
15753. The method of item 15503 wherein the combining is performed by spraying the implant into a solution that comprises the agent, a polymer and a solvent that will dissolve the implant.
15754. A method as in any one of items 15503-15753, wherein the device is a ventriculopleural shunt.
15755. A method as in any one of items 15503-15753, wherein the device is a jugular vein shunt.
15756. A method as in any one of items 15503-15753, wherein the device is a vena cava shunt.
15757. A method as in any one of items 15503-15753, wherein the device is a ventriculoperitoneal shunt.
15758. A method as in any one of items 15503-15753, wherein the device is a gallbladder shunt.
15759. A method as in any one of items 15503-15753, wherein the device is a peritoneum shunt.
15760. A method as in any one of items 15503-15753, wherein the device is an external ventricular drainage device.
15761. A method as in any one of items 15503-15753, wherein the device is an intracranial pressure monitoring device. 15762. A method as in any one of items 15503-15753, wherein the device is an intracranial pressure monitoring device.
15763. A method as in any one of items 15503-15753, wherein the device is a dural patch.
15764. A method as in any one of items 15503-15753, wherein the device is an implant to prevent epidural fibrosis post- laminectomy.
15765. A method as in any one of items 15503-15753, wherein the device is a device for continuous subarachnoid infusion.
15766. A method as in any one of items 15503-15753, wherein the device is a drainage shunt useful for draining fluids in the brain.
15767. A method as in any one of items 15503-15753, wherein the device is a pressure monitoring device.
15768. A method of making a medical device comprising: combining an inferior vena cava filter (e.g., an implant) and an anti-scarring agent or a composition comprising an anti-scarring agent, wherein the agent inhibits scarring between the device and a host into which the device is implanted.
15769. The method of item 15768 wherein the agent is an adensosine A2A receptor antagonist.
15770. The method of item 15768 wherein the agent is an AKT inhibitor. 15771. The method of item 15768 wherein the agent is an alpha 2 integrin antagonist, wherein the alpha 2 integrin antagonist is Pharmaprojects No. 5754 (Merck KgaA).
15772. The method of item 15768 wherein the agent is an alpha 4 integrin antagonist.
15773. The method of item 15768 wherein the agent is an alpha 7 nicotinic receptor agonist.
15774. The method of item 15768 wherein the agent is an angiogenesis inhibitor selected from the group consisting of AG-12,958 (Pfizer), ATN-161 (Attenuon LLC), neovastat, an angiogenesis inhibitor from Jerina AG (Germany), NM-3 (Mercian), VGA-1155 (Taisho), FCE-26644 (Pfizer), FCE-26950 (Pfizer), FPMA (Meiji Daries), FR-111142 (Fujisawa), GGTI-298, GM-1306 (Ligand), GPA-1734 (Novartis), NNC-47-0011 (Novo Nordisk), herbamycin (Nippon Kayaku), lenalidomide (Celegene), IP-10 (NIH), ABT-828 (Abbott), KIN-841 (Tokushima University, Japan), SF-1126 (Semafore Pharmaceuticals), laminin technology (NIH), CHIR-258 (Chiron), NVP-AEW541 (Novartis), NVP-AEW541 (Novartis), Vt16907 (Alchemia), OXI-8007 (Oxigene), EG-3306 (Ark Therapeutics), Maspin (Arriva), ABT- 567 (Abbott), PPI-2458 (Praecis Pharmaceuticals), CC-5079, CC-4089 (Celgene), HIF-1 alpha inhibitors (Xenova), S-247 (Pfizer), AP-23573 (Ariad), AZD-9935 (Astra Zeneca), mebendazole (Introgen Therapeutics), MetAP-2 inhibitors (GlaxoSmithKline), AG-615 (Angiogene Pharmaceuticals), Tie-2 antagonists (Hybrigenics), NC-381, CYC-381, NC-169, NC-219, NC-383, NC-384, NC-407 (Lorus Therapeutics), ATN-224 (Attenuon), ON-01370 (Onconova), Vitronectin antagonists (Amgen), SDX-103 (Salmedix), Vitronectin antagonists (Shire), CHP (Riemser), TEK (Amgen), Anecortave acetate (Alcon), T46.2 (Matrix Therapeutics), HG-2 (Heptagen), TEM antagonists (Genzyme), Oxi-4500 (Oxigene), ATN-161 (Attenuon), WX-293 (Wilex), M-2025 (Metris Therapeutics), Alphastatin (BioActa), YH-16 (Yantai Rongchang), BIBF-1120 (Boehringer Ingelheim), BAY-57-9352 (Bayer), AS- 1404 (Cancer Research Technology), SC-77964 (Pfizer), glycomimetics (BioTie Therapies), TIE-2 Inhibitors (Ontogen), DIMI, Octamer (Octamer), ABR-215050 (Active Biotech), ABT-518 (Abbott), KDR inhibitors (Abbott), BSF-466895 (Abbott), SCH-221153 (Schering-Plough), DAC:antiangiogenic (ConjuChem), TFPI (EntreMed), AZD-2171 (Astra-Zenaca), CDC-394 (Celgene), LY290293 (EIi Lilly), IDN-5390 (Indena), Kdr Kinase Inhibitors (Merck), CT-113020, CT-116433, CT-116563, CT-31890, CT-32228) (Cell Therapeutics), A-299620 (Abbott), TWEAK Inhibitor (Amgen), VEGF modulators (Johnson and Johnson), Tum-N53, tumstatin (Genzyme), Thios- 1 , Thios-2 (Thios Pharmaceuticals), MV-6401 (Miravant Medical Technologies), Spisulosine (PharmaMar), CEP-7055 (Cephalon), AUV-201 (Auvation), LM-609 (EIi Lilly), SKF-106615 (AnorMED), Oglufanide disodium (Cytran), BW-114 (Pharminox), Calreticulin (NIH), WX-678 (Wilex), SD-7784 (Pfizer), WX-UK1 (Wilex), SH-268 (Schering AG), 2-Me-PGA (Celgene), S- . 137 (Pfizer), ZD-6126 (Angiogene Pharmaceuticals), SG-292 (SignalGen), Benefin (Lane Labs), A6, A36 (Angstrom), SB-2723005 (GlaxoSmithKline), SC-7 (Cell Therapeutics), ZEN-014 (AEterna Zentaris), 2-methoxyestradiol (EntreMed), NK-130119 (Nippon Kayaku), CC-10004 (Celgene), AVE- 8062A (Ajinomoto), Tacedinaline (Pfizer), Actinonin (Tokyo Metropolitan Institute of Medical Science), Lenalidomide (Celgene), VGA-1155, BTO-956 (SRI International), ER-68203-00 (Eisai), CT-6685 (UCB), JKC-362 (Phoenix Pharmaceuticals), DMI-3798 (DMI Biosciences, Angiomate (Ipsen), ZD-6474 (AstraZeneca), CEP-5214 (Cephalon), Canstatin (Genzyme), NM-3 (Mercian), Oridigm (MediQuest Therapeutics), Exherin (Adherex), BLS-0597 (Boston Life Sciences), PTC-299 (PTC Therapeutics), NPI-2358 (Nereus Pharmaceuticals), CGP-79787 (Novartis), AEE-788 (Novartis), CKD-732 (Chong Kun Dang), CP-564959 (OSI Pharmaceuticals), CM-101 (CarboMed), CT-2584, CT3501 (Cell Therapeutics), combretastatin and analogues and derivatives thereof (Oxigene), Rebimastat (Bristol-Meyers Squibb), Dextrin 2-sulfate (ML Laboratories), Cilengitide (Merk KGaA), NSC-706704 (Pharminox), KRN- 951 (Kirin Brewery), Ukrain, NSC-631570 (Nowicky Pharma), Tecogalan sodium (Daiichi Pharmaceutical), Tz-93 (Tsumura), TBC-1635 (Encysive Pharmaceuticals), TAN-1120 (Takeda), Semaxanib (Pfizer), BDI-7800 (Biopharmacopae), SD-186, SD-983 (Bristol-Meyers Squibb), SB-223245 (GlaxoSmithKline), SC-236 (Pfizer), RWJ-590973 (Johnson and Johnson), ILX-1850 (Genzyme), SC-68488, S-836 (Pfizer), CG-55069-11 (CuraGen), Ki-23057 (Kirin Brewery), CCX-700 (Chemoentryx), Pegaptanib octasodium (Giled Sciences), ANGIOCOL (available from Biostratum Inc.), or an analogue or derivative thereof.
15775. The method of item 15768 wherein the agent is an apoptosis antagonist.
15776. The method of item 15768 wherein the agent is _ an apoptosis activator.
15777. The method of item 15768 wherein the agent is a beta 1 integrin antagonist.
15778. The method of item 15768 wherein the agent is a beta tubulin inhibitor.
15779. The method of item 15768 wherein the agent is a blocker of enzyme production in Hepatitis C.
15780. The method of item 15768 wherein the agent is a Bruton's tyrosine kinase inhibitor. 15781. The method of item 15768 wherein the agent is a calcineurin inhibitor.
15782. The method of item 15768 wherein the agent is a caspase 3 inhibitor.
15783. The method of item 15768 wherein the agent is a CC chemokine receptor antagonist.
15784. The method of item 15768 wherein the agent is a cell cycle inhibitor selected from the group consisting of SNS-595 (Sunesis), synthadotin, KRX-0403, and an analogue or derivative thereof.
15785. The method of item 15768 wherein the agent is a cathepsin B inhibitor.
15786. The method of item 15768 wherein the agent is a cathepsin K inhibitor, wherein the cathepsin K inhibitor is 462795 (GlaxoSmithKline), INPL-022-D6 (Amura Therapeutics), or an analogue or derivative thereof.
15787. The method of item 15768 wherein the agent is a cathepsin L inhibitor.
15788. The method of item 15768 wherein the agent is a CD40 antagonist.
15789. The method of item 15768 wherein the agent is a chemokine receptor agonist. 15790. The method of item 15768 wherein the agent is a chymase inhibitor.
15791. The method of item 15768 wherein the agent is a collagenase antagonist.
15792. The method of item 15768 wherein the agent is a CXCR antagonist.
15793. The method of item 15768 wherein the agent is a cyclin dependent kinase inhibitor selected from the group consisting of a CDK-1 inhibitor, a CDK-2 inhibitor, a CDK- 4 inhibitor, a CDK-6 inhibitor, a CAK1 inhibitor from GPC Biotech and Bristol-Myers Squibb, RGB-286199 (GPC Biotech), an anticancer agent from Astex Technology, a CAK1 inhibitor from GPC Biotech, a CDK inhibitor from Sanofi-Aventis, a CDK1/CDK2 inhibitor from Amgen, a CDK2 inhibitor from SUGEN-2 (Pfizer), a hearing loss therapy agent (Sound Pharmaceuticals), PD-0332991 (Pfizer), RGB-286199 (GPC Biotech), Ro-0505124 (Hoffmann-La Roche), a Ser/Thr kinase inhibitor from Lilly (EIi Lilly), CVT-2584 (CAS No. 199986-75- 9) (CV Therapeutics), and an analogue or derivative thereof.
15794. The method of item 15768 wherein the agent is a cyclooxygenase 1 inhibitor.
15795. The method of item 15768 wherein the agent is a DHFR inhibitor.
15796. The method of item 15768 wherein the agent is a dual integrin inhibitor. 15797. The method of item 15768 wherein the agent is an elastase inhibitor.
15798. The method of item 15768 wherein the agent is an elongation factor-1 alpha inhibitor.
15799. The method of item 15768 wherein the agent is an endothelial growth factor antagonist.
15800. The method of item 15768 wherein the agent is an endothelial growth factor receptor kinase inhibitor selected from the group consisting of sorafenib tosylate (Bayer), AAL-993 (Novartis), ABP-309 (Novartis), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), E- 7080 (Eisai), EG-3306 (Ark Therapeutics), EXEL-2880 (Exelixis), GW- 654652 (GlaxoSmithKline), a KDR inhibitor from LG Life Sciences, CT-6685 and CT-6729 (UCB), KRN-633 and KRN-951 (Kirin Brewery), OSI-930 (OSI Pharmaceuticals), SP-5.2 (Supratek Pharma), SU-11657 (Pfizer), a Tie-2 antagonist (Hybrigenics), a VEGFR-2 kinase inhibitor (Bristol-Myers Squibb), XL-647 (Exelixis), a KDR inhibitor from Abbott Laboratories, sorafenib tosylate, and an analogue or derivative thereof.
15801. The method of item 15768 wherein the agent is an endotoxin antagonist.
15802. The method of item 15768 wherein the agent is an epothilone and tubulin binder.
15803. The method of item 15768 wherein the agent is an estrogen receptor antagonist. 15804. The method of item 15768 wherein the agent is an FGF inhibitor.
15805. The method of item 15768 wherein the agent is a farnexyl transferase inhibitor.
15806. The method of item 15768 wherein the agent is farnesyltransferase inhibitor selected from the group of A-197574 (Abbott), a famesyltransferase inhibitor from Servier, FPTIII (Strathclyde Institute for Drug R), LB-42908 (LG Life Sciences), Pharmaprojects No. 5063 (Genzyme), Pharmaprojects No. 5597 (Ipsen), Yissum Project No. B-1055 (Yissum), and an analogue or derivative thereof.
15807. The method of item 15768 wherein the agent is an FLT-3 kinase inhibitor.
15808. The method of item 15768 wherein the agent is an FGF receptor kinase inhibitor.
15809. The method of item 15768 wherein the agent is a fibrinogen antagonist selected from the group consisting of AUV-201 (Auvation), MG-13926 (Sanofi-Aventis), plasminogen activator (CAS No. 105913-11-9) (from Sanofi-Aventis or UCB), plasminogen activator-2 (tPA- 2) (Sanofi-Aventis), pro-urokinase (CAS No. 82657-92-9) (Sanofi-Aventis), and an analogue or derivative thereof.
15810. The method of item 15768 wherein the agent is a heat shock protein 90 antagonist selected from the group consisting of SRN- 005 (Sirenade), geldanamycin, NSC-33050 (17-allylaminogeldanamycin; 17- AAG), 17-dimethylaminoethylamino-17-demethoxy-geldanamycin (17- DMAG), rifabutin (rifamycin XIV, r,4-didehydro-1-deoxy-1 ,4-dihydro-5'-(2- methylpropyl)-1-oxo-), and an analogue or derivative thereof.
15811. The method of item 15768 wherein the agent is a histone deacetylase inhibitor.
15812. The method of item 15768 wherein the agent is an HMGCoA reductase inhibitor selected from the group consisting of an atherosclerosis therapeutic from Lipid Sciences, ATI-16000 (ARYx Therapeutics), KS-01-019 (Kos Pharmaceuticals), Pharmaprojects No. 2197 (Sanofi-Aventi), RP 61969 (Sanofi-Aventis), and an analogue or derivative thereof.
15813. The method of item 15768 wherein the agent is an ICAM inhibitor.
15814. The method of item 15768 wherein the agent is an IL, ICE and IRAK antagonist, wherein the antagonist is a CJ-14877, CP- 424174 (Pfizer), NF-61 (Negma-Lerads), and an analogue or derivative thereof.
15815. The method of item 15768 wherein the agent is an IL-2 inhibitor.
15816. The method of item 15768 wherein the agent is an immunosuppressant selected from the group consisting of teriflunomide (Sanofi Aventis), chlorsulfaquinoxalone (NSC-339004), chlorsulfaquinoxalone sulfate, CS-712 (Sankyo), ismomultin alfa (CAS No. 457913-93-8) (Akzo Nobel), antiallergics from GenPat77, antiinflammatories or AT-005 (Androclus Therapeutics), autoimmune disease therapeutics from EpiVax, BN-007 (Bone), budesonide (CAS No. 51333-22- 3) (MAP Pharmaceuticals), CO-14 (Genzyme), edratide (CAS No. 433922- 67-9) (Teva), EP-314 (Enanta), eprovafen (CAS No. 101335-99-3) (Sanofi- Aventis), HWA-131 (CAS No. 118788-41-3) (Sanofi-Aventis), immunomodulators from MerLion Pharmaceuticals, immunosuppressives from Alchemia, IPL-12 (Inflazyme), MDL-9563 (CAS No. 27086-86-8) (Sanofi-Aventis), Pharmaprojects No. 2330 (Sanofi-Aventis), Pharmaprojects No. 6426 (Abgenix), PXS-25 (Pharmaxis), rosmarinic acid (CAS No. 20283-92-5) (Sanofi-Aventis), RP 42927 or RP 54745 (CAS No. 135330-08-4) (Sanofi-Aventis), SGN-35 (Seattle Genetics), ST-1959 (Sigma-Tau), type I diabetes therapy from SYNX Pharma, UNIL-88 (Debiopharm), VP-025 (Vasogen), VR-694 (Vectura), PRTX-001 (Protalex), and an analogue or derivative thereof.
15817. The method of item 15768 wherein the agent is an IMPDH (inosine monophosphate).
15818. The method of item 15768 wherein the agent is an integrin antagonist.
15819. The method of item 15768 wherein the agent is an interleukin antagonist.
15820. The method of item 15768 wherein the agent is an inhibitor of type III receptor tyrosine kinase.
15821. The method of item 15768 wherein the agent is an irreversible inhibitor of enzyme methionine aminopeptidase type 2.
15822. The method of item 15768 wherein the agent is an isozyme selective delta protein kinase C inhibitor. 15823. The method of item 15768 wherein the agent a
JAK3 enzyme inhibitor.
15824. The method of item 15768 wherein the agent is a
JNK inhibitor.
15825. The method of item 15768 wherein the agent is a kinase inhibitor.
15826. The method of item 15768 wherein the agent is kinesin antagonist.
15827. The method of item 15768 wherein the agent is a kinesin antagonist.
15828. The method of item 15768 wherein the agent is a leukotriene inhibitor and antagonist selected from the group consisting of ambicromil (CAS No. 58805-38-2) (Sanofi-Aventis), amelubant (CAS No. 346735-24-8) (Boehringer Ingelheim), DW-1141 (Dong Wha), ebselen (Daiichi Pharmaceutical), ibudilast (Kyorin), leucotriene inhibitors from Sanofi-Aventis, lymphotoxin-beta receptor (LT-D) from Biogen Idee, Pharmaprojects No. 1535 and 2728 (CAS No. 119340-33-9) (Sanofi- Aventis), R-112 (Rigel), Rev-5367 (CAS No. 92532-05-3) (Sanofi-Aventis), RG-14893 (CAS No. 141835-49-6) (Sanofi-Aventis), RG-5901-A (CAS No. 101910-24-1), 92532-23-5, RP 66153 (CAS No. 142422-79-5), RP 66364 (CAS No.186916-92-5), RP 69698 (CAS No. 141748-00-7) (Sanofi-Aventis), SC-411930 (Pfizer), SC-41930 (CAS No. 120072-59-5) (Pfizer), SC-50605 (CAS No. 138828-39-4) (Pfizer), SC-51146 (CAS No. 141059-52-1), SC- 53228 (CAS No. 153633-01-3) (Pfizer), spaglumic acid (ZY-15106) (CAS No.3106-85-2), 80619-64-3 (Novartis), tipredane (CAS No. 85197-77-9) (Bristol-Myers Squibb), U-75302 (CAS No. 119477-85-9) (Pfizer), and analogue or derivative thereof.
15829. The method of item 15768 wherein the agent is an MAP kinase inhibitor.
15830. The method of item 15768 wherein the agent is a matrix metalloproteinase inhibitor.
15831. The method of item 15768 wherein the agent is an MCP-CCR2 inhibitor.
15832. The method of item 15768 wherein the agent is an mTOR inhibitor.
15833. The method of item 15768 wherein the agent is an mTOR kinase inhibitor.
15834. The method of item 15768 wherein the agent is a microtubule inhibitor selected from the group consisting of antibody- maytansinoid conjugates from Biogen Idee, colchicines (MantiCore Pharmaceuticals), anticancer immunoconjugates from Johnson & Johnson, DIME from Octamer, gni-1f (GNI), huC242-DM4, huMy9-6-DM1 (ImmunoGen), IDN-5404 (Indena), IMO-098, IMOderm (Imotep), mebendazole (Introgen Therapeutics), microtubule poisons from Cambridge Enterprise, paclitaxel such as LOTAX from Aphios (CAS No. 33069-62-4), Genexol-PM from Samyang, Pharmaprojects No. 6383 (Tapestry Pharmaceuticals), RPR-112378 (Sanofi-Aventis), SGN-75 (Seattle Genetics), SPL-7435 (Starpharma), SSR-250411 (Sanofi-Aventis), trastuzumab-DM1 (Genentech), vinorelbine, and an analogue or derivative thereof. 15835. The method of item 15768 wherein the agent is an MIF inhibitor.
15836. The method of item 15768 wherein the agent is an MMP inhibitor.
15837. The method of item 15768 wherein the agent is a neurokinin (NK) antagonist selected from the group consisting of anthrotainin (CAS No. 148084-40-6) (Sanofi-Aventis), an IBS thereapeutic from ArQuIe, MDL-105212A (CAS No. 167261-60-1) (Ssanofi-Aventis), Pharmaprojects No. 2744, 3258 (CAS No. 139167-47-8) 4006, 4201 , or 5986 (Sanofi-Aventis), RP 67580 (CAS No. 135911-02-3), SR-144190 (CAS No. 201152-86-5), SSR-240600, SSR-241586 (Sanofi-Aventis), TKA-457 (Novartis), vestipitant mesylate (CAS No. 334476-64-1) (GlaxoSmithKline), Win-64821 (Sanofi-Aventis), PRX-96026 (Predix Pharmaceuticals), and an analogue or derivative thereof.
15838. The method of item 15768 wherein the agent is an NF kappa B inhibitor selected from the group consisting of AVE-0545 or AVE-0547 (Sanofi-Aventis), bortezomib (CAS No. 179324-69-7) (Millennium Pharmaceuticals), dexanabinol (CAS No. 112924-45-5) (Pharmos), dexlipotam (Viatris), Pharmaprojects No. 6283 (INDRA) (OXiGENE), IPL- 576092 (CAS No. 137571-30-3) (Inflazyme), NFKB decoy (Corgentech), NFKB decoy oligo (AnGes MG), NFKB's from Ariad, osteoporosis treatments or S5 (F005) from Fulcrum Pharmaceuticals, P61 (Phytopharm), R-flurbiprofen (CAS No. 5104-49-4) (Encore Pharmaceuticals), and an analogue or derivative thereof.
15839. The method of item 15768 wherein the agent is a nitric oxide agonist. 15840. The method of item 15768 wherein the agent is an ornithine decarboxylase inhibitor.
15841. The method of item 15768 wherein the agent is a p38 MAP kinase inhibitor selected from the group consisting of AZD-6703 (AstraZeneca), JX-401 (Jexys Pharmaceuticals), BMS-2 (Bristol-Myers Squibb), a p38 MAP kinase inhibitor from Novartis, a p38-alpha MAP kinase inhibitor from Amphora, Pharmaprojects No. 5704 (Pharmacopeia), RPR- 200765A (Sanofi-Aventis), SD-282 (Johnson & Johnson), TAK-715 (Takeda), and an analogue or derivative thereof.
15842. The method of item 15768 wherein the agent is a palmitoyl-protein thioesterase inhibitor.
15843. The method of item 15768 wherein the agent is a PDGF receptor kinase inhibitor selected from the group consisting of AAL- 993, AMN-107, or ABP-309 (Novartis), AMG-706 (Amgen), BAY-57-9352 (Bayer), CDP-860 (UCB), E-7080 (Eisai), imatinib (CAS No. 152459-95-5) (Novartis), OSI-930 (OSl Pharmaceuticals), RPR-127963E (Sanofi-Aventis), RWJ-540973 (Johnson & Johnson), sorafenib tosylate (Bayer), SU-11657 (Pfizer), tandutinib (CAS No. 387867-13-2) (Millennium Pharmaceuticals), vatalanib (Novartis), ZK-CDK (Schering AG), and an analogue or derivative thereof.
15844. The method of item 15768 wherein the agent is (-)-halofenate (Metabolex), AMG-131 (Amgen), antidiabetics from Japan Tobacco, AZD-4619, AZD-8450, AZD-8677 (AstraZeneca), DRF-10945, balaglitazone (Dr Reddy's), CS-00088, CS-00098 (Chipscreen Biosciences), E-3030 (Eisai), etalocib (CAS No. 161172-51-6) (EIi Lilly), GSK-641597 (Ligand), GSK-677954 (GlaxoSmithKline), GW-409544 (Ligand), GW- 590735 (GlaxoSmithKline), K-111 (Hoffmann-La Roche), LY-518674 (EIi Lilly), LY-674 (Ligand), LY-929 (Ligand), MC-3001 , MC-3002 (MaxoCore Pharmaceuticals), metformin HCI + pioglitazone (CAS No. 1115-70-4 and 112529-15-4), ACTOPLUS MET from Andrx), muraglitazar (CAS No. 331741-94-7) (Bristol-Myers Squibb), naveglitazar (Ligand), oleoylethanolamide (Kadmus Pharmaceuticals), ONO-5129, pioglitazone hydrochloride (CAS No. 111025-46-8 and 112529-15-4) (Takeda), PLX-204 (Plexxikon), PPAR agonists from Genfit, PPAR delta agonists from EIi Lilly, PPAR-alpha agonists from CrystalGenomics, PPAR-gamma modulators and PPAR-β modulators from CareX, rosiglitazone maleate (CAS No. 122324- 73-4 or 155141-29-0) (GlaxoSmithKline), rosiglitazone maleate/glimepir (CAS No. 155141-29-0 and 93479-97-1), AVANDARYL, rosiglitazone maleate/metformin extend (CAS No. 155141-29-0 and 657-24-9), AVANDAMET, rosiglitazone maleate+metformin, AVANDAMET (GiaxoSmithKline), tesaglitazar (AstraZeneca), and an analogue or derivative thereof.
15845. The method of item 15768 wherein the agent is a phosphatase inhibitor.
15846. The method of item 15768 wherein the agent is a phosphodiesterase (PDE) inhibitor selected from the group consisting of avanafil (Tanabe Seiyaku), dasantafil (CAS No. 569351-91-3) (Schering- Plough), A-906119 (CAS No. 134072-58-5), DL-850 (Sanofi-Aventis), GRC- 3015, GRC-3566, GRC-3886 (Glenmark), HWA-153 (CAS No. 56395-66-5) (Sanofi-Aventis), hydroxypumafentrine (Altana), IBFB-130011 , IBFB-14-016, IBFB-140301 , IBFB-150007, IBFB-211913 (IBFB Pharma), L-826141 (Merck & Co), medorinone (CAS No. 88296-61-1) (Sanofi-Aventis), MEM- 1917 (Memory Pharmaceuticals), ND-1251 (Neuro3d), PDE inhibitors from ICOS, PDE IV inhibitors from Memory Pharmaceuticals and CrystalGenomics, Pharmaprojects No. 2742 and 6141 (Sanofi-Aventis), QAD-171 (Novartis), RHC-2963 (CAS No. 76993-12-9 and 76993-14-1), RPR-117658, RPR-122818 derivatives, SR-24870 , and RPR-132294 (Sanofi-Aventis), SK-350 (ln2Gen), stroke therapy agents from deCODE Genetics, TAS-203 (Taiho), tofimilast (CAS No. 185954-27-2) (Pfizer), UK- 371800 (Pfizer), WIN-65579 (CAS No. 158020-82-7) (Sanofi-Aventis), IBFB- 130020 (IBFB Pharma), OPC-6535 (CAS No. 145739-56-6) (Otsuka), a phosphodiesterase III inhibitor, enoximone, a phosphodiesterase IV inhibitor, fosfosal, Atopik (Barrier Therapeutics), triflusal, a phosphodiesterase V inhibitor, and an analogue or derivative thereof.
15847. The method of item 15768 wherein the agent is a PKC inhibitor.
15848. The method of item 15768 wherein the agent is a platelet activating factor antagonist.
15849. The method of item 15768 wherein the agent is.a platelet-derived growth factor receptor kinase inhibitor.
15850. The method of item 15768 wherein the agent is a prolyl hydroxylase inhibitor.
15851. The method of item 15768 wherein the agent is a polymorphonuclear neutrophil inhibitor.
15852. The method of item 15768 wherein the agent is a protein kinase B inhibitor.
15853. The method of item 15768 wherein the agent is a protein kinase C stimulant. 15854. The method of item 15768 wherein the agent is a purine nucleoside analogue.
15855. The method of item 15768 wherein the agent is a purinoreceptor P2X antagonist.
15856. The method of item 15768 wherein the agent is a Raf kinase inhibitor.
15857. The method of item 15768 wherein the agent is a reversible inhibitor of ErbB1 and ErbB2.
15858. The method of item 15768 wherein the agent is a ribonucleoside triphosphate reductase inhibitor.
15859. The method of item 15768 wherein the agent is an SDF-1 antagonist.
15860. The method of item 15768 wherein the agent is a sheddase inhibitor.
15861. The method of item 15768 wherein the agent is an SRC inhibitor.
15862. The method of item 15768 wherein the agent is a stromelysin inhibitor.
15863. The method of item 15768 wherein the agent is an Syk kinase inhibitor. 15864. The method of item 15768 wherein the agent is a telomerase inhibitor.
15865. The method of item 15768 wherein the agent is a TGF beta inhibitor selected from the group consisting of pirfenidone (CAS No. 53179-13-8) (MARNAC)1 tranilast (CAS No. 53902-12-8) (Kissei), IN- 1130 (ln2Gen), mannose-6-phosphate (BTG), TGF-β antagonists from Inflazyme (Pharmaprojects No. 6075), TGF-β antagonists from Sydney, non-industrial source), TGF-β I receptor kinase inhibitors from EIi Lilly, TGF- β receptor inhibitors from Johnson & Johnson, and an analogue or derivative thereof.
15866. The method of item 15768 wherein the agent is a TNFα antagonist or TACE inhibitor selected from the group consisting of adalimumab (CAS No. 331731-18-1) (Cambridge Antibody Technology), AGIX-4207 (AtheroGenics), AGT-1 (Advanced Biotherapy), an antiinflammatory from Borean Pharma, Cellzome, or Paradigm Therapeutics, anti-inflammatory vaccine (TNF-alpha kinoid) from Neovacs, humanized anti-TNF antibody or an anti-TNF MAb (CB0006) Celltech (UCB), apratastat (CAS No. 287445-51-0) (Wyeth), BMS-561392 (Bristol-Myers Squibb), BN- 006 (Bone), certolizumab pegol (CAS No. 428863-50-7 or CH-138 (UCB), cilomilast (CAS No. 153259-65-5) (GlaxoSmithKline), CR-1 (Nuada Pharmaceuticals), CRx-119 (CombinatoRx), D-5410 (UCB), dacopafant (CAS No. 125372-33-0) (Sanofi-Aventis), dersalazine (CAS No. 188913-57- 7/188913-58-8) (Uriach), etanercept (CAS No. 185243-69-0) (Amgen), ethyl pyruvate (Critical (Critical Therapeutics), golimumab (CAS No. 476181-74-5) (Johnson & Johnson), hormono-immunotherapy from Ipsen, CDP571 (e.g., Humicade from UCB), IC-485 (ICOS), infliximab (CAS No. 170277-31-3) (Johnson & Johnson), IP-751 (Manhattan Pharmaceuticals), ISIS-104838 (CAS No. 250755-32-9) (ISIS Pharmaceuticals), lenalidomide (CAS No. 191732-72-6) (Celgene), lentinan (CAS No. 37339-90-5) (Ajinomoto), MDL- 201112 (CAS No. 142130-73-2) (Sanofi-Aventis), medroxyprogesterone (CAS No. 520-85-4) (InKine Pharmaceutical), N-acetylcysteine (CAS No. 616-91-1) (Zambon), NBE-P2 (DIREVO Biotech), nerelimomab (CAS No. 162774-06-3) (Chiron), OM-294DP (OM PHARMA), onercept (CAS No. 199685-57-9) (Yeda), PASSTNF-alpha (Verigen), pentoxifylline or oxypentifylline (Sanofi-Aventis), Pharmaprojects No. 4091 , 4241 , 4295, or 4488 (Sanofi-Aventis), Pharmaprojects No. 5480 (Amgen), Pharmaprojects No. 6749 (Cengent), pirfenidone (CAS No. 53179-13-8) (MARNAC), RPR- 132294 (Sanofi-Aventis), S5 (F002) (Fulcrum Pharmaceuticals), simvastatin (CAS No. 79902-63-9) (Merck & Co), STA-6292 (Synta Pharmaceuticals), tacrolimus (CAS No. 104987-11-3) (Fujisawa LifeCycle Pharma), talactoferrin alfa (CAS No. 308240-58-6) (Agennix), thalidomide (CAS No. 50-35-1) (Celgene), TNF antagonists form ProStrakan, and Synergen, TNF inhibitors (Amgen), TNF-alpha antagonists from Dynavax Technologies and Jerina AG (Germany), TNF-alpha inhibitors from IBFB Pharma and Xencor (Xencor), torbafylline (CAS No. 105102-21-4) (Sanofi-Aventis), UR-1505 (Uriach), VT-346 (Viron Therapeutics), YSIL6 (Y's Therapeutics), YSTH2 (Y's Therapeutics), NPI-1302a-3 (Nereus Pharmaceuticals, a TNF antagonist from Jerina AG (Germany), dersalazine, and an analogue or derivative thereof.
15867. The method of item 15768 wherein the agent is a Toll receptor inhibitor.
15868. The method of item 15768 wherein the agent is a tubulin antagonist.
15869. The method of item 15768 wherein the agent is a tyrosine kinase inhibitor selected from the group consisting of SU-011248, SUTENT from Pfizer Inc. (New York, NY), BMS-354825, PN-355 (Paracelsian Pharmaceuticals), AGN-199659 (Allergan), AAL-993 or ABP- 309 (Novartis), adaphostin (NIH), AEE-788 (Novartis), AG-013736 (OSl Pharmaceuticals), AG-13736 (Pfizer), ALT-110 (Alteris Therapeutics), AMG- 706 (Amgen), anticancer MAbs from Xencor, anti-EGFrvlll MAbs from Abgenix, anti-HER2 MAb from Abiogen, AZD-2171 or AZD-9935 (AstraZeneca), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), CEP-5214 (Cephalon), CEP-7055 (Cephalon), cetuximab (ImCIone Systems), CHIR-200131 and CHIR-258 (Chiron), CP-547632 (OSI Pharmaceuticals), CP-724714 (Pfizer), CT-301 (Creabilis Therapeutics), D- 69491 (Baxter International), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EGFR/ErbB2 inhibitors from Array BioPharma, erlotinib (CAS No. 183319- 69-9) (OSI Pharmaceuticals), EXEL-2880 (Exelixis), FK-778 (Sanofi- Aventis) , gefitinib (CAS No. 184475-35-2) (AstraZeneca), GW-2286 or GW- 654652 (GlaxoSmithKline), her2/neu antigen from AlphaVax, HER-2/neu inhibitor from Generex, Herzyme (Medipad) (Sirna Therapeutics), HKI-272 (Wyeth), HuMax-EGFr (Genmab), idronoxil (CAS No. 81267-65-4) (Novogen), IGF-1 inhibitors from Ontogen, IMC-11F8 (ImCIone Systems), kahalalide F (CAS No. 149204-42-2) (PharmaMar), KDR inhibitor from LG Life Sciences, KDR inhibitors from Abbott Laboratories, KDR kinase inhibitors (UCB), Kdr kinase inhibitors from Merck & Co, KRN-633 and KRN- 951 (Kirin Brewery), KSB-102 (Xenova), lapatinib ditosylate (CAS No. 388082-78-8) (GlaxoSmithKline), matuzumab (Merck KGaA), MDX-214 (Medarex), ME-103 (Pharmexa), MED-A300 (Gerolymatos), MNAC-13 (Lay Line Genomics), nimotuzumab (Center of Molecular Immunology), NSC- 330507 or NSC-707545 (NIH), NV-50 (Novogen), OSI-930 (OSI Pharmaceuticals), panitumumab (Abgenix), pelitinib (CAS No. 287933-82-7) (Wyeth), pertuzumab (CAS No. 380610-27-5) (Genentech), Pharmaprojects No. 3985 (Sanofi-Aventis), prostate cancer therapeutics from Sequenom (SQPC35, SQPC36, SQPC90), removab and remoxab (Trion Pharma), RG- 13022 (CAS No. 136831-48-6), RG-13291 (CAS No. 138989-50-1), or RG- 14620 (CAS No. 136831-49-7) (Sanofi-Aventis), RM-6427 (Romark), RNAi breast cancer therapy from Benitec, RP 53801 (CAS No. 125882-88-4) (Sanofi-Aventis), sorafenib tosylate (Bayer), SU-11657 (Pfizer), Tie-2 antagonists from Semaia (Hybrigenics), Tie-2 inhibitors from Ontogen, trastuzumab (CAS No. 180288-69-1) (Genentech), tyrosine kinase inhibitors from Sanofi-Aventis, U3-1287, U3-1565, U3-1784, U3-1800 (U3 Pharma), vatalanib (Novartis), VEGFR-2 kinase inhibitor from Bristol-Myers Squibb, XL-647 (Exelixis), ZD-6474 (AstraZeneca), ZK-CDK (Schering AG), an EGFR tyrosine kinase inhibitor, EKB-569 (Wyeth), and an analogue or derivative thereof.
15870. The method of item 15768 wherein the agent is a VEGF inhibitor.
15871. The method of item 15768 wherein the agent is a vitamin D receptor agonist.
15872. The method of item 15768 wherein the agent is ZD-6474 (an angiogenesis inhibitor).
15873. The method of item 15768 wherein the agent is AP-23573 (an mTOR inhibitor).
15874. The method of item 15768 wherein the agent is synthadotin (a tubulin antagonist).
15875. The method of item 15768 wherein the agent is S-0885 (a collagenase inhibitor).
15876. The method of item 15768 wherein the agent is aplidine (an elongation factor-1 alpha inhibitor). 15877. The method of item 15768 wherein the agent is ixabepilone (an epithilone).
15878. The method of item 15768 wherein the agent is IDN-5390 (an angiogenesis inhibitor).
15879. The method of item 15768 wherein the agent is SB-2723005 (an angiogenesis inhibitor).
15880. The method of item 15768 wherein the agent is ABT-518 (an angiogenesis inhibitor).
15881. The method of item 15768 wherein the agent is combretastatin (an angiogenesis inhibitor).
15882. The method of item 15768 wherein the agent is anecortave acetate (an angiogenesis inhibitor).
15883. The method of item 15768 wherein the agent is SB-715992 (a kinesin antagonist).
15884. The method of item 15768 wherein the agent is temsirolimus (an mTOR inhibitor).
15885. The method of item 15768 wherein the agent is adalimumab (a TNFα antagonist).
15886. The method of item 15768, wherein the composition comprises a polymer. 15887. The method of item 15768, wherein the composition comprises a polymeric carrier.
15888. The method of item 15768 wherein the anti- scarring agent inhibits adhesion between the device and a host into which the device is implanted.
15889. The method of item 15768 wherein the device delivers the anti-scarring agent locally to tissue proximate to the device.
15890. The method of item 15768 wherein the device has a coating that comprises the anti-scarring agent.
15891. The method of item 15768, wherein the device has a coating that comprises the agent and is disposed on a surface of the implant.
15892. The method of item 15768, wherein the device has a coating that comprises the agent and directly contacts the implant.
15893. The method of item 15768, wherein the device has a coating that comprises the agent and indirectly contacts the implant.
15894. The method of item 15768, wherein the device has a coating that comprises the agent and partially covers the implant.
15895. The method of item 15768, wherein the device has a coating that comprises the agent and completely covers the implant.
15896. The method of item 15768, wherein the device has a uniform coating. 15897. The method of item 15768, wherein the device has a non-uniform coating.
15898. The method of item 15768, wherein the device has a discontinuous coating.
15899. The method of item 15768, wherein the device has a patterned coating.
15900. The method of item 15768, wherein the device has a coating with a thickness of 100 Dm or less.
15901. The method of item 15768, wherein the device has a coating with a thickness of 10 Dm or less.
15902. The method of item 15768, wherein the device- has a coating, and the coating adheres to the surface of the implant upon deployment of the implant.
15903. The method of item 15768, wherein the device has a coating, and wherein the coating is stable at room temperature for a period of 1 year.
15904. The method of item 15768, wherein the device has a coating, and wherein the anti-scarring agent is present in the coating in an amount ranging between about 0.0001% to about 1% by weight.
15905. The method of item 15768, wherein the device has a coating, and wherein the anti-scarring agent is present in the coating in an amount ranging between about 1% to about 10% by weight. 15906. The method of item 15768, wherein the device has a coating, and wherein the anti-scarring agent is present in the coating in an amount ranging between about 10% to about 25% by weight.
15907. The method of item 15768, wherein the device has a coating, and wherein the anti-scarring agent is present in the coating in an amount ranging between about 25% to about 70% by weight.
15908. The method of item 15768, wherein the device has a coating, and wherein the coating further comprises a polymer.
15909. The method of item 15768, wherein the device has a first coating having a first composition and a second coating having a second composition.
15910. The method of item 15768, wherein tha.device has a first coating having a first composition and a second coating having a second composition, wherein the first composition and the second composition are different.
15911. The method of item 15768, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a copolymer.
15912. The method of item 15768, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a block copolymer.
15913. The method of item 15768, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a random copolymer. 15914. The method of item 15768, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a biodegradable polymer.
15915. The method of item 15768, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a non-biodegradable polymer.
15916. The method of item 15768, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a hydrophilic polymer.
15917. The method of item 15768, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a hydrophobic polymer.
15918. The method of item 15768, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a polymer having hydrophilic domains.
15919. The method of item 15768, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a polymer having hydrophobic domains.
15920. The method of item 15768, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a non-conductive polymer.
15921. The method of item 15768, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises an elastomer. 15922. The method of item 15768, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a hydrogel.
15923. The method of item 15768, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a silicone polymer.
15924. The method of item 15768, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a hydrocarbon polymer.
15925. The method of item 15768, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a styrene-derived polymer.
15926. The method of item 15768, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a butadiene polymer.
15927. The method of item 15768, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a macromer.
15928. The method of item 15768, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a poly(ethylene glycol) polymer.
15929. The method of item 15768 wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises an amorphous polymer. 15930. The method of item 15768, wherein the device comprises a lubricious coating.
15931. The method of item 15768 wherein the anti- scarring agent is located within pores or holes of the device.
15932. The method of item 15768 wherein the anti- scarring agent is located within a channel, lumen, or divet of the device.
15933. The method of item 15768, wherein the device comprises a second pharmaceutically active agent.
15934. The method of item 15768 wherein the device comprises an anti-inflammatory agent.
15935. The method of item 15768 wherein the device comprises an agent that inhibits infection.
15936. The method of item 15768 wherein the device comprises an agent that inhibits infection, and wherein the agent is an anthracycline.
15937. The method of item 15768 wherein the device comprises an agent that inhibits infection, and wherein the agent is doxorubicin.
15938. The method of item 15768 wherein the device comprises an agent that inhibits infection, and wherein the agent is mitoxantrone. 15939. The method of item 15768 wherein the device comprises an agent that inhibits infection, and wherein the agent is a fluoropyrimidine.
15940. The method of item 15768 wherein the device comprises an agent that inhibits infection, and wherein the agent is 5- fluorouracil (5-FU).
15941. The method of item 15768 wherein the device comprises an agent that inhibits infection, and wherein the agent is a folic acid antagonist.
15942. The method of item 15768 wherein the device comprises an agent that inhibits infection, and wherein the agent is methotrexate.
15943. The method of item 15768 wherein the device comprises an agent that inhibits infection, and wherein the agent is a podophylotoxin.
15944. The method of item 15768 wherein the device comprises an agent that inhibits infection, and wherein the agent is etoposide.
15945. The method of item 15768 wherein the device comprises an agent that inhibits infection, and wherein the agent is a camptothecin.
15946. The method of item 15768 wherein the device comprises an agent that inhibits infection, and wherein the agent is a hydroxyurea. 15947. The method of item 15768 wherein the device comprises an agent that inhibits infection, and wherein the agent is a platinum complex.
15948. The method of item 15768 wherein the device comprises an agent that inhibits infection, and wherein the agent is cisplatin.
15949. The method of item 15768, further comprising an anti-thrombotic agent.
15950. The method of item 15768 wherein the device comprises a visualization agent.
15951. The method of item 15768 wherein the device comprises a visualization agent, wherein the visualization agent is a radiopaque material, and wherein the radiopaque material comprises a metal, a halogenated compound, or a barium containing compound.
15952. The method of item 15768 wherein the device comprises a visualization agent, wherein the visualization agent is a radiopaque material, and wherein the radiopaque material comprises barium, tantalum, or technetium.
15953. The method of item 15768 wherein the device comprises a visualization agent, and wherein the visualization agent is a MRI responsive material.
15954. The method of item 15768 wherein the device comprises a visualization agent, and wherein the visualization agent comprises a gadolinium chelate. 15955. The method of item 15768 wherein the device comprises a visualization agent, and wherein the visualization agent comprises iron, magnesium, manganese, copper, or chromium.
15956. The method of item 15768 wherein the device comprises a visualization agent, and wherein the visualization agent comprises an iron oxide compound.
15957. The method of item 15768 wherein the device comprises a visualization agent, and wherein the visualization agent comprises a dye, pigment, or colorant.
15958. The method of item 15768 wherein the device comprises an echogenic material.
15959. The method of item 15768 wherein the device comprises an echogenic material, and wherein the echogenic material is in the form of a coating.
15960. The method of item 15768 wherein the device is sterile.
15961. The method of item 15768 wherein the anti- scarring agent is released into tissue in the vicinity of the device after deployment of the device.
15962. The method of item 15768 wherein the anti- scarring agent is released into tissue in the vicinity of the device after deployment of the device, and wherein the tissue is connective tissue. 15963. The method of item 15768 wherein the anti- scarring agent is released into tissue in the vicinity of the device after deployment of the device, and wherein the tissue is muscle tissue.
15964. The method of item 15768 wherein the anti- scarring agent is released into tissue in the vicinity of the device after deployment of the device, and wherein the tissue is nerve tissue.
15965. The method of item 15768 wherein the anti- scarring agent is released into tissue in the vicinity of the device after deployment of the device, and wherein the tissue is epithelium tissue.
15966. The method of item 15768 wherein the anti- scarring agent is released in effective concentrations from the device over a period ranging from the time of deployment of the device to about 1 year.
15967. The method of item 15768 wherein the anti- scarring agent is released in effective concentrations from the device over a period ranging from about 1 month to 6 months.
15968. The method of item 15768 wherein the anti- scarring agent is released in effective concentrations from the device over a period ranging from about 1 - 90 days.
15969. The method of item 15768 wherein the anti- scarring agent is released in effective concentrations from the device at a constant rate.
15970. The method of item 15768 wherein the anti- scarring agent is released in effective concentrations from the device at an increasing rate. 15971. The method of item 15768 wherein the anti- scarring agent is released in effective concentrations from the device at a decreasing rate.
15972. The method of item 15768 wherein the anti- scarring agent is released in effective concentrations from the composition comprising the anti-scarring agent by diffusion over a period ranging from the time of deployment of the device to about 90 days.
15973. The method of item 15768 wherein the anti- scarring agent is released in effective concentrations from the composition comprising the anti-scarring agent by erosion of the composition over a period ranging from the time of deployment of the device to about 90 days.
15974. The method of item 15768 wherein the device comprises about 0.01 Dg to about 10 Dg of the anti-scarring agent.
15975. The method of item 15768 wherein the device comprises about 10 Dg to about 10 mg of the anti-scarring agent.
15976. The method of item 15768 wherein the device comprises about 10 mg to about 250 mg of the anti-scarring agent.
15977. The method of item 15768 wherein the device comprises about 250 mg to about 1000 mg of the anti-scarring agent.
15978. The method of item 15768 wherein the device comprises about 1000 mg to about 2500 mg of the anti-scarring agent. 15979. The method of item 15768 wherein a surface of the device comprises less than 0.01 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
15980. The method of item 15768 wherein a surface of the device comprises about 0.01 Dg to about 1 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
15981. The method of item 15768 wherein a surface of the device comprises about 1 Dg to about 10 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
15982. The method of item 15768 wherein a surface of the device comprises about 10 Dg to about 250 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
15983. The method of item 15768 wherein a surface of the device comprises about 250 Dg to about 1000 Dg of the anti-scarring agent of anti-scarring agent per mm2 of device surface to which the anti- scarring agent is applied.
15984. The method of item 15768 wherein a surface of the device comprises about 1000 Dg to about 2500 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
15985. The method of item 15768 wherein the combining is performed by direct affixing the agent or the composition to the implant. 15986. The method of item 15768 wherein the combining is performed by spraying the agent or the component onto the implant.
15987. The method of item 15768 wherein the combining is performed by electrospraying the agent or the composition onto the implant.
15988. The method of item 15768 wherein the combining is performed by dipping the implant into a solution comprising the agent or the composition.
15989. The method of item 15768 wherein the combining is performed by covalently attaching the agent or the composition to the implant.
15990. The method of item 15768 wherein the combining is performed by non-covalently attaching the agent or the composition to the implant.
15991. The method of item 15768 wherein the combining is performed by coating the implant with a substance that contains the agent or the composition.
15992. The method of item 15768 wherein the combining is performed by coating the implant with a substance that absorbs the agent.
15993. The method of item 15768 wherein the combining is performed by interweaving a thread composed of, or coated with, the agent or the composition. 15994. The method of item 15768 wherein the combining is performed by covering all the implant with a sleeve that contains the agent or the composition.
15995. The method of item 15768 wherein the combining is performed by covering a portion of the implant with a sleeve that contains the agent or the composition.
15996. The method of item 15768 wherein the combining is performed by covering all the implant with a cover that contains the agent or the composition.
15997. The method of item 15768 wherein the combining is performed by covering a portion of the implant with a cover that contains the agent or the composition.
15998. The method of item 15768 wherein the combining is performed by covering all the implant with an electrospun fabric that contains the agent or the composition.
15999. The method of item 15768 wherein the combining is performed by covering a portion of the implant with an electrospun fabric that contains the agent or the composition.
16000. The method of item 15768 wherein the combining is performed by covering all the implant with a mesh that contains the agent or the composition.
16001. The method of item 15768 wherein the combining is performed by covering a portion of the implant with a mesh that contains the agent or the composition. 16002. The method of item 15768 wherein the combining is performed by constructing all the implant with the agent or the composition.
16003. The method of item 15768 wherein the combining is performed by constructing a portion of the implant with the agent or the composition.
16004. The method of item 15768 wherein the combining is performed by impregnating the implant with the agent or the composition.
16005. The method of item 15768 wherein the combining is performed by constructing all of the implant from a degradable polymer that releases the agent.
16006. The method of item 15768 wherein the combining is performed by constructing a portion of the implant from a degradable polymer that releases the agent.
16007. The method of item 15768 wherein the combining is performed by dipping the implant into a solution that comprise the agent and an inert solvent for the implant.
16008. The method of item 15768 wherein the combining is performed by dipping the implant into a solution that comprises the agent and a solvent that will swell the implant.
16009. The method of item 15768 wherein the combining is performed by dipping the implant into a solution that comprises the agent and a solvent that will dissolve the implant. 16010. The method of item 15768 wherein the combining is performed by dipping the implant into a solution that comprises the agent, a polymer and an inert solvent for the implant.
16011. The method of item 15768 wherein the combining is performed by dipping the implant into a solution that comprises the agent, a polymer and a solvent that will swell the implant.
16012. The method of item 15768 wherein the combining is performed by dipping the implant into a solution that comprises the agent, a polymer and a solvent that will dissolve the implant.
16013. The method of item 15768 wherein the combining is performed by spraying the implant into a solution that comprises the agent and an inert solvent for the implant.
16014. The method of item 15768 wherein the combining is performed by spraying the implant into a solution that comprises the agent and a solvent that will swell the implant.
16015. The method of item 15768 wherein the combining is performed by spraying the implant into a solution that comprises the agent and a solvent that will dissolve the implant.
16016. The method of item 15768 wherein the combining is performed by spraying the implant into a solution that comprises the agent, a polymer and an inert solvent for the implant.
16017. The method of item 15768 wherein the combining is performed by spraying the implant into a solution that comprises the agent, a polymer and a solvent that will swell the implant. 16018. The method of item 15768 wherein the combining is performed by spraying the implant into a solution that comprises the agent, a polymer and a solvent that will dissolve the implant.
16019. A method as in any one of items 15768-16018, wherein the device is a vascular filter.
16020. A method as in any one of items 15768-16018, wherein the device is a blood filter.
16021. A method as in any one of items 15768-16018, wherein the device is a caval filter.
16022. A method as in any one of items 15768-16018, wherein the device is a vena cava filter.
16023. A method as in any one of items 15768-16018, wherein the device is a thrombus filter.
16024. A method as in any one of items 15768-16018, wherein the device is an antimigration filter.
16025. A method as in any one of items 15768-16018, wherein the device is a percutaneous filter system.
16026. A method as in any one of items 15768-16018, wherein the device is an intravascular trap.
16027. A method as in any one of items 15768-16018, wherein the device is an intravascular filter. 16028. A method as in any one of items 15768-16018, wherein the device is a clot filter.
16029. A method as in any one of items 15768-16018, wherein the device is a vein filter.
16030. A method as in any one of items 15768-16018, wherein the device is a body vessel filter.
16031. A method of making a medical device comprising: combining a gastrointestinal device (e.g., an implant) and an anti-scarring agent or a composition comprising an anti-scarring agent, wherein the agent inhibits scarring between the device and a host into which the device is implanted.
16032. The method of item 16031 wherein the agent is an adensosine A2A receptor antagonist.
16033. The method of item 16031 wherein the agent is an AKT inhibitor.
16034. The method of item 16031 wherein the agent is an alpha 2 integrin antagonist, wherein the alpha 2 integrin antagonist is Pharmaprojects No. 5754 (Merck KgaA).
16035. The method of item 16031 wherein the agent is an alpha 4 integrin antagonist.
16036. The method of item 16031 wherein the agent is an alpha 7 nicotinic receptor agonist. 16037. The method of item 16031 wherein the agent is an angiogenesis inhibitor selected from the group consisting of AG-12,958 (Pfizer), ATN-161 (Attenuon LLC), neovastat, an angiogenesis inhibitor from Jerina AG (Germany), NM-3 (Mercian), VGA-1155 (Taisho), FCE-26644 (Pfizer), FCE-26950 (Pfizer), FPMA (Meiji Daries), FR-111142 (Fujisawa), GGTI-298, GM-1306 (Ligand), GPA-1734 (Novartis), NNC-47-0011 (Novo Nordisk), herbamycin (Nippon Kayaku), lenalidomide (Celegene), IP-10 (NIH), ABT-828 (Abbott), KIN-841 (Tokushima University, Japan), SF-1126 (Semafore Pharmaceuticals), laminin technology (NIH), CHIR-258 (Chiron), NVP-AEW541 (Novartis), NVP-AEW541 (Novartis), Vt16907 (Alchemia), OXI-8007 (Oxigene), EG-3306 (Ark Therapeutics), Maspin (Arriva), ABT- 567 (Abbott), PPI-2458 (Praecis Pharmaceuticals), CC-5079, CC-4089 (Celgene), HIF-1alpha inhibitors (Xenova), S-247 (Pfizer), AP-23573 (Ariad), AZD-9935 (Astra Zeneca), mebendazole (Introgen Therapeutics), MetAP-2 inhibitors (GlaxoSmithKline), AG-615 (Angiogene Pharmaceuticals), Tie-2 antagonists (Hybπgenics), NC-381 , CYC-381 , NC-169, NC-219, NC-383, NC-384, NC-407 (Lorus Therapeutics), ATN-224 (Attenuon), ON-01370 (Onconova), Vitronectin antagonists (Amgen), SDX-103 (Salmedix), Vitronectin antagonists (Shire), CHP (Riemser), TEK (Amgen), Anecortave acetate (Alcon), T46.2 (Matrix Therapeutics), HG-2 (Heptagen), TEM antagonists (Genzyme), Oxi-4500 (Oxigene), ATN-161 (Attenuon), WX-293 (Wilex), M-2025 (Metris Therapeutics), Alphastatin (BioActa), YH-16 (Yantai Rongchang), BIBF-1120 (Boehringer Ingelheim), BAY-57-9352 (Bayer), AS- 1404 (Cancer Research Technology), SC-77964 (Pfizer), glycomimetics (BioTie Therapies), TIE-2 Inhibitors (Ontogen), DIMI, Octamer (Octamer), ABR-215050 (Active Biotech), ABT-518 (Abbott), KDR inhibitors (Abbott), BSF-466895 (Abbott), SCH-221153 (Schering-Plough), DACrantiangiogenic (ConjuChem), TFPI (EntreMed), AZD-2171 (Astra-Zenaca), CDC-394 (Celgene), LY290293 (EIi Lilly), IDN-5390 (Indena), Kdr Kinase Inhibitors (Merck), CT-113020, CT-116433, CT-116563, CT-31890, CT-32228) (Cell Therapeutics), A-299620 (Abbott), TWEAK Inhibitor (Amgen), VEGF modulators (Johnson and Johnson), Tum-N53, tumstatin (Genzyme), Thios- 1, Thios-2 (Thios Pharmaceuticals), MV-6401 (Miravant Medical Technologies), Spisulosine (PharmaMar), CEP-7055 (Cephalon), AUV-201 (Auvation), LM-609 (EIi Lilly), SKF-106615 (AnorMED), Oglufanide disodium (Cytran), BW-114 (Pharminox), Calreticulin (NIH), WX-678 (Wilex), SD-7784 (Pfizer), WX-UK1 (Wilex), SH-268 (Schering AG), 2-Me-PGA (Celgene), S- 137 (Pfizer), ZD-6126 (Angiogene Pharmaceuticals), SG-292 (SignalGen), Benefin (Lane Labs), A6, A36 (Angstrom), SB-2723005 (GlaxoSmithKline), SC-7 (Cell Therapeutics), ZEN-014 (AEterna Zentaris), 2-methoxyestradiol (EntreMed), NK-130119 (Nippon Kayaku), CC-10004 (Celgene), AVE- 8062A (Ajinomoto), Tacedinaline (Pfizer), Actinonin (Tokyo Metropolitan Institute of Medical Science), Lenalidomide (Celgene), VGA-1155, BTO-956 (SRI International), ER-68203-00 (Eisai), CT-6685 (UCB), JKC-362 (Phoenix Pharmaceuticals), DMI-3798 (DMI Biosciences, Angiomate (Ipsen), ZD-6474 (AstraZeneca), CEP-5214 (Cephalon), Canstatin (Genzyme), NM-3 (Mercian), Oridigm (MediQuest Therapeutics), Exherin (Adherex), BLS-0597 (Boston Life Sciences), PTC-299 (PTC Therapeutics), NPI-2358 (Nereus Pharmaceuticals), CGP-79787 (Novartis), AEE-788 (Novartis), CKD-732 (Chong Kun Dang), CP-564959 (OSI Pharmaceuticals), CM-101 (CarboMed), CT-2584, CT3501 (Cell Therapeutics), combretastatin and analogues and derivatives thereof (Oxigene), Rebimastat (Bristol-Meyers Squibb), Dextrin 2-sulfate (ML Laboratories), Cilengitide (Merk KGaA), NSC-706704 (Pharminox), KRN- 951 (Kirin Brewery), Ukrain, NSC-631570 (Nowicky Pharma), Tecogalan sodium (Daiichi Pharmaceutical), Tz-93 (Tsumura), TBC-1635 (Encysive Pharmaceuticals), TAN-1120 (Takeda), Semaxanib (Pfizer), BDI-7800 (Biopharmacopae), SD-186, SD-983 (Bristol-Meyers Squibb), SB-223245 (GlaxoSmithKline), SC-236 (Pfizer), RWJ-590973 (Johnson and Johnson), ILX-1850 (Genzyme), SC-68488, S-836 (Pfizer), CG-55069-11 (CuraGen), Ki-23057 (Kirin Brewery), CCX-700 (Chemoentryx), Pegaptanib octasodium (Giled Sciences), ANGIOCOL (available from Biostratum Inc.), or an analogue or derivative thereof.
16038. The method of item 16031 wherein the agent is an apoptosis antagonist.
16039. The method of item 16031 wherein the agent is an apoptosis activator.
16040. The method of item 16031 wherein the agent is a beta 1 integrin antagonist.
16041. The method of item 16031 wherein the agent is a beta tubulin inhibitor.
16042. The method of item 16031 wherein the agent is a blocker of enzyme production in Hepatitis C.
16043. The method of item 16031 wherein the agent is a Bruton's tyrosine kinase inhibitor.
16044. The method of item 16031 wherein the agent is a calcineurin inhibitor.
16045. The method of item 16031 wherein the agent is a caspase 3 inhibitor.
16046. The method of item 16031 wherein the agent is a CC chemokine receptor antagonist. 16047. The method of item 16031 wherein the agent is a cell cycle inhibitor selected from the group consisting of SNS-595 (Sunesis), synthadotin, KRX-0403, and an analogue or derivative thereof.
16048. The method of item 16031 wherein the agent is a cathepsin B inhibitor.
16049. The method of item 16031 wherein the agent is a cathepsin K inhibitor, wherein the cathepsin K inhibitor is 462795 (GlaxoSmithKline), INPL-022-D6 (Amura Therapeutics), or an analogue or derivative thereof.
16050. The method of item 16031 wherein the agent is a cathepsin L inhibitor.
16051. The method of item 16031 wherein the agent is a CD40 antagonist.
16052. The method of item 16031 wherein the agent is a chemokine receptor agonist.
16053. The method of item 16031 wherein the agent is a chymase inhibitor.
16054. The method of item 16031 wherein the agent is a collagenase antagonist.
16055. The method of item 16031 wherein the agent is a CXCR antagonist. 16056. The method of item 16031 wherein the agent is a cyclin dependent kinase inhibitor selected from the group consisting of a CDK-1 inhibitor, a CDK-2 inhibitor, a CDK- 4 inhibitor, a CDK-6 inhibitor, a CAK1 inhibitor from GPC Biotech and Bristol-Myers Squibb, RGB-286199 (GPC Biotech), an anticancer agent from Astex Technology, a CAK1 inhibitor from GPC Biotech, a CDK inhibitor from Sanofi-Aventis, a CDK1/CDK2 inhibitor from Amgen, a CDK2 inhibitor from SUGEN-2 (Pfizer), a hearing loss therapy agent (Sound Pharmaceuticals), PD-0332991 (Pfizer), RGB-286199 (GPC Biotech), Ro-0505124 (Hoffmann-La Roche), a Ser/Thr kinase inhibitor from Lilly (EIi Lilly), CVT-2584 (CAS No. 199986-75- 9) (CV Therapeutics), and an analogue or derivative thereof.
16057. The method of item 16031 wherein the agent is a cyclooxygenase 1 inhibitor.
16058. The method of item 16031 wherein the agent is a DHFR inhibitor.
16059. The method of item 16031 wherein the agent is a dual integrin inhibitor.
16060. The method of item 16031 wherein the agent is an elastase inhibitor.
16061. The method of item 16031 wherein the agent is an elongation factor-1 alpha inhibitor.
16062. The method of item 16031 wherein the agent is an endothelial growth factor antagonist. 16063. The method of item 16031 wherein the agent is an endothelial growth factor receptor kinase inhibitor selected from the group consisting of sorafenib tosylate (Bayer), AAL-993 (Novartis), ABP-309 (Novartis), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), E- 7080 (Eisai), EG-3306 (Ark Therapeutics), EXEL-2880 (Exelixis), GW- 654652 (GlaxoSmithKline), a KDR inhibitor from LG Life Sciences, CT-6685 and CT-6729 (UCB), KRN-633 and KRN-951 (Kirin Brewery), OSI-930 (OSI Pharmaceuticals), SP-5.2 (Supratek Pharma), SU-11657 (Pfizer), a Tie-2 antagonist (Hybrigenics), a VEGFR-2 kinase inhibitor (Bristol-Myers Squibb), XL-647 (Exelixis), a KDR inhibitor from Abbott Laboratories, sorafenib tosylate, and an analogue or derivative thereof.
16064. The method of item 16031 wherein the agent is an endotoxin antagonist.
16065. The method of item 16031 wherein the agent is an epothilone and tubulin binder.
16066. The method of item 16031 wherein the agent is an estrogen receptor antagonist.
16067. The method of item 16031 wherein the agent is an FGF inhibitor.
16068. The method of item 16031 wherein the agent is a famexyl transferase inhibitor.
16069. The method of item 16031 wherein the agent is farnesyltransferase inhibitor selected from the group of A-197574 (Abbott), a farnesyltransferase inhibitor from Servier, FPTIII (Strathclyde Institute for Drug R), LB-42908 (LG Life Sciences), Pharmaprojects No. 5063 (Genzyme), Pharmaprojects No. 5597 (Ipsen), Yissum Project No. B-1055 (Yissum), and an analogue or derivative thereof.
16070. The method of item 16031 wherein the agent is an FLT-3 kinase inhibitor.
16071. The method of item 16031 wherein the agent is an FGF receptor kinase inhibitor.
16072. The method of item 16031 wherein the agent is a fibrinogen antagonist selected from the group consisting of AUV-201 (Auvation), MG-13926 (Sanofi-Aventis), plasminogen activator (CAS No. 105913-11-9) (from Sanofi-Aventis or UCB), plasminogen activator-2 (tPA- 2) (Sanofi-Aventis), pro-urokinase (CAS No. 82657-92-9) (Sanofi-Aventis), and an analogue or derivative thereof.
16073. The method of item 16031 wherein the agent is a heat shock protein 90 antagonist selected from the group consisting of SRN- 005 (Sirenade), geldanamycin, NSC-33050 (17-allylaminogeldanamycin; 17- AAG), 17-dimethylaminoethylamino-17-demethoxy-geldanamycin (17- DMAG), rifabutin (rifamycin XIV, 1',4-didehydro-1-deoxy-1 ,4-dihydro-5'-(2- methylpropyl)-1-oxo-), and an analogue or derivative thereof.
16074. The method of item 16031 wherein the agent is a histone deacetylase inhibitor.
16075. The method of item 16031 wherein the agent is an HMGCoA reductase inhibitor selected from the group consisting of an atherosclerosis therapeutic from Lipid Sciences, ATI- 16000 (ARYx Therapeutics), KS-01-019 (Kos Pharmaceuticals), Pharmaprojects No. 2197 (Sanofi-Aventi), RP 61969 (Sanofi-Aventis), and an analogue or derivative thereof.
16076. The method of item 16031 wherein the agent is an ICAM inhibitor.
16077. The method of item 16031 wherein the agent is an IL, ICE and IRAK antagonist, wherein the antagonist is a CJ-14877, CP- 424174 (Pfizer), NF-61 (Negma-Lerads), and an analogue or derivative thereof.
16078. The method of item 16031 wherein the agent is an IL-2 inhibitor.
16079. The method of item 16031 wherein the agent is an immunosuppressant selected from the group consisting of teriflunomide (Sanofi Aventis), chlorsulfaquinoxalone (NSC-339004), chlorsulfaquinoxalone sulfate, CS-712 (Sankyo), ismomultin alfa (CAS No. 457913-93-8) (Akzo Nobel), antiallergics from GenPat77, antiinflammatories or AT-005 (Androclus Therapeutics), autoimmune disease therapeutics from EpiVax, BN-007 (Bone), budesonide (CAS No. 51333-22- 3) (MAP Pharmaceuticals), CO-14 (Genzyme), edratide (CAS No. 433922- 67-9) (Teva), EP-314 (Enanta), eprovafen (CAS No. 101335-99-3) (Sanofi- Aventis), HWA-131 (CAS No. 118788-41-3) (Sanofi-Aventis), immunomodulators from MerLion Pharmaceuticals, immunosuppressives from Alchemia, IPL-12 (Inflazyme), MDL-9563 (CAS No. 27086-86-8) (Sanofi-Aventis), Pharmaprojects No. 2330 (Sanofi-Aventis), Pharmaprojects No. 6426 (Abgenix), PXS-25 (Pharmaxis), rosmarinic acid (CAS No. 20283-92-5) (Sanofi-Aventis), RP 42927 or RP 54745 (CAS No. 135330-08-4) (Sanofi-Aventis), SGN-35 (Seattle Genetics), ST-1959 (Sigma-Tau), type I diabetes therapy from SYNX Pharma, UNIL-88 (Debiopharm), VP-025 (Vasogen), VR-694 (Vectura), PRTX-001 (Protalex), and an analogue or derivative thereof.
16080. The method of item 16031 wherein the agent is an IMPDH (inosine monophosphate).
16081. The method of item 16031 wherein the agent is an integrin antagonist.
16082. The method of item 16031 wherein the agent is an interleukin antagonist.
16083. The method of item 16031 wherein the agent is an inhibitor of type III receptor tyrosine kinase.
16084. The method of item 16031 wherein the agent is an irreversible inhibitor of enzyme methionine aminopeptidase type 2.
16085. The method of item 16031 wherein the agent is an isozyme selective delta protein kinase C inhibitor.
16086. The method of item 16031 wherein the agent a JAK3 enzyme inhibitor.
16087. The method of item 16031 wherein the agent is a JNK inhibitor.
16088. The method of item 16031 wherein the agent is a kinase inhibitor. 16089. The method of item 16031 wherein the agent is kinesin antagonist.
16090. The method of item 16031 wherein the agent is a kinesin antagonist.
16091. The method of item 16031 wherein the agent is a leukotriene inhibitor and antagonist selected from the group consisting of ambicromil (CAS No. 58805-38-2) (Sanofi-Aventis), amelubant (CAS No. 346735-24-8) (Boehringer Ingelheim), DW-1141 (Dong Wha), ebselen (Daiichi Pharmaceutical), ibudilast (Kyorin), leucotriene inhibitors from Sanofi-Aventis, lymphotoxin-beta receptor (LT-D) from Biogen Idee, Pharmaprojects No. 1535 and 2728 (CAS No. 119340-33-9) (Sanofi- Aventis), R-112 (Rigel), Rev-5367 (CAS No. 92532-05-3) (Sanofi-Aventis), RG-14893 (CAS No. 141835-49-6) (Sanofi-Aventis), RG-5901-A (CAS No. 101910-24-1), 92532-23-5, RP 66153 (CAS No. 142422-79-5), RP 66364 (CAS No.186916-92-5), RP 69698 (CAS No. 141748-00-7) (Sanofi-Aventis), SC-411930 (Pfizer), SC-41930 (CAS No. 120072-59-5) (Pfizer), SC-50605 (CAS No. 138828-39-4) (Pfizer), SC-51146 (CAS No. 141059-52-1), SC- 53228 (CAS No. 153633-01-3) (Pfizer), spaglumic acid (ZY-15106) (CAS No.3106-85-2), 80619-64-3 (Novartis), tipredane (CAS No. 85197-77-9) (Bristol-Myers Squibb), U-75302 (CAS No. 119477-85-9) (Pfizer), and analogue or derivative thereof.
16092. The method of item 16031 wherein the agent is an MAP kinase inhibitor.
16093. The method of item 16031 wherein the agent is a matrix metalloproteinase inhibitor. 16094. The method of item 16031 wherein the agent is an MCP-CCR2 inhibitor.
16095. The method of item 16031 wherein the agent is an mTOR inhibitor.
16096. The method of item 16031 wherein the agent is an mTOR kinase inhibitor.
16097. The method of item 16031 wherein the agent is a microtubule inhibitor selected from the group consisting of antibody- maytansinoid conjugates from Biogen Idee, colchicines (MantiCore Pharmaceuticals), anticancer immunoconjugates from Johnson & Johnson, DIME from Octamer, gni-1f (GNI), huC242-DM4, huMy9-6-DM1 (ImmunoGen), IDN-5404 (Indena), IMO-098, IMOderm (Imotep), mebendazole (Introgen Therapeutics), microtubule poisons from Cambridge Enterprise, paclitaxel such as LOTAX from Aphios (CAS No. 33069-62-4), Genexol-PM from Samyang, Pharmaprojects No. 6383 (Tapestry Pharmaceuticals), RPR-112378 (Sanofi-Aventis), SGN-75 (Seattle Genetics), SPL-7435 (Starpharma), SSR-250411 (Sanofi-Aventis), trastuzumab-DM1 (Genentech), vinorelbine, and an analogue or derivative thereof.
16098. The method of item 16031 wherein the agent is an MIF inhibitor.
16099. The method of item 16031 wherein the agent is an MMP inhibitor.
16100. The method of item 16031 wherein the agent is a neurokinin (NK) antagonist selected from the group consisting of anthrotainin (CAS No. 148084-40-6) (Sanofi-Aventis), an IBS thereapeutic from ArQuIe, MDL-105212A (CAS No. 167261-60-1) (Ssanofi-Aventis), Pharmaprojects No. 2744, 3258 (CAS No. 139167-47-8) 4006, 4201 , or 5986 (Sanofi-Aventis), RP 67580 (CAS No. 135911-02-3), SR-144190 (CAS No. 201152-86-5), SSR-240600, SSR-241586 (Sanofi-Aventis), TKA-457 (Novartis), vestipitant mesylate (CAS No. 334476-64-1) (GlaxoSmithKline), Win-64821 (Sanofi-Aventis), PRX-96026 (Predix Pharmaceuticals), and an analogue or derivative thereof.
16101. The method of item 16031 wherein the agent is an NF kappa B inhibitor selected from the group consisting of AVE-0545 or AVE-0547 (Sanofi-Aventis), bortezomib (CAS No. 179324-69-7) (Millennium Pharmaceuticals), dexanabinol (CAS No. 112924-45-5) (Pharmos), dexlipotam (Viatris), Pharmaprojects No. 6283 (INDRA) (OXiGENE), IPL- 576092 (CAS No. 137571-30-3) (Inflazyme), NFKB decoy (Corgentech), NFKB decoy oligo (AnGes MG), NFKB's from Ariad, osteoporosis treatments or S5 (F005) from Fulcrum Pharmaceuticals, P61 (Phytopharm), R-flurbiprofen (CAS No. 5104-49-4) (Encore Pharmaceuticals), and an analogue or derivative thereof.
16102. The method of item 16031 wherein the agent is a nitric oxide agonist.
16103. The method of item 16031 wherein the agent is an ornithine decarboxylase inhibitor.
16104. The method of item 16031 wherein the agent is a p38 MAP kinase inhibitor selected from the group consisting of AZD-6703 (AstraZeneca), JX-401 (Jexys Pharmaceuticals), BMS-2 (Bristol-Myers Squibb), a p38 MAP kinase inhibitor from Novartis, a p38-alpha MAP kinase inhibitor from Amphora, Pharmaprojects No. 5704 (Pharmacopeia), RPR- 200765A (Sanofi-Aventis), SD-282 (Johnson & Johnson), TAK-715 (Takeda), and an analogue or derivative thereof.
16105. The method of item 16031 wherein the agent is a palmitoyl-protein thioesterase inhibitor.
16106. The method of item 16031 wherein the agent is a PDGF receptor kinase inhibitor selected from the group consisting of AAL- 993, AMN-107, or ABP-309 (Novartis), AMG-706 (Amgen), BAY-57-9352 (Bayer), CDP-860 (UCB), E-7080 (Eisai), imatinib (CAS No. 152459-95-5) (Novartis), OSI-930 (OSI Pharmaceuticals), RPR-127963E (Sanofi-Aventis), RWJ-540973 (Johnson & Johnson), sorafenib tosylate (Bayer), SU-11657 (Pfizer), tandutinib (CAS No. 387867-13-2) (Millennium Pharmaceuticals), vatalanib (Novartis), ZK-CDK (Schering AG), and an analogue or derivative thereof.
16107. The method of item 16031 wherein the agent is (-)-halofenate (Metabolex), AMG-131 (Amgen), antidiabetics from Japan Tobacco, AZD-4619, AZD-8450, AZD-8677 (AstraZeneca), DRF-10945, balaglitazone (Dr Reddy's), CS-00088, CS-00098 (Chipscreen Biosciences), E-3030 (Eisai), etalocib (CAS No. 161172-51-6) (EIi Lilly), GSK-641597 (Ligand), GSK-677954 (GlaxoSmithKline), GW-409544 (Ligand), GW- 590735 (GlaxoSmithKline), K-111 (Hoffmann-La Roche), LY-518674 (EIi Lilly), LY-674 (Ligand), LY-929 (Ligand), MC-3001, MC-3002 (MaxoCore Pharmaceuticals), metformin HCI + pioglitazone (CAS No. 1115-70-4 and
112529-15-4), ACTOPLUS MET from Andrx), muraglitazar (CAS No. 331741-94-7) (Bristol-Myers Squibb), naveglitazar (Ligand), oleoylethanolamide (Kadmus Pharmaceuticals), ONO-5129, pioglitazone hydrochloride (CAS No. 111025-46-8 and 112529-15-4) (Takeda), PLX-204 (Plexxikon), PPAR agonists from Genfit, PPAR delta agonists from EIi Lilly, PPAR-alpha agonists from CrystalGenomics, PPAR-gamma modulators and PPAR-β modulators from CareX, rosiglitazone maleate (CAS No. 122324- 73-4 or 155141-29-0) (GlaxoSmithKline), rosiglitazone maleate/glimepir (CAS No. 155141-29-0 and 93479-97-1), AVANDARYL1 rosiglitazone maleate/metformin extend (CAS No. 155141-29-0 and 657-24-9), AVANDAMET, rosiglitazone maleate+metformin, AVANDAMET (GlaxoSmithKline), tesaglitazar (AstraZeneca), and an analogue or derivative thereof.
16108. The method of item 16031 wherein the agent is a phosphatase inhibitor.
16109. The method of item 16031 wherein the agent is a phosphodiesterase (PDE) inhibitor selected from the group consisting of avanafil (Tanabe Seiyaku), dasantafil (CAS No. 569351-91-3) (Schering- Plough), A-906119 (CAS No. 134072-58-5), DL-850 (Sanofi-Aventis), GRC- 3015, GRC-3566, GRC-3886 (Glenmark), HWA-153 (CAS No. 56395-66-5) (Sanofi-Aventis), hydroxypumafentrine (Altana), IBFB-130011 , IBFB-14-016, IBFB-140301 , IBFB-150007, IBFB-211913 (IBFB Pharma), L-826141 (Merck & Co), medorinone (CAS No. 88296-61-1) (Sanofi-Aventis), MEM- 1917 (Memory Pharmaceuticals), ND-1251 (Neuro3d), PDE inhibitors from ICOS, PDE IV inhibitors from Memory Pharmaceuticals and CrystalGenomics, Pharmaprojects No. 2742 and 6141 (Sanofi-Aventis), QAD-171 (Novartis), RHC-2963 (CAS No. 76993-12-9 and 76993-14-1), RPR-117658, RPR-122818 derivatives, SR-24870 , and RPR-132294 (Sanofi-Aventis), SK-350 (ln2Gen), stroke therapy agents from deCODE Genetics, TAS-203 (Taiho), tofimilast (CAS No. 185954-27-2) (Pfizer), UK- 371800 (Pfizer), WIN-65579 (CAS No. 158020-82-7) (Sanofi-Aventis), IBFB- 130020 (IBFB Pharma), OPC-6535 (CAS No. 145739-56-6) (Otsuka), a phosphodiesterase III inhibitor, enoximone, a phosphodiesterase IV inhibitor, fosfosal, Atopik (Barrier Therapeutics), triflusal, a phosphodiesterase V inhibitor, and an analogue or derivative thereof. 16110. The method of item 16031 wherein the agent is a PKC inhibitor.
16111. The method of item 16031 wherein the agent is a platelet activating factor antagonist.
16112. The method of item 16031 wherein the agent is a platelet-derived growth factor receptor kinase inhibitor.
16113. The method of item 16031 wherein the agent is a prolyl hydroxylase inhibitor.
16114. The method of item 16031 wherein the agent is a polymorphonuclear neutrophil inhibitor.
16115. The method of item 16031 wherein the agent is a protein kinase B inhibitor.
16116. The method of item 16031 wherein the agent is a protein kinase C stimulant.
16117. The method of item 16031 wherein the agent is a purine nucleoside analogue.
16118. The method of item 16031 wherein the agent is a purinoreceptor P2X antagonist.
16119. The method of item 16031 wherein the agent is a Raf kinase inhibitor. 16120. The method of item 16031 wherein the agent is a reversible inhibitor of ErbB1 and ErbB2.
16121. The method of item 16031 wherein the agent is a ribonucleoside triphosphate reductase inhibitor.
16122. The method of item 16031 wherein the agent is an SDF-1 antagonist.
16123. The method of item 16031 wherein the agent is a sheddase inhibitor.
16124. The method of item 16031 wherein the agent is an SRC inhibitor.
16125. The method of item 16031 wherein the agent is.a stromelysin inhibitor.
16126. The method of item 16031 wherein the agent is an Syk kinase inhibitor.
16127. The method of item 16031 wherein the agent is a telomerase inhibitor.
16128. The method of item 16031 wherein the agent is a TGF beta inhibitor selected from the group consisting of pirfenidone (CAS No. 53179-13-8) (MARNAC), tranilast (CAS No. 53902-12-8) (Kissei), IN-
1130 (ln2Gen), mannose-6-phosphate (BTG), TGF-β antagonists from Inflazyme (Pharmaprojects No. 6075), TGF-β antagonists from Sydney, non-industrial source), TGF-β I receptor kinase inhibitors from EIi Lilly, TGF- β receptor inhibitors from Johnson & Johnson, and an analogue or derivative thereof.
16129. The method of item 16031 wherein the agent is a
TNFα antagonist or TACE inhibitor selected from the group consisting of adalimumab (CAS No. 331731-18-1) (Cambridge Antibody Technology), AGIX-4207 (AtheroGenics), AGT-1 (Advanced Biotherapy), an antiinflammatory from Borean Pharma, Cellzome, or Paradigm Therapeutics, anti-inflammatory vaccine (TNF-alpha kinoid) from Neovacs, humanized anti-TNF antibody or an anti-TNF MAb (CB0006) Celltech (UCB), apratastat (CAS No. 287445-51-0) (Wyeth), BMS-561392 (Bristol-Myers Squibb), BN- 006 (Bone), certolizumab pegol (CAS No. 428863-50-7 or CH-138 (UCB), cilomilast (CAS No. 153259-65-5) (GlaxoSmithKline), CR-1 (Nuada Pharmaceuticals), CRx-119 (CombinatoRx), D-5410 (UCB), dacopafant (CAS No. 125372-33-0) (Sanofi-Aventis), dersalazine (CAS No. 188913-57- 7/188913-58-8) (Uriach), etanercept (CAS No. 185243-69-0) (Amgen), ethyl pyruvate (Critical (Critical Therapeutics), golimumab (CAS No. 476181-74-5) (Johnson & Johnson), hormono-immunotherapy from Ipsen, CDP571 (e.g., Humicade from UCB), IC-485 (ICOS), infliximab (CAS No. 170277-31-3) (Johnson & Johnson), IP-751 (Manhattan Pharmaceuticals), ISIS-104838 (CAS No. 250755-32-9) (ISIS Pharmaceuticals), lenalidomide (CAS No. 191732-72-6) (Celgene), lentinan (CAS No. 37339-90-5) (Ajinomoto), MDL- 201112 (CAS No. 142130-73-2) (Sanofi-Aventis), medroxyprogesterone (CAS No. 520-85-4) (InKine Pharmaceutical), N-acetylcysteine (CAS No. 616-91-1) (Zambon), NBE-P2 (DIREVO Biotech), nerelimomab (CAS No. 162774-06-3) (Chiron), OM-294DP (OM PHARMA), onercept (CAS No. 199685-57-9) (Yeda), PASSTNF-alpha (Verigen), pentoxifylline or oxypentifylline (Sanofi-Aventis), Pharmaprojects No. 4091 , 4241, 4295, or 4488 (Sanofi-Aventis), Pharmaprojects No. 5480 (Amgen), Pharmaprojects No. 6749 (Cengent), pirfenidone (CAS No. 53179-13-8) (MARNAC), RPR- 132294 (Sanofi-Aventis), S5 (F002) (Fulcrum Pharmaceuticals), simvastatin (CAS No. 79902-63-9) (Merck & Co), STA-6292 (Synta Pharmaceuticals), tacrolimus (CAS No. 104987-11-3) (Fujisawa LifeCycle Pharma), talactoferrin alfa (CAS No. 308240-58-6) (Agennix), thalidomide (CAS No. 50-35-1) (Celgene), TNF antagonists form ProStrakan, and Synergen, TNF inhibitors (Amgen), TNF-alpha antagonists from Dynavax Technologies and Jerina AG (Germany), TNF-alpha inhibitors from IBFB Pharma and Xencor (Xencor), torbafylline (CAS No. 105102-21-4) (Sanofi-Aventis), UR-1505 (Uriach), VT-346 (Viron Therapeutics), YSIL6 (Y's Therapeutics), YSTH2 (Y's Therapeutics), NPI-1302a-3 (Nereus Pharmaceuticals, a TNF antagonist from Jerina AG (Germany), dersalazine, and an analogue or derivative thereof.
16130. The method of item 16031 wherein the agent is a Toll receptor inhibitor.
16131. The method of item 16031 wherein the agent is a tubulin antagonist.
16132. The method of item 16031 wherein the agent is a tyrosine kinase inhibitor selected from the group consisting of SU-011248, SUTENT from Pfizer Inc. (New York, NY), BMS-354825, PN-355 (Paracelsian Pharmaceuticals), AGN-199659 (Allergan), AAL-993 or ABP- 309 (Novartis), adaphostin (NIH), AEE-788 (Novartis), AG-013736 (OSI Pharmaceuticals), AG-13736 (Pfizer), ALT-110 (Alteris Therapeutics), AMG- 706 (Amgen), anticancer MAbs from Xencor, anti-EGFrvlll MAbs from Abgenix, anti-HER2 MAb from Abiogen, AZD-2171 or AZD-9935 (AstraZeneca), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), CEP-5214 (Cephalon), CEP-7055 (Cephalon), cetuximab (ImClone Systems), CHIR-200131 and CHIR-258 (Chiron), CP-547632 (OSI Pharmaceuticals), CP-724714 (Pfizer), CT-301 (Creabilis Therapeutics), D- 69491 (Baxter International), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EGFR/ErbB2 inhibitors from Array BioPharma, erlotinib (CAS No. 183319- 69-9) (OSI Pharmaceuticals), EXEL-2880 (Exelixis), FK-778 (Sanofi- Aventis) , gefitinib (CAS No. 184475-35-2) (AstraZeneca), GW-2286 or GW- 654652 (GlaxoSmithKline), her2/neu antigen from AlphaVax, HER-2/neu inhibitor from Generex, Herzyme (Medipad) (Sirna Therapeutics), HKI-272 (Wyeth), HuMax-EGFr (Genmab), idronoxil (CAS No. 81267-65-4) (Novogen), IGF-1 inhibitors from Ontogen, IMC-11F8 (ImCIone Systems), kahalalide F (CAS No. 149204-42-2) (PharmaMar), KDR inhibitor from LG Life Sciences, KDR inhibitors from Abbott Laboratories, KDR kinase inhibitors (UCB), Kdr kinase inhibitors from Merck & Co1 KRN-633 and KRN- 951 (Kirin Brewery), KSB-102 (Xenova), lapatinib ditosylate (CAS No. 388082-78-8) (GlaxoSmithKline), matuzumab (Merck KGaA), MDX-214 (Medarex), ME-103 (Pharmexa), MED-A300 (Gerolymatos), MNAC-13 (Lay Line Genomics), nimotuzumab (Center of Molecular Immunology), NSC- 330507 or NSC-707545 (NIH), NV-50 (Novogen), OSI-930 (OSI Pharmaceuticals), panitumumab (Abgenix), pelitinib (CAS. No. 287933-82-7) (Wyeth), pertuzumab (CAS No. 380610-27-5) (Genentech), Pharmaprojects No. 3985 (Sanofi-Aventis), prostate cancer therapeutics from Sequenom (SQPC35, SQPC36, SQPC90), removab and remoxab (Trion Pharma), RG- 13022 (CAS No. 136831-48-6), RG-13291 (CAS No. 138989-50-1), or RG- 14620 (CAS No. 136831-49-7) (Sanofi-Aventis), RM-6427 (Romark), RNAi breast cancer therapy from Benitec, RP 53801 (CAS No. 125882-88-4) (Sanofi-Aventis), sorafenib tosylate (Bayer), SU-11657 (Pfizer), Tie-2 antagonists from Semaia (Hybrigenics), Tie-2 inhibitors from Ontogen, trastuzumab (CAS No. 180288-69-1) (Genentech), tyrosine kinase inhibitors from Sanofi-Aventis, U3-1287, U3-1565, U3-1784, U3-1800 (U3 Pharma), vatalanib (Novartis), VEGFR-2 kinase inhibitor from Bristol-Myers Squibb, XL-647 (Exelixis), ZD-6474 (AstraZeneca), ZK-CDK (Schering AG), an EGFR tyrosine kinase inhibitor, EKB-569 (Wyeth), and an analogue or derivative thereof. 16133. The method of item 16031 wherein the agent is a VEGF inhibitor.
16134. The method of item 16031 wherein the agent is a vitamin D receptor agonist.
16135. The method of item 16031 wherein the agent is ZD-6474 (an angiogenesis inhibitor).
16136. The method of item 16031 wherein the agent is AP-23573 (an mTOR inhibitor).
16137. The method of item 16031 wherein the agent is synthadotin (a tubulin antagonist).
16138. The method of item 16031 wherein the agentis S-0885 (a collagenase inhibitor).
16139. The method of item 16031 wherein the agent is aplidine (an elongation factor-1 alpha inhibitor).
16140. The method of item 16031 wherein the agent is ixabepilone (an epithilone).
16141. The method of item 16031 wherein the agent is IDN-5390 (an angiogenesis inhibitor).
16142. The method of item 16031 wherein the agent is SB-2723005 (an angiogenesis inhibitor). 16143. The method of item 16031 wherein the agent is ABT-518 (an angiogenesis inhibitor).
16144. The method of item 16031 wherein the agent is combretastatin (an angiogenesis inhibitor).
16145. The method of item 16031 wherein the agent is anecortave acetate (an angiogenesis inhibitor).
16146. The method of item 16031 wherein the agent is SB-715992 (a kinesin antagonist).
16147. The method of item 16031 wherein the agent is temsirolimus (an mTOR inhibitor).
16148. The method of item 16031 wherein the agent is adalimumab (a TNFα antagonist).
16149. The method of item 16031 , wherein the composition comprises a polymer.
16150. The method of item 16031, wherein the composition comprises a polymeric carrier.
16151. The method of item 16031 wherein the anti- scarring agent inhibits adhesion between the device and a host into which the device is implanted.
16152. The method of item 16031 wherein the device delivers the anti-scarring agent locally to tissue proximate to the device. 16153. The method of item 16031 wherein the device has a coating that comprises the anti-scarring agent.
16154. The method of item 16031 , wherein the device has a coating that comprises the agent and is disposed on a surface of the implant.
16155. The method of item 16031 , wherein the device has a coating that comprises the agent and directly contacts the implant.
16156. The method of item 16031 , wherein the device has a coating that comprises the agent and indirectly contacts the implant.
16157. The method of item 16031 , wherein the device has a coating that comprises the agent and partially covers the implant.
16158. The method of item 16031 , wherein the device has a coating that comprises the agent and completely covers the implant.
16159. The method of item 16031 , wherein the device has a uniform coating.
16160. The method of item 16031 , wherein the device has a non-uniform coating.
16161. The method of item 16031, wherein the device has a discontinuous coating.
16162. The method of item 16031, wherein the device has a patterned coating. 16163. The method of item 16031 , wherein the device has a coating with a thickness of 100 Dm or less.
16164. The method of item 16031 , wherein the device has a coating with a thickness of 10 Gm or less.
16165. The method of item 16031 , wherein the device has a coating, and the coating adheres to the surface of the implant upon deployment of the implant.
16166. The method of item 16031 , wherein the device has a coating, and wherein the coating is stable at room temperature for a period of 1 year.
16167. The method of item 16031, wherein the device has a coating, and wherein the anti-scarring agent is present in the coating in an amount ranging between about 0.0001% to about 1% by weight.
16168. The method of item 16031, wherein the device has a coating, and wherein the anti-scarring agent is present in the coating in an amount ranging between about 1 % to about 10% by weight.
16169. The method of item 16031 , wherein the device has a coating, and wherein the anti-scarring agent is present in the coating in an amount ranging between about 10% to about 25% by weight.
16170. The method of item 16031 , wherein the device has a coating, and wherein the anti-scarring agent is present in the coating in an amount ranging between about 25% to about 70% by weight. 16171. The method of item 16031 , wherein the device has a coating, and wherein the coating further comprises a polymer.
16172. The method of item 16031 , wherein the device has a first coating having a first composition and a second coating having a second composition.
16173. The method of item 16031 , wherein the device has a first coating having a first composition and a second coating having a second composition, wherein the first composition and the second composition are different.
16174. The method of item 16031 , wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a copolymer.
16175. The method of item 16031 , wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a block copolymer.
16176. The method of item 16031 , wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a random copolymer.
16177. The method of item 16031 , wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a biodegradable polymer.
16178. The method of item 16031 , wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a non-biodegradable polymer. 16179. The method of item 16031 , wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a hydrophilic polymer.
16180. The method of item 16031, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a hydrophobic polymer.
16181. The method of item 16031, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a polymer having hydrophilic domains.
16182. The method of item 16031, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a polymer having hydrophobic domains.
16183. The method of item 16031 , wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a non-conductive polymer.
16184. The method of item 16031, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises an elastomer.
16185. The method of item 16031 , wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a hydrogel.
16186. The method of item 16031, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a silicone polymer. 16187. The method of item 16031 , wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a hydrocarbon polymer.
16188. The method of item 16031 , wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a styrene-derived polymer.
16189. The method of item 16031 , wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a butadiene polymer.
16190. The method of item 16031 , wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a macromer.
16191. The method of item 16031, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a poly(ethylene glycol) polymer.
16192. The method of item 16031 wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises an amorphous polymer.
16193. The method of item 16031 , wherein the device comprises a lubricious coating.
16194. The method of item 16031 wherein the anti- scarring agent is located within pores or holes of the device. 16195. The method of item 16031 wherein the anti- scarring agent is located within a channel, lumen, or divet of the device.
16196. The method of item 16031 , wherein the device comprises a second pharmaceutically active agent.
16197. The method of item 16031 wherein the device comprises an anti-inflammatory agent.
16198. The method of item 16031 wherein the device comprises an agent that inhibits infection.
16199. The method of item 16031 wherein the device comprises an agent that inhibits infection, and wherein the agent is an anthracycline.
16200. The method of item 16031 wherein the device comprises an agent that inhibits infection, and wherein the agent is doxorubicin.
16201. The method of item 16031 wherein the device comprises an agent that inhibits infection, and wherein the agent is mitoxantrone.
16202. The method of item 16031 wherein the device comprises an agent that inhibits infection, and wherein the agent is a fluoropyrimidine.
16203. The method of item 16031 wherein the device comprises an agent that inhibits infection, and wherein the agent is 5- fluorouracil (5-FU). 16204. The method of item 16031 wherein the device comprises an agent that inhibits infection, and wherein the agent is a folic acid antagonist.
16205. The method of item 16031 wherein the device comprises an agent that inhibits infection, and wherein the agent is methotrexate.
16206. The method of item 16031 wherein the device comprises an agent that inhibits infection, and wherein the agent is a podophylotoxin.
16207. The method of item 16031 wherein the device comprises an agent that inhibits infection, and wherein the agent is etoposide.
16208. The method of item 16031 wherein the device comprises an agent that inhibits infection, and wherein the agent is a camptothecin.
16209. The method of item 16031 wherein the device comprises an agent that inhibits infection, and wherein the agent is a hydroxyurea.
16210. The method of item 16031 wherein the device comprises an agent that inhibits infection, and wherein the agent is a platinum complex.
16211. The method of item 16031 wherein the device comprises an agent that inhibits infection, and wherein the agent is cisplatin. 16212. The method of item 16031 , further comprising an anti-thrombotic agent.
16213. The method of item 16031 wherein the device comprises a visualization agent.
16214. The method of item 16031 wherein the device comprises a visualization agent, wherein the visualization agent is a radiopaque material, and wherein the radiopaque material comprises a metal, a halogenated compound, or a barium containing compound.
16215. The method of item 16031 wherein the device comprises a visualization agent, wherein the visualization agent is a radiopaque material, and wherein the radiopaque material comprises barium, tantalum, or technetium.
16216. The method of item 16031 wherein the device comprises a visualization agent, and wherein the visualization agent is a MRI responsive material.
16217. The method of item 16031 wherein the device comprises a visualization agent, and wherein the visualization agent comprises a gadolinium chelate.
16218. The method of item 16031 wherein the device comprises a visualization agent, and wherein the visualization agent comprises iron, magnesium, manganese, copper, or chromium.
16219. The method of item 16031 wherein the device comprises a visualization agent, and wherein the visualization agent comprises an iron oxide compound. 16220. The method of item 16031 wherein the device comprises a visualization agent, and wherein the visualization agent comprises a dye, pigment, or colorant.
16221. The method of item 16031 wherein the device comprises an echogenic material.
16222. The method of item 16031 wherein the device comprises an echogenic material, and wherein the echogenic material is in the form of a coating.
16223. The method of item 16031 wherein the device is sterile.
16224. The method of item 16031 wherein the anti- scarring agent is released into tissue in the vicinity of the device after deployment of the device.
16225. The method of item 16031 wherein the anti- scarring agent is released into tissue in the vicinity of the device after deployment of the device, and wherein the tissue is connective tissue.
16226. The method of item 16031 wherein the anti- scarring agent is released into tissue in the vicinity of the device after deployment of the device, and wherein the tissue is muscle tissue.
16227. The method of item 16031 wherein the anti- scarring agent is released into tissue in the vicinity of the device after deployment of the device, and wherein the tissue is nerve tissue. 16228. The method of item 16031 wherein the anti- scarring agent is released into tissue in the vicinity of the device after deployment of the device, and wherein the tissue is epithelium tissue.
16229. The method of item 16031 wherein the anti- scarring agent is released in effective concentrations from the device over a period ranging from the time of deployment of the device to about 1 year.
16230. The method of item 16031 wherein the anti- scarring agent is released in effective concentrations from the device over a period ranging from about 1 month to 6 months.
16231. The method of item 16031 wherein the anti- scarring agent is released in effective concentrations from the device over a period ranging from about 1 - 90 days.
16232. The method of item 16031 wherein the anti- scarring agent is released in effective concentrations from the device at a constant rate.
16233. The method of item 16031 wherein the anti- scarring agent is released in effective concentrations from the device at an increasing rate.
16234. The method of item 16031 wherein the anti- scarring agent is released in effective concentrations from the device at a decreasing rate.
16235. The method of item 16031 wherein the anti- scarring agent is released in effective concentrations from the composition comprising the anti-scarring agent by diffusion over a period ranging from the time of deployment of the device to about 90 days.
16236. The method of item 16031 wherein the anti- scarring agent is released in effective concentrations from the composition comprising the anti-scarring agent by erosion of the composition over a period ranging from the time of deployment of the device to about 90 days.
16237. The method of item 16031 wherein the device comprises about 0.01 Dg to about 10 Gg of the anti-scarring agent.
16238. The method of item 16031 wherein the device comprises about 10 Dg to about 10 mg of the anti-scarring agent.
16239. The method of item 16031 wherein the device comprises about 10 mg to about 250 mg of the anti-scarring agent.
16240. The method of item 16031 wherein the device comprises about 250 mg to about 1000 mg of the anti-scarring agent.
16241. The method of item 16031 wherein the device comprises about 1000 mg to about 2500 mg of the anti-scarring agent.
16242. The method of item 16031 wherein a surface of the device comprises less than 0.01 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
16243. The method of item 16031 wherein a surface of the device comprises about 0.01 Dg to about 1 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied. 16244. The method of item 16031 wherein a surface of the device comprises about 1 Dg to about 10 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
16245. The method of item 16031 wherein a surface of the device comprises about 10 Dg to about 250 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
16246. The method of item 16031 wherein a surface of the device comprises about 250 Dg to about 1000 Dg of the anti-scarring agent of anti-scarring agent per mm2 of device surface to which the anti- scarring agent is applied.
16247. The method of item 16031 wherein a surface of the device comprises about 1000 Dg to about 2500 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
16248. The method of item 16031 wherein the combining is performed by direct affixing the agent or the composition to the implant.
16249. The method of item 16031 wherein the combining is performed by spraying the agent or the component onto the implant.
16250. The method of item 16031 wherein the combining is performed by electrospraying the agent or the composition onto the implant. 16251. The method of item 16031 wherein the combining is performed by dipping the implant into a solution comprising the agent or the composition.
16252. The method of item 16031 wherein the combining is performed by covalently attaching the agent or the composition to the implant.
16253. The method of item 16031 wherein the combining is performed by non-covalently attaching the agent or the composition to the implant.
16254. The method of item 16031 wherein the combining is performed by coating the implant with a substance that contains the agent or the composition.
16255. The method of item 16031 wherein the combining is performed by coating the implant with a substance that absorbs the agent.
16256. The method of item 16031 wherein the combining is performed by interweaving a thread composed of, or coated with, the agent or the composition.
16257. The method of item 16031 wherein the combining is performed by covering all the implant with a sleeve that contains the agent or the composition.
16258. The method of item 16031 wherein the combining is performed by covering a portion of the implant with a sleeve that contains the agent or the composition. 16259. The method of item 16031 wherein the combining is performed by covering all the implant with a cover that contains the agent or the composition.
16260. The method of item 16031 wherein the combining is performed by covering a portion of the implant with a cover that contains the agent or the composition.
16261. The method of item 16031 wherein the combining is performed by covering all the implant with an electrospun fabric that contains the agent or the composition.
16262. The method of item 16031 wherein the combining is performed by covering a portion of the implant with an electrospun fabric that contains the agent or the composition.
16263. The method of item 16031 wherein the combining is performed by covering all the implant with a mesh that contains the agent or the composition.
16264. The method of item 16031 wherein the combining is performed by covering a portion of the implant with a mesh that contains the agent or the composition.
16265. The method of item 16031 wherein the combining is performed by constructing all the implant with the agent or the composition.
16266. The method of item 16031 wherein the combining is performed by constructing a portion of the implant with the agent or the composition. 16267. The method of item 16031 wherein the combining is performed by impregnating the implant with the agent or the composition.
16268. The method of item 16031 wherein the combining is performed by constructing all of the implant from a degradable polymer that releases the agent.
16269. The method of item 16031 wherein the combining is performed by constructing a portion of the implant from a degradable polymer that releases the agent.
16270. The method of item 16031 wherein the combining is performed by dipping the implant into a solution that comprise the agent and an inert solvent for the implant.
16271. The method of item 16031 wherein the combining is performed by dipping the implant into a solution that comprises the agent and a solvent that will swell the implant.
16272. The method of item 16031 wherein the combining is performed by dipping the implant into a solution that comprises the agent and a solvent that will dissolve the implant.
16273. The method of item 16031 wherein the combining is performed by dipping the implant into a solution that comprises the agent, a polymer and an inert solvent for the implant.
16274. The method of item 16031 wherein the combining is performed by dipping the implant into a solution that comprises the agent, a polymer and a solvent that will swell the implant. 16275. The method of item 16031 wherein the combining is performed by dipping the implant into a solution that comprises the agent, a polymer and a solvent that will dissolve the implant.
16276. The method of item 16031 wherein the combining is performed by spraying the implant into a solution that comprises the agent and an inert solvent for the implant.
16277. The method of item 16031 wherein the combining is performed by spraying the implant into a solution that comprises the agent and a solvent that will swell the implant.
16278. The method of item 16031 wherein the combining is performed by spraying the implant into a solution that comprises the agent and a solvent that will dissolve the implant.
16279. The method of item 16031 wherein the combining is performed by spraying the implant into a solution that comprises the agent, a polymer and an inert solvent for the implant.
16280. The method of item 16031 wherein the combining is performed by spraying the implant into a solution that comprises the agent, a polymer and a solvent that will swell the implant.
16281. The method of item 16031 wherein the combining is performed by spraying the implant into a solution that comprises the agent, a polymer and a solvent that will dissolve the implant.
16282. A method as in any one of items 16031-16281 , wherein the device is a drainage tube. 16283. A method as in any one of items 16031-16281 , wherein the device is a feeding tube.
16284. A method as in any one of items 16031-16281, wherein the device is a portosystemic shunt.
16285. A method as in any one of items 16031-16281 , wherein the device is a shunt for ascite.
16286. A method as in any one of items 16031-16281 , wherein the device is a nasogastric or nasoenteral tube.
16287. A method as in any one of items 16031-16281 , wherein the device is a gastrostomy or percutaneous feeding tube.
16288. A method as in any one of items 16031-16281 , wherein the device is a jejunostomy endoscopic tube.
16289. A method as in any one of items 16031-16281 , wherein the device is a colostomy device.
16290. A method as in any one of items 16031-16281 , wherein the device is a biliary T-tube.
16291. A method as in any one of items 16031-16281 , wherein the device is a biopsy forceps.
16292. A method as in any one of items 16031-16281 , wherein the device is a biliary stone removal device. 16293. A method as in any one of items 16031-16281 , wherein the device is an endoscopic retrograde cholangiopancretography device.
16294. A method as in any one of items 16031-16281 , wherein the device is a dilation balloon.
16295. A method as in any one of items 16031 -16281 , wherein the device is an enteral feeding device.
16296. A method as in any one of items 16031-16281 , wherein the device is a stent.
16297. A method as in any one of items 16031-16281 , wherein the device is a low profile device.
16298. A method as in any one of items 16031-16281 , wherein the device is a virtual colonoscopy device.
16299. A method as in any one of items 16031-16281 , wherein the device is a capsule endoscope.
16300. A method as in any one of items 16031-16281 , wherein the device is a retrieval device.
16301. A method as in any one of items 16031-16281 , wherein the device is a gastrointestinal device adapted for examining the interior of the gastrointestinal tract. 16302. A method as in any one of items 16031-16281 , wherein the device is a gastrointestinal device adapted for irrigation or aspiration of the gastrointestinal tract.
16303. A method as in any one of items 16031-16281 , wherein the device is a colostomy device.
16304. A method as in any one of items 16031-16281 , wherein the device is a mechanical hemostatic device adapted for control of gastrointestinal bleeding.
16305. A method as in any one of items 16031-16281 , wherein the device is a gastrointestinal device adapted for cleaning blocked gastrointestinal tract.
16306. A method as in any one of items 16031-16281, wherein the device is a gastrointestinal device for providing communication between two bodily systems.
16307. A method as in any one of items 16031-16281 , wherein the device is a portosystemic shunt.
16308. A method as in any one of items 16031-16281 , wherein the device is a dilation catheter.
16309. A method of making a medical device comprising: combining a central venous catheter (e.g., an implant) and an anti-scarring agent or a composition comprising an anti-scarring agent, wherein the agent inhibits scarring between the device and a host into which the device is implanted. 16310. The method of item 16309 wherein the agent is an adensosine A2A receptor antagonist.
16311. The method of item 16309 wherein the agent is an AKT inhibitor.
16312. The method of item 16309 wherein the agent is an alpha 2 integrin antagonist, wherein the alpha 2 integrin antagonist is Pharmaprojects No. 5754 (Merck KgaA).
16313. The method of item 16309 wherein the agent is an alpha 4 integrin antagonist.
16314. The method of item 16309 wherein the agent is an alpha 7 nicotinic receptor agonist.
16315. The method of item 16309 wherein the agent is an angiogenesis inhibitor selected from the group consisting of AG-12,958 (Pfizer), ATN-161 (Attention LLC), neovastat, an angiogenesis inhibitor from Jerina AG (Germany), NM-3 (Mercian), VGA-1155 (Taisho), FCE-26644 (Pfizer), FCE-26950 (Pfizer), FPMA (Meiji Daries), FR-111142 (Fujisawa), GGTI-298, GM-1306 (Llgand), GPA-1734 (Novartis), NNC-47-0011 (Novo Nordisk), herbamycin (Nippon Kayaku), lenalidomide (Celegene), IP-10 (NIH), ABT-828 (Abbott), KIN-841 (Tokushima University, Japan), SF-1126 (Semafore Pharmaceuticals), laminin technology (NIH), CHIR-258 (Chiron), NVP-AEW541 (Novartis), NVP-AEW541 (Novartis), Vt16907 (Alchemia), OXI-8007 (Oxigene), EG-3306 (Ark Therapeutics), Maspin (Arriva), ABT- 567 (Abbott), PPI-2458 (Praecis Pharmaceuticals), CC-5079, CC-4089 (Celgene), HIF-1 alpha inhibitors (Xenova), S-247 (Pfizer), AP-23573 (Ariad), AZD-9935 (Astra Zeneca), mebendazole (Introgen Therapeutics), MetAP-2 inhibitors (GlaxoSmithKline), AG-615 (Angiogene Pharmaceuticals), Tie-2 antagonists (Hybrigenics), NC-381 , CYC-381 , NC-169, NC-219, NC-383, NC-384, NC-407 (Lorus Therapeutics), ATN-224 (Attenuon), ON-01370 (Onconova), Vitronectin antagonists (Amgen), SDX-103 (Salmedix), Vitronectin antagonists (Shire), CHP (Riemser), TEK (Amgen), Anecortave acetate (Alcon), T46.2 (Matrix Therapeutics), HG-2 (Heptagen), TEM antagonists (Genzyme), Oxi-4500 (Oxigene), ATN-161 (Attenuon), WX-293 (Wilex), M-2025 (Metris Therapeutics), Alphastatin (BioActa), YH-16 (Yantai Rongchang), BIBF-1120 (Boehringer Ingelheim), BAY-57-9352 (Bayer), AS- 1404 (Cancer Research Technology), SC-77964 (Pfizer), glycomimetics (BioTie Therapies), TIE-2 Inhibitors (Ontogen), DIMI, Octamer (Octamer), ABR-215050 (Active Biotech), ABT-518 (Abbott), KDR inhibitors (Abbott), BSF-466895 (Abbott), SCH-221153 (Schering-Plough), DAC:antiangiogenic (ConjuChem), TFPI (EntreMed), AZD-2171 (Astra-Zenaca), CDC-394 (Celgene), LY290293 (EIi Lilly), IDN-5390 (Indena), Kdr Kinase Inhibitors (Merck), CT-113020, CT-116433, CT-116563, CT-31890, CT-32228) (Cell Therapeutics), A-299620 (Abbott), TWEAK Inhibitor (Amgen), VEGF modulators (Johnson and Johnson), Tum-N53, tumstatin (Genzyme), Thios- 1 , Thios-2 (Thios Pharmaceuticals), MV-6401 (Miravant Medical Technologies), Spisulosine (PharmaMar), CEP-7055 (Cephalon), AUV-201 (Auvation), LM-609 (EIi Lilly), SKF-106615 (AnorMED), Oglufanide disodium (Cytran), BW-114 (Pharminox), Calreticulin (NIH), WX-678 (Wilex), SD-7784 (Pfizer), WX-UK1 (Wilex), SH-268 (Schering AG), 2-Me-PGA (Celgene), S- 137 (Pfizer), ZD-6126 (Angiogene Pharmaceuticals), SG-292 (SignalGen), Benefin (Lane Labs), A6, A36 (Angstrom), SB-2723005 (GlaxoSmithKline), SC-7 (Cell Therapeutics), ZEN-014 (AEterna Zentaris), 2-methoxyestradiol (EntreMed), NK-130119 (Nippon Kayaku), CC-10004 (Celgene), AVE- 8062A (Ajinomoto), Tacedinaline (Pfizer), Actinonin (Tokyo Metropolitan Institute of Medical Science), Lenalidomide (Celgene), VGA-1155, BTO-956 (SRI International), ER-68203-00 (Eisai), CT-6685 (UCB), JKC-362 (Phoenix Pharmaceuticals), DMI-3798 (DMI Biosciences, Angiomate (Ipsen), ZD-6474 (AstraZeneca), CEP-5214 (Cephalon), Canstatin (Genzyme), NM-3 (Mercian), Oridigm (MediQuest Therapeutics), Exherin (Adherex), BLS-0597 (Boston Life Sciences), PTC-299 (PTC Therapeutics), NPI-2358 (Nereus Pharmaceuticals), CGP-79787 (Novartis), AEE-788 (Novartis), CKD-732 (Chong Kun Dang), CP-564959 (OSI Pharmaceuticals), CM-101 (CarboMed), CT-2584, CT3501 (Cell Therapeutics), combretastatin and analogues and derivatives thereof (Oxigene), Rebimastat (Bristol-Meyers Squibb), Dextrin 2-sulfate (ML Laboratories), Cilengitide (Merk KGaA), NSC-706704 (Pharminox), KRN- 951 (Kirin Brewery), Ukrain, NSC-631570 (Nowicky Pharma), Tecogalan sodium (Daiichi Pharmaceutical), Tz-93 (Tsumura), TBC-1635 (Encysive Pharmaceuticals), TAN-1120 (Takeda), Semaxanib (Pfizer), BDI-7800 (Biopharmacopae), SD-186, SD-983 (Bristol-Meyers Squibb), SB-223245 (GlaxoSmithKline), SC-236 (Pfizer), RWJ-590973 (Johnson and Johnson), ILX-1850 (Genzyme), SC-68488, S-836 (Pfizer), CG-55069-11 (CuraGen), Ki-23057 (Kirin Brewery), CCX-700 (Chemoentryx), Pegaptanib octasodium (Giled Sciences), ANGIOCOL (available from Biostratum Inc.), or an analogue or derivative thereof.
16316. The method of item 16309 wherein the agent is an apoptosis antagonist.
16317. The method of item 16309 wherein the agent is an apoptosis activator.
16318. The method of item 16309 wherein the agent is a beta 1 integrin antagonist.
16319. The method of item 16309 wherein the agent is a beta tubulin inhibitor. 16320. The method of item 16309 wherein the agent is a blocker of enzyme production in Hepatitis C.
16321. The method of item 16309 wherein the agent is a Bruton's tyrosine kinase inhibitor.
16322. The method of item 16309 wherein the agent is a calcineurin inhibitor.
16323. The method of item 16309 wherein the agent is a caspase 3 inhibitor.
16324. The method of item 16309 wherein the agent is a CC chemokine receptor antagonist.
16325. The method of item 16309 wherein the agent is a cell cycle inhibitor selected from the group consisting of SNS-595 (Sunesis), synthadotin, KRX-0403, and an analogue or derivative thereof.
16326. The method of item 16309 wherein the agent is a cathepsin B inhibitor.
16327. The method of item 16309 wherein the agent is a cathepsin K inhibitor, wherein the cathepsin K inhibitor is 462795 (GlaxoSmithKline), INPL-022-D6 (Amura Therapeutics), or an analogue or derivative thereof.
16328. The method of item 16309 wherein the agent is a cathepsin L inhibitor. 16329. The method of item 16309 wherein the agent is a CD40 antagonist.
16330. The method of item 16309 wherein the agent is a chemokine receptor agonist.
16331. The method of item 16309 wherein the agent is a chymase inhibitor.
16332. The method of item 16309 wherein the agent is a collagenase antagonist.
16333. The method of item 16309 wherein the agent is a CXCR antagonist.
16334. The method of item 16309 wherein the agent is a cyclin dependent kinase inhibitor selected from the group consisting of a CDK-1 inhibitor, a CDK-2 inhibitor, a CDK- 4 inhibitor, a CDK-6 inhibitor, a CAK1 inhibitor from GPC Biotech and Bristol-Myers Squibb, RGB-286199 (GPC Biotech), an anticancer agent from Astex Technology, a CAK1 inhibitor from GPC Biotech, a CDK inhibitor from Sanofi-Aventis, a CDK1/CDK2 inhibitor from Amgen, a CDK2 inhibitor from SUGEN-2 (Pfizer), a hearing loss therapy agent (Sound Pharmaceuticals), PD-0332991 (Pfizer), RGB-286199 (GPC Biotech), Ro-0505124 (Hoffmann-La Roche), a Ser/Thr kinase inhibitor from Lilly (EIi Lilly), CVT-2584 (CAS No. 199986-75- 9) (CV Therapeutics), and an analogue or derivative thereof.
16335. The method of item 16309 wherein the agent is a cyclooxygenase 1 inhibitor. 16336. The method of item 16309 wherein the agent is a DHFR inhibitor.
16337. The method of item 16309 wherein the agent is a dual integrin inhibitor.
16338. The method of item 16309 wherein the agent is an elastase inhibitor.
16339. The method of item 16309 wherein the agent is an elongation factor-1 alpha inhibitor.
16340. The method of item 16309 wherein the agent is an endothelial growth factor antagonist.
16341. The method of item 16309 wherein the agent is an endothelial growth factor receptor kinase inhibitor selected from the group consisting of sorafenib tosylate (Bayer), AAL-993 (Novartis), ABP-309 (Novartis), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), E- 7080 (Eisai), EG-3306 (Ark Therapeutics), EXEL-2880 (Exelixis), GW- 654652 (GlaxoSmithKline), a KDR inhibitor from LG Life Sciences, CT-6685 and CT-6729 (UCB), KRN-633 and KRN-951 (Kirin Brewery), OSI-930 (OSI Pharmaceuticals), SP-5.2 (Supratek Pharma), SU-11657 (Pfizer), a Tie-2 antagonist (Hybrigenics), a VEGFR-2 kinase inhibitor (Bristol-Myers Squibb), XL-647 (Exelixis), a KDR inhibitor from Abbott Laboratories, sorafenib tosylate, and an analogue or derivative thereof.
16342. The method of item 16309 wherein the agent is an endotoxin antagonist. 16343. The method of item 16309 wherein the agent is an epothilone and tubulin binder.
16344. The method of item 16309 wherein the agent is an estrogen receptor antagonist.
16345. The method of item 16309 wherein the agent is an FGF inhibitor.
16346. The method of item 16309 wherein the agent is a farnexyl transferase inhibitor.
16347. The method of item 16309 wherein the agent is farnesyltransferase inhibitor selected from the group of A-197574 (Abbott), a famesyltransferase inhibitor from Servier, FPTIlI (Strathclyde Institute for Drug R), LB-42908 (LG Life Sciences), Pharmaprojects No. 5063 (Genzyme), Pharmaprojects No. 5597 (Ipsen), Yissum Project No. B-1055 (Yissum), and an analogue or derivative thereof.
16348. The method of item 16309 wherein the agent is an FLT-3 kinase inhibitor.
16349. The method of item 16309 wherein the agent is an FGF receptor kinase inhibitor.
16350. The method of item 16309 wherein the agent is a fibrinogen antagonist selected from the group consisting of AUV-201 (Auvation), MG-13926 (Sanofi-Aventis), plasminogen activator (CAS No. 105913-11-9) (from Sanofi-Aventis or UCB), plasminogen activator-2 (tPA- 2) (Sanofi-Aventis), pro-urokinase (CAS No. 82657-92-9) (Sanofi-Aventis), and an analogue or derivative thereof. 16351. The method of item 16309 wherein the agent is a heat shock protein 90 antagonist selected from the group consisting of SRN- 005 (Sirenade), geldanamycin, NSC-33050 (17-allylaminogeldanamycin; 17- AAG), ^-dimethylaminoethylamino-^-demethoxy-geldanamycin (17- DMAG), rifabutin (rifamycin XIV, 1\4-didehydro-1-deoxy-1 ,4-dihydro-5'-(2- methylpropyl)-1-oxo-), and an analogue or derivative thereof.
16352. The method of item 16309 wherein the agent is a histone deacetylase inhibitor.
16353. The method of item 16309 wherein the agent is an HMGCoA reductase inhibitor selected from the group consisting of an atherosclerosis therapeutic from Lipid Sciences, ATI-16000 (ARYx Therapeutics), KS-01-019 (Kos Pharmaceuticals), Pharmaprojects No. 2197 (Sanofi-Aventi), RP 61969 (Sanofi-Aventis), and an analogue or derivative thereof.
16354. The method of item 16309 wherein the agent is an ICAM inhibitor.
16355. The method of item 16309 wherein the agent is an IL, ICE and IRAK antagonist, wherein the antagonist is a CJ-14877, CP- 424174 (Pfizer), NF-61 (Negma-Lerads), and an analogue or derivative thereof.
16356. The method of item 16309 wherein the agent is an IL-2 inhibitor.
16357. The method of item 16309 wherein the agent is an immunosuppressant selected from the group consisting of teriflunomide (Sanofi Aventis), chlorsulfaquinoxalone (NSC-339004), chlorsulfaquinoxalone sulfate, CS-712 (Sankyo), ismomultin alfa (CAS No. 457913-93-8) (Akzo Nobel), antiallergics from GenPat77, antiinflammatories or AT-005 (Androclus Therapeutics), autoimmune disease therapeutics from EpiVax, BN-007 (Bone), budesonide (CAS No. 51333-22- 3) (MAP Pharmaceuticals), CO-14 (Genzyme), edratide (CAS No. 433922- 67-9) (Teva), EP-314 (Enanta), eprovafen (CAS No. 101335-99-3) (Sanofi- Aventis), HWA-131 (CAS No. 118788-41-3) (Sanofi-Aventis), immunomodulators from MerLion Pharmaceuticals, immunosuppressives from Alchemia, IPL-12 (Inflazyme), MDL-9563 (CAS No. 27086-86-8) (Sanofi-Aventis), Pharmaprojects No. 2330 (Sanofi-Aventis), Pharmaprojects No. 6426 (Abgenix), PXS-25 (Pharmaxis), rosmarinic acid (CAS No. 20283-92-5) (Sanofi-Aventis), RP 42927 or RP 54745 (CAS No. 135330-08-4) (Sanofi-Aventis), SGN-35 (Seattle Genetics), ST-1959 (Sigma-Tau), type I diabetes therapy from SYNX Pharma, UNIL-88 (Debiopharm), VP-025 (Vasogen), VR-694 (Vectura), PRTX-001 (Protalex), and an analogue or derivative thereof.
16358. The method of item 16309 wherein the agent is an IMPDH (inosine monophosphate).
16359. The method of item 16309 wherein the agent is an integrin antagonist.
16360. The method of item 16309 wherein the agent is an interleukin antagonist.
16361. The method of item 16309 wherein the agent is an inhibitor of type III receptor tyrosine kinase.
16362. The method of item 16309 wherein the agent is an irreversible inhibitor of enzyme methionine aminopeptidase type 2. 16363. The method of item 16309 wherein the agent is an isozyme selective delta protein kinase C inhibitor.
16364. The method of item 16309 wherein the agent a JAK3 enzyme inhibitor.
16365. The method of item 16309 wherein the agent is a
JNK inhibitor.
16366. The method of item 16309 wherein the agent is a kinase inhibitor.
16367. The method of item 16309 wherein the agent is kinesin antagonist.
16368. The method of item 16309 wherein the agent is a kinesin antagonist.
16369. The method of item 16309 wherein the agent is a leukotriene inhibitor and antagonist selected from the group consisting of ambicromil (CAS No. 58805-38-2) (Sanofi-Aventis), amelubant (CAS No. 346735-24-8) (Boehringer Ingelheim), DW-1141 (Dong Wha), ebselen (Daiichi Pharmaceutical), ibudilast (Kyorin), leucotriene inhibitors from Sanofi-Aventis, lymphotoxin-beta receptor (LT-D) from Biogen Idee, Pharmaprojects No. 1535 and 2728 (CAS No. 119340-33-9) (Sanofi- Aventis), R-112 (Rigel), Rev-5367 (CAS No. 92532-05-3) (Sanofi-Aventis), RG-14893 (CAS No. 141835-49-6) (Sanofi-Aventis), RG-5901-A (CAS No. 101910-24-1), 92532-23-5, RP 66153 (CAS No. 142422-79-5), RP 66364 (CAS No.186916-92-5), RP 69698 (CAS No. 141748-00-7) (Sanofi-Aventis), SC-411930 (Pfizer), SC-41930 (CAS No. 120072-59-5) (Pfizer), SC-50605 (CAS No. 138828-39-4) (Pfizer), SC-51146 (CAS No. 141059-52-1), SC- 53228 (CAS No. 153633-01-3) (Pfizer), spaglumic acid (ZY-15106) (CAS No.3106-85-2), 80619-64-3 (Novartis), tipredane (CAS No. 85197-77-9) (Bristol-Myers Squibb), U-75302 (CAS No. 119477-85-9) (Pfizer), and analogue or derivative thereof.
16370. The method of item 16309 wherein the agent is an MAP kinase inhibitor.
16371. The method of item 16309 wherein the agent is a matrix metalloproteinase inhibitor.
16372. The method of item 16309 wherein the agent is an MCP-CCR2 inhibitor.
16373. The method of item 16309 wherein the agent is an mTOR inhibitor.
16374. The method of item 16309 wherein the agent is an mTOR kinase inhibitor.
16375. The method of item 16309 wherein the agent is a microtubule inhibitor selected from the group consisting of antibody- maytansinoid conjugates from Biogen Idee, colchicines (MantiCore Pharmaceuticals), anticancer immunoconjugates from Johnson & Johnson, DIME from Octamer, gni-1f (GNI), huC242-DM4, huMy9-6-DM1 (ImmunoGen), IDN-5404 (Indena), IMO-098, IMOderm (Imotep), mebendazole (Introgen Therapeutics), microtubule poisons from Cambridge Enterprise, paclitaxel such as LOTAX from Aphios (CAS No. 33069-62-4), GenexoI-PM from Samyang, Pharmaprojects No. 6383 (Tapestry Pharmaceuticals), RPR-112378 (Sanofi-Aventis), SGN-75 (Seattle Genetics), SPL-7435 (Starpharma), SSR-250411 (Sanofi-Aventis), trastuzumab-DM1 (Genentech), vinoreibine, and an analogue or derivative thereof.
16376. The method of item 16309 wherein the agent is an MIF inhibitor.
16377. The method of item 16309 wherein the agent is an MMP inhibitor.
16378. The method of item 16309 wherein the agent is a neurokinin (NK) antagonist selected from the group consisting of anthrotainin (CAS No. 148084-40-6) (Sanofi-Aventis), an IBS thereapeutic from ArQuIe, MDL-105212A (CAS No. 167261-60-1) (Ssanofi-Aventis), Pharmaprojects No. 2744, 3258 (CAS No. 139167-47-8) 4006, 4201, or 5986 (Sanofi-Aventis), RP 67580 (CAS No. 135911-02-3), SR-144190 (CAS No. 201152-86-5), SSR-240600, SSR-241586 (Sanofi-Aventis), TKA-457 (Novartis), vestipitant mesylate (CAS No. 334476-64-1) (GlaxoSmithKline), Win-64821 (Sanofi-Aventis), PRX-96026 (Predix Pharmaceuticals), and an analogue or derivative thereof.
16379. The method of item 16309 wherein the agent is an NF kappa B inhibitor selected from the group consisting of AVE-0545 or AVE-0547 (Sanofi-Aventis), bortezomib (CAS No. 179324-69-7) (Millennium Pharmaceuticals), dexanabinol (CAS No. 112924-45-5) (Pharmos), dexlipotam (Viatris), Pharmaprojects No. 6283 (INDRA) (OXiGENE), IPL- 576092 (CAS No. 137571-30-3) (Inflazyme), NFKB decoy (Corgentech), NFKB decoy oligo (AnGes MG), NFKB's from Ariad, osteoporosis treatments or S5 (F005) from Fulcrum Pharmaceuticals, P61 (Phytopharm), R-flurbiprofen (CAS No. 5104-49-4) (Encore Pharmaceuticals), and an analogue or derivative thereof. 16380. The method of item 16309 wherein the agent is a nitric oxide agonist.
16381. The method of item 16309 wherein the agent is an ornithine decarboxylase inhibitor.
16382. The method of item 16309 wherein the agent is a p38 MAP kinase inhibitor selected from the group consisting of AZD-6703 (AstraZeneca), JX-401 (Jexys Pharmaceuticals), BMS-2 (Bristol-Myers Squibb), a p38 MAP kinase inhibitor from Novartis, a p38-alpha MAP kinase inhibitor from Amphora, Pharmaprojects No. 5704 (Pharmacopeia), RPR- 200765A (Sanofi-Aventis), SD-282 (Johnson & Johnson), TAK-715 (Takeda), and an analogue or derivative thereof.
16383. The method of item 16309 wherein the agent is a palmitoyl-protein thioesterase inhibitor.
16384. The method of item 16309 wherein the agent is a PDGF receptor kinase inhibitor selected from the group consisting of AAL- 993, AMN-107, or ABP-309 (Novartis), AMG-706 (Amgen), BAY-57-9352 (Bayer), CDP-860 (UCB), E-7080 (Eisai), imatinib (CAS No. 152459-95-5) (Novartis), OSI-930 (OSI Pharmaceuticals), RPR-127963E (Sanofi-Aventis), RWJ-540973 (Johnson & Johnson), sorafenib tosylate (Bayer), SU-11657 (Pfizer), tandutinib (CAS No. 387867-13-2) (Millennium Pharmaceuticals), vatalanib (Novartis), ZK-CDK (Schering AG), and an analogue or derivative thereof.
16385. The method of item 16309 wherein the agent is (-)-halofenate (Metabolex), AMG-131 (Amgen), antidiabetics from Japan Tobacco, AZD-4619, AZD-8450, AZD-8677 (AstraZeneca), DRF-10945, balaglitazone (Dr Reddy's), CS-00088, CS-00098 (Chipscreen Biosciences), E-3030 (Eisai), etalocib (CAS No. 161172-51-6) (EIi Lilly), GSK-641597 (Ligand), GSK-677954 (GlaxoSmithKline), GW-409544 (Ligand), GW- 590735 (GlaxoSmithKline), K-111 (Hoffmann-La Roche), LY-518674 (EIi Lilly), LY-674 (Ligand), LY-929 (Ligand), MC-3001 , MC-3002 (MaxoCore Pharmaceuticals), metformin HCl + pioglitazone (CAS No. 1115-70-4 and 112529-15-4), ACTOPLUS MET from Andrx), muraglitazar (CAS No. 331741-94-7) (Bristol-Myers Squibb), naveglitazar (Ligand), oleoylethanolamide (Kadmus Pharmaceuticals), ONO-5129, pioglitazone hydrochloride (CAS No. 111025-46-8 and 112529-15-4) (Takeda), PLX-204 (Plexxikon), PPAR agonists from Genfit, PPAR delta agonists from EIi Lilly, PPAR-alpha agonists from CrystalGenomics, PPAR-gamma modulators and PPAR-β modulators from CareX, rosiglitazone maleate (CAS No. 122324- 73-4 or 155141-29-0) (GlaxoSmithKline), rosiglitazone maleate/glimepir (CAS No. 155141-29-0 and 93479-97-1), AVANDARYL, rosiglitazone maleate/metformin extend (CAS No. 155141-29-0 and 657-24-9), AVANDAMET, rosiglitazone maleate+metformin, AVANDAMET (GlaxoSmithKline), tesaglitazar (AstraZeneca), and an analogue or derivative thereof.
16386. The method of item 16309 wherein the agent is a phosphatase inhibitor.
16387. The method of item 16309 wherein the agent is a phosphodiesterase (PDE) inhibitor selected from the group consisting of avanafil (Tanabe Seiyaku), dasantafil (CAS No. 569351-91-3) (Schering- Plough), A-906119 (CAS No. 134072-58-5), DL-850 (Sanofi-Aventis), GRC- 3015, GRC-3566, GRC-3886 (Glenmark), HWA-153 (CAS No. 56395-66-5) (Sanofi-Aventis), hydroxypumafentrine (Altana), IBFB-130011 , IBFB-14-016, IBFB-140301 , IBFB-150007, IBFB-211913 (IBFB Pharma), L-826141 (Merck & Co), medorinone (CAS No. 88296-61-1) (Sanofi-Aventis), MEM- 1917 (Memory Pharmaceuticals), ND-1251 (Neuro3d), PDE inhibitors from ICOS1 PDE IV inhibitors from Memory Pharmaceuticals and CrystalGenomics, Pharmaprojects No. 2742 and 6141 (Sanofi-Aventis), QAD-171 (Novartis), RHC-2963 (CAS No. 76993-12-9 and 76993-14-1), RPR-117658, RPR-122818 derivatives, SR-24870 , and RPR-132294 (Sanofi-Aventis), SK-350 (ln2Gen), stroke therapy agents from deCODE Genetics, TAS-203 (Taiho), tofimilast (CAS No. 185954-27-2) (Pfizer), UK- 371800 (Pfizer), WIN-65579 (CAS No. 158020-82-7) (Sanofi-Aventis), IBFB- 130020 (IBFB Pharma), OPC-6535 (CAS No. 145739-56-6) (Otsuka), a phosphodiesterase III inhibitor, enoximone, a phosphodiesterase IV inhibitor, fosfosal, Atopik (Barrier Therapeutics), triflusal, a phosphodiesterase V inhibitor, and an analogue or derivative thereof.
16388. The method of item 16309 wherein the agent is a PKC inhibitor.
16389. The method of item 16309 wherein the agent is a platelet activating factor antagonist.
16390. The method of item 16309 wherein the agent is a platelet-derived growth factor receptor kinase inhibitor.
16391. The method of item 16309 wherein the agent is a prolyl hydroxylase inhibitor.
16392. The method of item 16309 wherein the agent is a polymorphonuclear neutrophil inhibitor.
16393. The method of item 16309 wherein the agent is a protein kinase B inhibitor. 16394. The method of item 16309 wherein the agent is a protein kinase C stimulant.
16395. The method of item 16309 wherein the agent is a purine nucleoside analogue.
16396. The method of item 16309 wherein the agent is a purinoreceptor P2X antagonist.
16397. The method of item 16309 wherein the agent is a Raf kinase inhibitor.
16398. The method of item 16309 wherein the agent is a reversible inhibitor of ErbB1 and ErbB2.
16399. The method of item 16309 wherein the agent is. a ribonucleoside triphosphate reductase inhibitor.
16400. The method of item 16309 wherein the agent is an SDF-1 antagonist.
16401. The method of item 16309 wherein the agent is a sheddase inhibitor.
16402. The method of item 16309 wherein the agent is an SRC inhibitor.
16403. The method of item 16309 wherein the agent is a stromelysin inhibitor. 16404. The method of item 16309 wherein the agent is an Syk kinase inhibitor.
16405. The method of item 16309 wherein the agent is a telomerase inhibitor.
16406. The method of item 16309 wherein the agent is a TGF beta inhibitor selected from the group consisting of pirfenidone (CAS No. 53179-13-8) (MARNAC), tranilast (CAS No. 53902-12-8) (Kissei), IN- 1130 (ln2Gen), mannose-6-phosphate (BTG), TGF-β antagonists from Inflazyme (Pharmaprojects No. 6075), TGF-β antagonists from Sydney, non-industrial source), TGF-β I receptor kinase inhibitors from Eli Lilly, TGF- β receptor inhibitors from Johnson & Johnson, and an analogue or derivative thereof.
16407. The method of item 16309 wherein the agent is a TNFα antagonist or TACE inhibitor selected from the group consisting of adalimumab (CAS No. 331731-18-1) (Cambridge Antibody Technology), AGIX-4207 (AtheroGenics), AGT-1 (Advanced Biotherapy), an antiinflammatory from Borean Pharma, Cellzome, or Paradigm Therapeutics, anti-inflammatory vaccine (TNF-alpha kinoid) from Neovacs, humanized anti-TNF antibody or an anti-TNF MAb (CB0006) Celltech (UCB), apratastat (CAS No. 287445-51-0) (Wyeth), BMS-561392 (Bristol-Myers Squibb), BN- 006 (Bone), certolizumab pegol (CAS No. 428863-50-7 or CH-138 (UCB), cilomilast (CAS No. 153259-65-5) (GlaxoSmithKline), CR-1 (Nuada Pharmaceuticals), CRx-119 (CombinatoRx), D-5410 (UCB), dacopafant (CAS No. 125372-33-0) (Sanofi-Aventis), dersalazine (CAS No. 188913-57- 7/188913-58-8) (Uriach), etanercept (CAS No. 185243-69-0) (Amgen), ethyl pyruvate (Critical (Critical Therapeutics), golimumab (CAS No. 476181-74-5) (Johnson & Johnson), hormono-immunotherapy from Ipsen, CDP571 (e.g., Humicade from UCB), IC-485 (ICOS), infliximab (CAS No. 170277-31-3) (Johnson & Johnson), IP-751 (Manhattan Pharmaceuticals), ISIS-104838 (CAS No. 250755-32-9) (ISIS Pharmaceuticals), lenalidomide (CAS No. 191732-72-6) (Celgene), lentinan (CAS No. 37339-90-5) (Ajinomoto), MDL- 201112 (CAS No. 142130-73-2) (Sanofi-Aventis), medroxyprogesterone (CAS No. 520-85-4) (InKine Pharmaceutical), N-acetylcysteine (CAS No. 616-91-1) (Zambon), NBE-P2 (DIREVO Biotech), nerelimomab (CAS No. 162774-06-3) (Chiron), OM-294DP (OM PHARMA), onercept (CAS No. 199685-57-9) (Yeda), PASSTN F-alpha (Verigen), pentoxifylline or oxypentifylline (Sanofi-Aventis), Pharmaprojects No. 4091, 4241, 4295, or 4488 (Sanofi-Aventis), Pharmaprojects No. 5480 (Amgen), Pharmaprojects No. 6749 (Cengent), pirfenidone (CAS No. 53179-13-8) (MARNAC), RPR- 132294 (Sanofi-Aventis), S5 (F002) (Fulcrum Pharmaceuticals), simvastatin (CAS No. 79902-63-9) (Merck & Co), STA-6292 (Synta Pharmaceuticals), tacrolimus (CAS No. 104987-11-3) (Fujisawa LifeCycle Pharma), talactoferrin alfa (CAS No. 308240-58-6) (Agennix), thalidomide (CAS No. 50-35-1) (Celgene), TNF antagonists form ProStrakan, and Synergen, TNF inhibitors (Amgen), TNF-alpha antagonists from Dynavax Technologies and Jerina AG (Germany), TNF-alpha inhibitors from IBFB Pharma and Xencor (Xencor), torbafylline (CAS No. 105102-21-4) (Sanofi-Aventis), UR-1505 (Uriach), VT-346 (Viron Therapeutics), YSIL6 (Y's Therapeutics), YSTH2 (Y's Therapeutics), NPI-1302a-3 (Nereus Pharmaceuticals, a TNF antagonist from Jerina AG (Germany), dersalazine, and an analogue or derivative thereof.
16408. The method of item 16309 wherein the agent is a Toll receptor inhibitor.
16409. The method of item 16309 wherein the agent is a tubulin antagonist. 16410. The method of item 16309 wherein the agent is a tyrosine kinase inhibitor selected from the group consisting of SU-011248, SUTENT from Pfizer Inc. (New York, NY), BMS-354825, PN-355 (Paracelsian Pharmaceuticals), AGN-199659 (Allergan), AAL-993 or ABP- 309 (Novartis), adaphostin (NlH), AEE-788 (Novartis), AG-013736 (OSI Pharmaceuticals), AG-13736 (Pfizer), ALT-110 (Alteris Therapeutics), AMG- 706 (Amgen), anticancer MAbs from Xencor, anti-EGFrvlll MAbs from Abgenix, anti-HER2 MAb from Abiogen, AZD-2171 or AZD-9935 (AstraZeneca), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), CEP-5214 (Cephalon), CEP-7055 (Cephalon), cetuximab (ImClone Systems), CHIR-200131 and CHIR-258 (Chiron), CP-547632 (OSI Pharmaceuticals), CP-724714 (Pfizer), CT-301 (Creabilis Therapeutics), D- 69491 (Baxter International), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EGFR/ErbB2 inhibitors from Array BioPharma, erlotinib (CAS No. 183319- 69-9) (OSI Pharmaceuticals), EXEL-2880 (Exelixis), FK-778 (Sanofi- Aventis) , gefrtinib (CAS No. 184475-35-2) (AstraZeneca), GW-2286 or GW- 654652 (GlaxoSmithKline), her2/neu antigen from AlphaVax, HER-2/neu inhibitor from Generex, Herzyme (Medipad) (Sirna Therapeutics), HKI-272 (Wyeth), HuMax-EGFr (Genmab), idronoxil (CAS No. 81267-65-4) (Novogen), IGF-1 inhibitors from Ontogen, IMC-11 F8 (ImClone Systems), kahalalide F (CAS No. 149204-42-2) (PharmaMar), KDR inhibitor from LG Life Sciences, KDR inhibitors from Abbott Laboratories, KDR kinase inhibitors (UCB), Kdr kinase inhibitors from Merck & Co, KRN-633 and KRN- 951 (Kirin Brewery), KSB-102 (Xenova), lapatinib ditosylate (CAS No. 388082-78-8) (GlaxoSmithKline), matuzumab (Merck KGaA), MDX-214 (Medarex), ME-103 (Pharmexa), MED-A300 (Gerolymatos), MNAC-13 (Lay Line Genomics), nimotuzumab (Center of Molecular Immunology), NSC- 330507 or NSC-707545 (NlH), NV-50 (Novogen), OSI-930 (OSI Pharmaceuticals), panitumumab (Abgenix), pelitinib (CAS No. 287933-82-7) (Wyeth), pertuzumab (CAS No. 380610-27-5) (Genentech), Pharmaprojects No. 3985 (Sanofi-Aventis), prostate cancer therapeutics from Sequenom (SQPC35, SQPC36, SQPC90), removab and remoxab (Trion Pharma), RG- 13022 (CAS No. 136831-48-6), RG-13291 (CAS No. 138989-50-1), or RG- 14620 (CAS No. 136831-49-7) (Sanofi-Aventis), RM-6427 (Romark), RNAi breast cancer therapy from Benitec, RP 53801 (CAS No. 125882-88-4) (Sanofi-Aventis), sorafenib tosylate (Bayer), SU-11657 (Pfizer), Tie-2 antagonists from Semaia (Hybrigenics), Tie-2 inhibitors from Ontogen, trastuzumab (CAS No. 180288-69-1) (Genentech), tyrosine kinase inhibitors from Sanofi-Aventis, U3-1287, U3-1565, U3-1784, U3-1800 (U3 Pharma), vatalanib (Novartis), VEGFR-2 kinase inhibitor from Bristol-Myers Squibb, XL-647 (Exelixis), ZD-6474 (AstraZeneca), ZK-CDK (Schering AG), an EGFR tyrosine kinase inhibitor, EKB-569 (Wyeth), and an analogue or derivative thereof.
16411. The method of item 16309 wherein the agent is a VEGF inhibitor.
16412. The method of item 16309 wherein the agent is a vitamin D receptor agonist.
16413. The method of item 16309 wherein the agent is ZD-6474 (an angiogenesis inhibitor).
16414. The method of item 16309 wherein the agent is AP-23573 (an mTOR inhibitor).
16415. The method of item 16309 wherein the agent is synthadotin (a tubulin antagonist).
16416. The method of item 16309 wherein the agent is S-0885 (a collagenase inhibitor). 16417. The method of item 16309 wherein the agent is aplidine (an elongation factor-1 alpha inhibitor).
16418. The method of item 16309 wherein the agent is ixabepilone (an epithilone).
16419. The method of item 16309 wherein the agent is IDN-5390 (an angiogenesis inhibitor).
16420. The method of item 16309 wherein the agent is SB-2723005 (an angiogenesis inhibitor).
16421. The method of item 16309 wherein the agent is ABT-518 (an angiogenesis inhibitor).
16422. The method of item 16309 wherein the agent is combretastatin (an angiogenesis inhibitor).
16423. The method of item 16309 wherein the agent is anecortave acetate (an angiogenesis inhibitor).
16424. The method of item 16309 wherein the agent is SB-715992 (a kinesin antagonist).
16425. The method of item 16309 wherein the agent is temsirolimus (an mTOR inhibitor).
16426. The method of item 16309 wherein the agent is adalimumab (a TNFα antagonist). 16427. The method of item 16309, wherein the composition comprises a polymer.
16428. The method of item 16309, wherein the composition comprises a polymeric carrier.
16429. The method of item 16309 wherein the anti- scarring agent inhibits adhesion between the device and a host into which the device is implanted.
16430. The method of item 16309 wherein the device delivers the anti-scarring agent locally to tissue proximate to the device.
16431. The method of item 16309 wherein the device has a coating that comprises the anti-scarring agent.
16432. The method of item 16309, wherein the device has a coating that comprises the agent and is disposed on a surface of the implant.
16433. The method of item 16309, wherein the device has a coating that comprises the agent and directly contacts the implant.
16434. The method of item 16309, wherein the device has a coating that comprises the agent and indirectly contacts the implant.
16435. The method of item 16309, wherein the device has a coating that comprises the agent and partially covers the implant.
16436. The method of item 16309, wherein the device has a coating that comprises the agent and completely covers the implant. 16437. The method of item 16309, wherein the device has a uniform coating.
16438. The method of item 16309, wherein the device has a non-uniform coating.
16439. The method of item 16309, wherein the device has a discontinuous coating.
16440. The method of item 16309, wherein the device has a patterned coating.
16441. The method of item 16309, wherein the device has a coating with a thickness of 100 Dm or less.
16442. The method of item 16309, wherein the device has a coating with a thickness of 10 Dm or less.
16443. The method of item 16309, wherein the device has a coating, and the coating adheres to the surface of the implant upon deployment of the implant.
16444. The method of item 16309, wherein the device has a coating, and wherein the coating is stable at room temperature for a period of 1 year.
16445. The method of item 16309, wherein the device has a coating, and wherein the anti-scarring agent is present in the coating in an amount ranging between about 0.0001% to about 1% by weight. 16446. The method of item 16309, wherein the device has a coating, and wherein the anti-scarring agent is present in the coating in an amount ranging between about 1% to about 10% by weight.
16447. The method of item 16309, wherein the device has a coating, and wherein the anti-scarring agent is present in the coating in an amount ranging between about 10% to about 25% by weight.
16448. The method of item 16309, wherein the device has a coating, and wherein the anti-scarring agent is present in the coating in an amount ranging between about 25% to about 70% by weight.
16449. The method of item 16309, wherein the device has a coating, and wherein the coating further comprises a polymer.
16450. The method of item 16309, wherein the device has a first coating having a first composition and a second coating having a second composition.
16451. The method of item 16309, wherein the device has a first coating having a first composition and a second coating having a second composition, wherein the first composition and the second composition are different.
16452. The method of item 16309, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a copolymer.
16453. The method of item 16309, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a block copolymer. 16454. The method of item 16309, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a random copolymer.
16455. The method of item 16309, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a biodegradable polymer.
16456. The method of item 16309, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a non-biodegradable polymer.
16457. The method of item 16309, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a hydrophilic polymer.
16458. The method of item 16309, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a hydrophobic polymer.
16459. The method of item 16309, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a polymer having hydrophilic domains.
16460. The method of item 16309, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a polymer having hydrophobic domains.
16461. The method of item 16309, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a non-conductive polymer. 16462. The method of item 16309, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises an elastomer.
16463. The method of item 16309, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a hydrogel.
16464. The method of item 16309, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a silicone polymer.
16465. The method of item 16309, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a hydrocarbon polymer.
16466. The method of item 16309, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a styrene-derived polymer.
16467. The method of item 16309, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a butadiene polymer.
16468. The method of item 16309, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a macromer.
16469. The method of item 16309, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a poly(ethylene glycol) polymer. 16470. The method of item 16309 wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises an amorphous polymer.
16471. The method of item 16309, wherein the device comprises a lubricious coating.
16472. The method of item 16309 wherein the anti- scarring agent is located within pores or holes of the device.
16473. The method of item 16309 wherein the anti- scarring agent is located within a channel, lumen, or divet of the device.
16474. The method of item 16309, wherein the device comprises a second pharmaceutically active agent.
16475. The method of item 16309 wherein the device comprises an anti-inflammatory agent.
16476. The method of item 16309 wherein the device comprises an agent that inhibits infection.
16477. The method of item 16309 wherein the device comprises an agent that inhibits infection, and wherein the agent is an anthracycline.
16478. The method of item 16309 wherein the device comprises an agent that inhibits infection, and wherein the agent is doxorubicin. 16479. The method of item 16309 wherein the device comprises an agent that inhibits infection, and wherein the agent is • mitoxantrone.
16480. The method of item 16309 wherein the device comprises an agent that inhibits infection, and wherein the agent is a fluoropyrimidine.
16481. The method of item 16309 wherein the device comprises an agent that inhibits infection, and wherein the agent is 5- fluorouracil (5-FU).
16482. The method of item 16309 wherein the device comprises an agent that inhibits infection, and wherein the agent is a folic acid antagonist.
16483. The method of item 16309 wherein the device comprises an agent that inhibits infection, and wherein the agent is methotrexate.
16484. The method of item 16309 wherein the device comprises an agent that inhibits infection, and wherein the agent is a podophylotoxin.
16485. The method of item 16309 wherein the device comprises an agent that inhibits infection, and wherein the agent is etoposide.
16486. The method of item 16309 wherein the device comprises an agent that inhibits infection, and wherein the agent is a camptothecin. 16487. The method of item 16309 wherein the device comprises an agent that inhibits infection, and wherein the agent is a hydroxyurea.
16488. The method of item 16309 wherein the device comprises an agent that inhibits infection, and wherein the agent is a platinum complex.
16489. The method of item 16309 wherein the device comprises an agent that inhibits infection, and wherein the agent is cisplatin.
16490. The method of item 16309, further comprising an anti-thrombotic agent.
16491. The method of item 16309 wherein the device comprises a visualization agent.
16492. The method of item 16309 wherein the device comprises a visualization agent, wherein the visualization agent is a radiopaque material, and wherein the radiopaque material comprises a metal, a halogenated compound, or a barium containing compound.
16493. The method of item 16309 wherein the device comprises a visualization agent, wherein the visualization agent is a radiopaque material, and wherein the radiopaque material comprises barium, tantalum, or technetium.
16494. The method of item 16309 wherein the device comprises a visualization agent, and wherein the visualization agent is a MRI responsive material. 16495. The method of item 16309 wherein the device comprises a visualization agent, and wherein the visualization agent comprises a gadolinium chelate.
16496. The method of item 16309 wherein the device comprises a visualization agent, and wherein the visualization agent comprises iron, magnesium, manganese, copper, or chromium.
16497. The method of item 16309 wherein the device comprises a visualization agent, and wherein the visualization agent comprises an iron oxide compound.
16498. The method of item 16309 wherein the device comprises a visualization agent, and wherein the visualization agent comprises a dye, pigment, or colorant.
16499. The method of item 16309 wherein the device comprises an echogenic material.
16500. The method of item 16309 wherein the device comprises an echogenic material, and wherein the echogenic material is in the form of a coating.
16501. The method of item 16309 wherein the device is sterile.
16502. The method of item 16309 wherein the anti- scarring agent is released into tissue in the vicinity of the device after deployment of the device. 16503. The method of item 16309 wherein the anti- scarring agent is released into tissue in the vicinity of the device after deployment of the device, and wherein the tissue is connective tissue.
16504. The method of item 16309 wherein the anti- scarring agent is released into tissue in the vicinity of the device after deployment of the device, and wherein the tissue is muscle tissue.
16505. The method of item 16309 wherein the anti- scarring agent is released into tissue in the vicinity of the device after deployment of the device, and wherein the tissue is nerve tissue.
16506. The method of item 16309 wherein the anti- scarring agent is released into tissue in the vicinity of the device after deployment of the device, and wherein the tissue is epithelium tissue.
16507. The method of item 16309 wherein the anti- scarring agent is released in effective concentrations from the device over a period ranging from the time of deployment of the device to about 1 year.
16508. The method of item 16309 wherein the anti- scarring agent is released in effective concentrations from the device over a period ranging from about 1 month to 6 months.
16509. The method of item 16309 wherein the anti- scarring agent is released in effective concentrations from the device over a period ranging from about 1 - 90 days.
16510. The method of item 16309 wherein the anti- scarring agent is released in effective concentrations from the device at a constant rate. 16511. The method of item 16309 wherein the anti- scarring agent is released in effective concentrations from the device at an increasing rate.
16512. The method of item 16309 wherein the anti- scarring agent is released in effective concentrations from the device at a decreasing rate.
16513. The method of item 16309 wherein the anti- scarring agent is released in effective concentrations from the composition comprising the anti-scarring agent by diffusion over a period ranging from the time of deployment of the device to about 90 days.
16514. The method of item 16309 wherein the anti- scarring agent is released in effective concentrations from the composition comprising the anti-scarring agent by erosion of the composition over a period ranging from the time of deployment of the device to about 90 days.
16515. The method of item 16309 wherein the device comprises about 0.01 Dg to about 10 Dg of the anti-scarring agent.
16516. The method of item 16309 wherein the device comprises about 10 Dg to about 10 mg of the anti-scarring agent.
16517. The method of item 16309 wherein the device comprises about 10 mg to about 250 mg of the anti-scarring agent.
16518. The method of item 16309 wherein the device comprises about 250 mg to about 1000 mg of the anti-scarring agent. 16519. The method of item 16309 wherein the device comprises about 1000 mg to about 2500 mg of the anti-scarring agent.
16520. The method of item 16309 wherein a surface of the device comprises less than 0.01 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
16521. The method of item 16309 wherein a surface of the device comprises about 0.01 Dg to about 1 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
16522. The method of item 16309 wherein a surface of the device comprises about 1 Dg to about 10 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
16523. The method of item 16309 wherein a surface of the device comprises about 10 Dg to about 250 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
16524. The method of item 16309 wherein a surface of the device comprises about 250 Dg to about 1000 Dg of the anti-scarring agent of anti-scarring agent per mm2 of device surface to which the anti- scarring agent is applied.
16525. The method of item 16309 wherein a surface of the device comprises about 1000 Dg to about 2500 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
16526. The method of item 16309 wherein the combining is performed by direct affixing the agent or the composition to the implant. 16527. The method of item 16309 wherein the combining is performed by spraying the agent or the component onto the implant.
16528. The method of item 16309 wherein the combining is performed by electrospraying the agent or the composition onto the implant.
16529. The method of item 16309 wherein the combining is performed by dipping the implant into a solution comprising the agent or the composition.
16530. The method of item 16309 wherein the combining is performed by covalently attaching the agent or the composition to the implant.
16531. The method of item 16309 wherein the combining is performed by non-covalently attaching the agent or the composition to the implant.
16532. The method of item 16309 wherein the combining is performed by coating the implant with a substance that contains the agent or the composition.
16533. The method of item 16309 wherein the combining is performed by coating the implant with a substance that absorbs the agent.
16534. The method of item 16309 wherein the combining is performed by interweaving a thread composed of, or coated with, the agent or the composition. 16535. The method of item 16309 wherein the combining is performed by covering all the implant with a sleeve that contains the agent or the composition.
16536. The method of item 16309 wherein the combining is performed by covering a portion of the implant with a sleeve that contains the agent or the composition.
16537. The method of item 16309 wherein the combining is performed by covering all the implant with a cover that contains the agent or the composition.
16538. The method of item 16309 wherein the combining is performed by covering a portion of the implant with a cover that contains the agent or the composition.
16539. The method of item 16309 wherein the combining is performed by covering all the implant with an electrospun fabric that contains the agent or the composition.
16540. The method of item 16309 wherein the combining is performed by covering a portion of the implant with an electrospun fabric that contains the agent or the composition.
16541. The method of item 16309 wherein the combining is performed by covering all the implant with a mesh that contains the agent or the composition.
16542. The method of item 16309 wherein the combining is performed by covering a portion of the implant with a mesh that contains the agent or the composition. 16543. The method of item 16309 wherein the combining is performed by constructing all the implant with the agent or the composition.
16544. The method of item 16309 wherein the combining is performed by constructing a portion of the implant with the agent or the composition.
16545. The method of item 16309 wherein the combining is performed by impregnating the implant with the agent or the composition.
16546. The method of item 16309 wherein the combining is performed by constructing all of the implant from a degradable polymer that releases the agent.
16547. The method of item 16309 wherein the combining is performed by constructing a portion of the implant from a degradable polymer that releases the agent.
16548. The method of item 16309 wherein the combining is performed by dipping the implant into a solution that comprise the agent and an inert solvent for the implant.
16549. The method of item 16309 wherein the combining is performed by dipping the implant into a solution that comprises the agent and a solvent that will swell the implant.
16550. The method of item 16309 wherein the combining is performed by dipping the implant into a solution that comprises the agent and a solvent that will dissolve the implant. 16551. The method of item 16309 wherein the combining is performed by dipping the implant into a solution that comprises the agent, a polymer and an inert solvent for the implant.
16552. The method of item 16309 wherein the combining is performed by dipping the implant into a solution that comprises the agent, a polymer and a solvent that will swell the implant.
16553. The method of item 16309 wherein the combining is performed by dipping the implant into a solution that comprises the agent, a polymer and a solvent that will dissolve the implant.
16554. The method of item 16309 wherein the combining is performed by spraying the implant into a solution that comprises the agent and an inert solvent for the implant.
16555. The method of item 16309 wherein the combining is performed by spraying the implant into a solution that comprises the agent and a solvent that will swell the implant.
16556. The method of item 16309 wherein the combining is performed by spraying the implant into a solution that comprises the agent and a solvent that will dissolve the implant.
16557. The method of item 16309 wherein the combining is performed by spraying the implant into a solution that comprises the agent, a polymer and an inert solvent for the implant.
16558. The method of item 16309 wherein the combining is performed by spraying the implant into a solution that comprises the agent, a polymer and a solvent that will swell the implant. 16559. The method of item 16309 wherein the combining is performed by spraying the implant into a solution that comprises the agent, a polymer and a solvent that will dissolve the implant.
16560. A method as in any one of items 16309-16559, wherein the device is a central venous catheter with a cuff.
16561. A method as in any one of items 16309-16559, wherein the device is a central venous catheter without a cuff.
16562. A method as in any one of items 16309-16559, wherein the device is a central venous catheter with a flange.
16563. A method as in any one of items 16309-16559, wherein the device is a central venous catheter without a flange.
16564. A method as in any one of items 16309-16559, wherein the device is a central venous catheter adapted for providing access to the circulatory system.
16565. A method as in any one of items 16309-16559, wherein the device is a central venous catheter adapted for providing multiple conduits for accessing the circulatory system.
16566. A method as in any one of items 16309-16559, wherein the device is a central venous catheter comprising a means for preventing infection as a result of long term use.
16567. A method as in any one of items 16309-16559, wherein the device is a central venous catheter adaptable for being used with an apparatus that provides a means of controlling the injection or withdrawl of bodily fluids through the central venous catheter.
16568. A method as in any one of items 16309-16559, wherein the device is a parenteral nutrition catheter.
16569. A method as in any one of items 16309-16559, wherein the device is a peripherally inserted central venous catheter.
16570. A method as in any one of items 16309-16559, wherein the device is a flow directed balloon tipped pulmonary artery catheter.
16571. A method as in any one of items 16309-16559, wherein the device is a long term central venous access catheter.
16572. A method of making a medical device comprising: combining a ventricular assist device (e.g., an implant) and an anti-scarring agent or a composition comprising an anti-scarring agent, wherein the agent inhibits scarring between the device and a host into which the device is implanted.
16573. The method of item 16572 wherein the agent is an adensosine A2A receptor antagonist.
16574. The method of item 16572 wherein the agent is an AKT inhibitor.
16575. The method of item 16572 wherein the agent is an alpha 2 integrin antagonist, wherein the alpha 2 integrin antagonist is Pharmaprojects No. 5754 (Merck KgaA). 16576. The method of item 16572 wherein the agent is an alpha 4 integrin antagonist.
16577. The method of item 16572 wherein the agent is an alpha 7 nicotinic receptor agonist.
16578. The method of item 16572 wherein the agent is an angiogenesis inhibitor selected from the group consisting of AG-12,958 (Pfizer), ATN-161 (Attention LLC), neovastat, an angiogenesis inhibitor from Jerina AG (Germany), NM-3 (Mercian), VGA-1155 (Taisho), FCE-26644 (Pfizer), FCE-26950 (Pfizer), FPMA (Meiji Daries), FR-111142 (Fujisawa), GGTI-298, GM-1306 (Ligand), GPA-1734 (Novartis), NNC-47-0011 (Novo Nordisk), herbamycin (Nippon Kayaku), lenalidomide (Celegene), IP-10 (NIH), ABT-828 (Abbott), KIN-841 (Tokushima University, Japan), SF-1126 (Semafore Pharmaceuticals), laminin technology (NIH), CHIR-258 (Chiron), NVP-AEW541 (Novartis), NVP-AEW541 (Novartis), VM6907 (Alchemia), OXI-8007 (Oxigene), EG-3306 (Ark Therapeutics), Maspin (Arriva), ABT- 567 (Abbott), PPI-2458 (Praecis Pharmaceuticals), CC-5079, CC-4089 (Celgene), HIF-1 alpha inhibitors (Xenova), S-247 (Pfizer), AP-23573 (Ariad), AZD-9935 (Astra Zeneca), mebendazole (Introgen Therapeutics), MetAP-2 inhibitors (GlaxoSmithKline), AG-615 (Angiogene Pharmaceuticals), Tie-2 antagonists (Hybrigenics), NC-381 , CYC-381 , NC-169, NC-219, NC-383, NC-384, NC-407 (Lorus Therapeutics), ATN-224 (Attenuon), ON-01370 (Onconova), Vitronectin antagonists (Amgen), SDX-103 (Salmedix), Vitronectin antagonists (Shire), CHP (Riemser), TEK (Amgen), Anecortave acetate (Alcon), T46.2 (Matrix Therapeutics), HG-2 (Heptagen), TEM antagonists (Genzyme), Oxi-4500 (Oxigene), ATN-161 (Attenuon), WX-293 (Wilex), M-2025 (Metris Therapeutics), Alphastatin (BioActa), YH-16 (Yantai Rongchang), B1BF-1120 (Boehringer Ingelheim), BAY-57-9352 (Bayer), AS- 1404 (Cancer Research Technology), SC-77964 (Pfizer), glycomimetics (BioTie Therapies), TIE-2 Inhibitors (Ontogen), DIMI, Octamer (Octamer), ABR-215050 (Active Biotech), ABT-518 (Abbott), KDR inhibitors (Abbott), BSF-466895 (Abbott), SCH-221153 (Schering-Plough), DAC:antiangiogenic (ConjuChem), TFPI (EntreMed), AZD-2171 (Astra-Zenaca), CDC-394 (Celgene), LY290293 (EIi Lilly), IDN-5390 (Indena), Kdr Kinase Inhibitors (Merck), CT-113020, CT-116433, CT-116563, CT-31890, CT-32228) (Cell Therapeutics), A-299620 (Abbott), TWEAK Inhibitor (Amgen), VEGF modulators (Johnson and Johnson), Tum-N53, tumstatin (Genzyme), Thios- 1 , Thios-2 (Thios Pharmaceuticals), MV-6401 (Miravant Medical Technologies), Spisulosine (PharmaMar), CEP-7055 (Cephalon), AUV-201 (Auvation), LM-609 (EIi Lilly), SKF-106615 (AnorMED), Oglufanide disodium (Cytran), BW-114 (Pharminox), Calreticulin (NIH), WX-678 (Wilex), SD-7784 (Pfizer), WX-UK1 (Wilex), SH-268 (Schering AG), 2-Me-PGA (Celgene), S- 137 (Pfizer), ZD-6126 (Angiogene Pharmaceuticals), SG-292 (SignalGen), Benefin (Lane Labs), A6, A36 (Angstrom), SB-2723005 (GlaxoSmithKline), SC-7 (Cell Therapeutics), ZEN-014 (AEterna Zentaris), 2-methoxyestradiol (EntreMed), NK-130119 (Nippon Kayaku), CC-10004 (Celgene), AVE- 8062A (Ajinomoto), Tacedinaline (Pfizer), Actinonin (Tokyo Metropolitan Institute of Medical Science), Lenalidomide (Celgene), VGA-1155, BTO-956 (SRI International), ER-68203-00 (Eisai), CT-6685 (UCB), JKC-362 (Phoenix Pharmaceuticals), DMI-3798 (DMI Biosciences, Angiomate (Ipsen), ZD-6474 (AstraZeneca), CEP-5214 (Cephalon), Canstatin (Genzyme), NM-3 (Mercian), Oridigm (MediQuest Therapeutics), Exherin (Adherex), BLS-0597 (Boston Life Sciences), PTC-299 (PTC Therapeutics), NPI-2358 (Nereus Pharmaceuticals), CGP-79787 (Novartis), AEE-788 (Novartis), CKD-732 (Chong Kun Dang), CP-564959 (OSI Pharmaceuticals), CM-101 (CarboMed), CT-2584, CT3501 (Cell Therapeutics), combretastatin and analogues and derivatives thereof (Oxigene), Rebimastat (Bristol-Meyers Squibb), Dextrin 2-sulfate (ML Laboratories), Cilengitide (Merk KGaA), NSC-706704 (Pharminox), KRN- 951 (Kirin Brewery), Ukrain, NSC-631570 (Nowicky Pharma), Tecogalan sodium (Daiichi Pharmaceutical), Tz-93 (Tsumura), TBC-1635 (Encysive Pharmaceuticals), TAN-1120 (Takeda), Semaxanib (Pfizer), BDI-7800 (Biopharmacopae), SD-186, SD-983 (Bristol-Meyers Squibb), SB-223245 (GlaxoSmithKline), SC-236 (Pfizer), RWJ-590973 (Johnson and Johnson), ILX-1850 (Genzyme), SC-68488, S-836 (Pfizer), CG-55069-11 (CuraGen), Ki-23057 (Kirin Brewery), CCX-700 (Chemoentryx), Pegaptanib octasodium (Giled Sciences), ANGIOCOL (available from Biostratum Inc.), or an analogue or derivative thereof.
16579. The method of item 16572 wherein the agent is an apoptosis antagonist.
16580. The method of item 16572 wherein the agent is an apoptosis activator.
16581. The method of item 16572 wherein the agent is a beta 1 integrin antagonist.,
16582. The method of item 16572 wherein the agent is a beta tubulin inhibitor.
16583. The method of item 16572 wherein the agent is a blocker of enzyme production in Hepatitis C.
16584. The method of item 16572 wherein the agent is a Bruton's tyrosine kinase inhibitor.
16585. The method of item 16572 wherein the agent is a calcineurin inhibitor.
16586. The method of item 16572 wherein the agent is a caspase 3 inhibitor. 16587. The method of item 16572 wherein the agent is a CC chemokine receptor antagonist.
16588. The method of item 16572 wherein the agent is a cell cycle inhibitor selected from the group consisting of SNS-595 (Sunesis), synthadotin, KRX-0403, and an analogue or derivative thereof.
16589. The method of item 16572 wherein the agent is a cathepsin B inhibitor.
16590. The method of item 16572 wherein the agent is a cathepsin K inhibitor, wherein the cathepsin K inhibitor is 462795 (GlaxoSmithKline), INPL-022-D6 (Amura Therapeutics), or an analogue or derivative thereof.
16591. The method of item 16572 wherein the agent is a cathepsin L inhibitor.
16592. The method of item 16572 wherein the agent is a CD40 antagonist.
16593. The method of item 16572 wherein the agent is a chemokine receptor agonist.
16594. The method of item 16572 wherein the agent is a chymase inhibitor.
16595. The method of item 16572 wherein the agent is a collagenase antagonist. 16596. The method of item 16572 wherein the agent is a CXCR antagonist.
16597. The method of item 16572 wherein the agent is a cyclin dependent kinase inhibitor selected from the group consisting of a CDK-1 inhibitor, a CDK-2 inhibitor, a CDK- 4 inhibitor, a CDK-6 inhibitor, a CAK1 inhibitor from GPC Biotech and Bristol-Myers Squibb, RGB-286199 (GPC Biotech), an anticancer agent from Astex Technology, a CAK1 inhibitor from GPC Biotech, a CDK inhibitor from Sanofi-Aventis, a CDK1/CDK2 inhibitor from Amgen, a CDK2 inhibitor from SUGEN-2 (Pfizer), a hearing loss therapy agent (Sound Pharmaceuticals), PD-0332991 (Pfizer), RGB-286199 (GPC Biotech), Ro-0505124 (Hoffmann-La Roche), a Ser/Thr kinase inhibitor from Lilly (EIi Lilly), CVT-2584 (CAS No. 199986-75- 9) (CV Therapeutics), and an analogue or derivative thereof.
16598. The method of item 16572 wherein the agent is a cyclooxygenase 1 inhibitor.
16599. The method of item 16572 wherein the agent is a DHFR inhibitor.
16600. The method of item 16572 wherein the agent is a dual integrin inhibitor.
16601. The method of item 16572 wherein the agent is an elastase inhibitor.
16602. The method of item 16572 wherein the agent is an elongation factor-1 alpha inhibitor. 16603. The method of item 16572 wherein the agent is an endothelial growth factor antagonist.
16604. The method of item 16572 wherein the agent is an endothelial growth factor receptor kinase inhibitor selected from the group consisting of sorafenib tosylate (Bayer), AAL-993 (Novartis), ABP-309 (Novartis), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), E- 7080 (Eisai), EG-3306 (Ark Therapeutics), EXEL-2880 (Exelixis), GW- 654652 (GlaxoSmithKline), a KDR inhibitor from LG Life Sciences, CT-6685 and CT-6729 (UCB), KRN-633 and KRN-951 (Kirin Brewery), OSI-930 (OSI Pharmaceuticals), SP-5.2 (Supratek Pharma), SU-11657 (Pfizer), a Tie-2 antagonist (Hybrigenics), a VEGFR-2 kinase inhibitor (Bristol-Myers Squibb), XL-647 (Exelixis), a KDR inhibitor from Abbott Laboratories, sorafenib tosylate, and an analogue or derivative thereof.
16605. The method of item 16572 wherein the agent is an endotoxin antagonist.
16606. The method of item 16572 wherein the agent is an epothilone and tubulin binder.
16607. The method of item 16572 wherein the agent is an estrogen receptor antagonist.
16608. The method of item 16572 wherein the agent is an FGF inhibitor.
16609. The method of item 16572 wherein the agent is a farnexyl transferase inhibitor. 16610. The method of item 16572 wherein the agent is famesyltransferase inhibitor selected from the group of A-197574 (Abbott), a farnesyltransferase inhibitor from Servier, FPTIII (Strathclyde Institute for Drug R), LB-42908 (LG Life Sciences), Pharmaprojects No. 5063 (Genzyme), Pharmaprojects No. 5597 (Ipsen), Yissum Project No. B-1055 (Yissum), and an analogue or derivative thereof.
16611. The method of item 16572 wherein the agent is an FLT-3 kinase inhibitor.
16612. The method of item 16572 wherein the agent is an FGF receptor kinase inhibitor.
16613. The method of item 16572 wherein the agent is a fibrinogen antagonist selected from the group consisting of AUV-201 (Auvation), MG-13926 (Sanofi-Aventis), plasminogen activator (CAS No. 105913-11-9) (from Sanofi-Aventis or UCB), plasminogen activator-2 (tPA- 2) (Sanofi-Aventis), pro-urokinase (CAS No. 82657-92-9) (Sanofi-Aventis), and an analogue or derivative thereof.
16614. The method of item 16572 wherein the agent is a heat shock protein 90 antagonist selected from the group consisting of SRN- 005 (Sirenade), geldanamycin, NSC-33050 (17-allylaminogeldanamycin; 17- AAG), 17-dimethylaminoethylamino-17-demethoxy-geldanamycin (17- DMAG), rifabutin (rifamycin XIV, 1',4-didehydro-1-deoxy-1 ,4-dihydro-5'-(2- methylpropyl)-1-oxo-), and an analogue or derivative thereof.
16615. The method of item 16572 wherein the agent is a histone deacetylase inhibitor. 16616. The method of item 16572 wherein the agent is an HMGCoA reductase inhibitor selected from the group consisting of an atherosclerosis therapeutic from Lipid Sciences, ATI-16000 (ARYx Therapeutics), KS-01-019 (Kos Pharmaceuticals), Pharmaprojects No. 2197 (Sanofi-Aventi), RP 61969 (Sanofi-Aventis), and an analogue or derivative thereof.
16617. The method of item 16572 wherein the agent is an ICAM inhibitor.
16618. The method of item 16572 wherein the agent is an IL, ICE and IRAK antagonist, wherein the antagonist is a CJ-14877, CP- 424174 (Pfizer), NF-61 (Negma-Lerads), and an analogue or derivative thereof.
16619. The method of item 16572 wherein the agent is an IL-2 inhibitor.
16620. The method of item 16572 wherein the agent is an immunosuppressant selected from the group consisting of teriflunomide (Sanofi Aventis), chlorsulfaquinoxalone (NSC-339004), chlorsulfaquinoxalone sulfate, CS-712 (Sankyo), ismomultin alfa (CAS No. 457913-93-8) (Akzo Nobel), antiallergics from GenPat77, antiinflammatories or AT-005 (Androclus Therapeutics), autoimmune disease therapeutics from EpiVax, BN-007 (Bone), budesonide (CAS No. 51333-22- 3) (MAP Pharmaceuticals), CO-14 (Genzyme), edratide (CAS No. 433922- 67-9) (Teva), EP-314 (Enanta), eprovafen (CAS No. 101335-99-3) (Sanofi- Aventis), HWA-131 (CAS No. 118788-41-3) (Sanofi-Aventis), immunomodulators from MerLion Pharmaceuticals, immunosuppressives from Alchemia, IPL-12 (Inflazyme), MDL-9563 (CAS No. 27086-86-8) (Sanofi-Aventis), Pharmaprojects No. 2330 (Sanofi-Aventis), Pharmaprojects No. 6426 (Abgenix), PXS-25 (Pharmaxis), rosmarinic acid (CAS No. 20283-92-5) (Sanofi-Aventis), RP 42927 or RP 54745 (CAS No. 135330-08-4) (Sanofi-Aventis), SGN-35 (Seattle Genetics), ST-1959 (Sigma-Tau), type I diabetes therapy from SYNX Pharma, UNIL-88 (Debiopharm), VP-025 (Vasogen), VR-694 (Vectura), PRTX-001 (Protalex), and an analogue or derivative thereof.
16621. The method of item 16572 wherein the agent is an IMPDH (inosine monophosphate).
16622. The method of item 16572 wherein the agent is an integrin antagonist.
16623. The method of item 16572 wherein the agent is an interleukin antagonist.
16624. The method of item 16572 wherein the agent is an inhibitor of type III receptor tyrosine kinase.
16625. The method of item 16572 wherein the agent is an irreversible inhibitor of enzyme methionine aminopeptidase type 2.
16626. The method of item 16572 wherein the agent is an isozyme selective delta protein kinase C inhibitor.
16627. The method of item 16572 wherein the agent a JAK3 enzyme inhibitor.
16628. The method of item 16572 wherein the agent is a JNK inhibitor. 16629. The method of item 16572 wherein the agent is a kinase inhibitor.
16630. The method of item 16572 wherein the agent is kinesin antagonist.
16631. The method of item 16572 wherein the agent is a kinesin antagonist.
16632. The method of item 16572 wherein the agent is a leukotriene inhibitor and antagonist selected from the group consisting of ambicromil (CAS No. 58805-38-2) (Sanofi-Aventis), amelubant (CAS No. 346735-24-8) (Boehringer Ingelheim), DW-1141 (Dong Wha), ebselen (Daiichi Pharmaceutical), ibudilast (Kyorin), leucotriene inhibitors from Sanofi-Aventis, lymphotoxin-beta receptor (LT-D) from Biogen Idee, Pharmaprojects No. 1535 and 2728 (CAS No. 119340-33-9) (Sanofi- Aventis), R-112 (Rigel), Rev-5367 (CAS No. 92532-05-3) (Sanofi-Aventis), RG-14893 (CAS No. 141835-49-6) (Sanofi-Aventis), RG-5901-A (CAS No. 101910-24-1), 92532-23-5, RP 66153 (CAS No. 142422-79-5), RP 66364 (CAS No.186916-92-5), RP 69698 (CAS No. 141748-00-7) (Sanofi-Aventis), SC-411930 (Pfizer), SC-41930 (CAS No. 120072-59-5) (Pfizer), SC-50605 (CAS No. 138828-39-4) (Pfizer), SC-51146 (CAS No. 141059-52-1), SC- 53228 (CAS No. 153633-01-3) (Pfizer), spaglumic acid (ZY-15106) (CAS No.3106-85-2), 80619-64-3 (Novartis), tipredane (CAS No. 85197-77-9) (Bristol-Myers Squibb), U-75302 (CAS No. 119477-85-9) (Pfizer), and analogue or derivative thereof.
16633. The method of item 16572 wherein the agent is an MAP kinase inhibitor. 16634. The method of item 16572 wherein the agent is a matrix metalloproteinase inhibitor.
16635. The method of item 16572 wherein the agent is an MCP-CCR2 inhibitor.
16636. The method of item 16572 wherein the agent is an mTOR inhibitor.
16637. The method of item 16572 wherein the agent is an mTOR kinase inhibitor.
16638. The method of item 16572 wherein the agent is a microtubule inhibitor selected from the group consisting of antibody- maytansinoid conjugates from Biogen Idee, colchicines (MantiCore Pharmaceuticals), anticancer immunoconjugates from Johnson & Johnson, DIME from Octamer, gni-1f (GNI), huC242-DM4, huMy9-6-DM1 (ImmunoGen), IDN-5404 (Indena), IMO-098, IMOderm (Imotep), mebendazole (Introgen Therapeutics), microtubule poisons from Cambridge Enterprise, paclitaxel such as LOTAX from Aphios (CAS No. 33069-62-4), Genexol-PM from Samyang, Pharmaprojects No. 6383 (Tapestry Pharmaceuticals), RPR-112378 (Sanofi-Aventis), SGN-75 (Seattle Genetics), SPL-7435 (Starpharma), SSR-250411 (Sanofi-Aventis), trastuzumab-DM1 (Genentech), vinorelbine, and an analogue or derivative thereof.
16639. The method of item 16572 wherein the agent is an MIF inhibitor.
16640. The method of item 16572 wherein the agent is an MMP inhibitor. 16641. The method of item 16572 wherein the agent is a neurokinin (NK) antagonist selected from the group consisting of anthrotainin (CAS No. 148084-40-6) (Sanofi-Aventis), an IBS thereapeutic from ArQuIe, MDL-105212A (CAS No. 167261-60-1) (Ssanofi-Aventis), Pharmaprojects No. 2744, 3258 (CAS No. 139167-47-8) 4006, 4201 , or 5986 (Sanofi-Aventis), RP 67580 (CAS No. 135911-02-3), SR-144190 (CAS No. 201152-86-5), SSR-240600, SSR-241586 (Sanofi-Aventis), TKA-457 (Novartis), vestipitant mesylate (CAS No. 334476-64-1) (GlaxoSmithKline), Win-64821 (Sanofi-Aventis), PRX-96026 (Predix Pharmaceuticals), and an analogue or derivative thereof.
16642. The method of item 16572 wherein the agent is an NF kappa B inhibitor selected from the group consisting of AVE-0545 or AVE-0547 (Sanofi-Aventis), bortezomib (CAS No. 179324-69-7) (Millennium Pharmaceuticals), dexanabinol (CAS No. 112924-45-5) (Pharmos), dexlipotam (Viatris), Pharmaprojects No. 6283 (INDRA) (OXiGENE), IPL- 576092 (CAS No. 137571-30-3) (Inflazyme), NFKB decoy (Corgentech), NFKB decoy oligo (AnGes MG), NFKB's from Ariad, osteoporosis treatments or S5 (F005) from Fulcrum Pharmaceuticals, P61 (Phytopharm), R-flurbiprofen (CAS No. 5104-49-4) (Encore Pharmaceuticals), and an analogue or derivative thereof.
16643. The method of item 16572 wherein the agent is a nitric oxide agonist.
16644. The method of item 16572 wherein the agent is an ornithine decarboxylase inhibitor.
16645. The method of item 16572 wherein the agent is a p38 MAP kinase inhibitor selected from the group consisting of AZD-6703 (AstraZeneca), JX-401 (Jexys Pharmaceuticals), BMS-2 (Bristol-Myers Squibb), a p38 MAP kinase inhibitor from Novartis, a p38-alpha MAP kinase inhibitor from Amphora, Pharmaprojects No. 5704 (Pharmacopeia), RPR- 200765A (Sanofi-Aventis), SD-282 (Johnson & Johnson), TAK-715 (Takeda), and an analogue or derivative thereof.
16646. The method of item 16572 wherein the agent is a palmitoyl-protein thioesterase inhibitor.
16647. The method of item 16572 wherein the agent is a PDGF receptor kinase inhibitor selected from the group consisting of AAL- 993, AMN-107, or ABP-309 (Novartis), AMG-706 (Amgen), BAY-57-9352 (Bayer), CDP-860 (UCB), E-7080 (Eisai), imatinib (CAS No. 152459-95-5) (Novartis), OSI-930 (OSI Pharmaceuticals), RPR-127963E (Sanofi-Aventis), RWJ-540973 (Johnson & Johnson), sorafenib tosylate (Bayer), SU-11657 (Pfizer), tandutinib (CAS No. 387867-13-2) (Millennium Pharmaceuticals), vatalanib (Novartis), ZK-CDK (Schering AG), and an analogue or derivative thereof.
16648. The method of item 16572 wherein the agent is (-)-halofenate (Metabolex), AMG-131 (Amgen), antidiabetics from Japan Tobacco, AZD-4619, AZD-8450, AZD-8677 (AstraZeneca), DRF-10945, balaglitazone (Dr Reddy's), CS-00088, CS-00098 (Chipscreen Biosciences), E-3030 (Eisai), etalocib (CAS No. 161172-51-6) (EIi Lilly), GSK-641597 (Ligand), GSK-677954 (GlaxoSmithKline), GW-409544 (Ligand), GW- 590735 (GlaxoSmithKline), K-111 (Hoffmann-La Roche), LY-518674 (EIi Lilly), LY-674 (Ligand), LY-929 (Ligand), MC-3001, MC-3002 (MaxoCore Pharmaceuticals), metformin HCI + pioglitazone (CAS No. 1115-70-4 and
112529-15-4), ACTOPLUS MET from Andrx), muraglitazar (CAS No. 331741-94-7) (Bristol-Myers Squibb), naveglitazar (Ligand), oleoylethanolamide (Kadmus Pharmaceuticals), ONO-5129, pioglitazone hydrochloride (CAS No. 111025-46-8 and 112529-15-4) (Takeda), PLX-204 (Plexxikon), PPAR agonists from Genfit, PPAR delta agonists from EIi Lilly, PPAR-alpha agonists from CrystalGenomics, PPAR-gamma modulators and PPAR-β modulators from CareX, rosiglitazone maleate (CAS No. 122324- 73-4 or 155141-29-0) (GlaxoSmithKline), rosiglitazone maleate/glimepir (CAS No. 155141-29-0 and 93479-97-1), AVANDARYL, rosiglitazone maleate/metformin extend (CAS No. 155141-29-0 and 657-24-9), AVANDAMET, rosiglitazone maleate+metformin, AVANDAMET (GlaxoSmithKline), tesaglitazar (AstraZeneca), and an analogue or derivative thereof.
16649. The method of item 16572 wherein the agent is a phosphatase inhibitor.
16650. The method of item 16572 wherein the agent is a phosphodiesterase (PDE) inhibitor selected from the group consisting of avanafil (Tanabe Seiyaku), dasantafil (CAS No. 569351-91-3) (Schering- Plough), A-906119 (CAS No. 134072-58-5), DL-850 (Sanofi-Aventis), GRC- 3015, GRC-3566, GRC-3886 (Glenmark), HWA-153 (CAS No. 56395-66-5) (Sanofi-Aventis), hydroxypumafentrine (Altana), IBFB-130011, IBFB-14-016, IBFB-140301 , IBFB-150007, IBFB-211913 (IBFB Pharma), L-826141 (Merck & Co), medorinone (CAS No. 88296-61-1) (Sanofi-Aventis), MEM- 1917 (Memory Pharmaceuticals), ND-1251 (Neuro3d), PDE inhibitors from ICOS, PDE IV inhibitors from Memory Pharmaceuticals and CrystalGenomics, Pharmaprojects No. 2742 and 6141 (Sanofi-Aventis), QAD-171 (Novartis), RHC-2963 (CAS No. 76993-12-9 and 76993-14-1), RPR-117658, RPR-122818 derivatives, SR-24870 , and RPR-132294 (Sanofi-Aventis), SK-350 (ln2Gen), stroke therapy agents from deCODE Genetics, TAS-203 (Taiho), tofimilast (CAS No. 185954-27-2) (Pfizer), UK- 371800 (Pfizer), WIN-65579 (CAS No. 158020-82-7) (Sanofi-Aventis), IBFB- 130020 (IBFB Pharma), OPC-6535 (CAS No. 145739-56-6) (Otsuka), a phosphodiesterase III inhibitor, enoximone, a phosphodiesterase IV inhibitor, fosfosal, Atopik (Barrier Therapeutics), triflusal, a phosphodiesterase V inhibitor, and an analogue or derivative thereof.
16651. The method of item 16572 wherein the agent is a PKC inhibitor.
16652. The method of item 16572 wherein the agent is a platelet activating factor antagonist.
16653. The method of item 16572 wherein the agent is a platelet-derived growth factor receptor kinase inhibitor.
16654. The method of item 16572 wherein the agent is a prolyl hydroxylase inhibitor.
16655. The method of item 16572 wherein the agent is a polymorphonuclear neutrophil inhibitor.
16656. The method of item 16572 wherein the agent is a protein kinase B inhibitor.
16657. The method of item 16572 wherein the agent is a protein kinase C stimulant.
16658. The method of item 16572 wherein the agent is a purine nucleoside analogue.
16659. The method of item 16572 wherein the agent is a purinoreceptor P2X antagonist. 16660. The method of item 16572 wherein the agent is a Raf kinase inhibitor.
16661. The method of item 16572 wherein the agent is a reversible inhibitor of ErbB1 and ErbB2.
16662. The method of item 16572 wherein the agent is a ribonucleoside triphosphate reductase inhibitor.
16663. The method of item 16572 wherein the agent is an SDF-1 antagonist.
16664. The method of item 16572 wherein the agent is a sheddase inhibitor.
16665. The method of item 16572 wherein the agent is an SRC inhibitor.
16666. The method of item 16572 wherein the agent is a stromelysin inhibitor.
16667. The method of item 16572 wherein the agent is an Syk kinase inhibitor.
16668. The method of item 16572 wherein the agent is a telomerase inhibitor.
16669. The method of item 16572 wherein the agent is a TGF beta inhibitor selected from the group consisting of pirfenidone (CAS No. 53179-13-8) (MARNAC), tranϋast (CAS No. 53902-12-8) (Kissei), IN- 1130 (ln2Gen), mannose-6-phosphate (BTG), TGF-β antagonists from Inflazyme (Pharmaprojects No. 6075), TGF-β antagonists from Sydney, non-industrial source), TGF-β I receptor kinase inhibitors from EIi Lilly, TGF- β receptor inhibitors from Johnson & Johnson, and an analogue or derivative thereof.
16670. The method of item 16572 wherein the agent is a
TNFα antagonist or TACE inhibitor selected from the group consisting of adalimumab (CAS No. 331731-18-1) (Cambridge Antibody Technology), AGIX-4207 (AtheroGenics), AGT-1 (Advanced Biotherapy), an antiinflammatory from Borean Pharma, Cellzome, or Paradigm Therapeutics, anti-inflammatory vaccine (TNF-alpha kinoid) from Neovacs, humanized anti-TNF antibody or an anti-TNF MAb (CB0006) Celltech (UCB), apratastat (CAS No. 287445-51-0) (Wyeth), BMS-561392 (Bristol-Myers Squibb), BN- 006 (Bone), certolizumab pegol (CAS No. 428863-50-7 or CH-138 (UCB), cilomilast (CAS No. 153259-65-5) (GlaxoSmithKline), CR-1 (Nuada Pharmaceuticals), CRx-119 (CombinatoRx), D-5410 (UCB), dacopafant (CAS No. 125372-33-0) (Sanofi-Aventis), dersalazine (CAS No. 188913-57- 7/188913-58-8) (Uriach), etanercept (CAS No. 185243-69-0) (Amgen), ethyl pyruvate (Critical (Critical Therapeutics), golimumab (CAS No. 476181-74-5) (Johnson & Johnson), hormono-immunotherapy from Ipsen, CDP571 (e.g., Humicade from UCB), IC-485 (ICOS), infliximab (CAS No. 170277-31-3) (Johnson & Johnson), IP-751 (Manhattan Pharmaceuticals), ISIS-104838 (CAS No. 250755-32-9) (ISIS Pharmaceuticals), lenalidomide (CAS No. 191732-72-6) (Celgene), lentinan (CAS No. 37339-90-5) (Ajinomoto), MDL- 201112 (CAS No. 142130-73-2) (Sanofi-Aventis), medroxyprogesterone (CAS No. 520-85-4) (InKine Pharmaceutical), N-acetylcysteine (CAS No. 616-91-1) (Zambon), NBE-P2 (DIREVO Biotech), nerelimomab (CAS No. 162774-06-3) (Chiron), OM-294DP (OM PHARMA), onercept (CAS No. 199685-57-9) (Yeda), PASSTNF-alpha (Verigen), pentoxifylline or oxypentifylline (Sanofi-Aventis), Pharmaprojects No. 4091 , 4241, 4295, or 4488 (Sanofi-Aventis), Pharmaprojects No. 5480 (Amgen), Pharmaprojects No. 6749 (Cengent), pirfenidone (CAS No. 53179-13-8) (MARNAC), RPR- 132294 (Sanofi-Aventis), S5 (F002) (Fulcrum Pharmaceuticals), simvastatin (CAS No. 79902-63-9) (Merck & Co), STA-6292 (Synta Pharmaceuticals), tacrolimus (CAS No. 104987-11-3) (Fujisawa LifeCycle Pharma), talactoferrin alfa (CAS No. 308240-58-6) (Agennix), thalidomide (CAS No. 50-35-1) (Celgene), TNF antagonists form ProStrakan, and Synergen, TNF inhibitors (Amgen), TNF-alpha antagonists from Dynavax Technologies and Jerina AG (Germany), TNF-alpha inhibitors from IBFB Pharma and Xencor (Xencor), torbafylline (CAS No. 105102-21-4) (Sanofi-Aventis), UR-1505 (Uriach), VT-346 (Viron Therapeutics), YSIL6 (Y's Therapeutics), YSTH2 (Y's Therapeutics), NPI-1302a-3 (Nereus Pharmaceuticals, a TNF antagonist from Jerina AG (Germany), dersalazine, and an analogue or derivative thereof.
16671. The method of item 16572 wherein the agent is a Toll receptor inhibitor.
16672. The method of item 16572 wherein the agent is a tubulin antagonist.
16673. The method of item 16572 wherein the agent is a tyrosine kinase inhibitor selected from the group consisting of SU-011248, SUTENT from Pfizer Inc. (New York, NY), BMS-354825, PN-355 (Paracelsian Pharmaceuticals), AGN-199659 (Allergan), AAL-993 or ABP- 309 (Novartis), adaphostin (NIH), AEE-788 (Novartis), AG-013736 (OSI Pharmaceuticals), AG-13736 (Pfizer), ALT-110 (Alteris Therapeutics), AMG- 706 (Amgen), anticancer MAbs from Xencor, anti-EGFrvlll MAbs from Abgenix, anti-HER2 MAb from Abiogen, AZD-2171 or AZD-9935 (AstraZeneca), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), CEP-5214 (Cephalon), CEP-7055 (Cephalon), cetuximab (ImClone Systems), CHIR-200131 and CHIR-258 (Chiron), CP-547632 (OSI Pharmaceuticals), CP-724714 (Pfizer), CT-301 (Creabilis Therapeutics), D- 69491 (Baxter International), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EGFR/ErbB2 inhibitors from Array BioPharma, erlotinib (CAS No. 183319- 69-9) (OSI Pharmaceuticals), EXEL-2880 (Exelixis), FK-778 (Sanofi- Aventis) , gefitinib (CAS No. 184475-35-2) (AstraZeneca), GW-2286 or GW- 654652 (GlaxoSmithKline), her2/neu antigen from AlphaVax, HER-2/neu inhibitor from Generex, Herzyme (Medipad) (Sirna Therapeutics), HKI-272 (Wyeth), HuMax-EGFr (Genmab), idronoxil (CAS No. 81267-65-4) (Novogen), IGF-1 inhibitors from Ontogen, IMC-11F8 (ImCIone Systems), kahalalide F (CAS No. 149204-42-2) (PharmaMar), KDR inhibitor from LG Life Sciences, KDR inhibitors from Abbott Laboratories, KDR kinase inhibitors (UCB), Kdr kinase inhibitors from Merck & Co, KRN-633 and KRN- 951 (Kirin Brewery), KSB-102 (Xenova), lapatinib ditosylate (CAS No. 388082-78-8) (GlaxoSmithKline), matuzumab (Merck KGaA), MDX-214 (Medarex), ME-103 (Pharmexa), MED-A300 (Gerolymatos), MNAC-13 (Lay Line Genomics), nimotuzumab (Center of Molecular Immunology), NSC- 330507 or NSC-707545 (NIH), NV-50 (Novogen), OSI-930 (OSI Pharmaceuticals), panitumumab (Abgenix), pelitinib (CAS No. 287933-82-7) (Wyeth), pertuzumab (CAS No. 380610-27-5) (Genentech), Pharmaprojects No. 3985 (Sanofi-Aventis), prostate cancer therapeutics from Sequenom (SQPC35, SQPC36, SQPC90), removab and remoxab (Trion Pharma), RG- 13022 (CAS No. 136831-48-6), RG-13291 (CAS No. 138989-50-1), or RG- 14620 (CAS No. 136831-49-7) (Sanofi-Aventis), RM-6427 (Romark), RNAi breast cancer therapy from Benitec, RP 53801 (CAS No. 125882-88-4) (Sanofi-Aventis), sorafenib tosylate (Bayer), SU-11657 (Pfizer), Tie-2 antagonists from Semaia (Hybrigenics), Tie-2 inhibitors from Ontogen, trastuzumab (CAS No. 180288-69-1) (Genentech), tyrosine kinase inhibitors from Sanofi-Aventis, U3-1287, U3-1565, U3-1784, U3-1800 (U3 Pharma), vatalanib (Novartis), VEGFR-2 kinase inhibitor from Bristol-Myers Squibb, XL-647 (Exelixis), ZD-6474 (AstraZeneca), ZK-CDK (Schering AG), an EGFR tyrosine kinase inhibitor, EKB-569 (Wyeth), and an analogue or derivative thereof.
16674. The method of item 16572 wherein the agent is a VEGF inhibitor.
16675. The method of item 16572 wherein the agent is a vitamin D receptor agonist.
16676. The method of item 16572 wherein the agent is ZD-6474 (an angiogenesis inhibitor).
16677. The method of item 16572 wherein the agent is AP-23573 (an mTOR inhibitor).
16678. The method of item 16572 wherein the agent is synthadotin (a tubulin antagonist).
16679. The method of item 16572 wherein the agent is S-0885 (a collagenase inhibitor).
16680. The method of item 16572 wherein the agent is aplidine (an elongation factor-1 alpha inhibitor).
16681. The method of item 16572 wherein the agent is ixabepilone (an epithilone).
16682. The method of item 16572 wherein the agent is IDN-5390 (an angiogenesis inhibitor). 16683. The method of item 16572 wherein the agent is SB-2723005 (an angiogenesis inhibitor).
16684. The method of item 16572 wherein the agent is ABT-518 (an angiogenesis inhibitor).
16685. The method of item 16572 wherein the agent is combretastatin (an angiogenesis inhibitor).
16686. The method of item 16572 wherein the agent is anecortave acetate (an angiogenesis inhibitor).
16687. The method of item 16572 wherein the agent is SB-715992 (a kinesin antagonist).
16688. The method of item 16572 wherein the agent is temsirolimus (an mTOR inhibitor).
16689. The method of item 16572 wherein the agent is adalimumab (a TNFα antagonist).
16690. The method of item 16572, wherein the composition comprises a polymer.
16691. The method of item 16572, wherein the composition comprises a polymeric carrier.
16692. The method of item 16572 wherein the anti- scarring agent inhibits adhesion between the device and a host into which the device is implanted. 16693. The method of item 16572 wherein the device delivers the anti-scarring agent locally to tissue proximate to the device.
16694. The method of item 16572 wherein the device has a coating that comprises the anti-scarring agent.
16695. The method of item 16572, wherein the device has a coating that comprises the agent and is disposed on a surface of the implant.
16696. The method of item 16572, wherein the device has a coating that comprises the agent and directly contacts the implant.
16697. The method of item 16572, wherein the device has a coating that comprises the agent and indirectly contacts the implant.
16698. The method of item 16572, wherein the device has a coating that comprises the agent and partially covers the implant.
16699. The method of item 16572, wherein the device has a coating that comprises the agent and completely covers the implant.
16700. The method of item 16572, wherein the device has a uniform coating.
16701. The method of item 16572, wherein the device has a non-uniform coating.
16702. The method of item 16572, wherein the device has a discontinuous coating. 16703. The method of item 16572, wherein the device has a patterned coating.
16704. The method of item 16572, wherein the device has a coating with a thickness of 100 Dm or less.
16705. The method of item 16572, wherein the device has a coating with a thickness of 10 Dm or less.
16706. The method of item 16572, wherein the device has a coating, and the coating adheres to the surface of the implant upon deployment of the implant.
16707. The method of item 16572, wherein the device has a coating, and wherein the coating is stable at room temperature for a period of 1 year.
16708. The method of item 16572, wherein the device has a coating, and wherein the anti-scarring agent is present in the coating in an amount ranging between about 0.0001% to about 1% by weight.
16709. The method of item 16572, wherein the device has a coating, and wherein the anti-scarring agent is present in the coating in an amount ranging between about 1 % to about 10% by weight.
16710. The method of item 16572, wherein the device has a coating, and wherein the anti-scarring agent is present in the coating in an amount ranging between about 10% to about 25% by weight. 16711. The method of item 16572, wherein the device has a coating, and wherein the anti-scarring agent is present in the coating in an amount ranging between about 25% to about 70% by weight.
16712. The method of item 16572, wherein the device has a coating, and wherein the coating further comprises a polymer.
16713. The method of item 16572, wherein the device has a first coating having a first composition and a second coating having a second composition.
16714. The method of item 16572, wherein the device has a first coating having a first composition and a second coating having a second composition, wherein the first composition and the second composition are different.
16715. The method of item 16572, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a copolymer.
16716. The method of item 16572, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a block copolymer.
16717. The method of item 16572, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a random copolymer.
16718. The method of item 16572, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a biodegradable polymer. 16719. The method of item 16572, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a non-biodegradable polymer.
16720. The method of item 16572, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a hydrophilic polymer.
16721. The method of item 16572, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a hydrophobic polymer.
16722. The method of item 16572, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a polymer having hydrophilic domains.
16723. The method of item 16572, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a polymer having hydrophobic domains.
16724. The method of item 16572, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a non-conductive polymer.
16725. The method of item 16572, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises an elastomer.
16726. The method of item 16572, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a hydrogel. 16727. The method of item 16572, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a silicone polymer.
16728. The method of item 16572, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a hydrocarbon polymer.
16729. The method of item 16572, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a styrene-derived polymer.
16730. The method of item 16572, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a butadiene polymer.
16731. The method of item 16572, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a macromer.
16732. The method of item 16572, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a poly(ethylene glycol) polymer.
16733. The method of item 16572 wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises an amorphous polymer.
16734. The method of item 16572, wherein the device comprises a lubricious coating. 16735. The method of item 16572 wherein the anti- scarring agent is located within pores or holes of the device.
16736. The method of item 16572 wherein the anti- scarring agent is located within a channel, lumen, or divet of the device.
16737. The method of item 16572, wherein the device comprises a second pharmaceutically active agent.
16738. The method of item 16572 wherein the device comprises an anti-inflammatory agent.
16739. The method of item 16572 wherein the device comprises an agent that inhibits infection.
16740. The method of item 16572 wherein the device comprises an agent that inhibits infection, and wherein the agent is an anthracycline.
16741. The method of item 16572 wherein the device comprises an agent that inhibits infection, and wherein the agent is doxorubicin.
16742. The method of item 16572 wherein the device comprises an agent that inhibits infection, and wherein the agent is mitoxantrone.
16743. The method of item 16572 wherein the device comprises an agent that inhibits infection, and wherein the agent is a fluoropyrimidine. 16744. The method of item 16572 wherein the device comprises an agent that inhibits infection, and wherein the agent is 5- fluorouracil (5-FU).
16745. The method of item 16572 wherein the device comprises an agent that inhibits infection, and wherein the agent is a folic acid antagonist.
16746. The method of item 16572 wherein the device comprises an agent that inhibits infection, and wherein the agent is methotrexate.
16747. The method of item 16572 wherein the device comprises an agent that inhibits infection, and wherein the agent is a podophylotoxin.
16748. The method of item 16572 wherein the device comprises an agent that inhibits infection, and wherein the agent is etoposide.
16749. The method of item 16572 wherein the device comprises an agent that inhibits infection, and wherein the agent is a camptothecin.
16750. The method of item 16572 wherein the device comprises an agent that inhibits infection, and wherein the agent is a hydroxyurea.
16751. The method of item 16572 wherein the device comprises an agent that inhibits infection, and wherein the agent is a platinum complex. 16752. The method of item 16572 wherein the device comprises an agent that inhibits infection, and wherein the agent is cisplatin.
16753. The method of item 16572, further comprising an anti-thrombotic agent.
16754. The method of item 16572 wherein the device comprises a visualization agent.
16755. The method of item 16572 wherein the device comprises a visualization agent, wherein the visualization agent is a radiopaque material, and wherein the radiopaque material comprises a metal, a halogenated compound, or a barium containing compound.
16756. The method of item 16572 wherein the device comprises a visualization agent, wherein the visualization agent is a radiopaque material, and wherein the radiopaque material comprises barium, tantalum, or technetium.
16757. The method of item 16572 wherein the device comprises a visualization agent, and wherein the visualization agent is a MRI responsive material.
16758. The method of item 16572 wherein the device comprises a visualization agent, and wherein the visualization agent comprises a gadolinium chelate.
16759. The method of item 16572 wherein the device comprises a visualization agent, and wherein the visualization agent comprises iron, magnesium, manganese, copper, or chromium. 16760. The method of item 16572 wherein the device comprises a visualization agent, and wherein the visualization agent comprises an iron oxide compound.
16761. The method of item 16572 wherein the device comprises a visualization agent, and wherein the visualization agent comprises a dye, pigment, or colorant.
16762. The method of item 16572 wherein the device comprises an echogenic material.
16763. The method of item 16572 wherein the device comprises an echogenic material, and wherein the echogenic material is in the form of a coating.
16764. The method of item 16572 wherein the device is sterile.
16765. The method of item 16572 wherein the anti- scarring agent is released into tissue in the vicinity of the device after deployment of the device.
16766. The method of item 16572 wherein the anti- scarring agent is released into tissue in the vicinity of the device after deployment of the device, and wherein the tissue is connective tissue.
16767. The method of item 16572 wherein the anti- scarring agent is released into tissue in the vicinity of the device after deployment of the device, and wherein the tissue is muscle tissue. 16768. The method of item 16572 wherein the anti- scarring agent is released into tissue in the vicinity of the device after deployment of the device, and wherein the tissue is nerve tissue.
16769. The method of item 16572 wherein the anti- scarring agent is released into tissue in the vicinity of the device after deployment of the device, and wherein the tissue is epithelium tissue.
16770. The method of item 16572 wherein the anti- scarring agent is released in effective concentrations from the device over a period ranging from the time of deployment of the device to about 1 year.
16771. The method of item 16572 wherein the anti- scarring agent is released in effective concentrations from the device over a period ranging from about 1 month to 6 months.
16772. The method of item 16572 wherein the anti- scarring agent is released in effective concentrations from the device over a period ranging from about 1 - 90 days.
16773. The method of item 16572 wherein the anti- scarring agent is released in effective concentrations from the device at a constant rate.
16774. The method of item 16572 wherein the anti- scarring agent is released in effective concentrations from the device at an increasing rate.
16775. The method of item 16572 wherein the anti- scarring agent is released in effective concentrations from the device at a decreasing rate. 16776. The method of item 16572 wherein the anti- scarring agent is released in effective concentrations from the composition comprising the anti-scarring agent by diffusion over a period ranging from the time of deployment of the device to about 90 days.
16777. The method of item 16572 wherein the anti- scarring agent is released in effective concentrations from the composition comprising the anti-scarring agent by erosion of the composition over a period ranging from the time of deployment of the device to about 90 days.
16778. The method of item 16572 wherein the device comprises about 0.01 Dg to about 10 Dg of the anti-scarring agent.
16779. The method of item 16572 wherein the device comprises about 10 Dg to about 10 mg of the anti-scarring agent.
16780. The method of item 16572 wherein the device comprises about 10 mg to about 250 mg of the anti-scarring agent.
16781. The method of item 16572 wherein the device comprises about 250 mg to about 1000 mg of the anti-scarring agent.
16782. The method of item 16572 wherein the device comprises about 1000 mg to about 2500 mg of the anti-scarring agent.
16783. The method of item 16572 wherein a surface of the device comprises less than 0.01 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied. 16784. The method of item 16572 wherein a surface of the device comprises about 0.01 Dg to about 1 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
16785. The method of item 16572 wherein a surface of the device comprises about 1 Dg to about 10 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
16786. The method of item 16572 wherein a surface of the device comprises about 10 Dg to about 250 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
16787. The method of item 16572 wherein a surface of the device comprises about 250 Dg to about 1000 Dg of the anti-scarring agent of anti-scarring agent per mm2 of device surface to which the anti- scarring agent is applied.
16788. The method of item 16572 wherein a surface of the device comprises about 1000 Dg to about 2500 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
16789. The method of item 16572 wherein the combining is performed by direct affixing the agent or the composition to the implant.
16790. The method of item 16572 wherein the combining is performed by spraying the agent or the component onto the implant. 16791. The method of item 16572 wherein the combining is performed by electrospraying the agent or the composition onto the implant.
16792. The method of item 16572 wherein the combining is performed by dipping the implant into a solution comprising the agent or the composition.
16793. The method of item 16572 wherein the combining is performed by covalently attaching the agent or the composition to the implant.
16794. The method of item 16572 wherein the combining is performed by non-covalently attaching the agent or the composition to the implant.
16795. The method of item 16572 wherein the combining is performed by coating the implant with a substance that contains the agent or the composition.
16796. The method of item 16572 wherein the combining is performed by coating the implant with a substance that absorbs the agent.
16797. The method of item 16572 wherein the combining is performed by interweaving a thread composed of, or coated with, the agent or the composition.
16798. The method of item 16572 wherein the combining is performed by covering all the implant with a sleeve that contains the agent or the composition. 16799. The method of item 16572 wherein the combining is performed by covering a portion of the implant with a sleeve that contains the agent or the composition.
16800. The method of item 16572 wherein the combining is performed by covering all the implant with a cover that contains the agent or the composition.
16801. The method of item 16572 wherein the combining is performed by covering a portion of the implant with a cover that contains the agent or the composition.
16802. The method of item 16572 wherein the combining is performed by covering all the implant with an electrospun fabric that contains the agent or the composition.
16803. The method of item 16572 wherein the combining is performed by covering a portion of the implant with an electrospun fabric that contains the agent or the composition.
16804. The method of item 16572 wherein the combining is performed by covering all the implant with a mesh that contains the agent or the composition.
16805. The method of item 16572 wherein the combining is performed by covering a portion of the implant with a mesh that contains the agent or the composition.
16806. The method of item 16572 wherein the combining is performed by constructing all the implant with the agent or the composition. 16807. The method of item 16572 wherein the combining is performed by constructing a portion of the implant with the agent or the composition.
16808. The method of item 16572 wherein the combining is performed by impregnating the implant with the agent or the composition.
16809. The method of item 16572 wherein the combining is performed by constructing all of the implant from a degradable polymer that releases the agent.
16810. The method of item 16572 wherein the combining is performed by constructing a portion of the implant from a degradable polymer that releases the agent.
16811. The method of item 16572 wherein the combining is performed by dipping the implant into a solution that comprise the agent and an inert solvent for the implant.
16812. The method of item 16572 wherein the combining is performed by dipping the implant into a solution that comprises the agent and a solvent that will swell the implant.
16813. The method of item 16572 wherein the combining is performed by dipping the implant into a solution that comprises the agent and a solvent that will dissolve the implant.
16814. The method of item 16572 wherein the combining is performed by dipping the implant into a solution that comprises the agent, a polymer and an inert solvent for the implant. 16815. The method of item 16572 wherein the combining is performed by dipping the implant into a solution that comprises the agent, a polymer and a solvent that will swell the implant.
16816. The method of item 16572 wherein the combining is performed by dipping the implant into a solution that comprises the agent, a polymer and a solvent that will dissolve the implant.
16817. The method of item 16572 wherein the combining is performed by spraying the implant into a solution that comprises the agent and an inert solvent for the implant.
16818. The method of item 16572 wherein the combining is performed by spraying the implant into a solution that comprises the agent and a solvent that will swell the implant.
16819. The method of item 16572 wherein the combining is performed by spraying the implant into a solution that comprises the agent and a solvent that will dissolve the implant.
16820. The method of item 16572 wherein the combining is performed by spraying the implant into a solution that comprises the agent, a polymer and an inert solvent for the implant.
16821. The method of item 16572 wherein the combining is performed by spraying the implant into a solution that comprises the agent, a polymer and a solvent that will swell the implant.
16822. The method of item 16572 wherein the combining is performed by spraying the implant into a solution that comprises the agent, a polymer and a solvent that will dissolve the implant. 16823. A method as in any one of items 16572-16822, wherein the device is a left ventricular assist device.
16824. A method as in any one of items 16572-16822, wherein the device is a right ventricular assist device.
16825. A method as in any one of items 16572-16822, wherein the device is a biventricular assist device.
16826. A method as in any one of items 16572-16822, wherein the device is a cardiac assist device.
16827. A method as in any one of items 16572-16822, wherein the device is a mechanical assist device.
16828. A method as in any one of items 16572-16822, wherein the device is an artificial cardiac assist device.
16829. A method as in any one of items 16572-16822, wherein the device is an implantable heart assist system.
16830. A method as in any one of items 16572-16822, wherein the device is a heart assist pump.
16831. A method as in any one of items 16572-16822, wherein the device is an intra-ventricular cardiac assist device.
16832. A method of making a medical device comprising: combining a spinal implant and an anti-scarring agent or a composition comprising an anti-scarring agent, wherein the agent inhibits scarring between the device and a host into which the device is implanted. 16833. The method of item 16832 wherein the agent is an adensosine A2A receptor antagonist.
16834. The method of item 16832 wherein the agent is an AKT inhibitor.
16835. The method of item 16832 wherein the agent is an alpha 2 integrin antagonist, wherein the alpha 2 integrin antagonist is Pharmaprojects No. 5754 (Merck KgaA).
16836. The method of item 16832 wherein the agent is an alpha 4 integrin antagonist.
16837. The method of item 16832 wherein the agent is an alpha 7 nicotinic receptor agonist.
16838. The method of item 16832 wherein the agent is an angiogenesis inhibitor selected from the group consisting of AG-12,958 (Pfizer), ATN-161 (Attention LLC)1 neovastat, an angiogenesis inhibitor from Jerina AG (Germany), NM-3 (Mercian), VGA-1155 (Taisho), FCE-26644 (Pfizer), FCE-26950 (Pfizer), FPMA (Meiji Daries), FR-111142 (Fujisawa), GGTI-298, GM-1306 (Ligand), GPA-1734 (Novartis), NNC-47-0011 (Novo Nordisk), herbamycin (Nippon Kayaku), lenalidomide (Celegene), IP-10 (NIH), ABT-828 (Abbott), KIN-841 (Tokushima University, Japan), SF-1126 (Semafore Pharmaceuticals), laminin technology (NIH), CHIR-258 (Chiron), NVP-AEW541 (Novartis), NVP-AEW541 (Novartis), Vt16907 (Alchemia), OXI-8007 (Oxigene), EG-3306 (Ark Therapeutics), Maspin (Arriva), ABT- 567 (Abbott), PPI-2458 (Praecis Pharmaceuticals), CC-5079, CC-4089 (Celgene), HIF-1 alpha inhibitors (Xenova), S-247 (Pfizer), AP-23573 (Ariad), AZD-9935 (Astra Zeneca), mebendazole (Introgen Therapeutics), MetAP-2 inhibitors (GlaxoSmithKline), AG-615 (Angiogene Pharmaceuticals), Tie-2 antagonists (Hybrigenics), NC-381 , CYC-381 , NC-169, NC-219, NC-383, NC-384, NC-407 (Lorus Therapeutics), ATN-224 (Attention), ON-01370 (Onconova), Vitronectin antagonists (Amgen), SDX-103 (Salmedix), Vitronectin antagonists (Shire), CHP (Riemser), TEK (Amgen), Anecortave acetate (Alcon), T46.2 (Matrix Therapeutics), HG-2 (Heptagen), TEM antagonists (Genzyme), Oxi-4500 (Oxigene), ATN-161 (Attenuon), WX-293 (Wilex), M-2025 (Metris Therapeutics), Alphastatin (BioActa), YH-16 (Yantai Rongchang), BIBF-1120 (Boehringer Ingelheim), BAY-57-9352 (Bayer), AS- 1404 (Cancer Research Technology), SC-77964 (Pfizer), glycomimetics (BioTie Therapies), TIE-2 Inhibitors (Ontogen), DIMI, Octamer (Octamer), ABR-215050 (Active Biotech), ABT-518 (Abbott), KDR inhibitors (Abbott), BSF-466895 (Abbott), SCH-221153 (Schering-Plough), DAC:antiangiogenic (ConjuChem), TFPI (EntreMed), AZD-2171 (Astra-Zenaca), CDC-394 (Celgene), LY290293 (EIi Lilly), IDN-5390 (Indena), Kdr Kinase Inhibitors (Merck), CT-113020, CT-116433, CT-116563, CT-31890, CT-32228) (Cell Therapeutics), A-299620 (Abbott), TWEAK Inhibitor (Amgen), VEGF modulators (Johnson and Johnson), Tum-N53, tumstatin (Genzyme), Thios- 1 , Thios-2 (Thios Pharmaceuticals), MV-6401 (Miravant Medical Technologies), Spisulosine (PharmaMar), CEP-7055 (Cephalon), AUV-201 (Auvation), LM-609 (EIi Lilly), SKF-106615 (AnorMED), Oglufanide disodium (Cytran), BW-114 (Pharminox), Calreticulin (NIH), WX-678 (Wilex), SD-7784 (Pfizer), WX-UK1 (Wilex), SH-268 (Schering AG), 2-Me-PGA (Celgene), S- 137 (Pfizer), ZD-6126 (Angiogene Pharmaceuticals), SG-292 (SignalGen), Benefin (Lane Labs), A6, A36 (Angstrom), SB-2723005 (GlaxoSmithKline), SC-7 (Cell Therapeutics), ZEN-014 (AEterna Zentaris), 2-methoxyestradiol (EntreMed), NK-130119 (Nippon Kayaku), CC-10004 (Celgene), AVE- 8062A (Ajinomoto), Tacedinaline (Pfizer), Actinonin (Tokyo Metropolitan Institute of Medical Science), Lenalidomide (Celgene), VGA-1155, BTO-956 (SRI International), ER-68203-00 (Eisai), CT-6685 (UCB), JKC-362 (Phoenix Pharmaceuticals), DMI-3798 (DMI Biosciences, Angiomate (Ipsen), ZD-6474 (AstraZeneca), CEP-5214 (Cephalon), Canstatin (Genzyme), NM-3 (Mercian), Oridigm (MediQuest Therapeutics), Exherin (Adherex), BLS-0597 (Boston Life Sciences), PTC-299 (PTC Therapeutics), NPI-2358 (Nereus Pharmaceuticals), CGP-79787 (Novartis), AEE-788 (Novartis), CKD-732 (Chong Kun Dang), CP-564959 (OSI Pharmaceuticals), CM-101 (CarboMed), CT-2584, CT3501 (Cell Therapeutics), combretastatin and analogues and derivatives thereof (Oxigene), Rebimastat (Bristol-Meyers Squibb), Dextrin 2-sulfate (ML Laboratories), Cilengitide (Merk KGaA), NSC-706704 (Pharminox), KRN- 951 (Kirin Brewery), Ukrain, NSC-631570 (Nowicky Pharma), Tecogalan sodium (Daiichi Pharmaceutical), Tz-93 (Tsumura), TBC-1635 (Encysive Pharmaceuticals), TAN-1120 (Takeda), Semaxanib (Pfizer), BDI-7800 (Biopharmacopae), SD-186, SD-983 (Bristol-Meyers Squibb), SB-223245 (GlaxoSmithKline), SC-236 (Pfizer), RWJ-590973 (Johnson and Johnson), ILX-1850 (Genzyme), SC-68488, S-836 (Pfizer), CG-55069-11 (CuraGen), Ki-23057 (Kirin Brewery), CCX-700 (Chemoentryx), Pegaptanib octasodium (Giled Sciences), ANGIOCOL (available from Biostratum Inc.), or an analogue or derivative thereof.
16839. The method of item 16832 wherein the agent is an apoptosis antagonist.
16840. The method of item 16832 wherein the agent is an apoptosis activator.
16841. The method of item 16832 wherein the agent is a beta 1 integrin antagonist.
16842. The method of item 16832 wherein the agent is a beta tubulin inhibitor. 16843. The method of item 16832 wherein the agent is a blocker of enzyme production in Hepatitis C.
16844. The method of item 16832 wherein the agent is a Bruton's tyrosine kinase inhibitor.
16845. The method of item 16832 wherein the agent is a calcineurin inhibitor.
16846. The method of item 16832 wherein the agent is a caspase 3 inhibitor.
16847. The method of item 16832 wherein the agent is a CC chemokine receptor antagonist.
16848. The method of item 16832 wherein the agent is a cell cycle inhibitor selected from the group consisting of SNS-595 (Sunesis), synthadotin, KRX-0403, and an analogue or derivative thereof.
16849. The method of item 16832 wherein the agent is a cathepsin B inhibitor.
16850. The method of item 16832 wherein the agent is a cathepsin K inhibitor, wherein the cathepsin K inhibitor is 462795 (GlaxoSmithKline), INPL-022-D6 (Amura Therapeutics), or an analogue or derivative thereof.
16851. The method of item 16832 wherein the agent is a cathepsin L inhibitor. 16852. The method of item 16832 wherein the agent is a CD40 antagonist.
16853. The method of item 16832 wherein the agent is a chemokine receptor agonist.
16854. The method of item 16832 wherein the agent is a chymase inhibitor.
16855. The method of item 16832 wherein the agent is a collagenase antagonist.
16856. The method of item 16832 wherein the agent is a CXCR antagonist.
16857. The method of item 16832 wherein the agent is a cyclin dependent kinase inhibitor selected from the group consisting of a CDK-1 inhibitor, a CDK-2 inhibitor, a CDK- 4 inhibitor, a CDK-6 inhibitor, a CAK1 inhibitor from GPC Biotech and Bristol-Myers Squibb, RGB-286199 (GPC Biotech), an anticancer agent from Astex Technology, a CAK1 inhibitor from GPC Biotech, a CDK inhibitor from Sanofi-Aventis, a CDK1/CDK2 inhibitor from Amgen, a CDK2 inhibitor from SUGEN-2 (Pfizer), a hearing loss therapy agent (Sound Pharmaceuticals), PD-0332991 (Pfizer), RGB-286199 (GPC Biotech), Ro-0505124 (Hoffmann-La Roche), a Ser/Thrkinase inhibitor from Lilly (EIi Lilly), CVT-2584 (CAS No. 199986-75- 9) (CV Therapeutics), and an analogue or derivative thereof.
16858. The method of item 16832 wherein the agent is a cyclooxygenase 1 inhibitor. 16859. The method of item 16832 wherein the agent is a DHFR inhibitor.
16860. The method of item 16832 wherein the agent is a dual integrin inhibitor.
16861. The method of item 16832 wherein the agent is an elastase inhibitor.
16862. The method of item 16832 wherein the agent is an elongation factor-1 alpha inhibitor.
16863. The method of item 16832 wherein the agent is an endothelial growth factor antagonist.
16864. The method of item 16832 wherein the agent is an endothelial growth factor receptor kinase inhibitor selected from the group consisting of sorafenib tosylate (Bayer), AAL-993 (Novartis), ABP-309 (Novartis), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), E- 7080 (Eisai), EG-3306 (Ark Therapeutics), EXEL-2880 (Exelixis), GW- 654652 (GlaxoSmithKline), a KDR inhibitor from LG Life Sciences, CT-6685 and CT-6729 (UCB), KRN-633 and KRN-951 (Kirin Brewery), OSI-930 (OSI Pharmaceuticals), SP-5.2 (Supratek Pharma), SU-11657 (Pfizer), a Tie-2 antagonist (Hybrigenics), a VEGFR-2 kinase inhibitor (Bristol-Myers Squibb), XL-647 (Exelixis), a KDR inhibitor from Abbott Laboratories, sorafenib tosylate, and an analogue or derivative thereof.
16865. The method of item 16832 wherein the agent is an endotoxin antagonist. 16866. The method of item 16832 wherein the agent is an epothilone and tubulin binder.
16867. The method of item 16832 wherein the agent is an estrogen receptor antagonist.
16868. The method of item 16832 wherein the agent is an FGF inhibitor.
16869. The method of item 16832 wherein the agent is a famexyl transferase inhibitor.
16870. The method of item 16832 wherein the agent is famesyltransferase inhibitor selected from the group of A-197574 (Abbott), a famesyltransferase inhibitor from Servier, FPTIII (Strathclyde Institute for Drug R), LB-42908 (LG Life Sciences), Pharmaprojects No. 5063 (Genzyme), Pharmaprojects No. 5597 (Ipsen), Yissum Project No. B-1055 (Yissum), and an analogue or derivative thereof.
16871. The method of item 16832 wherein the agent is an FLT-3 kinase inhibitor.
16872. The method of item 16832 wherein the agent is an FGF receptor kinase inhibitor.
16873. The method of item 16832 wherein the agent is a fibrinogen antagonist selected from the group consisting of AUV-201 (Auvation), MG-13926 (Sanofi-Aventis), plasminogen activator (CAS No. 105913-11-9) (from Sanofi-Aventis or UCB), plasminogen activator-2 (tPA- 2) (Sanofi-Aventis), pro-urokinase (CAS No. 82657-92-9) (Sanofi-Aventis), and an analogue or derivative thereof. 16874. The method of item 16832 wherein the agent is a heat shock protein 90 antagonist selected from the group consisting of SRN- 005 (Sirenade), geldanamycin, NSC-33050 (17-allylaminogeldanamycin; 17- AAG), ^-dimethylaminoethylamino-^-demethoxy-geldanamycin (17- DMAG), rifabutin (rifamycin XIV, 1\4-didehydro-1-deoxy-1 ,4-dihydro-5'-(2- methylpropyl)-1~oxo-), and an analogue or derivative thereof.
16875. The method of item 16832 wherein the agent is a histone deacetylase inhibitor.
16876. The method of item 16832 wherein the agent is an HMGCoA reductase inhibitor selected from the group consisting of an atherosclerosis therapeutic from Lipid Sciences, ATI-16000 (ARYx Therapeutics), KS-01-019 (Kos Pharmaceuticals), Pharmaprojects No. 2197 (Sanofi-Aventi), RP 61969 (Sanofi-Aventis), and an analogue or derivative thereof.
16877. The method of item 16832 wherein the agent is an ICAM inhibitor.
16878. The method of item 16832 wherein the agent is an IL, ICE and IRAK antagonist, wherein the antagonist is a CJ-14877, CP- 424174 (Pfizer), NF-61 (Negma-Lerads), and an analogue or derivative thereof.
16879. The method of item 16832 wherein the agent is an IL-2 inhibitor.
16880. The method of item 16832 wherein the agent is an immunosuppressant selected from the group consisting of teriflunomide (Sanofi Aventis), chlorsulfaquinoxalone (NSC-339004), chlorsulfaquinoxalone sulfate, CS-712 (Sankyo), ismomultin alfa (CAS No. 457913-93-8) (Akzo Nobel), antiallergics from GenPat77, antiinflammatories or AT-005 (Androclus Therapeutics), autoimmune disease therapeutics from EpiVax, BN-007 (Bone), budesonide (CAS No. 51333-22- 3) (MAP Pharmaceuticals), CO-14 (Genzyme), edratide (CAS No. 433922- 67-9) (Teva), EP-314 (Enanta), eprovafen (CAS No. 101335-99-3) (Sanofi- Aventis), HWA-131 (CAS No. 118788-41-3) (Sanofi-Aventis), immunomodulators from MerLion Pharmaceuticals, immunosuppressives from Alchemia, IPL-12 (Inflazyme), MDL-9563 (CAS No. 27086-86-8) (Sanofi-Aventis), Pharmaprojects No. 2330 (Sanofi-Aventis), Pharmaprojects No. 6426 (Abgenix), PXS-25 (Pharmaxis), rosmarinic acid (CAS No. 20283-92-5) (Sanofi-Aventis), RP 42927 or RP 54745 (CAS No. 135330-08-4) (Sanofi-Aventis), SGN-35 (Seattle Genetics), ST-1959 (Sigma-Tau), type I diabetes therapy from SYNX Pharma, UNIL-88 (Debiopharm), VP-025 (Vasogen), VR-694 (Vectura), PRTX-001 (Protalex), and an analogue or derivative thereof.
16881. The method of item 16832 wherein the agent is an IMPDH (inosine monophosphate).
16882. The method of item 16832 wherein the agent is an integrin antagonist.
16883. The method of item 16832 wherein the agent is an interleukin antagonist.
16884. The method of item 16832 wherein the agent is an inhibitor of type III receptor tyrosine kinase.
16885. The method of item 16832 wherein the agent is an irreversible inhibitor of enzyme methionine aminopeptidase type 2. 16886. The method of item 16832 wherein the agent is an isozyme selective delta protein kinase C inhibitor.
16887. The method of item 16832 wherein the agent a JAK3 enzyme inhibitor.
16888. The method of item 16832 wherein the agent is a
JNK inhibitor.
16889. The method of item 16832 wherein the agent is a kinase inhibitor.
16890. The method of item 16832 wherein the agent is kinesin antagonist.
16891. The method of item 16832 wherein the agent is a kinesin antagonist.
16892. The method of item 16832 wherein the agent is a leukotriene inhibitor and antagonist selected from the group consisting of ambicromil (CAS No. 58805-38-2) (Sanofi-Aventis), amelubant (CAS No. 346735-24-8) (Boehringer Ingelheim), DW-1141 (Dong Wha), ebselen (Daiichi Pharmaceutical), ibudilast (Kyorin), leucotriene inhibitors from Sanofi-Aventis, lymphotoxin-beta receptor (LT-D) from Biogen Idee, Pharmaprojects No. 1535 and 2728 (CAS No. 119340-33-9) (Sanofi- Aventis), R-112 (Rigel), Rev-5367 (CAS No. 92532-05-3) (Sanofi-Aventis), RG-14893 (CAS No. 141835-49-6) (Sanofi-Aventis), RG-5901-A (CAS No. 101910-24-1), 92532-23-5, RP 66153 (CAS No. 142422-79-5), RP 66364 (CAS No.186916-92-5), RP 69698 (CAS No. 141748-00-7) (Sanofi-Aventis), SC-411930 (Pfizer), SC-41930 (CAS No. 120072-59-5) (Pfizer), SC-50605 (CAS No. 138828-39-4) (Pfizer), SC-51146 (CAS No. 141059-52-1), SC- 53228 (CAS No. 153633-01-3) (Pfizer), spaglumic acid (ZY-15106) (CAS No.3106-85-2), 80619-64-3 (Novartis), tipredane (CAS No. 85197-77-9) (Bristol-Myers Squibb), U-75302 (CAS No. 119477-85-9) (Pfizer), and analogue or derivative thereof.
16893. The method of item 16832 wherein the agent is an MAP kinase inhibitor.
16894. The method of item 16832 wherein the agent is a matrix metalloproteinase inhibitor.
16895. The method of item 16832 wherein the agent is an MCP-CCR2 inhibitor.
16896. The method of item 16832 wherein the agent is an mTOR inhibitor.
16897. The method of item 16832 wherein the agent is an mTOR kinase inhibitor.
16898. The method of item 16832 wherein the agent is a microtubule inhibitor selected from the group consisting of antibody- maytansinoid conjugates from Biogen Idee, colchicines (MantiCore Pharmaceuticals), anticancer immunoconjugates from Johnson & Johnson, DIME from Octamer, gni-1f (GNI), huC242-DM4, huMy9-6-DM1 (ImmunoGen), IDN-5404 (Indena), IMO-098, IMOderm (Imotep), mebendazole (Introgen Therapeutics), microtubule poisons from Cambridge Enterprise, paclitaxel such as LOTAX from Aphios (CAS No. 33069-62-4), Genexol-PM from Samyang, Pharmaprojects No. 6383 (Tapestry Pharmaceuticals), RPR-112378 (Sanofi-Aventis), SGN-75 (Seattle Genetics), SPL-7435 (Starpharma), SSR-250411 (Sanofi-Aventis), trastuzumab-DM1 (Genentech), vinorelbine, and an analogue or derivative thereof.
16899. The method of item 16832 wherein the agent is an MIF inhibitor.
16900. The method of item 16832 wherein the agent is an MMP inhibitor.
16901. The method of item 16832 wherein the agent is a neurokinin (NK) antagonist selected from the group consisting of anthrotainin (CAS No. 148084-40-6) (Sanofi-Aventis), an IBS thereapeutic from ArQuIe, MDL-105212A (CAS No. 167261-60-1) (Ssanofi-Aventis), Pharmaprojects No. 2744, 3258 (CAS No. 139167-47-8) 4006, 4201, or 5986 (Sanofi-Aventis), RP 67580 (CAS No. 135911-02-3), SR-144190 (CAS No. 201152-86-5), SSR-240600, SSR-241586 (Sanofi-Aventis), TKA-457 (Novartis), vestipitant mesylate (CAS No. 334476-64-1) (GlaxoSmithKline), Win-64821 (Sanofi-Aventis), PRX-96026 (Predix Pharmaceuticals), and an analogue or derivative thereof.
16902. The method of item 16832 wherein the agent is an NF kappa B inhibitor selected from the group consisting of AVE-0545 or AVE-0547 (Sanofi-Aventis), bortezomib (CAS No. 179324-69-7) (Millennium Pharmaceuticals), dexanabinol (CAS No. 112924-45-5) (Pharmos), dexlipotam (Viatris), Pharmaprojects No. 6283 (INDRA) (OXiGENE), IPL- 576092 (CAS No. 137571-30-3) (Inflazyme), NFKB decoy (Corgentech), NFKB decoy oligo (AnGes MG), NFKB's from Ariad, osteoporosis treatments or S5 (F005) from Fulcrum Pharmaceuticals, P61 (Phytopharm), R-flurbiprofen (CAS No. 5104-49-4) (Encore Pharmaceuticals), and an analogue or derivative thereof. 16903. The method of item 16832 wherein the agent is a nitric oxide agonist.
16904. The method of item 16832 wherein the agent is an ornithine decarboxylase inhibitor.
16905. The method of item 16832 wherein the agent is a p38 MAP kinase inhibitor selected from the group consisting of AZD-6703 (AstraZeneca), JX-401 (Jexys Pharmaceuticals), BMS-2 (Bristol-Myers Squibb), a p38 MAP kinase inhibitor from Novartis, a p38-alpha MAP kinase inhibitor from Amphora, Pharmaprojects No. 5704 (Pharmacopeia), RPR- 200765A (Sanofi-Aventis), SD-282 (Johnson & Johnson), TAK-715 (Takeda), and an analogue or derivative thereof.
16906. The method of item 16832 wherein the agent is a palmitoyl-protein thioesterase inhibitor.
16907. The method of item 16832 wherein the agent is a PDGF receptor kinase inhibitor selected from the group consisting of AAL- 993, AMN-107, or ABP-309 (Novartis), AMG-706 (Amgen), BAY-57-9352 (Bayer), CDP-860 (UCB), E-7080 (Eisai), imatinib (CAS No. 152459-95-5) (Novartis), OSI-930 (OSI Pharmaceuticals), RPR-127963E (Sanofi-Aventis), RWJ-540973 (Johnson & Johnson), sorafenib tosylate (Bayer), SU-11657 (Pfizer), tandutinib (CAS No. 387867-13-2) (Millennium Pharmaceuticals), vatalanib (Novartis), ZK-CDK (Schering AG), and an analogue or derivative thereof.
16908. The method of item 16832 wherein the agent is (-)-halofenate (Metabolex), AMG-131 (Amgen), antidiabetics from Japan Tobacco, AZD-4619, AZD-8450, AZD-8677 (AstraZeneca), DRF-10945, balaglitazone (Dr Reddy's), CS-00088, CS-00098 (Chipscreen Biosciences), E-3030 (Eisai), etalocib (CAS No. 161172-51-6) (EIi Lilly), GSK-641597 (Ligand), GSK-677954 (GlaxoSmithKline), GW-409544 (Ligand), GW- 590735 (GlaxoSmithKline), K-111 (Hoffmann-La Roche), LY-518674 (EIi Lilly), LY-674 (Ligand), LY-929 (Ligand), MC-3001, MC-3002 (MaxoCore Pharmaceuticals), metformin HCI + pioglitazone (CAS No. 1115-70-4 and 112529-15-4), ACTOPLUS MET from Andrx), muraglitazar (CAS No. 331741-94-7) (Bristol-Myers Squibb), naveglitazar (Ligand), oleoylethanolamide (Kadmus Pharmaceuticals), ONO-5129, pioglitazone hydrochloride (CAS No. 111025-46-8 and 112529-15-4) (Takeda), PLX-204 (Plexxikon), PPAR agonists from Genfit, PPAR delta agonists from EIi Lilly, PPAR-alpha agonists from CrystalGenomics, PPAR-gamma modulators and PPAR-β modulators from CareX, rosiglitazone maleate (CAS No. 122324- 73-4 or 155141-29-0) (GlaxoSmithKline), rosiglitazone maleate/glimepir (CAS No. 155141-29-0 and 93479-97-1), AVANDARYL1 rosiglitazone maleate/metformin extend (CAS No. 155141-29-0 and 657-24-9), AVANDAMET, rosiglitazone maleate+metformin, AVANDAMET (GlaxoSmithKline), tesaglitazar (AstraZeneca), and an analogue or derivative thereof.
16909. The method of item 16832 wherein the agent is a phosphatase inhibitor.
16910. The method of item 16832 wherein the agent is a phosphodiesterase (PDE) inhibitor selected from the group consisting of avanafil (Tanabe Seiyaku), dasantafil (CAS No. 569351-91-3) (Schering- Plough), A-906119 (CAS No. 134072-58-5), DL-850 (Sanofi-Aventis), GRC- 3015, GRC-3566, GRC-3886 (Glenmark), HWA-153 (CAS No. 56395-66-5) (Sanofi-Aventis), hydroxypumafentrine (Altana), IBFB-130011 , IBFB-14-016, IBFB-140301 , IBFB-150007, IBFB-211913 (IBFB Pharma), L-826141 (Merck & Co), medorinone (CAS No. 88296-61-1) (Sanofi-Aventis), MEM- 1917 (Memory Pharmaceuticals), ND-1251 (Neuro3d), PDE inhibitors from ICOS, PDE IV inhibitors from Memory Pharmaceuticals and CrystalGenomics, Pharmaprojects No. 2742 and 6141 (Sanofi-Aventis), QAD-171 (Novartis), RHC-2963 (CAS No. 76993-12-9 and 76993-14-1), RPR-117658, RPR-122818 derivatives, SR-24870 , and RPR-132294 (Sanofi-Aventis), SK-350 (ln2Gen), stroke therapy agents from deCODE Genetics, TAS-203 (Taiho), tofimilast (CAS No. 185954-27-2) (Pfizer), UK- 371800 (Pfizer), WIN-65579 (CAS No. 158020-82-7) (Sanofi-Aventis), IBFB- 130020 (IBFB Pharma), OPC-6535 (CAS No. 145739-56-6) (Otsuka), a phosphodiesterase III inhibitor, enoximone, a phosphodiesterase IV inhibitor, fosfosal, Atopik (Barrier Therapeutics), triflusal, a phosphodiesterase V inhibitor, and an analogue or derivative thereof.
16911. The method of item 16832 wherein the agent is a PKC inhibitor.
16912. The method of item 16832 wherein the agent is a platelet activating factor antagonist.
16913. The method of item 16832 wherein the agent is a platelet-derived growth factor receptor kinase inhibitor.
16914. The method of item 16832 wherein the agent is a prolyl hydroxylase inhibitor.
16915. The method of item 16832 wherein the agent is a polymorphonuclear neutrophil inhibitor.
16916. The method of item 16832 wherein the agent is a protein kinase B inhibitor. 16917. The method of item 16832 wherein the agent is a protein kinase C stimulant.
16918. The method of item 16832 wherein the agent is a purine nucleoside analogue.
16919. The method of item 16832 wherein the agent is a purinoreceptor P2X antagonist.
16920. The method of item 16832 wherein the agent is a Raf kinase inhibitor.
16921. The method of item 16832 wherein the agent is a reversible inhibitor of ErbB1 and ErbB2.
16922. The method of item 16832 wherein the agent is a ribonucleoside triphosphate reductase inhibitor.
16923. The method of item 16832 wherein the agent is an SDF-1 antagonist.
16924. The method of item 16832 wherein the agent is a sheddase inhibitor.
16925. The method of item 16832 wherein the agent is an SRC inhibitor.
16926. The method of item 16832 wherein the agent is a stromelysin inhibitor. 16927. The method of item 16832 wherein the agent is an Syk kinase inhibitor.
16928. The method of item 16832 wherein the agent is a telomerase inhibitor.
16929. The method of item 16832 wherein the agent is a TGF beta inhibitor selected from the group consisting of pirfenidone (CAS No. 53179-13-8) (MARNAC), tranilast (CAS No. 53902-12-8) (Kissei), IN- 1130 (ln2Gen), mannose-6-phosphate (BTG), TGF-β antagonists from Inflazyme (Pharmaprojects No. 6075), TGF-β antagonists from Sydney, non-industrial source), TGF-β I receptor kinase inhibitors from EIi Lilly, TGF- β receptor inhibitors from Johnson & Johnson, and an analogue or derivative thereof.
16930. The method of item 16832 wherein the agent is a TNFα antagonist or TACE inhibitor selected from the group consisting of adalimumab (CAS No. 331731-18-1) (Cambridge Antibody Technology), AGIX-4207 (AtheroGenics), AGT-1 (Advanced Biotherapy), an antiinflammatory from Borean Pharma, Cellzome, or Paradigm Therapeutics, anti-inflammatory vaccine (TNF-alpha kinoid) from Neovacs, humanized anti-TNF antibody or an anti-TNF MAb (CB0006) Celltech (UCB), apratastat (CAS No. 287445-51-0) (Wyeth), BMS-561392 (Bristol-Myers Squibb), BN- 006 (Bone), certolizumab pegol (CAS No. 428863-50-7 or CH-138 (UCB)1 cilomilast (CAS No. 153259-65-5) (GlaxoSmithKline), CR-1 (Nuada Pharmaceuticals), CRx-119 (CombinatoRx), D-5410 (UCB), dacopafant (CAS No. 125372-33-0) (Sanofi-Aventis), dersalazine (CAS No. 188913-57- 7/188913-58-8) (Uriach), etanercept (CAS No. 185243-69-0) (Amgen), ethyl pyruvate (Critical (Critical Therapeutics), golimumab (CAS No. 476181-74-5) (Johnson & Johnson), hormono-immunotherapy from Ipsen, CDP571 (e.g., Humicade from UCB), IC-485 (ICOS), infliximab (CAS No. 170277-31-3) (Johnson & Johnson), IP-751 (Manhattan Pharmaceuticals), ISIS-104838 (CAS No. 250755-32-9) (ISIS Pharmaceuticals), lenalidomide (CAS No. 191732-72-6) (Celgene), lentinan (CAS No. 37339-90-5) (Ajinomoto), MDL- 201112 (CAS No. 142130-73-2) (Sanofi-Aventis), medroxyprogesterone (CAS No. 520-85-4) (InKine Pharmaceutical), N-acetylcysteine (CAS No. 616-91-1) (Zambon), NBE-P2 (DIREVO Biotech), nerelimomab (CAS No. 162774-06-3) (Chiron), OM-294DP (OM PHARMA), onercept (CAS No. 199685-57-9) (Yeda), PASSTNF-alpha (Verigen), pentoxifylline or oxypentifylline (Sanofi-Aventis), Pharmaprojects No. 4091 , 4241, 4295, or 4488 (Sanofi-Aventis), Pharmaprojects No. 5480 (Amgen), Pharmaprojects No. 6749 (Cengent), pirfenidone (CAS No. 53179-13-8) (MARNAC), RPR- 132294 (Sanofi-Aventis), S5 (F002) (Fulcrum Pharmaceuticals), simvastatin (CAS No. 79902-63-9) (Merck & Co), STA-6292 (Synta Pharmaceuticals), tacrolimus (CAS No. 104987-11-3) (Fujisawa LifeCycle Pharma), talactoferrin alfa (CAS No. 308240-58-6) (Agennix), thalidomide (CAS No. 50-35-1) (Celgene), TNF antagonists form ProStrakan, and Synergen, TNF inhibitors (Amgen), TNF-alpha antagonists from Dynavax Technologies and Jerina AG (Germany), TNF-alpha inhibitors from IBFB Pharma and Xencor (Xencor), torbafylline (CAS No. 105102-21-4) (Sanofi-Aventis), UR-1505 (Uriach), VT-346 (Viron Therapeutics), YSIL6 (Y1S Therapeutics), YSTH2 (Y's Therapeutics), NPI-1302a-3 (Nereus Pharmaceuticals, a TNF antagonist from Jerina AG (Germany), dersalazine, and an analogue or derivative thereof.
16931. The method of item 16832 wherein the agent is a Toll receptor inhibitor.
16932. The method of item 16832 wherein the agent is a tubulin antagonist. 16933. The method of item 16832 wherein the agent is a tyrosine kinase inhibitor selected from the group consisting of SU-011248, SUTENT from Pfizer Inc. (New York, NY), BMS-354825, PN-355 (Paracelsian Pharmaceuticals), AGN-199659 (Allergan), AAL-993 or ABP- 309 (Novartis), adaphostin (NIH), AEE-788 (Novartis), AG-013736 (OSI Pharmaceuticals), AG-13736 (Pfizer), ALT-110 (Alteris Therapeutics), AMG- 706 (Amgen), anticancer MAbs from Xencor, anti-EGFrvlll MAbs from Abgenix, anti-HER2 MAb from Abiogen, AZD-2171 or AZD-9935 (AstraZeneca), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), CEP-5214 (Cephalon), CEP-7055 (Cephalon), cetuximab (ImClone Systems), CHIR-200131 and CHIR-258 (Chiron), CP-547632 (OSI Pharmaceuticals), CP-724714 (Pfizer), CT-301 (Creabilis Therapeutics), D- 69491 (Baxter International), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EGFR/ErbB2 inhibitors from Array BioPharma, erlotinib (CAS No. 183319- 69-9) (OSI Pharmaceuticals), EXEL-2880 (Exelixis), FK-778 (Sanofi- Aventis) , gefitinib (CAS No. 184475-35-2) (AstraZeneca), GW-2286 or GW- 654652 (GlaxoSmithKline), her2/neu antigen from AlphaVax, HER-2/neu inhibitor from Generex, Herzyme (Medipad) (Sirna Therapeutics), HKI-272 (Wyeth), HuMax-EGFr (Genmab), idronoxil (CAS No. 81267-65-4) (Novogen), IGF-1 inhibitors from Ontogen, IMC-11F8 (ImClone Systems), kahalalide F (CAS No. 149204-42-2) (PharmaMar), KDR inhibitor from LG Life Sciences, KDR inhibitors from Abbott Laboratories, KDR kinase inhibitors (UCB), Kdr kinase inhibitors from Merck & Co, KRN-633 and KRN- 951 (Kirin Brewery), KSB-102 (Xenova), lapatinib ditosylate (CAS No. 388082-78-8) (GlaxoSmithKline), matuzumab (Merck KGaA), MDX-214 (Medarex), ME-103 (Pharmexa), MED-A300 (Gerolymatos), MNAC-13 (Lay Line Genomics), nimotuzumab (Center of Molecular Immunology), NSC- 330507 or NSC-707545 (NIH), NV-50 (Novogen), OSI-930 (OSI Pharmaceuticals), panitumumab (Abgenix), pelitinib (CAS No. 287933-82-7) (Wyeth), pertuzumab (CAS No. 380610-27-5) (Genentech), Pharmaprojects No. 3985 (Sanofi-Aventis), prostate cancer therapeutics from Sequenom (SQPC35, SQPC36, SQPC90), removab and remoxab (Trion Pharma), RG- 13022 (CAS No. 136831-48-6), RG-13291 (CAS No. 138989-50-1), or RG- 14620 (CAS No. 136831-49-7) (Sanofi-Aventis), RM-6427 (Romark), RNAi breast cancer therapy from Benitec, RP 53801 (CAS No. 125882-88-4) (Sanofi-Aventis), sorafenib tosylate (Bayer), SU-11657 (Pfizer), Tie-2 antagonists from Semaia (Hybrigenics), Tie-2 inhibitors from Ontogen, trastuzumab (CAS No. 180288-69-1) (Genentech), tyrosine kinase inhibitors from Sanofi-Aventis, U3-1287, U3-1565, U3-1784, U3-1800 (U3 Pharma), vatalanib (Novartis), VEGFR-2 kinase inhibitor from Bristol-Myers Squibb, XL-647 (Exelixis), ZD-6474 (AstraZeneca), ZK-CDK (Schering AG), an EGFR tyrosine kinase inhibitor, EKB-569 (Wyeth), and an analogue or derivative thereof.
16934. The method of item 16832 wherein the agent is a VEGF inhibitor.
16935. The method of item 16832 wherein the agent is a vitamin D receptor agonist.
16936. The method of item 16832 wherein the agent is ZD-6474 (an angiogenesis inhibitor).
16937. The method of item 16832 wherein the agent is AP-23573 (an mTOR inhibitor).
16938. The method of item 16832 wherein the agent is synthadotin (a tubulin antagonist).
16939. The method of item 16832 wherein the agent is S-0885 (a collagenase inhibitor). 16940. The method of item 16832 wherein the agent is aplidine (an elongation factor-1 alpha inhibitor).
16941. The method of item 16832 wherein the agent is ixabepilone (an epithilone).
16942. The method of item 16832 wherein the agent is IDN-5390 (an angiogenesis inhibitor).
16943. The method of item 16832 wherein the agent is SB-2723005 (an angiogenesis inhibitor).
16944. The method of item 16832 wherein the agent is ABT-518 (an angiogenesis inhibitor).
16945. The method of item 16832 wherein the agent is combretastatin (an angiogenesis inhibitor).
16946. The method of item 16832 wherein the agent is anecortave acetate (an angiogenesis inhibitor).
16947. The method of item 16832 wherein the agent is SB-715992 (a kinesin antagonist).
16948. The method of item 16832 wherein the agent is temsirolimus (an mTOR inhibitor).
16949. The method of item 16832 wherein the agent is adalimumab (a TNFα antagonist). 16950. The method of item 16832, wherein the composition comprises a polymer.
16951. The method of item 16832, wherein the composition comprises a polymeric carrier.
16952. The method of item 16832 wherein the anti- scarring agent inhibits adhesion between the device and a host into which the device is implanted.
16953. The method of item 16832 wherein the device delivers the anti-scarring agent locally to tissue proximate to the device.
16954. The method of item 16832 wherein the device has a coating that comprises the anti-scarring agent.
16955. The method of item 16832, wherein the device has a coating that comprises the agent and is disposed on a surface of the implant.
16956. The method of item 16832, wherein the device has a coating that comprises the agent and directly contacts the implant.
16957. The method of item 16832, wherein the device has a coating that comprises the agent and indirectly contacts the implant.
16958. The method of item 16832, wherein the device has a coating that comprises the agent and partially covers the implant.
16959. The method of item 16832, wherein the device has a coating that comprises the agent and completely covers the implant. 16960. The method of item 16832, wherein the device has a uniform coating.
16961. The method of item 16832, wherein the device has a non-uniform coating.
16962. The method of item 16832, wherein the device has a discontinuous coating.
16963. The method of item 16832, wherein the device has a patterned coating.
16964. The method of item 16832, wherein the device has a coating with a thickness of 100 Dm or less.
16965. The method of item 16832, wherein the device has a coating with a thickness of 10 Dm or less.
16966. The method of item 16832, wherein the device has a coating, and the coating adheres to the surface of the implant upon deployment of the implant.
16967. The method of item 16832, wherein the device has a coating, and wherein the coating is stable at room temperature for a period of 1 year.
16968. ' The method of item 16832, wherein the device has a coating, and wherein the anti-scarring agent is present in the coating in an amount ranging between about 0.0001% to about 1% by weight. 16969. The method of item 16832, wherein the device has a coating, and wherein the anti-scarring agent is present in the coating in an amount ranging between about 1% to about 10% by weight.
16970. The method of item 16832, wherein the device has a coating, and wherein the anti-scarring agent is present in the coating in an amount ranging between about 10% to about 25% by weight.
16971. The method of item 16832, wherein the device has a coating, and wherein the anti-scarring agent is present in the coating in an amount ranging between about 25% to about 70% by weight.
16972. The method of item 16832, wherein the device has a coating, and wherein the coating further comprises a polymer.
16973. The method of item 16832, wherein the device has a first coating having a first composition and a second coating having a second composition.
16974. The method of item 16832, wherein the device has a first coating having a first composition and a second coating having a second composition, wherein the first composition and the second composition are different.
16975. The method of item 16832, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a copolymer.
16976. The method of item 16832, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a block copolymer. 16977. The method of item 16832, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a random copolymer.
16978. The method of item 16832, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a biodegradable polymer.
16979. The method of item 16832, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a non-biodegradable polymer.
16980. The method of item 16832, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a hydrophilic polymer.
16981. The method of item 16832, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a hydrophobic polymer.
16982. The method of item 16832, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a polymer having hydrophilic domains.
16983. The method of item 16832, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a polymer having hydrophobic domains.
16984. The method of item 16832, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a non-conductive polymer. 16985. The method of item 16832, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises an elastomer.
16986. The method of item 16832, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a hydrogel.
16987. The method of item 16832, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a silicone polymer.
16988. The method of item 16832, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a hydrocarbon polymer.
16989. The method of item 16832, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a styrene-derived polymer.
16990. The method of item 16832, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a butadiene polymer.
16991. The method of item 16832, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a macromer.
16992. The method of item 16832, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a poly(ethylene glycol) polymer. 16993. The method of item 16832 wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises an amorphous polymer.
16994. The method of item 16832, wherein the device comprises a lubricious coating.
16995. The method of item 16832 wherein the anti- scarring agent is located within pores or holes of the device.
16996. The method of item 16832 wherein the anti- scarring agent is located within a channel, lumen, or divet of the device.
16997. The method of item 16832, wherein the device comprises a second pharmaceutically active agent.
16998. The method of item 16832 wherein the device comprises an anti-inflammatory agent.
16999. The method of item 16832 wherein the device comprises an agent that inhibits infection.
17000. The method of item 16832 wherein the device comprises an agent that inhibits infection, and wherein the agent is an anthracycline.
17001. The method of item 16832 wherein the device comprises an agent that inhibits infection, and wherein the agent is doxorubicin. 17002. The method of item 16832 wherein the device comprises an agent that inhibits infection, and wherein the agent is mitoxantrone.
17003. The method of item 16832 wherein the device comprises an agent that inhibits infection, and wherein the agent is a fluoropyrimidine.
17004. The method of item 16832 wherein the device comprises an agent that inhibits infection, and wherein the agent is 5- fluorouracil (5-FU).
17005. The method of item 16832 wherein the device comprises an agent that inhibits infection, and wherein the agent is a folic acid antagonist.
17006. The method of item 16832 wherein the device comprises an agent that inhibits infection, and wherein the agent is methotrexate.
17007. The method of item 16832 wherein the device comprises an agent that inhibits infection, and wherein the agent is a podophylotoxin.
17008. The method of item 16832 wherein the device comprises an agent that inhibits infection, and wherein the agent is etoposide.
17009. The method of item 16832 wherein the device comprises an agent that inhibits infection, and wherein the agent is a camptothecin. 17010. The method of item 16832 wherein the device comprises an agent that inhibits infection, and wherein the agent is a hydroxyurea.
17011. The method of item 16832 wherein the device comprises an agent that inhibits infection, and wherein the agent is a platinum complex.
MOM. The method of item 16832 wherein the device comprises an agent that inhibits infection, and wherein the agent is cisplatin.
17013. The method of item 16832, further comprising an anti-thrombotic agent.
17014. The method of item 16832 wherein the device comprises a visualization agent.
17015. The method of item 16832 wherein the device comprises a visualization agent, wherein the visualization agent is a radiopaque material, and wherein the radiopaque material comprises a metal, a halogenated compound, or a barium containing compound.
17016. The method of item 16832 wherein the device comprises a visualization agent, wherein the visualization agent is a radiopaque material, and wherein the radiopaque material comprises barium, tantalum, or technetium.
17017. The method of item 16832 wherein the device comprises a visualization agent, and wherein the visualization agent is a MRI responsive material. 17018. The method of item 16832 wherein the device comprises a visualization agent, and wherein the visualization agent comprises a gadolinium chelate.
17019. The method of item 16832 wherein the device comprises a visualization agent, and wherein the visualization agent comprises iron, magnesium, manganese, copper, or chromium.
17020. The method of item 16832 wherein the device comprises a visualization agent, and wherein the visualization agent comprises an iron oxide compound.
17021. The method of item 16832 wherein the device comprises a visualization agent, and wherein the visualization agent comprises a dye, pigment, or colorant.
17022. The method of item 16832 wherein the device comprises an echogenic material.
17023. The method of item 16832 wherein the device comprises an echogenic material, and wherein the echogenic material is in the form of a coating.
17024. The method of item 16832 wherein the device is sterile.
17025. The method of item 16832 wherein the anti- scarring agent is released into tissue in the vicinity of the device after deployment of the device. 17026. The method of item 16832 wherein the anti- scarring agent is released into tissue in the vicinity of the device after deployment of the device, and wherein the tissue is connective tissue.
17027. The method of item 16832 wherein the anti- scarring agent is released into tissue in the vicinity of the device after deployment of the device, and wherein the tissue is muscle tissue.
17028. The method of item 16832 wherein the anti- scarring agent is released into tissue in the vicinity of the device after deployment of the device, and wherein the tissue is nerve tissue.
17029. The method of item 16832 wherein the anti- scarring agent is released into tissue in the vicinity of the device after deployment of the device, and wherein the tissue is epithelium tissue.
17030. The method of item 16832 wherein the anti- scarring agent is released in effective concentrations from the device over a period ranging from the time of deployment of the device to about 1 year.
17031. The method of item 16832 wherein the anti- scarring agent is released in effective concentrations from the device over a period ranging from about 1 month to 6 months.
17032. The method of item 16832 wherein the anti- scarring agent is released in effective concentrations from the device over a period ranging from about 1 - 90 days.
17033. The method of item 16832 wherein the anti- scarring agent is released in effective concentrations from the device at a constant rate. 17034. The method of item 16832 wherein the anti- scarring agent is released in effective concentrations from the device at an increasing rate.
17035. The method of item 16832 wherein the anti- scarring agent is released in effective concentrations from the device at a decreasing rate.
17036. The method of item 16832 wherein the anti- scarring agent is released in effective concentrations from the composition comprising the anti-scarring agent by diffusion over a period ranging from the time of deployment of the device to about 90 days.
17037. The method of item 16832 wherein the anti- scarring agent is released in effective concentrations from the composition comprising the anti-scarring agent by erosion of the composition over a period ranging from the time of deployment of the device to about 90 days.
17038. The method of item 16832 wherein the device comprises about 0.01 Dg to about 10 Dg of the anti-scarring agent.
17039. The method of item 16832 wherein the device comprises about 10 Dg to about 10 mg of the anti-scarring agent.
17040. The method of item 16832 wherein the device comprises about 10 mg to about 250 mg of the anti-scarring agent.
17041. The method of item 16832 wherein the device comprises about 250 mg to about 1000 mg of the anti-scarring agent. 17042. The method of item 16832 wherein the device comprises about 1000 mg to about 2500 mg of the anti-scarring agent.
17043. The method of item 16832 wherein a surface of the device comprises less than 0.01 Og of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
17044. The method of item 16832 wherein a surface of the device comprises about 0.01 Og to about 1 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
17045. The method of item 16832 wherein a surface of the device comprises about 1 Dg to about 10 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
17046. The method of item 16832 wherein a surface of the device comprises about 10 Dg to about 250 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
17047. The method of item 16832 wherein a surface of the device comprises about 250 Dg to about 1000 Dg of the anti-scarring agent of anti-scarring agent per mm2 of device surface to which the anti- scarring agent is applied.
17048. The method of item 16832 wherein a surface of the device comprises about 1000 Dg to about 2500 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
17049. The method of item 16832 wherein the combining is performed by direct affixing the agent or the composition to the implant. 17050. The method of item 16832 wherein the combining is performed by spraying the agent or the component onto the implant.
17051. The method of item 16832 wherein the combining is performed by electrospraying the agent or the composition onto the implant.
17052. The method of item 16832 wherein the combining is performed by dipping the implant into a solution comprising the agent or the composition.
17053. The method of item 16832 wherein the combining is performed by covalently attaching the agent or the composition to the implant.
17054. The method of item 16832 wherein the combining is performed by non-covalently attaching the agent or the composition to the implant.
17055. The method of item 16832 wherein the combining is performed by coating the implant with a substance that contains the agent or the composition.
17056. The method of item 16832 wherein the combining is performed by coating the implant with a substance that absorbs the agent.
17057. The method of item 16832 wherein the combining is performed by interweaving a thread composed of, or coated with, the agent or the composition. 17058. The method of item 16832 wherein the combining is performed by covering all the implant with a sleeve that contains the agent or the composition.
17059. The method of item 16832 wherein the combining is performed by covering a portion of the implant with a sleeve that contains the agent or the composition.
17060. The method of item 16832 wherein the combining is performed by covering all the implant with a cover that contains the agent or the composition.
17061. The method of item 16832 wherein the combining is performed by covering a portion of the implant with a cover that contains the agent or the composition.
17062. The method of item 16832 wherein the combining is performed by covering all the implant with an electrospun fabric that contains the agent or the composition.
17063. The method of item 16832 wherein the combining is performed by covering a portion of the implant with an electrospun fabric that contains the agent or the composition.
17064. The method of item 16832 wherein the combining is performed by covering all the implant with a mesh that contains the agent or the composition.
17065. The method of item 16832 wherein the combining is performed by covering a portion of the implant with a mesh that contains the agent or the composition. 17066. The method of item 16832 wherein the combining is performed by constructing all the implant with the agent or the composition.
17067. The method of item 16832 wherein the combining is performed by constructing a portion of the implant with the agent or the composition.
17068. The method of item 16832 wherein the combining is performed by impregnating the implant with the agent or the composition.
17069. The method of item 16832 wherein the combining is performed by constructing all of the implant from a degradable polymer that releases the agent.
17070. The method of item 16832 wherein the combining is performed by constructing a portion of the implant from a degradable polymer that releases the agent.
17071. The method of item 16832 wherein the combining is performed by dipping the implant into a solution that comprise the agent and an inert solvent for the implant.
17072. The method of item 16832 wherein the combining is performed by dipping the implant into a solution that comprises the agent and a solvent that will swell the implant.
17073. The method of item 16832 wherein the combining is performed by dipping the implant into a solution that comprises the agent and a solvent that will dissolve the implant. 17074. The method of item 16832 wherein the combining is performed by dipping the implant into a solution that comprises the agent, a polymer and an inert solvent for the implant.
17075. The method of item 16832 wherein the combining is performed by dipping the implant into a solution that comprises the agent, a polymer and a solvent that will swell the implant.
17076. The method of item 16832 wherein the combining is performed by dipping the implant into a solution that comprises the agent, a polymer and a solvent that will dissolve the implant.
17077. The method of item 16832 wherein the combining is performed by spraying the implant into a solution that comprises the agent and an inert solvent for the implant.
17078. The method of item 16832 wherein the combining is performed by spraying the implant into a solution that comprises the agent and a solvent that will swell the implant.
17079. The method of item 16832 wherein the combining is performed by spraying the implant into a solution that comprises the agent and a solvent that will dissolve the implant.
17080. The method of item 16832 wherein the combining is performed by spraying the implant into a solution that comprises the agent, a polymer and an inert solvent for the implant.
17081. The method of item 16832 wherein the combining is performed by spraying the implant into a solution that comprises the agent, a polymer and a solvent that will swell the implant. 17082. The method of item 16832 wherein the combining is performed by spraying the implant into a solution that comprises the agent, a polymer and a solvent that will dissolve the implant.
17083. A method as in any one of items 16832-17082, wherein the device is a spinal disc.
17084. A method as in any one of items 16832-17082, wherein the device is a vertebral implant.
17085. A method as in any one of items 16832-17082, wherein the device is a vertebral disc prosthesis.
17086. A method as in any one of items 16832-17082, wherein the device is a lumbar disc implant.
17087. A method as in any one of items 16832-17082, wherein the device is a cervical disc implant.
17088. A method as in any one of items 16832-17082, wherein the device is an intervertebral disc.
17089. A method as in any one of items 16832-17082, wherein the device is a spinal prosthesis.
17090. A method as in any one of items 16832-17082, wherein the device is an artificial disc.
17091. A method as in any one of items 16832-17082, wherein the device is a spinal disc endoprosthesis. 17092. A method as in any one of items 16832-17082, wherein the device is an intervertebral implant.
17093. A method as in any one of items 16832-17082, wherein the device is an implantable spinal graft.
17094. A method as in any one of items 16832-17082, wherein the device is an implantable bone graft.
17095. A method as in any one of items 16832-17082, wherein the device is an artificial lumbar disc.
17096. A method as in any one of items 16832-17082, wherein the device is a spinal nucleus implant.
17097. A method as in any one of items 16832-17082, wherein the device is an intervertebral disc spacer.
17098. A method as in any one of items 16832-17082, wherein the device is a fusion cage.
17099. A method as in any one of items 16832-17082, wherein the device is a fusion basket.
17100. A method as in any one of items 16832-17082, wherein the device is a fusion cage apparatus.
17101. A method as in any one of items 16832-17082, wherein the device is an interbody cage. 17102. A method as in any one of items 16832-17082, wherein the device is an interbody implant.
17103. A method as in any one of items 16832-17082, wherein the device is a fusion cage.
17104. A method as in any one of items 16832-17082, wherein the device is an anchoring device.
17105. A method as in any one of items 16832-17082, wherein the device is a bone fixation apparatus.
17106. A method as in any one of items 16832-17082, wherein the device is a fusion stabilization chamber.
17107. A method as in any one of items 16832-17082, wherein the device is an anchoring bone plate.
17108. A method as in any one of items 16832-17082, wherein the device is a bone screw.
17109. A method of making a medical device comprising: combining an implant that provides a surgical adhesion barrier and an anti-scarring agent or a composition comprising an anti-scarring agent, wherein the agent inhibits scarring between the device and a host into which the device is implanted.
17110. The method of item 17109 wherein the agent is an adensosine A2A receptor antagonist. 17111. The method of item 17109 wherein the agent is an AKT inhibitor.
17112. The method of item 17109 wherein the agent is an alpha 2 integrin antagonist, wherein the alpha 2 integrin antagonist is Pharmaprojects No. 5754 (Merck KgaA).
17113. The method of item 17109 wherein the agent is an alpha 4 integrin antagonist.
17114. The method of item 17109 wherein the agent is an alpha 7 nicotinic receptor agonist.
17115. The method of item 17109 wherein the agent is an angiogenesis inhibitor selected from the group consisting of AG-12,958 (Pfizer), ATN-161 (Attention LLC), neovastat, an angiogenesis inhibitor from Jerina AG (Germany), NM-3 (Mercian), VGA-1155 (Taisho), FCE-26644 (Pfizer), FCE-26950 (Pfizer), FPMA (Meiji Daries), FR-111142 (Fujisawa), GGTI-298, GM-1306 (Ligand), GPA-1734 (Novartis), NNC-47-0011 (Novo Nordisk), herbamycin (Nippon Kayaku), lenalidomide (Celegene), IP-10 (NIH), ABT-828 (Abbott), KIN-841 (Tokushima University, Japan), SF-1126 (Semafore Pharmaceuticals), laminin technology (NIH), CHIR-258 (Chiron), NVP-AEW541 (Novartis), NVP-AEW541 (Novartis), Vt16907 (Alchemia), OXI-8007 (Oxigene), EG-3306 (Ark Therapeutics), Maspin (Arriva), ABT- 567 (Abbott), PPI-2458 (Praecis Pharmaceuticals), CC-5079, CC-4089 (Celgene), HIF-1 alpha inhibitors (Xenova), S-247 (Pfizer), AP-23573 (Ariad), AZD-9935 (Astra Zeneca), mebendazole (Introgen Therapeutics), MetAP-2 inhibitors (GlaxoSmithKline), AG-615 (Angiogene Pharmaceuticals), Tie-2 antagonists (Hybrigenics), NC-381 , CYC-381, NC-169, NC-219, NC-383, NC-384, NC-407 (Lorus Therapeutics), ATN-224 (Attenuon), ON-01370 (Onconova), Vitronectin antagonists (Amgen), SDX-103 (Salmedix), Vitronectin antagonists (Shire), CHP (Riemser), TEK (Amgen), Anecortave acetate (Alcon), T46.2 (Matrix Therapeutics), HG-2 (Heptagen), TEM antagonists (Genzyme), Oxi-4500 (Oxigene), ATN-161 (Attenuon), WX-293 (Wilex), M-2025 (Metris Therapeutics), Alphastatin (BioActa), YH-16 (Yantai Rongchang), BIBF-1120 (Boehringer Ingelheim), BAY-57-9352 (Bayer), AS- 1404 (Cancer Research Technology), SC-77964 (Pfizer), glycomimetics (BioTie Therapies), TIE-2 Inhibitors (Ontogen), DIMI, Octamer (Octamer), ABR-215050 (Active Biotech), ABT-518 (Abbott), KDR inhibitors (Abbott), BSF-466895 (Abbott), SCH-221153 (Schering-Plough), DAC.antiangiogenic (ConjuChem), TFPI (EntreMed), AZD-2171 (Astra-Zenaca), CDC-394 (Celgene), LY290293 (EIi Lilly), IDN-5390 (Indena), Kdr Kinase Inhibitors (Merck), CT-113020, CT-116433, CT-116563, CT-31890, CT-32228) (Cell Therapeutics), A-299620 (Abbott), TWEAK Inhibitor (Amgen), VEGF modulators (Johnson and Johnson), Tum-N53, tumstatin (Genzyme), Thios- 1 , Thios-2 (Thios Pharmaceuticals), MV-6401 (Miravant Medical Technologies), Spisulosine (PharmaMar), CEP-7055 (Cephalon), AUV-201 (Auvation), LM-609 (EIi Lilly), SKF-106615 (AnorMED), Oglufanide disodium (Cytran), BW-114 (Pharminox), Calreticulin (NIH), WX-678 (Wilex), SD-7784 (Pfizer), WX-UK1 (Wilex), SH-268 (Schering AG), 2-Me-PGA (Celgene), S- 137 (Pfizer), ZD-6126 (Angiogene Pharmaceuticals), SG-292 (SignalGen), Benefin (Lane Labs), A6, A36 (Angstrom), SB-2723005 (GlaxoSmithKline), SC-7 (Cell Therapeutics), ZEN-014 (AEtema Zentaris), 2-methoxyestradiol (EntreMed), NK-130119 (Nippon Kayaku), CC-10004 (Celgene), AVE- 8062A (Ajinomoto), Tacedinaline (Pfizer), Actinonin (Tokyo Metropolitan Institute of Medical Science), Lenalidomide (Celgene), VGA-1155, BTO-956 (SRI International), ER-68203-00 (Eisai), CT-6685 (UCB), JKC-362 (Phoenix Pharmaceuticals), DMI-3798 (DMI Biosciences, Angiomate (Ipsen), ZD-6474 (AstraZeneca), CEP-5214 (Cephalon), Canstatin (Genzyme), NM-3 (Mercian), Oridigm (Med iQuest Therapeutics), Exherin (Adherex), BLS-0597 (Boston Life Sciences), PTC-299 (PTC Therapeutics), NPI-2358 (Nereus Pharmaceuticals), CGP-79787 (Novartis), AEE-788 (Novartis), CKD-732 (Chong Kun Dang), CP-564959 (OSI Pharmaceuticals), CM-101 (CarboMed), CT-2584, CT3501 (Cell Therapeutics), combretastatin and analogues and derivatives thereof (Oxigene), Rebimastat (Bristol-Meyers Squibb), Dextrin 2-sulfate (ML Laboratories), Cilengitide (Merk KGaA), NSC-706704 (Pharminox), KRN- 951 (Kirin Brewery), Ukrain, NSC-631570 (Nowicky Pharma), Tecogalan sodium (Daiichi Pharmaceutical), Tz-93 (Tsumura), TBC-1635 (Encysive Pharmaceuticals), TAN-1120 (Takeda), Semaxanib (Pfizer), BDI-7800 (Biopharmacopae), SD-186, SD-983 (Bristol-Meyers Squibb), SB-223245 (GlaxoSmithKline), SC-236 (Pfizer), RWJ-590973 (Johnson and Johnson), ILX-1850 (Genzyme), SC-68488, S-836 (Pfizer), CG-55069-11 (CuraGen), Ki-23057 (Kirin Brewery), CCX-700 (Chemoentryx), Pegaptanib octasodium (Giled Sciences), ANGIOCOL (available from Biostratum Inc.), or an analogue or derivative thereof.
17116. The method of item 17109 wherein the agent is an apoptosis antagonist.
17117. The method of item 17109 wherein the agent is an apoptosis activator.
17118. The method of item 17109 wherein the agent is a beta 1 integrin antagonist.
17119. The method of item 17109 wherein the agent is a beta tubulin inhibitor.
17120. The method of item 17109 wherein the agent is a blocker of enzyme production in Hepatitis C. 17121. The method of item 17109 wherein the agent is a Bruton's tyrosine kinase inhibitor.
17122. The method of item 17109 wherein the agent is a calcineurin inhibitor.
17123. The method of item 17109 wherein the agent is a caspase 3 inhibitor.
17124. The method of item 17109 wherein the agent is a CC chemokine receptor antagonist.
17125. The method of item 17109 wherein the agent is a cell cycle inhibitor selected from the group consisting of SNS-595 (Sunesis), synthadotin, KRX-0403, and an analogue or derivative thereof.
17126. The method of item 17109 wherein the agent is a cathepsin B inhibitor.
17127. The method of item 17109 wherein the agent is a cathepsin K inhibitor, wherein the cathepsin K inhibitor is 462795 (GlaxoSmithKline), INPL-022-D6 (Amura Therapeutics), or an analogue or derivative thereof.
17128. The method of item 17109 wherein the agent is a cathepsin L inhibitor.
17129. The method of item 17109 wherein the agent is a CD40 antagonist. 17130. The method of item 17109 wherein the agent is a chemokine receptor agonist.
17131. The method of item 17109 wherein the agent is a chymase inhibitor.
17132. The method of item 17109 wherein the agent is a collagenase antagonist.
17133. The method of item 17109 wherein the agent is a CXCR antagonist.
17134. The method of item 17109 wherein the agent is a cyclin dependent kinase inhibitor selected from the group consisting of a CDK-1 inhibitor, a CDK-2 inhibitor, a CDK- 4 inhibitor, a CDK-6 inhibitor, a CAK1 inhibitor from GPC Biotech and Bristol-Myers Squibb, RGB-286199 (GPC Biotech), an anticancer agent from Astex Technology, a CAK1 inhibitor from GPC Biotech, a CDK inhibitor from Sanofi-Aventis, a CDK1/CDK2 inhibitor from Amgen, a CDK2 inhibitor from SUGEN-2 (Pfizer), a hearing loss therapy agent (Sound Pharmaceuticals), PD-0332991 (Pfizer), RGB-286199 (GPC Biotech), Ro-0505124 (Hoffmann-La Roche), a Ser/Thr kinase inhibitor from Lilly (EIi Lilly), CVT-2584 (CAS No. 199986-75- 9) (CV Therapeutics), and an analogue or derivative thereof.
17135. The method of item 17109 wherein the agent is a cyclooxygenase 1 inhibitor.
17136. The method of item 17109 wherein the agent is a DHFR inhibitor. 17137. The method of item 17109 wherein the agent is a dual integrin inhibitor.
17138. The method of item 17109 wherein the agent is an elastase inhibitor.
17139. The method of item 17109 wherein the agent is an elongation factor-1 alpha inhibitor.
17140. The method of item 17109 wherein the agent is an endothelial growth factor antagonist.
17141. The method of item 17109 wherein the agent is an endothelial growth factor receptor kinase inhibitor selected from the group consisting of sorafenib tosylate (Bayer), AAL-993 (Novartis), ABP-309 (Novartis), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), E- 7080 (Eisai), EG-3306 (Ark Therapeutics), EXEL-2880 (Exelixis), GW- 654652 (GlaxoSmithKline), a KDR inhibitor from LG Life Sciences, CT-6685 and CT-6729 (UCB), KRN-633 and KRN-951 (Kirin Brewery), OSI-930 (OSI Pharmaceuticals), SP-5.2 (Supratek Pharma), SU-11657 (Pfizer), a Tie-2 antagonist (Hybrigenics), a VEGFR-2 kinase inhibitor (Bristol-Myers Squibb), XL-647 (Exelixis), a KDR inhibitor from Abbott Laboratories, sorafenib tosylate, and an analogue or derivative thereof.
17142. The method of item 17109 wherein the agent is an endotoxin antagonist.
17143. The method of item 17109 wherein the agent is an epothilone and tubulin binder. 17144. The method of item 17109 wherein the agent is an estrogen receptor antagonist.
17145. The method of item 17109 wherein the agent is an FGF inhibitor.
17146. The method of item 17109 wherein the agent is a famexyl transferase inhibitor.
17147. The method of item 17109 wherein the agent is farnesyltransferase inhibitor selected from the group of A-197574 (Abbott), a famesyltransferase inhibitor from Servier, FPTIII (Strathclyde Institute for Drug R), LB-42908 (LG Life Sciences), Pharmaprojects No. 5063 (Genzyme), Pharmaprojects No. 5597 (Ipsen), Yissum Project No. B-1055 (Yissum), and an analogue or derivative thereof.
17148. The method of item 17109 wherein the agent is an FLT-3 kinase inhibitor.
17149. The method of item 17109 wherein the agent is an FGF receptor kinase inhibitor.
17150. The method of item 17109 wherein the agent is a fibrinogen antagonist selected from the group consisting of AUV-201 (Auvation), MG-13926 (Sanofi-Aventis), plasminogen activator (CAS No. 105913-11-9) (from Sanofi-Aventis or UCB), plasminogen activator-2 (tPA- 2) (Sanofi-Aventis), pro-urokinase (CAS No. 82657-92-9) (Sanofi-Aventis), and an analogue or derivative thereof.
17151. The method of item 17109 wherein the agent is a heat shock protein 90 antagonist selected from the group consisting of SRN- 005 (Sirenade), geldanamycin, NSC-33050 (17-allylaminogeldanamycin; 17- AAG), ^-dimethylaminoethylamino-^-demethoxy-geldanamycin (17- DMAG), rifabutin (rifamycin XIV, 1',4-didehydro-1-deoxy-1 ,4-dihydro-5'-(2- methylpropyl)-1-oxo-), and an analogue or derivative thereof.
17152. The method of item 17109 wherein the agent is a histone deacetylase inhibitor.
17153. The method of item 17109 wherein the agent is an HMGCoA reductase inhibitor selected from the group consisting of an atherosclerosis therapeutic from Lipid Sciences, ATI-16000 (ARYx Therapeutics), KS-01-019 (Kos Pharmaceuticals), Pharmaprojects No. 2197 (Sanofi-Aventi), RP 61969 (Sanofi-Aventis), and an analogue or derivative thereof.
17154. The method of item 17109 wherein the agent is an ICAM inhibitor.
17155. The method of item 17109 wherein the agent is an IL, ICE and IRAK antagonist, wherein the antagonist is a CJ-14877, CP- 424174 (Pfizer), NF-61 (Negma-Lerads), and an analogue or derivative thereof.
17156. The method of item 17109 wherein the agent is an IL-2 inhibitor.
17157. The method of item 17109 wherein the agent is an immunosuppressant selected from the group consisting of teriflunomide (Sanofi Aventis), chlorsulfaquinoxalone (NSC-339004), chlorsulfaquinoxalone sulfate, CS-712 (Sankyo), ismomultin alfa (CAS No. 457913-93-8) (Akzo Nobel), antiallergics from GenPat.77, anti- inflammatories or AT-005 (Androclus Therapeutics), autoimmune disease therapeutics from EpiVax, BN-007 (Bone), budesonide (CAS No. 51333-22- 3) (MAP Pharmaceuticals), CO-14 (Genzyme), edratide (CAS No. 433922- 67-9) (Teva), EP-314 (Enanta), eprovafen (CAS No. 101335-99-3) (Sanofi- Aventis), HWA-131 (CAS No. 118788-41-3) (Sanofi-Aventis), immunomodulators from MerLion Pharmaceuticals, immunosuppressives from Alchemia, IPL-12 (Inflazyme), MDL-9563 (CAS No. 27086-86-8) (Sanofi-Aventis), Pharmaprojects No. 2330 (Sanofi-Aventis), Pharmaprojects No. 6426 (Abgenix), PXS-25 (Pharmaxis), rosmarinic acid (CAS No. 20283-92-5) (Sanofi-Aventis), RP 42927 or RP 54745 (CAS No. 135330-08-4) (Sanofi-Aventis), SGN-35 (Seattle Genetics), ST-1959 (Sigma-Tau), type I diabetes therapy from SYNX Pharma, UNIL-88 (Debiopharm), VP-025 (Vasogen), VR-694 (Vectura), PRTX-001 (Protalex), and an analogue or derivative thereof.
17158. The method of item 17109 wherein the agent is an IMPDH (inosine monophosphate).
17159. The method of item 17109 wherein the agent is an integrin antagonist.
17160. The method of item 17109 wherein the agent is an interleukin antagonist.
17161. The method of item 17109 wherein the agent is an inhibitor of type III receptor tyrosine kinase.
17162. The method of item 17109 wherein the agent is an irreversible inhibitor of enzyme methionine aminopeptidase type 2. 17163. The method of item 17109 wherein the agent is an isozyme selective delta protein kinase C inhibitor.
17164. The method of item 17109 wherein the agent a JAK3 enzyme inhibitor.
17165. The method of item 17109 wherein the agent is a
JNK inhibitor.
17166. The method of item 17109 wherein the agent is a kinase inhibitor.
17167. The method of item 17109 wherein the agent is kinesin antagonist.
17168. The method of item 17109 wherein the agent is a kinesin antagonist.
17169. The method of item 17109 wherein the agent is a leukotriene inhibitor and antagonist selected from the group consisting of ambicromil (CAS No. 58805-38-2) (Sanofi-Aventis), amelubant (CAS No. 346735-24-8) (Boehringer Ingelheim), DW-1141 (Dong Wha), ebselen (Daiichϊ Pharmaceutical), ibudilast (Kyorin), leucotriene inhibitors from Sanofi-Aventis, lymphotoxin-beta receptor (LT-D) from Biogen Idee, Pharmaprojects No. 1535 and 2728 (CAS No. 119340-33-9) (Sanofi- Aventis), R-112 (Rigel), Rev-5367 (CAS No. 92532-05-3) (Sanofi-Aventis), RG-14893 (CAS No. 141835-49-6) (Sanofi-Aventis), RG-5901-A (CAS No. 101910-24-1), 92532-23-5, RP 66153 (CAS No. 142422-79-5), RP 66364 (CAS No.186916-92-5), RP 69698 (CAS No. 141748-00-7) (Sanofi-Aventis), SC-411930 (Pfizer), SC-41930 (CAS No. 120072-59-5) (Pfizer), SC-50605 (CAS No. 138828-39-4) (Pfizer), SC-51146 (CAS No. 141059-52-1), SC- 53228 (CAS No. 153633-01-3) (Pfizer), spaglumic acid (ZY-15106) (CAS No.3106-85-2), 80619-64-3 (Novartis), tipredane (CAS No. 85197-77-9) (Bristol-Myers Squibb), U-75302 (CAS No. 119477-85-9) (Pfizer), and analogue or derivative thereof.
17170. The method of item 17109 wherein the agent is an MAP kinase inhibitor.
17171. The method of item 17109 wherein the agent is a matrix metalloproteinase inhibitor.
17172. The method of item 17109 wherein the agent is an MCP-CCR2 inhibitor.
17173. The method of item 17109 wherein the agent is an mTOR inhibitor.
17174. The method of item 17109 wherein the agent is an mTOR kinase inhibitor.
17175. The method of item 17109 wherein the agent is a microtubule inhibitor selected from the group consisting of antibody- maytansinoid conjugates from Biogen Idee, colchicines (MantiCore Pharmaceuticals), anticancer immunoconjugates from Johnson & Johnson, DIME from Octamer, gni-1f (GNI), huC242-DM4, huMy9-6-DM1 (ImmunoGen), IDN-5404 (Indena), IMO-098, IMOderm (Imotep), mebendazole (Introgen Therapeutics), microtubule poisons from Cambridge Enterprise, paclitaxel such as LOTAX from Aphios (CAS No. 33069-62-4), Genexol-PM from Samyang, Pharmaprojects No. 6383 (Tapestry Pharmaceuticals), RPR-112378 (Sanofi-Aventis), SGN-75 (Seattle Genetics), SPL-7435 (Starpharma), SSR-250411 (Sanofi-Aventis), trastuzumab-DM1 (Genentech), vinorelbine, and an analogue or derivative thereof.
17176. The method of item 17109 wherein the agent is an MIF inhibitor.
17177. The method of item 17109 wherein the agent is an MMP inhibitor.
17178. The method of item 17109 wherein the agent is a neurokinin (NK) antagonist selected from the group consisting of anthrotainin (CAS No. 148084-40-6) (Sanofi-Aventis), an IBS thereapeutic from ArQuIe, MDL-105212A (CAS No. 167261-60-1) (Ssanofi-Aventis), Pharmaprojects No. 2744, 3258 (CAS No. 139167-47-8) 4006, 4201 , or 5986 (Sanofi-Aventis), RP 67580 (CAS No. 135911-02-3), SR-144190 (CAS No. 201152-86-5), SSR-240600, SSR-241586 (Sanofi-Aventis), TKA-457 (Novartis), vestipitant mesylate (CAS No. 334476-64-1) (GlaxoSmithKline), Win-64821 (Sanofi-Aventis), PRX-96026 (Predix Pharmaceuticals), and an analogue or derivative thereof.
17179. The method of item 17109 wherein the agent is an NF kappa B inhibitor selected from the group consisting of AVE-0545 or AVE-0547 (Sanofi-Aventis), bortezomib (CAS No. 179324-69-7) (Millennium Pharmaceuticals), dexanabinol (CAS No. 112924-45-5) (Pharmos), dexlipotam (Viatris), Pharmaprojects No. 6283 (INDRA) (OXiGENE), IPL- 576092 (CAS No. 137571-30-3) (Inflazyme), NFKB decoy (Corgentech), NFKB decoy oligo (AnGes MG), NFKB's from Ariad, osteoporosis treatments or S5 (F005) from Fulcrum Pharmaceuticals, P61 (Phytopharm), R-flurbiprofen (CAS No. 5104-49-4) (Encore Pharmaceuticals), and an analogue or derivative thereof. 17180. The method of item 17109 wherein the agent is a nitric oxide agonist.
17181. The method of item 17109 wherein the agent is an ornithine decarboxylase inhibitor.
17182. The method of item 17109 wherein the agent is a p38 MAP kinase inhibitor selected from the group consisting of AZD-6703 (AstraZeneca), JX-401 (Jexys Pharmaceuticals), BMS-2 (Bristol-Myers Squibb), a p38 MAP kinase inhibitor from Novartis, a p38-alpha MAP kinase inhibitor from Amphora, Pharmaprojects No. 5704 (Pharmacopeia), RPR- 200765A (Sanofi-Aventis), SD-282 (Johnson & Johnson), TAK-715 (Takeda), and an analogue or derivative thereof.
17183. The method of item 17109 wherein the agent is a palmitoyl-protein thioesterase inhibitor.
17184. The method of item 17109 wherein the agent is a PDGF receptor kinase inhibitor selected from the group consisting of AAL- 993, AMN-107, or ABP-309 (Novartis), AMG-706 (Amgen), BAY-57-9352 (Bayer), CDP-860 (UCB), E-7080 (Eisai), imatinib (CAS No. 152459-95-5) (Novartis), OSI-930 (OSI Pharmaceuticals), RPR-127963E (Sanofi-Aventis), RWJ-540973 (Johnson & Johnson), sorafenib tosylate (Bayer), SU-11657 (Pfizer), tandutinib (CAS No. 387867-13-2) (Millennium Pharmaceuticals), vatalanib (Novartis), ZK-CDK (Schering AG), and an analogue or derivative thereof.
17185. The method of item 17109 wherein the agent is (-)-halofenate (Metabolex), AMG-131 (Amgen), antidiabetics from Japan Tobacco, AZD-4619, AZD-8450, AZD-8677 (AstraZeneca), DRF-10945, balaglitazone (Dr Reddy's), CS-00088, CS-00098 (Chipscreen Biosciences), E-3030 (Eisai), etalocib (CAS No. 161172-51-6) (EIi Lilly), GSK-641597 (Ligand), GSK-677954 (GlaxoSmithKline), GW-409544 (Ligand), GW- 590735 (GlaxoSmithKline), K-111 (Hoffmann-La Roche), LY-518674 (EIi Lilly), LY-674 (Ligand), LY-929 (Ligand), MC-3001, MC-3002 (MaxoCore Pharmaceuticals), metformin HCI + pioglitazone (CAS No. 1115-70-4 and 112529-15-4), ACTOPLUS MET from Andrx), muraglitazar (CAS No. 331741-94-7) (Bristol-Myers Squibb), naveglitazar (Ligand), oleoylethanolamide (Kadmus Pharmaceuticals), ONO-5129, pioglitazone hydrochloride (CAS No. 111025-46-8 and 112529-15-4) (Takeda), PLX-204 (Plexxikon), PPAR agonists from Genfit, PPAR delta agonists from EIi Lilly, PPAR-alpha agonists from CrystalGenomics, PPAR-gamma modulators and PPAR-β modulators from CareX, rosiglitazone maleate (CAS No. 122324- 73-4 or 155141-29-0) (GlaxoSmithKline), rosiglitazone maleate/glimepir (CAS No. 155141-29-0 and 93479-97-1), AVANDARYL, rosiglitazone maleate/metformin extend (CAS No. 155141-29-0 and 657-24-9), AVANDAMET, rosiglitazone maleate+metformin, AVANDAMET (GlaxoSmithKline), tesaglitazar (AstraZeneca), and an analogue or derivative thereof.
17186. The method of item 17109 wherein the agent is a phosphatase inhibitor.
17187. The method of item 17109 wherein the agent is a phosphodiesterase (PDE) inhibitor selected from the group consisting of avanafil (Tanabe Seiyaku), dasantafil (CAS No. 569351-91-3) (Schering- Plough), A-906119 (CAS No. 134072-58-5), DL-850 (Sanofi-Aventis), GRC- 3015, GRC-3566, GRC-3886 (Glenmark), HWA-153 (CAS No. 56395-66-5) (Sanofi-Aventis), hydroxypumafentrine (Altana), IBFB-130011 , IBFB-14-016, IBFB-140301 , IBFB-150007, IBFB-211913 (IBFB Pharma), L-826141 (Merck & Co), medorinone (CAS No. 88296-61-1) (Sanofi-Aventis), MEM- 1917 (Memory Pharmaceuticals), ND-1251 (Neuro3d), PDE inhibitors from ICOS, PDE IV inhibitors from Memory Pharmaceuticals and CrystalGenomics, Pharmaprojects No. 2742 and 6141 (Sanofi-Aventis), QAD-171 (Novartis), RHC-2963 (CAS No. 76993-12-9 and 76993-14-1), RPR-117658, RPR-122818 derivatives, SR-24870 , and RPR-132294 (Sanofi-Aventis), SK-350 (ln2Gen), stroke therapy agents from deCODE Genetics, TAS-203 (Taiho), tofimilast (CAS No. 185954-27-2) (Pfizer), UK- 371800 (Pfizer), WIN-65579 (CAS No. 158020-82-7) (Sanofi-Aventis), IBFB- 130020 (IBFB Pharma), OPC-6535 (CAS No. 145739-56-6) (Otsuka), a phosphodiesterase III inhibitor, enoximone, a phosphodiesterase IV inhibitor, fosfosal, Atopik (Barrier Therapeutics), triflusal, a phosphodiesterase V inhibitor, and an analogue or derivative thereof.
17188. The method of item 17109 wherein the agent is a PKC inhibitor.
17189. The method of item 17109 wherein the agent is a platelet activating factor antagonist.
17190. The method of item 17109 wherein the agent is a platelet-derived growth factor receptor kinase inhibitor.
17191. The method of item 17109 wherein the agent is a prolyl hydroxylase inhibitor.
17192. The method of item 17109 wherein the agent is a polymorphonuclear neutrophil inhibitor.
17193. The method of item 17109 wherein the agent is a protein kinase B inhibitor. 17194. The method of item 17109 wherein the agent is a protein kinase C stimulant.
17195. The method of item 17109 wherein the agent is a purine nucleoside analogue.
17196. The method of item 17109 wherein the agent is a purinoreceptor P2X antagonist.
17197. The method of item 17109 wherein the agent is a Raf kinase inhibitor.
17198. The method of item 17109 wherein the agent is a reversible inhibitor of ErbB1 and ErbB2.
17199. The method of item 17109 wherein the agent is a ribonucleoside triphosphate reductase inhibitor.
17200. The method of item 17109 wherein the agent is an SDF-1 antagonist.
17201. The method of item 17109 wherein the agent is a sheddase inhibitor.
17202. The method of item 17109 wherein the agent is an SRC inhibitor.
17203. The method of item 17109 wherein the agent is a stromelysin inhibitor. 17204. The method of item 17109 wherein the agent is an Syk kinase inhibitor.
17205. The method of item 17109 wherein the agent is a telomerase inhibitor.
17206. The method of item 17109 wherein the agent is a TGF beta inhibitor selected from the group consisting of pirfenidone (CAS No. 53179-13-8) (MARNAC), tranilast (CAS No. 53902-12-8) (Kissei), IN- 1130 (ln2Gen), mannose-6-phosphate (BTG), TGF-β antagonists from Inflazyme (Pharmaprojects No. 6075), TGF-β antagonists from Sydney, non-industrial source), TGF-β I receptor kinase inhibitors from EIi Lilly, TGF- β receptor inhibitors from Johnson & Johnson, and an analogue or derivative thereof.
17207. The method of item 17109 wherein the agent is a TNFα antagonist or TACE inhibitor selected from the group consisting of adalimumab (CAS No. 331731-18-1) (Cambridge Antibody Technology), AGIX-4207 (AtheroGenics), AGT-1 (Advanced Biotherapy), an antiinflammatory from Borean Pharma, Cellzome, or Paradigm Therapeutics, anti-inflammatory vaccine (TNF-alpha kinoid) from Neovacs, humanized anti-TNF antibody or an anti-TNF MAb (CB0006) Celltech (UCB), apratastat (CAS No. 287445-51-0) (Wyeth), BMS-561392 (Bristol-Myers Squibb), BN- 006 (Bone), certolizumab pegol (CAS No. 428863-50-7 or CH-138 (UCB), cilomilast (CAS No. 153259-65-5) (GlaxoSmithKline), CR-1 (Nuada Pharmaceuticals), CRx-119 (CombinatoRx), D-5410 (UCB), dacopafant (CAS No. 125372-33-0) (Sanofi-Aventis), dersalazine (CAS No. 188913-57- 7/188913-58-8) (Uriach), etanercept (CAS No. 185243-69-0) (Amgen), ethyl pyruvate (Critical (Critical Therapeutics), golimumab (CAS No. 476181-74-5) (Johnson & Johnson), hormono-immunotherapy from Ipsen, CDP571 (e.g., Humicade from UCB), IC-485 (ICOS), infliximab (CAS No. 170277-31-3) (Johnson & Johnson), IP-751 (Manhattan Pharmaceuticals), ISIS-104838 (CAS No. 250755-32-9) (ISIS Pharmaceuticals), lenalidomide (CAS No. 191732-72-6) (Celgene), lentinan (CAS No. 37339-90-5) (Ajinomoto), MDL- 201112 (CAS No. 142130-73-2) (Sanofi-Aventis), medroxyprogesterone (CAS No. 520-85-4) (InKine Pharmaceutical), N-acetylcysteine (CAS No. 616-91-1) (Zambon), NBE-P2 (DIREVO Biotech), nerelimomab (CAS No. 162774-06-3) (Chiron), OM-294DP (OM PHARMA), onercept (CAS No. 199685-57-9) (Yeda), PASSTNF-alpha (Verigen), pentoxifylline or oxypentifylline (Sanofi-Aventis), Pharmaprojects No. 4091, 4241 , 4295, or 4488 (Sanofi-Aventis), Pharmaprojects No. 5480 (Amgen), Pharmaprojects No. 6749 (Cengent), pirfenidone (CAS No. 53179-13-8) (MARNAC), RPR- 132294 (Sanofi-Aventis), S5 (F002) (Fulcrum Pharmaceuticals), simvastatin (CAS No. 79902-63-9) (Merck & Co), STA-6292 (Synta Pharmaceuticals), tacrolimus (CAS No. 104987-11-3) (Fujisawa LifeCycle Pharma), talactoferrin alfa (CAS No. 308240-58-6) (Agennix), thalidomide (CAS No. 50-35-1) (Celgene), TNF antagonists form ProStrakan, and Synergen, TNF inhibitors (Amgen), TNF-alpha antagonists from Dynavax Technologies and Jerina AG (Germany), TNF-alpha inhibitors from IBFB Pharma and Xencor (Xencor), torbafylline (CAS No. 105102-21-4) (Sanofi-Aventis), UR-1505 (Uriach), VT-346 (Viron Therapeutics), YSIL6 (Y1S Therapeutics), YSTH2 (Y's Therapeutics), NPI-1302a-3 (Nereus Pharmaceuticals, a TNF antagonist from Jerina AG (Germany), dersalazine, and an analogue or derivative thereof.
17208. The method of item 17109 wherein the agent is a Toll receptor inhibitor.
17209. The method of item 17109 wherein the agent is a tubulin antagonist. 17210. The method of item 17109 wherein the agent is a tyrosine kinase inhibitor selected from the group consisting of SU-011248, SUTENT from Pfizer Inc. (New York, NY), BMS-354825, PN-355 (Paracelsian Pharmaceuticals), AGN-199659 (Allergan), AAL-993 or ABP- 309 (Novartis), adaphostin (NIH), AEE-788 (Novartis), AG-013736 (OSI Pharmaceuticals), AG-13736 (Pfizer), ALT-110 (Alteris Therapeutics), AMG- 706 (Amgen), anticancer MAbs from Xencor, anti-EGFrvlll MAbs from Abgenix, anti-HER2 MAb from Abiogen, AZD-2171 or AZD-9935 (AstraZeneca), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), CEP-5214 (Cephalon), CEP-7055 (Cephalon), cetuximab (ImClone Systems), CHIR-200131 and CHIR-258 (Chiron), CP-547632 (OSI Pharmaceuticals), CP-724714 (Pfizer), CT-301 (Creabilis Therapeutics), D- 69491 (Baxter International), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EGFR/ErbB2 inhibitors from Array BioPharma, erlotinib (CAS No. 183319- 69-9) (OSI Pharmaceuticals), EXEL-2880 (Exelixis), FK-778 (Sanofi- Aventis) , gefitinib (CAS No. 184475-35-2) (AstraZeneca), GW-2286 or GW- 654652 (GlaxoSmithKline), her2/neu antigen from AlphaVax, HER-2/neu inhibitor from Generex, Herzyme (Medipad) (Sirna Therapeutics), HKI-272 (Wyeth), HuMax-EGFr (Genmab), idronoxil (CAS No. 81267-65-4) (Novogen), IGF-1 inhibitors from Ontogen, IMC-11F8 (ImClone Systems), kahalalide F (CAS No. 149204-42-2) (PharmaMar), KDR inhibitor from LG Life Sciences, KDR inhibitors from Abbott Laboratories, KDR kinase inhibitors (UCB), Kdr kinase inhibitors from Merck & Co, KRN-633 and KRN- 951 (Kirin Brewery), KSB-102 (Xenova), lapatinib ditosylate (CAS No. 388082-78-8) (GlaxoSmithKline), matuzumab (Merck KGaA), MDX-214 (Medarex), ME-103 (Pharmexa), MED-A300 (Gerolymatos), MNAC-13 (Lay Line Genomics), nimotuzumab (Center of Molecular Immunology), NSC- 330507 or NSC-707545 (NIH), NV-50 (Novogen), OSI-930 (OSI Pharmaceuticals), panitumumab (Abgenix), pelitinib (CAS No. 287933-82-7) (Wyeth), pertuzumab (CAS No. 380610-27-5) (Genentech), Pharmaprojects No. 3985 (Sanofi-Aventis), prostate cancer therapeutics from Sequenom (SQPC35, SQPC36, SQPC90), removab and remoxab (Trion Pharma), RG- 13022 (CAS No. 136831-48-6), RG-13291 (CAS No. 138989-50-1), or RG- 14620 (CAS No. 136831-49-7) (Sanofi-Aventis), RM-6427 (Romark), RNAi breast cancer therapy from Benitec, RP 53801 (CAS No. 125882-88-4) (Sanofi-Aventis), sorafenib tosylate (Bayer), SU-11657 (Pfizer), Tie-2 antagonists from Semaia (Hybrigenics), Tie-2 inhibitors from Ontogen, trastuzumab (CAS No. 180288-69-1) (Genentech), tyrosine kinase inhibitors from Sanofi-Aventis, U3-1287, U3-1565, U3-1784, U3-1800 (U3 Pharma), vatalanib (Novartis), VEGFR-2 kinase inhibitor from Bristol-Myers Squibb, XL-647 (Exelixis), ZD-6474 (AstraZeneca), ZK-CDK (Schering AG), an EGFR tyrosine kinase inhibitor, EKB-569 (Wyeth), and an analogue or derivative thereof.
17211. The method of item 17109 wherein the agent is a VEGF inhibitor.
17212. The method of item 17109 wherein the agent is a vitamin D receptor agonist.
17213. The method of item 17109 wherein the agent is ZD-6474 (an angiogenesis inhibitor).
17214. The method of item 17109 wherein the agent is AP-23573 (an mTOR inhibitor).
17215. The method of item 17109 wherein the agent is synthadotin (a tubulin antagonist).
17216. The method of item 17109 wherein the agent is S-0885 (a collagenase inhibitor). 17217. The method of item 17109 wherein the agent is aplidine (an elongation factor-1 alpha inhibitor).
17218. The method of item 17109 wherein the agent is ixabepilone (an epithilone).
17219. The method of item 17109 wherein the agent is IDN-5390 (an angiogenesis inhibitor).
17220. The method of item 17109 wherein the agent is SB-2723005 (an angiogenesis inhibitor).
17221. The method of item 17109 wherein the agent is ABT-518 (an angiogenesis inhibitor).
17222. The method of item 17109 wherein the agent is combretastatin (an angiogenesis inhibitor).
17223. The method of item 17109 wherein the agent is anecortave acetate (an angiogenesis inhibitor).
17224. The method of item 17109 wherein the agent is SB-715992 (a kinesin antagonist).
17225. The method of item 17109 wherein the agent is temsirolimus (an mTOR inhibitor).
17226. The method of item 17109 wherein the agent is adalimumab (a TNFα antagonist). 17227. The method of item 17109, wherein the composition comprises a polymer.
17228. The method of item 17109, wherein the composition comprises a polymeric carrier.
17229. The method of item 17109 wherein the anti- scarring agent inhibits adhesion between the device and a host into which the device is implanted.
17230. The method of item 17109 wherein the device delivers the anti-scarring agent locally to tissue proximate to the device.
17231. The method of item 17109 wherein the device has a coating that comprises the anti-scarring agent.
17232. The method of item 17109, wherein the device has a coating that comprises the agent and is disposed on a surface of the implant.
17233. The method of item 17109, wherein the device has a coating that comprises the agent and directly contacts the implant.
17234. The method of item 17109, wherein the device has a coating that comprises the agent and indirectly contacts the implant.
17235. The method of item 17109, wherein the device has a coating that comprises the agent and partially covers the implant.
17236. The method of item 17109, wherein the device has a coating that comprises the agent and completely covers the implant. 17237. The method of item 17109, wherein the device has a uniform coating.
17238. The method of item 17109, wherein the device has a non-uniform coating.
17239. The method of item 17109, wherein the device has a discontinuous coating.
17240. The method of item 17109, wherein the device has a patterned coating.
17241. The method of item 17109, wherein the device has a coating with a thickness of 100 Dm or less.
17242. The method of item 17109, wherein the device has a coating with a thickness of 10 Dm or less.
17243. The method of item 17109, wherein the device has a coating, and the coating adheres to the surface of the implant upon deployment of the implant.
17244. The method of item 17109, wherein the device has a coating, and wherein the coating is stable at room temperature for a period of 1 year.
17245. The method of item 17109, wherein the device has a coating, and wherein the anti-scarring agent is present in the coating in an amount ranging between about 0.0001% to about 1% by weight. 17246. The method of item 17109, wherein the device has a coating, and wherein the anti-scarring agent is present in the coating in an amount ranging between about 1% to about 10% by weight.
17247. The method of item 17109, wherein the device has a coating, and wherein the anti-scarring agent is present in the coating in an amount ranging between about 10% to about 25% by weight.
17248. The method of item 17109, wherein the device has a coating, and wherein the anti-scarring agent is present in the coating in an amount ranging between about 25% to about 70% by weight.
17249. The method of item 17109, wherein the device has a coating, and wherein the coating further comprises a polymer.
17250. The method of item 17109, wherein the device has a first coating having a first composition and a second coating having a second composition.
17251. The method of item 17109, wherein the device has a first coating having a first composition and a second coating having a second composition, wherein the first composition and the second composition are different.
17252. The method of item 17109, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a copolymer.
17253. The method of item 17109, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a block copolymer. 17254. The method of item 17109, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a random copolymer.
17255. The method of item 17109, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a biodegradable polymer.
17256. The method of item 17109, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a non-biodegradable polymer.
17257. The method of item 17109, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a hydrophilic polymer.
17258. The method of item 17109, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a hydrophobic polymer.
17259. The method of item 17109, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a polymer having hydrophilic domains.
17260. The method of item 17109, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a polymer having hydrophobic domains.
17261. The method of item 17109, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a non-conductive polymer. 17262. The method of item 17109, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises an elastomer.
17263. The method of item 17109, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a hydrogel.
17264. The method of item 17109, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a silicone polymer.
17265. The method of item 17109, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a hydrocarbon polymer.
17266. The method of item 17109, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a styrene-derived polymer.
17267. The method of item 17109, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a butadiene polymer.
17268. The method of item 17109, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a macromer.
17269. The method of item 17109, wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises a poly(ethylene glycol) polymer. 17270. The method of item 17109 wherein the composition comprises a polymeric carrier, and wherein the polymeric carrier comprises an amorphous polymer.
17271. The method of item 17109, wherein the device comprises a lubricious coating.
17272. The method of item 17109 wherein the anti- scarring agent is located within pores or holes of the device.
17273. The method of item 17109 wherein the anti- scarring agent is located within a channel, lumen, or divet of the device.
17274. The method of item 17109, wherein the device comprises a second pharmaceutically active agent.
17275. The method of item 17109 wherein the device comprises an anti-inflammatory agent.
17276. The method of item 17109 wherein the device comprises an agent that inhibits infection.
17277. The method of item 17109 wherein the device comprises an agent that inhibits infection, and wherein the agent is an anthracycline.
17278. The method of item 17109 wherein the device comprises an agent that inhibits infection, and wherein the agent is doxorubicin. 17279. The method of item 17109 wherein the device comprises an agent that inhibits infection, and wherein the agent is mitoxantrone.
17280. The method of item 17109 wherein the device comprises an agent that inhibits infection, and wherein the agent is a fluoropyrimidine.
17281. The method of item 17109 wherein the device comprises an agent that inhibits infection, and wherein the agent is 5- fluorouracil (5-FU).
17282. The method of item 17109 wherein the device comprises an agent that inhibits infection, and wherein the agent is a folic acid antagonist.
17283. The method of item 17109 wherein the device comprises an agent that inhibits infection, and wherein the agent is methotrexate.
17284. The method of item 17109 wherein the device comprises an agent that inhibits infection, and wherein the agent is a podophylotoxin.
17285. The method of item 17109 wherein the device comprises an agent that inhibits infection, and wherein the agent is etoposide.
17286. The method of item 17109 wherein the device comprises an agent that inhibits infection, and wherein the agent is a camptothecin. 17287. The method of item 17109 wherein the device comprises an agent that inhibits infection, and wherein the agent is a hydroxyurea.
17288. The method of item 17109 wherein the device comprises an agent that inhibits infection, and wherein the agent is a platinum complex.
17289. The method of item 17109 wherein the device comprises an agent that inhibits infection, and wherein the agent is cisplatin.
17290. The method of item 17109, further comprising an anti-thrombotic agent.
17291. The method of item 17109 wherein the device comprises a visualization agent.
17292. The method of item 17109 wherein the device comprises a visualization agent, wherein the visualization agent is a radiopaque material, and wherein the radiopaque material comprises a metal, a halogenated compound, or a barium containing compound.
17293. The method of item 17109 wherein the device comprises a visualization agent, wherein the visualization agent is a radiopaque material, and wherein the radiopaque material comprises barium, tantalum, or technetium.
17294. The method of item 17109 wherein the device comprises a visualization agent, and wherein the visualization agent is a MRI responsive material. 17295. The method of item 17109 wherein the device comprises a visualization agent, and wherein the visualization agent comprises a gadolinium chelate.
17296. The method of item 17109 wherein the device comprises a visualization agent, and wherein the visualization agent comprises iron, magnesium, manganese, copper, or chromium.
17297. The method of item 17109 wherein the device comprises a visualization agent, and wherein the visualization agent comprises an iron oxide compound.
17298. The method of item 17109 wherein the device comprises a visualization agent, and wherein the visualization agent comprises a dye, pigment, or colorant.
17299. The method of item 17109 wherein the device comprises an echogenic material.
17300. The method of item 17109 wherein the device comprises an echogenic material, and wherein the echogenic material is in the form of a coating.
17301. The method of item 17109 wherein the device is sterile.
17302. The method of item 17109 wherein the anti- scarring agent is released into tissue in the vicinity of the device after deployment of the device. 17303. The method of item 17109 wherein the anti- scarring agent is released into tissue in the vicinity of the device after deployment of the device, and wherein the tissue is connective tissue.
17304. The method of item 17109 wherein the anti- scarring agent is released into tissue in the vicinity of the device after deployment of the device, and wherein the tissue is muscle tissue.
17305. The method of item 17109 wherein the anti- scarring agent is released into tissue in the vicinity of the device after deployment of the device, and wherein the tissue is nerve tissue.
17306. The method of item 17109 wherein the anti- scarring agent is released into tissue in the vicinity of the device after deployment of the device, and wherein the tissue is epithelium tissue.
17307. The method of item 17109 wherein the anti- scarring agent is released in effective concentrations from the device over a period ranging from the time of deployment of the device to about 1 year.
17308. The method of item 17109 wherein the anti- scarring agent is released in effective concentrations from the device over a period ranging from about 1 month to 6 months.
17309. ' The method of item 17109 wherein the anti- scarring agent is released in effective concentrations from the device over a period ranging from about 1 - 90 days.
17310. The method of item 17109 wherein the anti- scarring agent is released in effective concentrations from the device at a constant rate. 17311. The method of item 17109 wherein the anti- scarring agent is released in effective concentrations from the device at an increasing rate.
17312. The method of item 17109 wherein the anti- scarring agent is released in effective concentrations from the device at a decreasing rate.
17313. The method of item 17109 wherein the anti- scarring agent is released in effective concentrations from the composition comprising the anti-scarring agent by diffusion over a period ranging from the time of deployment of the device to about 90 days.
17314. The method of item 17109 wherein the anti- scarring agent is released in effective concentrations from the composition comprising the anti-scarring agent by erosion of the composition over a period ranging from the time of deployment of the device to about 90 days.
17315. The method of item 17109 wherein the device comprises about 0.01 Dg to about 10 Dg of the anti-scarring agent.
17316. The method of item 17109 wherein the device comprises about 10 Dg to about 10 mg of the anti-scarring agent.
17317. The method of item 17109 wherein the device comprises about 10 mg to about 250 mg of the anti-scarring agent.
17318. The method of item 17109 wherein the device comprises about 250 mg to about 1000 mg of the anti-scarring agent. 17319. The method of item 17109 wherein the device comprises about 1000 mg to about 2500 mg of the anti-scarring agent.
17320. The method of item 17109 wherein a surface of the device comprises less than 0.01 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
17321. The method of item 17109 wherein a surface of the device comprises about 0.01 Dg to about 1 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
17322. The method of item 17109 wherein a surface of the device comprises about 1 Dg to about 10 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
17323. The method of item 17109 wherein a surface of the device comprises about 10 Dg to about 250 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
17324. The method of item 17109 wherein a surface of the device comprises about 250 Dg to about 1000 Dg of the anti-scarring agent of anti-scarring agent per mm2 of device surface to which the anti- scarring agent is applied.
17325. The method of item 17109 wherein a surface of the device comprises about 1000 Dg to about 2500 Dg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
17326. The method of item 17109 wherein the combining is performed by direct affixing the agent or the composition to the implant. 17327. The method of item 17109 wherein the combining is performed by spraying the agent or the component onto the implant.
17328. The method of item 17109 wherein the combining is performed by electrospraying the agent or the composition onto the implant.
17329. The method of item 17109 wherein the combining is performed by dipping the implant into a solution comprising the agent or the composition.
17330. The method of item 17109 wherein the combining is performed by covalently attaching the agent or the composition to the implant.
17331. The method of item 17109 wherein the combining is performed by non-covalently attaching the agent or the composition to the implant.
17332. The method of item 17109 wherein the combining is performed by coating the implant with a substance that contains the agent or the composition.
17333. The method of item 17109 wherein the combining is performed by coating the implant with a substance that absorbs the agent.
17334. The method of item 17109 wherein the combining is performed by interweaving a thread composed of, or coated with, the agent or the composition. 17335. The method of item 17109 wherein the combining is performed by covering all the implant with a sleeve that contains the agent or the composition.
17336. The method of item 17109 wherein the combining is performed by covering a portion of the implant with a sleeve that contains the agent or the composition.
17337. The method of item 17109 wherein the combining is performed by covering all the implant with a cover that contains the agent or the composition.
17338. The method of item 17109 wherein the combining is performed by covering a portion of the implant with a cover that contains the agent or the composition.
17339. The method of item 17109 wherein the combining is performed by covering all the implant with an electrospun fabric that contains the agent or the composition.
17340. The method of item 17109 wherein the combining is performed by covering a portion of the implant with an electrospun fabric that contains the agent or the composition.
17341. The method of item 17109 wherein the combining is performed by covering all the implant with a mesh that contains the agent or the composition.
17342. The method of item 17109 wherein the combining is performed by covering a portion of the implant with a mesh that contains the agent or the composition. 17343. The method of item 17109 wherein the combining is performed by constructing all the implant with the agent or the composition.
17344. The method of item 17109 wherein the combining is performed by constructing a portion of the implant with the agent or the composition.
17345. The method of item 17109 wherein the combining is performed by impregnating the implant with the agent or the composition.
17346. The method of item 17109 wherein the combining is performed by constructing all of the implant from a degradable polymer that releases the agent.
17347. The method of item 17109 wherein the combining is performed by constructing a portion of the implant from a degradable polymer that releases the agent.
17348. The method of item 17109 wherein the combining is performed by dipping the implant into a solution that comprise the agent and an inert solvent for the implant.
17349. The method of item 17109 wherein the combining is performed by dipping the implant into a solution that comprises the agent and a solvent that will swell the implant.
17350. The method of item 17109 wherein the combining is performed by dipping the implant into a solution that comprises the agent and a solvent that will dissolve the implant. 17351. The method of item 17109 wherein the combining is performed by dipping the implant into a solution that comprises the agent, a polymer and an inert solvent for the implant.
17352. The method of item 17109 wherein the combining is performed by dipping the implant into a solution that comprises the agent, a polymer and a solvent that will swell the implant.
17353. The method of item 17109 wherein the combining is performed by dipping the implant into a solution that comprises the agent, a polymer and a solvent that will dissolve the implant.
17354. The method of item 17109 wherein the combining is performed by spraying the implant into a solution that comprises the agent and an inert solvent for the implant.
17355. The method of item 17109 wherein the combining is performed by spraying the implant into a solution that comprises the agent and a solvent that will swell the implant.
17356. The method of item 17109 wherein the combining is performed by spraying the implant into a solution that comprises the agent and a solvent that will dissolve the implant.
17357. The method of item 17109 wherein the combining is performed by spraying the implant into a solution that comprises the agent, a polymer and an inert solvent for the implant.
17358. The method of item 17109 wherein the combining is performed by spraying the implant into a solution that comprises the agent, a polymer and a solvent that will swell the implant. 17359. The method of item 17109 wherein the combining is performed by spraying the implant into a solution that comprises the agent, a polymer and a solvent that will dissolve the implant.
17360. A method for implanting a medical device comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with i) an anti-fibrotic agent, ii) an anti-infective agent, iii) a polymer, iv) a composition comprising an anti-fibrotic agent and a polymer, v) a composistion comprising an anti-infective agent and a polymer, or vi) a composition comprising an anti-fibrotic agent, an anti- infective agent and a polymer, and (b) implanting the medical device into the host.
17361. The method of item 17360 wherein the anti- fibrotic agent is an adensosine A2A receptor antagonist.
17362. The method of item 17360 wherein the anti- fibrotic agent is an AKT inhibitor.
17363. The method of item 17360 wherein the anti- fibrotic agent is an alpha 2 integrin antagonist, wherein the alpha 2 integrin antagonist is Pharmaprojects No. 5754 (Merck KgaA).
17364. The method of item 17360 wherein the anti- fibrotic agent is an alpha 4 integrin antagonist.
17365. The method of item 17360 wherein the anti- fibrotic agent is an alpha 7 nicotinic receptor agonist.
17366. The method of item 17360 wherein the anti- fibrotic agent is an angiogenesis inhibitor selected from the group consisting of AG-12,958 (Pfizer), ATN-161 (Attenuon LLC), neovastat, an angiogenesis inhibitor from Jerina AG (Germany), NM-3 (Mercian), VGA-1155 (Taisho), FCE-26644 (Pfizer), FCE-26950 (Pfizer), FPMA (Meiji Daries), FR-111142 (Fujisawa), GGTI-298, GM-1306 (Ligand), GPA-1734 (Novartis), NNC-47- 0011 (Novo Nordisk), herbamycin (Nippon Kayaku), lenalidomide (Celegene), IP-10 (NIH), ABT-828 (Abbott), KIN-841 (Tokushima University, Japan), SF-1126 (Semafore Pharmaceuticals), laminin technology (NIH), CHIR-258 (Chiron), NVP-AEW541 (Novartis), NVP-AEW541 (Novartis), Vt16907 (Alchemia), OXI-8007 (Oxigene), EG-3306 (Ark Therapeutics), Maspin (Arriva), ABT-567 (Abbott), PPI-2458 (Praecis Pharmaceuticals), CC-5079, CC-4089 (Celgene), HIF-1 alpha inhibitors (Xenova), S-247 (Pfizer), AP-23573 (Ariad), AZD-9935 (Astra Zeneca), mebendazole (Introgen Therapeutics), MetAP-2 inhibitors (GlaxoSmithKline), AG-615 (Angiogene Pharmaceuticals), Tie-2 antagonists (Hybrigenics), NC-381 , CYC-381 , NC-169, NC-219, NC-383, NC-384, NC-407 (Lorus Therapeutics), ATN-224 (Attenuon), ON-01370 (Onconova), Vitronectin antagonists (Amgen), SDX-103 (Salmedix), Vitronectin antagonists (Shire), CHP (Riemser), TEK (Amgen), Anecortave acetate (Alcon), T46.2 (Matrix Therapeutics), HG-2 (Heptagen), TEM antagonists (Genzyme), Oxi-4500 (Oxigene), ATN-161 (Attenuon), WX-293 (Wilex), M-2025 (Metris Therapeutics), Alphastatin (BioActa), YH-16 (Yantai Rongchang), BIBF- 1120 (Boehringer Ingelheim), BAY-57-9352 (Bayer), AS-1404 (Cancer Research Technology), SC-77964 (Pfizer), glycomimetics (BioTie Therapies), TIE-2 Inhibitors (Ontogen), DIMI, Octamer (Octamer), ABR- 215050 (Active Biotech), ABT-518 (Abbott), KDR inhibitors (Abbott), BSF- 466895 (Abbott), SCH-221153 (Schering-Plough), DAC:antiangiogenic (ConjuChem), TFPI (EntreMed), AZD-2171 (Astra-Zenaca), CDC-394 (Celgene), LY290293 (EIi Lilly), IDN-5390 (Indena), Kdr Kinase Inhibitors (Merck), CT-113020, CT-116433, CT-116563, CT-31890, CT-32228) (Cell Therapeutics), A-299620 (Abbott), TWEAK Inhibitor (Amgen), VEGF modulators (Johnson and Johnson), Tum-N53, tumstatin (Genzyme), Thios- 1, Thios-2 (Thios Pharmaceuticals), MV-6401 (Miravant Medical Technologies), Spisulosine (PharmaMar), CEP-7055 (Cephalon), AUV-201 (Auvation), LM-609 (EIi Lilly), SKF-106615 (AnorMED), Oglufanide disodium (Cytran), BW-114 (Pharminox), Calreticulin (NIH), WX-678 (Wilex), SD-7784 (Pfizer), WX-UK1 (Wilex), SH-268 (Schering AG), 2-Me-PGA (Celgene), S- 137 (Pfizer), ZD-6126 (Angiogene Pharmaceuticals), SG-292 (SignalGen), Benefin (Lane Labs), A6, A36 (Angstrom), SB-2723005 (GlaxoSmithKline), SC-7 (Cell Therapeutics), ZEN-014 (AEterna Zentaris), 2-methoxyestradiol (EntreMed), NK-130119 (Nippon Kayaku), CC-10004 (Celgene), AVE- 8062A (Ajinomoto), Tacedinaline (Pfizer), Actinonin (Tokyo Metropolitan Institute of Medical Science), Lenalidomide (Celgene), VGA-1155, BTO-956 (SRI International), ER-68203-00 (Eisai), CT-6685 (UCB), JKC-362 (Phoenix Pharmaceuticals), DMI-3798 (DMI Biosciences, Angiomate (Ipsen), ZD-6474 (AstraZeneca), CEP-5214 (Cephalon), Canstatin (Genzyme), NM-3 (Mercian), Oridigm (MediQuest Therapeutics), Exherin (Adherex), BLS-0597 (Boston Life Sciences), PTC-299 (PTC Therapeutics), NPI-2358 (Nereus Pharmaceuticals), CGP-79787 (Novartis), AEE-788 (Novartis), CKD-732 (Chong Kun Dang), CP-564959 (OSI Pharmaceuticals), CM-101 (CarboMed), CT-2584, CT3501 (Cell Therapeutics), combretastatin and analogues and derivatives thereof (Oxigene), Rebimastat (Bristol-Meyers Squibb), Dextrin 2-sulfate (ML Laboratories), Cilengitide (Merk KGaA), NSC-706704 (Pharminox), KRN- 951 (Kirin Brewery), Ukrain, NSC-631570 (Nowicky Pharma), Tecogalan sodium (Daiichi Pharmaceutical), Tz-93 (Tsumura), TBC-1635 (Encysive Pharmaceuticals), TAN-1120 (Takeda), Semaxanib (Pfizer), BDI-7800 (Biopharmacopae), SD-186, SD-983 (Bristol-Meyers Squibb), SB-223245 (GlaxoSmithKline), SC-236 (Pfizer), RWJ-590973 (Johnson and Johnson), ILX-1850 (Genzyme), SC-68488, S-836 (Pfizer), CG-55069-11 (CuraGen), Ki-23057 (Kirin Brewery), CCX-700 (Chemoentryx), Pegaptanib octasodium (Giled Sciences), ANGIOCOL (available from Biostratum Inc.), or an analogue or derivative thereof. 17367. The method of item 17360 wherein the anti- fibrotic agent is an apoptosis antagonist.
17368. The method of item 17360 wherein the anti- fibrotic agent is an apoptosis activator.
17369. The method of item 17360 wherein the anti- fibrotic agent is a beta 1 integrin antagonist.
17370. The method of item 17360 wherein the anti- fibrotic agent is a beta tubulin inhibitor.
17371. The method of item 17360 wherein the anti- fibrotic agent is a blocker of enzyme production in Hepatitis C.
17372. The method of item 17360 wherein the anti- fibrotic agent is a Bruton's tyrosine kinase inhibitor.
17373. The method of item 17360 wherein the anti- fibrotic agent is a calcineurin inhibitor.
17374. The method of item 17360 wherein the anti- fibrotic agent is a caspase 3 inhibitor.
17375. The method of item 17360 wherein the anti- fibrotic agent is a CC chemokine receptor antagonist.
17376. The method of item 17360 wherein the anti- fibrotic agent is a cell cycle inhibitor selected from the group consisting of SNS-595 (Sunesis), synthadotin, KRX-0403, and an analogue or derivative thereof. 17377. The method of item 17360 wherein the anti- fibrotic agent is a cathepsin B inhibitor.
17378. The method of item 17360 wherein the anti- fibrotic agent is a cathepsin K inhibitor, wherein the cathepsin K inhibitor is 462795 (GlaxoSmithKline), INPL-022-D6 (Amura Therapeutics), or an analogue or derivative thereof.
17379. The method of item 17360 wherein the anti- fibrotic agent is a cathepsin L inhibitor.
17380. The method of item 17360 wherein the anti- fibrotic agent is a CD40 antagonist.
17381. The method of item 17360 wherein the anti- fibrotic agent is a chemokine receptor agonist.
17382. The method of item 17360 wherein the anti- fibrotic agent is a chymase inhibitor.
17383. The method of item 17360 wherein the anti- fibrotic agent is a collagenase antagonist.
17384. The method of item 17360 wherein the anti- fibrotic agent is a CXCR antagonist.
17385. The method of item 17360 wherein the anti- fibrotic agent is a cyclin dependent kinase inhibitor selected from the group consisting of a CDK- 1 inhibitor, a CDK-2 inhibitor, a CDK- 4 inhibitor, a CDK-6 inhibitor, a CAK1 inhibitor from GPC Biotech and Bristol-Myers Squibb, RGB-286199 (GPC Biotech), an anticancer agent from Astex Technology, a CAK1 inhibitor from GPC Biotech, a CDK inhibitor from Sanofi-Aventis, a CDK1/CDK2 inhibitor from Amgen, a CDK2 inhibitor from SUGEN-2 (Pfizer), a hearing loss therapy agent (Sound Pharmaceuticals), PD-0332991 (Pfizer), RGB-286199 (GPC Biotech), Ro-0505124 (Hoffmann- La Roche), a Ser/Thr kinase inhibitor from Lilly (EIi Lilly), CVT-2584 (CAS No. 199986-75-9) (CV Therapeutics), and an analogue or derivative thereof.
17386. The method of item 17360 wherein the anti- fibrotic agent is a cyclooxygenase 1 inhibitor.
17387. The method of item 17360 wherein the anti- fibrotic agent is a DHFR inhibitor.
17388. The method of item 17360 wherein the anti- fibrotic agent is a dual integrin inhibitor.
17389. The method of item 17360 wherein the anti- fibrotic agent is an elastase inhibitor.
17390. The method of item 17360 wherein the anti- fibrotic agent is an elongation factor-1 alpha inhibitor.
17391. The method of item 17360 wherein the anti- fibrotic agent is an endothelial growth factor antagonist.
17392. The method of item 17360 wherein the anti- fibrotic agent is an endothelial growth factor receptor kinase inhibitor selected from the group consisting of sorafenib tosylate (Bayer), AAL-993 (Novartis), ABP-309 (Novartis), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EXEL- 2880 (Exelixis), GW-654652 (GlaxoSmithKline), a KDR inhibitor from LG Life Sciences, CT-6685 and CT-6729 (UCB)1 KRN-633 and KRN-951 (Kirin Brewery), OSI-930 (OSI Pharmaceuticals), SP-5.2 (Supratek Pharma), SU- 11657 (Pfizer), a Tie-2 antagonist (Hybrigenics), a VEGFR-2 kinase inhibitor (Bristol-Myers Squibb), XL-647 (Exelixis), a KDR inhibitor from Abbott Laboratories, sorafenib tosylate, and an analogue or derivative thereof.
17393. The method of item 17360 wherein the anti- fibrotic agent is an endotoxin antagonist.
17394. The method of item 17360 wherein the anti- fibrotic agent is an epothilone and tubulin binder.
17395. The method of item 17360 wherein the anti- fibrotic agent is an estrogen receptor antagonist.
17396. The method of item 17360 wherein the anti- fibrotic agent is an FGF inhibitor.
17397. The method of item 17360 wherein the anti- fibrotic agent is a farnexyl transferase inhibitor.
17398. The method of item 17360 wherein the anti- fibrotic agent is farnesyltransferase inhibitor selected from the group of A- 197574 (Abbott), a farnesyltransferase inhibitor from Servier, FPTIII (Strathclyde Institute for Drug R), LB-42908 (LG Life Sciences), Pharmaprojects No. 5063 (Genzyme), Pharmaprojects No. 5597 (Ipsen), Yissum Project No. B-1055 (Yissum), and an analogue or derivative thereof.
17399. The method of item 17360 wherein the anti- fibrotic agent is an FLT-3 kinase inhibitor. 17400. The method of item 17360 wherein the anti- fibrotic agent is an FGF receptor kinase inhibitor.
17401. The method of item 17360 wherein the anti- fibrotic agent is a fibrinogen antagonist selected from the group consisting of AUV-201 (Auvation), MG-13926 (Sanofi-Aventis), plasminogen activator (CAS No. 105913-11-9) (from Sanofi-Aventis or UCB), plasminogen activator-2 (tPA-2) (Sanofi-Aventis), pro-urokinase (CAS No. 82657-92-9) (Sanofi-Aventis), and an analogue or derivative thereof.
17402. The method of item 17360 wherein the anti- fibrotic agent is a heat shock protein 90 antagonist selected from the group consisting of SRN-005 (Sirenade), geldanamycin, NSC-33050 (17- allylaminogeldanamycin; 17-AAG), 17-dimethylaminoethylamino-17- demethoxy-geldanamycin (17-DMAG), rifabutin (rifamycin XIV, 1',4- didehydro-1-deoxy-1 ,4-dihydro-5'-(2-methylpropyl)-1-oxo-), and an analogue or derivative thereof.
17403. The method of item 17360 wherein the anti- fibrotic agent is a histone deacetylase inhibitor.
17404. The method of item 17360 wherein the anti- fibrotic agent is an HMGCoA reductase inhibitor selected from the group consisting of an atherosclerosis therapeutic from Lipid Sciences, ATM 6000 (ARYx Therapeutics), KS-01-019 (Kos Pharmaceuticals), Pharmaprojects No. 2197 (Sanofi-Aventi), RP 61969 (Sanofi-Aventis), and an analogue or derivative thereof.
17405. The method of item 17360 wherein the anti- fibrotic agent is an ICAM inhibitor. 17406. The method of item 17360 wherein the anti- fibrotic agent is an IL, ICE and IRAK antagonist, wherein the antagonist is a CJ-14877, CP-424174 (Pfizer), NF-61 (Negma-Lerads), and an analogue or derivative thereof.
17407. The method of item 17360 wherein the anti- fibrotic agent is an IL-2 inhibitor.
17408. The method of item 17360 wherein the anti- fibrotic agent is an immunosuppressant selected from the group consisting of teriflunomide (Sanofi Aventis), chlorsulfaquinoxalone (NSC-339004), chlorsulfaquinoxalone sulfate, CS-712 (Sankyo), ismomultin alfa (CAS No. 457913-93-8) (Akzo Nobel), antiallergics from GenPat77, antiinflammatories or AT-005 (Androclus Therapeutics), autoimmune disease therapeutics from EpiVax, BN-007 (Bone), budesonide (CAS No. 51333-22- 3) (MAP Pharmaceuticals), CO-14 (Genzyme), edratide (CAS No. 433922- 67-9) (Teva), EP-314 (Enanta), eprovafen (CAS No. 101335-99-3) (Sanofi- Aventis), HWA-131 (CAS No. 118788-41-3) (Sanofi-Aventis), immunomodulators from MerLion Pharmaceuticals, immunosuppressives from Alchemia, IPL-12 (Inflazyme), MDL-9563 (CAS No. 27086-86-8) (Sanofi-Aventis), Pharmaprojects No. 2330 (Sanofi-Aventis), Pharmaprojects No. 6426 (Abgenix), PXS-25 (Pharmaxis), rosmarinic acid (CAS No. 20283-92-5) (Sanofi-Aventis), RP 42927 or RP 54745 (CAS No. 135330-08-4) (Sanofi-Aventis), SGN-35 (Seattle Genetics), ST-1959 (Sigma-Tau), type I diabetes therapy from SYNX Pharma, UNIL-88 (Debiopharm), VP-025 (Vasogen), VR-694 (Vectura), PRTX-001 (Protalex), and an analogue or derivative thereof.
17409. The method of item 17360 wherein the anti- fibrotic agent is an IMPDH (inosine monophosphate). 17410. The method of item 17360 wherein the anti- fibrotic agent is an integrin antagonist.
17411. The method of item 17360 wherein the anti- fibrotic agent is an interleukin antagonist.
17412. The method of item 17360 wherein the anti- fibrotic agent is an inhibitor of type III receptor tyrosine kinase.
17413. The method of item 17360 wherein the anti- fibrotic agent is an irreversible inhibitor of enzyme methionine aminopeptidase type 2.
17414. The method of item 17360 wherein the anti- fibrotic agent is an isozyme selective delta protein kinase C inhibitor.
17415. The method of item 17360 wherein the anti- fibrotic agent a JAK3 enzyme inhibitor.
17416. The method of item 17360 wherein the anti- fibrotic agent is a JNK inhibitor.
17417. The method of item 17360 wherein the anti- fibrotic agent is a kinase inhibitor.
17418. The method of item 17360 wherein the anti- fibrotic agent is kinesin antagonist.
17419. The method of item 17360 wherein the anti- fibrotic agent is a kinesin antagonist. 17420. The method of item 17360 wherein the anti- fibrotic agent is a leukotriene inhibitor and antagonist selected from the group consisting of ambicromil (CAS No. 58805-38-2) (Sanofi-Aventis), amelubant (CAS No. 346735-24-8) (Boehringer Ingelheim), DW-1141 (Dong Wha), ebselen (Daiichi Pharmaceutical), ibudilast (Kyorin), leucotriene inhibitors from Sanofi-Aventis, lymphotoxin-beta receptor (LT-D) from Biogen Idee, Pharmaprojects No. 1535 and 2728 (CAS No. 119340-33-9) (Sanofi-Aventis), R-112 (Rigel), Rev-5367 (CAS No. 92532-05-3) (Sanofi- Aventis), RG-14893 (CAS No. 141835-49-6) (Sanofi-Aventis), RG-5901-A (CAS No. 101910-24-1), 92532-23-5, RP 66153 (CAS No. 142422-79-5), RP 66364 (CAS No.186916-92-5), RP 69698 (CAS No. 141748-00-7) (Sanofi-Aventis), SC-411930 (Pfizer), SC-41930 (CAS No. 120072-59-5) (Pfizer), SC-50605 (CAS No. 138828-39-4) (Pfizer), SC-51146 (CAS No. 141059-52-1), SC-53228 (CAS No. 153633-01-3) (Pfizer), spaglumic acid (ZY-15106) (CAS No.3106-85-2), 80619-64-3 (Novartis), tipredane (CAS No. 85197-77-9) (Bristol-Myers Squibb), U-75302 (CAS No. 119477-85-9) (Pfizer), and analogue or derivative thereof.
17421. The method of item 17360 wherein the anti- fibrotic agent is an MAP kinase inhibitor.
17422. The method of item 17360 wherein the anti- fibrotic agent is a matrix metalloproteinase inhibitor.
17423. The method of item 17360 wherein the anti- fibrotic agent is an MCP-CCR2 inhibitor.
17424. The method of item 17360 wherein the anti- fibrotic agent is an mTOR inhibitor. 17425. The method of item 17360 wherein the anti- fibrotic agent is an mTOR kinase inhibitor.
17426. The method of item 17360 wherein the anti- fibrotic agent is a microtubule inhibitor selected from the group consisting of antibody-maytansinoid conjugates from Biogen Idee, colchicines (MantiCore Pharmaceuticals), anticancer immunoconjugates from Johnson & Johnson, DIME from Octamer, gni-1f (GNI), huC242-DM4, huMy9-6-DM1 (ImmunoGen), IDN-5404 (Indena), IMO-098, IMOderm (Imotep), mebendazole (Introgen Therapeutics), microtubule poisons from Cambridge Enterprise, paclitaxel such as LOTAX from Aphios (CAS No. 33069-62-4), Genexol-PM from Samyang, Pharmaprojects No. 6383 (Tapestry Pharmaceuticals), RPR-112378 (Sanofi-Aventis), SGN-75 (Seattle Genetics), SPL-7435 (Starpharma), SSR-250411 (Sanofi-Aventis), trastuzumab-DM1 (Genentech), vinorelbine, and an analogue or derivative thereof.
17427. The method of item 17360 wherein the anti- fibrotic agent is an MIF inhibitor.
17428. The method of item 17360 wherein the anti- fibrotic agent is an MMP inhibitor.
17429. The method of item 17360 wherein the anti- fibrotic agent is a neurokinin (NK) antagonist selected from the group consisting of anthrotainin (CAS No. 148084-40-6) (Sanofi-Aventis), an IBS thereapeutic from ArQuIe, MDL-105212A (CAS No. 167261-60-1) (Ssanofi- Aventis), Pharmaprojects No. 2744, 3258 (CAS No. 139167-47-8) 4006, 4201 , or 5986 (Sanofi-Aventis), RP 67580 (CAS No. 135911-02-3), SR- 144190 (CAS No. 201152-86-5), SSR-240600, SSR-241586 (Sanofi- Aventis), TKA-457 (Novartis), vestipitant mesylate (CAS No. 334476-64-1) (GlaxoSmithKline), Win-64821 (Sanofi-Aventis), PRX-96026 (Predix Pharmaceuticals), and an analogue or derivative thereof.
17430. The method of item 17360 wherein the anti- fibrotic agent is an NF kappa B inhibitor selected from the group consisting of AVE-0545 or AVE-0547 (Sanofi-Aventis), bortezomib (CAS No. 179324- 69-7) (Millennium Pharmaceuticals), dexanabinol (CAS No. 112924-45-5) (Pharmos), dexlipotam (Viatris), Pharmaprojects No. 6283 (INDRA) (OXiGENE), IPL-576092 (CAS No. 137571-30-3) (Inflazyme), NFKB decoy (Corgentech), NFKB decoy oligo (AnGes MG), NFKB's from Ariad, osteoporosis treatments or S5 (F005) from Fulcrum Pharmaceuticals, P61 (Phytopharm), R-flurbiprofen (CAS No. 5104-49-4) (Encore Pharmaceuticals), and an analogue or derivative thereof.
17431. The method of item 17360 wherein the anti- fibrotic agent is a nitric oxide agonist.
17432. The method of item 17360 wherein the anti- fibrotic agent is an ornithine decarboxylase inhibitor.
17433. The method of item 17360 wherein the anti- fibrotic agent is a p38 MAP kinase inhibitor selected from the group consisting of AZD-6703 (AstraZeneca), JX-401 (Jexys Pharmaceuticals), BMS-2 (Bristol-Myers Squibb), a p38 MAP kinase inhibitor from Novartis, a p38-alpha MAP kinase inhibitor from Amphora, Pharmaprojects No. 5704 (Pharmacopeia), RPR-200765A (Sanofi-Aventis), SD-282 (Johnson & Johnson), TAK-715 (Takeda), and an analogue or derivative thereof.
17434. The method of item 17360 wherein the anti- fibrotic agent is a palmitoyl-protein thioesterase inhibitor. 17435. The method of item 17360 wherein the anti- fibrotic agent is a PDGF receptor kinase inhibitor selected from the group consisting of AAL-993, AMN-107, or ABP-309 (Novartis), AMG-706 (Amgen), BAY-57-9352 (Bayer), CDP-860 (UCB), E-7080 (Eisai), imatinib (CAS No. 152459-95-5) (Novartis), OSI-930 (OSI Pharmaceuticals), RPR- 127963E (Sanofi-Aventis), RWJ-540973 (Johnson & Johnson), sorafenib tosylate (Bayer), SU-11657 (Pfizer), tandutinib (CAS No. 387867-13-2) (Millennium Pharmaceuticals), vatalanib (Novartis), ZK-CDK (Schering AG), and an analogue or derivative thereof.
17436. The method of item 17360 wherein the anti- fibrotic agent is (-)-halofenate (Metabolex), AMG-131 (Amgen), antidiabetics from Japan Tobacco, AZD-4619, AZD-8450, AZD-8677 (AstraZeneca), DRF-10945, balaglitazone (Dr Reddy's), CS-00088, CS-00098 (Chipscreen Biosciences), E-3030 (Eisai), etalocib (CAS No. 161172-51-6) (EIi Lilly), GSK-641597 (Ligand), GSK-677954 (GlaxoSmithKline), GW-409544 (Ligand), GW-590735 (GlaxoSmithKline), K-111 (Hoffmann-La Roche), LY- 518674 (EIi Lilly), LY-674 (Ligand), LY-929 (Ligand), MC-3001, MC-3002 (MaxoCore Pharmaceuticals), metformin HCI + pioglitazone (CAS No. 1115- 70-4 and 112529-15-4), ACTOPLUS MET from Andrx), muraglitazar (CAS No. 331741-94-7) (Bristol-Myers Squibb), naveglitazar (Ligand), oleoylethanolamide (Kadmus Pharmaceuticals), ONO-5129, pioglitazone hydrochloride (CAS No. 111025-46-8 and 112529-15-4) (Takeda), PLX-204 (Plexxikon), PPAR agonists from Genfit, PPAR delta agonists from EIi Lilly, PPAR-alpha agonists from CrystalGenomics, PPAR-gamma modulators and PPAR-β modulators from CareX, rosiglitazone maleate (CAS No. 122324- 73-4 or 155141-29-0) (GlaxoSmithKline), rosiglitazone maleate/glimepir (CAS No. 155141-29-0 and 93479-97-1), AVANDARYL, rosiglitazone maleate/metformin extend (CAS No. 155141-29-0 and 657-24-9), AVANDAMET, rosiglitazone maleate+metformin, AVANDAMET (GlaxoSmithKline), tesaglitazar (AstraZeneca), and an analogue or derivative thereof.
17437. The method of item 17360 wherein the anti- fibrotic agent is a phosphatase inhibitor.
17438. The method of item 17360 wherein the anti- fibrotic agent is a phosphodiesterase (PDE) inhibitor selected from the group consisting of avanafil (Tanabe Seiyaku), dasantafil (CAS No. 569351- 91-3) (Schering-Plough), A-906119 (CAS No. 134072-58-5), DL-850 (Sanofi-Aventis), GRC-3015, GRC-3566, GRC-3886 (Glenmark), HWA-153 (CAS No. 56395-66-5) (Sanofi-Aventis), hydroxypumafentrine (Altana), IBFB-130011, IBFB-14-016, IBFB-140301, IBFB-150007, IBFB-211913 (IBFB Pharma), L-826141 (Merck & Co), medorinone (CAS No. 88296-61-1) (Sanofi-Aventis), MEM-1917 (Memory Pharmaceuticals), ND-1251 (Neuro3d), PDE inhibitors from ICOS, PDE IV inhibitors from Memory Pharmaceuticals and CrystalGenomics, Pharmaprojects No. 2742 and 6141 (Sanofi-Aventis), QAD-171 (Novartis), RHC-2963 (CAS No. 76993-12-9 and 76993-14-1), RPR-117658, RPR-122818 derivatives, SR-24870 , and RPR- 132294 (Sanofi-Aventis), SK-350 (ln2Gen), stroke therapy agents from deCODE Genetics, TAS-203 (Taiho), tofimilast (CAS No. 185954-27-2) (Pfizer), UK-371800 (Pfizer), WIN-65579 (CAS No. 158020-82-7) (Sanofi- Aventis), IBFB-130020 (IBFB Pharma), OPC-6535 (CAS No. 145739-56-6) (Otsuka), a phosphodiesterase III inhibitor, enoximone, a phosphodiesterase IV inhibitor, fosfosal, Atopik (Barrier Therapeutics), triflusal, a phosphodiesterase V inhibitor, and an analogue or derivative thereof.
17439. The method of item 17360 wherein the anti- fibrotic agent is a PKC inhibitor. 17440. The method of item 17360 wherein the anti- fibrotic agent is a platelet activating factor antagonist.
17441. The method of item 17360 wherein the anti- fibrotic agent is a platelet-derived growth factor receptor kinase inhibitor.
17442. The method of item 17360 wherein the anti- fibrotic agent is a prolyl hydroxylase inhibitor.
17443. The method of item 17360 wherein the anti- fibrotic agent is a polymorphonuclear neutrophil inhibitor.
17444. The method of item 17360 wherein the anti- fibrotic agent is a protein kinase B inhibitor.
17445. The method of item 17360 wherein the anti- fibrotic agent is a protein kinase C stimulant.
17446. The method of item 17360 wherein the anti- fibrotic agent is a purine nucleoside analogue.
17447. The method of item 17360 wherein the anti- fibrotic agent is a purinoreceptor P2X antagonist.
17448. The method of item 17360 wherein the anti- fibrotic agent is a Raf kinase inhibitor.
17449. The method of item 17360 wherein the anti- fibrotic agent is a reversible inhibitor of ErbB1 and ErbB2. 17450. The method of item 17360 wherein the anti- fibrotic agent is a ribonucleoside triphosphate reductase inhibitor.
17451. The method of item 17360 wherein the anti- fibrotic agent is an SDF-1 antagonist.
17452. The method of item 17360 wherein the anti- fibrotic agent is a sheddase inhibitor.
17453. The method of item 17360 wherein the anti- fibrotic agent is an SRC inhibitor.
17454. The method of item 17360 wherein the anti- fibrotic agent is a stromelysin inhibitor.
17455. The method of item 17360 wherein the anti- fibrotic agent is an Syk kinase inhibitor.
17456. The method of item 17360 wherein the anti- fibrotic agent is a telomerase inhibitor.
17457. The method of item 17360 wherein the anti- fibrotic agent is a TGF beta inhibitor selected from the group consisting of pirfenidone (CAS No. 53179-13-8) (MARNAC), tranilast (CAS No. 53902- 12-8) (Kissei), IN-1130 (ln2Gen), mannose-6-phosphate (BTG), TGF-β antagonists from Inflazyme (Pharmaprojects No. 6075), TGF-β antagonists from Sydney, non-industrial source), TGF-β I receptor kinase inhibitors from EIi Lilly, TGF-β receptor inhibitors from Johnson & Johnson, and an analogue or derivative thereof. 17458. The method of item 17360 wherein the anti- fibrotic agent is a TNFα antagonist or TACE inhibitor selected from the group consisting of adalimumab (CAS No. 331731-18-1) (Cambridge Antibody Technology), AGIX-4207 (AtheroGenics), AGT-1 (Advanced Biotherapy), an anti-inflammatory from Borean Pharma, Cellzome, or Paradigm Therapeutics, anti-inflammatory vaccine (TNF-alpha kinoid) from Neovacs, humanized anti-TNF antibody or an anti-TNF MAb (CB0006) Celltech (UCB), apratastat (CAS No. 287445-51-0) (Wyeth), BMS-561392 (Bristol-Myers Squibb), BN-006 (Bone), certolizumab pegol (CAS No. 428863-50-7 or CH-138 (UCB), cilomilast (CAS No. 153259-65-5) (GlaxoSmithKline), CR-1 (Nuada Pharmaceuticals), CRx-119 (CombinatoRx), D-5410 (UCB), dacopafant (CAS No. 125372-33-0) (Sanofi- Aventis), dersalazine (CAS No. 188913-57-7/188913-58-8) (Uriach), etanercept (CAS No. 185243-69-0) (Amgen), ethyl pyruvate (Critical (Critical Therapeutics), golimumab (CAS No. 476181-74-5) (Johnson & Johnson), hormono-immunotherapy from Ipsen, CDP571 (e.g., Humicade from UCB), IC-485 (ICOS), infliximab (CAS No. 170277-31-3) (Johnson & Johnson), IP- 751 (Manhattan Pharmaceuticals), ISIS-104838 (CAS No. 250755-32-9) (ISIS Pharmaceuticals), lenalidomide (CAS No. 191732-72-6) (Celgene), lentinan (CAS No. 37339-90-5) (Ajinomoto), MDL-201112 (CAS No. 142130-73-2) (Sanofi-Aventis), medroxyprogesterone (CAS No. 520-85-4) (InKine Pharmaceutical), N-acetylcysteine (CAS No. 616-91-1) (Zambon), NBE-P2 (DIREVO Biotech), nerelimomab (CAS No. 162774-06-3) (Chiron), OM-294DP (OM PHARMA), onercept (CAS No. 199685-57-9) (Yeda), PASSTNF-alpha (Verigen), pentoxifylline or oxypentifylline (Sanofi-Aventis), Pharmaprojects No. 4091, 4241 , 4295, or 4488 (Sanofi-Aventis), Pharmaprojects No. 5480 (Amgen), Pharmaprojects No. 6749 (Cengent), pirfenidone (CAS No. 53179-13-8) (MARNAC), RPR-132294 (Sanofi- Aventis), S5 (F002) (Fulcrum Pharmaceuticals), simvastatin (CAS No. 79902-63-9) (Merck & Co), STA-6292 (Synta Pharmaceuticals), tacrolimus (CAS No. 104987-11-3) (Fujisawa LifeCycle Pharma), talactoferrin alfa (CAS No. 308240-58-6) (Agennix), thalidomide (CAS No. 50-35-1) (Celgene), TNF antagonists form ProStrakan, and Synergen, TNF inhibitors (Amgen), TNF-alpha antagonists from Dynavax Technologies and Jerina AG (Germany), TNF-alpha inhibitors from IBFB Pharma and Xencor (Xencor), torbafylline (CAS No. 105102-21-4) (Sanofi-Aventis), UR-1505 (Uriach), VT-346 (Viron Therapeutics), YSIL6 (Y's Therapeutics), YSTH2 (Y's Therapeutics), NPI-1302a-3 (Nereus Pharmaceuticals, a TNF antagonist from Jerina AG (Germany), dersalazine, and an analogue or derivative thereof.
17459. The method of item 17360 wherein the anti- fibrotic agent is a Toll receptor inhibitor.
17460. The method of item 17360 wherein the anti- fibrotic agent is a tubulin antagonist.
17461. The method of item 17360 wherein the anti- fibrotic agent is a tyrosine kinase inhibitor selected from the group consisting of SU-011248, SUTENT from Pfizer Inc. (New York, NY), BMS-354825, PN- 355 (Paracelsian Pharmaceuticals), AGN-199659 (Allergan), AAL-993 or ABP-309 (Novartis), adaphostin (NIH), AEE-788 (Novartis), AG-013736 (OSI Pharmaceuticals), AG-13736 (Pfizer), ALT-110 (Alteris Therapeutics), AMG-706 (Amgen), anticancer MAbs from Xencor, anti-EGFrvlll MAbs from Abgenix, anti-HER2 MAb from Abiogen, AZD-2171 or AZD-9935 (AstraZeneca), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), CEP-5214 (Cephalon), CEP-7055 (Cephalon), cetuximab (ImClone Systems), CHIR-200131 and CHIR-258 (Chiron), CP-547632 (OSI Pharmaceuticals), CP-724714 (Pfizer), CT-301 (Creabilis Therapeutics), D- 69491 (Baxter International), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EGFR/ErbB2 inhibitors from Array BioPharma, erlotinib (CAS No. 183319- 69-9) (OSI Pharmaceuticals), EXEL-2880 (Exelixis), FK-778 (Sanofi- Aventis) , gefitinib (CAS No. 184475-35-2) (AstraZeneca), GW-2286 or GW- 654652 (GlaxoSmithKline), her2/neu antigen from AlphaVax, HER-2/neu inhibitor from Generex, Herzyme (Medipad) (Sirna Therapeutics), HKI-272 (Wyeth), HuMax-EGFr (Genmab), idronoxil (CAS No. 81267-65-4) (Novogen), IGF-1 inhibitors from Ontogen, IMC-11F8 (ImCIone Systems), kahalalide F (CAS No. 149204-42-2) (PharmaMar), KDR inhibitor from LG Life Sciences, KDR inhibitors from Abbott Laboratories, KDR kinase inhibitors (UCB), Kdr kinase inhibitors from Merck & Co, KRN-633 and KRN- 951 (Kirin Brewery), KSB-102 (Xenova), lapatinib ditosylate (CAS No. 388082-78-8) (GlaxoSmithKline), matuzumab (Merck KGaA), MDX-214 (Medarex), ME-103 (Pharmexa), MED-A300 (Gerolymatos), MNAC-13 (Lay Line Genomics), nimotuzumab (Center of Molecular Immunology), NSC- 330507 or NSC-707545 (NIH), NV-50 (Novogen), OSI-930 (OSI Pharmaceuticals), panitumumab (Abgenix), pelitinib (CAS No. 287933-82-7) (Wyeth), pertuzumab (CAS No. 380610-27-5) (Genentech), Pharmaprojects No. 3985 (Sanofi-Aventis), prostate cancer therapeutics from Sequenom (SQPC35, SQPC36, SQPC90), removab and remoxab (Trion Pharma), RG- 13022 (CAS No. 136831-48-6), RG-13291 (CAS No. 138989-50-1), or RG- 14620 (CAS No. 136831-49-7) (Sanofi-Aventis), RM-6427 (Romark), RNAi breast cancer therapy from Benitec, RP 53801 (CAS No. 125882-88-4) (Sanofi-Aventis), sorafenib tosylate (Bayer), SU-11657 (Pfizer), Tie-2 antagonists from Semaia (Hybrigenics), Tie-2 inhibitors from Ontogen, trastuzumab (CAS No. 180288-69-1) (Genentech), tyrosine kinase inhibitors from Sanofi-Aventis, U3-1287, U3-1565, U3-1784, U3-1800 (U3 Pharma), vatalanib (Novartis), VEGFR-2 kinase inhibitor from Bristol-Myers Squibb, XL-647 (Exelixis), ZD-6474 (AstraZeneca), ZK-CDK (Schering AG), an EGFR tyrosine kinase inhibitor, EKB-569 (Wyeth), and an analogue or derivative thereof.
17462. The method of item 17360 wherein the anti- fibrotic agent is a VEGF inhibitor. 17463. The method of item 17360 wherein the anti- fibrotic agent is a vitamin D receptor agonist.
17464. The method of item 17360 wherein the anti- fibrotic agent is ZD-6474 (an angiogenesis inhibitor).
17465. The method of item 17360 wherein the anti- fibrotic agent is AP-23573 (an mTOR inhibitor).
17466. The method of item 17360 wherein the anti- fibrotic agent is synthadotin (a tubulin antagonist).
17467. The method of item 17360 wherein the anti- fibrotic agent is S-0885 (a collagenase inhibitor).
17468. The method of item 17360 wherein the anti- fibrotic agent is aplidine (an elongation factor-1 alpha inhibitor).
17469. The method of item 17360 wherein the anti- fibrotic agent is ixabepilone (an epithilone).
17470. The method of item 17360 wherein the anti- fibrotic agent is IDN-5390 (an angiogenesis inhibitor).
17471. The method of item 17360 wherein the anti- fibrotic agent is SB-2723005 (an angiogenesis inhibitor).
17472. The method of item 17360 wherein the anti- fibrotic agent is ABT-518 (an angiogenesis inhibitor). 17473. The method of item 17360 wherein the anti- fibrotic agent is combretastatin (an angiogenesis inhibitor).
17474. The method of item 17360 wherein the anti- fibrotic agent is anecortave acetate (an angiogenesis inhibitor).
17475. The method of item 17360 wherein the anti- fibrotic agent is SB-715992 (a kinesin antagonist).
17476. The method of item 17360 wherein the anti- fibrotic agent is temsirolimus (an mTOR inhibitor).
17477. The method of item 17360 wherein the anti- fibrotic agent is adalimumab (a TNFα antagonist).
17478. The method of item 17360 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with an anti-fibrotic agent, and (b) implanting the medical device into the host.
17479. The method of item 17360 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with an anti-infective agent, and (b) implanting the medical device into the host.
17480. The method of item 17360 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with a polymer; and (b) implanting the medical device into the host. 17481. The method of item 17360 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with a composition comprising an anti-fibrotic agent and a polymer, and (b) implanting the medical device into the host.
17482. The method of item 17360 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with a composition comprising an anti-infective agent and a polymer, and (b) implanting the medical device into the host.
17483. The method of item 17360 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with a composition comprising an anti-fibrotic agent, an anti- infective agent and a polymer, and (b) implanting the medical device into the host.
17484. The method of item 17360 wherein the anti- infective agent is an anthracycline.
17485. The method of item 17360 wherein the anti- infective agent is doxorubicin.
17486. The method of item 17360 wherein the anti- infective agent is is mitoxantrone.
17487. The method of item 17360 wherein the anti- infective agent is a fluoropyrimidine.
17488. The method of item 17360 wherein the anti- infective agent is 5-fluorouracil (5-FU). 17489. The method of item 17360 wherein the anti- infective agent is a folic acid antagonist.
17490. The method of item 17360 wherein the anti- infective agent is methotrexate.
17491. The method of item 17360 wherein the anti- infective agent is a podophylotoxin.
17492. The method of item 17360 wherein the anti- infective agent is etoposide.
17493. The method of item 17360 wherein the anti- infective agent is camptothecin.
17494. The method of item 17360 wherein the anti- infective agent is a hydroxyurea.
17495. The method of item 17360 wherein the anti- infective agent is a platinum complex.
17496. The method of item 17360 wherein the anti- infective agent is cisplatin.
17497. The method of item 17360 wherein the composition comprises an anti-thrombotic agent.
17498. The method of item 17360 wherein the polymer is formed from reactants comprising a naturally occurring polymer. 17499. The method of item 17360 wherein the polymer reactants comprising protein.
17500. The method of item 17360 wherein the polymer reactants comprising carbohydrate.
17501. The method of item 17360 wherein the polymer reactants comprising biodegradable polymer.
17502. The method of item 17360 wherein the polymer reactants comprising nonbiodegradable polymer.
17503. The method of item 17360 wherein the polymer reactants comprising collagen.
17504. The method of item 17360 wherein the polymer reactants comprising methylated collagen.
17505. The method of item 17360 wherein the polymer reactants comprising fibrinogen.
17506. The method of item 17360 wherein the polymer reactants comprising thrombin.
17507. The method of item 17360 wherein the polymer reactants comprising blood plasma.
17508. The method of item 17360 wherein the polymer reactants comprising calcium salt. 17509. The method of item 17360 wherein the polymer reactants comprising an antifibronolytic agent.
17510. The method of item 17360 wherein the polymer reactants comprising fibrinogen analog.
17511. The method of item 17360 wherein the polymer reactants comprising albumin.
17512. The method of item 17360 wherein the polymer reactants comprising plasminogen.
17513. The method of item 17360 wherein the polymer reactants comprising von Willebrands factor.
17514. The method of item 17360 wherein the polymer reactants comprising Factor VIII.
17515. The method of item 17360 wherein the polymer reactants comprising hypoallergenic collagen.
17516. The method of item 17360 wherein the polymer reactants comprising atelopeptidic collagen.
17517. The method of item 17360 wherein the polymer reactants comprising telopeptide collagen.
17518. The method of item 17360 wherein the polymer reactants comprising crosslinked collagen. 17519. The method of item 17360 wherein the polymer is formed from reactants comprising aprotinin.
17520. The method of item 17360 wherein the polymer is formed from reactants comprising epsilon-amino-n-caproic acid.
17521. The method of item 17360 wherein the polymer is formed from reactants comprising gelatin.
17522. The method of item 17360 wherein the polymer is formed from reactants comprising protein conjugates.
17523. The method of item 17360 wherein the polymer is formed from reactants comprising gelatin conjugates.
17524. The method of item 17360 wherein the polymer is formed from reactants comprising a synthetic polymer.
17525. The method of item 17360 wherein the polymer is formed from reactants comprising a synthetic isocyanate-containing compound.
17526. The method of item 17360 wherein the polymer is formed from reactants comprising a synthetic thiol-containing compound.
17527. The method of item 17360 wherein the polymer is formed from reactants comprising a synthetic compound containing at least two thiol groups. 17528. The method of item 17360 wherein the polymer is formed from reactants comprising a synthetic compound containing at least three thiol groups.
17529. The method of item 17360 wherein the polymer is formed from reactants comprising a synthetic compound containing at least four thiol groups.
17530. The method of item 17360 wherein the polymer is formed from reactants comprising a synthetic amino-containing compound.
17531. The method of item 17360 wherein the polymer is formed from reactants comprising a synthetic compound containing at least two amino groups.
17532. The method of item 17360 wherein the polymer is formed from reactants comprising a synthetic compound containing at least three amino groups.
17533. The method of item 17360 wherein the polymer is formed from reactants comprising a synthetic compound containing at least four amino groups.
17534. The method of item 17360 wherein the polymer is formed from reactants comprising a synthetic compound comprising a carbonyl-oxygen-succinimidyl group.
17535. The method of item 17360 wherein the polymer is formed from reactants comprising a synthetic compound comprising at least two carbonyl-oxygen-succinimidyl groups. 17536. The method of item 17360 wherein the polymer is formed from reactants comprising a synthetic compound comprising at least three carbonyl-oxygen-succinimidyl groups.
17537. The method of item 17360 wherein the polymer is formed from reactants comprising a synthetic compound comprising at least four carbonyl-oxygen-succinimidyl groups.
17538. The method of item 17360 wherein the polymer is formed from reactants comprising a synthetic polyalkylene oxide- containing compound.
17539. The method of item 17360 wherein the polymer is formed from reactants comprising a synthetic compound comprising both polyalkylene oxide and biodegradable polyester blocks.
17540. The method of item 17360 wherein the polymer is formed from reactants comprising a synthetic polyalkylene oxide- containing compound having reactive amino groups.
17541. The method of item 17360 wherein the polymer is formed from reactants comprising a synthetic polyalkylene oxide- containing compound having reactive thiol groups.
17542. The method of item 17360 wherein the polymer is formed from reactants comprising a synthetic polyalkylene oxide- containing compound having reactive carbonyl-oxygen-succinimidyl groups.
17543. The method of item 17360 wherein the polymer is formed from reactants comprising a synthetic compound comprising a biodegradable polyester block. 17544. The method of item 17360 wherein the polymer is formed from reactants comprising a synthetic polymer formed in whole or part from lactic acid or lactide.
17545. The method of item 17360 wherein the polymer is formed from reactants comprising a synthetic polymer formed in whole or part from glycolic acid or glycolide.
17546. The method of item 17360 wherein the polymer is formed from reactants comprising polylysine.
17547. The method of item 17360 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound comprising a polyalkylene oxide portion.
17548. The method of item 17360 wherein the polymer is formed from reactants comprising (a) protein and (b) polylysine.
17549. The method of item 17360 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound having at least four thiol groups.
17550. The method of item 17360 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound having at least four amino groups.
17551. The method of item 17360 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound having at least four carbonyl-oxygen-succinimide groups. 17552. The method of item 17360 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound having a biodegradable region formed from reactants selected from lactic acid, lactide, glycolic acid, glycolide, and epison-caprolactone.
17553. The method of item 17360 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound comprising a polyalkylene oxide portion.
17554. The method of item 17360 wherein the polymer is formed from reactants comprising (a) collagen and (b) polylysine.
17555. The method of item 17360 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound having at least four thiol groups.
17556. The method of item 17360 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound having at least four amino groups.
17557. The method of item 17360 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound having at least four carbonyl-oxygen-succinimide groups.
17558. The method of item 17360 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound having a biodegradable region formed from reactants selected from lactic acid, lactide, glycolic acid, glycolide, and epison-caprolactone. 17559. The method of item 17360 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound comprising a polyalkylene oxide portion.
17560. The method of item 17360 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) polylysine.
17561. The method of item 17360 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound having at least four thiol groups.
17562. The method of item 17360 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound having at least four amino groups.
17563. The method of item 17360 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound having at least four carbonyl-oxygen-succinimide groups.
17564. The method of item 17360 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound having a biodegradable region formed from reactants selected from lactic acid, lactide, glycolic acid, glycolide, and epison-caprolactone.
17565. The method of item 17360 wherein the polymer is formed from reactants comprising hyaluronic acid.
17566. The method of item 17360 wherein the polymer is formed from reactants comprising a hyaluronic acid derivative. 17567. The method of item 17360 wherein the polymer is formed from reactants comprising pentaerythritol poly(ethylene glycol)ether tetra-sulfhydryl of number average molecular weight between 3,000 and 30,000.
17568. The method of item 17360 wherein the polymer is formed from reactants comprising pentaerythritol poly(ethylene glycol)ether tetra-amino of number average molecular weight between 3,000 and 30,000.
17569. The method of item 17360 wherein the polymer is formed from reactants comprising (a) a synthetic compound having a number average molecular weight between 3,000 and 30,000 and comprising a polyalkylene oxide region and multiple nucleophilic groups, and (b) a synthetic compound having a number average molecular weight between 3,000 and 30,000 and comprising a polyalkylene oxide region and multiple electrophilic groups.
17570. The method of item 17360 wherein the composition comprises a colorant.
17571. The method of item 17360 wherein the composition is sterile.
17572. A method as in any one of items 17360-17571 , wherein the device is an intravascular device.
17573. A method as in any one of items 17360-17571 , wherein the device is a gastrointestinal stent. 17574. A method as in any one of items 17360-17571 , wherein the device is a tracheal and bronchial stent.
17575. A method as in any one of items 17360-17571, wherein the device is a genital urinary stent.
17576. A method as in any one of items 17360-17571 , wherein the device is an ear and nose stent.
17577. A method as in any one of items 17360-17571 , wherein the device is an ear ventilation.
17578. A method as in any one of items 17360-17571 , wherein the device is an intraocular implant.
17579. A method as in any one of items 17360-17571 , wherein the device is a vascular graft.
17580. A method as in any one of items 17360-17571 , wherein the device comprises a film or a mesh.
17581. A method as in any one of items 17360-17571 , wherein the device is a glaucoma drainage device.
17582. A method as in any one of items 17360-17571 , wherein the device is a prosthetic heart valve or a component thereof.
17583. A method as in any one of items 17360-17571 , wherein the device is a penile implant. 17584. A method as in any one of items 17360-17571 , wherein the device is an endotracheal or tracheostomy tube.
17585. A method as in any one of items 17360-17571, wherein the device is a peritoneal dialysis catheter.
17586. A method as in any one of items 17360-17571 , wherein the device is a central nervous system shunt or a pressure monitoring device.
17587. A method as in any one of items 17360-17571 , wherein the device is an inferior vena cava filter.
17588. A method as in any one of items 17360-17571 , wherein the device is a gastrointestinal device.
17589. A method as in any one of items 17360-17571 , wherein the device is a central venous catheter.
17590. A method as in any one of items 17360-17571 , wherein the device is a ventricular assist device.
17591. A method as in any one of items 17360-17571 , wherein the device is a spinal implant.
17592. A method as in any one of items 17360-17571 , wherein the device is an implantable electrical device.
17593. A method as in any one of items 17360-17571 , wherein the device is an implantable sensor. 17594. A method as in any one of items 17360-17571 , wherein the device is an implantable pump.
17595. A method as in any one of items 17360-17571, wherein the device is a soft tissue implant.
17596. A method for implanting a medical device comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with i) an anti-fibrotic agent, ii) an anti-infective agent, iii) a polymer, iv) a composition comprising an anti-fibrotic agent and a polymer, v) a composistion comprising an anti-infective agent and a polymer, or vi) a composition comprising an anti-fibrotic agent, an anti- infective agent and a polymer, and (b) implanting the medical device into the host, wherein the medical device is an intravascular device.
17597. The method of item 17596 wherein the anti- fibrotic agent is an adensosine A2A receptor antagonist.
17598. The method of item 17596 wherein the anti- fibrotic agent is an AKT inhibitor.
17599. The method of item 17596 wherein the anti- fibrotic agent is an alpha 2 integrin antagonist, wherein the alpha 2 integrin antagonist is Pharmaprojects No. 5754 (Merck KgaA).
17600. The method of item 17596 wherein the anti- fibrotic agent is an alpha 4 integrin antagonist.
17601. The method of item 17596 wherein the anti- fibrotic agent is an alpha 7 nicotinic receptor agonist. 17602. The method of item 17596 wherein the anti- fibrotic agent is an angiogenesis inhibitor selected from the group consisting of AG-12,958 (Pfizer), ATN-161 (Attenuon LLC), neovastat, an angiogenesis inhibitor from Jerina AG (Germany), NM-3 (Mercian), VGA-1155 (Taisho), FCE-26644 (Pfizer), FCE-26950 (Pfizer), FPMA (Meiji Daries), FR-111142 (Fujisawa), GGTI-298, GM-1306 (Ligand), GPA-1734 (Novartis), NNC-47- 0011 (Novo Nordisk), herbamycin (Nippon Kayaku), lenalidomide (Celegene), IP-10 (NIH), ABT-828 (Abbott), KIN-841 (Tokushima University, Japan), SF-1126 (Semafore Pharmaceuticals), laminin technology (NIH), CHIR-258 (Chiron), NVP-AEW541 (Novartis), NVP-AEW541 (Novartis), Vt16907 (Alchemia), OXI-8007 (Oxigene), EG-3306 (Ark Therapeutics), Maspin (Arriva), ABT-567 (Abbott), PPI-2458 (Praecis Pharmaceuticals), CC-5079, CC-4089 (Celgene), HIF-1 alpha inhibitors (Xenova), S-247 (Pfizer), AP-23573 (Ariad), AZD-9935 (Astra Zeneca), mebendazole (Introgen Therapeutics), MetAP-2 inhibitors (GlaxoSmithKline), AG-615 (Angiogene Pharmaceuticals), Tie-2 antagonists (Hybrigenics), NC-381 , CYC-381 , NC-169, NC-219, NC-383, NC-384, NC-407 (Lorus Therapeutics), ATN-224 (Attenuon), ON-01370 (Onconova), Vitronectin antagonists (Amgen), SDX-103 (Salmedix), Vitronectin antagonists (Shire), CHP (Riemser), TEK (Amgen), Anecortave acetate (Alcon), T46.2 (Matrix Therapeutics), HG-2 (Heptagen), TEM antagonists (Genzyme), Oxi-4500 (Oxigene), ATN-161 (Attenuon), WX-293 (Wilex), M-2025 (Metris Therapeutics), Alphastatin (BioActa), YH-16 (Yantai Rongchang), BIBF- 1120 (Boehringer Ingelheim), BAY-57-9352 (Bayer), AS-1404 (Cancer Research Technology), SC-77964 (Pfizer), glycomimetics (BioTie Therapies), TIE-2 Inhibitors (Ontogen), DIMI, Octamer (Octamer), ABR- 215050 (Active Biotech), ABT-518 (Abbott), KDR inhibitors (Abbott), BSF- 466895 (Abbott), SCH-221153 (Schering-Plough), DAC:antiangiogenic (ConjuChem), TFPI (EntreMed), AZD-2171 (Astra-Zenaca), CDC-394 (Celgene), LY290293 (EIi Lilly), IDN-5390 (Indena), Kdr Kinase Inhibitors (Merck), CT-113020, CT-116433, CT-116563, CT-31890, CT-32228) (Cell Therapeutics), A-299620 (Abbott), TWEAK Inhibitor (Amgen), VEGF modulators (Johnson and Johnson), Tum-N53, tumstatin (Genzyme), Thios- 1 , Thios-2 (Thios Pharmaceuticals), MV-6401 (Miravant Medical Technologies), Spisulosine (PharmaMar), CEP-7055 (Cephalon), AUV-201 (Auvation), LM-609 (EIi Lilly), SKF-106615 (AnorMED), Oglufanide disodium (Cytran), BW-114 (Pharminox), Calreticulin (NIH), WX-678 (Wilex), SD-7784 (Pfizer), WX-UK1 (Wilex), SH-268 (Schering AG), 2-Me-PGA (Celgene), S- 137 (Pfizer), ZD-6126 (Angiogene Pharmaceuticals), SG-292 (SignalGen), Benefin (Lane Labs), A6, A36 (Angstrom), SB-2723005 (GlaxoSmithKline), SC-7 (Cell Therapeutics), ZEN-014 (AEterna Zentaris), 2-methoxyestradiol (EntreMed), NK-130119 (Nippon Kayaku), CC-10004 (Celgene), AVE- 8062A (Ajinomoto), Tacedinaline (Pfizer), Actinonin (Tokyo Metropolitan Institute of Medical Science), Lenalidomide (Celgene), VGA-1155, BTO-956 (SRI International), ER-68203-00 (Eisai), CT-6685 (UCB), JKC-362 (Phoenix Pharmaceuticals), DMI-3798 (DMI Biosciences, Angiomate (Ipsen), ZD-6474 (AstraZeneca), CEP-5214 (Cephalon), Canstatin (Genzyme), NM-3 (Mercian), Oridigm (MediQuest Therapeutics), Exherin (Adherex), BLS-0597 (Boston Life Sciences), PTC-299 (PTC Therapeutics), NPI-2358 (Nereus Pharmaceuticals), CGP-79787 (Novartis), AEE-788 (Novartis), CKD-732 (Chong Kun Dang), CP-564959 (OSI Pharmaceuticals), CM-101 (CarboMed), CT-2584, CT3501 (Cell Therapeutics), combretastatin and analogues and derivatives thereof (Oxigene), Rebimastat (Bristol-Meyers Squibb), Dextrin 2-sulfate (ML Laboratories), Cilengitide (Merk KGaA), NSC-706704 (Pharminox), KRN- 951 (Kirin Brewery), Ukrain, NSC-631570 (Nowicky Pharma), Tecogalan sodium (Daiichi Pharmaceutical), Tz-93 (Tsumura), TBC-1635 (Encysive Pharmaceuticals), TAN-1120 (Takeda), Semaxanib (Pfizer), BDI-7800 (Biopharmacopae), SD-186, SD-983 (Bristol-Meyers Squibb), SB-223245 (GlaxoSmithKline), SC-236 (Pfizer), RWJ-590973 (Johnson and Johnson), ILX-1850 (Genzyme), SC-68488, S-836 (Pfizer), CG-55069-11 (CuraGen), Ki-23057 (Kirin Brewery), CCX-700 (Chemoentryx), Pegaptanib octasodium (Giled Sciences), ANGIOCOL (available from Biostratum Inc.), or an analogue or derivative thereof.
17603. The method of item 17596 wherein the anti- fibrotic agent is an apoptosis antagonist.
17604. The method of item 17596 wherein the anti- fibrotic agent is an apoptosis activator.
17605. The method of item 17596 wherein the anti- fibrotic agent is a beta 1 integrin antagonist.
17606. The method of item 17596 wherein the anti- fibrotic agent is a beta tubulin inhibitor.
17607. The method of item 17596 wherein the anti- fibrotic agent is a blocker of enzyme production in Hepatitis C.
17608. The method of item 17596 wherein the anti- fibrotic agent is a Bruton's tyrosine kinase inhibitor.
17609. The method of item 17596 wherein the anti- fibrotic agent is a calcineurin inhibitor.
17610. The method of item 17596 wherein the anti- fibrotic agent is a caspase 3 inhibitor.
17611. The method of item 17596 wherein the anti- fibrotic agent is a CC chemokine receptor antagonist. 17612. The method of item 17596 wherein the anti- fibrotic agent is a cell cycle inhibitor selected from the group consisting of SNS-595 (Sunesis), synthadotin, KRX-0403, and an analogue or derivative thereof.
17613. The method of item 17596 wherein the anti- fibrotic agent is a cathepsin B inhibitor.
17614. The method of item 17596 wherein the anti- fibrotic agent is a cathepsin K inhibitor, wherein the cathepsin K inhibitor is 462795 (GlaxoSmithKline), INPL-022-D6 (Amura Therapeutics), or an analogue or derivative thereof.
17615. The method of item 17596 wherein the anti- fibrotic agent is a cathepsin L inhibitor.
17616. The method of item 17596 wherein the anti- fibrotic agent is a CD40 antagonist.
17617. The method of item 17596 wherein the anti- fibrotic agent is a chemokine receptor agonist.
17618. The method of item 17596 wherein the anti- fibrotic agent is a chymase inhibitor.
17619. The method of item 17596 wherein the anti- fibrotic agent is a collagenase antagonist.
17620. The method of item 17596 wherein the anti- fibrotic agent is a CXCR antagonist. 17621. The method of item 17596 wherein the anti- fibrotic agent is a cyclin dependent kinase inhibitor selected from the group consisting of a CDK-1 inhibitor, a CDK-2 inhibitor, a CDK- 4 inhibitor, a CDK-6 inhibitor, a CAK1 inhibitor from GPC Biotech and Bristol-Myers Squibb, RGB-286199 (GPC Biotech), an anticancer agent from Astex Technology, a CAK1 inhibitor from GPC Biotech, a CDK inhibitor from Sanofi-Aventis, a CDK1/CDK2 inhibitor from Amgen, a CDK2 inhibitor from SUGEN-2 (Pfizer), a hearing loss therapy agent (Sound Pharmaceuticals), PD-0332991 (Pfizer), RGB-286199 (GPC Biotech), Ro-0505124 (Hoffmann- La Roche), a Ser/Thr kinase inhibitor from Lilly (EIi Lilly), CVT-2584 (CAS No. 199986-75-9) (CV Therapeutics), and an analogue or derivative thereof.
17622. The method of item 17596 wherein the anti- fibrotic agent is a cyclooxygenase 1 inhibitor.
17623. The method of item 17596 wherein the anti- fibrotic agent is a DHFR inhibitor.
17624. The method of item 17596 wherein the anti- fibrotic agent is a dual integrin inhibitor.
17625. The method of item 17596 wherein the anti- fibrotic agent is an elastase inhibitor.
17626. The method of item 17596 wherein the anti- fibrotic agent is an elongation factor-1 alpha inhibitor.
17627. The method of item 17596 wherein the anti- fibrotic agent is an endothelial growth factor antagonist. 17628. The method of item 17596 wherein the anti- fibrotic agent is an endothelial growth factor receptor kinase inhibitor selected from the group consisting of sorafenib tosylate (Bayer), AAL-993 (Novartis), ABP-309 (Novartis), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EXEL- 2880 (Exelixis), GW-654652 (GlaxoSmithKline), a KDR inhibitor from LG Life Sciences, CT-6685 and CT-6729 (UCB), KRN-633 and KRN-951 (Kirin Brewery), OSI-930 (OSI Pharmaceuticals), SP-5.2 (Supratek Pharma), SU-
11657 (Pfizer), a Tie-2 antagonist (Hybrigenics), a VEGFR-2 kinase inhibitor (Bristol-Myers Squibb), XL-647 (Exelixis), a KDR inhibitor from Abbott Laboratories, sorafenib tosylate, and an analogue or derivative thereof.
17629. The method of item 17596 wherein the anti- fibrotic agent is an endotoxin antagonist.
17630. The method of item 17596 wherein the anti- fibrotic agent is an epothilone and tubulin binder.
17631. The method of item 17596 wherein the anti- fibrotic agent is an estrogen receptor antagonist.
17632. The method of item 17596 wherein the anti- fibrotic agent is an FGF inhibitor.
17633. The method of item 17596 wherein the anti- fibrotic agent is a farnexyl transferase inhibitor.
17634. The method of item 17596 wherein the anti- fibrotic agent is farnesyltransferase inhibitor selected from the group of A- 197574 (Abbott), a farnesyltransferase inhibitor from Servier, FPTIlI (Strathclyde Institute for Drug R), LB-42908 (LG Life Sciences), Pharmaprojects No. 5063 (Genzyme), Pharmaprojects No. 5597 (Ipsen), Yissum Project No. B-1055 (Yissum), and an analogue or derivative thereof.
17635. The method of item 17596 wherein the anti- fibrotic agent is an FLT-3 kinase inhibitor.
17636. The method of item 17596 wherein the anti- fibrotic agent is an FGF receptor kinase inhibitor.
17637. The method of item 17596 wherein the anti- fibrotic agent is a fibrinogen antagonist selected from the group consisting of AUV-201 (Auvation), MG-13926 (Sanofi-Aventis), plasminogen activator (CAS No. 105913-11-9) (from Sanofi-Aventis or UCB), plasminogen activator-2 (tPA-2) (Sanofi-Aventis), pro-urokinase (CAS No. 82657-92-9) (Sanofi-Aventis), and an analogue or derivative thereof.
17638. The method of item 17596 wherein the anti- fibrotic agent is a heat shock protein 90 antagonist selected from the group consisting of SRN-005 (Sirenade), geldanamycin, NSC-33050 (17- allylaminogeldanamycin; 17-AAG), 17-dimethylaminoethylamino-17- demethoxy-geldanamycin (17-DMAG), rifabutin (rifamycin XIV, 1',4- didehydro-1-deoxy-1,4-dihydro-5'-(2-methylpropyl)-1-oxo-), and an analogue or derivative thereof.
17639. The method of item 17596 wherein the anti- fibrotic agent is a histone deacetylase inhibitor.
17640. The method of item 17596 wherein the anti- fibrotic agent is an HMGCoA reductase inhibitor selected from the group consisting of an atherosclerosis therapeutic from Lipid Sciences, ATM 6000 (ARYx Therapeutics), KS-01-019 (Kos Pharmaceuticals), Pharmaprojects No. 2197 (Sanofi-Aventi), RP 61969 (Sanofi-Aventis), and an analogue or derivative thereof.
17641. The method of item 17596 wherein the anti- fibrotic agent is an ICAM inhibitor.
17642. The method of item 17596 wherein the anti- fibrotic agent is an IL, ICE and IRAK antagonist, wherein the antagonist is a CJ-14877, CP-424174 (Pfizer), NF-61 (Negma-Lerads), and an analogue or derivative thereof.
17643. The method of item 17596 wherein the anti- fibrotic agent is an IL-2 inhibitor.
17644. The method of item 17596 wherein the anti- fibrotic agent is an immunosuppressant selected from the group consisting of teriflunomide (Sanofi Aventis), chlorsulfaquinoxalone (NSC-339004), chlorsulfaquinoxalone sulfate, CS-712 (Sankyo), ismomultin alfa (CAS No. 457913-93-8) (Akzo Nobel), antiallergics from GenPat77, antiinflammatories or AT-005 (Androclus Therapeutics), autoimmune disease therapeutics from EpiVax, BN-007 (Bone), budesonide (CAS No. 51333-22- 3) (MAP Pharmaceuticals), CO-14 (Genzyme), edratide (CAS No. 433922- 67-9) (Teva), EP-314 (Enanta), eprovafen (CAS No. 101335-99-3) (Sanofi- Aventis), HWA-131 (CAS No. 118788-41-3) (Sanofi-Aventis), immunomodulators from MerLion Pharmaceuticals, immunosuppressives from Alchemia, IPL-12 (Inflazyme), MDL-9563 (CAS No. 27086-86-8) (Sanofi-Aventis), Pharmaprojects No. 2330 (Sanofi-Aventis), Pharmaprojects No. 6426 (Abgenix), PXS-25 (Pharmaxis), rosmarinic acid (CAS No. 20283-92-5) (Sanofi-Aventis), RP 42927 or RP 54745 (CAS No. 135330-08-4) (Sanofi-Aventis), SGN-35 (Seattle Genetics), ST-1959 (Sigma-Tau), type I diabetes therapy from SYNX Pharma, UNIL-88 (Debiopharm), VP-025 (Vasogen), VR-694 (Vectura), PRTX-001 (Protalex), and an analogue or derivative thereof.
17645. The method of item 17596 wherein the anti- fibrotic agent is an IMPDH (inosine monophosphate).
17646. The method of item 17596 wherein the anti- fibrotic agent is an integrin antagonist.
17647. The method of item 17596 wherein the anti- fibrotic agent is an interleukin antagonist.
17648. The method of item 17596 wherein the anti- fibrotic agent is an inhibitor of type III receptor tyrosine kinase.
17649. The method of item 17596 wherein the anti- fibrotic agent is an irreversible inhibitor of enzyme methionine aminopeptidase type 2.
17650. The method of item 17596 wherein the anti- fibrotic agent is an isozyme selective delta protein kinase C inhibitor.
17651. The method of item 17596 wherein the anti- fibrotic agent a JAK3 enzyme inhibitor.
17652. The method of item 17596 wherein the anti- fibrotic agent is a JNK inhibitor.
17653. The method of item 17596 wherein the anti- fibrotic agent is a kinase inhibitor. 17654. The method of item 17596 wherein the anti- fibrotic agent is kinesin antagonist.
17655. The method of item 17596 wherein the anti- fibrotic agent is a kinesin antagonist.
17656. The method of item 17596 wherein the anti- fibrotic agent is a leukotriene inhibitor and antagonist selected from the group consisting of ambicromil (CAS No. 58805-38-2) (Sanofi-Aventis), amelubant (CAS No. 346735-24-8) (Boehringer Ingelheim), DW-1141 (Dong Wha), ebselen (Daiichi Pharmaceutical), ibudilast (Kyorin), leucotriene inhibitors from Sanofi-Aventis, lymphotoxin-beta receptor (LT-D) from Biogen Idee, Pharmaprojects No. 1535 and 2728 (CAS No. 119340-33-9) (Sanofi-Aventis), R-112 (Rigel), Rev-5367 (CAS No. 92532-05-3) (Sanofi- Aventis), RG-14893 (CAS No. 141835-49-6) (Sanofi-Aventis), RG-5901-A (CAS No. 101910-24-1), 92532-23-5, RP 66153 (CAS No. 142422-79-5), RP 66364 (CAS No.186916-92-5), RP 69698 (CAS No. 141748-00-7) (Sanofi-Aventis), SC-411930 (Pfizer), SC-41930 (CAS No. 120072-59-5) (Pfizer), SC-50605 (CAS No. 138828-39-4) (Pfizer), SC-51146 (CAS No. 141059-52-1), SC-53228 (CAS No. 153633-01-3) (Pfizer), spaglumic acid (ZY-15106) (CAS No.3106-85-2), 80619-64-3 (Novartis), tipredane (CAS No. 85197-77-9) (Bristol-Myers Squibb), U-75302 (CAS No. 119477-85-9) (Pfizer), and analogue or derivative thereof.
17657. The method of item 17596 wherein the anti- fibrotic agent is an MAP kinase inhibitor.
17658. The method of item 17596 wherein the anti- fibrotic agent is a matrix metalloproteinase inhibitor. 17659. The method of item 17596 wherein the anti- fibrotic agent is an MCP-CCR2 inhibitor.
17660. The method of item 17596 wherein the anti- fibrotic agent is an mTOR inhibitor.
17661. The method of item 17596 wherein the anti- fibrotic agent is an mTOR kinase inhibitor.
17662. The method of item 17596 wherein the anti- fibrotic agent is a microtubule inhibitor selected from the group consisting of antibody-maytansinoid conjugates from Biogen Idee, colchicines (MantiCore Pharmaceuticals), anticancer immunoconjugates from Johnson & Johnson, DIME from Octamer, gni-1f (GNI), huC242-DM4, huMy9-6-DM1 (ImmunoGen), lDN-5404 (Indena), IMO-098, IMOderm (Imotep), mebendazole (Introgen Therapeutics), microtubule poisons from Cambridge Enterprise, paclitaxel such as LOTAX from Aphios (CAS No. 33069-62-4), Genexol-PM from Samyang, Pharmaprojects No. 6383 (Tapestry Pharmaceuticals), RPR-112378 (Sanofi-Aventis), SGN-75 (Seattle Genetics), SPL-7435 (Starpharma), SSR-250411 (Sanofi-Aventis), trastuzumab-DM1 (Genentech), vinorelbine, and an analogue or derivative thereof.
17663. The method of item 17596 wherein the anti- fibrotic agent is an MIF inhibitor.
17664. The method of item 17596 wherein the anti- fibrotic agent is an MMP inhibitor.
17665. The method of item 17596 wherein the anti- fibrotic agent is a neurokinin (NK) antagonist selected from the group consisting of anthrotainin (CAS No. 148084-40-6) (Sanofi-Aventis), an IBS thereapeutic from ArQuIe, MDL-105212A (CAS No. 167261-60-1) (Ssanofi- Aventis), Pharmaprojects No. 2744, 3258 (CAS No. 139167-47-8) 4006, 4201 , or 5986 (Sanofi-Aventis), RP 67580 (CAS No. 135911-02-3), SR- 144190 (CAS No. 201152-86-5), SSR-240600, SSR-241586 (Sanofi- Aventis), TKA-457 (Novartis), vestipitant mesylate (CAS No. 334476-64-1) (GlaxoSmithKline), Win-64821 (Sanofi-Aventis), PRX-96026 (Predix Pharmaceuticals), and an analogue or derivative thereof.
17666. The method of item 17596 wherein the anti- fibrotic agent is an NF kappa B inhibitor selected from the group consisting of AVE-0545 or AVE-0547 (Sanofi-Aventis), bortezomib (CAS No. 179324- 69-7) (Millennium Pharmaceuticals), dexanabinol (CAS No. 112924-45-5) (Pharmos), dexlipotam (Viatris), Pharmaprojects No. 6283 (INDRA) (OXiGENE), IPL-576092 (CAS No. 137571-30-3) (Inflazyme), NFKB decoy (Corgentech), NFKB decoy oligo (AnGes MG), NFKB's from Ariad, osteoporosis treatments or S5 (F005) from Fulcrum Pharmaceuticals, P61 (Phytopharm), R-flurbiprofen (CAS No. 5104-49-4) (Encore Pharmaceuticals), and an analogue or derivative thereof.
17667. The method of item 17596 wherein the anti- fibrotic agent is a nitric oxide agonist.
17668. The method of item 17596 wherein the anti- fibrotic agent is an ornithine decarboxylase inhibitor.
17669. The method of item 17596 wherein the anti- fibrotic agent is a p38 MAP kinase inhibitor selected from the group consisting of AZD-6703 (AstraZeneca), JX-401 (Jexys Pharmaceuticals), BMS-2 (Bristol-Myers Squibb), a p38 MAP kinase inhibitor from Novartis, a p38-alpha MAP kinase inhibitor from Amphora, Pharmaprojects No. 5704 (Pharmacopeia), RPR-200765A (Sanofi-Aventis), SD-282 (Johnson & Johnson), TAK-715 (Takeda), and an analogue or derivative thereof.
17670. The method of item 17596 wherein the anti- fibrotic agent is a palmitoyl-protein thioesterase inhibitor.
17671. The method of item 17596 wherein the anti- fibrotic agent is a PDGF receptor kinase inhibitor selected from the group consisting of AAL-993, AMN-107, or ABP-309 (Novartis), AMG-706 (Amgen), BAY-57-9352 (Bayer), CDP-860 (UCB), E-7080 (Eisai), imatinib (CAS No. 152459-95-5) (Novartis), OSI-930 (OSI Pharmaceuticals), RPR- 127963E (Sanofi-Aventis), RWJ-540973 (Johnson & Johnson), sorafenib tosylate (Bayer), SU-11657 (Pfizer), tandutinib (CAS No. 387867-13-2) (Millennium Pharmaceuticals), vatalanib (Novartis), ZK-CDK (Schering AG), and an analogue or derivative thereof.
17672. The method of item 17596 wherein the anti- fibrotic agent is (-)-halofenate (Metabolex), AMG-131 (Amgen), antidiabetics from Japan Tobacco, AZD-4619, AZD-8450, AZD-8677 (AstraZeneca), DRF-10945, balaglitazone (Dr Reddy's), CS-00088, CS-00098 (Chipscreen Biosciences), E-3030 (Eisai), etalocib (CAS No. 161172-51-6) (EIi Lilly), GSK-641597 (Ligand), GSK-677954 (GlaxoSmithKline), GW-409544 (Ligand), GW-590735 (GlaxoSmithKline), K-111 (Hoffmann-La Roche), LY- 518674 (EIi Lilly), LY-674 (Ligand), LY-929 (Ligand), MC-3001, MC-3002 (MaxoCore Pharmaceuticals), metformin HCI + pioglitazone (CAS No. 1115- 70-4 and 112529-15-4), ACTOPLUS MET from Andrx), muraglitazar (CAS No. 331741-94-7) (Bristol-Myers Squibb), naveglitazar (Ligand), oleoylethanolamide (Kadmus Pharmaceuticals), ONO-5129, pioglitazone hydrochloride (CAS No. 111025-46-8 and 112529-15-4) (Takeda), PLX-204 (Plexxikon), PPAR agonists from Genfit, PPAR delta agonists from EIi Lilly, PPAR-alpha agonists from CrystalGenomics, PPAR-gamma modulators and PPAR-β modulators from CareX, rosiglitazone maleate (CAS No. 122324- 73-4 or 155141-29-0) (GlaxoSmithKline), rosiglitazone maleate/glimepir (CAS No. 155141-29-0 and 93479-97-1), AVANDARYL, rosiglitazone maleate/metformin extend (CAS No. 155141-29-0 and 657-24-9), AVANDAMET, rosiglitazone maleate+metformin, AVANDAMET (GlaxoSmithKline), tesaglitazar (AstraZeneca), and an analogue or derivative thereof.
17673. The method of item 17596 wherein the anti- fibrotic agent is a phosphatase inhibitor.
17674. The method of item 17596 wherein the anti- fibrotic agent is a phosphodiesterase (PDE) inhibitor selected from the group consisting of avanafil (Tanabe Seiyaku), dasantafil (CAS No. 569351- 91-3) (Schering-Plough), A-906119 (CAS No. 134072-58-5), DL-850 (Sanofi-Aventis), GRC-3015, GRC-3566, GRC-3886 (Glenmark), HWA-153 (CAS No. 56395-66-5) (Sanofi-Aventis), hydroxypumafentrine (Altana), IBFB-130011 , IBFB-14-016, IBFB-140301 , IBFB-150007, IBFB-211913 (IBFB Pharma), L-826141 (Merck & Co), medorinone (CAS No. 88296-61-1) (Sanofi-Aventis), MEM-1917 (Memory Pharmaceuticals), ND-1251 (Neuro3d), PDE inhibitors from ICOS, PDE IV inhibitors from Memory Pharmaceuticals and CrystalGenomics, Pharmaprojects No. 2742 and 6141 (Sanofi-Aventis), QAD-171 (Novartis), RHC-2963 (CAS No. 76993-12-9 and 76993-14-1), RPR-117658, RPR-122818 derivatives, SR-24870 , and RPR- 132294 (Sanofi-Aventis), SK-350 (ln2Gen), stroke therapy agents from deCODE Genetics, TAS-203 (Taiho), tofimilast (CAS No. 185954-27-2) (Pfizer), UK-371800 (Pfizer), WIN-65579 (CAS No. 158020-82-7) (Sanofi- Aventis), IBFB-130020 (IBFB Pharma), OPC-6535 (CAS No. 145739-56-6) (Otsuka), a phosphodiesterase III inhibitor, enoximone, a phosphodiesterase IV inhibitor, fosfosal, Atopik (Barrier Therapeutics), triflusal, a phosphodiesterase V inhibitor, and an analogue or derivative thereof.
17675. The method of item 17596 wherein the anti- fibrotic agent is a PKC inhibitor.
17676. The method of item 17596 wherein the anti- fibrotic agent is a platelet activating factor antagonist.
17677. The method of item 17596 wherein the anti- fibrotic agent is a platelet-derived growth factor receptor kinase inhibitor.
17678. The method of item 17596 wherein the anti- fibrotic agent is a prolyl hydroxylase inhibitor.
17679. The method of item 17596 wherein the anti- fibrotic agent is a polymorphonuclear neutrophil inhibitor.
17680. The method of item 17596 wherein the anti- fibrotic agent is a protein kinase B inhibitor.
17681. The method of item 17596 wherein the anti- fibrotic agent is a protein kinase C stimulant.
17682. The method of item 17596 wherein the anti- fibrotic agent is a purine nucleoside analogue.
17683. The method of item 17596 wherein the anti- fibrotic agent is a purinoreceptor P2X antagonist. 17684. The method of item 17596 wherein the anti- fibrotic agent is a Raf kinase inhibitor.
17685. The method of item 17596 wherein the anti- fibrotic agent is a reversible inhibitor of ErbB1 and ErbB2.
17686. The method of item 17596 wherein the anti- fibrotic agent is a ribonucleoside triphosphate reductase inhibitor.
17687. The method of item 17596 wherein the anti- fibrotic agent is an SDF-1 antagonist.
17688. The method of item 17596 wherein the anti- fibrotic agent is a sheddase inhibitor.
17689. The method of item 17596 wherein the anti- fibrotic agent is an SRC inhibitor.
17690. The method of item 17596 wherein the anti- fibrotic agent is a stromelysin inhibitor.
17691. The method of item 17596 wherein the anti- fibrotic agent is an Syk kinase inhibitor.
17692. The method of item 17596 wherein the anti- fibrotic agent is a telomerase inhibitor.
17693. The method of item 17596 wherein the anti- fibrotic agent is a TGF beta inhibitor selected from the group consisting of pirfenidone (CAS No. 53179-13-8) (MARNAC), tranilast (CAS No. 53902- 12-8) (Kissei), IN-1130 (ln2Gen), mannose-6-phosphate (BTG), TGF-β antagonists from Inflazyme (Pharmaprojects No. 6075), TGF-β antagonists from Sydney, non-industrial source), TGF-β I receptor kinase inhibitors from EIi Lilly, TGF-β receptor inhibitors from Johnson & Johnson, and an analogue or derivative thereof.
17694. The method of item 17596 wherein the anti- fibrotic agent is a TNFα antagonist or TACE inhibitor selected from the group consisting of adalimumab (CAS No. 331731-18-1) (Cambridge Antibody Technology), AGIX-4207 (AtheroGenics), AGT-1 (Advanced Biotherapy), an anti-inflammatory from Borean Pharma, Cellzome, or Paradigm Therapeutics, anti-inflammatory vaccine (TNF-alpha kinoid) from Neovacs, humanized anti-TNF antibody or an anti-TNF MAb (CB0006) Celltech (UCB), apratastat (CAS No. 287445-51-0) (Wyeth), BMS-561392 (Bristol-Myers Squibb), BN-006 (Bone), certolizumab pegol (CAS No. 428863-50-7 or CH-138 (UCB), cilomilast (CAS No. 153259-65-5) (GlaxoSmithKline), CR-1 (Nuada Pharmaceuticals), CRx-119 (CombinatoRx), D-5410 (UCB), dacopafant (CAS No. 125372-33-0) (Sanofi- Aventis), dersalazine (CAS No. 188913-57-7/188913-58-8) (Uriach), etanercept (CAS No. 185243-69-0) (Amgen), ethyl pyruvate (Critical (Critical Therapeutics), golimumab (CAS No. 476181-74-5) (Johnson & Johnson), hormono-immunotherapy from Ipsen, CDP571 (e.g., Humicade from UCB), IC-485 (ICOS), infliximab (CAS No. 170277-31-3) (Johnson & Johnson), IP- 751 (Manhattan Pharmaceuticals), ISIS-104838 (CAS No. 250755-32-9) (ISIS Pharmaceuticals), lenalidomide (CAS No. 191732-72-6) (Celgene), lentinan (CAS No. 37339-90-5) (Ajinomoto), MDL-201112 (CAS No. 142130-73-2) (Sanofi-Aventis), medroxyprogesterone (CAS No. 520-85-4) (InKine Pharmaceutical), N-acetylcysteine (CAS No. 616-91-1) (Zambon), NBE-P2 (DIREVO Biotech), nerelimomab (CAS No. 162774-06-3) (Chiron), OM-294DP (OM PHARMA), onercept (CAS No. 199685-57-9) (Yeda), PASSTNF-alpha (Verigen), pentoxifylline or oxypentifylline (Sanofi-Aventis), Pharmaprojects No. 4091 , 4241 , 4295, or 4488 (Sanofi-Aventis), Pharmaprojects No. 5480 (Amgen), Pharmaprojects No. 6749 (Cengent), pirfenidone (CAS No. 53179-13-8) (MARNAC), RPR-132294 (Sanofi- Aventis), S5 (F002) (Fulcrum Pharmaceuticals), simvastatin (CAS No. 79902-63-9) (Merck & Co), STA-6292 (Synta Pharmaceuticals), tacrolimus (CAS No. 104987-11-3) (Fujisawa LifeCycle Pharma), talactoferrin alfa (CAS No. 308240-58-6) (Agennix), thalidomide (CAS No. 50-35-1) (Celgene), TNF antagonists form ProStrakan, and Synergen, TNF inhibitors (Amgen), TNF-alpha antagonists from Dynavax Technologies and Jerina AG (Germany), TNF-alpha inhibitors from IBFB Pharma and Xencor (Xencor), torbafylline (CAS No. 105102-21-4) (Sanofi-Aventis), UR-1505 (Uriach), VT-346 (Viron Therapeutics), YSIL6 (Y's Therapeutics), YSTH2 (Y's Therapeutics), NPI-1302a-3 (Nereus Pharmaceuticals, a TNF antagonist from Jerina AG (Germany), dersalazine, and an analogue or derivative thereof.
17695. The method of item 17596 wherein the anti- fibrotic agent is a Toll receptor inhibitor.
17696. The method of item 17596 wherein the anti- fibrotic agent is a tubulin antagonist.
17697. The method of item 17596 wherein the anti- fibrotic agent is a tyrosine kinase inhibitor selected from the group consisting of SU-011248, SUTENT from Pfizer Inc. (New York, NY), BMS-354825, PN- 355 (Paracelsian Pharmaceuticals), AGN-199659 (Allergan), AAL-993 or ABP-309 (Novartis), adaphostin (NIH), AEE-788 (Novartis), AG-013736 (OSI Pharmaceuticals), AG-13736 (Pfizer), ALT-110 (Alteris Therapeutics), AMG-706 (Amgen), anticancer MAbs from Xencor, anti-EGFrvlll MAbs from Abgenix, anti-HER2 MAb from Abiogen, AZD-2171 or AZD-9935 (AstraZeneca), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), CEP-5214 (Cephalon), CEP-7055 (Cephalon), cetuximab (ImClone Systems), CHIR-200131 and CHIR-258 (Chiron), CP-547632 (OSI Pharmaceuticals), CP-724714 (Pfizer), CT-301 (Creabilis Therapeutics), D- 69491 (Baxter International), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EGFR/ErbB2 inhibitors from Array BioPharma, erlotinib (CAS No. 183319- 69-9) (OSI Pharmaceuticals), EXEL-2880 (Exelixis), FK-778 (Sanofi- Aventis) , gefitinib (CAS No. 184475-35-2) (AstraZeneca), GW-2286 or GW- 654652 (GlaxoSmithKline), her2/neu antigen from AlphaVax, HER-2/neu inhibitor from Generex, Herzyme (Medipad) (Sirna Therapeutics), HKI-272 (Wyeth), HuMax-EGFr (Genmab), idronoxil (CAS No. 81267-65-4) (Novogen), IGF-1 inhibitors from Ontogen, IMC-11F8 (ImClone Systems), kahalalide F (CAS No. 149204-42-2) (PharmaMar), KDR inhibitor from LG Life Sciences, KDR inhibitors from Abbott Laboratories, KDR kinase inhibitors (UCB), Kdr kinase inhibitors from Merck & Co, KRN-633 and KRN- 951 (Kirin Brewery), KSB-102 (Xenova), lapatinib ditosylate (CAS No. 388082-78-8) (GlaxoSmithKline), matuzumab (Merck KGaA), MDX-214 (Medarex), ME-103 (Pharmexa), MED-A300 (Gerolymatos), MNAC-13 (Lay Line Genomics), nimotuzumab (Center of Molecular Immunology), NSC- 330507 or NSC-707545 (NIH), NV-50 (Novogen), OSI-930 (OSI Pharmaceuticals), panitumumab (Abgenix), pelitinib (CAS No. 287933-82-7) (Wyeth), pertuzumab (CAS No. 380610-27-5) (Genentech), Pharmaprojects No. 3985 (Sanofi-Aventis), prostate cancer therapeutics from Sequenom (SQPC35, SQPC36, SQPC90), removab and remoxab (Trion Pharma), RG- 13022 (CAS No. 136831-48-6), RG-13291 (CAS No. 138989-50-1), or RG- 14620 (CAS No. 136831-49-7) (Sanofi-Aventis), RM-6427 (Romark), RNAi breast cancer therapy from Benitec, RP 53801 (CAS No. 125882-88-4) (Sanofi-Aventis), sorafenib tosylate (Bayer), SU-11657 (Pfizer), Tie-2 antagonists from Semaia (Hybrigenics), Tie-2 inhibitors from Ontogen, trastuzumab (CAS No. 180288-69-1) (Genentech), tyrosine kinase inhibitors from Sanofi-Aventis, U3-1287, U3-1565, U3-1784, U3-1800 (U3 Pharma), vatalanib (Novartis), VEGFR-2 kinase inhibitor from Bristol-Myers Squibb, XL-647 (Exelixis), ZD-6474 (AstraZeneca), ZK-CDK (Schering AG), an EGFR tyrosine kinase inhibitor, EKB-569 (Wyeth), and an analogue or derivative thereof.
17698. The method of item 17596 wherein the anti- fibrotic agent is a VEGF inhibitor.
17699. The method of item 17596 wherein the anti- fibrotic agent is a vitamin D receptor agonist.
17700. The method of item 17596 wherein the anti- fibrotic agent is ZD-6474 (an angiogenesis inhibitor).
17701. The method of item 17596 wherein the anti- fibrotic agent is AP-23573 (an mTOR inhibitor).
17702. The method of item 17596 wherein the anti- fibrotic agent is synthadotin (a tubulin antagonist).
17703. The method of item 17596 wherein the anti- fibrotic agent is S-0885 (a collagenase inhibitor).
17704. The method of item 17596 wherein the anti- fibrotic agent is aplidine (an elongation factor-1 alpha inhibitor).
17705. The method of item 17596 wherein the anti- fibrotic agent is ixabepilone (an epithilone).
17706. The method of item 17596 wherein the anti- fibrotic agent is IDN-5390 (an angiogenesis inhibitor). 17707. The method of item 17596 wherein the anti- fibrotic agent is SB-2723005 (an angiogenesis inhibitor).
17708. The method of item 17596 wherein the anti- fibrotic agent is ABT-518 (an angiogenesis inhibitor).
17709. The method of item 17596 wherein the anti- fibrotic agent is combretastatin (an angiogenesis inhibitor).
17710. The method of item 17596 wherein the anti- fibrotic agent is anecortave acetate (an angiogenesis inhibitor).
17711. The method of item 17596 wherein the anti- fibrotic agent is SB-715992 (a kinesin antagonist).
17712. The method of item 17596 wherein the anti- fibrotic agent is temsirolimus (an mTOR inhibitor).
17713. The method of item 17596 wherein the anti- fibrotic agent is adalimumab (a TNFα antagonist).
17714. The method of item 17596 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with an anti-fibrotic agent, and (b) implanting the medical device into the host.
17715. The method of item 17596 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with an anti-infective agent, and (b) implanting the medical device into the host. 17716. The method of item 17596 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with a polymer; and (b) implanting the medical device into the host.
17717. The method of item 17596 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with a composition comprising an anti-fibrotic agent and a polymer, and (b) implanting the medical device into the host.
17718. The method of item 17596 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with a composition comprising an anti-infective agent and a polymer, and (b) implanting the medical device into the host.
17719. The method of item 17596 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with a composition comprising an anti-fibrotic agent, an anti- infective agent and a polymer, and (b) implanting the medical device into the host.
17720. The method of item 17596 wherein the anti- infective agent is an anthracycline.
17721. The method of item 17596 wherein the anti- infective agent is doxorubicin.
17722. The method of item 17596 wherein the anti- infective agent is is mitoxantrone. 17723. The method of item 17596 wherein the anti- infective agent is a fluoropyrimidine.
17724. The method of item 17596 wherein the anti- infective agent is 5-fluorouracil (5-FU).
17725. The method of item 17596 wherein the anti- infective agent is a folic acid antagonist.
17726. The method of item 17596 wherein the anti- infective agent is methotrexate.
17727 '. The method of item 17596 wherein the anti- infective agent is a podophylotoxtn.
17728. The method of item 17596 wherein the anti- infective agent is etoposide.
17729. The method of item 17596 wherein the anti- infective agent is camptothecin.
17730. The method of item 17596 wherein the anti- infective agent is a hydroxyurea.
17731. The method of item 17596 wherein the anti- infective agent is a platinum complex.
17732. The method of item 17596 wherein the anti- infective agent is cisplatin. 17733. The method of item 17596 wherein the composition comprises an antithrombotic agent.
17734. The method of item 17596 wherein the polymer is formed from reactants comprising a naturally occurring polymer.
17735. The method of item 17596 wherein the polymer is formed from reactants comprising protein.
17736. The method of item 17596 wherein the polymer is formed from reactants comprising carbohydrate.
17737. The method of item 17596 wherein the polymer is formed from reactants comprising biodegradable polymer.
17738. The method of item 17596 wherein the polymer is formed from reactants comprising nonbiodegradable polymer.
17739. The method of item 17596 wherein the polymer is formed from reactants comprising collagen.
17740. The method of item 17596 wherein the polymer is formed from reactants comprising methylated collagen.
17741. The method of item 17596 wherein the polymer is formed from reactants comprising fibrinogen.
17742. The method of item 17596 wherein the polymer is formed from reactants comprising thrombin. 17743. The method of item 17596 wherein the polymer 4 reactants comprising blood plasma.
17744. The method of item 17596 wherein the polymer reactants comprising calcium salt.
17745. The method of item 17596 wherein the polymer reactants comprising an antifibronolytic agent.
17746. The method of item 17596 wherein the polymer reactants comprising fibrinogen analog.
17747. The method of item 17596 wherein the polymer reactants comprising albumin.
17748. The method of item 17596 wherein the polymer reactants comprising plasminogen.
17749. The method of item 17596 wherein the polymer reactants comprising von Willebrands factor.
17750. The method of item 17596 wherein the polymer reactants comprising Factor VIII.
17751. The method of item 17596 wherein the polymer reactants comprising hypoallergenic collagen.
17752. The method of item 17596 wherein the polymer reactants comprising atelopeptidic collagen. 17753. The method of item 17596 wherein the polymer is formed from reactants comprising telopeptide collagen.
17754. The method of item 17596 wherein the polymer is formed from reactants comprising crosslinked collagen.
17755. The method of item 17596 wherein the polymer is formed from reactants comprising aprotinin.
17756. The method of item 17596 wherein the polymer is formed from reactants comprising epsilon-amino-n-caproic acid.
17757. The method of item 17596 wherein the polymer is formed from reactants comprising gelatin.
17758. The method of item 17596 wherein the polymer is formed from reactants comprising protein conjugates.
17759. The method of item 17596 wherein the polymer is formed from reactants comprising gelatin conjugates.
17760. The method of item 17596 wherein the polymer is formed from reactants comprising a synthetic polymer.
17761. The- method of item 17596 wherein the polymer is formed from reactants comprising a synthetic isocyanate-containing compound.
17762. The method of item 17596 wherein the polymer is formed from reactants comprising a synthetic thiol-containing compound. 17763. The method of item 17596 wherein the polymer is formed from reactants comprising a synthetic compound containing at least two thiol groups.
17764. The method of item 17596 wherein the polymer is formed from reactants comprising a synthetic compound containing at least three thiol groups.
17765. The method of item 17596 wherein the polymer is formed from reactants comprising a synthetic compound containing at least four thiol groups.
17766. The method of item 17596 wherein the polymer is formed from reactants comprising a synthetic amino-containing compound.
17767. The method of item 17596 wherein the polymer is formed from reactants comprising a synthetic compound containing at least two amino groups.
17768. The method of item 17596 wherein the polymer is formed from reactants comprising a synthetic compound containing at least three amino groups.
17769. The method of item 17596 wherein the polymer is formed from reactants comprising a synthetic compound containing at least four amino groups.
17770. The method of item 17596 wherein the polymer is formed from reactants comprising a synthetic compound comprising a carbonyl-oxygen-succinimidyl group. 17771. The method of item 17596 wherein the polymer is formed from reactants comprising a synthetic compound comprising at least two carbonyl-oxygen-succinimidyl groups.
17772. The method of item 17596 wherein the polymer is formed from reactants comprising a synthetic compound comprising at least three carbonyl-oxygen-succinimidyl groups.
17773. The method of item 17596 wherein the polymer is formed from reactants comprising a synthetic compound comprising at least four carbonyl-oxygen-succinimidyl groups.
17774. The method of item 17596 wherein the polymer is formed from reactants comprising a synthetic polyalkylene oxide- containing compound.
17775. The method of item 17596 wherein the polymer is formed from reactants comprising a synthetic compound comprising both polyalkylene oxide and biodegradable polyester blocks.
17776. The method of item 17596 wherein the polymer is formed from reactants comprising a synthetic polyalkylene oxide- containing compound having reactive amino groups.
17777. The method of item 17596 wherein the polymer is formed from reactants comprising a synthetic polyalkylene oxide- containing compound having reactive thiol groups.
17778. The method of item 17596 wherein the polymer is formed from reactants comprising a synthetic polyalkylene oxide- containing compound having reactive carbonyl-oxygen-succinimidyl groups. 17779. The method of item 17596 wherein the polymer is formed from reactants comprising a synthetic compound comprising a biodegradable polyester block.
17780. The method of item 17596 wherein the polymer is formed from reactants comprising a synthetic polymer formed in whole or part from lactic acid or lactide.
17781. The method of item 17596 wherein the polymer is formed from reactants comprising a synthetic polymer formed in whole or part from glycolic acid or glycolide.
17782. The method of item 17596 wherein the polymer is formed from reactants comprising polylysine.
17783. The method of item 17596 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound comprising a polyalkylene oxide portion.
17784. The method of item 17596 wherein the polymer is formed from reactants comprising (a) protein and (b) polylysine.
17785. The method of item 17596 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound having at least four thiol groups.
17786. The method of item 17596 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound having at least four amino groups. 17787. The method of item 17596 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound having at least four carbonyl-oxygen-succinimide groups.
17788. The method of item 17596 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound having a biodegradable region formed from reactants selected from lactic acid, lactide, glycolic acid, glycolide, and epison-caprolactone.
17789. The method of item 17596 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound comprising a polyalkylene oxide portion.
17790. The method of item 17596 wherein the polymer is formed from reactants comprising (a) collagen and (b) polylysine.
17791. The method of item 17596 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound having at least four thiol groups.
17792. The method of item 17596 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound having at least four amino groups.
17793. The method of item 17596 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound having at least four carbonyl-oxygen-succinimide groups.
17794. The method of item 17596 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound having a biodegradable region formed from reactants selected from lactic acid, lactide, glycolic acid, glycolide, and epison-caprolactone.
17795. The method of item 17596 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound comprising a polyalkylene oxide portion.
17796. The method of item 17596 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) polylysine.
17797. The method of item 17596 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound having at least four thiol groups.
17798. The method of item 17596 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound having at least four amino groups.
17799. The method of item 17596 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound having at least four carbonyl-oxygen-succinimide groups.
17800. The method of item 17596 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound having a biodegradable region formed from reactants selected from lactic acid, lactide, glycolic acid, glycolide, and epison-caprolactone.
17801. The method of item 17596 wherein the polymer is formed from reactants comprising hyaluronic acid. 17802. The method of item 17596 wherein the polymer is formed from reactants comprising a hyaluronic acid derivative.
17803. The method of item 17596 wherein the polymer is formed from reactants comprising pentaerythritol poly(ethylene glycol)ether tetra-sulfhydryl of number average molecular weight between 3,000 and 30,000.
17804. The method of item 17596 wherein the polymer is formed from reactants comprising pentaerythritol poly(ethylene glycol)ether tetra-amino of number average molecular weight between 3,000 and 30,000.
17805. The method of item 17596 wherein the polymer is formed from reactants comprising (a) a synthetic compound having a number average molecular weight between 3,000 and 30,000 and comprising a polyalkylene oxide region and multiple nucleophilic groups, and (b) a synthetic compound having a number average molecular weight between 3,000 and 30,000 and comprising a polyalkylene oxide region and multiple electrophilic groups.
17806. The method of item 17596 wherein the composition comprises a colorant.
17807. The method of item 17596 wherein the composition is sterile.
17808. A method as in any one of items 17596-17807, wherein the device is a catheter. 17809. A method as in any one of items 17596-17807, wherein the device is a balloon catheter.
17810. A method as in any one of items 17596-17807, wherein the device is a balloon.
17811. A method as in any one of items 17596-17807, wherein the device is a stent graft.
17812. A method as in any one of items 17596-17807, wherein the device is a guidewire.
17813. A method as in any one of items 17596-17807, wherein the device is a stent.
17814. A method as in any one of items 17596-17807, wherein the device is an intravascular stent.
17815. A method as in any one of items 17596-17807, wherein the device is a metallic stent.
17816. A method as in any one of items 17596-17807, wherein the device is a polymeric stent.
17817. A method as in any one of items 17596-17807, wherein the device is a biodegradable stent.
17818. A method as in any one of items 17596-17807, wherein the device is a non-biodegradable stent. 17819. A method as in any one of items 17596-17807, wherein the device is a self-expandable stent.
17820. A method as in any one of items 17596-17807, wherein the device is a balloon expandable stent.
17821. A method as in any one of items 17596-17807, wherein the device is a covered stent.
17822. A method as in any one of items 17596-17807, wherein the device is a drug eluting stent.
17823. A method as in any one of items 17596-17807, wherein the device is a stent that comprises a radio-opaque material.
17824. A method as in any one of items 17596-17807, wherein the device is a stent that comprises an echogenic material.
17825. A method as in any one of items 17596-17807, wherein the device is a stent that comprises an MRI responsive material.
17826. A method as in any one of items 17596-17807, wherein the device is an anastomotic connector device.
17827. A method as in any one of items 17596-17807, wherein the device is an artery to artery anastomotic connector device.
17828. A method as in any one of items 17596-17807, wherein the device is a vein to artery anastomotic connector device. 17829. A method as in any one of items 17596-17807, wherein the device is an artery to vein anastomotic connector device.
17830. A method as in any one of items 17596-17807, wherein the device is an artery to synthetic graft anastomotic connector device.
17831. A method as in any one of items 17596-17807, wherein the device is a synthetic graft to artery anastomotic connector device.
17832. A method as in any one of items 17596-17807, wherein the device is a vein to synthetic graft anastomotic connector device.
17833. A method as in any one of items 17596-17807, wherein the device is a synthetic graft to vein anastomotic connector device.
17834. A method as in any one of items 17596-17807, wherein the device is a vascular clip.
17835. A method as in any one of items 17596-17807, wherein the device is a vascular suture.
17836. A method as in any one of items 17596-17807, wherein the device is a vascular clamp.
17837. A method as in any one of items 17596-17807, wherein the device is a suturing device.
17838. A method as in any one of items 17596-17807, wherein the device is an anastomotic coupler. 17839. A method as in any one of items 17596-17807, wherein the device is an automated or modified suture device.
17840. A method as in any one of items 17596-17807, wherein the device is a micromechanical anastomotic connector device.
17841. A method as in any one of items 17596-17807, wherein the device is an anastomotic coupling device that facilitates automated attachment of a graft or vessel to an aperature or orifice in a target vessel without the use of sutures or staples.
17842. A method as in any one of items 17596-17807, wherein the device is an anastomotic coupling device that comprises a tubular graft conduit and may be placed in a side wall of a target vessel so that the tubular graft conduit may be extended from the target vessel.
17843. A method as in any one of items 17596-17807, wherein the device is an anastomotic coupler in the form of a frame.
17844. A method as in any one of items 17596-17807, wherein the device is an anastomotic coupler in a ring-like form.
17845. A method as in any one of items 17596-17807, wherein the device is a resorbable anastomotic coupler.
17846. A method as in any one of items 17596-17807, wherein the device is an anastomotic coupler that comprises a bioabsorbable and elastomeric material. 17847. A method as in any one of items 17596-17807, wherein the device is an anastomotic coupler adapted to connect a first blood vessel with a second blood vessel with a graft vessel.
17848. A method as in any one of items 17596-17807, wherein the device is an anastomotic coupler adapted to connect a first blood vessel with a second blood vessel without a graft vessel.
17849. A method as in any one of items 17596-17807, wherein the device is an anastomotic coupler that is incorporated in the design of a vascular graft.
17850. A method as in any one of items 17596-17807, wherein the device is an anastomotic coupler that comprises a graft that incorporates a fixation mechanism.
17851. A method as in any one of items 17596-17807, wherein the device is an anastomotic coupler that comprises a compressible, expandable fitting for securing the ends of a bypass graft to two vessels.
17852. A method as in any one of items 17596-17807, wherein the device is an anastomotic coupler that comprises a pair of coupling disc members for joining two vessels in an end to end or end to side fashion.
17853. A method as in any one of items 17596-17807, wherein the device is a proximal aoritic connector.
17854. A method as in any one of items 17596-17807, wherein the device is a distal coronary connector. 17855. A method as in any one of items 17596-17807, wherein the device is a bypass device made of a biocompatible material.
17856. A method as in any one of items 17596-17807, wherein the device is a bypass device made of at least partially a metal or metal alloy.
17857. A method as in any one of items 17596-17807, wherein the device is a bypass device made of at least partially a synthetic polymer.
17858. A method as in any one of items 17596-17807, wherein the device is a bypass device made of at least partially naturally derived polymer.
17859. A method as in any one of items 17596-17807, wherein the device is a tubular anastomotic coupler that comprises a tubular structure that may be attached directly to a proximal blood vessel.
17860. A method as in any one of items 17596-17807, wherein the device is a tubular anastomotic coupler that comprises a tubular structure that may be attached directly to a distal blood vessel.
17861. A method as in any one of items 17596-17807, wherein the device is a tubular anastomotic coupler that has a proximal end attachable to a proximal vessel and a distal end attachable to a bypass graft.
17862. A method as in any one of items 17596-17807, wherein the device is a tubular anastomotic coupler that has a proximal end attachable to a graft vessel that is secured to a proximal blood vessel and a distal end attachable to a distal blood vessel.
17863. A method as in any one of items 17596-17807, wherein the device is an anastomotic connector device adapted for end to end anastomosis procedures.
17864. A method as in any one of items 17596-17807, wherein the device is an anastomotic stent.
17865. A method as in any one of items 17596-17807, wherein the device is an anastomotic sleeve.
17866. A method as in any one of items 17596-17807, wherein the device is an anastomotic connector device adapted for end to side anastomosis procedures.
17867. A method as in any one of items 17596-17807, wherein the device is a single lumen bypass device.
17868. A method as in any one of items 17596-17807, wherein the device is a multi-lumen bypass device.
17869. A method as in any one of items 17596-17807, wherein the device is an anastomotic coupling device that comprises a single tubular portion that may be used as a shunt to divert blood from a source vessel to a graft vessel.
17870. A method as in any one of items 17596-17807, wherein the device is an anastomotic coupling device that comprises more than one tubular potion, and wherein at least one tubular potion may be used as a shunt for diverting blood between a source vessel and a target vessel.
17871. A method as in any one of items 17596-17807, wherein the device is an anastomotic connector device that comprises a tubular portion, and wherein one or more ends of the tubular portion may be inserted into the end or into the side of one or more blood vessels.
17872. A method as in any one of items 17596-17807, wherein the device is a multi-lumen anastomotic connector device that at least one arm of the device may be attached to a graft vessel.
17873. A method as in any one of items 17596-17807, wherein the device is an anastomic connect device that includes three or more tubular arms that extend from a junction site.
17874. A method as in any one of items 17596-17807, wherein the device is a multi-lumen anastomotic connector device is generally T-shaped.
17875. A method as in any one of items 17596-17807, wherein the device is a multi-lumen anastomotic connector device is generally Y-shaped.
17876. A method as in any one of items 17596-17807, wherein the device is an anastomotic connector device that comprises a tube for bypassing blood flow directly from a portion of a heart to a coronary artery. 17877. A method as in any one of items 17596-17807, wherein the device is an anastomotic connector device that comprises a network of interconnected tubular conduits.
17878. A method as in any one of items 17596-17807, wherein the device is an anastomotic connector device that is configured with two or more termini that provide a vessel interface without the need for sutures and a fluid communication through an intersecting lumen.
17879. A method for implanting a medical device comprinsing: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with i) an anti-fibrotic agent, ii) an anti-infective agent, iii) a polymer, iv) a composition comprising an anti-fibrotic agent and a polymer, v) a composition comprising an anti-infective agent and a polymer, or vi) a composition comprising an anti-fibrotic agent, an anti- infective agent and a polymer, and (b) implanting the medical device into the host, wherein the medical device is a gastrointestinal stent.
17880. The method of item 17879 wherein the anti- fibrotic agent is an adensosine A2A receptor antagonist.
17881. The method of item 17879 wherein the anti- fibrotic agent is an AKT inhibitor.
17882. The method of item 17879 wherein the anti- fibrotic agent is an alpha 2 integrin antagonist, wherein the alpha 2 integrin antagonist is Pharmaprojects No. 5754 (Merck KgaA).
17883. The method of item 17879 wherein the anti- fibrotic agent is an alpha 4 integrin antagonist. 17884. The method of item 17879 wherein the anti- fibrotic agent is an alpha 7 nicotinic receptor agonist.
17885. The method of item 17879 wherein the anti- fibrotic agent is an angiogenesis inhibitor selected from the group consisting of AG-12,958 (Pfizer), ATN-161 (Attenuon LLC), neovastat, an angiogenesis inhibitor from Jerina AG (Germany), NM-3 (Mercian), VGA-1155 (Taisho), FCE-26644 (Pfizer), FCE-26950 (Pfizer), FPMA (Meiji Daries), FR-111142 (Fujisawa), GGTI-298, GM-1306 (Ligand), GPA-1734 (Novartis), NNC-47- 0011 (Novo Nordisk), herbamycin (Nippon Kayaku), lenalidomide (Celegene), IP-10 (NIH), ABT-828 (Abbott), KIN-841 (Tokushima University, Japan), SF-1126 (Semafore Pharmaceuticals), laminin technology (NIH), CHIR-258 (Chiron), NVP-AEW541 (Novartis), NVP-AEW541 (Novartis), Vt16907 (Alchemia), OXI-8007 (Oxigene), EG-3306 (Ark Therapeutics), Maspin (Arriva), ABT-567 (Abbott), PPI-2458 (Praecis Pharmaceuticals), CC-5079, CC-4089 (Celgene), HIF-1 alpha inhibitors (Xenova), S-247 (Pfizer), AP-23573 (Ariad), AZD-9935 (Astra Zeneca), mebendazole (Introgen Therapeutics), MetAP-2 inhibitors (GlaxoSmithKline), AG-615 (Angiogene Pharmaceuticals), Tie-2 antagonists (Hybrigenics), NC-381 , CYC-381 , NC-169, NC-219, NC-383, NC-384, NC-407 (Lorus Therapeutics), ATN-224 (Attenuon), ON-01370 (Onconova), Vitronectin antagonists (Amgen), SDX-103 (Salmedix), Vitronectin antagonists (Shire), CHP (Riemser), TEK (Amgen), Anecortave acetate (Alcon), T46.2 (Matrix Therapeutics), HG-2 (Heptagen), TEM antagonists (Genzyme), Oxi-4500 (Oxigene), ATN-161 (Attenuon), WX-293 (Wilex), M-2025 (Metris Therapeutics), Alphastatin (BioActa), YH-16 (Yantai Rongchang), BIBF- 1120 (Boehringer Ingelheim), BAY-57-9352 (Bayer), AS-1404 (Cancer Research Technology), SC-77964 (Pfizer), glycomimetics (BioTie Therapies), TIE-2 Inhibitors (Ontogen), DIMI, Octamer (Octamer), ABR- 215050 (Active Biotech), ABT-518 (Abbott), KDR inhibitors (Abbott), BSF- 466895 (Abbott), SCH-221153 (Schering-Plough), DAC:antiangiogenic (ConjuChem), TFPI (EntreMed), AZD-2171 (Astra-Zenaca), CDC-394 (Celgene), LY290293 (EIi Lilly), IDN-5390 (Indena), Kdr Kinase Inhibitors (Merck), CT-113020, CT-116433, CT-116563, CT-31890, CT-32228) (Cell Therapeutics), A-299620 (Abbott), TWEAK Inhibitor (Amgen), VEGF modulators (Johnson and Johnson), Tum-N53, tumstatin (Genzyme), Thios- 1 , Thios-2 (Thios Pharmaceuticals), MV-6401 (Miravant Medical Technologies), Spisulosine (PharmaMar), CEP-7055 (Cephalon), AUV-201 (Auvation), LM-609 (EIi Lilly), SKF-106615 (AnorMED), Oglufanide disodium (Cytran), BW-114 (Pharminox), Calreticulin (NIH), WX-678 (Wilex), SD-7784 (Pfizer), WX-UK1 (Wilex), SH-268 (Schering AG), 2-Me-PGA (Celgene), S- 137 (Pfizer), ZD-6126 (Angiogene Pharmaceuticals), SG-292 (SignalGen), Benefin (Lane Labs), A6, A36 (Angstrom), SB-2723005 (GlaxoSmithKline), SC-7 (Cell Therapeutics), ZEN-014 (AEterna Zentaris), 2-methoxyestradiol (EntreMed), NK-130119 (Nippon Kayaku), CC-10004 (Celgene), AVE- 8062A (Ajinomoto), Tacedinaline (Pfizer), Actinonin (Tokyo Metropolitan Institute of Medical Science), Lenalidomide (Celgene), VGA-1155, BTO-956 (SRI International), ER-68203-00 (Eisai), CT-6685 (UCB), JKC-362 (Phoenix Pharmaceuticals), DMI-3798 (DMI Biosciences, Angiomate (Ipsen), ZD-6474 (AstraZeneca), CEP-5214 (Cephalon), Canstatin (Genzyme), NM-3 (Mercian), Oridigm (MediQuest Therapeutics), Exherin (Adherex), BLS-0597 (Boston Life Sciences), PTC-299 (PTC Therapeutics), NPI-2358 (Nereus Pharmaceuticals), CGP-79787 (Novartis), AEE-788 (Novartis), CKD-732 (Chong Kun Dang), CP-564959 (OSI Pharmaceuticals), CM-101 (CarboMed), CT-2584, CT3501 (Cell Therapeutics), combretastatin and analogues and derivatives thereof (Oxigene), Rebimastat (Bristol-Meyers Squibb), Dextrin 2-sulfate (ML Laboratories), Cilengitide (Merk KGaA), NSC-706704 (Pharminox), KRN- 951 (Kirin Brewery), Ukrain, NSC-631570 (Nowicky Pharma), Tecogalan — sodium (Daiichi Pharmaceutical), Tz-93 (Tsumura), TBC-1635 (Encysive Pharmaceuticals), TAN-1120 (Takeda), Semaxanib (Pfizer), BDl-7800 (Biopharmacopae), SD-186, SD-983 (Bristol-Meyers Squibb), SB-223245 (GlaxoSmithKline), SC-236 (Pfizer), RWJ-590973 (Johnson and Johnson), ILX-1850 (Genzyme), SC-68488, S-836 (Pfizer), CG-55069-11 (CuraGen), Ki-23057 (Kirin Brewery), CCX-700 (Chemoentryx), Pegaptanib octasodium (Giled Sciences), ANGIOCOL (available from Biostratum Inc.), or an analogue or derivative thereof.
17886. The method of item 17879 wherein the anti- fibrotic agent is an apoptosis antagonist.
17887. The method of item 17879 wherein the anti- fibrotic agent is an apoptosis activator.
17888. The method of item 17879 wherein the anti- fibrotic agent is a beta 1 integrin antagonist.
17889. The method of item 17879 wherein the anti- fibrotic agent is a beta tubulin inhibitor.
17890. The method of item 17879 wherein the anti- fibrotic agent is a blocker of enzyme production in Hepatitis C.
17891. The method of item 17879 wherein the anti- fibrotic agent is a Bruton's tyrosine kinase inhibitor.
17892. The method of item 17879 wherein the anti- fibrotic agent is a calcineurin inhibitor.
17893. The method of item 17879 wherein the anti- fibrotic agent is a caspase 3 inhibitor. 17894. The method of item 17879 wherein the anti- fibrotic agent is a CC chemokine receptor antagonist.
17895. The method of item 17879 wherein the anti- fibrotic agent is a cell cycle inhibitor selected from the group consisting of SNS-595 (Sunesis), synthadotin, KRX-0403, and an analogue or derivative thereof.
17896. The method of item 17879 wherein the anti- fibrotic agent is a cathepsin B inhibitor.
17897. The method of item 17879 wherein the anti- fibrotic agent is a cathepsin K inhibitor, wherein the cathepsin K inhibitor is 462795 (GlaxoSmithKline), INPL-022-D6 (Amura Therapeutics), or an analogue or derivative thereof.
17898. The method of item 17879 wherein the anti- fibrotic agent is a cathepsin L inhibitor.
17899. The method of item 17879 wherein the anti- fibrotic agent is a CD40 antagonist.
17900. The method of item 17879 wherein the anti- fibrotic agent is a chemokine receptor agonist.
17901. The method of item 17879 wherein the anti- fibrotic agent is a chymase inhibitor.
17902. The method of item 17879 wherein the anti- fibrotic agent is a collagenase antagonist. 17903. The method of item 17879 wherein the anti- fibrotic agent is a CXCR antagonist.
17904. The method of item 17879 wherein the anti- fibrotic agent is a cyclin dependent kinase inhibitor selected from the group consisting of a CDK-1 inhibitor, a CDK-2 inhibitor, a CDK- 4 inhibitor, a CDK-6 inhibitor, a CAK1 inhibitor from GPC Biotech and Bristol-Myers Squibb, RGB-286199 (GPC Biotech), an anticancer agent from Astex Technology, a CAK1 inhibitor from GPC Biotech, a CDK inhibitor from Sanofi-Aventis, a CDK1/CDK2 inhibitor from Amgen, a CDK2 inhibitor from SUGEN-2 (Pfizer), a hearing loss therapy agent (Sound Pharmaceuticals), PD-0332991 (Pfizer), RGB-286199 (GPC Biotech), Ro-0505124 (Hoffmann- La Roche), a Ser/Thr kinase inhibitor from Lilly (EIi Lilly), CVT-2584 (CAS No. 199986-75-9) (CV Therapeutics), and an analogue or derivative thereof.
17905. The method of item 17879 wherein the anti- fibrotic agent is a cyclooxygenase 1 inhibitor.
17906. The method of item 17879 wherein the anti- fibrotic agent is a DHFR inhibitor.
17907. The method of item 17879 wherein the anti- fibrotic agent is a dual integrin inhibitor.
17908. The method of item 17879 wherein the anti- fibrotic agent is an elastase inhibitor.
17909. The method of item 17879 wherein the anti- fibrotic agent is an elongation factor-1 alpha inhibitor. 17910. The method of item 17879 wherein the anti- fibrotic agent is an endothelial growth factor antagonist.
17911. The method of item 17879 wherein the anti- fibrotic agent is an endothelial growth factor receptor kinase inhibitor selected from the group consisting of sorafenib tosylate (Bayer), AAL-993 (Novartis), ABP-309 (Novartis), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EXEL- 2880 (Exelixis), GW-654652 (GlaxoSmithKline), a KDR inhibitor from LG Life Sciences, CT-6685 and CT-6729 (UCB), KRN-633 and KRN-951 (Kirin Brewery), OSI-930 (OSI Pharmaceuticals), SP-5.2 (Supratek Pharma), SU-
11657 (Pfizer), a Tie-2 antagonist (Hybrigenics), a VEGFR-2 kinase inhibitor (Bristol-Myers Squibb), XL-647 (Exelixis), a KDR inhibitor from Abbott Laboratories, sorafenib tosylate, and an analogue or derivative thereof.
17912. The method of item 17879 wherein the anti- fibrotic agent is an endotoxin antagonist.
17913. The method of item 17879 wherein the anti- fibrotic agent is an epothilone and tubulin binder.
17914. The method of item 17879 wherein the anti- fibrotic agent is an estrogen receptor antagonist.
17915. The method of item 17879 wherein the anti- fibrotic agent is an FGF inhibitor.
17916. The method of item 17879 wherein the anti- fibrotic agent is a farnexyl transferase inhibitor. 17917. The method of item 17879 wherein the anti- fibrotic agent is farnesyltransferase inhibitor selected from the group of A- 197574 (Abbott), a farnesyltransferase inhibitor from Servier, FPTIII (Strathclyde Institute for Drug R)1 LB-42908 (LG Life Sciences), Pharmaprojects No. 5063 (Genzyme), Pharmaprojects No. 5597 (Ipsen), Yissum Project No. B-1055 (Yissum), and an analogue or derivative thereof.
17918. The method of item 17879 wherein the anti- fibrotic agent is an FLT-3 kinase inhibitor.
17919. The method of item 17879 wherein the anti- fibrotic agent is an FGF receptor kinase inhibitor.
17920. The method of item 17879 wherein the anti- fibrotic agent is a fibrinogen antagonist selected from the group consisting of AUV-201 (Auvation), MG-13926 (Sanofi-Aventis), plasminogen activator (CAS No. 105913-11-9) (from Sanofi-Aventis or UCB), plasminogen activator-2 (tPA-2) (Sanofi-Aventis), pro-urokinase (CAS No. 82657-92-9) (Sanofi-Aventis), and an analogue or derivative thereof.
17921. The method of item 17879 wherein the anti- fibrotic agent is a heat shock protein 90 antagonist selected from the group consisting of SRN-005 (Sirenade), geldanamycin, NSC-33050 (17- allylaminogeldanamycin; 17-AAG), 17-dimethylaminoethylamino-17- demethoxy-geldanamycin (17-DMAG), rifabutin (rifamycin XIV, 1',4- didehydro-1-deoxy-1,4-dihydro-5'-(2-methylpropyl)-1-oxo-), and an analogue or derivative thereof.
17922. The method of item 17879 wherein the anti- fibrotic agent is a histone deacetylase inhibitor. 17923. The method of item 17879 wherein the anti- fibrotic agent is an HMGCoA reductase inhibitor selected from the group consisting of an atherosclerosis therapeutic from Lipid Sciences, ATI-16000 (ARYx Therapeutics), KS-01-019 (Kos Pharmaceuticals), Pharmaprojects No. 2197 (Sanofi-Aventi), RP 61969 (Sanofi-Aventis), and an analogue or derivative thereof.
17924. The method of item 17879 wherein the anti- fibrotic agent is an ICAM inhibitor.
17925. The method of item 17879 wherein the anti- fibrotic agent is an IL, ICE and IRAK antagonist, wherein the antagonist is a CJ-14877, CP-424174 (Pfizer), NF-61 (Negma-Lerads), and an analogue or derivative thereof.
17926. The method of item 17879 wherein the anti- fibrotic agent is an IL-2 inhibitor.
17927. The method of item 17879 wherein the anti- fibrotic agent is an immunosuppressant selected from the group consisting of teriflunomide (Sanofi Aventis), chlorsulfaquinoxalone (NSC-339004), chlorsulfaquinoxalone sulfate, CS-712 (Sankyo), ismomultin alfa (CAS No. 457913-93-8) (Akzo Nobel), antiallergics from GenPat77, antiinflammatories or AT-005 (Androclus Therapeutics), autoimmune disease therapeutics from EpiVax, BN-007 (Bone), budesonide (CAS No. 51333-22- 3) (MAP Pharmaceuticals), CO-14 (Genzyme), edratide (CAS No. 433922- 67-9) (Teva), EP-314 (Enanta), eprovafen (CAS No. 101335-99-3) (Sanofi- Aventis), HWA-131 (CAS No. 118788-41-3) (Sanofi-Aventis), immunomodulators from MerLion Pharmaceuticals, immunosuppressives from Alchemia, IPL-12 (Inflazyme), MDL-9563 (CAS No. 27086-86-8) (Sanofi-Aventis), Pharmaprojects No. 2330 (Sanofi-Aventis), Pharmaprojects No. 6426 (Abgenix), PXS-25 (Pharmaxis), rosmarinic acid (CAS No. 20283-92-5) (Sanofi-Aventis), RP 42927 or RP 54745 (CAS No. 135330-08-4) (Sanofi-Aventis), SGN-35 (Seattle Genetics), ST-1959 (Sigma-Tau), type I diabetes therapy from SYNX Pharma, UNIL-88 (Debiopharm), VP-025 (Vasogen), VR-694 (Vectura), PRTX-001 (Protalex), and an analogue or derivative thereof.
17928. The method of item 17879 wherein the anti- fibrotic agent is an IMPDH (inosine monophosphate).
17929. The method of item 17879 wherein the anti- fibrotic agent is an integrin antagonist.
17930. The method of item 17879 wherein the anti- fibrotic agent is an interleukin antagonist.
17931. The method of item 17879 wherein the anti- fibrotic agent is an inhibitor of type III receptor tyrosine kinase.
17932. The method of item 17879 wherein the anti- fibrotic agent is an irreversible inhibitor of enzyme methionine aminopeptidase type 2.
17933. The method of item 17879 wherein the anti- fibrotic agent is an isozyme selective delta protein kinase C inhibitor.
17934. The method of item 17879 wherein the anti- fibrotic agent a JAK3 enzyme inhibitor.
17935. The method of item 17879 wherein the anti- fibrotic agent is a JNK inhibitor. 17936. The method of item 17879 wherein the anti- fibrotic agent is a kinase inhibitor.
17937. The method of item 17879 wherein the anti- fibrotic agent is kinesin antagonist.
17938. The method of item 17879 wherein the anti- fibrotic agent is a kinesin antagonist.
17939. The method of item 17879 wherein the anti- fibrotic agent is a leukotriene inhibitor and antagonist selected from the group consisting of ambicromil (CAS No. 58805-38-2) (Sanofi-Aventis), amelubant (CAS No. 346735-24-8) (Boehringer Ingelheim), DW-1141 (Dong Wha), ebselen (Daiichi Pharmaceutical), ibudilast (Kyorin), leucotriene inhibitors from Sanofi-Aventis, lymphotoxin-beta receptor (LT-D) from Biogen Idee, Pharmaprojects No. 1535 and 2728 (CAS No. 119340-33-9) (Sanofi-Aventis), R-112 (Rigel), Rev-5367 (CAS No. 92532-05-3) (Sanofi- Aventis), RG-14893 (CAS No. 141835-49-6) (Sanofi-Aventis), RG-5901-A (CAS No. 101910-24-1), 92532-23-5, RP 66153 (CAS No. 142422-79-5), RP 66364 (CAS No.186916-92-5), RP 69698 (CAS No. 141748-00-7) (Sanofi-Aventis), SC-411930 (Pfizer), SC-41930 (CAS No. 120072-59-5) (Pfizer), SC-50605 (CAS No. 138828-39-4) (Pfizer), SC-51146 (CAS No. 141059-52-1), SC-53228 (CAS No. 153633-01-3) (Pfizer), spaglumic acid (ZY-15106) (CAS No.3106-85-2), 80619-64-3 (Novartis), tipredane (CAS No. 85197-77-9) (Bristol-Myers Squibb), U-75302 (CAS No. 119477-85-9) (Pfizer), and analogue or derivative thereof.
17940. The method of item 17879 wherein the anti- fibrotic agent is an MAP kinase inhibitor. 17941. The method of item 17879 wherein the anti- fibrotic agent is a matrix metalloproteinase inhibitor.
17942. The method of item 17879 wherein the anti- fibrotic agent is an MCP-CCR2 inhibitor.
17943. The method of item 17879 wherein the anti- fibrotic agent is an mTOR inhibitor.
17944. The method of item 17879 wherein the anti- fibrotic agent is an mTOR kinase inhibitor.
17945. The method of item 17879 wherein the anti- fibrotic agent is a microtubule inhibitor selected from the group consisting of antibody-maytansinoid conjugates from Biogen Idee, colchicines (MantiCore Pharmaceuticals), anticancer immunoconjugates from Johnson & Johnson, DIME from Octamer, gni-1f (GNI), huC242-DM4, huMy9-6-DM1 (ImmunoGen), IDN-5404 (Indena), IMO-098, IMOderm (Imotep), mebendazole (Introgen Therapeutics), microtubule poisons from Cambridge Enterprise, paclitaxel such as LOTAX from Aphios (CAS No. 33069-62-4), Genexol-PM from Samyang, Pharmaprojects No. 6383 (Tapestry Pharmaceuticals), RPR-112378 (Sanofi-Aventis), SGN-75 (Seattle Genetics), SPL-7435 (Starpharma), SSR-250411 (Sanofi-Aventis), trastuzumab-DM1 (Genentech), vinorelbine, and an analogue or derivative thereof.
17946. The method of item 17879 wherein the anti- fibrotic agent is an MIF inhibitor.
17947. The method of item 17879 wherein the anti- fibrotic agent is an MMP inhibitor. 17948. The method of item 17879 wherein the anti- fibrotic agent is a neurokinin (NK) antagonist selected from the group consisting of anthrotainin (CAS No. 148084-40-6) (Sanofi-Aventis), an IBS thereapeutic from ArQuIe1 MDL-105212A (CAS No. 167261-60-1) (Ssanofi- Aventis), Pharmaprojects No. 2744, 3258 (CAS No. 139167-47-8) 4006, 4201 , or 5986 (Sanofi-Aventis), RP 67580 (CAS No. 135911-02-3), SR- 144190 (CAS No. 201152-86-5), SSR-240600, SSR-241586 (Sanofi- Aventis), TKA-457 (Novartis), vestipitant mesylate (CAS No. 334476-64-1) (GlaxoSmithKline), Win-64821 (Sanofi-Aventis), PRX-96026 (Predix Pharmaceuticals), and an analogue or derivative thereof.
17949. The method of item 17879 wherein the anti- fibrotic agent is an NF kappa B inhibitor selected from the group consisting of AVE-0545 or AVE-0547 (Sanofi-Aventis), bortezomib (CAS No. 179324- 69-7) (Millennium Pharmaceuticals), dexanabinol (CAS No. 112924-45-5) (Pharmos), dexlipotam (Viatris), Pharmaprojects No. 6283 (INDRA) (OXiGENE), IPL-576092 (CAS No. 137571-30-3) (Inflazyme), NFKB decoy (Corgentech), NFKB decoy oligo (AnGes MG), NFKB's from Ariad, osteoporosis treatments or S5 (F005) from Fulcrum Pharmaceuticals, P61 (Phytopharm), R-flurbiprofen (CAS No. 5104-49-4) (Encore Pharmaceuticals), and an analogue or derivative thereof.
17950. The method of item 17879 wherein the anti- fibrotic agent is a nitric oxide agonist.
17951. The method of item 17879 wherein the anti- fibrotic agent is an ornithine decarboxylase inhibitor.
17952. The method of item 17879 wherein the anti- fibrotic agent is a p38 MAP kinase inhibitor selected from the group consisting of AZD-6703 (AstraZeneca), JX-401 (Jexys Pharmaceuticals), BMS-2 (Bristol-Myers Squibb), a p38 MAP kinase inhibitor from Novartis, a p38-alpha MAP kinase inhibitor from Amphora, Pharmaprojects No. 5704 (Pharmacopeia), RPR-200765A (Sanofi-Aventis), SD-282 (Johnson & Johnson), TAK-715 (Takeda), and an analogue or derivative thereof.
17953. The method of item 17879 wherein the anti- fibrotic agent is a palmitoyl-protein thioesterase inhibitor.
17954. The method of item 17879 wherein the anti- fibrotic agent is a PDGF receptor kinase inhibitor selected from the group consisting of AAL-993, AMN-107, or ABP-309 (Novartis), AMG-706 (Amgen), BAY-57-9352 (Bayer), CDP-860 (UCB), E-7080 (Eisai), imatinib (CAS No. 152459-95-5) (Novartis), OSI-930 (OSI Pharmaceuticals), RPR- 127963E (Sanofi-Aventis), RWJr540973 (Johnson & Johnson), sorafenib tosylate (Bayer), SU-11657 (Pfizer), tandutinib (CAS No. 387867-13-2) (Millennium Pharmaceuticals), vatalanib (Novartis), ZK-CDK (Schering AG), and an analogue or derivative thereof.
17955. The method of item 17879 wherein the anti- fibrotic agent is (-)-halofenate (Metabolex), AMG-131 (Amgen), antidiabetics from Japan Tobacco, AZD-4619, AZD-8450, AZD-8677 (AstraZeneca), DRF-10945, balaglitazone (Dr Reddy's), CS-00088, CS-00098 (Chipscreen Biosciences), E-3030 (Eisai), etalocib (CAS No. 161172-51-6) (EIi Lilly), GSK-641597 (Ligand), GSK-677954 (GlaxoSmithKline), GW-409544 (Ligand), GW-590735 (GlaxoSmithKline), K-111 (Hoffmann-La Roche), LY- 518674 (EIi Lilly), LY-674 (Ligand), LY-929 (Ligand), MC-3001 , MC-3002 (MaxoCore Pharmaceuticals), metformin HCI + pioglitazone (CAS No. 1115- 70-4 and 112529-15-4), ACTOPLUS MET from Andrx), muraglitazar (CAS No. 331741-94-7) (Bristol-Myers Squibb), naveglitazar (Ligand), oleoylethanolamide (Kadmus Pharmaceuticals), ONO-5129, pioglitazone hydrochloride (CAS No. 111025-46-8 and 112529-15-4) (Takeda), PLX-204 (Plexxikon), PPAR agonists from Genfit, PPAR delta agonists from EIi Lilly, PPAR-alpha agonists from CrystalGenomics, PPAR-gamma modulators and PPAR-β modulators from CareX, rosiglitazone maleate (CAS No. 122324- 73-4 or 155141-29-0) (GlaxoSmithKline), rosiglitazone maleate/glimepir (CAS No. 155141-29-0 and 93479-97-1), AVANDARYL, rosiglitazone maleate/metformin extend (CAS No. 155141-29-0 and 657-24-9), AVANDAMET, rosiglitazone maleate+metformin, AVANDAMET (GlaxoSmithKline), tesaglitazar (AstraZeneca), and an analogue or derivative thereof.
17956. The method of item 17879 wherein the anti- fibrotic agent is a phosphatase inhibitor.
17957. The method of item 17879 wherein the anti- fibrotic agent is a phosphodiesterase (PDE) inhibitor selected from the group consisting of avanafil (Tanabe Seiyaku), dasantafil (CAS No. 569351- 91-3) (Schering-Plough), A-906119 (CAS No. 134072-58-5), DL-850 (Sanofi-Aventis), GRC-3015, GRC-3566, GRC-3886 (Glenmark), HWA-153 (CAS No. 56395-66-5) (Sanofi-Aventis), hydroxypumafentrine (Altana), IBFB-130011 , IBFB-14-016, IBFB-140301 , IBFB-150007, IBFB-211913 (IBFB Pharma), L-826141 (Merck & Co), medorinone (CAS No. 88296-61-1) (Sanofi-Aventis), MEM-1917 (Memory Pharmaceuticals), ND-1251 (Neuro3d), PDE inhibitors from ICOS, PDE IV inhibitors from Memory Pharmaceuticals and CrystalGenomics, Pharmaprojects No. 2742 and 6141 (Sanofi-Aventis), QAD-171 (Novartis), RHC-2963 (CAS No. 76993-12-9 and 76993-14-1), RPR-117658, RPR-122818 derivatives, SR-24870 , and RPR- 132294 (Sanofi-Aventis), SK-350 (ln2Gen), stroke therapy agents from deCODE Genetics, TAS-203 (Taiho), tofimilast (CAS No. 185954-27-2) (Pfizer), UK-371800 (Pfizer), WIN-65579 (CAS No. 158020-82-7) (Sanofi- Aventis), IBFB-130020 (IBFB Pharma), OPC-6535 (CAS No. 145739-56-6) (Otsuka), a phosphodiesterase III inhibitor, enoximone, a phosphodiesterase IV inhibitor, fosfosal, Atopik (Barrier Therapeutics), triflusal, a phosphodiesterase V inhibitor, and an analogue or derivative thereof.
17958. The method of item 17879 wherein the anti- fibrotic agent is a PKC inhibitor.
17959. The method of item 17879 wherein the anti- fibrotic agent is a platelet activating factor antagonist.
17960. The method of item 17879 wherein the anti- fibrotic agent is a platelet-derived growth factor receptor kinase inhibitor.
17961. The method of item 17879 wherein the anti- fibrotic agent is a prolyl hydroxylase inhibitor.
17962. The method of item 17879 wherein the anti- fibrotic agent is a polymorphonuclear neutrophil inhibitor.
17963. The method of item 17879 wherein the anti- fibrotic agent is a protein kinase B inhibitor.
17964. The method of item 17879 wherein the anti- fibrotic agent is a protein kinase C stimulant.
17965. The method of item 17879 wherein the anti- fibrotic agent is a purine nucleoside analogue.
17966. The method of item 17879 wherein the anti- fibrotic agent is a purinoreceptor P2X antagonist. 17967. The method of item 17879 wherein the anti- fibrotic agent is a Raf kinase inhibitor.
17968. The method of item 17879 wherein the anti- fibrotic agent is a reversible inhibitor of ErbB1 and ErbB2.
17969. The method of item 17879 wherein the anti- fibrotic agent is a ribonucleoside triphosphate reductase inhibitor.
17970. The method of item 17879 wherein the anti- fibrotic agent is an SDF-1 antagonist.
17971. The method of item 17879 wherein the anti- fibrotic agent is a sheddase inhibitor.
17972. The method of item 17879 wherein the anti- fibrotic agent is an SRC inhibitor.
17973. The method of item 17879 wherein the anti- fibrotic agent is a stromelysin inhibitor.
17974. The method of item 17879 wherein the anti- fibrotic agent is an Syk kinase inhibitor.
17975. The method of item 17879 wherein the anti- fibrotic agent is a telomerase inhibitor.
17976. The method of item 17879 wherein the anti- fibrotic agent is a TGF beta inhibitor selected from the group consisting of pirfenidone (CAS No. 53179-13-8) (MARNAC), tranilast (CAS No. 53902- 12-8) (Kissei), IN-1130 (ln2Gen), mannose-6-phosphate (BTG), TGF-β antagonists from Inflazyme (Pharmaprojects No. 6075), TGF-(I antagonists from Sydney, non-industrial source), TGF-β I receptor kinase inhibitors from EIi Lilly, TGF-β receptor inhibitors from Johnson & Johnson, and an analogue or derivative thereof.
17977. The method of item 17879 wherein the anti- fibrotic agent is a TNFα antagonist or TACE inhibitor selected from the group consisting of adalimumab (CAS No. 331731-18-1) (Cambridge Antibody Technology), AGIX-4207 (AtheroGenics), AGT-1 (Advanced Biotherapy), an anti-inflammatory from Borean Pharma, Cellzome, or Paradigm Therapeutics, anti-inflammatory vaccine (TNF-alpha kinoid) from Neovacs, humanized anti-TNF antibody or an anti-TNF MAb (CB0006) Celltech (UCB), apratastat (CAS No. 287445-51-0) (Wyeth), BMS-561392 (Bristol-Myers Squibb), BN-006 (Bone), certolizumab pegol (CAS No. 428863-50-7 or CH-138 (UCB), cilomilast (CAS No. 153259-65-5) (GlaxoSmithKline), CR-1 (Nuada Pharmaceuticals), CRx-119 (CombinatoRx), D-5410 (UCB), dacopafant (CAS No. 125372-33-0) (Sanofi- Aventis), dersalazine (CAS No. 188913-57-7/188913-58-8) (Uriach), etanercept (CAS No. 185243-69-0) (Amgen), ethyl pyruvate (Critical (Critical Therapeutics), golimumab (CAS No. 476181-74-5) (Johnson & Johnson), hormono-immunotherapy from Ipsen, CDP571 (e.g., Humicade from UCB), IC-485 (ICOS), infliximab (CAS No. 170277-31-3) (Johnson & Johnson), IP- 751 (Manhattan Pharmaceuticals), ISIS-104838 (CAS No. 250755-32-9) (ISIS Pharmaceuticals), lenalidomide (CAS No. 191732-72-6) (Celgene), lentinan (CAS No. 37339-90-5) (Ajinomoto), MDL-201112 (CAS No. 142130-73-2) (Sanofi-Aventis), medroxyprogesterone (CAS No. 520-85-4) (InKine Pharmaceutical), N-acetylcysteine (CAS No. 616-91-1) (Zambon), NBE-P2 (DIREVO Biotech), nerelimomab (CAS No. 162774-06-3) (Chiron), OM-294DP (OM PHARMA), onercept (CAS No. 199685-57-9) (Yeda), PASSTNF-alpha (Verigen), pentoxifylline or oxypentifylline (Sanofi-Aventis), Pharmaprojects No. 4091 , 4241, 4295, or 4488 (Sanofi-Aventis), Pharmaprojects No. 5480 (Amgen), Pharmaprojects No. 6749 (Cengent), pirfenidone (CAS No. 53179-13-8) (MARNAC), RPR-132294 (Sanofi- Aventis), S5 (F002) (Fulcrum Pharmaceuticals), simvastatin (CAS No. 79902-63-9) (Merck & Co), STA-6292 (Synta Pharmaceuticals), tacrolimus (CAS No. 104987-11-3) (Fujisawa LifeCycle Pharma), talactoferrin alfa (CAS No. 308240-58-6) (Agennix), thalidomide (CAS No. 50-35-1) (Celgene), TNF antagonists form ProStrakan, and Synergen, TNF inhibitors (Amgen), TNF-alpha antagonists from Dynavax Technologies and Jerina AG (Germany), TNF-alpha inhibitors from IBFB Pharma and Xencor (Xencor), torbafylline (CAS No. 105102-21-4) (Sanofi-Aventis), UR-1505 (Uriach), VT-346 (Viron Therapeutics), YSIL6 (Y1S Therapeutics), YSTH2 (Y's Therapeutics), NPI-1302a-3 (Nereus Pharmaceuticals, a TNF antagonist from Jerina AG (Germany), dersalazine, and an analogue or derivative thereof.
17978. The method of item 17879 wherein the anti- fibrotic agent is a Toll receptor inhibitor.
17979. The method of item 17879 wherein the anti- fibrotic agent is a tubulin antagonist.
17980. The method of item 17879 wherein the anti- fibrotic agent is a tyrosine kinase inhibitor selected from the group consisting of SU-011248, SUTENT from Pfizer Inc. (New York, NY), BMS-354825, PN- 355 (Paracelsian Pharmaceuticals), AGN-199659 (Allergan), AAL-993 or ABP-309 (Novartis), adaphostin (NIH), AEE-788 (Novartis), AG-013736 (OSI Pharmaceuticals), AG-13736 (Pfizer), ALT-110 (Alteris Therapeutics), AMG-706 (Amgen), anticancer MAbs from Xencor, anti-EGFrvlll MAbs from Abgenix, anti-HER2 MAb from Abiogen, AZD-2171 or AZD-9935 (AstraZeneca), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), CEP-5214 (Cephalon), CEP-7055 (Cephalon), cetuximab (ImClone Systems), CHIR-200131 and CHIR-258 (Chiron), CP-547632 (OSI Pharmaceuticals), CP-724714 (Pfizer), CT-301 (Creabilis Therapeutics), D- 69491 (Baxter International), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EGFR/ErbB2 inhibitors from Array BioPharma, erlotinib (CAS No. 183319- 69-9) (OSI Pharmaceuticals), EXEL-2880 (Exelixis), FK-778 (Sanofi- Aventis) , gefitinib (CAS No. 184475-35-2) (AstraZeneca), GW-2286 or GW- 654652 (GlaxoSmithKline), her2/neu antigen from AlphaVax, HER-2/neu inhibitor from Generex, Herzyme (Medipad) (Sirna Therapeutics), HKI-272 (Wyeth), HuMax-EGFr (Genmab), idronoxil (CAS No. 81267-65-4) (Novogen), IGF-1 inhibitors from Ontogen, IMC-11F8 (ImClone Systems), kahalalide F (CAS No. 149204-42-2) (PharmaMar), KDR inhibitor from LG Life Sciences, KDR inhibitors from Abbott Laboratories, KDR kinase inhibitors (UCB), Kdr kinase inhibitors from Merck & Co, KRN-633 and KRN- 951 (Kirin Brewery), KSB-102 (Xenova), lapatinib ditosylate (CAS No. 388082-78-8) (GlaxoSmithKline), matuzumab (Merck KGaA), MDX-214 (Medarex), ME-103 (Pharmexa), MED-A300 (Gerolymatos), MNAC-13 (Lay Line Genomics), nimotuzumab (Center of Molecular Immunology), NSC- 330507 or NSC-707545 (NIH), NV-50 (Novogen), OSI-930 (OSI Pharmaceuticals), panitumumab (Abgenix), pelitinib (CAS No. 287933-82-7) (Wyeth), pertuzumab (CAS No. 380610-27-5) (Genentech), Pharmaprojects No. 3985 (Sanofi-Aventis), prostate cancer therapeutics from Sequenom (SQPC35, SQPC36, SQPC90), removab and remoxab (Trion Pharma), RG- 13022 (CAS No. 136831-48-6), RG-13291 (CAS No. 138989-50-1), or RG- 14620 (CAS No. 136831-49-7) (Sanofi-Aventis), RM-6427 (Romark), RNAi breast cancer therapy from Benitec, RP 53801 (CAS No. 125882-88-4) (Sanofi-Aventis), sorafenib tosylate (Bayer), SU-11657 (Pfizer), Tie-2 antagonists from Semaia (Hybrigenics), Tie-2 inhibitors from Ontogen, trastuzumab (CAS No. 180288-69-1) (Genentech), tyrosine kinase inhibitors from Sanofi-Aventis, U3-1287, U3-1565, U3-1784, U3-1800 (U3 Pharma), vatalanib (Novartis), VEGFR-2 kinase inhibitor from Bristol-Myers Squibb, XL-647 (Exelixis), ZD-6474 (AstraZeneca), ZK-CDK (Schering AG), an EGFR tyrosine kinase inhibitor, EKB-569 (Wyeth), and an analogue or derivative thereof.
17981. The method of item 17879 wherein the anti- fibrotic agent is a VEGF inhibitor.
17982. The method of item 17879 wherein the anti- fibrotic agent is a vitamin D receptor agonist.
17983. The method of item 17879 wherein the anti- fibrotic agent is ZD-6474 (an angiogenesis inhibitor).
17984. The method of item 17879 wherein the anti- fibrotic agent is AP-23573 (an mTOR inhibitor).
17985. The method of item 17879 wherein the anti- fibrotic agent is synthadotin (a tubulin antagonist).
17986. The method of item 17879 wherein the anti- fibrotic agent is S-0885 (a collagenase inhibitor).
17987. The method of item 17879 wherein the anti- fibrotic agent is aplidine (an elongation factor-1 alpha inhibitor).
17988. The method of item 17879 wherein the anti- fibrotic agent is ixabepilone (an epithilone).
17989. The method of item 17879 wherein the anti- fibrotic agent is IDN-5390 (an angiogenesis inhibitor). 17990. The method of item 17879 wherein the anti- fibrotic agent is SB-2723005 (an angiogenesis inhibitor).
17991. The method of item 17879 wherein the anti- fibrotic agent is ABT-518 (an angiogenesis inhibitor).
17992. The method of item 17879 wherein the anti- fibrotic agent is combretastatin (an angiogenesis inhibitor).
17993. The method of item 17879 wherein the anti- fibrotic agent is anecortave acetate (an angiogenesis inhibitor).
17994. The method of item 17879 wherein the anti- fibrotic agent is SB-715992 (a kinesin antagonist).
17995. The method of item 17879 wherein the anti- fibrotic agent is temsirolimus (an mTOR inhibitor).
17996. The method of item 17879 wherein the anti- fibrotic agent is adalimumab (a TNFα antagonist).
17997. The method of item 17879 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with an anti-fibrotic agent, and (b) implanting the medical device into the host.
17998. The method of item 17879 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with an anti-infective agent, and (b) implanting the medical device into the host. 17999. The method of item 17879 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with a polymer; and (b) implanting the medical device into the host.
18000. The method of item 17879 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with a composition comprising an anti-fibrotic agent and a polymer, and (b) implanting the medical device into the host.
18001. The method of item 17879 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with a composition comprising an anti-infective agent and a polymer, and (b) implanting the medical device into the host.
18002. The method of item 17879 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with a composition comprising an anti-fibrotic agent, an anti- infective agent and a polymer, and (b) implanting the medical device into the host.
18003. The method of item 17879 wherein the anti- infective agent is an anthracycline.
18004. The method of item 17879 wherein the anti- infective agent is doxorubicin.
18005. The method of item 17879 wherein the anti- infective agent is is mitoxantrone. 18006. The method of item 17879 wherein the anti- infective agent is a fluoropyrimidine.
18007. The method of item 17879 wherein the anti- infective agent is 5-fluorouracil (5-FU).
18008. The method of item 17879 wherein the anti- infective agent is a folic acid antagonist.
18009. The method of item 17879 wherein the anti- infective agent is methotrexate.
18010. The method of item 17879 wherein the anti- infective agent is a podophylotoxin.
18011. The method of item 17879 wherein the anti- infective agent is etoposide.
18012. The method of item 17879 wherein the anti- infective agent is camptothecin.
18013. The method of item 17879 wherein the anti- infective agent is a hydroxyurea.
18014. The method of item 17879 wherein the anti- infective agent is a platinum complex.
18015. The method of item 17879 wherein the anti- infective agent is cisplatin. 18016. The method of item 17879 wherein the composition comprises an antithrombotic agent.
18017. The method of item 17879 wherein the polymer is formed from reactants comprising a naturally occurring polymer.
18018. The method of item 17879 wherein the polymer is formed from reactants comprising protein.
18019. The method of item 17879 wherein the polymer is formed from reactants comprising carbohydrate.
18020. The method of item 17879 wherein the polymer is formed from reactants comprising biodegradable polymer.
18021. The method of item 17879 wherein the polymer is formed from reactants comprising nonbiodegradable polymer.
18022. The method of item 17879 wherein the polymer is formed from reactants comprising collagen.
18023. The method of item 17879 wherein the polymer is formed from reactants comprising methylated collagen.
18024. The method of item 17879 wherein the polymer is formed from reactants comprising fibrinogen.
18025. The method of item 17879 wherein the polymer is formed from reactants comprising thrombin. 18026. The method of item 17879 wherein the polymer reactants comprising blood plasma.
18027. The method of item 17879 wherein the polymer reactants comprising calcium salt.
18028. The method of item 17879 wherein the polymer reactants comprising an antifibronolytic agent.
18029. The method of item 17879 wherein the polymer reactants comprising fibrinogen analog.
18030. The method of item 17879 wherein the polymer reactants comprising albumin.
18031. The method of item 17879 wherein the polymer reactants comprising plasminogen.
18032. The method of item 17879 wherein the polymer reactants comprising von Willebrands factor.
18033. The method of item 17879 wherein the polymer reactants comprising Factor VIII.
18034. The method of item 17879 wherein the polymer reactants comprising hypoallergenic collagen.
18035. The method of item 17879 wherein the polymer reactants comprising atelopeptidic collagen. 18036. The method of item 17879 wherein the polymer is formed from reactants comprising telopeptide collagen.
18037. The method of item 17879 wherein the polymer is formed from reactants comprising crosslinked collagen.
18038. The method of item 17879 wherein the polymer is formed from reactants comprising aprotinin.
18039. The method of item 17879 wherein the polymer is formed from reactants comprising epsilon-amino-n-caproic acid.
18040. The method of item 17879 wherein the polymer is formed from reactants comprising gelatin.
18041. The method of item 17879 wherein the polymer is formed from reactants comprising protein conjugates.
18042. The method of item 17879 wherein the polymer is formed from reactants comprising gelatin conjugates.
18043. The method of item 17879 wherein the polymer is formed from reactants comprising a synthetic polymer.
18044. The method of item 17879 wherein the polymer is formed from reactants comprising a synthetic isocyanate-containing compound.
18045. The method of item 17879 wherein the polymer is formed from reactants comprising a synthetic thiol-containing compound. 18046. The method of item 17879 wherein the polymer is formed from reactants comprising a synthetic compound containing at least two thiol groups.
18047. The method of item 17879 wherein the poiymer is formed from reactants comprising a synthetic compound containing at least three thiol groups.
18048. The method of item 17879 wherein the polymer is formed from reactants comprising a synthetic compound containing at least four thiol groups.
18049. The method of item 17879 wherein the polymer is formed from reactants comprising a synthetic amino-containing compound.
18050. The method of item 17879 wherein the polymer is formed from reactants comprising a synthetic compound containing at least two amino groups.
18051. The method of item 17879 wherein the polymer is formed from reactants comprising a synthetic compound containing at least three amino groups.
18052. The method of item 17879 wherein the polymer is formed from reactants comprising a synthetic compound containing at least four amino groups.
18053. The method of item 17879 wherein the polymer is formed from reactants comprising a synthetic compound comprising a carbonyl-oxygen-succinimidyl group. 18054. The method of item 17879 wherein the polymer is formed from reactants comprising a synthetic compound comprising at least two carbonyl-oxygen-succinimidyl groups.
18055. The method of item 17879 wherein the polymer is formed from reactants comprising a synthetic compound comprising at least three carbonyl-oxygen-succinimidyl groups.
18056. The method of item 17879 wherein the polymer is formed from reactants comprising a synthetic compound comprising at least four carbonyl-oxygen-succinimidyl groups.
18057. The method of item 17879 wherein the polymer is formed from reactants comprising a synthetic polyalkylene oxide- containing compound.
18058. The method of item 17879 wherein the polymer is formed from reactants comprising a synthetic compound comprising both polyalkylene oxide and biodegradable polyester blocks.
18059. The method of item 17879 wherein the polymer is formed from reactants comprising a synthetic polyalkylene oxide- containing compound having reactive amino groups.
18060. The method of item 17879 wherein the polymer is formed from reactants comprising a synthetic polyalkylene oxide- containing compound having reactive thiol groups.
18061. The method of item 17879 wherein the polymer is formed from reactants comprising a synthetic polyalkylene oxide- containing compound having reactive carbonyl-oxygen-succinimidyl groups. 18062. The method of item 17879 wherein the polymer is formed from reactants comprising a synthetic compound comprising a biodegradable polyester block.
18063. The method of item 17879 wherein the polymer is formed from reactants comprising a synthetic polymer formed in whole or part from lactic acid or lactide.
18064. The method of item 17879 wherein the polymer is formed from reactants comprising a synthetic polymer formed in whole or part from glycolic acid or glycolide.
18065. The method of item 17879 wherein the polymer is formed from reactants comprising polylysine.
18066. The method of item 17879 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound comprising a polyalkylene oxide portion.
18067. The method of item 17879 wherein the polymer is formed from reactants comprising (a) protein and (b) polylysine.
18068. The method of item 17879 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound having at least four thiol groups.
18069. The method of item 17879 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound having at least four amino groups. 18070. The method of item 17879 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound having at least four carbonyl-oxygen-succinimide groups.
18071. The method of item 17879 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound having a biodegradable region formed from reactants selected from lactic acid, lactide, glycolic acid, glycolide, and epison-caprolactone.
18072. The method of item 17879 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound comprising a polyalkylene oxide portion.
18073. The method of item 17879 wherein the polymer is formed from reactants comprising (a) collagen and (b) polylysine.
18074. The method of item 17879 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound having at least four thiol groups.
18075. The method of item 17879 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound having at least four amino groups.
18076. The method of item 17879 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound having at least four carbonyl-oxygen-succinimide groups.
18077. The method of item 17879 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound having a biodegradable region formed from reactants selected from lactic acid, lactide, glycolic acid, glycolide, and epison-caprolactone.
18078. The method of item 17879 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound comprising a polyalkylene oxide portion.
18079. The method of item 17879 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) polylysine.
18080. The method of item 17879 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound having at least four thiol groups.
18081. The method of item 17879 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound having at least four amino groups.
18082. The method of item 17879 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound having at least four carbonyl-oxygen-succinimide groups.
18083. The method of item 17879 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound having a biodegradable region formed from reactants selected from lactic acid, lactide, glycolic acid, glycolide, and epison-caprolactone.
18084. The method of item 17879 wherein the polymer is formed from reactants comprising hyaluronic acid. 18085. The method of item 17879 wherein the polymer is formed from reactants comprising a hyaluronic acid derivative.
18086. The method of item 17879 wherein the polymer is formed from reactants comprising pentaerythritol poly(ethylene glycol)ether tetra-sulfhydryl of number average molecular weight between 3,000 and 30,000.
18087. The method of item 17879 wherein the polymer is formed from reactants comprising pentaerythritol poly(ethylene glycol)ether tetra-amino of number average molecular weight between 3,000 and 30,000.
18088. The method of item 17879 wherein the polymer is formed from reactants comprising (a) a synthetic compound having a number average molecular weight between 3,000 and 30,000 and comprising a polyalkylene oxide region and multiple nucleophilic groups, and (b) a synthetic compound having a number average molecular weight between 3,000 and 30,000 and comprising a polyalkylene oxide region and multiple electrophilic groups.
18089. The method of item 17879 wherein the composition comprises a colorant.
18090. The method of item 17879 wherein the composition is sterile.
18091. A method as in any one of items 17879-18090, wherein the device is an esophageal stent. 18092. A method as in any one of items 17879-18090, wherein the device is a biliary stent.
18093. A method as in any one of items 17879-18090, wherein the device is a colonic stent.
18094. A method as in any one of items 17879-18090, wherein the device is a pancreatic stent.
18095. A method for implanting a medical device comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with i) an anti-fibrotic agent, ii) an anti-infective agent, iii) a polymer, iv) a composition comprising an anti-fibrotic agent and a polymer, v) a composition comprising an anti-infective agent and a polymer, or vi) a composition comprising an anti-fibrotic agent, an anti- infective agent and a polymer, and (b) implanting the medical device into the host, wherein the medical device is a tracheal and bronchial stent.
18096. The method of item 18095 wherein the anti- fibrotic agent is an adensosine A2A receptor antagonist.
18097. The method of item 18095 wherein the anti- fibrotic agent is an AKT inhibitor.
18098. The method of item 18095 wherein the anti- fibrotic agent is an alpha 2 integrin antagonist, wherein the alpha 2 integrin antagonist is Pharmaprojects No. 5754 (Merck KgaA).
18099. The method of item 18095 wherein the anti- fibrotic agent is an alpha 4 integrin antagonist. 18100. The method of item 18095 wherein the anti- fibrotic agent is an alpha 7 nicotinic receptor agonist.
18101. The method of item 18095 wherein the anti- fibrotic agent is an angiogenesis inhibitor selected from the group consisting of AG-12,958 (Pfizer), ATN-161 (Attention LLC), neovastat, an angiogenesis inhibitor from Jerina AG (Germany), NM-3 (Mercian), VGA-1155 (Taisho), FCE-26644 (Pfizer), FCE-26950 (Pfizer), FPMA (Meiji Daries), FR-111142 (Fujisawa), GGTI-298, GM-1306 (Ligand), GPA-1734 (Novartis), NNC-47- 0011 (Novo Nordisk), herbamycin (Nippon Kayaku), lenalidomide (Celegene), IP-10 (NIH), ABT-828 (Abbott), KIN-841 (Tokushima University, Japan), SF-1126 (Semafore Pharmaceuticals), laminin technology (NIH), CHIR-258 (Chiron), NVP-AEW541 (Novartis), NVP-AEW541 (Novartis), Vt16907 (Alchemia), OXI-8007 (Oxigene), EG-3306 (Ark Therapeutics), Maspin (Arriva), ABT-567 (Abbott), PPI-2458 (Praecis Pharmaceuticals), CC-5079, CC-4089 (Celgene), HIF-1 alpha inhibitors (Xenova), S-247 (Pfizer), AP-23573 (Ariad), AZD-9935 (Astra Zeneca), mebendazole (Introgen Therapeutics), MetAP-2 inhibitors (GlaxoSmithKline), AG-615 (Angiogene Pharmaceuticals), Tie-2 antagonists (Hybrigenics), NC-381 , CYC-381, NC-169, NC-219, NC-383, NC-384, NC-407 (Lorus Therapeutics), ATN-224 (Attenuon), ON-01370 (Onconova), Vitronectin antagonists (Amgen), SDX-103 (Salmedix), Vitronectin antagonists (Shire), CHP (Riemser), TEK (Amgen), Anecortave acetate (Alcon), T46.2 (Matrix Therapeutics), HG-2 (Heptagen), TEM antagonists (Genzyme), Oxi-4500 (Oxigene), ATN-161 (Attenuon), WX-293 (Wilex), M-2025 (Metris Therapeutics), Alphastatin (BioActa), YH-16 (Yantai Rongchang), BIBF- 1120 (Boehringer Ingelheim), BAY-57-9352 (Bayer), AS-1404 (Cancer Research Technology), SC-77964 (Pfizer), glycomimetics (BioTie Therapies), TIE-2 Inhibitors (Ontogen), DIMI, Octamer (Octamer), ABR- 215050 (Active Biotech), ABT-518 (Abbott), KDR inhibitors (Abbott), BSF- 466895 (Abbott), SCH-221153 (Schering-Plough), DAC:antiangiogenic (ConjuChem), TFPI (EntreMed), AZD-2171 (Astra-Zenaca), CDC-394 (Celgene), LY290293 (EIi Lilly), IDN-5390 (Indena), Kdr Kinase Inhibitors (Merck), CT-113020, CT-116433, CT-116563, CT-31890, CT-32228) (Cell Therapeutics), A-299620 (Abbott), TWEAK Inhibitor (Amgen), VEGF modulators (Johnson and Johnson), Tum-N53, tumstatin (Genzyme), Thios- 1 , Thios-2 (Thios Pharmaceuticals), MV-6401 (Miravant Medical Technologies), Spisulosine (PharmaMar), CEP-7055 (Cephalon), AUV-201 (Auvation), LM-609 (EIi Lilly), SKF-106615 (AnorMED), Oglufanide disodium (Cytran), BW-114 (Pharminox), Calreticulin (NIH), WX-678 (Wilex), SD-7784 (Pfizer), WX-UK1 (Wilex), SH-268 (Schering AG), 2-Me-PGA (Celgene), S- 137 (Pfizer), ZD-6126 (Angiogene Pharmaceuticals), SG-292 (SignalGen), Benefin (Lane Labs), A6, A36 (Angstrom), SB-2723005 (GlaxoSmithKline), SC-7 (Cell Therapeutics), ZEN-014 (AEterna Zentaris), 2-methoxyestradiol (EntreMed), NK-130119 (Nippon Kayaku), CC-10004 (Celgene), AVE- 8062A (Ajinomoto), Tacedinaline (Pfizer), Actinonin (Tokyo Metropolitan Institute of Medical Science), Lenalidomide (Celgene), VGA-1155, BTO-956 (SRI International), ER-68203-00 (Eisai), CT-6685 (UCB), JKC-362 (Phoenix Pharmaceuticals), DMI-3798 (DMI Biosciences, Angiomate (Ipsen), ZD-6474 (AstraZeneca), CEP-5214 (Cephalon), Canstatin (Genzyme), NM-3 (Mercian), Oridigm (MediQuest Therapeutics), Exherin (Adherex), BLS-0597 (Boston Life Sciences), PTC-299 (PTC Therapeutics), NPI-2358 (Nereus Pharmaceuticals), CGP-79787 (Novartis), AEE-788 (Novartis), CKD-732 (Chong Kun Dang), CP-564959 (OSI Pharmaceuticals), CM-101 (CarboMed), CT-2584, CT3501 (Cell Therapeutics), combretastatin and analogues and derivatives thereof (Oxigene), Rebimastat (Bristol-Meyers Squibb), Dextrin 2-sulfate (ML Laboratories), Cilengitide (Merk KGaA), NSC-706704 (Pharminox), KRN- 951 (Kirin Brewery), Ukrain, NSC-631570 (Nowicky Pharma), Tecogalan sodium (Daiichi Pharmaceutical), Tz-93 (Tsumura), TBC-1635 (Encysive Pharmaceuticals), TAN-1120 (Takeda), Semaxanib (Pfizer), BDI-7800 (Biopharmacopae), SD-186, SD-983 (Bristol-Meyers Squibb), SB-223245 (GlaxoSmithKline), SC-236 (Pfizer), RWJ-590973 (Johnson and Johnson), ILX-1850 (Genzyme), SC-68488, S-836 (Pfizer), CG-55069-11 (CuraGen), Ki-23057 (Kirin Brewery), CCX-700 (Chemoentryx), Pegaptanib octasodium (Giled Sciences), ANGIOCOL (available from Biostratum Inc.), or an analogue or derivative thereof.
18102. The method of item 18095 wherein the anti- fibrotic agent is an apoptosis antagonist.
18103. The method of item 18095 wherein the anti- fibrotic agent is an apoptosis activator.
18104. The method of item 18095 wherein the anti- fibrotic agent is a beta 1 integrin antagonist.
18105. The method of item 18095 wherein the anti- fibrotic agent is a beta tubulin inhibitor.
18106. The method of item 18095 wherein the anti- fibrotic agent is a blocker of enzyme production in Hepatitis C.
18107. The method of item 18095 wherein the anti- fibrotic agent is a Bruton's tyrosine kinase inhibitor.
18108. The method of item 18095 wherein the anti- fibrotic agent is a calcineurin inhibitor.
18109. The method of item 18095 wherein the anti- fibrotic agent is a caspase 3 inhibitor. 18110. The method of item 18095 wherein the anti- fibrotic agent is a CC chemokine receptor antagonist.
18111. The method of item 18095 wherein the anti- fibrotic agent is a cell cycle inhibitor selected from the group consisting of SNS-595 (Sunesis), synthadotin, KRX-0403, and an analogue or derivative thereof.
18112. The method of item 18095 wherein the anti- fibrotic agent is a cathepsin B inhibitor.
18113. The method of item 18095 wherein the anti- fibrotic agent is a cathepsin K inhibitor, wherein the cathepsin K inhibitor is 462795 (GlaxoSmithKline), INPL-022-D6 (Amura Therapeutics), or an analogue or derivative thereof.
18114. The method of item 18095 wherein the anti- fibrotic agent is a cathepsin L inhibitor.
18115. The method of item 18095 wherein the anti- fibrotic agent is a CD40 antagonist.
18116. The method of item 18095 wherein the anti- fibrotic agent is a chemokine receptor agonist.
18117. The method of item 18095 wherein the anti- fibrotic agent is a chymase inhibitor.
18118. The method of item 18095 wherein the anti- fibrotic agent is a collagenase antagonist. 18119. The method of item 18095 wherein the anti- fibrotic agent is a CXCR antagonist.
18120. The method of item 18095 wherein the anti- fibrotic agent is a cyclin dependent kinase inhibitor selected from the group consisting of a CDK-1 inhibitor, a CDK-2 inhibitor, a CDK- 4 inhibitor, a CDK-6 inhibitor, a CAK1 inhibitor from GPC Biotech and Bristol-Myers Squibb, RGB-286199 (GPC Biotech), an anticancer agent from Astex Technology, a CAK1 inhibitor from GPC Biotech, a CDK inhibitor from Sanofi-Aventis, a CDK1/CDK2 inhibitor from Amgen, a CDK2 inhibitor from SUGEN-2 (Pfizer), a hearing loss therapy agent (Sound Pharmaceuticals), PD-0332991 (Pfizer), RGB-286199 (GPC Biotech), Ro-0505124 (Hoffmann- La Roche), a Ser/Thr kinase inhibitor from Lilly (EIi Lilly), CVT-2584 (CAS No. 199986-75-9) (CV Therapeutics), and an analogue or derivative thereof.
18121. The method of item 18095 wherein the anti- fibrotic agent is a cyclooxygenase 1 inhibitor.
18122. The method of item 18095 wherein the anti- fibrotic agent is a DHFR inhibitor.
18123. The method of item 18095 wherein the anti- fibrotic agent is a dual integrin inhibitor.
18124. The method of item 18095 wherein the anti- fibrotic agent is an elastase inhibitor.
18125. The method of item 18095 wherein the anti- fibrotic agent is an elongation factor- 1 alpha inhibitor. 18126. The method of item 18095 wherein the anti- fibrotic agent is an endothelial growth factor antagonist.
18127. The method of item 18095 wherein the anti- fibrotic agent is an endothelial growth factor receptor kinase inhibitor selected from the group consisting of sorafenib tosylate (Bayer), AAL-993 (Novartis), ABP-309 (Novartis), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EXEL- 2880 (Exelixis), GW-654652 (GlaxoSmithKline), a KDR inhibitor from LG Life Sciences, CT-6685 and CT-6729 (UCB), KRN-633 and KRN-951 (Kirin Brewery), OSI-930 (OSI Pharmaceuticals), SP-5.2 (Supratek Pharma), SU-
11657 (Pfizer), a Tie-2 antagonist (Hybrigenics), a VEGFR-2 kinase inhibitor (Bristol-Myers Squibb), XL-647 (Exelixis), a KDR inhibitor from Abbott Laboratories, sorafenib tosylate, and an analogue or derivative thereof.
18128. The method of item 18095 wherein the anti- fibrotic agent is an endotoxin antagonist.
18129. The method of item 18095 wherein the anti- fibrotic agent is an epothilone and tubulin binder.
18130. The method of item 18095 wherein the anti- fibrotic agent is an estrogen receptor antagonist.
18131. The method of item 18095 wherein the anti- fibrotic agent is an FGF inhibitor.
18132. The method of item 18095 wherein the anti- fibrotic agent is a farnexyl transferase inhibitor. 18133. The method of item 18095 wherein the anti- fibrotic agent is farnesyltransferase inhibitor selected from the group of A- 197574 (Abbott), a farnesyltransferase inhibitor from Servier, FPTIII (Strathclyde Institute for Drug R), LB-42908 (LG Life Sciences), Pharmaprojects No. 5063 (Genzyme), Pharmaprojects No. 5597 (Ipsen), Yissum Project No. B-1055 (Yissum), and an analogue or derivative thereof.
18134. The method of item 18095 wherein the anti- fibrotic agent is an FLT-3 kinase inhibitor.
18135. The method of item 18095 wherein the anti- fibrotic agent is an FGF receptor kinase inhibitor.
18136. The method of item 18095 wherein the anti- fibrotic agent is a fibrinogen antagonist selected from the group consisting of AUV-201 (Auvation), MG-13926 (Sanofi-Aventis), plasminogen activator (CAS No. 105913-11-9) (from Sanofi-Aventis or UCB), plasminogen activator-2 (tPA-2) (Sanofi-Aventis), pro-urokinase (CAS No. 82657-92-9) (Sanofi-Aventis), and an analogue or derivative thereof.
18137. The method of item 18095 wherein the anti- fibrotic agent is a heat shock protein 90 antagonist selected from the group consisting of SRN-005 (Sirenade), geldanamycin, NSC-33050 (17- allylaminogeldanamycin; 17-AAG), 17-dimethylaminoethylamino-17- demethoxy-geldanamycin (17-DMAG), rifabutin (rifamycin XIV, 1',4- didehydro-1-deoxy-1 ,4-dihydro-5'-(2-methylpropyl)-1-oxo-), and an analogue or derivative thereof.
18138. The method of item 18095 wherein the anti- fibrotic agent is a histone deacetylase inhibitor. 18139. The method of item 18095 wherein the anti- fibrotic agent is an HMGCoA reductase inhibitor selected from the group consisting of an atherosclerosis therapeutic from Lipid Sciences, ATI-16000 (ARYx Therapeutics), KS-01-019 (Kos Pharmaceuticals), Pharmaprojects No. 2197 (Sanofi-Aventi), RP 61969 (Sanofi-Aventis), and an analogue or derivative thereof.
18140. The method of item 18095 wherein the anti- fibrotic agent is an ICAM inhibitor.
18141. The method of item 18095 wherein the anti- fibrotic agent is an IL, ICE and IRAK antagonist, wherein the antagonist is a CJ-14877, CP-424174 (Pfizer), NF-61 (Negma-Lerads), and an analogue or derivative thereof.
18142. The method of item 18095 wherein the anti- fibrotic agent is an IL-2 inhibitor.
18143. The method of item 18095 wherein the anti- fibrotic agent is an immunosuppressant selected from the group consisting of teriflunomide (Sanofi Aventis), chlorsulfaquinoxalone (NSC-339004), chlorsulfaquinoxalone sulfate, CS-712 (Sankyo), ismomultin alfa (CAS No. 457913-93-8) (Akzo Nobel), antiallergics from GenPat77, antiinflammatories or AT-005 (Androclus Therapeutics), autoimmune disease therapeutics from EpiVax, BN-007 (Bone), budesonide (CAS No. 51333-22- 3) (MAP Pharmaceuticals), CO-14 (Genzyme), edratide (CAS No. 433922- 67-9) (Teva), EP-314 (Enanta), eprovafen (CAS No. 101335-99-3) (Sanofi- Aventis), HWA-131 (CAS No. 118788-41-3) (Sanofi-Aventis), immunomodulators from MerLion Pharmaceuticals, immunosuppressives from Alchemia, IPL-12 (Inflazyme), MDL-9563 (CAS No. 27086-86-8) (Sanofi-Aventis), Pharmaprojects No. 2330 (Sanofi-Aventis), Pharmaprojects No. 6426 (Abgenix), PXS-25 (Pharmaxis), rosmarinic acid (CAS No. 20283-92-5) (Sanofi-Aventis), RP 42927 or RP 54745 (CAS No. 135330-08-4) (Sanofi-Aventis), SGN-35 (Seattle Genetics), ST-1959 (Sigma-Tau), type I diabetes therapy from SYNX Pharma, UNIL-88 (Debiopharm), VP-025 (Vasogen), VR-694 (Vectura), PRTX-001 (Protalex), and an analogue or derivative thereof.
18144. The method of item 18095 wherein the anti- fibrotic agent is an IMPDH (inosine monophosphate).
18145. The method of item 18095 wherein the anti- fibrotic agent is an integrin antagonist.
18146. The method of item 18095 wherein the anti- fibrotic agent is an interleukin antagonist.
18147. The method of item 18095 wherein the anti- fibrotic agent is an inhibitor of type III receptor tyrosine kinase.
18148. The method of item 18095 wherein the anti- fibrotic agent is an irreversible inhibitor of enzyme methionine aminopeptidase type 2.
18149. The method of item 18095 wherein the anti- fibrotic agent is an isozyme selective delta protein kinase C inhibitor.
18150. The method of item 18095 wherein the anti- fibrotic agent a JAK3 enzyme inhibitor.
18151. The method of item 18095 wherein the anti- fibrotic agent is a JNK inhibitor. 18152. The method of item 18095 wherein the anti- fibrotic agent is a kinase inhibitor.
18153. The method of item 18095 wherein the anti- fibrotic agent is kinesin antagonist.
18154. The method of item 18095 wherein the anti- fibrotic agent is a kinesin antagonist.
18155. The method of item 18095 wherein the anti- fibrotic agent is a leukotriene inhibitor and antagonist selected from the group consisting of ambicromil (CAS No. 58805-38-2) (Sanofi-Aventis), amelubant (CAS No. 346735-24-8) (Boehringer Ingelheim), DW-1141 (Dong Wha), ebselen (Daiichi Pharmaceutical), ibudilast (Kyorin), leucotriene inhibitors from Sanofi-Aventis, lymphotoxin-beta receptor (LT-D) from Biogen Idee, Pharmaprojects No. 1535 and 2728 (CAS No. 119340-33-9) (Sanofi-Aventis), R-112 (Rigel), Rev-5367 (CAS No. 92532-05-3) (Sanofi- Aventis), RG-14893 (CAS No. 141835-49-6) (Sanofi-Aventis), RG-5901-A (CAS No. 101910-24-1), 92532-23-5, RP 66153 (CAS No. 142422-79-5), RP 66364 (CAS No.186916-92-5), RP 69698 (CAS No. 141748-00-7) (Sanofi-Aventis), SC-411930 (Pfizer), SC-41930 (CAS No. 120072-59-5) (Pfizer), SC-50605 (CAS No. 138828-39-4) (Pfizer), SC-51146 (CAS No. 141059-52-1), SC-53228 (CAS No. 153633-01-3) (Pfizer), spaglumic acid (ZY-15106) (CAS No.3106-85-2), 80619-64-3 (Novartis), tipredane (CAS No. 85197-77-9) (Bristol-Myers Squibb), U-75302 (CAS No. 119477-85-9) (Pfizer), and analogue or derivative thereof.
18156. The method of item 18095 wherein the anti- fibrotic agent is an MAP kinase inhibitor. 18157. The method of item 18095 wherein the anti- fibrotic agent is a matrix metalloproteinase inhibitor.
18158. The method of item 18095 wherein the anti- fibrotic agent is an MCP-CCR2 inhibitor.
18159. The method of item 18095 wherein the anti- fibrotic agent is an mTOR inhibitor.
18160. The method of item 18095 wherein the anti- fibrotic agent is an mTOR kinase inhibitor.
18161. The method of item 18095 wherein the anti- fibrotic agent is a microtubule inhibitor selected from the group consisting of antibody-maytansinoid conjugates from Biogen Idee, colchicines (MantiCore Pharmaceuticals), anticancer immunoconjugates from Johnson & Johnson, DIME from Octamer, gni-1f (GNI), huC242-DM4, huMy9-6-DM1 (ImmunoGen), IDN-5404 (Indena), IMO-098, IMOderm (Imotep), mebendazole (Introgen Therapeutics), microtubule poisons from Cambridge Enterprise, paclitaxel such as LOTAX from Aphios (CAS No. 33069-62-4), Genexol-PM from Samyang, Pharmaprojects No. 6383 (Tapestry Pharmaceuticals), RPR-112378 (Sanofi-Aventis), SGN-75 (Seattle Genetics), SPL-7435 (Starpharma), SSR-250411 (Sanofi-Aventis), trastuzumab-DM1 (Genentech), vinorelbine, and an analogue or derivative thereof.
18162. The method of item 18095 wherein the anti- fibrotic agent is an MlF inhibitor.
18163. The method of item 18095 wherein the anti- fibrotic agent is an MMP inhibitor. 18164. The method of item 18095 wherein the anti- fibrotic agent is a neurokinin (NK) antagonist selected from the group consisting of anthrotainin (CAS No. 148084-40-6) (Sanofi-Aventis), an IBS thereapeutic from ArQuIe, MDL-105212A (CAS No. 167261-60-1) (Ssanofi- Aventis), Pharmaprojects No. 2744, 3258 (CAS No. 139167-47-8) 4006, 4201 , or 5986 (Sanofi-Aventis), RP 67580 (CAS No. 135911-02-3), SR- 144190 (CAS No. 201152-86-5), SSR-240600, SSR-241586 (Sanofi- Aventis), TKA-457 (Novartis), vestipitant mesylate (CAS No. 334476-64-1) (GlaxoSmithKline), Win-64821 (Sanofi-Aventis), PRX-96026 (Predix Pharmaceuticals), and an analogue or derivative thereof.
18165. The method of item 18095 wherein the anti- fibrotic agent is an NF kappa B inhibitor selected from the group consisting of AVE-0545 or AVE-0547 (Sanofi-Aventis), bortezomib (CAS No. 179324- 69-7) (Millennium Pharmaceuticals), dexanabinol (CAS No. 112924-45-5) (Pharmos), dexlipotam (Viatris), Pharmaprojects No. 6283 (INDRA) (OXiGENE), IPL-576092 (CAS No. 137571-30-3) (Inflazyme), NFKB decoy (Corgentech), NFKB decoy oligo (AnGes MG), NFKB's from Ariad, osteoporosis treatments or S5 (F005) from Fulcrum Pharmaceuticals, P61 (Phytopharm), R-flurbiprofen (CAS No. 5104-49-4) (Encore Pharmaceuticals), and an analogue or derivative thereof.
18166. The method of item 18095 wherein the anti- fibrotic agent is a nitric oxide agonist.
18167. The method of item 18095 wherein the anti- fibrotic agent is an ornithine decarboxylase inhibitor.
18168. The method of item 18095 wherein the anti- fibrotic agent is a p38 MAP kinase inhibitor selected from the group consisting of AZD-6703 (AstraZeneca), JX-401 (Jexys Pharmaceuticals), BMS-2 (Bristol-Myers Squibb), a p38 MAP kinase inhibitor from Novartis, a p38-alpha MAP kinase inhibitor from Amphora, Pharmaprojects No. 5704 (Pharmacopeia), RPR-200765A (Sanofi-Aventis), SD-282 (Johnson & Johnson), TAK-715 (Takeda), and an analogue or derivative thereof.
18169. The method of item 18095 wherein the anti- fibrotic agent is a palmitoyl-protein thioesterase inhibitor.
18170. The method of item 18095 wherein the anti- fibrotic agent is a PDGF receptor kinase inhibitor selected from the group consisting of AAL-993, AMN-107, or ABP-309 (Novartis), AMG-706 (Amgen), BAY-57-9352 (Bayer), CDP-860 (UCB), E-7080 (Eisai), imatinib (CAS No. 152459-95-5) (Novartis), OSI-930 (OSi Pharmaceuticals), RPR- 127963E (Sanofi-Aventis), RWJ-540973 (Johnson & Johnson), sorafenib tosylate (Bayer), SU-11657 (Pfizer), tandutinib (CAS No. 387867-13-2) (Millennium Pharmaceuticals), vatalanib (Novartis), ZK-CDK (Schering AG), and an analogue or derivative thereof.
18171. The method of item 18095 wherein the anti- fibrotic agent is (-)-halofenate (Metabolex), AMG-131 (Amgen), antidiabetics from Japan Tobacco, AZD-4619, AZD-8450, AZD-8677 (AstraZeneca), DRF-10945, balaglitazone (Dr Reddy's), CS-00088, CS-00098 (Chipscreen Biosciences), E-3030 (Eisai), etalocib (CAS No. 161172-51-6) (EIi Lilly), GSK-641597 (Ligand), GSK-677954 (GlaxoSmithKline), GW-409544 (Ligand), GW-590735 (GlaxoSmithKline), K-111 (Hoffmann-La Roche), LY- 518674 (EIi Lilly), LY-674 (Ligand), LY-929 (Ligand), MC-3001 , MC-3002 (MaxoCore Pharmaceuticals), metformin HCI + pioglitazone (CAS No. 1115- 70-4 and 112529-15-4), ACTOPLUS MET from Andrx), muraglitazar (CAS No. 331741-94-7) (Bristol-Myers Squibb), naveglitazar (Ligand), oleoylethanolamide (Kadmus Pharmaceuticals), ONO-5129, pioglitazone hydrochloride (CAS No. 111025-46-8 and 112529-15-4) (Takeda), PLX-204 (Plexxikon), PPAR agonists from Genfit, PPAR delta agonists from EIi Lilly, PPAR-alpha agonists from CrystalGenomics, PPAR-gamma modulators and PPAR-β modulators from CareX, rosiglitazone maleate (CAS No. 122324- 73-4 or 155141-29-0) (GlaxoSmithKline), rosiglitazone maleate/glimepir (CAS No. 155141-29-0 and 93479-97-1), AVANDARYL1 rosiglitazone maleate/metformin extend (CAS No. 155141-29-0 and 657-24-9), AVANDAMET, rosiglitazone maleate+metformin, AVANDAMET (GlaxoSmithKline), tesaglitazar (AstraZeneca), and an analogue or derivative thereof.
18172. The method of item 18095 wherein the anti- fibrotic agent is a phosphatase inhibitor.
18173. The method of item 18095 wherein the anti- fibrotic agent is a phosphodiesterase (PDE) inhibitor selected from the group consisting of avanafil (Tanabe Seiyaku), dasantafil (CAS No. 569351- 91-3) (Schering-Plough), A-906119 (CAS No. 134072-58-5), DL-850 (Sanofi-Aventis), GRC-3015, GRC-3566, GRC-3886 (Glenmark), HWA-153 (CAS No. 56395-66-5) (Sanofi-Aventis), hydroxypumafentrine (Altana), IBFB-130011 , IBFB-14-016, IBFB-140301 , IBFB-150007, IBFB-211913 (IBFB Pharma), L-826141 (Merck & Co), medorinone (CAS No. 88296-61-1) (Sanofi-Aventis), MEM-1917 (Memory Pharmaceuticals), ND-1251 (Neuro3d), PDE inhibitors from ICOS, PDE IV inhibitors from Memory Pharmaceuticals and CrystalGenomics, Pharmaprojects No. 2742 and 6141 (Sanofi-Aventis), QAD-171 (Novartis), RHC-2963 (CAS No. 76993-12-9 and 76993-14-1), RPR-117658, RPR-122818 derivatives, SR-24870 , and RPR- 132294 (Sanofi-Aventis), SK-350 (ln2Gen), stroke therapy agents from deCODE Genetics, TAS-203 (Taiho), tofimilast (CAS No. 185954-27-2) (Pfizer), UK-371800 (Pfizer), WIN-65579 (CAS No. 158020-82-7) (Sanofi- Aventis), IBFB-130020 (IBFB Pharma), OPC-6535 (CAS No. 145739-56-6) (Otsuka), a phosphodiesterase III inhibitor, enoximone, a phosphodiesterase IV Inhibitor, fosfosal, Atopik (Barrier Therapeutics), triflusal, a phosphodiesterase V inhibitor, and an analogue or derivative thereof.
18174. The method of item 18095 wherein the anti- fibrotic agent is a PKC inhibitor.
18175. The method of item 18095 wherein the anti- fibrotic agent is a platelet activating factor antagonist.
18176. The method of item 18095 wherein the anti- fibrotic agent is a platelet-derived growth factor receptor kinase inhibitor.
18177. The method of item 18095 wherein the anti- fibrotic agent is a prolyl hydroxylase inhibitor.
18178. The method of item 18095 wherein the anti- fibrotic agent is a polymorphonuclear neutrophil inhibitor.
18179. The method of item 18095 wherein the anti- fibrotic agent is a protein kinase B inhibitor.
18180. The method of item 18095 wherein the anti- fibrotic agent is a protein kinase C stimulant.
18181. The method of item 18095 wherein the anti- fibrotic agent is a purine nucleoside analogue.
18182. The method of item 18095 wherein the anti- fibrotic agent is a purinoreceptor P2X antagonist. 18183. The method of item 18095 wherein the anti- fibrotic agent is a Raf kinase inhibitor.
18184. The method of item 18095 wherein the anti- fibrotic agent is a reversible inhibitor of ErbB1 and ErbB2.
18185. The method of item 18095 wherein the anti- fibrotic agent is a ribonucleoside triphosphate reductase inhibitor.
18186. The method of item 18095 wherein the anti- fibrotic agent is an SDF-1 antagonist.
18187. The method of item 18095 wherein the anti- fibrotic agent is a sheddase inhibitor.
18188. The method of item 18095 wherein the anti- fibrotic agent is an SRC inhibitor.
18189. The method of item 18095 wherein the anti- fibrotic agent is a stromelysin inhibitor.
18190. The method of item 18095 wherein the anti- fibrotic agent is an Syk kinase inhibitor.
18191. The method of item 18095 wherein the anti- fibrotic agent is a telomerase inhibitor.
18192. The method of item 18095 wherein the anti- fibrotic agent is a TGF beta inhibitor selected from the group consisting of pirfenidone (CAS No. 53179-13-8) (MARNAC), tranilast (CAS No. 53902- 12-8) (Kissei), IN-1130 (ln2Gen), mannose-6-phosphate (BTG), TGF-β antagonists from Inflazyme (Pharmaprojects No. 6075), TGF-β antagonists from Sydney, non-industrial source), TGF-β I receptor kinase inhibitors from EIi Lilly, TGF-β receptor inhibitors from Johnson & Johnson, and an analogue or derivative thereof.
18193. The method of item 18095 wherein the anti- fibrotic agent is a TNFα antagonist or TACE inhibitor selected from the group consisting of adalimumab (CAS No. 331731-18-1) (Cambridge Antibody Technology), AGIX-4207 (AtheroGenics), AGT-1 (Advanced Biotherapy), an anti-inflammatory from Borean Pharma, Cellzome, or Paradigm Therapeutics, anti-inflammatory vaccine (TNF-alpha kinoid) from Neovacs, humanized anti-TNF antibody or an anti-TNF MAb (CB0006) Celltech (UCB), apratastat (CAS No. 287445-51-0) (Wyeth), BMS-561392 (Bristol-Myers Squibb), BN-006 (Bone), certolizumab pegol (CAS No. 428863-50-7 or CH-138 (UCB), cilomilast (CAS No. 153259-65-5) (GlaxoSmithKline), CR-1 (Nuada Pharmaceuticals), CRx-119 (CombinatoRx), D-5410 (UCB), dacopafant (CAS No. 125372-33-0) (Sanofi- Aventis), dersalazine (CAS No. 188913-57-7/188913-58-8) (Uriach), etanercept (CAS No. 185243-69-0) (Amgen), ethyl pyruvate (Critical (Critical Therapeutics), golimumab (CAS No. 476181-74-5) (Johnson & Johnson), hormono-immunotherapy from Ipsen, CDP571 (e.g., Humicade from UCB), IC-485 (ICOS), infliximab (CAS No. 170277-31-3) (Johnson & Johnson), IP- 751 (Manhattan Pharmaceuticals), ISIS-104838 (CAS No. 250755-32-9) (ISIS Pharmaceuticals), lenalidomide (CAS No. 191732-72-6) (Celgene), lentinan (CAS No. 37339-90-5) (Ajinomoto), MDL-201112 (CAS No. 142130-73-2) (Sanofi-Aventis), medroxyprogesterone (CAS No. 520-85-4) (InKine Pharmaceutical), N-acetylcysteine (CAS No. 616-91-1) (Zambon), NBE-P2 (DIREVO Biotech), nerelimomab (CAS No. 162774-06-3) (Chiron), OM-294DP (OM PHARMA), onercept (CAS No. 199685-57-9) (Yeda), PASSTNF-alpha (Verigen), pentoxifylline or oxypentifylline (Sanofi-Aventis), Pharmaprojects No. 4091, 4241, 4295, or 4488 (Sanofi-Aventis), Pharmaprojects No. 5480 (Amgen), Pharmaprojects No. 6749 (Cengent), pirfenidone (CAS No. 53179-13-8) (MARNAC), RPFM 32294 (Sanofi- Aventis), S5 (F002) (Fulcrum Pharmaceuticals), simvastatin (CAS No. 79902-63-9) (Merck & Co), STA-6292 (Synta Pharmaceuticals), tacrolimus (CAS No. 104987-11-3) (Fujisawa LifeCycle Pharma), talactoferrin alfa (CAS No. 308240-58-6) (Agennix), thalidomide (CAS No. 50-35-1) (Celgene), TNF antagonists form ProStrakan, and Synergen, TNF inhibitors (Amgen), TNF-alpha antagonists from Dynavax Technologies and Jerina AG (Germany), TNF-alpha inhibitors from IBFB Pharma and Xencor (Xencor), torbafylline (CAS No. 105102-21-4) (Sanofi-Aventis), UR-1505 (Uriach), VT-346 (Viron Therapeutics), YSIL6 (Y1S Therapeutics), YSTH2 (Y's Therapeutics), NPI-1302a-3 (Nereus Pharmaceuticals, a TNF antagonist from Jerina AG (Germany), dersalazine, and an analogue or derivative thereof.
18194. The method of item 18095 wherein the anti- fibrotic agent is a Toll receptor inhibitor.
18195. The method of item 18095 wherein the anti- fibrotic agent is a tubulin antagonist.
18196. The method of item 18095 wherein the anti- fibrotic agent is a tyrosine kinase inhibitor selected from the group consisting of SU-011248, SUTENT from Pfizer Inc. (New York, NY), BMS-354825, PN- 355 (Paracelsian Pharmaceuticals), AGN-199659 (Allergan), AAL-993 or ABP-309 (Novartis), adaphostin (NIH), AEE-788 (Novartis), AG-013736 (OSI Pharmaceuticals), AG-13736 (Pfizer), ALT-110 (Alteris Therapeutics), AMG-706 (Amgen), anticancer MAbs from Xencor, anti-EGFrvlll MAbs from Abgenix, anti-HER2 MAb from Abiogen, AZD-2171 or AZD-9935 (AstraZeneca), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), CEP-5214 (Cephalon), CEP-7055 (Cephalon), cetuximab (ImClone Systems), CHIR-200131 and CHIR-258 (Chiron), CP-547632 (OSI Pharmaceuticals), CP-724714 (Pfizer), CT-301 (Creabilis Therapeutics), D- 69491 (Baxter International), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EGFR/ErbB2 inhibitors from Array BioPharma, erlotinib (CAS No. 183319- 69-9) (OSI Pharmaceuticals), EXEL-2880 (Exelixis), FK-778 (Sanofi- Aventis) , gefitinib (CAS No. 184475-35-2) (AstraZeneca), GW-2286 or GW- 654652 (GlaxoSmithKline), her2/neu antigen from AlphaVax, HER-2/neu inhibitor from Generex, Herzyme (Medipad) (Sirna Therapeutics), HKI-272 (Wyeth), HuMax-EGFr (Genmab), idronoxil (CAS No. 81267-65-4) (Novogen), IGF- 1 inhibitors from Ontogen, IMC-11 F8 (ImClone Systems), kahalalide F (CAS No. 149204-42-2) (PharmaMar), KDR inhibitor from LG Life Sciences, KDR inhibitors from Abbott Laboratories, KDR kinase inhibitors (UCB), Kdr kinase inhibitors from Merck & Co, KRN-633 and KRN- 951 (Kirin Brewery), KSB-102 (Xenova), lapatinib ditosylate (CAS No. 388082-78-8) (GlaxoSmithKline), matuzumab (Merck KGaA), MDX-214 (Medarex), ME-103 (Pharmexa), MED-A300 (Gerolymatos), MNAC-13 (Lay Line Genomics), nimotuzumab (Center of Molecular Immunology), NSC- 330507 or NSC-707545 (NIH), NV-50 (Novogen), OSI-930 (OSI Pharmaceuticals), panitumumab (Abgenix), pelitinib (CAS No. 287933-82-7) (Wyeth), pertuzumab (CAS No. 380610-27-5) (Genentech), Pharmaprojects No. 3985 (Sanofi-Aventis), prostate cancer therapeutics from Sequenom (SQPC35, SQPC36, SQPC90), removab and remoxab (Trion Pharma), RG- 13022 (CAS No. 136831-48-6), RG-13291 (CAS No. 138989-50-1), or RG- 14620 (CAS No. 136831-49-7) (Sanofi-Aventis), RM-6427 (Romark), RNAi breast cancer therapy from Benitec, RP 53801 (CAS No. 125882-88-4) (Sanofi-Aventis), sorafenib tosylate (Bayer), SU-11657 (Pfizer), Tie-2 antagonists from Semaia (Hybrigenics), Tie-2 inhibitors from Ontogen, trastuzumab (CAS No. 180288-69-1) (Genentech), tyrosine kinase inhibitors from Sanofi-Aventis, U3-1287, U3-1565, U3-1784, U3-1800 (U3 Pharma), vatalanib (Novartis), VEGFR-2 kinase inhibitor from Bristol-Myers Squibb, XL-647 (Exelixis), ZD-6474 (AstraZeneca), ZK-CDK (Schering AG), an EGFR tyrosine kinase inhibitor, EKB-569 (Wyeth), and an analogue or derivative thereof.
18197. The method of item 18095 wherein the anti- fibrotic agent is a VEGF inhibitor.
18198. The method of item 18095 wherein the anti- fibrotic agent is a vitamin D receptor agonist.
18199. The method of item 18095 wherein the anti- fibrotic agent is ZD-6474 (an angiogenesis inhibitor).
18200. The method of item 18095 wherein the anti- fibrotic agent is AP-23573 (an mTOR inhibitor).
18201. The method of item 18095 wherein the anti- fibrotic agent is synthadotin (a tubulin antagonist).
18202. The method of item 18095 wherein the anti- fibrotic agent is S-0885 (a collagenase inhibitor).
18203. The method of item 18095 wherein the anti- fibrotic agent is aplidine (an elongation factor-1 alpha inhibitor).
18204. The method of item 18095 wherein the anti- fibrotic agent is ixabepilone (an epithilone).
18205. The method of item 18095 wherein the anti- fibrotic agent is IDN-5390 (an angiogenesis inhibitor). 18206. The method of item 18095 wherein the anti- fibrotic agent is SB-2723005 (an angiogenesis inhibitor).
18207. The method of item 18095 wherein the anti- fibrotic agent is ABT-518 (an angiogenesis inhibitor).
18208. The method of item 18095 wherein the anti- fibrotic agent is combretastatin (an angiogenesis inhibitor).
18209. The method of item 18095 wherein the anti- fibrotic agent is anecortave acetate (an angiogenesis inhibitor).
18210. The method of item 18095 wherein the anti- fibrotic agent is SB-715992 (a kinesin antagonist).
18211. The method of item 18095 wherein the anti- fibrotic agent is temsirolimus (an mTOR inhibitor).
18212. The method of item 18095 wherein the anti- fibrotic agent is adalimumab (a TNFα antagonist).
18213. The method of item 18095 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with an anti-fibrotic agent, and (b) implanting the medical device into the host.
18214. The method of item 18095 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with an anti-infective agent, and (b) implanting the medical device into the host. 18215. The method of item 18095 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with a polymer; and (b) implanting the medical device into the host.
18216. The method of item 18095 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with a composition comprising an anti-fibrotic agent and a polymer, and (b) implanting the medical device into the host.
18217. The method of item 18095 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with a composition comprising an anti-infective agent and a polymer, and (b) implanting the medical device into the host.
18218. The method of item 18095 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with a composition comprising an anti-fibrotic agent, an anti- infective agent and a polymer, and (b) implanting the medical device into the host.
18219. The method of item 18095 wherein the anti- infective agent is an anthracycline.
18220. The method of item 18095 wherein the anti- infective agent is doxorubicin.
18221. The method of item 18095 wherein the anti- infective agent is is mitoxantrone. 18222. The method of item 18095 wherein the anti- infective agent is a fluoropyrimidine.
18223. The method of item 18095 wherein the anti- infective agent is 5-fluorouracil (5-FU).
18224. The method of item 18095 wherein the anti- infective agent is a folic acid antagonist.
18225. The method of item 18095 wherein the anti- infective agent is methotrexate.
18226. The method of item 18095 wherein the anti- infective agent is a podophylotoxin.
18227. The method of item 18095 wherein the anti- infective agent is etoposide.
18228. The method of item 18095 wherein the anti- infective agent is camptothecin.
18229. The method of item 18095 wherein the anti- infective agent is a hydroxyurea.
18230. The method of item 18095 wherein the anti- infective agent is a platinum complex.
18231. The method of item 18095 wherein the anti- infective agent is cisplatin. 18232. The method of item 18095 wherein the composition comprises an anti-thrombotic agent.
18233. The method of item 18095 wherein the polymer is formed from reactants comprising a naturally occurring polymer.
18234. The method of item 18095 wherein the polymer is formed from reactants comprising protein.
18235. The method of item 18095 wherein the polymer is formed from reactants comprising carbohydrate.
18236. The method of item 18095 wherein the polymer is formed from reactants comprising biodegradable polymer.
18237. The method of item 18095 wherein the polymer is formed from reactants comprising nonbiodegradable polymer.
18238. The method of item 18095 wherein the polymer is formed from reactants comprising collagen.
18239. The method of item 18095 wherein the polymer is formed from reactants comprising methylated collagen.
18240. The method of item 18095 wherein the polymer is formed from reactants comprising fibrinogen.
18241. The method of item 18095 wherein the polymer is formed from reactants comprising thrombin. 18242. The method of item 18095 wherein the polymer reactants comprising blood plasma.
18243. The method of item 18095 wherein the polymer reactants comprising calcium salt.
18244. The method of item 18095 wherein the polymer reactants comprising an antifibronolytic agent.
18245. The method of item 18095 wherein the polymer reactants comprising fibrinogen analog.
18246. The method of item 18095 wherein the polymer reactants comprising albumin.
18247. The method of item 18095 wherein the polymer reactants comprising plasminogen.
18248. The method of item 18095 wherein the polymer reactants comprising von Willebrands factor.
18249. The method of item 18095 wherein the polymer reactants comprising Factor VIII.
18250. The method of item 18095 wherein the polymer reactants comprising hypoallergenic collagen.
18251. The method of item 18095 wherein the polymer reactants comprising atelopeptidic collagen. 18252. The method of item 18095 wherein the polymer is formed from reactants comprising telopeptide collagen.
18253. The method of item 18095 wherein the polymer is formed from reactants comprising crosslinked collagen.
18254. The method of item 18095 wherein the polymer is formed from reactants comprising aprotinin.
18255. The method of item 18095 wherein the polymer is formed from reactants comprising epsilon-amino-n-caproic acid.
18256. The method of item 18095 wherein the polymer is formed from reactants comprising gelatin.
18257. The method of item 18095 wherein the polymer is formed from reactants comprising protein conjugates.
18258. The method of item 18095 wherein the polymer is formed from reactants comprising gelatin conjugates.
18259. The method of item 18095 wherein the polymer is formed from reactants comprising a synthetic polymer.
18260. The method of item 18095 wherein the polymer is formed from reactants comprising a synthetic isocyanate-containing compound.
18261. The method of item 18095 wherein the polymer is formed from reactants comprising a synthetic thiol-containing compound. 18262. The method of item 18095 wherein the polymer is formed from reactants comprising a synthetic compound containing at least two thiol groups.
18263. The method of item 18095 wherein the polymer is formed from reactants comprising a synthetic compound containing at least three thiol groups.
18264. The method of item 18095 wherein the polymer is formed from reactants comprising a synthetic compound containing at least four thiol groups.
18265. The method of item 18095 wherein the polymer is formed from reactants comprising a synthetic amino-containing compound.
18266. The method of item 18095 wherein the polymer is formed from reactants comprising a synthetic compound containing at least two amino groups.
18267. The method of item 18095 wherein the polymer is formed from reactants comprising a synthetic compound containing at least three amino groups.
18268. The method of item 18095 wherein the polymer is formed from reactants comprising a synthetic compound containing at least four amino groups.
18269. The method of item 18095 wherein the polymer is formed from reactants comprising a synthetic compound comprising a carbonyl-oxygen-succinimidyl group. 18270. The method of item 18095 wherein the polymer is formed from reactants comprising a synthetic compound comprising at least two carbonyl-oxygen-succinimidyl groups.
18271. The method of item 18095 wherein the polymer is formed from reactants comprising a synthetic compound comprising at least three carbonyl-oxygen-succinimidyl groups.
18272. The method of item 18095 wherein the polymer is formed from reactants comprising a synthetic compound comprising at least four carbonyl-oxygen-succinimidyl groups.
18273. The method of item 18095 wherein the polymer is formed from reactants comprising a synthetic polyalkylene oxide- containing compound.
18274. The method of item 18095 wherein the polymer is formed from reactants comprising a synthetic compound comprising both polyalkylene oxide and biodegradable polyester blocks.
18275. The method of item 18095 wherein the polymer is formed from reactants comprising a synthetic polyalkylene oxide- containing compound having reactive amino groups.
18276. The method of item 18095 wherein the polymer is formed from reactants comprising a synthetic polyalkylene oxide- containing compound having reactive thiol groups.
18277. The method of item 18095 wherein the polymer is formed from reactants comprising a synthetic polyalkylene oxide- containing compound having reactive carbonyl-oxygen-succinimidyl groups. 18278. The method of item 18095 wherein the polymer is formed from reactants comprising a synthetic compound comprising a biodegradable polyester block.
18279. The method of item 18095 wherein the polymer is formed from reactants comprising a synthetic polymer formed in whole or part from lactic acid or lactide.
18280. The method of item 18095 wherein the polymer is formed from reactants comprising a synthetic polymer formed in whole or part from glycolic acid or glycolide.
18281. The method of item 18095 wherein the polymer is formed from reactants comprising polylysine.
18282. The method of item 18095 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound comprising a polyalkylene oxide portion.
18283. The method of item 18095 wherein the polymer is formed from reactants comprising (a) protein and (b) polylysine.
18284. The method of item 18095 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound having at least four thiol groups.
18285. The method of item 18095 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound having at least four amino groups. 18286. The method of item 18095 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound having at least four carbonyl-oxygen-succinimide groups.
18287. The method of item 18095 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound having a biodegradable region formed from reactants selected from lactic acid, lactide, glycolic acid, glycolide, and epison-caprolactone.
18288. The method of item 18095 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound comprising a polyalkylene oxide portion.
18289. The method of item 18095 wherein the polymer is formed from reactants comprising (a) collagen and (b) polylysine.
18290. The method of item 18095 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound having at least four thiol groups.
18291. The method of item 18095 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound having at least four amino groups.
18292. The method of item 18095 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound having at least four carbonyl-oxygen-succinimide groups.
18293. The method of item 18095 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound having a biodegradable region formed from reactants selected from lactic acid, lactide, glycolic acid, glycolide, and epison-caprolactone.
18294. The method of item 18095 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound comprising a polyalkylene oxide portion.
18295. The method of item 18095 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) polylysine.
18296. The method of item 18095 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound having at least four thiol groups.
18297. The method of item 18095 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound having at least four amino groups.
18298. The method of item 18095 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound having at least four carbonyl-oxygen-succinimide groups.
18299. The method of item 18095 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound having a biodegradable region formed from reactants selected from lactic acid, lactide, glycolic acid, glycolide, and epison-caprolactone.
18300. The method of item 18095 wherein the polymer is formed from reactants comprising hyaluronic acid. 18301. The method of item 18095 wherein the polymer is formed from reactants comprising a hyaluronic acid derivative.
18302. The method of item 18095 wherein the polymer is formed from reactants comprising pentaerythritol poly(ethylene glycol)ether tetra-sulfhydryl of number average molecular weight between 3,000 and 30,000.
18303. The method of item 18095 wherein the polymer is formed from reactants comprising pentaerythritol poly(ethylene glycol)ether tetra-amino of number average molecular weight between 3,000 and 30,000.
18304. The method of item 18095 wherein the polymer is formed from reactants comprising (a) a synthetic compound having a number average molecular weight between 3,000 and 30,000 and comprising a polyalkylene oxide region and multiple nucleophilic groups, and (b) a synthetic compound having a number average molecular weight between 3,000 and 30,000 and comprising a polyaikylene oxide region and multiple electrophilic groups.
18305. The method of item 18095 wherein the composition comprises a colorant.
18306. The method of item 18095 wherein the composition is sterile.
18307. A method as in any one of items 18095-18306, wherein the device is a tracheal stent. 18308. A method as in any one of items 18095-18306, wherein the device is a bronchial stent.
18309. A method as in any one of items 18095-18306, wherein the device is a metallic tracheal stent.
18310. A method as in any one of items 18095-18306, wherein the device is a metallic bronchial stent.
18311. A method as in any one of items 18095-18306, wherein the device is a polymeric tracheal stent.
18312. A method as in any one of items 18095-18306, wherein the device is a polymeric bronchial stent.
18313. A method for implanting a medical device comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with i) an anti-fibrotic agent, ii) an anti-infective agent, iii) a polymer, iv) a composition comprising an anti-fibrotic agent and a polymer, v) a composition comprising an anti-infective agent and a polymer, or vi) a composition comprising an anti-fibrotic agent, an anti- infective agent and a polymer, and (b) implanting the medical device into the host, wherein the medical device is a genital urinary stent.
18314. The method of item 18313 wherein the anti- fibrotic agent is an adensosine A2A receptor antagonist.
18315. The method of item 18313 wherein the anti- fibrotic agent is an AKT inhibitor. 18316. The method of item 18313 wherein the anti- fibrotic agent is an alpha 2 integrin antagonist, wherein the alpha 2 integrin antagonist is Pharmaprojects No. 5754 (Merck KgaA).
18317. The method of item 18313 wherein the anti- fibrotic agent is an alpha 4 integrin antagonist.
18318. The method of item 18313 wherein the anti- fibrotic agent is an alpha 7 nicotinic receptor agonist.
18319. The method of item 18313 wherein the anti- fibrotic agent is an angiogenesis inhibitor selected from the group consisting of AG-12,958 (Pfizer), ATN-161 (Attention LLC), neovastat, an angiogenesis inhibitor from Jerina AG (Germany), NM-3 (Mercian), VGA-1155 (Taisho), FCE-26644 (Pfizer), FCE-26950 (Pfizer), FPMA (Meiji Daries), FR-111142 (Fujisawa), GGTI-298, GM-1306 (Ligand), GPA-1734 (Novartis), NNC-47- 0011 (Novo Nordisk), herbamycin (Nippon Kayaku), lenalidomide (Celegene), IP-10 (NIH), ABT-828 (Abbott), K1N-841 (Tokushima University, Japan), SF-1126 (Semafore Pharmaceuticals), laminin technology (NIH), CHIR-258 (Chiron), NVP-AEW541 (Novartis), NVP-AEW541 (Novartis), Vt16907 (Alchemia), OXI-8007 (Oxigene), EG-3306 (Ark Therapeutics), Maspin (Arriva), ABT-567 (Abbott), PPI-2458 (Praecis Pharmaceuticals), CC-5079, CC-4089 (Celgene), HIF-1 alpha inhibitors (Xenova), S-247 (Pfizer), AP-23573 (Ariad), AZD-9935 (Astra Zeneca), mebendazole (Introgen Therapeutics), MetAP-2 inhibitors (GlaxoSmithKline), AG-615 (Angiogene Pharmaceuticals), Tie-2 antagonists (Hybrigenics), NC-381, CYC-381, NC-169, NC-219, NC-383, NC-384, NC-407 (Lorus Therapeutics), ATN-224 (Attenuon), ON-01370 (Onconova), Vitronectin antagonists (Amgen), SDX-103 (Salmedix), Vitronectin antagonists (Shire), CHP (Riemser), TEK (Amgen), Anecortave acetate (Alcon), T46.2 (Matrix Therapeutics), HG-2 (Heptagen), TEM antagonists (Genzyme), Oxi-4500 (Oxigene), ATN-161 (Attention), WX-293 (Wilex), M-2025 (Metris Therapeutics), Alphastatin (BioActa), YH-16 (Yantai Rongchang), BIBF- 1120 (Boehringer Ingelheim), BAY-57-9352 (Bayer), AS-1404 (Cancer Research Technology), SC-77964 (Pfizer), glycomimetics (BioTie Therapies), TIE-2 Inhibitors (Ontogen), DIMI, Octamer (Octamer), ABR- 215050 (Active Biotech), ABT-518 (Abbott), KDR inhibitors (Abbott), BSF- 466895 (Abbott), SCH-221153 (Schering-Plough), DAC:antiangiogenic (ConjuChem), TFPI (EntreMed), AZD-2171 (Astra-Zenaca), CDC-394 (Celgene), LY290293 (EIi Lilly), IDN-5390 (Indena), Kdr Kinase Inhibitors (Merck), CT-113020, CT-116433, CT-116563, CT-31890, CT-32228) (Cell Therapeutics), A-299620 (Abbott), TWEAK Inhibitor (Amgen), VEGF modulators (Johnson and Johnson), Tum-N53, tumstatin (Genzyme), Thios- 1 , Thios-2 (Thios Pharmaceuticals), MV-6401 (Miravant Medical Technologies), Spisulosine (PharmaMar), CEP-7055 (Cephalon), AUV-201 (Auvation), LM-609 (EIi Lilly), SKF-106615 (AnorMED), Oglufanide disodium (Cytran), BW-114 (Pharminox), Calreticulin (NIH), WX-678 (Wilex), SD-7784 (Pfizer), WX-UK1 (Wilex), SH-268 (Schering AG), 2-Me-PGA (Celgene), S- 137 (Pfizer), ZD-6126 (Angiogene Pharmaceuticals), SG-292 (SignalGen), Benefin (Lane Labs), A6, A36 (Angstrom), SB-2723005 (GlaxoSmithKline), SC-7 (Cell Therapeutics), ZEN-014 (AEterna Zentaris), 2-methoxyestradiol (EntreMed), NK-130119 (Nippon Kayaku), CC-10004 (Celgene), AVE- 8062A (Ajinomoto), Tacedinaline (Pfizer), Actinonin (Tokyo Metropolitan Institute of Medical Science), Lenalidomide (Celgene), VGA-1155, BTO-956 (SRI International), ER-68203-00 (Eisai), CT-6685 (UCB), JKC-362 (Phoenix Pharmaceuticals), DMI-3798 (DMl Biosciences, Angiomate (Ipsen), ZD-6474 (AstraZeneca), CEP-5214 (Cephalon), Canstatin (Genzyme), NM-3 (Mercian), Oridigm (MediQuest Therapeutics), Exherin (Adherex), BLS-0597 (Boston Life Sciences), PTC-299 (PTC Therapeutics), NPI-2358 (Nereus Pharmaceuticals), CGP-79787 (Novartis), AEE-788 (Novartis), CKD-732 (Chong Kun Dang), CP-564959 (OSI Pharmaceuticals), CM-101 (CarboMed), CT-2584, CT3501 (Cell Therapeutics), combretastatin and analogues and derivatives thereof (Oxigene), Rebimastat (Bristol-Meyers Squibb), Dextrin 2-sulfate (ML Laboratories), Cilengitide (Merk KGaA), NSC-706704 (Pharminox), KRN- 951 (Kirin Brewery), Ukrain, NSC-631570 (Nowicky Pharma), Tecogalan sodium (Daiichi Pharmaceutical), Tz-93 (Tsumura), TBC-1635 (Encysive Pharmaceuticals), TAN-1120 (Takeda), Semaxanib (Pfizer), BDI-7800 (Biopharmacopae), SD-186, SD-983 (Bristol-Meyers Squibb), SB-223245 (GlaxoSmithKline), SC-236 (Pfizer), RWJ-590973 (Johnson and Johnson), ILX-1850 (Genzyme), SC-68488, S-836 (Pfizer), CG-55069-11 (CuraGen), Ki-23057 (Kirin Brewery), CCX-700 (Chemoentryx), Pegaptanib octasodium (Giled Sciences), ANGIOCOL (available from Biostratum Inc.), or an analogue or derivative thereof.
18320. The method of item 18313 wherein the anti- fibrotic agent is an apoptosis antagonist.
18321. The method of item 18313 wherein the anti- fibrotic agent is an apoptosis activator.
18322. The method of item 18313 wherein the anti- fibrotic agent is a beta 1 integrin antagonist.
18323. The method of item 18313 wherein the anti- fibrotic agent is a beta tubulin inhibitor.
18324. The method of item 18313 wherein the anti- fibrotic agent is a blocker of enzyme production in Hepatitis C.
18325. The method of item 18313 wherein the anti- fibrotic agent is a Bruton's tyrosine kinase inhibitor. 18326. The method of item 18313 wherein the anti- fibrotic agent is a calcineurin inhibitor.
18327. The method of item 18313 wherein the anti- fibrotic agent is a caspase 3 inhibitor.
18328. The method of item 18313 wherein the anti- fibrotic agent is a CC chemokine receptor antagonist.
18329. The method of item 18313 wherein the anti- fibrotic agent is a ceil cycle inhibitor selected from the group consisting of SNS-595 (Sunesis), synthadotin, KRX-0403, and an analogue or derivative thereof.
18330. The method of item 18313 wherein the anti- fibrotic agent is a cathepsin B inhibitor.
18331. The method of item 18313 wherein the anti- fibrotic agent is a cathepsin K inhibitor, wherein the cathepsin K inhibitor is 462795 (GlaxoSmithKline), INPL-022-D6 (Amura Therapeutics), or an analogue or derivative thereof.
18332. The method of item 18313 wherein the anti- fibrotic agent is a cathepsin L inhibitor.
18333. The method of item 18313 wherein the anti- fibrotic agent is a CD40 antagonist.
18334. The method of item 18313 wherein the anti- fibrotic agent is a chemokine receptor agonist. 18335. The method of item 18313 wherein the anti- fibrotic agent is a chymase inhibitor.
18336. The method of item 18313 wherein the anti- fibrotic agent is a collagenase antagonist.
18337. The method of item 18313 wherein the anti- fibrotic agent is a CXCR antagonist.
18338. The method of item 18313 wherein the anti- fibrotic agent is a cyclin dependent kinase inhibitor selected from the group consisting of a CDK-1 inhibitor, a CDK-2 inhibitor, a CDK- 4 inhibitor, a CDK-6 inhibitor, a CAK1 inhibitor from GPC Biotech and Bristol-Myers Squibb, RGB-286199 (GPC Biotech), an anticancer agent from Astex Technology, a CAK1 inhibitor from GPC Biotech, a CDK inhibitor from Sanofi-Aventis, a CDK1/CDK2 inhibitor from Amgen, a CDK2 inhibitor from SUGEN-2 (Pfizer), a hearing loss therapy agent (Sound Pharmaceuticals), PD-0332991 (Pfizer), RGB-286199 (GPC Biotech), Ro-0505124 (Hoffmann- La Roche), a Ser/Thr kinase inhibitor from Lilly (EIi Lilly), CVT-2584 (CAS No. 199986-75-9) (CV Therapeutics), and an analogue or derivative thereof.
18339. The method of item 18313 wherein the anti- fibrotic agent is a cyclooxygenase 1 inhibitor.
18340. The method of item 18313 wherein the anti- fibrotic agent is a DHFR inhibitor.
18341. The method of item 18313 wherein the anti- fibrotic agent is a dual integrin inhibitor. 18342. The method of item 18313 wherein the anti- fibrotic agent is an elastase inhibitor.
18343. The method of item 18313 wherein the anti- fibrotic agent is an elongation factor-1 alpha inhibitor.
18344. The method of item 18313 wherein the anti- fibrotic agent is an endothelial growth factor antagonist.
18345. The method of item 18313 wherein the anti- fibrotic agent is an endothelial growth factor receptor kinase inhibitor selected from the group consisting of sorafenib tosylate (Bayer), AAL-993 (Novartis), ABP-309 (Novartis), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EXEL- 2880 (Exelixis), GW-654652 (GlaxoSmithKline), a KDR inhibitor from LG Life Sciences, CT-6685 and CT-6729 (UCB), KRN-633 and KRN-951 (Kirin Brewery), OSI-930 (OSI Pharmaceuticals), SP-5.2 (Supratek Pharma), SU- 11657 (Pfizer), a Tie-2 antagonist (Hybrigenics), a VEGFR-2 kinase inhibitor (Bristol-Myers Squibb), XL-647 (Exelixis), a KDR inhibitor from Abbott Laboratories, sorafenib tosylate, and an analogue or derivative thereof.
18346. The method of item 18313 wherein the anti- fibrotic agent is an endotoxin antagonist.
18347. The method of item 18313 wherein the anti- fibrotic agent is an epothilone and tubulin binder.
18348. The method of item 18313 wherein the anti- fibrotic agent is an estrogen receptor antagonist. 18349. The method of item 18313 wherein the anti- fibrotic agent is an FGF inhibitor.
18350. The method of item 18313 wherein the anti- fibrotic agent is a farnexyl transferase inhibitor.
18351. The method of item 18313 wherein the anti- fibrotic agent is farnesyltransferase inhibitor selected from the group of A- 197574 (Abbott), a farnesyltransferase inhibitor from Servier, FPTIII (Strathclyde Institute for Drug R), LB-42908 (LG Life Sciences), Pharmap rejects No. 5063 (Genzyme), Pharmaprojects No. 5597 (Ipsen), Yissum Project No. B-1055 (Yissum), and an analogue or derivative thereof.
18352. The method of item 18313 wherein the anti- fibrotic agent is an FLT-3 kinase inhibitor.
18353. The method of item 18313 wherein the anti- fibrotic agent is an FGF receptor kinase inhibitor.
18354. The method of item 18313 wherein the anti- fibrotic agent is a fibrinogen antagonist selected from the group consisting of AUV-201 (Auvation), MG-13926 (Sanofi-Aventis), plasminogen activator (CAS No. 105913-11-9) (from Sanofi-Aventis or UCB), plasminogen activator-2 (tPA-2) (Sanofi-Aventis), pro-urokinase (CAS No. 82657-92-9) (Sanofi-Aventis), and an analogue or derivative thereof.
18355. The method of item 18313 wherein the anti- fibrotic agent is a heat shock protein 90 antagonist selected from the group consisting of SRN-005 (Sirenade), geldanamycin, NSC-33050 (17- aliylamin'ogeldanamycin; 17-AAG), 17-dimethylaminoethylamino-17- demethoxy-geldanamycin (17-DMAG), rifabutin (rifamycin XIV, 1',4- didehydro-1-deoxy-1,4-dihydro-5'-(2-methylpropyl)-1-oxo-), and an analogue or derivative thereof.
18356. The method of item 18313 wherein the anti- fibrotic agent is a histone deacetylase inhibitor.
18357. The method of item 18313 wherein the anti- fibrotic agent is an HMGCoA reductase inhibitor selected from the group consisting of an atherosclerosis therapeutic from Lipid Sciences, ATI-16000 (ARYx Therapeutics), KS-01-019 (Kos Pharmaceuticals), Pharmaprojects No. 2197 (Sanofi-Aventi), RP 61969 (Sanofi-Aventis), and an analogue or derivative thereof.
18358. The method of item 18313 wherein the anti- fibrotic agent is an ICAM inhibitor.
18359. The method of item 18313 wherein the anti- fibrotic agent is an IL, ICE and IRAK antagonist, wherein the antagonist is a CJ-14877, CP-424174 (Pfizer), NF-61 (Negma-Lerads), and an analogue or derivative thereof.
18360. The method of item 18313 wherein the anti- fibrotic agent is an IL-2 inhibitor.
18361. The method of item 18313 wherein the anti- fibrotic agent is an immunosuppressant selected from the group consisting of teriflunomide (Sanofi Aventis), chlorsulfaquinoxalone (NSC-339004), chlorsulfaquinoxalone sulfate, CS-712 (Sankyo), ismomultin alfa (CAS No. 457913-93-8) (Akzo Nobel), antiallergics from GenPat77, antiinflammatories or AT-005 (Androclus Therapeutics), autoimmune disease therapeutics from EpiVax, BN-007 (Bone), budesonide (CAS No. 51333-22- 3) (MAP Pharmaceuticals), CO-14 (Genzyme), edratide (CAS No. 433922- 67-9) (Teva), EP-314 (Enanta), eprovafen (CAS No. 101335-99-3) (Sanofi- Aventis), HWA-131 (CAS No. 118788-41-3) (Sanofi-Aventis), immunomodulators from MerLion Pharmaceuticals, immunosuppressives from Alchemia, IPL-12 (Inflazyme), MDL-9563 (CAS No. 27086-86-8) (Sanofi-Aventis), Pharmaprojects No. 2330 (Sanofi-Aventis), Pharmaprojects No. 6426 (Abgenix), PXS-25 (Pharmaxis), rosmarinic acid (CAS No. 20283-92-5) (Sanofi-Aventis), RP 42927 or RP 54745 (CAS No. 135330-08-4) (Sanofi-Aventis), SGN-35 (Seattle Genetics), ST-1959 (Sigma-Tau), type I diabetes therapy from SYNX Pharma, UNIL-88 (Debiopharm), VP-025 (Vasogen), VR-694 (Vectura), PRTX-001 (Protalex), and an analogue or derivative thereof.
18362. The method of item 18313 wherein the anti- fibrotic agent is an IMPDH (inosine monophosphate).
18363. The method of item 18313 wherein the anti- fibrotic agent is an integrin antagonist.
18364. The method of item 18313 wherein the anti- fibrotic agent is an interleukin antagonist.
18365. The method of item 18313 wherein the anti- fibrotic agent is an inhibitor of type III receptor tyrosine kinase.
18366. The method of item 18313 wherein the anti- fibrotic agent is an irreversible inhibitor of enzyme methionine aminopeptidase type 2.
18367. The method of item 18313 wherein the anti- fibrotic agent is an isozyme selective delta protein kinase C inhibitor. 18368. The method of item 18313 wherein the anti- fibrotic agent a JAK3 enzyme inhibitor.
18369. The method of item 18313 wherein the anti- fibrotic agent is a JNK inhibitor.
18370. The method of item 18313 wherein the anti- fibrotic agent is a kinase inhibitor.
18371. The method of item 18313 wherein the anti- fibrotic agent is kinesin antagonist.
18372. The method of item 18313 wherein the anti- fibrotic agent is a kinesin antagonist.
18373. The method of item 18313 wherein the anti- fibrotic agent is a leukotriene inhibitor and antagonist selected from the group consisting of ambicromil (CAS No. 58805-38-2) (Sanofi-Aventis), amelubant (CAS No. 346735-24-8) (Boehringer Ingelheim), DW-1141 (Dong Wha), ebselen (Daiichi Pharmaceutical), ibudilast (Kyorin), leucotriene inhibitors from Sanofi-Aventis, lymphotoxin-beta receptor (LT-D) from Biogen Idee, Pharmaprojects No. 1535 and 2728 (CAS No. 119340-33-9) (Sanofi-Aventis), R-112 (Rigel), Rev-5367 (CAS No. 92532-05-3) (Sanofi- Aventis), RG-14893 (CAS No. 141835-49-6) (Sanofi-Aventis), RG-5901-A (CAS No. 101910-24-1), 92532-23-5, RP 66153 (CAS No. 142422-79-5), RP 66364 (CAS No.186916-92-5), RP 69698 (CAS No. 141748-00-7) (Sanofi-Aventis), SC-411930 (Pfizer), SC-41930 (CAS No. 120072-59-5) (Pfizer), SC-50605 (CAS No. 138828-39-4) (Pfizer), SC-51146 (CAS No. 141059-52-1), SC-53228 (CAS No. 153633-01-3) (Pfizer), spaglumic acid (ZY-15106) (CAS No.3106-85-2), 80619-64-3 (Novartis), tipredane (CAS No. 85197-77-9) (Bristol-Myers Squibb), U-75302 (CAS No. 119477-85-9) (Pfizer), and analogue or derivative thereof.
18374. The method of item 18313 wherein the anti- fibrotic agent is an MAP kinase inhibitor.
18375. The method of item 18313 wherein the anti- fibrotic agent is a matrix metalloproteinase inhibitor.
18376. The method of item 18313 wherein the anti- fibrotic agent is an MCP-CCR2 inhibitor.
18377. The method of item 18313 wherein the anti- fibrotic agent is an mTOR inhibitor.
18378. The method of item 18313 wherein the anti- fibrotic agent is an mTOR kinase inhibitor.
18379. The method of item 18313 wherein the anti- fibrotic agent is a microtubule inhibitor selected from the group consisting of antibody-maytansinoid conjugates from Biogen Idee, colchicines (MantiCore Pharmaceuticals), anticancer immunoconjugates from Johnson & Johnson, DIME from Octamer, gni-1f (GNI), huC242-DM4, huMy9-6-DM1 (ImmunoGen), IDN-5404 (Indena), IMO-098, IMOderm (Imotep), mebendazole (Introgen Therapeutics), microtubule poisons from Cambridge Enterprise, paclitaxel such as LOTAX from Aphios (CAS No. 33069-62-4), Genexol-PM from Samyang, Pharmaprojects No. 6383 (Tapestry Pharmaceuticals), RPR-112378 (Sanofi-Aventis), SGN-75 (Seattle Genetics), SPL-7435 (Starpharma), SSR-250411 (Sanofi-Aventis), trastuzumab-DM1 (Genentech), vinorelbine, and an analogue or derivative thereof. 18380. The method of item 18313 wherein the anti- fibrotic agent is an MIF inhibitor.
18381. The method of item 18313 wherein the anti- fibrotic agent is an MMP inhibitor.
18382. The method of item 18313 wherein the anti- fibrotic agent is a neurokinin (NK) antagonist selected from the group consisting of anthrotainin (CAS No. 148084-40-6) (Sanofi-Aventis), an IBS thereapeutic from ArQuIe, MDL-105212A (CAS No. 167261-60-1) (Ssanofi- Aventis), Pharmaprojects No. 2744, 3258 (CAS No. 139167-47-8) 4006, 4201 , or 5986 (Sanofi-Aventis), RP 67580 (CAS No. 135911-02-3), SR- 144190 (CAS No. 201152-86-5), SSR-240600, SSR-241586 (Sanofi- Aventis), TKA-457 (Novartis), vestipitant mesylate (CAS No. 334476-64-1) (GlaxoSmithKline), Win-64821 (Sanofi-Aventis), PRX-96026 (Predix Pharmaceuticals), and an analogue or derivative thereof.
18383. The method of item 18313 wherein the anti- fibrotic agent is an NF kappa B inhibitor selected from the group consisting of AVE-0545 or AVE-0547 (Sanofi-Aventis), bortezomib (CAS No. 179324- 69-7) (Millennium Pharmaceuticals), dexanabinol (CAS No. 112924-45-5) (Pharmos), dexlipotam (Viatris), Pharmaprojects No. 6283 (INDRA) (OXiGENE), IPL-576092 (CAS No. 137571-30-3) (Inflazyme), NFKB decoy (Corgentech), NFKB decoy oligo (AnGes MG), NFKB's from Ariad, osteoporosis treatments or S5 (F005) from Fulcrum Pharmaceuticals, P61 (Phytopharm), R-flurbiprofen (CAS No. 5104-49-4) (Encore Pharmaceuticals), and an analogue or derivative thereof.
18384. The method of item 18313 wherein the anti- fibrotic agent is a nitric oxide agonist. 18385. The method of item 18313 wherein the anti- fibrotic agent is an ornithine decarboxylase inhibitor.
18386. The method of item 18313 wherein the anti- fibrotic agent is a p38 MAP kinase inhibitor selected from the group consisting of AZD-6703 (AstraZeneca), JX-401 (Jexys Pharmaceuticals), BMS-2 (Bristol-Myers Squibb), a p38 MAP kinase inhibitor from Novartis, a p38-alpha MAP kinase inhibitor from Amphora, Pharmaprojects No. 5704 (Pharmacopeia), RPR-200765A (Sanofi-Aventis), SD-282 (Johnson & Johnson), TAK-715 (Takeda), and an analogue or derivative thereof.
18387. The method of item 18313 wherein the anti- fibrotic agent is a palmitoyl-protein thioesterase inhibitor.
18388. The method of item 18313 wherein the anti- fibrotic agent is a PDGF receptor kinase inhibitor selected from the group consisting of AAL-993, AMN-107, or ABP-309 (Novartis), AMG-706 (Amgen), BAY-57-9352 (Bayer), CDP-860 (UCB), E-7080 (Eisai), imatinib (CAS No. 152459-95-5) (Novartis), OSI-930 (OSI Pharmaceuticals), RPR- 127963E (Sanofi-Aventis), RWJ-540973 (Johnson & Johnson), sorafenib tosylate (Bayer), SU-11657 (Pfizer), tandutinib (CAS No. 387867-13-2) (Millennium Pharmaceuticals), vatalanib (Novartis), ZK-CDK (Schering AG), and an analogue or derivative thereof.
18389. The method of item 18313 wherein the anti- fibrotic agent is (-)-halofenate (Metabolex), AMG-131 (Amgen), antidiabetics from Japan Tobacco, AZD-4619, AZD-8450, AZD-8677 (AstraZeneca), DRF-10945, balaglitazone (Dr Reddy's), CS-00088, CS-00098 (Chipscreen Biosciences), E-3030 (Eisai), etalocib (CAS No. 161172-51-6) (EIi Lilly), GSK-641597 (Ligand), GSK-677954 (GlaxoSmithKline), GW-409544 (Ligand), GW-590735 (GlaxoSmithKline), K-111 (Hoffmann-La Roche), LY- 518674 (EIi Lilly), LY-674 (Ligand), LY-929 (Ligand), MC-3001 , MC-3002 (MaxoCore Pharmaceuticals), metformin HCI + pioglitazone (CAS No. 1115- 70-4 and 112529-15-4), ACTOPLUS MET from Andrx), muraglitazar (CAS No. 331741-94-7) (Bristol-Myers Squibb), naveglitazar (Ligand), oleoylethanolamide (Kadmus Pharmaceuticals), ONO-5129, pioglitazone hydrochloride (CAS No. 111025-46-8 and 112529-15-4) (Takeda), PLX-204 (Plexxikon), PPAR agonists from Genfit, PPAR delta agonists from EIi Lilly, PPAR-alpha agonists from CrystalGenomics, PPAR-gamma modulators and PPAR-β modulators from CareX, rosiglitazone maleate (CAS No. 122324- 73-4 or 155141-29-0) (GlaxoSmithKline), rosiglitazone maleate/glimepir (CAS No. 155141-29-0 and 93479-97-1), AVANDARYL, rosiglitazone maleate/metformin extend (CAS No. 155141-29-0 and 657-24-9), AVANDAMET, rosiglitazone maleate+metformin, AVANDAMET (GlaxoSmithKline), tesaglitazar (AstraZeneca), and an analogue or derivative thereof.
18390. The method of item 18313 wherein the anti- fibrotic agent is a phosphatase inhibitor.
18391. The method of item 18313 wherein the anti- fibrotic agent is a phosphodiesterase (PDE) inhibitor selected from the group consisting of avanafil (Tanabe Seiyaku), dasantafil (CAS No. 569351- 91-3) (Schering-Plough), A-906119 (CAS No. 134072-58-5), DL-850 (Sanofi-Aventis), GRC-3015, GRC-3566, GRC-3886 (Glenmark), HWA-153 (CAS No. 56395-66-5) (Sanofi-Aventis), hydroxypumafentrine (Altana), IBFB-130011 , IBFB-14-016, IBFB-140301 , IBFB-150007, IBFB-211913 (IBFB Pharma), L-826141 (Merck & Co), medorinone (CAS No. 88296-61-1) (Sanofi-Aventis), MEM-1917 (Memory Pharmaceuticals), ND-1251 (Neuro3d), PDE inhibitors from ICOS, PDE IV inhibitors from Memory Pharmaceuticals and CrystalGenomics, Pharmaprojects No. 2742 and 6141 (Sanofi-Aventis), QAD-171 (Novartis), RHC-2963 (CAS No. 76993-12-9 and 76993-14-1), RPR-117658, RPR-122818 derivatives, SR-24870 , and RPR- 132294 (Sanofi-Aventis), SK-350 (ln2Gen), stroke therapy agents from deCODE Genetics, TAS-203 (Taiho), tofimilast (CAS No. 185954-27-2) (Pfizer), UK-371800 (Pfizer), WIN-65579 (CAS No. 158020-82-7) (Sanofi- Aventis), IBFB-130020 (IBFB Pharma), OPC-6535 (CAS No. 145739-56-6) (Otsuka), a phosphodiesterase III inhibitor, enoximone, a phosphodiesterase IV inhibitor, fosfosal, Atopik (Barrier Therapeutics), triflusal, a phosphodiesterase V inhibitor, and an analogue or derivative thereof.
18392. The method of item 18313 wherein the anti- fibrotic agent is a PKC inhibitor.
18393. The method of item 18313 wherein the anti- fibrotic agent is a platelet activating factor antagonist.
18394. The method of item 18313 wherein the anti- fibrotic agent is a platelet-derived growth factor receptor kinase inhibitor.
18395. The method of item 18313 wherein the anti- fibrotic agent is a prolyl hydroxylase inhibitor.
18396. The method of item 18313 wherein the anti- fibrotic agent is a polymorphonuclear neutrophil inhibitor.
18397. The method of item 18313 wherein the anti- fibrotic agent is a protein kinase B inhibitor.
18398. The method of item 18313 wherein the anti- fibrotic agent is a protein kinase C stimulant. 18399. The method of item 18313 wherein the anti- fibrotic agent is a purine nucleoside analogue.
18400. The method of item 18313 wherein the anti- fibrotic agent is a purinoreceptor P2X antagonist.
18401. The method of item 18313 wherein the anti- fibrotic agent is a Raf kinase inhibitor.
18402. The method of item 18313 wherein the anti- fibrotic agent is a reversible inhibitor of ErbB1 and ErbB2.
18403. The method of item 18313 wherein the anti- fibrotic agent is a ribonucleoside triphosphate reductase inhibitor.
18404. The method of item 18313 wherein the anti- fibrotic agent is an SDF-1 antagonist.
18405. The method of item 18313 wherein the anti- fibrotic agent is a sheddase inhibitor.
18406. The method of item 18313 wherein the anti- fibrotic agent is an SRC inhibitor.
18407. The method of item 18313 wherein the anti- fibrotic agent is a stromelysin inhibitor.
18408. The method of item 18313 wherein the anti- fibrotic agent is an Syk kinase inhibitor. 18409. The method of item 18313 wherein the anti- fibrotic agent is a telomerase inhibitor.
18410. The method of item 18313 wherein the anti- fibrotic agent is a TGF beta inhibitor selected from the group consisting of pirfenidone (CAS No. 53179-13-8) (MARNAC), tranilast (CAS No. 53902- 12-8) (Kissei), IN-1130 (ln2Gen), mannose-6-phosphate (BTG), TGF-β antagonists from Inflazyme (Pharmaprojects No. 6075), TGF-β antagonists from Sydney, non-industrial source), TGF-β I receptor kinase inhibitors from EIi Lilly, TGF-β receptor inhibitors from Johnson & Johnson, and an analogue or derivative thereof.
18411. The method of item 18313 wherein the anti- fibrotic agent is a TNFα antagonist or TACE inhibitor selected from the group consisting of adalimumab (CAS No. 331731-18-1) (Cambridge Antibody Technology), AGIX-4207 (AtheroGenics), AGT-1 (Advanced Biotherapy), an anti-inflammatory from Borean Pharma, Cellzome, or Paradigm Therapeutics, anti-inflammatory vaccine (TNF-alpha kinoid) from Neovacs, humanized anti-TNF antibody or an anti-TNF MAb (CB0006) Celltech (UCB), apratastat (CAS No. 287445-51-0) (Wyeth), BMS-561392 (Bristol-Myers Squibb), BN-006 (Bone), certolizumab pegol (CAS No. 428863-50-7 or CH-138 (UCB), cilomilast (CAS No. 153259-65-5) (GlaxoSmithKline), CR-1 (Nuada Pharmaceuticals), CRx-119 (CombinatoRx), D-5410 (UCB), dacopafant (CAS No. 125372-33-0) (Sanofi- Aventis), dersalazine (CAS No. 188913-57-7/188913-58-8) (Uriach), etanercept (CAS No. 185243-69-0) (Amgen), ethyl pyruvate (Critical (Critical Therapeutics), golimumab (CAS No. 476181-74-5) (Johnson & Johnson), hormono-immunotherapy from Ipsen, CDP571 (e.g., Humicade from UCB), IC-485 (ICOS), infliximab (CAS No. 170277-31-3) (Johnson & Johnson), IP- 751 (Manhattan Pharmaceuticals), ISIS-104838 (CAS No. 250755-32-9) (ISIS Pharmaceuticals), lenalidomide (CAS No. 191732-72-6) (Celgene), lentinan (CAS No. 37339-90-5) (Ajinomoto), MDL-201112 (CAS No. 142130-73-2) (Sanofi-Aventis), medroxyprogesterone (CAS No. 520-85-4) (InKine Pharmaceutical), N-acetylcysteine (CAS No. 616-91-1) (Zambon), NBE-P2 (DIREVO Biotech), nerelimomab (CAS No. 162774-06-3) (Chiron), OM-294DP (OM PHARMA), onercept (CAS No. 199685-57-9) (Yeda), PASSTNF-alpha (Verigen), pentoxifylline or oxypentifylline (Sanofi-Aventis), Pharmaprojects No. 4091 , 4241 , 4295, or 4488 (Sanofi-Aventis), Pharmaprojects No. 5480 (Amgen), Pharmaprojects No. 6749 (Cengent), pirfenidone (CAS No. 53179-13-8) (MARNAC), RPR-132294 (Sanofi- Aventis), S5 (F002) (Fulcrum Pharmaceuticals), simvastatin (CAS No. 79902-63-9) (Merck & Co), STA-6292 (Synta Pharmaceuticals), tacrolimus (CAS No. 104987-11-3) (Fujisawa LifeCycle Pharma), talactoferrin alfa (CAS No. 308240-58-6) (Agennix), thalidomide (CAS No. 50-35-1) (Celgene), TNF antagonists form ProStrakan, and Synergen, TNF inhibitors (Amgen), TNF-alpha antagonists from Dynavax Technologies and Jerina AG (Germany), TNF-alpha inhibitors from IBFB Pharma and Xencor (Xencor), torbafylline (CAS No. 105102-21-4) (Sanofi-Aventis), UR-1505 (Uriach), VT-346 (Viron Therapeutics), YSIL6 (Y1S Therapeutics), YSTH2 (Y's Therapeutics), NPI-1302a-3 (Nereus Pharmaceuticals, a TNF antagonist from Jerina AG (Germany), dersalazine, and an analogue or derivative thereof.
18412. The method of item 18313 wherein the anti- fibrotic agent is a Toll receptor inhibitor.
18413. The method of item 18313 wherein the anti- fibrotic agent is a tubulin antagonist.
18414. The method of item 18313 wherein the anti- fibrotic agent is a tyrosine kinase inhibitor selected from the group consisting of SU-011248, SUTENT from Pfizer Inc. (New York, NY), BMS-354825, PN- 355 (Paracelsian Pharmaceuticals), AGN-199659 (Allergan), AAL-993 or ABP-309 (Novartis), adaphostin (NIH), AEE-788 (Novartis), AG-013736 (OSI Pharmaceuticals), AG-13736 (Pfizer), ALT-110 (Alteris Therapeutics), AMG-706 (Amgen), anticancer MAbs from Xencor, anti-EGFrvlll MAbs from Abgenix, anti-HER2 MAb from Abiogen, AZD-2171 or AZD-9935 (AstraZeneca), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), CEP-5214 (Cephalon), CEP-7055 (Cephalon), cetuximab (ImCIone Systems), CHIR-200131 and CHIR-258 (Chiron), CP-547632 (OSI Pharmaceuticals), CP-724714 (Pfizer), CT-301 (Creabilis Therapeutics), D- 69491 (Baxter International), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EGFR/ErbB2 inhibitors from Array BioPharma, erlotinib (CAS No. 183319- 69-9) (OSI Pharmaceuticals), EXEL-2880 (Exelixis), FK-778 (Sanofi- Aventis) , gefitinib (CAS No. 184475-35-2) (AstraZeneca), GW-2286 or GW- 654652 (GlaxoSmithKline), her2/neu antigen from AlphaVax, HER-2/neu inhibitor from Generex, Herzyme (Medipad) (Sirna Therapeutics), HKI-272 (Wyeth), HuMax-EGFr (Genmab), idronoxil (CAS No. 81267-65-4) (Novogen), IGF-1 inhibitors from Ontogen, IMC-11F8 (ImCIone Systems), kahalalide F (CAS No. 149204-42-2) (PharmaMar), KDR inhibitor from LG Life Sciences, KDR inhibitors from Abbott Laboratories, KDR kinase inhibitors (UCB), Kdr kinase inhibitors from Merck & Co, KRN-633 and KRN- 951 (Kirin Brewery), KSB-102 (Xenova), lapatinib ditosylate (CAS No. 388082-78-8) (GlaxoSmithKline), matuzumab (Merck KGaA), MDX-214 (Medarex), ME-103 (Pharmexa), MED-A300 (Gerolymatos), MNAC-13 (Lay Line Genomics), nimotuzumab (Center of Molecular Immunology), NSC- 330507 or NSC-707545 (NIH), NV-50 (Novogen), OSI-930 (OSI Pharmaceuticals), panitumumab (Abgenix), pelitinib (CAS No. 287933-82-7) (Wyeth), pertuzumab (CAS No. 380610-27-5) (Genentech), Pharmaprojects No. 3985 (Sanofi-Aventis), prostate cancer therapeutics from Sequenom (SQPC35, SQPC36, SQPC90), removab and remoxab (Trion Pharma), RG- 13022 (CAS No. 136831-48-6), RG-13291 (CAS No. 138989-50-1), or RG- 14620 (CAS No. 136831-49-7) (Sanofi-Aventis), RM-6427 (Romark), RNAi breast cancer therapy from Benitec, RP 53801 (CAS No. 125882-88-4) (Sanofi-Aventis), sorafenib tosylate (Bayer), SU-11657 (Pfizer), Tie-2 antagonists from Semaia (Hybrigenics), Tie-2 inhibitors from Ontogen, trastuzumab (CAS No. 180288-69-1) (Genentech), tyrosine kinase inhibitors from Sanofi-Aventis, U3-1287, U3-1565, U3-1784, U3-1800 (U3 Pharma), vatalanib (Novartis), VEGFR-2 kinase inhibitor from Bristol-Myers Squibb, XL-647 (Exelixis), ZD-6474 (AstraZeneca), ZK-CDK (Schering AG), an EGFR tyrosine kinase inhibitor, EKB-569 (Wyeth), and an analogue or derivative thereof.
18415. The method of item 18313 wherein the anti- fibrotic agent is a VEGF inhibitor.
18416. The method of item 18313 wherein the anti- fibrotic agent is a vitamin D receptor agonist.
18417. The method of item 18313 wherein the anti- fibrotic agent is ZD-6474 (an angiogenesis inhibitor).
18418. The method of item 18313 wherein the anti- fibrotic agent is AP-23573 (an mTOR inhibitor).
18419. The method of item 18313 wherein the anti- fibrotic agent is synthadotin (a tubulin antagonist).
18420. The method of item 18313 wherein the anti- fibrotic agent is S-0885 (a collagenase inhibitor).
18421. The method of item 18313 wherein the anti- fibrotic agent is aplidine (an elongation factor-1 alpha inhibitor). 18422. The method of item 18313 wherein the anti- fibrotic agent is ixabepilone (an epithilone).
18423. The method of item 18313 wherein the anti- fibrotic agent is IDN-5390 (an angiogenesis inhibitor).
18424. The method of item 18313 wherein the anti- fibrotic agent is SB-2723005 (an angiogenesis inhibitor).
18425. The method of item 18313 wherein the anti- fibrotic agent is ABT-518 (an angiogenesis inhibitor).
18426. The method of item 18313 wherein the anti- fibrotic agent is combretastatin (an angiogenesis inhibitor).
18427. The method of item 18313 wherein the anti- fibrotic agent is anecortave acetate (an angiogenesis inhibitor).
18428. The method of item 18313 wherein the anti- fibrotic agent is SB-715992 (a kinesin antagonist).
18429. The method of item 18313 wherein the anti- fibrotic agent is temsirolimus (an mTOR inhibitor).
18430. The method of item 18313 wherein the anti- fibrotic agent is adalimumab (a TNFα antagonist).
18431. The method of item 18313 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with an anti-fibrotic agent, and (b) implanting the medical device into the host. 18432. The method of item 18313 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with an anti-infective agent, and (b) implanting the medical device into the host.
18433. The method of item 18313 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with a polymer; and (b) implanting the medical device into the host.
18434. The method of item 18313 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with a composition comprising an anti-fibrotic agent and a polymer, and (b) implanting the medical device into the host.
18435. The method of item 18313 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with a composition comprising an anti-infective agent and a polymer, and (b) implanting the medical device into the host.
18436. The method of item 18313 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with a composition comprising an anti-fibrotic agent, an anti- infective agent and a polymer, and (b) implanting the medical device into the host.
18437. The method of item 18313 wherein the anti- infective agent is an anthracycline.
18438. The method of item 18313 wherein the anti- infective agent is doxorubicin. 18439. The method of item 18313 wherein the anti- infective agent is is mitoxantrone.
18440. The method of item 18313 wherein the anti- infective agent is a fluoropyrimidine.
18441. The method of item 18313 wherein the anti- infective agent is 5-fluorouracil (5-FU).
18442. The method of item 18313 wherein the anti- infective agent is a folic acid antagonist.
18443. The method of item 18313 wherein the anti- infective agent is methotrexate.
18444. The method of item 18313 wherein the anti- infective agent is a podophylotoxin.
18445. The method of item 18313 wherein the anti- infective agent is etoposide.
18446. The method of item 18313 wherein the anti- infective agent is camptothecin.
18447. The method of item 18313 wherein the anti- infective agent is a hydroxyurea.
18448. The method of item 18313 wherein the anti- infective agent is a platinum complex. 18449. The method of item 18313 wherein the anti- infective agent is cisplatin.
18450. The method of item 18313 wherein the composition comprises an anti-thrombotic agent.
18451. The method of item 18313 wherein the polymer is formed from reactants comprising a naturally occurring polymer.
18452. The method of item 18313 wherein the polymer is formed from reactants comprising protein.
18453. The method of item 18313 wherein the polymer is formed from reactants comprising carbohydrate.
18454. The method of item 18313 wherein the polymer is formed from reactants comprising biodegradable polymer.
18455. The method of item 18313 wherein the polymer is formed from reactants comprising nonbiodegradable polymer.
18456. The method of item 18313 wherein the polymer is formed from reactants comprising collagen.
18457. The method of item 18313 wherein the polymer is formed from reactants comprising methylated collagen.
18458. The method of item 18313 wherein the polymer is formed from reactants comprising fibrinogen. 18459. The method of item 18313 wherein the polymer reactants comprising thrombin.
18460. The method of item 18313 wherein the polymer reactants comprising blood plasma.
18461. The method of item 18313 wherein the polymer reactants comprising calcium salt.
18462. The method of item 18313 wherein the polymer reactants comprising an antifibronolytic agent.
18463. The method of item 18313 wherein the polymer reactants comprising fibrinogen analog.
18464. The method of item 18313 wherein the polymer reactants comprising albumin.
18465. The method of item 18313 wherein the polymer reactants comprising plasminogen.
18466. The method of item 18313 wherein the polymer reactants comprising von Willebrands factor.
18467. The method of item 18313 wherein the polymer reactants comprising Factor VIII.
18468. The method of item 18313 wherein the polymer reactants comprising hypoallergenic collagen. 18469. The method of item 18313 wherein the polymer is formed from reactants comprising atelopeptidic collagen.
18470. The method of item 18313 wherein the polymer is formed from reactants comprising telopeptide collagen.
18471. The method of item 18313 wherein the polymer is formed from reactants comprising crosslinked collagen.
18472. The method of item 18313 wherein the polymer is formed from reactants comprising aprotinin.
18473. The method of item 18313 wherein the polymer is formed from reactants comprising epsilon-amino-n-caproic acid.
18474. The method of item 18313 wherein the polymer is formed from reactants comprising gelatin.
18475. The method of item 18313 wherein the polymer is formed from reactants comprising protein conjugates.
18476. The method of item 18313 wherein the polymer is formed from reactants comprising gelatin conjugates.
18477. The method of item 18313 wherein the polymer is formed from reactants comprising a synthetic polymer.
18478. The method of item 18313 wherein the polymer is formed from reactants comprising a synthetic isocyanate-containing compound. 18479. The method of item 18313 wherein the polymer is formed from reactants comprising a synthetic thiol-containing compound.
18480. The method of item 18313 wherein the polymer is formed from reactants comprising a synthetic compound containing at least two thiol groups.
18481. The method of item 18313 wherein the polymer is formed from reactants comprising a synthetic compound containing at least three thiol groups.
18482. The method of item 18313 wherein the polymer is formed from reactants comprising a synthetic compound containing at least four thiol groups.
18483. The method of item 18313 wherein the polymer is formed from reactants comprising a synthetic amino-containing compound.
18484. The method of item 18313 wherein the polymer is formed from reactants comprising a synthetic compound containing at least two amino groups.
18485. The method of item 18313 wherein the polymer is formed from reactants comprising a synthetic compound containing at least three amino groups.
18486. The method of item 18313 wherein the polymer is formed from reactants comprising a synthetic compound containing at least four amino groups. 18487. The method of item 18313 wherein the polymer is formed from reactants comprising a synthetic compound comprising a carbonyl-oxygen-succinimidyl group.
18488. The method of item 18313 wherein the polymer is formed from reactants comprising a synthetic compound comprising at least two carbonyl-oxygen-succinimidyl groups.
18489. The method of item 18313 wherein the polymer is formed from reactants comprising a synthetic compound comprising at least three carbonyl-oxygen-succinimidyl groups.
18490. The method of item 18313 wherein the polymer is formed from reactants comprising a synthetic compound comprising at least four carbonyl-oxygen-succinimidyl groups.
18491. The method of item 18313 wherein the polymer is formed from reactants comprising a synthetic polyalkylene oxide- containing compound.
18492. The method of item 18313 wherein the polymer is formed from reactants comprising a synthetic compound comprising both polyalkylene oxide and biodegradable polyester blocks.
18493. The method of item 18313 wherein the polymer is formed from reactants comprising a synthetic polyalkylene oxide- containing compound having reactive amino groups.
18494. The method of item 18313 wherein the polymer is formed from reactants comprising a synthetic polyalkylene oxide- containing compound having reactive thiol groups. 18495. The method of item 18313 wherein the polymer is formed from reactants comprising a synthetic polyalkylene oxide- containing compound having reactive carbonyl-oxygen-succinimidyl groups.
18496. The method of item 18313 wherein the polymer is formed from reactants comprising a synthetic compound comprising a biodegradable polyester block.
18497. The method of item 18313 wherein the polymer is formed from reactants comprising a synthetic polymer formed in whole or part from lactic acid or lactide.
18498. The method of item 18313 wherein the polymer is formed from reactants comprising a synthetic polymer formed in whole or part from glycolic acid or glycolide.
18499. The method of item 18313 wherein the polymer is formed from reactants comprising polylysine.
18500. The method of item 18313 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound comprising a polyalkylene oxide portion.
18501. The method of item 18313 wherein the polymer is formed from reactants comprising (a) protein and (b) polylysine.
18502. The method of item 18313 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound having at least four thiol groups. 18503. The method of item 18313 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound having at least four amino groups.
18504. The method of item 18313 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound having at least four carbonyl-oxygen-succinimide groups.
18505. The method of item 18313 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound having a biodegradable region formed from reactants selected from lactic acid, lactide, glycolic acid, glycolide, and epison-caprolactone.
18506. The method of item 18313 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound comprising a polyalkylene oxide portion.
18507. The method of item 18313 wherein the polymer is formed from reactants comprising (a) collagen and (b) polylysine.
18508. The method of item 18313 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound having at least four thiol groups.
18509. The method of item 18313 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound having at least four amino groups.
18510. The method of item 18313 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound having at least four carbonyi-oxygen-succinimide groups. 18511. The method of item 18313 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound having a biodegradable region formed from reactants selected from lactic acid, lactide, glycolic acid, glycolide, and epison-caprolactone.
18512. The method of item 18313 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound comprising a polyalkylene oxide portion.
18513. The method of item 18313 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) polylysine.
18514. The method of item 18313 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound having at least four thiol groups.
18515. The method of item 18313 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound having at least four amino groups.
18516. The method of item 18313 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound having at least four carbonyl-oxygen-succinimide groups.
18517. The method of item 18313 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound having a biodegradable region formed from reactants selected from lactic acid, lactide, glycolic acid, glycolide, and epison-caprolactone. 18518. The method of item 18313 wherein the polymer is formed from reactants comprising hyaluronic acid.
18519. The method of item 18313 wherein the polymer is formed from reactants comprising a hyaluronic acid derivative.
18520. The method of item 18313 wherein the polymer is formed from reactants comprising pentaerythritol poly(ethylene glycol)ether tetra-sulfhydryl of number average molecular weight between 3,000 and 30,000.
18521. The method of item 18313 wherein the polymer is formed from reactants comprising pentaerythritol poly(ethylene glycol)ether tetra-amino of number average molecular weight between 3,000 and 30,000.
18522. The method of item 18313 wherein the polymer is formed from reactants comprising (a) a synthetic compound having a number average molecular weight between 3,000 and 30,000 and comprising a polyalkylene oxide region and multiple nucleophilic groups, and (b) a synthetic compound having a number average molecular weight between 3,000 and 30,000 and comprising a polyalkylene oxide region and multiple electrophilic groups.
18523. The method of item 18313 wherein the composition comprises a colorant.
18524. The method of item 18313 wherein the composition is sterile. 18525. A method as in any one of items 18313-18524, wherein the device is a ureteric stent.
18526. A method as in any one of items 18313-18524, wherein the device is a urethral stent.
18527. A method as in any one of items 18313-18524, wherein the device is a fallopian tube stent.
18528. A method as in any one of items 18313-18524, wherein the device is a prostate stent.
18529. A method as in any one of items 18313-18524, wherein the device is a metallic genital urinary stent.
18530. A method as in any one of items 18313-18524, wherein the device is a polymeric genital urinary stent.
18531. A method for implanting a medical device comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with i) an anti-fibrotic agent, ii) an anti-infective agent, iii) a polymer, iv) a composition comprising an anti-fibrotic agent and a polymer, v) a composition comprising an anti-infective agent and a polymer, and (b) implanting the medical device into the host, wherein the medical device is an ear and nose stent.
18532. The method of item 18531 wherein the anti- fibrotic agent is an adensosine A2A receptor antagonist.
18533. The method of item 18531 wherein the anti- fibrotic agent is an AKT inhibitor. 18534. The method of item 18531 wherein the anti- fibrotic agent is an alpha 2 integrin antagonist, wherein the alpha 2 integrin antagonist is Pharmaprojects No. 5754 (Merck KgaA).
18535. The method of item 18531 wherein the anti- fibrotic agent is an alpha 4 integrin antagonist.
18536. The method of item 18531 wherein the anti- fibrotic agent is an alpha 7 nicotinic receptor agonist.
18537. The method of item 18531 wherein the anti- fibrotic agent is an angiogenesis inhibitor selected from the group consisting of AG-12,958 (Pfizer), ATN-161 (Attenuon LLC), neovastat, an angiogenesis inhibitor from Jerina AG (Germany), NM-3 (Mercian), VGA-1155 (Taisho), FCE-26644 (Pfizer), FCE-26950 (Pfizer), FPMA (Meiji Daries), FR-111142 (Fujisawa), GGTI-298, GM-1306 (Ligand), GPA-1734 (Novartis), NNC-47- 0011 (Novo Nordisk), herbamycin (Nippon Kayaku), lenalidomide (Celegene), IP-10 (NIH), ABT-828 (Abbott), KIN-841 (Tokushima University, Japan), SF-1126 (Semafore Pharmaceuticals), laminin technology (NIH), CHIR-258 (Chiron), NVP-AEW541 (Novartis), NVP-AEW541 (Novartis), Vt16907 (Alchemia), OXI-8007 (Oxigene), EG-3306 (Ark Therapeutics), Maspin (Arriva), ABT-567 (Abbott), PPI-2458 (Praecis Pharmaceuticals), CC-5079, CC-4089 (Celgene), HIF-1 alpha inhibitors (Xenova), S-247 (Pfizer), AP-23573 (Ariad), AZD-9935 (Astra Zeneca), mebendazole (Introgen Therapeutics), MetAP-2 inhibitors (GlaxoSmithKline), AG-615 (Angiogene Pharmaceuticals), Tie-2 antagonists (Hybrigenics), NC-381 , CYC-381, NC-169, NC-219, NC-383, NC-384, NC-407 (Lorus Therapeutics), ATN-224 (Attenuon), ON-01370 (Onconova), Vitronectin antagonists (Amgen), SDX-103 (Salmedix), Vitronectin antagonists (Shire), CHP (Riemser), TEK (Amgen), Anecortave acetate (Alcon), T46.2 (Matrix Therapeutics), HG-2 (Heptagen), TEM antagonists (Genzyme), Oxi-4500 (Oxigene), ATN-161 (Attenuon), WX-293 (Wilex), M-2025 (Metris Therapeutics), Alphastatin (BioActa), YH-16 (Yantai Rongchang), BIBF- 1120 (Boehringer Ingelheim), BAY-57-9352 (Bayer), AS-1404 (Cancer Research Technology), SC-77964 (Pfizer), glycomimetics (BioTie Therapies), TIE-2 Inhibitors (Ontogen), DIMI, Octamer (Octamer), ABR- 215050 (Active Biotech), ABT-518 (Abbott), KDR inhibitors (Abbott), BSF- 466895 (Abbott), SCH-221153 (Schering-Plough), DAC:antiangiogenic (ConjuChem), TFPI (EntreMed), AZD-2171 (Astra-Zenaca), CDC-394 (Celgene), LY290293 (EIi Lilly), IDN-5390 (Indena), Kdr Kinase Inhibitors (Merck), CT-113020, CT-116433, CT-116563, CT-31890, CT-32228) (Cell Therapeutics), A-299620 (Abbott), TWEAK Inhibitor (Amgen), VEGF modulators (Johnson and Johnson), Tum-N53, tumstatin (Genzyme), Thios- 1, Thios-2 (Thios Pharmaceuticals), MV-6401 (Miravant Medical Technologies), Spisulosine (PharmaMar), CEP-7055 (Cephalon), AUV-201 (Auvation), LM-609 (EIi Lilly), SKF-106615 (AnorMED), Oglufanide disodium (Cytran), BW-114 (Pharminox), Calreticulin (NIH), WX-678 (Wilex), SD-7784 (Pfizer), WX-UK1 (Wilex), SH-268 (Schering AG), 2-Me-PGA (Celgene), S- 137 (Pfizer), ZD-6126 (Angiogene Pharmaceuticals), SG-292 (SignalGen), Benefin (Lane Labs), A6, A36 (Angstrom), SB-2723005 (GlaxoSmithKline), SC-7 (Cell Therapeutics), ZEN-014 (AEtema Zentaris), 2-methoxyestradiol (EntreMed), NK-130119 (Nippon Kayaku), CC-10004 (Celgene), AVE- 8062A (Ajinomoto), Tacedinaline (Pfizer), Actinonin (Tokyo Metropolitan Institute of Medical Science), Lenalidomide (Celgene), VGA-1155, BTO-956 (SRI International), ER-68203-00 (Eisai), CT-6685 (UCB)1 JKC-362 (Phoenix Pharmaceuticals), DMI-3798 (DMI Biosciences, Angiomate (Ipsen), ZD-6474 (AstraZeneca), CEP-5214 (Cephalon), Canstatin (Genzyme), NM-3 (Mercian), Oridigm (MediQuest Therapeutics), Exherin (Adherex), BLS-0597 (Boston Life Sciences), PTC-299 (PTC Therapeutics), NPl-2358 (Nereus Pharmaceuticals), CGP-79787 (Novartis), AEE-788 (Novartis), CKD-732 (Chong Kun Dang), CP-564959 (OSI Pharmaceuticals), CM-101 (CarboMed), CT-2584, CT3501 (Cell Therapeutics), combretastatin and analogues and derivatives thereof (Oxigene), Rebimastat (Bristol-Meyers Squibb), Dextrin 2-sulfate (ML Laboratories), Cilengitide (Merk KGaA), NSC-706704 (Pharminox), KRN- 951 (Kirin Brewery), Ukrain, NSC-631570 (Nowicky Pharma), Tecogalan sodium (Daiichi Pharmaceutical), Tz-93 (Tsumura), TBC-1635 (Encysive Pharmaceuticals), TAN-1120 (Takeda), Semaxanib (Pfizer), BDI-7800 (Biopharmacopae), SD-186, SD-983 (Bristol-Meyers Squibb), SB-223245 (GlaxoSmithKline), SC-236 (Pfizer), RWJ-590973 (Johnson and Johnson), ILX-1850 (Genzyme), SC-68488, S-836 (Pfizer), CG-55069-11 (CuraGen), Ki-23057 (Kirin Brewery), CCX-700 (Chemoentryx), Pegaptanib octasodium (Giled Sciences), ANGIOCOL (available from Biostratum Inc.), or an analogue or derivative thereof.
18538. The method of item 18531 wherein the anti- fibrotic agent is an apoptosis antagonist.
18539. The method of item 18531 wherein the anti- fibrotic agent is an apoptosis activator.
18540. The method of item 18531 wherein the anti- fibrotic agent is a beta 1 integrin antagonist.
18541. The method of item 18531 wherein the anti- fibrotic agent is a beta tubulin inhibitor.
18542. The method of item 18531 wherein the anti- fibrotic agent is a blocker of enzyme production in Hepatitis C.
18543. The method of item 18531 wherein the anti- fibrotic agent is a Bruton's tyrosine kinase inhibitor. 18544. The method of item 18531 wherein the anti- fibrotic agent is a calcineurin inhibitor.
18545. The method of item 18531 wherein the anti- fibrotic agent is a caspase 3 inhibitor.
18546. The method of item 18531 wherein the anti- fibrotic agent is a CC chemokine receptor antagonist.
18547. The method of item 18531 wherein the anti- fibrotic agent is a cell cycle inhibitor selected from the group consisting of SNS-595 (Sunesis), synthadotin, KRX-0403, and an analogue or derivative thereof.
18548. The method of item 18531 wherein the anti- fibrotic agent is a cathepsin. B inhibitor.
18549. The method of item 18531 wherein the anti- fibrotic agent is a cathepsin K inhibitor, wherein the cathepsin K inhibitor is 462795 (GlaxoSmithKline), INPL-022-D6 (Amura Therapeutics), or an analogue or derivative thereof.
18550. The method of item 18531 wherein the anti- fibrotic agent is a cathepsin L inhibitor.
18551. The method of item 18531 wherein the anti- fibrotic agent is a CD40 antagonist.
18552. The method of item 18531 wherein the anti- fibrotic agent is a chemokine receptor agonist. 18553. The method of item 18531 wherein the anti- fibrotic agent is a chymase inhibitor.
18554. The method of item 18531 wherein the anti- fibrotic agent is a collagenase antagonist.
18555. The method of item 18531 wherein the anti- fibrotic agent is a CXCR antagonist.
18556. The method of item 18531 wherein the anti- fibrotic agent is a cyclin dependent kinase inhibitor selected from the group consisting of a CDK-1 inhibitor, a CDK-2 inhibitor, a CDK- 4 inhibitor, a CDK-6 inhibitor, a CAK1 inhibitor from GPC Biotech and Bristol-Myers Squibb, RGB-286199 (GPC Biotech), an anticancer agent from Astex Technology, a CAK1 inhibitor from GPC Biotech, a CDK inhibitor from Sanofi-Aventis, a CDK1/CDK2 inhibitor from Amgen, a CDK2 inhibitor from SUGEN-2 (Pfizer), a hearing loss therapy agent (Sound Pharmaceuticals), PD-0332991 (Pfizer), RGB-286199 (GPC Biotech), Ro-0505124 (Hoffmann- La Roche), a Ser/Thr kinase inhibitor from Lilly (EIi Lilly), CVT-2584 (CAS No. 199986-75-9) (CV Therapeutics), and an analogue or derivative thereof.
18557. The method of item 18531 wherein the anti- fibrotic agent is a cyclooxygenase 1 inhibitor.
18558. The method of item 18531 wherein the anti- fibrotic agent is a DHFR inhibitor.
18559. The method of item 18531 wherein the anti- fibrotic agent is a dual integrin inhibitor. 18560. The method of item 18531 wherein the anti- fibrotic agent is an elastase inhibitor.
18561. The method of item 18531 wherein the anti- fibrotic agent is an elongation factor-1 alpha inhibitor.
18562. The method of item 18531 wherein the anti- fibrotic agent is an endothelial growth factor antagonist.
18563. The method of item 18531 wherein the anti- fibrotic agent is an endothelial growth factor receptor kinase inhibitor selected from the group consisting of sorafenib tosylate (Bayer), AAL-993 (Novartis), ABP-309 (Novartis), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EXEL- 2880 (Exelixis), GW-654652 (GlaxoSmithKline), a KDR inhibitor from LG Life Sciences, CT-6685 and CT-6729 (UCB), KRN-633 and KRN-951 (Kirjn . Brewery), OSI-930 (OSI Pharmaceuticals), SP-5.2 (Supratek Pharma), SU- 11657 (Pfizer), a Tie-2 antagonist (Hybrigenics), a VEGFR-2 kinase inhibitor (Bristol-Myers Squibb), XL-647 (Exelixis), a KDR inhibitor from Abbott Laboratories, sorafenib tosylate, and an analogue or derivative thereof.
18564. The method of item 18531 wherein the anti- fibrotic agent is an endotoxin antagonist.
18565. The method of item 18531 wherein the anti- fibrotic agent is an epothilone and tubulin binder.
18566. The method of item 18531 wherein the anti- fibrotic agent is an estrogen receptor antagonist. 18567. The method of item 18531 wherein the anti- fibrotic agent is an FGF inhibitor.
18568. The method of item 18531 wherein the anti- fibrotic agent is a farnexyl transferase inhibitor.
18569. The method of item 18531 wherein the anti- fibrotic agent is farnesyltransferase inhibitor selected from the group of A- 197574 (Abbott), a farnesyltransferase inhibitor from Servier, FPTIII (Strathclyde Institute for Drug R), LB-42908 (LG Life Sciences), Pharmaprojects No. 5063 (Genzyme), Pharmaprojects No. 5597 (Ipsen), Yissum Project No. B-1055 (Yissum), and an analogue or derivative thereof.
18570. The method of item 18531 wherein the anti- fibrotic agent is an FLT-3 kinase inhibitor.
18571. The method of item 18531 wherein the anti- fibrotic agent is an FGF receptor kinase inhibitor.
18572. The method of item 18531 wherein the anti- fibrotic agent is a fibrinogen antagonist selected from the group consisting of AUV-201 (Auvation), MG-13926 (Sanofi-Aventis), plasminogen activator (CAS No. 105913-11-9) (from Sanofi-Aventis or UCB), plasminogen activator-2 (tPA-2) (Sanofi-Aventis), pro-urokinase (CAS No. 82657-92-9) (Sanofi-Aventis), and an analogue or derivative thereof.
18573. The method of item 18531 wherein the anti- fibrotic agent is a heat shock protein 90 antagonist selected from the group consisting of SRN-005 (Sirenade), geldanamycin, NSC-33050 (17- allylaminogeldanamycin; 17-AAG), 17-dimethylaminoethylamino-17- demethoxy-geldanamycin (17-DMAG), rifabutin (rifamycin XIV, 1',4- didehydro-1-deoxy-1,4-dihydro-5'-(2-methylpropyl)-1-oxo-), and an analogue or derivative thereof.
18574. The method of item 18531 wherein the anti- fibrotic agent is a histone deacetylase inhibitor.
18575. The method of item 18531 wherein the anti- fibrotic agent is an HMGCoA reductase inhibitor selected from the group consisting of an atherosclerosis therapeutic from Lipid Sciences, ATI-16000 (ARYx Therapeutics), KS-01-019 (Kos Pharmaceuticals), Pharmaprojects No. 2197 (Sanofi-Aventi), RP 61969 (Sanofi-Aventis), and an analogue or derivative thereof.
18576. The method of item 18531 wherein the anti- fibrotic agent is an ICAM inhibitor.
18577. The method of item 18531 wherein the anti- fibrotic agent is an IL, ICE and IRAK antagonist, wherein the antagonist is a CJ-14877, CP-424174 (Pfizer), NF-61 (Negma-Lerads), and an analogue or derivative thereof.
18578. The method of item 18531 wherein the anti- fibrotic agent is an IL-2 inhibitor.
18579. The method of item 18531 wherein the anti- fibrotic agent is an immunosuppressant selected from the group consisting of teriflunomide (Sanofi Aventis), chlorsulfaquinoxalone (NSC-339004), chlorsulfaquinoxalone sulfate, CS-712 (Sankyo), ismomultin alfa (CAS No. 457913-93-8) (Akzo Nobel), antiallergics from GenPat77, antiinflammatories or AT-005 (Androclus Therapeutics), autoimmune disease therapeutics from EpiVax, BN-007 (Bone), budesonide (CAS No. 51333-22- 3) (MAP Pharmaceuticals), CO-14 (Genzyme), edratide (CAS No. 433922- 67-9) (Teva), EP-314 (Enanta), eprovafen (CAS No. 101335-99-3) (Sanofi- Aventis), HWA-131 (CAS No. 118788-41-3) (Sanofi-Aventis), immunomodulators from MerLion Pharmaceuticals, immunosuppressives from Alchemia, IPL-12 (Inflazyme), MDL-9563 (CAS No. 27086-86-8) (Sanofi-Aventis), Pharmaprojects No. 2330 (Sanofi-Aventis), Pharmaprojects No. 6426 (Abgenix), PXS-25 (Pharmaxis), rosmarinic acid (CAS No. 20283-92-5) (Sanofi-Aventis), RP 42927 or RP 54745 (CAS No. 135330-08-4) (Sanofi-Aventis), SGN-35 (Seattle Genetics), ST-1959 (Sigma-Tau), type I diabetes therapy from SYNX Pharma, UNIL-88 (Debiopharm), VP-025 (Vasogen), VR-694 (Vectura), PRTX-001 (Protalex), and an analogue or derivative thereof.
18580. The method of item 18531 wherein the anti- fibrotic agent is an IMPDH (inosine monophosphate).
18581. The method of item 18531 wherein the anti- fibrotic agent is an integrin antagonist.
18582. The method of item 18531 wherein the anti- fibrotic agent is an interleukin antagonist.
18583. The method of item 18531 wherein the anti- fibrotic agent is an inhibitor of type III receptor tyrosine kinase.
18584. The method of item 18531 wherein the anti- fibrotic agent is an irreversible inhibitor of enzyme methionine aminopeptidase type 2.
18585. The method of item 18531 wherein the anti- fibrotic agent is an isozyme selective delta protein kinase C inhibitor. 18586. The method of item 18531 wherein the anti- fibrotic agent a JAK3 enzyme inhibitor.
18587. The method of item 18531 wherein the anti- fibrotic agent is a JNK inhibitor.
18588. The method of item 18531 wherein the anti- fibrotic agent is a kinase inhibitor.
18589. The method of item 18531 wherein the anti- fibrotic agent is kinesin antagonist.
18590. The method of item 18531 wherein the anti- fibrotic agent is a kinesin antagonist.
18591. The method of item 18531 wherein the anti- fibrotic agent is a leukotriene inhibitor and antagonist selected from the group consisting of ambicromil (CAS No. 58805-38-2) (Sanofi-Aventis), amelubant (CAS No. 346735-24-8) (Boehringer Ingelheim), DW-1141 (Dong Wha), ebselen (Daiichi Pharmaceutical), ibudilast (Kyorin), leucotriene inhibitors from Sanofi-Aventis, lymphotoxin-beta receptor (LT-D) from Biogen Idee, Pharmaprojects No. 1535 and 2728 (CAS No. 119340-33-9) (Sanofi-Aventis), R-112 (Rigel), Rev-5367 (CAS No. 92532-05-3) (Sanofi- Aventis), RG-14893 (CAS No. 141835-49-6) (Sanofi-Aventis), RG-5901-A (CAS No. 101910-24-1), 92532-23-5, RP 66153 (CAS No. 142422-79-5), RP 66364 (CAS No.186916-92-5), RP 69698 (CAS No. 141748-00-7) (Sanofi-Aventis), SC-411930 (Pfizer), SC-41930 (CAS No. 120072-59-5) (Pfizer), SC-50605 (CAS No. 138828-39-4) (Pfizer), SC-51146 (CAS No. 141059-52-1), SC-53228 (CAS No. 153633-01-3) (Pfizer), spaglumic acid (ZY-15106) (CAS No.3106-85-2), 80619-64-3 (Novartis), tipredane (CAS No. 85197-77-9) (Bristol-Myers Squibb), U-75302 (CAS No. 119477-85-9) (Pfizer), and analogue or derivative thereof.
18592. The method of item 18531 wherein the anti- fibrotic agent is an MAP kinase inhibitor.
18593. The method of item 18531 wherein the anti- fibrotic agent is a matrix metalloproteinase inhibitor.
18594. The method of item 18531 wherein the anti- fibrotic agent is an MCP-CCR2 inhibitor.
18595. The method of item 18531 wherein the anti- fibrotic agent is an mTOR inhibitor.
18596. The method of item 18531 wherein the anti- fibrotic agent is an mTOR kinase inhibitor.
18597. The method of item 18531 wherein the anti- fibrotic agent is a microtubule inhibitor selected from the group consisting of antibody-maytansinoid conjugates from Biogen Idee, colchicines (MantiCore Pharmaceuticals), anticancer immunoconjugates from Johnson & Johnson, DIME from Octamer, gni-1f (GNI), huC242-DM4, huMy9-6-DM1 (ImmunoGen), IDN-5404 (Indena), IMO-098, IMOderm (Imotep), mebendazole (Introgen Therapeutics), microtubule poisons from Cambridge Enterprise, paclitaxel such as LOTAX from Aphios (CAS No. 33069-62-4), Genexol-PM from Samyang, Pharmaprojects No. 6383 (Tapestry Pharmaceuticals), RPR-112378 (Sanofi-Aventis), SGN-75 (Seattle Genetics), SPL-7435 (Starpharma), SSR-250411 (Sanofi-Aventis), trastuzumab-DM1 (Genentech), vinorelbine, and an analogue or derivative thereof. 18598. The method of item 18531 wherein the anti- fibrotic agent is an MIF inhibitor.
18599. The method of item 18531 wherein the anti- fibrotic agent is an MMP inhibitor.
18600. The method of item 18531 wherein the anti- fibrotic agent is a neurokinin (NK) antagonist selected from the group consisting of anthrotainin (CAS No. 148084-40-6) (Sanofi-Aventis), an IBS thereapeutic from ArQuIe, MDL-105212A (CAS No. 167261-60-1) (Ssanofi- Aventis), Pharmaprojects No. 2744, 3258 (CAS No. 139167-47-8) 4006, 4201 , or 5986 (Sanofi-Aventis), RP 67580 (CAS No. 135911-02-3), SR- 144190 (CAS No. 201152-86-5), SSR-240600, SSR-241586 (Sanofi- Aventis), TKA-457 (Novartis), vestipitant mesylate (CAS No. 334476-64-1) (GlaxoSmithKline), Win-64821 (Sanofi-Aventis), PRX-96026 (Predix Pharmaceuticals), and an analogue or derivative thereof.
18601. The method of item 18531 wherein the anti- fibrotic agent is an NF kappa B inhibitor selected from the group consisting of AVE-0545 or AVE-0547 (Sanofi-Aventis), bortezomib (CAS No. 179324- 69-7) (Millennium Pharmaceuticals), dexanabinol (CAS No. 112924-45-5) (Pharmos), dexlipotam (Viatris), Pharmaprojects No. 6283 (INDRA) (OXiGENE), IPL-576092 (CAS No. 137571-30-3) (Inflazyme), NFKB decoy (Corgentech), NFKB decoy oligo (AnGes MG), NFKB's from Ariad, osteoporosis treatments or S5 (F005) from Fulcrum Pharmaceuticals, P61 (Phytopharm), R-flurbiprofen (CAS No. 5104-49-4) (Encore Pharmaceuticals), and an analogue or derivative thereof.
18602. The method of item 18531 wherein the anti- fibrotic agent is a nitric oxide agonist. 18603. The method of item 18531 wherein the anti- fibrotic agent is an ornithine decarboxylase inhibitor.
18604. The method of item 18531 wherein the anti- fibrotic agent is a p38 MAP kinase inhibitor selected from the group consisting of AZD-6703 (AstraZeneca), JX-401 (Jexys Pharmaceuticals), BMS-2 (Bristol-Myers Squibb), a p38 MAP kinase inhibitor from Novartis, a p38-alpha MAP kinase inhibitor from Amphora, Pharmaprojects No. 5704 (Pharmacopeia), RPR-200765A (Sanofi-Aventis), SD-282 (Johnson & Johnson), TAK-715 (Takeda), and an analogue or derivative thereof.
18605. The method of item 18531 wherein the anti- fibrotic agent is a palmitoyl-protein thioesterase inhibitor.
18606. The method of item 18531 wherein the anti- fibrotic agent is a PDGF receptor kinase inhibitor selected from the group consisting of AAL-993, AMN-107, or ABP-309 (Novartis), AMG-706 (Amgen), BAY-57-9352 (Bayer), CDP-860 (UCB), E-7080 (Eisai), imatinib (CAS No. 152459-95-5) (Novartis), OSI-930 (OSI Pharmaceuticals), RPR- 127963E (Sanofi-Aventis), RWJ-540973 (Johnson & Johnson), sorafenib tosylate (Bayer), SU-11657 (Pfizer), tandutinib (CAS No. 387867-13-2) (Millennium Pharmaceuticals), vatalanib (Novartis), ZK-CDK (Schering AG), and an analogue or derivative thereof.
18607. The method of item 18531 wherein the anti- fibrotic agent is (-)-halofenate (Metabolex), AMG-131 (Amgen), antidiabetics from Japan Tobacco, AZD-4619, AZD-8450, AZD-8677 (AstraZeneca), DRF-10945, balaglitazone (Dr Reddy's), CS-00088, CS-00098 (Chipscreen Biosciences), E-3030 (Eisai), etalocib (CAS No. 161172-51-6) (EIi Lilly), GSK-641597 (Ligand), GSK-677954 (GlaxoSmithKline), GW-409544 (Ligand), GW-590735 (GlaxoSmithKline), K-111 (Hoffmann-La Roche), LY- 518674 (EIi Lilly), LY-674 (Ligand), LY-929 (Ligand), MC-3001, MC-3002 (MaxoCore Pharmaceuticals), metformin HCI + pioglitazone (CAS No. 1115- 70-4 and 112529-15-4), ACTOPLUS MET from Andrx), muraglitazar (CAS No. 331741-94-7) (Bristol-Myers Squibb), naveglitazar (Ligand), oleoylethanolamide (Kadmus Pharmaceuticals), ONO-5129, pioglitazone hydrochloride (CAS No. 111025-46-8 and 112529-15-4) (Takeda), PLX-204 (Plexxikon), PPAR agonists from Genfit, PPAR delta agonists from EIi Lilly, PPAR-alpha agonists from CrystalGenomics, PPAR-gamma modulators and PPAR-β modulators from CareX, rosiglitazone maleate (CAS No. 122324- 73-4 or 155141-29-0) (GlaxoSmithKline), rosiglitazone maleate/glimepir (CAS No. 155141-29-0 and 93479-97-1), AVANDARYL, rosiglitazone maleate/metformin extend (CAS No. 155141-29-0 and 657-24-9), AVANDAMET, rosiglitazone maleate+metformin, AVANDAMET (GlaxoSmithKline), tesaglitazar (AstraZeneca), and an analogue or derivative thereof.
18608. The method of item 18531 wherein the anti- fibrotic agent is a phosphatase inhibitor.
18609. The method of item 18531 wherein the anti- fibrotic agent is a phosphodiesterase (PDE) inhibitor selected from the group consisting of avanafil (Tanabe Seiyaku), dasantafil (CAS No. 569351- 91-3) (Schering-Plough), A-906119 (CAS No. 134072-58-5), DL-850 (Sanofi-Aventis), GRC-3015, GRC-3566, GRC-3886 (Glenmark), HWA-153 (CAS No. 56395-66-5) (Sanofi-Aventis), hydroxypumafentrine (Altana), IBFB-130011, IBFB-14-016, IBFB-140301 , IBFB-150007, IBFB-211913 (IBFB Pharma), L-826141 (Merck & Co), medorinone (CAS No. 88296-61-1) (Sanofi-Aventis), MEM-1917 (Memory Pharmaceuticals), ND-1251 (Neuro3d), PDE inhibitors from ICOS, PDE IV inhibitors from Memory Pharmaceuticals and CrystalGenomics, Pharmaprojects No. 2742 and 6141 (Sanofi-Aventis), QAD-171 (Novartis), RHC-2963 (CAS No. 76993-12-9 and 76993-14-1), RPR-117658, RPR-122818 derivatives, SR-24870 , and RPR- 132294 (Sanofi-Aventis), SK-350 (ln2Gen), stroke therapy agents from deCODE Genetics, TAS-203 (Taiho), tofimilast (CAS No. 185954-27-2) (Pfizer), UK-371800 (Pfizer), WIN-65579 (CAS No. 158020-82-7) (Sanofi- Aventis), IBFB-130020 (IBFB Pharma), OPC-6535 (CAS No. 145739-56-6) (Otsuka), a phosphodiesterase III inhibitor, enoximone, a phosphodiesterase IV inhibitor, fosfosal, Atopik (Barrier Therapeutics), triflusal, a phosphodiesterase V inhibitor, and an analogue or derivative thereof.
18610. The method of item 18531 wherein the anti- fibrotic agent is a PKC inhibitor.
18611. The method of item 18531 wherein the anti- fibrotic agent is a platelet activating factor antagonist.
18612. The method of item 18531 wherein the anti- fibrotic agent is a platelet-derived growth factor receptor kinase inhibitor.
18613. The method of item 18531 wherein the anti- fibrotic agent is a prolyl hydroxylase inhibitor.
18614. The method of item 18531 wherein the anti- fibrotic agent is a polymorphonuclear neutrophil inhibitor.
18615. The method of item 18531 wherein the anti- fibrotic agent is a protein kinase B inhibitor.
18616. The method of item 18531 wherein the anti- fibrotic agent is a protein kinase C stimulant. 18617. The method of item 18531 wherein the anti- fibrotic agent is a purine nucleoside analogue.
18618. The method of item 18531 wherein the anti- fibrotic agent is a purinoreceptor P2X antagonist.
18619. The method of item 18531 wherein the anti- fibrotic agent is a Raf kinase inhibitor.
18620. The method of item 18531 wherein the anti- fibrotic agent is a reversible inhibitor of ErbB1 and ErbB2.
18621. The method of item 18531 wherein the anti- fibrotic agent is a ribonucleoside triphosphate reductase inhibitor.
18622. The method of item 18531 wherein the anti- fibrotic agent is an SDF-1 antagonist.
18623. The method of item 18531 wherein the anti- fibrotic agent is a sheddase inhibitor.
18624. The method of item 18531 wherein the anti- fibrotic agent is an SRC inhibitor.
18625. The method of item 18531 wherein the anti- fibrotic agent is a stromelysin inhibitor.
18626. The method of item 18531 wherein the anti- fibrotic agent is an Syk kinase inhibitor. 18627. The method of item 18531 wherein the anti- fibrotic agent is a telomerase inhibitor.
18628. The method of item 18531 wherein the anti- fibrotic agent is a TGF beta inhibitor selected from the group consisting of pirfenidone (CAS No. 53179-13-8) (MARNAC), tranilast (CAS No. 53902- 12-8) (Kissei), IN-1130 (ln2Gen), mannose-6-phosphate (BTG), TGF-β antagonists from Inflazyme (Pharmaprojects No. 6075), TGF-β antagonists from Sydney, non-industrial source), TGF-β I receptor kinase inhibitors from EIi Lilly, TGF-β receptor inhibitors from Johnson & Johnson, and an analogue or derivative thereof.
18629. The method of item 18531 wherein the anti- fibrotic agent is a TNFα antagonist or TACE inhibitor selected from the group consisting of adalimumab (CAS No. 331731-18-1) (Cambridge Antibody Technology), AGIX-4207 (AtheroGenics), AGT-1 (Advanced Biotherapy), an anti-inflammatory from Borean Pharma, Cellzome, or Paradigm Therapeutics, anti-inflammatory vaccine (TNF-alpha kinoid) from Neovacs, humanized anti-TNF antibody or an anti-TNF MAb (CB0006) Celltech (UCB), apratastat (CAS No. 287445-51-0) (Wyeth), BMS-561392 (Bristol-Myers Squibb), BN-006 (Bone), certolizumab pegol (CAS No. 428863-50-7 or CH-138 (UCB), cilomilast (CAS No. 153259-65-5) (GlaxoSmithKline), CR-1 (Nuada Pharmaceuticals), CRx-119 (CombinatoRx), D-5410 (UCB), dacopafant (CAS No. 125372-33-0) (Sanofi- Aventis), dersalazine (CAS No. 188913-57-7/188913-58-8) (Uriach), etanercept (CAS No. 185243-69-0) (Amgen), ethyl pyruvate (Critical (Critical Therapeutics), golimumab (CAS No. 476181-74-5) (Johnson & Johnson), hormono-immunotherapy from Ipsen, CDP571 {e.g., Humicade from UCB), IC-485 (ICOS), infliximab (CAS No. 170277-31-3) (Johnson & Johnson), IP- 751 (Manhattan Pharmaceuticals), ISIS-104838 (CAS No. 250755-32-9) (ISIS Pharmaceuticals), lenalidomide (CAS No. 191732-72-6) (Celgene), lentinan (CAS No. 37339-90-5) (Ajinomoto), MDL-201112 (CAS No. 142130-73-2) (Sanofi-Aventis), medroxyprogesterone (CAS No. 520-85-4) (InKine Pharmaceutical), N-acetylcysteine (CAS No. 616-91-1) (Zambon), NBE-P2 (DIREVO Biotech), nerelimomab (CAS No. 162774-06-3) (Chiron), OM-294DP (OM PHARMA), onercept (CAS No. 199685-57-9) (Yeda), PASSTNF-alpha (Verigen), pentoxifylline or oxypentifylline (Sanofi-Aventis), Pharmaprojects No. 4091 , 4241 , 4295, or 4488 (Sanofi-Aventis), Pharmaprojects No. 5480 (Amgen), Pharmaprojects No. 6749 (Cengent), pirfenidone (CAS No. 53179-13-8) (MARNAC), RPR- 132294 (Sanofi- Aventis), S5 (F002) (Fulcrum Pharmaceuticals), simvastatin (CAS No. 79902-63-9) (Merck & Co), STA-6292 (Synta Pharmaceuticals), tacrolimus (CAS No. 104987-11-3) (Fujisawa LifeCycle Pharma), talactoferrin alfa (CAS No. 308240-58-6) (Agennix), thalidomide (CAS No. 50-35-1) (Celgene), TNF antagonists form ProStrakan, and Synergen, TNF inhibitors (Amgen), TNF-alpha antagonists from Dynavax Technologies and Jerina AG (Germany), TNF-alpha inhibitors from IBFB Pharma and Xencor (Xencor), torbafylline (CAS No. 105102-21-4) (Sanofi-Aventis), UR-1505 (Uriach), VT-346 (Viron Therapeutics), YSIL6 (Y's Therapeutics), YSTH2 (Y's Therapeutics), NPI-1302a-3 (Nereus Pharmaceuticals, a TNF antagonist from Jerina AG (Germany), dersalazine, and an analogue or derivative thereof.
18630. The method of item 18531 wherein the anti- fibrotic agent is a Toll receptor inhibitor.
18631. The method of item 18531 wherein the anti- fibrotic agent is a tubulin antagonist.
18632. The method of item 18531 wherein the anti- fibrotic agent is a tyrosine kinase inhibitor selected from the group consisting of SU-011248, SUTENT from Pfizer Inc. (New York, NY), BMS-354825, PN- 355 (Paracelsian Pharmaceuticals), AGN-199659 (Allergan), AAL-993 or ABP-309 (Novartis), adaphostin (NIH), AEE-788 (Novartis), AG-013736 (OSI Pharmaceuticals), AG-13736 (Pfizer), ALT-110 (Alteris Therapeutics), AMG-706 (Amgen), anticancer MAbs from Xencor, anti-EGFrvlll MAbs from Abgenix, anti-HER2 MAb from Abiogen, AZD-2171 or AZD-9935 (AstraZeneca), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), CEP-5214 (Cephalon), CEP-7055 (Cephalon), cetuximab (ImCIone Systems), CHIR-200131 and CHIR-258 (Chiron), CP-547632 (OSI Pharmaceuticals), CP-724714 (Pfizer), CT-301 (Creabilis Therapeutics), D- 69491 (Baxter International), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EGFR/ErbB2 inhibitors from Array BioPharma, erlotinib (CAS No. 183319- 69-9) (OSI Pharmaceuticals), EXEL-2880 (Exelixis), FK-778 (Sanofi- Aventis) , gefitinib (CAS No. 184475-35-2) (AstraZeneca), GW-2286 or GW- 654652 (GlaxoSmithKline), her2/neu antigen from AlphaVax, HER-2/neu inhibitor from Generex, Herzyme (Medipad) (Sirna Therapeutics), HKI-272 (Wyeth), HuMax-EGFr (Genmab), idronoxil (CAS No. 81267-65-4). (Novogen), IGF-1 inhibitors from Ontogen, IMC-11F8 (ImCIone Systems), kahalalide F (CAS No. 149204-42-2) (PharmaMar), KDR inhibitor from LG Life Sciences, KDR inhibitors from Abbott Laboratories, KDR kinase inhibitors (UCB), Kdr kinase inhibitors from Merck & Co, KRN-633 and KRN- 951 (Kirin Brewery), KSB-102 (Xenova), lapatinib ditosylate (CAS No. 388082-78-8) (GlaxoSmithKline), matuzumab (Merck KGaA), MDX-214 (Medarex), ME-103 (Pharmexa), MED-A300 (Gerolymatos), MNAC-13 (Lay Line Genomics), nimotuzumab (Center of Molecular Immunology), NSC- 330507 or NSC-707545 (NIH), NV-50 (Novogen), OSI-930 (OSI Pharmaceuticals), panitumumab (Abgenix), pelitinib (CAS No. 287933-82-7) (Wyeth), pertuzumab (CAS No. 380610-27-5) (Genentech), Pharmaprojects No. 3985 (Sanofi-Aventis), prostate cancer therapeutics from Sequenom (SQPC35, SQPC36, SQPC90), removab and remoxab (Trion Pharma), RG- 13022 (CAS No. 136831-48-6), RG-13291 (CAS No. 138989-50-1), or RG- 14620 (CAS No. 136831-49-7) (Sanofi-Aventis), RM-6427 (Romark), RNAi breast cancer therapy from Benitec, RP 53801 (CAS No. 125882-88-4) (Sanofi-Aventis), sorafenib tosylate (Bayer), SlM 1657 (Pfizer), Tie-2 antagonists from Semaia (Hybrigenics), Tie-2 inhibitors from Ontogen, trastuzumab (CAS No. 180288-69-1) (Genentech), tyrosine kinase inhibitors from Sanofi-Aventis, U3-1287, U3-1565, U3-1784, U3-1800 (U3 Pharma), vatalanib (Novartis), VEGFR-2 kinase inhibitor from Bristol-Myers Squibb, XL-647 (Exelixis), ZD-6474 (AstraZeneca), ZK-CDK (Schering AG), an EGFR tyrosine kinase inhibitor, EKB-569 (Wyeth), and an analogue or derivative thereof.
18633. The method of item 18531 wherein the anti- fibrotic agent is a VEGF inhibitor.
18634. The method of item 18531 wherein the anti- fibrotic agent is a vitamin D receptor agonist.
18635. The method of item 18531 wherein the anti- fibrotic agent is ZD-6474 (an angiogenesis inhibitor).
18636. The method of item 18531 wherein the anti- fibrotic agent is AP-23573 (an mTOR inhibitor).
18637. The method of item 18531 wherein the anti- fibrotic agent is synthadotin (a tubulin antagonist).
18638. The method of item 18531 wherein the anti- fibrotic agent is S-0885 (a collagenase inhibitor).
18639. The method of item 18531 wherein the anti- fibrotic agent is aplidine (an elongation factor-1 alpha inhibitor). 18640. The method of item 18531 wherein the anti- fibrotic agent is ixabepilone (an epithilone).
18641. The method of item 18531 wherein the anti- fibrotic agent is IDN-5390 (an angiogenesis inhibitor).
18642. The method of item 18531 wherein the anti- fibrotic agent is SB-2723005 (an angiogenesis inhibitor).
18643. The method of item 18531 wherein the anti- fibrotic agent is ABT-518 (an angiogenesis inhibitor).
18644. The method of item 18531 wherein the anti- fibrotic agent is combretastatin (an angiogenesis inhibitor).
18645. The method of item 18531 wherein the anti- fibrotic agent is anecortave acetate (an angiogenesis inhibitor).
18646. The method of item 18531 wherein the anti- fibrotic agent is SB-715992 (a kinesin antagonist).
18647. The method of item 18531 wherein the anti- fibrotic agent is temsirolimus (an mTOR inhibitor).
18648. The method of item 18531 wherein the anti- fibrotic agent is adalimumab (a TNFα antagonist).
18649. The method of item 18531 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with an anti-fibrotic agent, and (b) implanting the medical device into the host. 18650. The method of item 18531 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with an anti-infective agent, and (b) implanting the medical device into the host.
18651. The method of item 18531 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with a polymer; and (b) implanting the medical device into the host.
18652. The method of item 18531 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with a composition comprising an anti-fibrotic agent and a polymer, and (b) implanting the medical device into the host.
18653. The method of item 18531 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with a composition comprising an anti-infective agent and a polymer, and (b) implanting the medical device into the host.
18654. The method of item 18531 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with a composition comprising an anti-fibrotic agent, an anti- infective agent and a polymer, and (b) implanting the medical device into the host.
18655. The method of item 18531 wherein the anti- infective agent is an anthracycline.
18656. The method of item 18531 wherein the anti- infective agent is doxorubicin. 18657. The method of item 18531 wherein the anti- infective agent is is mitoxantrone.
18658. The method of item 18531 wherein the anti- infective agent is a fluoropyrimidine.
18659. The method of item 18531 wherein the anti- infective agent is 5-fluorouracil (5-FU).
18660. The method of item 18531 wherein the anti- infective agent is a folic acid antagonist.
18661. The method of item 18531 wherein the anti- infective agent is methotrexate.
18662. The method of item 18531 wherein the anti- infective agent is a podophylotoxin.
18663. The method of item 18531 wherein the anti- infective agent is etoposide.
18664. The method of item 18531 wherein the anti- infective agent is camptothecin.
18665. The method of item 18531 wherein the anti- infective agent is a hydroxyurea.
18666. The method of item 18531 wherein the anti- infective agent is a platinum complex. 18667. The method of item 18531 wherein the anti- infective agent is cisplatin.
18668. The method of item 18531 wherein the composition comprises an anti-thrombotic agent.
18669. The method of item 18531 wherein the polymer is formed from reactants comprising a naturally occurring polymer.
18670. The method of item 18531 wherein the polymer is formed from reactants comprising protein.
18671. The method of item 18531 wherein the polymer is formed from reactants comprising carbohydrate.
18672. The method of item 18531 wherein the polymer is formed from reactants comprising biodegradable polymer.
18673. The method of item 18531 wherein the polymer is formed from reactants comprising nonbiodegradable polymer.
18674. The method of item 18531 wherein the polymer is formed from reactants comprising collagen.
18675. The method of item 18531 wherein the polymer is formed from reactants comprising methylated collagen.
18676. The method of item 18531 wherein the polymer is formed from reactants comprising fibrinogen. 18677. The method of item 18531 wherein the polymer reactants comprising thrombin.
18678. The method of item 18531 wherein the polymer reactants comprising blood plasma.
18679. The method of item 18531 wherein the polymer reactants comprising calcium salt.
18680. The method of item 18531 wherein the polymer reactants comprising an antifibronolytic agent.
18681. The method of item 18531 wherein the polymer reactants comprising fibrinogen analog.
18682. The method of item 18531 wherein the polymer reactants comprising albumin.
18683. The method of item 18531 wherein the polymer reactants comprising plasminogen.
18684. The method of item 18531 wherein the polymer reactants comprising von Willebrands factor.
18685. The method of item 18531 wherein the polymer reactants comprising Factor VIII.
18686. The method of item 18531 wherein the polymer reactants comprising hypoallergenic collagen. 18687. The method of item 18531 wherein the polymer is formed from reactants comprising atelopeptidic collagen.
18688. The method of item 18531 wherein the polymer is formed from reactants comprising telopeptide collagen.
18689. The method of item 18531 wherein the polymer is formed from reactants comprising crosslinked collagen.
18690. The method of item 18531 wherein the polymer is formed from reactants comprising aprotinin.
18691. The method of item 18531 wherein the polymer is formed from reactants comprising epsilon-amino-n-caproic acid.
18692. The method of item 18531 wherein the polymer . is formed from reactants comprising gelatin.
18693. The method of item 18531 wherein the polymer is formed from reactants comprising protein conjugates.
18694. The method of item 18531 wherein the polymer is formed from reactants comprising gelatin conjugates.
18695. The method of item 18531 wherein the polymer is formed from reactants comprising a synthetic polymer.
18696. The method of item 18531 wherein the polymer is formed from reactants comprising a synthetic isocyanate-containing compound. 18697. The method of item 18531 wherein the polymer is formed from reactants comprising a synthetic thiol-containing compound.
18698. The method of item 18531 wherein the polymer is formed from reactants comprising a synthetic compound containing at least two thiol groups.
18699. The method of item 18531 wherein the polymer is formed from reactants comprising a synthetic compound containing at least three thiol groups.
18700. The method of item 18531 wherein the polymer is formed from reactants comprising a synthetic compound containing at least four thiol groups.
18701. The method of item 18531 wherein the polymer is formed from reactants comprising a synthetic amino-containing compound.
18702. The method of item 18531 wherein the polymer is formed from reactants comprising a synthetic compound containing at least two amino groups.
18703. The method of item 18531 wherein the polymer is formed from reactants comprising a synthetic compound containing at least three amino groups.
18704. The method of item 18531 wherein the polymer is formed from reactants comprising a synthetic compound containing at least four amino groups. 18705. The method of item 18531 wherein the polymer is formed from reactants comprising a synthetic compound comprising a carbonyl-oxygen-succinimidyl group.
18706. The method of item 18531 wherein the polymer is formed from reactants comprising a synthetic compound comprising at least two carbonyl-oxygen-succinimidyl groups.
18707. The method of item 18531 wherein the polymer is formed from reactants comprising a synthetic compound comprising at least three carbonyl-oxygen-succinimidyl groups.
18708. The method of item 18531 wherein the polymer is formed from reactants comprising a synthetic compound comprising at least four carbonyl-oxygen-succinimidyl groups.
18709. The method of item 18531 wherein the polymer is formed from reactants comprising a synthetic polyalkylene oxide- containing compound.
18710. The method of item 18531 wherein the polymer is formed from reactants comprising a synthetic compound comprising both polyalkylene oxide and biodegradable polyester blocks.
18711. The method of item 18531 wherein the polymer is formed from reactants comprising a synthetic polyalkylene oxide- containing compound having reactive amino groups.
18712. The method of item 18531 wherein the polymer is formed from reactants comprising a synthetic polyalkylene oxide- containing compound having reactive thiol groups. 18713. The method of item 18531 wherein the polymer is formed from reactants comprising a synthetic polyalkylene oxide- containing compound having reactive carbonyl-oxygen-succinimidyl groups.
18714. The method of item 18531 wherein the polymer is formed from reactants comprising a synthetic compound comprising a biodegradable polyester block.
18715. The method of item 18531 wherein the polymer is formed from reactants comprising a synthetic polymer formed in whole or part from lactic acid or lactide.
18716. The method of item 18531 wherein the polymer is formed from reactants comprising a synthetic polymer formed in whole or part from glycolic acid or glycolide.
18717. The method of item 18531 wherein the polymer is formed from reactants comprising polylysine.
18718. The method of item 18531 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound comprising a polyalkylene oxide portion.
18719. The method of item 18531 wherein the polymer is formed from reactants comprising (a) protein and (b) polylysine.
18720. The method of item 18531 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound having at least four thiol groups. 18721. The method of item 18531 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound having at least four amino groups.
18722. The method of item 18531 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound having at least four carbonyl-oxygen-succinimide groups.
18723. The method of item 18531 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound having a biodegradable region formed from reactants selected from lactic acid, lactide, glycolic acid, glycolide, and epison-caprolactone.
18724. The method of item 18531 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound comprising a polyalkylene oxide portion.
18725. The method of item 18531 wherein the polymer is formed from reactants comprising (a) collagen and (b) polylysine.
18726. The method of item 18531 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound having at least four thiol groups.
18727. The method of item 18531 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound having at least four amino groups.
18728. The method of item 18531 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound having at least four carbonyl-oxygen-succinimide groups. 18729. The method of item 18531 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound having a biodegradable region formed from reactants selected from lactic acid, lactide, glycolic acid, glycolide, and epison-caprolactone.
18730. The method of item 18531 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound comprising a polyalkylene oxide portion.
18731. The method of item 18531 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) polylysine.
18732. The method of item 18531 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound having at least four thiol groups.
18733. The method of item 18531 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound having at least four amino groups.
18734. The method of item 18531 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound having at least four carbonyl-oxygen-succinimide groups.
18735. The method of item 18531 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound having a biodegradable region formed from reactants selected from lactic acid, lactide, glycolic acid, glycolide, and epison-caprolactone. 18736. The method of item 18531 wherein the polymer is formed from reactants comprising hyaluronic acid.
18737. The method of item 18531 wherein the polymer is formed from reactants comprising a hyaluronic acid derivative.
18738. The method of item 18531 wherein the polymer is formed from reactants comprising pentaerythritol poly(ethylene glycol)ether tetra-sulfhydryl of number average molecular weight between 3,000 and 30,000.
18739. The method of item 18531 wherein the polymer is formed from reactants comprising pentaerythritol poly(ethylene glycol)ether tetra-amino of number average molecular weight between 3,000 and 30,000.
18740. The method of item 18531 wherein the polymer is formed from reactants comprising (a) a synthetic compound having a number average molecular weight between 3,000 and 30,000 and comprising a polyalkylene oxide region and multiple nucleophilic groups, and (b) a synthetic compound having a number average molecular weight between 3,000 and 30,000 and comprising a polyalkylene oxide region and multiple electrophilic groups.
18741. The method of item 18531 wherein the composition comprises a colorant.
18742. The method of item 18531 wherein the composition is sterile. 18743. A method as in any one of items 18531-18742, wherein the device is a lacrimal duct stent.
18744. A method as in any one of items 18531-18742, wherein the device is an Eustachian tube stent.
18745. A method as in any one of items 18531-18742, wherein the device is a nasal stent.
18746. A method as in any one of items 18531-18742, wherein the device is a sinus stent.
18747. A method for implanting a medical device comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with i) an anti-fibrotic agent, ii) an anti-infective agent, iii) a polymer, iv) a composition comprising an anti-fibrotic agent and a polymer, v) a composition comprising an anti-infective agent and a polymer, or vi) a composition comprising an anti-fibrotic agent, an anti- infective agent and a polymer, and (b) implanting the medical device into the host, wherein the medical device is an ear ventilation tube.
18748. The method of item 18747 wherein the anti- fibrotic agent is an adensosine A2A receptor antagonist.
18749. The method of item 18747 wherein the anti- fibrotic agent is an AKT inhibitor.
18750. The method of item 18747 wherein the anti- fibrotic agent is an alpha 2 integrin antagonist, wherein the alpha 2 integrin antagonist is Pharmaprojects No. 5754 (Merck KgaA). 18751. The method of item 18747 wherein the anti- fibrotic agent is an alpha 4 integrin antagonist.
18752. The method of item 18747 wherein the anti- fibrotic agent is an alpha 7 nicotinic receptor agonist.
18753. The method of item 18747 wherein the anti- fibrotic agent is an angiogenesis inhibitor selected from the group consisting of AG-12,958 (Pfizer), ATN-161 (Attention LLC), neovastat, an angiogenesis inhibitor from Jerina AG (Germany), NM-3 (Mercian), VGA-1155 (Taisho), FCE-26644 (Pfizer), FCE-26950 (Pfizer), FPMA (Meiji Daries), FFM 11142 (Fujisawa), GGTI-298, GM-1306 (Ligand), GPA-1734 (Novartis), NNC-47- 0011 (Novo Nordisk), herbamycin (Nippon Kayaku), lenalidomide (Celegene), IP-10 (NIH), ABT-828 (Abbott), KIN-841 (Tokushima University, Japan), SF-1126 (Semafore Pharmaceuticals), laminin technology (NIH), CHIR-258 (Chiron), NVP-AEW541 (Novartis), NVP-AEW541 (Novartis), Vt16907 (Alchemia), OXI-8007 (Oxigene), EG-3306 (Ark Therapeutics), Maspin (AmVa), ABT-567 (Abbott), PPI-2458 (Praecis Pharmaceuticals), CC-5079, CC-4089 (Celgene), HIF-1 alpha inhibitors (Xenova), S-247 (Pfizer), AP-23573 (Ariad), AZD-9935 (Astra Zeneca), mebendazole (Introgen Therapeutics), MetAP-2 inhibitors (GlaxoSmithKline), AG-615 (Angiogene Pharmaceuticals), Tie-2 antagonists (Hybrigenics), NC-381 , CYC-381 , NC-169, NC-219, NC-383, NC-384, NC-407 (Lorus Therapeutics), ATN-224 (Attenuon), ON-01370 (Onconova), Vitronectin antagonists (Amgen), SDX-103 (Salmedix), Vitronectin antagonists (Shire), CHP (Riemser), TEK (Amgen), Anecortave acetate (Alcon), T46.2 (Matrix Therapeutics), HG-2 (Heptagen), TEM antagonists (Genzyme), Oxi-4500 (Oxigene), ATN-161 (Attenuon), WX-293 (Wilex), M-2025 (Metris Therapeutics), Alphastatin (BioActa), YH-16 (Yantai Rongchang), BIBF- 1120 (Boehringer Ingelheim), BAY-57-9352 (Bayer), AS-1404 (Cancer Research Technology), SC-77964 (Pfizer), glycomimetics (BioTie Therapies), TIE-2 Inhibitors (Ontogen), DIMI, Octamer (Octamer), ABR- 215050 (Active Biotech), ABT-518 (Abbott), KDR inhibitors (Abbott), BSF- 466895 (Abbott), SCH-221153 (Schering-Plough), DAC:antiangiogenic (ConjuChem), TFPI (EntreMed), AZD-2171 (Astra-Zenaca), CDC-394 (Celgene), LY290293 (EIi Lilly), IDN-5390 (Indena), Kdr Kinase Inhibitors (Merck), CT-113020, CT-116433, CT-116563, CT-31890, CT-32228) (Cell Therapeutics), A-299620 (Abbott), TWEAK Inhibitor (Amgen), VEGF modulators (Johnson and Johnson), Tum-N53, tumstatin (Genzyme), Thios- 1 , Thios-2 (Thios Pharmaceuticals), MV-6401 (Miravant Medical Technologies), Spisulosine (PharmaMar), CEP-7055 (Cephalon), AUV-201 (Auvation), LM-609 (EIi Lilly), SKF-106615 (AnorMED), Oglufanide disodium (Cytran), BW-114 (Pharminox), Calreticulin (NIH), WX-678 (Wilex), SD-7784 (Pfizer), WX-UK1 (Wilex), SH-268 (Schering AG), 2-Me-PGA (Celgene), S- 137 (Pfizer), ZD-6126 (Angiogene Pharmaceuticals), SG-292 (SignalGen), Benefin (Lane Labs), A6, A36 (Angstrom), SB-2723005 (GlaxoSmithKline), SC-7 (Cell Therapeutics), ZEN-014 (AEterna Zentaris), 2-methoxyestradiol (EntreMed), NK-130119 (Nippon Kayaku), CC-10004 (Celgene), AVE- 8062A (Ajinomoto), Tacedinaline (Pfizer), Actinonin (Tokyo Metropolitan Institute of Medical Science), Lenalidomide (Celgene), VGA-1155, BTO-956 (SRI International), ER-68203-00 (Eisai), CT-6685 (UCB), JKC-362 (Phoenix Pharmaceuticals), DMI-3798 (DMI Biosciences, Angiomate (Ipsen), ZD-6474 (AstraZeneca), CEP-5214 (Cephalon), Canstatin (Genzyme), NM-3 (Mercian), Oridigm (MediQuest Therapeutics), Exherin (Adherex), BLS-0597 (Boston Life Sciences), PTC-299 (PTC Therapeutics), NPI-2358 (Nereus Pharmaceuticals), CGP-79787 (Novartis), AEE-788 (Novartis), CKD-732 (Chong Kun Dang), CP-564959 (OSI Pharmaceuticals), CM-101 (CarboMed), CT-2584, CT3501 (Cell Therapeutics), combretastatin and analogues and derivatives thereof (Oxigene), Rebimastat (Bristol-Meyers Squibb), Dextrin 2-sulfate (ML Laboratories), Cilengitide (Merk KGaA), NSC-706704 (Pharminox), KRN- 951 (Kirin Brewery), Ukrain, NSC-631570 (Nowicky Pharma), Tecogalan sodium (Daiichi Pharmaceutical), Tz-93 (Tsumura), TBC-1635 (Encysive Pharmaceuticals), TAN-1120 (Takeda), Semaxanib (Pfizer), BDI-7800 (Biopharmacopae), SD-186, SD-983 (Bristol-Meyers Squibb), SB-223245 (GlaxoSmithKline), SC-236 (Pfizer), RWJ-590973 (Johnson and Johnson), ILX-1850 (Genzyme), SC-68488, S-836 (Pfizer), CG-55069-11 (CuraGen), Ki-23057 (Kirin Brewery), CCX-700 (Chemoentryx), Pegaptanib octasodium (Giled Sciences), ANGIOCOL (available from Biostratum Inc.), or an analogue or derivative thereof.
18754. The method of item 18747 wherein the anti- fibrotic agent is an apoptosis antagonist.
18755. The method of item 18747 wherein the anti- fibrotic agent is an apoptosis activator.
18756. The method of item 18747 wherein the anti- fibrotic agent is a beta 1 integrin antagonist.
18757. The method of item 18747 wherein the anti- fibrotic agent is a beta tubulin inhibitor.
18758. The method of item 18747 wherein the anti- fibrotic agent is a blocker of enzyme production in Hepatitis C.
18759. The method of item 18747 wherein the anti- fibrotic agent is a Bruton's tyrosine kinase inhibitor.
18760. The method of item 18747 wherein the anti- fibrotic agent is a calcineurin inhibitor. 18761. The method of item 18747 wherein the anti- fibrotic agent is a caspase 3 inhibitor.
18762. The method of item 18747 wherein the anti- fibrotic agent is a CC chemokine receptor antagonist.
18763. The method of item 18747 wherein the anti- fibrotic agent is a cell cycle inhibitor selected from the group consisting of SNS-595 (Sunesis), synthadotin, KRX-0403, and an analogue or derivative thereof.
18764. The method of item 18747 wherein the anti- fibrotic agent is a cathepsin B inhibitor.
18765. The method of item 18747 wherein the anti- fibrotic agent is a cathepsin K inhibitor, wherein the cathepsin K inhibitor is 462795 (GlaxoSmithKline), INPL-022-D6 (Amura Therapeutics), or an analogue or derivative thereof.
18766. The method of item 18747 wherein the anti- fibrotic agent is a cathepsin L inhibitor.
18767. The method of item 18747 wherein the anti- fibrotic agent is a CD40 antagonist.
18768. The method of item 18747 wherein the anti- fibrotic agent is a chemokine receptor agonist.
18769. The method of item 18747 wherein the anti- fibrotic agent is a chymase inhibitor. 18770. The method of item 18747 wherein the anti- fibrotic agent is a collagenase antagonist.
18771. The method of item 18747 wherein the anti- fibrotic agent is a CXCR antagonist.
18772. The method of item 18747 wherein the anti- fibrotic agent is a cyclin dependent kinase inhibitor selected from the group consisting of a CDK-1 inhibitor, a CDK-2 inhibitor, a CDK- 4 inhibitor, a CDK-6 inhibitor, a CAK1 inhibitor from GPC Biotech and Bristol-Myers Squibb, RGB-286199 (GPC Biotech), an anticancer agent from Astex Technology, a CAK1 inhibitor from GPC Biotech, a CDK inhibitor from Sanofi-Aventis, a CDK1/CDK2 inhibitor from Amgen, a CDK2 inhibitor from SUGEN-2 (Pfizer), a hearing loss therapy agent (Sound Pharmaceuticals), PD-0332991 (Pfizer), RGB-286199 (GPC Biotech), Ro-0505124 (Hoffmann- La Roche), a Ser/Thr kinase inhibitor from Lilly (EIi Lilly), CVT-2584 (CAS No. 199986-75-9) (CV Therapeutics), and an analogue or derivative thereof.
18773. The method of item 18747 wherein the anti- fibrotic agent is a cyclooxygenase 1 inhibitor.
18774. The method of item 18747 wherein the anti- fibrotic agent is a DHFR inhibitor.
18775. The method of item 18747 wherein the anti- fibrotic agent is a dual integrin inhibitor.
18776. The method of item 18747 wherein the anti- fibrotic agent is an elastase inhibitor. 18777. The method of item 18747 wherein the anti- fibrotic agent is an elongation factor-1 alpha inhibitor.
18778. The method of item 18747 wherein the anti- fibrotic agent is an endothelial growth factor antagonist.
18779. The method of item 18747 wherein the anti- fibrotic agent is an endothelial growth factor receptor kinase inhibitor selected from the group consisting of sorafenib tosylate (Bayer), AAL-993 (Novartis), ABP-309 (Novartis), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EXEL- 2880 (Exelixis), GW-654652 (GlaxoSmithKline), a KDR inhibitor from LG Life Sciences, CT-6685 and CT-6729 (UCB), KRN-633 and KRN-951 (Kirin Brewery), OSI-930 (OSI Pharmaceuticals), SP-5.2 (Supratek Pharma), SU-
11657 (Pfizer), a Tie-2 antagonist (Hybrigenics), a VEGFR-2 kinase inhibitor (Bristol-Myers Squibb), XL-647 (Exelixis), a KDR inhibitor from Abbott Laboratories, sorafenib tosylate, and an analogue or derivative thereof.
18780. The method of item 18747 wherein the anti- fibrotic agent is an endotoxin antagonist.
18781. The method of item 18747 wherein the anti- fibrotic agent is an epothilone and tubulin binder.
18782. The method of item 18747 wherein the anti- fibrotic agent is an estrogen receptor antagonist.
18783. The method of item 18747 wherein the anti- fibrotic agent is an FGF inhibitor. 18784. The method of item 18747 wherein the anti- fibrotic agent is a farnexyl transferase inhibitor.
18785. The method of item 18747 wherein the anti- fibrotic agent is famesyltransferase inhibitor selected from the group of A- 197574 (Abbott), a famesyltransferase inhibitor from Servier, FPTIII (Strathclyde Institute for Drug R), LB-42908 (LG Life Sciences), Pharmaprojects No. 5063 (Genzyme), Pharmaprojects No. 5597 (Ipsen), Yissum Project No. B-1055 (Yissum), and an analogue or derivative thereof.
18786. The method of item 18747 wherein the anti- fibrotic agent is an FLT-3 kinase inhibitor.
18787. The method of item 18747 wherein the anti- fibrotic agent is an FGF receptor kinase inhibitor.
18788. The method of item 18747 wherein the anti- fibrotic agent is a fibrinogen antagonist selected from the group consisting of AUV-201 (Auvation), MG-13926 (Sanofi-Aventis), plasminogen activator (CAS No. 105913-11-9) (from Sanofi-Aventis or UCB), plasminogen activator-2 (tPA-2) (Sanofi-Aventis), pro-urokinase (CAS No. 82657-92-9) (Sanofi-Aventis), and an analogue or derivative thereof.
18789. The method of item 18747 wherein the anti- fibrotic agent is a heat shock protein 90 antagonist selected from the group consisting of SRN-005 (Sirenade), geldanamycin, NSC-33050 (17- allylaminogeldanamycin; 17-AAG), 17-dimethylaminoethylamino-17- demethoxy-geldanamycin (17-DMAG), rifabutin (rifamycin XIV, 1',4- didehydro-1-deoxy-1,4-dihydro-5'-(2-methylpropyl)-1-oxo-), and an analogue or derivative thereof. 18790. The method of item 18747 wherein the anti- fibrotic agent is a histone deacetylase inhibitor.
18791. The method of item 18747 wherein the anti- fibrotic agent is an HMGCoA reductase inhibitor selected from the group consisting of an atherosclerosis therapeutic from Lipid Sciences, ATI-16000 (ARYx Therapeutics), KS-01-019 (Kos Pharmaceuticals), Pharmaprojects No. 2197 (Sanofi-Aventi), RP 61969 (Sanofi-Aventis), and an analogue or derivative thereof.
18792. The method of item 18747 wherein the anti- fibrotic agent is an ICAM inhibitor.
18793. The method of item 18747 wherein the anti- fibrotic agent is an IL, ICE and IRAK antagonist, wherein the antagonist is a CJ-14877, CP-424174 (Pfizer), NF-61 (Negma-Lerads), and an analogue or derivative thereof.
18794. The method of item 18747 wherein the anti- fibrotic agent is an IL-2 inhibitor.
18795. The method of item 18747 wherein the anti- fibrotic agent is an immunosuppressant selected from the group consisting of teriflunomide (Sanofi Aventis), chlorsulfaquinoxalone (NSC-339004), chlorsulfaquinoxalone sulfate, CS-712 (Sankyo), ismomultin alfa (CAS No. 457913-93-8) (Akzo Nobel), antiallergics from GenPat77, antiinflammatories or AT-005 (Androclus Therapeutics), autoimmune disease therapeutics from EpiVax, BN-007 (Bone), budesonide (CAS No. 51333-22- 3) (MAP Pharmaceuticals), CO-14 (Genzyme), edratide (CAS No. 433922- 67-9) (Teva), EP-314 (Enanta), eprovafen (CAS No. 101335-99-3) (Sanofi- Aventis), HWA-131 (CAS No. 118788-41-3) (Sanofi-Aventis), immunomodulators from MerLion Pharmaceuticals, immunosuppressives from Alchemia, IPL-12 (Inflazyme), MDL-9563 (CAS No. 27086-86-8) (Sanofi-Aventis), Pharmaprojects No. 2330 (Sanofi-Aventis), Pharmaprojects No. 6426 (Abgenix), PXS-25 (Pharmaxis), rosmarinic acid (CAS No. 20283-92-5) (Sanofi-Aventis), RP 42927 or RP 54745 (CAS No. 135330-08-4) (Sanofi-Aventis), SGN-35 (Seattle Genetics), ST-1959 (Sigma-Tau), type I diabetes therapy from SYNX Pharma, UNIL-88 (Debiopharm), VP-025 (Vasogen), VR-694 (Vectura), PRTX-001 (Protalex), and an analogue or derivative thereof.
18796. The method of item 18747 wherein the anti- fibrotic agent is an IMPDH (inosine monophosphate).
18797. The method of item 18747 wherein the anti- fibrotic agent is an integrin antagonist.
18798. The method of item 18747 wherein the anti- fibrotic agent is an interleukin antagonist.
18799. The method of item 18747 wherein the anti- fibrotic agent is an inhibitor of type III receptor tyrosine kinase.
18800. The method of item 18747 wherein the anti- fibrotic agent is an irreversible inhibitor of enzyme methionine aminopeptidase type 2.
18801. The method of item 18747 wherein the anti- fibrotic agent is an isozyme selective delta protein kinase C inhibitor.
18802. The method of item 18747 wherein the anti- fibrotic agent a JAK3 enzyme inhibitor. 18803. The method of item 18747 wherein the anti- fibrotic agent is a JNK inhibitor.
18804. The method of item 18747 wherein the anti- fibrotic agent is a kinase inhibitor.
18805. The method of item 18747 wherein the anti- fibrotic agent is kinesin antagonist.
18806. The method of item 18747 wherein the anti- fibrotic agent is a kinesin antagonist.
18807. The method of item 18747 wherein the anti- fibrotic agent is a leukotriene inhibitor and antagonist selected from the group consisting of ambicromil (CAS No. 58805-38-2) (Sanofi-Aventis), amelubant (CAS No. 346735-24-8) (Boehringer Ingelheim), DW-1141 {Dong Wha), ebselen (Daiichi Pharmaceutical), ibudilast (Kyorin), leucotriene inhibitors from Sanofi-Aventis, lymphotoxin-beta receptor (LT-D) from Biogen Idee, Pharmaprojects No. 1535 and 2728 (CAS No. 119340-33-9) (Sanofi-Aventis), R-112 (Rigel), Rev-5367 (CAS No. 92532-05-3) (Sanofi- Aventis), RG-14893 (CAS No. 141835-49-6) (Sanofi-Aventis), RG-5901-A (CAS No. 101910-24-1), 92532-23-5, RP 66153 (CAS No. 142422-79-5), RP 66364 (CAS No.186916-92-5), RP 69698 (CAS No. 141748-00-7) (Sanofi-Aventis), SC-411930 (Pfizer), SC-41930 (CAS No. 120072-59-5) (Pfizer), SC-50605 (CAS No. 138828-39-4) (Pfizer), SC-51146 (CAS No. 141059-52-1), SC-53228 (CAS No. 153633-01-3) (Pfizer), spaglumic acid (ZY-15106) (CAS No.3106-85-2), 80619-64-3 (Novartis), tipredane (CAS No. 85197-77-9) (Bristol-Myers Squibb), U-75302 (CAS No. 119477-85-9) (Pfizer), and analogue or derivative thereof. 18808. The method of item 18747 wherein the anti- fibrotic agent is an MAP kinase inhibitor.
18809. The method of item 18747 wherein the anti- fibrotic agent is a matrix metalloproteinase inhibitor.
18810. The method of item 18747 wherein the anti- fibrotic agent is an MCP-CCR2 inhibitor.
18811. The method of item 18747 wherein the anti- fibrotic agent is an mTOR inhibitor.
18812. The method of item 18747 wherein the anti- fibrotic agent is an mTOR kinase inhibitor.
18813. The method of item 18747 wherein the anti- fibrotic agent is a microtubule inhibitor selected from the group consisting of antibody-maytansinoid conjugates from Biogen Idee, colchicines (MantiCore Pharmaceuticals), anticancer immunoconjugates from Johnson & Johnson, DIME from Octamer, gni-1f (GNI), huC242-DM4, huMy9-6-DM1 (ImmunoGen), IDN-5404 (Indena), IMO-098, IMOderm (Imotep), mebendazole (Introgen Therapeutics), microtubule poisons from Cambridge Enterprise, paclitaxel such as LOTAX from Aphios (CAS No. 33069-62-4), Genexol-PM from Samyang, Pharmaprojects No. 6383 (Tapestry Pharmaceuticals), RPR-112378 (Sanofi-Aventis), SGN-75 (Seattle Genetics), SPL-7435 (Starpharma), SSR-250411 (Sanofi-Aventis), trastuzumab-DM1 (Genentech), vinorelbine, and an analogue or derivative thereof.
18814. The method of item 18747 wherein the anti- fibrotic agent is an MIF inhibitor. 18815. The method of item 18747 wherein the anti- fibrotic agent is an MMP inhibitor.
18816. The method of item 18747 wherein the anti- fibrotic agent is a neurokinin (NK) antagonist selected from the group consisting of anthrotainin (CAS No. 148084-40-6) (Sanofi-Aventis), an IBS thereapeutic from ArQuIe, MDL-105212A (CAS No. 167261-60-1) (Ssanofi- Aventis), Pharmaprojects No. 2744, 3258 (CAS No. 139167-47-8) 4006, 4201, or 5986 (Sanofi-Aventis), RP 67580 (CAS No. 135911-02-3), SR- 144190 (CAS No. 201152-86-5), SSR-240600, SSR-241586 (Sanofi- Aventis), TKA-457 (Novartis), vestipitant mesylate (CAS No. 334476-64-1) (GlaxoSmithKline), Win-64821 (Sanofi-Aventis), PRX-96026 (Predix Pharmaceuticals), and an analogue or derivative thereof.
18817. The method of item 18747 wherein the anti- fibrotic agent is an NF kappa B inhibitor selected from the group consisting of AVE-0545 or AVE-0547 (Sanofi-Aventis), bortezomib (CAS No. 179324- 69-7) (Millennium Pharmaceuticals), dexanabinol (CAS No. 112924-45-5) (Pharmos), dexlipotam (Viatris), Pharmaprojects No. 6283 (INDRA) (OXiGENE), IPL-576092 (CAS No. 137571-30-3) (Inflazyme), NFKB decoy (Corgentech), NFKB decoy oligo (AnGes MG), NFKB's from Ariad, osteoporosis treatments or S5 (F005) from Fulcrum Pharmaceuticals, P61 (Phytopharm), R-flurbiprofen (CAS No. 5104-49-4) (Encore Pharmaceuticals), and an analogue or derivative thereof.
18818. The method of item 18747 wherein the anti- fibrotic agent is a nitric oxide agonist.
18819. The method of item 18747 wherein the anti- fibrotic agent is an ornithine decarboxylase inhibitor. 18820. The method of item 18747 wherein the anti- fibrotic agent is a p38 MAP kinase inhibitor selected from the group consisting of AZD-6703 (AstraZeneca), JX-401 (Jexys Pharmaceuticals), BMS-2 (Bristol-Myers Squibb), a p38 MAP kinase inhibitor from Novartis, a p38-alpha MAP kinase inhibitor from Amphora, Pharmaprojects No. 5704 (Pharmacopeia), RPR-200765A (Sanofi-Aventis), SD-282 (Johnson & Johnson), TAK-715 (Takeda), and an analogue or derivative thereof.
18821. The method of item 18747 wherein the anti- fibrotic agent is a palmitoyl-protein thioesterase inhibitor.
18822. The method of item 18747 wherein the anti- fibrotic agent is a PDGF receptor kinase inhibitor selected from the group consisting of AAL-993, AMN-107, or ABP-309 (Novartis), AMG-706 (Amgen), BAY-57-9352 (Bayer), CDP-860 (UCB), E-7080 (Eisai), imatinib (CAS No. 152459-95-5) (Novartis), OSI-930 (OSI Pharmaceuticals), RPR- . 127963E (Sanofi-Aventis), RWJ-540973 (Johnson & Johnson), sorafenib tosylate (Bayer), SU-11657 (Pfizer), tandutinib (CAS No. 387867-13-2) (Millennium Pharmaceuticals), vatalanib (Novartis), ZK-CDK (Schering AG), and an analogue or derivative thereof.
18823. The method of item 18747 wherein the anti- fibrotic agent is (-)-halofenate (Metabolex), AMG-131 (Amgen), antidiabetics from Japan Tobacco, AZD-4619, AZD-8450, AZD-8677 (AstraZeneca), DRF-10945, balaglitazone (Dr Reddy's), CS-00088, CS-00098 (Chipscreen Biosciences), E-3030 (Eisai), etalocib (CAS No. 161172-51-6) (EIi Lilly), GSK-641597 (Ligand), GSK-677954 (GlaxoSmithKline), GW-409544 (Ligand), GW-590735 (GlaxoSmithKline), K-111 (Hoffmann-La Roche), LY- 518674 (EIi Lilly), LY-674 (Ligand), LY-929 (Ligand), MC-3001, MC-3002 (MaxoCore Pharmaceuticals), metformin HCl + pioglitazone (CAS No. 1115- 70-4 and 112529-15-4), ACTOPLUS MET from Andrx), muraglitazar (CAS No. 331741-94-7) (Bristol-Myers Squibb), naveglitazar (Ligand), oleoylethanolamide (Kadmus Pharmaceuticals), ONO-5129, pioglitazone hydrochloride (CAS No. 111025-46-8 and 112529-15-4) (Takeda), PLX-204 (Plexxikon), PPAR agonists from Genfit, PPAR delta agonists from EIi Lilly, PPAR-alpha agonists from CrystalGenomics, PPAR-gamma modulators and PPAR-β modulators from CareX, rosiglitazone maleate (CAS No. 122324- 73-4 or 155141-29-0) (GlaxoSmithKline), rosiglitazone maleate/glimepir (CAS No. 155141-29-0 and 93479-97-1), AVANDARYL, rosiglitazone maleate/metformin extend (CAS No. 155141-29-0 and 657-24-9), AVANDAMET, rosiglitazone maleate+metformin, AVANDAMET (GlaxoSmithKline), tesaglitazar (AstraZeneca), and an analogue or derivative thereof.
18824. The method of item 18747 wherein the anti- fibrotic agent is a phosphatase inhibitor.
18825. The method of item 18747 wherein the anti- fibrotic agent is a phosphodiesterase (PDE) inhibitor selected from the group consisting of avanafil (Tanabe Seiyaku), dasantafil (CAS No. 569351- 91-3) (Schering-Plough), A-906119 (CAS No. 134072-58-5), DL-850 (Sanofi-Aventis), GRC-3015, GRC-3566, GRC-3886 (Glenmark), HWA-153 (CAS No. 56395-66-5) (Sanofi-Aventis), hydroxypumafentrine (Altana), IBFB-130011 , IBFB-14-016, IBFB-140301, IBFB-150007, IBFB-211913 (IBFB Pharma), L-826141 (Merck & Co), medorinone (CAS No. 88296-61-1) (Sanofi-Aventis), MEM-1917 (Memory Pharmaceuticals), ND-1251 (Neuro3d), PDE inhibitors from ICOS, PDE IV inhibitors from Memory Pharmaceuticals and CrystalGenomics, Pharmaprojects No. 2742 and 6141 (Sanofi-Aventis), QAD-171 (Novartis), RHC-2963 (CAS No. 76993-12-9 and 76993-14-1), RPR-117658, RPR-122818 derivatives, SR-24870 , and RPR- 132294 (Sanofi-Aventis), SK-350 (ln2Gen), stroke therapy agents from deCODE Genetics, TAS-203 (Taiho), tofimilast (CAS No. 185954-27-2) (Pfizer), UK-371800 (Pfizer), WIN-65579 (CAS No. 158020-82-7) (Sanofi- Aventis), IBFB-130020 (IBFB Pharma), OPC-6535 (CAS No. 145739-56-6) (Otsuka), a phosphodiesterase III inhibitor, enoximone, a phosphodiesterase IV inhibitor, fosfosal, Atopik (Barrier Therapeutics), triflusal, a phosphodiesterase V inhibitor, and an analogue or derivative thereof.
18826. The method of item 18747 wherein the anti- fibrotic agent is a PKC inhibitor.
18827. The method of item 18747 wherein the anti- fibrotic agent is a platelet activating factor antagonist.
18828. The method of item 18747 wherein the anti- fibrotic agent is a platelet-derived growth factor receptor kinase inhibitor.
18829. The method of item 18747 wherein the anti- fibrotic agent is a prolyl hydroxylase inhibitor.
18830. The method of item 18747 wherein the anti- fibrotic agent is a polymorphonuclear neutrophil inhibitor.
18831. The method of item 18747 wherein the anti- fibrotic agent is a protein kinase B inhibitor.
18832. The method of item 18747 wherein the anti- fibrotic agent is a protein kinase C stimulant.
18833. The method of item 18747 wherein the anti- fibrotic agent is a purine nucleoside analogue. 18834. The method of item 18747 wherein the anti- fibrotic agent is a purinoreceptor P2X antagonist.
18835. The method of item 18747 wherein the anti- fibrotic agent is a Raf kinase inhibitor.
18836. The method of item 18747 wherein the anti- fibrotic agent is a reversible inhibitor of ErbB1 and ErbB2.
18837. The method of item 18747 wherein the anti- fibrotic agent is a ribonucleoside triphosphate reductase inhibitor.
18838. The method of item 18747 wherein the anti- fibrotic agent is an SDF-1 antagonist.
18839. The method of item 18747 wherein the anti- fibrotic agent is a sheddase inhibitor.
18840. The method of item 18747 wherein the anti- fibrotic agent is an SRC inhibitor.
18841. The method of item 18747 wherein the anti- fibrotic agent is a stromelysin inhibitor.
18842. The method of item 18747 wherein the anti- fibrotic agent is an Syk kinase inhibitor.
18843. The method of item 18747 wherein the anti- fibrotic agent is a telomerase inhibitor. 18844. The method of item 18747 wherein the anti- fibrotic agent is a TGF beta inhibitor selected from the group consisting of pirfenidone (CAS No. 53179-13-8) (MARNAC), tranilast (CAS No. 53902- 12-8) (Kissei), IN-1130 (ln2Gen), mannose-6-phosphate (BTG), TGF-β antagonists from Inflazyme (Pharmaprojects No. 6075), TGF-β antagonists from Sydney, non-industrial source), TGF-β I receptor kinase inhibitors from EIi Lilly, TGF-β receptor inhibitors from Johnson & Johnson, and an analogue or derivative thereof.
18845. The method of item 18747 wherein the anti- fibrotic agent is a TNFα antagonist or TACE inhibitor selected from the group consisting of adalimumab (CAS No. 331731-18-1) (Cambridge Antibody Technology), AGIX-4207 (AtheroGenics), AGT-1 (Advanced Biotherapy), an anti-inflammatory from Borean Pharma, Cellzome, or Paradigm Therapeutics, anti-inflammatory vaccine (TNF-alpha kinoid) from Neovacs, humanized anti-TNF antibody or an anti-TNF MAb (CB0006) Celltech (UCB), apratastat (CAS No. 287445-51-0) (Wyeth), BMS-561392 (Bristol-Myers Squibb), BN-006 (Bone), certolizumab pegol (CAS No. 428863-50-7 or CH-138 (UCB), cilomilast (CAS No. 153259-65-5) (GlaxoSmithKline), CR-1 (Nuada Pharmaceuticals), CRx-119 (CombinatoRx), D-5410 (UCB), dacopafant (CAS No. 125372-33-0) (Sanofi- Aventis), dersalazine (CAS No. 188913-57-7/188913-58-8) (Uriach), etanercept (CAS No. 185243-69-0) (Amgen), ethyl pyruvate (Critical (Critical Therapeutics), golimumab (CAS No. 476181-74-5) (Johnson & Johnson), hormono-immunotherapy from Ipsen, CDP571 (e.g., Humicade from UCB), IC-485 (ICOS), infliximab (CAS No. 170277-31-3) (Johnson & Johnson), IP- 751 (Manhattan Pharmaceuticals), ISIS-104838 (CAS No. 250755-32-9) (ISIS Pharmaceuticals), lenalidomide (CAS No. 191732-72-6) (Celgene), lentinan (CAS No. 37339-90-5) (Ajinomoto), MDL-201112 (CAS No. 142130-73-2) (Sanofi-Aventis), medroxyprogesterone (CAS No. 520-85-4) (InKine Pharmaceutical), N-acetylcysteine (CAS No. 616-91-1) (Zambon), NBE-P2 (DIREVO Biotech), nerelimomab (CAS No. 162774-06-3) (Chiron), OM-294DP (OM PHARMA), onercept (CAS No. 199685-57-9) (Yeda), PASSTNF-alpha (Verigen), pentoxifylline or oxypentifylline (Sanofi-Aventis), Pharmaprojects No. 4091 , 4241, 4295, or 4488 (Sanofi-Aventis), Pharmaprojects No. 5480 (Amgen), Pharmaprojects No. 6749 (Cengent), pirfenidone (CAS No. 53179-13-8) (MARNAC), RPR-132294 (Sanofi- Aventis), S5 (F002) (Fulcrum Pharmaceuticals), simvastatin (CAS No. 79902-63-9) (Merck & Co), STA-6292 (Synta Pharmaceuticals), tacrolimus (CAS No. 104987-11-3) (Fujisawa LifeCycle Pharma), talactoferrin alfa (CAS No. 308240-58-6) (Agennix), thalidomide (CAS No. 50-35-1) (Celgene), TNF antagonists form ProStrakan, and Synergen, TNF inhibitors (Amgen), TNF-alpha antagonists from Dynavax Technologies and Jerina AG (Germany), TNF-alpha inhibitors from IBFB Pharma and Xencor (Xencor), torbafylline (CAS No. 105102-21-4) (Sanofi-Aventis), UR-1505 (Uriach), VT-346 (Viron Therapeutics), YSIL6 (Y's Therapeutics), YSTH2 (Y's Therapeutics), NPI-1302a-3 (Nereus Pharmaceuticals, a TNF antagonist from Jerina AG (Germany), dersalazine, and an analogue or derivative thereof.
18846. The method of item 18747 wherein the anti- fibrotic agent is a Toll receptor inhibitor.
18847. The method of item 18747 wherein the anti- fibrotic agent is a tubulin antagonist.
18848. The method of item 18747 wherein the anti- fibrotic agent is a tyrosine kinase inhibitor selected from the group consisting of SU-011248, SUTENT from Pfizer Inc. (New York, NY), BMS-354825, PN- 355 (Paracelsian Pharmaceuticals), AGN-199659 (Allergan), AAL-993 or ABP-309 (Novartis), adaphostin (NIH), AEE-788 (Novartis), AG-013736 (OSI Pharmaceuticals), AG-13736 (Pfizer), ALT-110 (Alteris Therapeutics), AMG-706 (Amgen), anticancer MAbs from Xencor, anti-EGFrvlll MAbs from Abgenix, anti-HER2 MAb from Abiogen, AZD-2171 or A2D-9935 (AstraZeneca), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), CEP-5214 (Cephalon), CEP-7055 (Cephalon), cetuximab (ImCIone Systems), CHIR-200131 and CHIR-258 (Chiron), CP-547632 (OSI Pharmaceuticals), CP-724714 (Pfizer), CT-301 (Creabilis Therapeutics), D- 69491 (Baxter International), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EGFR/ErbB2 inhibitors from Array BioPharma, erlotinib (CAS No. 183319- 69-9) (OSI Pharmaceuticals), EXEL-2880 (Exelixis), FK-778 (Sanofi- Aventis) , gefitinib (CAS No. 184475-35-2) (AstraZeneca), GW-2286 or GW- 654652 (GlaxoSmithKline), her2/neu antigen from AlphaVax, HER-2/neu inhibitor from Generex, Herzyme (Medipad) (Sirna Therapeutics), HKI-272 (Wyeth), HuMax-EGFr (Genmab), idronoxil (CAS No. 81267-65-4) (Novogen), IGF-1 inhibitors from Ontogen, IMC-11F8 (ImCIone Systems), kahalalide F (CAS No. 149204-42-2) (PharmaMar), KDR inhibitor from LG Life Sciences, KDR inhibitors from Abbott Laboratories, KDR kinase inhibitors (UCB), Kdr kinase inhibitors from Merck & Co, KRN-633 and KRN- 951 (Kirin Brewery), KSB-102 (Xenova), lapatinib ditosylate (CAS No. 388082-78-8) (GlaxoSmithKline), matuzumab (Merck KGaA), MDX-214 (Medarex), ME-103 (Pharmexa), MED-A300 (Gerolymatos), MNAC-13 (Lay Line Genomics), nimotuzumab (Center of Molecular Immunology), NSC- 330507 or NSC-707545 (NIH), NV-50 (Novogen), OSI-930 (OSI Pharmaceuticals), panitumumab (Abgenix), pelitinib (CAS No. 287933-82-7) (Wyeth), pertuzumab (CAS No. 380610-27-5) (Genentech), Pharmaprojects No. 3985 (Sanofi-Aventis), prostate cancer therapeutics from Sequenom (SQPC35, SQPC36, SQPC90), removab and remoxab (Trion Pharma), RG- 13022 (CAS No. 136831-48-6), RG-13291 (CAS No. 138989-50-1), or RG- 14620 (CAS No. 136831-49-7) (Sanofi-Aventis), RM-6427 (Romark), RNAi breast cancer therapy from Benitec, RP 53801 (CAS No. 125882-88-4) (Sanofi-Aventis), sorafenib tosylate (Bayer), SU-11657 (Pfizer), Tie-2 antagonists from Semaia (Hybrigenics), Tie-2 inhibitors from Ontogen, trastuzumab (CAS No. 180288-69-1) (Genentech), tyrosine kinase inhibitors from Sanofi-Aventis, U3-1287, U3-1565, U3-1784, U3-1800 (U3 Pharma), vatalanib (Novartis), VEGFR-2 kinase inhibitor from Bristol-Myers Squibb, XL-647 (Exelixis), ZD-6474 (AstraZeneca), ZK-CDK (Schering AG), an EGFR tyrosine kinase inhibitor, EKB-569 (Wyeth), and an analogue or derivative thereof.
18849. The method of item 18747 wherein the anti- fibrotic agent is a VEGF inhibitor.
18850. The method of item 18747 wherein the anti- fibrotic agent is a vitamin D receptor agonist.
18851. The method of item 18747 wherein the anti- fibrotic agent is ZD-6474 (an angiogenesis inhibitor).
18852. The method of item 18747 wherein the anti- fibrotic agent is AP-23573 (an mTOR inhibitor).
18853. The method of item 18747 wherein the anti- fibrotic agent is synthadotin (a tubulin antagonist).
18854. The method of item 18747 wherein the anti- fibrotic agent is S-0885 (a collagenase inhibitor).
18855. The method of item 18747 wherein the anti- fibrotic agent is aplidine (an elongation factor-1 alpha inhibitor).
18856. The method of item 18747 wherein the anti- fibrotic agent is ixabepilone (an epithilone). 18857. The method of item 18747 wherein the anti- fibrotic agent is IDN-5390 (an angiogenesis inhibitor).
18858. The method of item 18747 wherein the anti- fibrotic agent is SB-2723005 (an angiogenesis inhibitor).
18859. The method of item 18747 wherein the anti- fibrotic agent is ABT-518 (an angiogenesis inhibitor).
18860. The method of item 18747 wherein the anti- fibrotic agent is combretastatin (an angiogenesis inhibitor).
18861. The method of item 18747 wherein the anti- fibrotic agent is anecortave acetate (an angiogenesis inhibitor).
18862. The method of item 18747 wherein the anti- fibrotic agent is SB-715992 (a kinesin antagonist).
18863. The method of item 18747 wherein the anti- fibrotic agent is temsirolimus (an mTOR inhibitor).
18864. The method of item 18747 wherein the anti- fibrotic agent is adalimumab (a TNFα antagonist).
18865. The method of item 18747 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with an anti-fibrotic agent, and (b) implanting the medical device into the host.
18866. The method of item 18747 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with an anti-infective agent, and (b) implanting the medical device into the host.
18867. The method of item 18747 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with a polymer; and (b) implanting the medical device into the host.
18868. The method of item 18747 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with a composition comprising an anti-fibrotic agent and a polymer, and (b) implanting the medical device into the host.
18869. The method of item 18747 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with a composition comprising an anti-infective agent and a . . _ polymer, and (b) implanting the medical device into the host.
18870. The method of item 18747 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with a composition comprising an anti-fibrotic agent, an anti- infective agent and a polymer, and (b) implanting the medical device into the host.
18871. The method of item 18747 wherein the anti- infective agent is an anthracycline.
18872. The method of item 18747 wherein the anti- infective agent is doxorubicin. 18873. The method of item 18747 wherein the anti- infective agent is is mitoxantrone.
18874. The method of item 18747 wherein the anti- infective agent is a fluoropyrimidine.
18875. The method of item 18747 wherein the anti- infective agent is 5-fluorouracil (5-FU).
18876. The method of item 18747 wherein the anti- infective agent is a folic acid antagonist.
18877. The method of item 18747 wherein the anti- infective agent is methotrexate.
18878. The method of item 18747 wherein the anti- infective agent is a podophylotoxin.
18879. The method of item 18747 wherein the anti- infective agent is etoposide.
18880. The method of item 18747 wherein the anti- infective agent is camptothecin.
18881. The method of item 18747 wherein the anti- infective agent is a hydroxyurea.
18882. The method of item 18747 wherein the anti- infective agent is a platinum complex. 18883. The method of item 18747 wherein the anti- infective agent is cisplatin.
18884. The method of item 18747 wherein the composition comprises an antithrombotic agent.
18885. The method of item 18747 wherein the polymer is formed from reactants comprising a naturally occurring polymer.
18886. The method of item 18747 wherein the polymer is formed from reactants comprising protein.
18887. The method of item 18747 wherein the polymer is formed from reactants comprising carbohydrate.
18888. The method of item 18747 wherein the polymer is formed from reactants comprising biodegradable polymer.
18889. The method of item 18747 wherein the polymer is formed from reactants comprising nonbiodegradable polymer.
18890. The method of item 18747 wherein the polymer is formed from reactants comprising collagen.
18891. The method of item 18747 wherein the polymer is formed from reactants comprising methylated collagen.
18892. The method of item 18747 wherein the polymer is formed from reactants comprising fibrinogen. 30
18893. The method of item 18747 wherein the polymer reactants comprising thrombin.
18894. The method of item 18747 wherein the polymer reactants comprising blood plasma.
18895. The method of item 18747 wherein the polymer reactants comprising calcium salt.
18896. The method of item 18747 wherein the polymer reactants comprising an antifibronolytic agent.
18897. The method of item 18747 wherein the polymer reactants comprising fibrinogen analog.
18898. The method of item 18747 wherein the polymer reactants comprising albumin.
18899. The method of item 18747 wherein the polymer reactants comprising plasminogen.
18900. The method of item 18747 wherein the polymer reactants comprising von Willebrands factor.
18901. The method of item 18747 wherein the polymer reactants comprising Factor VIII.
18902. The method of item 18747 wherein the polymer reactants comprising hypoallergenic collagen. 18903. The method of item 18747 wherein the polymer is formed from reactants comprising atelopeptidic collagen.
18904. The method of item 18747 wherein the polymer is formed from reactants comprising telopeptide collagen.
18905. The method of item 18747 wherein the polymer is formed from reactants comprising crosslinked collagen.
18906. The method of item 18747 wherein the polymer is formed from reactants comprising aprotinin.
18907. The method of item 18747 wherein the polymer is formed from reactants comprising epsilon-amino-n-caproic acid.
18908. The method of item 18747 wherein the polymer is formed from reactants comprising gelatin.
18909. The method of item 18747 wherein the polymer is formed from reactants comprising protein conjugates.
18910. The method of item 18747 wherein the polymer is formed from reactants comprising gelatin conjugates.
18911. The method of item 18747 wherein the polymer is formed from reactants comprising a synthetic polymer.
18912. The method of item 18747 wherein the polymer is formed from reactants comprising a synthetic isocyanate-containing compound. 18913. The method of item 18747 wherein the polymer is formed from reactants comprising a synthetic thiol-containing compound.
18914. The method of item 18747 wherein the polymer is formed from reactants comprising a synthetic compound containing at least two thiol groups.
18915. The method of item 18747 wherein the polymer is formed from reactants comprising a synthetic compound containing at least three thiol groups.
18916. The method of item 18747 wherein the polymer is formed from reactants comprising a synthetic compound containing at least four thiol groups.
18917. The method of item 18747 wherein the polymer is formed from reactants comprising a synthetic amino-containing compound.
18918. The method of item 18747 wherein the polymer is formed from reactants comprising a synthetic compound containing at least two amino groups.
18919. The method of item 18747 wherein the polymer is formed from reactants comprising a synthetic compound containing at least three amino groups.
18920. The method of item 18747 wherein the polymer is formed from reactants comprising a synthetic compound containing at least four amino groups. 18921. The method of item 18747 wherein the polymer is formed from reactants comprising a synthetic compound comprising a carbonyi-oxygen-succinimidyl group.
18922. The method of item 18747 wherein the polymer is formed from reactants comprising a synthetic compound comprising at least two carbonyi-oxygen-succinimidyl groups.
18923. The method of item 18747 wherein the polymer is formed from reactants comprising a synthetic compound comprising at least three carbonyi-oxygen-succinimidyl groups.
18924. The method of item 18747 wherein the polymer is formed from reactants comprising a synthetic compound comprising at least four carbonyi-oxygen-succinimidyl groups.
18925. The method of item 18747 wherein the polymer is formed from reactants comprising a synthetic polyalkylene oxide- containing compound.
18926. The method of item 18747 wherein the polymer is formed from reactants comprising a synthetic compound comprising both polyalkylene oxide and biodegradable polyester blocks.
18927. The method of item 18747 wherein the polymer is formed from reactants comprising a synthetic polyalkylene oxide- containing compound having reactive amino groups.
18928. The method of item 18747 wherein the polymer is formed from reactants comprising a synthetic polyalkylene oxide- containing compound having reactive thiol groups. 18929. The method of item 18747 wherein the polymer is formed from reactants comprising a synthetic polyalkylene oxide- containing compound having reactive carbonyl-oxygen-succinimidyl groups.
18930. The method of item 18747 wherein the polymer is formed from reactants comprising a synthetic compound comprising a biodegradable polyester block.
18931. The method of item 18747 wherein the polymer is formed from reactants comprising a synthetic polymer formed in whole or part from lactic acid or lactide.
18932. The method of item 18747 wherein the polymer is formed from reactants comprising a synthetic polymer formed in whole or part from glycolic acid or glycolide.
18933. The method of item 18747 wherein the polymer is formed from reactants comprising polylysine.
18934. The method of item 18747 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound comprising a polyalkylene oxide portion.
18935. The method of item 18747 wherein the polymer is formed from reactants comprising (a) protein and (b) polylysine.
18936. The method of item 18747 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound having at least four thiol groups. 18937. The method of item 18747 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound having at least four amino groups.
18938. The method of item 18747 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound having at least four carbonyl-oxygen-succinimide groups.
18939. The method of item 18747 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound having a biodegradable region formed from reactants selected from lactic acid, lactide, glycolic acid, glycolide, and epison-caprolactone.
18940. The method of item 18747 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound comprising a polyalkylene oxide portion.
18941. The method of item 18747 wherein the polymer is formed from reactants comprising (a) collagen and (b) polylysine.
18942. The method of item 18747 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound having at least four thiol groups.
18943. The method of item 18747 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound having at least four amino groups.
18944. The method of item 18747 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound having at least four carbonyl-oxygen-succinimide groups. 18945. The method of item 18747 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound having a biodegradable region formed from reactants selected from lactic acid, lactide, glycolic acid, glycolide, and epison-caprolactone.
18946. The method of item 18747 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound comprising a polyalkylene oxide portion.
18947. The method of item 18747 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) polylysine.
18948. The method of item 18747 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound having at least four thiol groups.
18949. The method of item 18747 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound having at least four amino groups.
18950. The method of item 18747 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound having at least four carbonyl-oxygen-succinimide groups.
18951. The method of item 18747 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound having a biodegradable region formed from reactants selected from lactic acid, lactide, glycolic acid, glycolide, and epison-caprolactone. 18952. The method of item 18747 wherein the polymer is formed from reactants comprising hyaluronic acid.
18953. The method of item 18747 wherein the polymer is formed from reactants comprising a hyaluronic acid derivative.
18954. The method of item 18747 wherein the polymer is formed from reactants comprising pentaerythritol poly(ethylene glycol)ether tetra-sulfhydryl of number average molecular weight between 3,000 and 30,000.
18955. The method of item 18747 wherein the polymer is formed from reactants comprising pentaerythritol poly(ethylene glycol)ether tetra-amino of number average molecular weight between 3,000 and 30,000.
18956. The method of item 18747 wherein the polymer is formed from reactants comprising (a) a synthetic compound having a number average molecular weight between 3,000 and 30,000 and comprising a polyalkylene oxide region and multiple nucleophilic groups, and (b) a synthetic compound having a number average molecular weight between 3,000 and 30,000 and comprising a polyalkylene oxide region and multiple electrophilic groups.
18957. The method of item 18747 wherein the composition comprises a colorant.
18958. The method of item 18747 wherein the composition is sterile. 18959. A method as in any one of items 18747-18958, wherein the device is a grommet shaped tube.
18960. A method as in any one of items 18747-18958, wherein the device is a T-tube.
18961. A method as in any one of items 18747-18958, wherein the device is a tympanostomy tube.
18962. A method as in any one of items 18747-18958, wherein the device is a drain tube.
18963. A method as in any one of items 18747-18958, wherein the device is a tympanic tube.
18964. A method as in any one of items 18747-18958, wherein the device is an otological tube.
18965. A method as in any one of items 18747-18958, wherein the device is a myringotomy tube.
18966. A method as in any one of items 18747-18958, wherein the device is an artificial Eustachian tube.
18967. A method as in any one of items 18747-18958, wherein the device is a medical device is an Eustachian tube prosthesis.
18968. A method as in any one of items 18747-18958, wherein the device is an Eustachian stent. 18969. A method for implanting a medical device comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with i) an anti-fibrotic agent, ii) an anti-infective agent, iii) a polymer, iv) a composition comprising an anti-fibrotic agent and a polymer, v) a composition comprising an anti-infective agent and a polymer, or vi) a composition comprising an anti-fibrotic agent, an anti- infective agent and a polymer, and (b) implanting the medical device into the host, wherein the medical device is an intraocular implant.
18970. The method of item 18969 wherein the anti- fibrotic agent is an adensosine A2A receptor antagonist.
18971. The method of item 18969 wherein the anti- fibrotic agent is an AKT inhibitor.
18972. The method of item 18969 wherein the anti- fibrotic agent is an alpha 2 integrin antagonist, wherein the alpha 2 integrin antagonist is Pharmaprojects No. 5754 (Merck KgaA).
18973. The method of item 18969 wherein the anti- fibrotic agent is an alpha 4 integrin antagonist.
18974. The method of item 18969 wherein the anti- fibrotic agent is an alpha 7 nicotinic receptor agonist.
18975. The method of item 18969 wherein the anti- fibrotic agent is an angiogenesis inhibitor selected from the group consisting of AG-12,958 (Pfizer), ATN-161 (Attenuon LLC), neovastat, an angiogenesis inhibitor from Jerina AG (Germany), NM-3 (Mercian), VGA-1155 (Taisho), FCE-26644 (Pfizer), FCE-26950 (Pfizer), FPMA (Meiji Daries), FR-111142 (Fujisawa), GGTI-298, GM-1306 (Ligand), GPA-1734 (Novartis), NNC-47- 0011 (Novo Nordisk), herbamycin (Nippon Kayaku), lenalidomide (Celegene), IP-10 (NIH), ABT-828 (Abbott), KIN-841 (Tokushima University, Japan), SF-1126 (Semafore Pharmaceuticals), laminin technology (NIH), CHIR-258 (Chiron), NVP-AEW541 (Novartis), NVP-AEW541 (Novartis), Vt16907 (Alchemia), OXI-8007 (Oxigene), EG-3306 (Ark Therapeutics), Maspin (Arriva), ABT-567 (Abbott), PPI-2458 (Praecis Pharmaceuticals), CC-5079, CC-4089 (Celgene), HIF-1 alpha inhibitors (Xenova), S-247 (Pfizer), AP-23573 (Ariad), AZD-9935 (Astra Zeneca), mebendazole (Introgen Therapeutics), MetAP-2 inhibitors (GlaxoSmithKline), AG-615 (Angiogene Pharmaceuticals), Tie-2 antagonists (Hybrigenics), NC-381 , CYC-381 , NC-169, NC-219, NC-383, NC-384, NC-407 (Lorus Therapeutics), ATN-224 (Attenuon), ON-01370 (Onconova), Vitronectin antagonists (Amgen), SDX-103 (Salmedix), Vitronectin antagonists (Shire), CHP (Riemser), TEK (Amgen), Anecortave acetate (Alcon), T46.2 (Matrix Therapeutics), HG-2 (Heptagen), TEM antagonists (Genzyme), Oxi-4500 (Oxigene), ATN-161 (Attenuon), WX-293 (Wilex), M-2025 (Metris Therapeutics), Alphastatin (BioActa), YH-16 (Yantai Rongchang), BIBF- 1120 (Boehringer Ingelheim), BAY-57-9352 (Bayer), AS-1404 (Cancer Research Technology), SC-77964 (Pfizer), glycomimetics (BioTie Therapies), TIE-2 Inhibitors (Ontogen), DIMI, Octamer (Octamer), ABR- 215050 (Active Biotech), ABT-518 (Abbott), KDR inhibitors (Abbott), BSF- 466895 (Abbott), SCH-221153 (Schering-Plough), DAC:antiangiogenic (ConjuChem), TFPI (EntreMed), AZD-2171 (Astra-Zenaca), CDC-394 (Celgene), LY290293 (EIi Lilly), IDN-5390 (Indena), Kdr Kinase Inhibitors (Merck), CT-113020, CT-116433, CT-116563, CT-31890, CT-32228) (Cell Therapeutics), A-299620 (Abbott), TWEAK Inhibitor (Amgen), VEGF modulators (Johnson and Johnson), Tum-N53, tumstatin (Genzyme), Thios- 1 , Thios-2 (Thios Pharmaceuticals), MV-6401 (Miravant Medical Technologies), Spisulosine (PharmaMar), CEP-7055 (Cephalon), AUV-201 (Auvation), LM-609 (EIi Lilly), SKF-106615 (AnorMED), Oglufanide disodium (Cytran), BW-114 (Pharminox), Calreticulin (NIH), WX-678 (Wilex), SD-7784 (Pfizer), WX-UK1 (Wilex), SH-268 (Schering AG), 2-Me-PGA (Celgene), S- 137 (Pfizer), ZD-6126 (Angiogene Pharmaceuticals), SG-292 (SignalGen), Benefin (Lane Labs), A6, A36 (Angstrom), SB-2723005 (GlaxoSmithKline), SC-7 (Cell Therapeutics), ZEN-014 (AEterna Zentaris), 2-methoxyestradiol (EntreMed), NK-130119 (Nippon Kayaku), CC-10004 (Celgene), AVE- 8062A (Ajinomoto), Tacedinaline (Pfizer), Actinonin (Tokyo Metropolitan Institute of Medical Science), Lenalidomide (Celgene), VGA-1155, BTO-956 (SRI International), ER-68203-00 (Eisai), CT-6685 (UCB), JKC-362 (Phoenix Pharmaceuticals), DMI-3798 (DMI Biosciences, Angiomate (Ipsen), ZD-6474 (AstraZeneca), CEP-5214 (Cephalon), Canstatin (Genzyme), NM-3 (Mercian), Oridigm (MediQuest Therapeutics), Exherin (Adherex), BLS-0597 (Boston Life Sciences), PTC-299 (PTC Therapeutics), NPI-2358 (Nereus Pharmaceuticals), CGP-79787 (Novartis), AEE-788 (Novartis), CKD-732 (Chong Kun Dang), CP-564959 (OSI Pharmaceuticals), CM-101 (CarboMed), CT-2584, CT3501 (Cell Therapeutics), combretastatin and analogues and derivatives thereof (Oxigene), Rebimastat (Bristol-Meyers Squibb), Dextrin 2-sulfate (ML Laboratories), Cilengitide (Merk KGaA), NSC-706704 (Pharminox), KRN- 951 (Kirin Brewery), Ukrain, NSC-631570 (Nowicky Pharma), Tecogalan sodium (Daiichi Pharmaceutical), Tz-93 (Tsumura), TBC-1635 (Encysive Pharmaceuticals), TAN-1120 (Takeda), Semaxanib (Pfizer), BDI-7800 (Biopharmacopae), SD-186, SD-983 (Bristol-Meyers Squibb), SB-223245 (GlaxoSmithKline), SC-236 (Pfizer), RWJ-590973 (Johnson and Johnson), ILX-1850 (Genzyme), SC-68488, S-836 (Pfizer), CG-55069-11 (CuraGen), Ki-23057 (Kirin Brewery), CCX-700 (Chemoentryx), Pegaptanib octasodium (Giled Sciences), ANGIOCOL (available from Biostratum Inc.), or an analogue or derivative thereof.
18976. The method of item 18969 wherein the anti- fibrotic agent is an apoptosis antagonist. 18977. The method of item 18969 wherein the anti- fibrotic agent is an apoptosis activator.
18978. The method of item 18969 wherein the anti- fibrotic agent is a beta 1 integrin antagonist.
18979. The method of item 18969 wherein the anti- fibrotic agent is a beta tubulin inhibitor.
18980. The method of item 18969 wherein the anti- fibrotic agent is a blocker of enzyme production in Hepatitis C.
18981. The method of item 18969 wherein the anti- fibrotic agent is a Bruton's tyrosine kinase inhibitor.
18982. The method of item 18969 wherein the anti- fibrotic agent is a calcineurin inhibitor.
18983. The method of item 18969 wherein the anti- fibrotic agent is a caspase 3 inhibitor.
18984. The method of item 18969 wherein the anti- fibrotic agent is a CC chemokine receptor antagonist.
18985. The method of item 18969 wherein the anti- fibrotic agent is a cell cycle inhibitor selected from the group consisting of SNS-595 (Sunesis), synthadotin, KRX-0403, and an analogue or derivative thereof.
18986. The method of item 18969 wherein the anti- fibrotic agent is a cathepsin B inhibitor. 18987. The method of item 18969 wherein the anti- fibrotic agent is a cathepsin K inhibitor, wherein the cathepsin K inhibitor is 462795 (GlaxoSmithKline), INPL-022-D6 (Amura Therapeutics), or an analogue or derivative thereof.
18988. The method of item 18969 wherein the anti- fibrotic agent is a cathepsin L inhibitor.
18989. - The method of item 18969 wherein the anti- fibrotic agent is a CD40 antagonist.
18990. The method of item 18969 wherein the anti- fibrotic agent is a chemokine receptor agonist.
18991. The method of item 18969 wherein the anti- fibrotic agent is a chymase inhibitor. ^
18992. The method of item 18969 wherein the anti- fibrotic agent is a collagenase antagonist.
18993. The method of item 18969 wherein the anti- fibrotic agent is a CXCR antagonist.
18994. The method of item 18969 wherein the anti- fibrotic agent is a cyclin dependent kinase inhibitor selected from the group consisting of a CDK-1 inhibitor, a CDK-2 inhibitor, a CDK- 4 inhibitor, a CDK-6 inhibitor, a CAK1 inhibitor from GPC Biotech and Bristol-Myers Squibb, RGB-286199 (GPC Biotech), an anticancer agent from Astex Technology, a CAK1 inhibitor from GPC Biotech, a CDK inhibitor from Sanofi-Aventis, a CDK1/CDK2 inhibitor from Amgen, a CDK2 inhibitor from SUGEN-2 (Pfizer), a hearing loss therapy agent (Sound Pharmaceuticals), PD-0332991 (Pfizer), RGB-286199 (GPC Biotech), Ro-0505124 (Hoffmann- La Roche), a Ser/Thr kinase inhibitor from Lilly (EIi Lilly), CVT-2584 (CAS No. 199986-75-9) (CV Therapeutics), and an analogue or derivative thereof.
18995. The method of item 18969 wherein the anti- fibrotic agent is a cyclooxygenase 1 inhibitor.
18996. The method of item 18969 wherein the anti- fibrotic agent is a DHFR inhibitor.
18997. The method of item 18969 wherein the anti- fibrotic agent is a dual integrin inhibitor.
18998. The method of item 18969 wherein the anti- fibrotic agent is an elastase inhibitor.
18999. The method of item 18969 wherein the anti- fibrotic agent is an elongation factor-1 alpha inhibitor.
19000. The method of item 18969 wherein the anti- fibrotic agent is an endothelial growth factor antagonist.
19001. The method of item 18969 wherein the anti- fibrotic agent is an endothelial growth factor receptor kinase inhibitor selected from the group consisting of sorafenib tosylate (Bayer), AAL-993 (Novartis), ABP-309 (Novartis), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EXEL- 2880 (Exelixis), GW-654652 (GlaxoSmithKline), a KDR inhibitor from LG Life Sciences, CT-6685 and CT-6729 (UCB), KRN-633 and KRN-951 (Kirin Brewery), OSI-930 (OSI Pharmaceuticals), SP-5.2 (Supratek Pharma), SU-
11657 (Pfizer), a Tie-2 antagonist (Hybrigenics), a VEGFR-2 kinase inhibitor (Bristol-Myers Squibb), XL-647 (Exelixis), a KDR inhibitor from Abbott Laboratories, sorafenib tosylate, and an analogue or derivative thereof.
19002. The method of item 18969 wherein the anti- fibrotic agent is an endotoxin antagonist.
19003. The method of item 18969 wherein the anti- fibrotic agent is an epothilone and tubulin binder.
19004. The method of item 18969 wherein the anti- fibrotic agent is an estrogen receptor antagonist.
19005. The method of item 18969 wherein the anti- fibrotic agent is an FGF inhibitor.
19006. The method of item 18969 wherein the anti- fibrotic agent is a farnexyl transferase inhibitor.
19007. The method of item 18969 wherein the anti- fibrotic agent is farnesyltransferase inhibitor selected from the group of A- 197574 (Abbott), a farnesyltransferase inhibitor from Servier, FPTIII (Strathclyde Institute for Drug R), LB-42908 (LG Life Sciences), Pharmaprojects No. 5063 (Genzyme), Pharmaprojects No. 5597 (Ipsen), Yissum Project No. B-1055 (Yissum), and an analogue or derivative thereof.
19008. The method of item 18969 wherein the anti- fibrotic agent is an FLT-3 kinase inhibitor.
19009. The method of item 18969 wherein the anti- fibrotic agent is an FGF receptor kinase inhibitor. 19010. The method of item 18969 wherein the anti- fibrotic agent is a fibrinogen antagonist selected from the group consisting of AUV-201 (Auvation), MG-13926 (Sanofi-Aventis), plasminogen activator (CAS No. 105913-11-9) (from Sanofi-Aventis or UCB), plasminogen activator-2 (tPA-2) (Sanofi-Aventis), pro-urokinase (CAS No. 82657-92-9) (Sanofi-Aventis), and an analogue or derivative thereof.
19011. The method of item 18969 wherein the anti- fibrotic agent is a heat shock protein 90 antagonist selected from the group consisting of SRN-005 (Sirenade), geldanamycin, NSC-33050 (17- allylaminogeldanamycin; 17-AAG), 17-dimethylaminoethylamino-17- demethoxy-geldanamycin (17-DMAG), rifabutin (rifamycin XIV, 1',4- didehydro-1-deoxy-1,4-dihydro-5'-(2-methylpropyl)-1-oxo-), and an analogue or derivative thereof.
19012. The method of item 18969 wherein the anti- fibrotic agent is a histone deacetylase inhibitor.
19013. The method of item 18969 wherein the anti- fibrotic agent is an HMGCoA reductase inhibitor selected from the group consisting of an atherosclerosis therapeutic from Lipid Sciences, ATI-16000 (ARYx Therapeutics), KS-01-019 (Kos Pharmaceuticals), Pharmaprojects No. 2197 (Sanofi-Aventi), RP 61969 (Sanofi-Aventis), and an analogue or derivative thereof.
19014. The method of item 18969 wherein the anti- fibrotic agent is an ICAM inhibitor.
19015. The method of item 18969 wherein the anti- fibrotic agent is an IL, ICE and IRAK antagonist, wherein the antagonist is a CJ-14877, CP-424174 (Pfizer), NF-61 (Negma-Lerads), and an analogue or derivative thereof.
19016. The method of item 18969 wherein the anti- fibrotic agent is an IL-2 inhibitor.
19017. The method of item 18969 wherein the anti- fibrotic agent is an immunosuppressant selected from the group consisting of teriflunomide (Sanofi Aventis), chlorsulfaquinoxalone (NSC-339004), chlorsulfaquinoxalone sulfate, CS-712 (Sankyo), ismomultin alfa (CAS No. 457913-93-8) (Akzo Nobel), antiallergics from GenPat77, antiinflammatories or AT-005 (Androclus Therapeutics), autoimmune disease therapeutics from EpiVax, BN-007 (Bone), budesonide (CAS No. 51333-22- 3) (MAP Pharmaceuticals), CO-14 (Genzyme), edratide (CAS No. 433922- 67-9) (Teva), EP-314 (Enanta), eprovafen (CAS No. 101335-99-3) (Sanofi- Aventis), HWA-131 (CAS No. 118788-41 -3). (Sanofj-Aventis), immunomodulators from MerLion Pharmaceuticals, immunosuppressives from Alchemia, IPL-12 (Inflazyme), MDL-9563 (CAS No. 27086-86-8) (Sanofi-Aventis), Pharmaprojects No. 2330 (Sanofi-Aventis), Pharmaprojects No. 6426 (Abgenix), PXS-25 (Pharmaxis), rosmarinic acid (CAS No. 20283-92-5) (Sanofi-Aventis), RP 42927 or RP 54745 (CAS No. 135330-08-4) (Sanofi-Aventis), SGN-35 (Seattle Genetics), ST-1959 (Sigma-Tau), type I diabetes therapy from SYNX Pharma, UNIL-88 (Debiopharm), VP-025 (Vasogen), VR-694 (Vectura), PRTX-001 (Protalex), and an analogue or derivative thereof.
19018. The method of item 18969 wherein the anti- fibrotic agent is an IMPDH (inosine monophosphate).
19019. The method of item 18969 wherein the anti- fibrotic agent is an integrin antagonist. 19020. The method of item 18969 wherein the anti- fibrotic agent is an interleukin antagonist.
19021. The method of item 18969 wherein the anti- fibrotic agent is an inhibitor of type III receptor tyrosine kinase.
19022. The method of item 18969 wherein the anti- fibrotic agent is an irreversible inhibitor of enzyme methionine aminopeptidase type 2.
19023. The method of item 18969 wherein the anti- fibrotic agent is an isozyme selective delta protein kinase C inhibitor.
19024. The method of item 18969 wherein the anti- fibrotic agent a JAK3 enzyme inhibitor.
19025. The method of item 18969 wherein the anti- fibrotic agent is a JNK inhibitor.
19026. The method of item 18969 wherein the anti- fibrotic agent is a kinase inhibitor.
19027. The method of item 18969 wherein the anti- fibrotic agent is kinesin antagonist.
19028. The method of item 18969 wherein the anti- fibrotic agent is a kinesin antagonist.
19029. The method of item 18969 wherein the anti- fibrotic agent is a leukotriene inhibitor and antagonist selected from the group consisting of ambicromil (CAS No. 58805-38-2) (Sanofi-Aventis), amelubant (CAS No. 346735-24-8) (Boehringer Ingelheim), DW-1141 (Dong Wha), ebselen (Daiichi Pharmaceutical), ibudilast (Kyorin), leucotriene inhibitors from Sanofi-Aventis, lymphotoxin-beta receptor (LT-D) from Biogen Idee, Pharmaprojects No. 1535 and 2728 (CAS No. 119340-33-9) (Sanofi-Aventis), R-112 (Rigel), Rev-5367 (CAS No. 92532-05-3) (Sanofi- Aventis), RG-14893 (CAS No. 141835-49-6) (Sanofi-Aventis), RG-5901-A (CAS No. 101910-24-1), 92532-23-5, RP 66153 (CAS No. 142422-79-5), RP 66364 (CAS No.186916-92-5), RP 69698 (CAS No. 141748-00-7) (Sanofi-Aventis), SC-411930 (Pfizer), SC-41930 (CAS No. 120072-59-5) (Pfizer), SC-50605 (CAS No. 138828-39-4) (Pfizer), SC-51146 (CAS No. 141059-52-1), SC-53228 (CAS No. 153633-01-3) (Pfizer), spaglumic acid (ZY-15106) (CAS No.3106-85-2), 80619-64-3 (Novartis), tipredane (CAS No. 85197-77-9) (Bristol-Myers Squibb), U-75302 (CAS No. 119477-85-9) (Pfizer), and analogue or derivative thereof.
19030. The method of item 18969 wherein the anti- . _ fibrotic agent is an MAP kinase inhibitor.
19031. The method of item 18969 wherein the anti- fibrotic agent is a matrix metalloproteinase inhibitor.
19032. The method of item 18969 wherein the anti- fibrotic agent is an MCP-CCR2 inhibitor.
19033. The method of item 18969 wherein the anti- fibrotic agent is an mTOR inhibitor.
19034. The method of item 18969 wherein the anti- fibrotic agent is an mTOR kinase inhibitor. 19035. The method of item 18969 wherein the anti- fibrotic agent is a microtubule inhibitor selected from the group consisting of antibody-maytansinoid conjugates from Biogen Idee, colchicines (MantiCore Pharmaceuticals), anticancer immunoconjugates from Johnson & Johnson, DIME from Octamer, gni-1f (GNI), huC242-DM4, huMy9-6-DM1 (ImmunoGen), IDN-5404 (Indena), IMO-098, IMOderm (Imotep), mebendazole (Introgen Therapeutics), microtubule poisons from Cambridge Enterprise, paclitaxel such as LOTAX from Aphios (CAS No. 33069-62-4), Genexol-PM from Samyang, Pharmaprojects No. 6383 (Tapestry Pharmaceuticals), RPR-112378 (Sanofi-Aventis), SGN-75 (Seattle Genetics), SPL-7435 (Starpharma), SSR-250411 (Sanofi-Aventis), trastuzumab-DM1 (Genentech), vinorelbine, and an analogue or derivative thereof.
19036. The method of item 18969 wherein the anti- fibrotic agent is an MIF inhibitor.
19037. The method of item 18969 wherein the anti- fibrotic agent is an MMP inhibitor.
19038. The method of item 18969 wherein the anti- fibrotic agent is a neurokinin (NK) antagonist selected from the group consisting of anthrotainin (CAS No. 148084-40-6) (Sanofi-Aventis), an IBS thereapeutic from ArQuIe, MDL-105212A (CAS No. 167261-60-1) (Ssanofi- Aventis), Pharmaprojects No. 2744, 3258 (CAS No. 139167-47-8) 4006, 4201 , or 5986 (Sanofi-Aventis), RP 67580 (CAS No. 135911-02-3), SR- 144190 (CAS No. 201152-86-5), SSR-240600, SSR-241586 (Sanofi- Aventis), TKA-457 (Novartis), vestipitant mesylate (CAS No. 334476-64-1) (GlaxoSmithKline), Win-64821 (Sanofi-Aventis), PRX-96026 (Predix Pharmaceuticals), and an analogue or derivative thereof. 19039. The method of item 18969 wherein the anti- fibrotic agent is an NF kappa B inhibitor selected from the group consisting of AVE-0545 or AVE-0547 (Sanofi-Aventis), bortezomib (CAS No. 179324- 69-7) (Millennium Pharmaceuticals), dexanabinol (CAS No. 112924-45-5) (Pharmos), dexlipotam (Viatris), Pharmaprojects No. 6283 (INDRA) (OXiGENE), IPL-576092 (CAS No. 137571-30-3) (Inflazyme), NFKB decoy (Corgentech), NFKB decoy oligo (AnGes MG), NFKB's from Ariad, osteoporosis treatments or S5 (F005) from Fulcrum Pharmaceuticals, P61 (Phytopharm), R-flurbiprofen (CAS No. 5104-49-4) (Encore Pharmaceuticals), and an analogue or derivative thereof.
19040. The method of item 18969 wherein the anti- fibrotic agent is a nitric oxide agonist.
19041. The method of item 18969 wherein the anti- fibrotic agent is an ornithine decarboxylase inhibitor.
19042. The method of item 18969 wherein the anti- fibrotic agent is a p38 MAP kinase inhibitor selected from the group consisting of AZD-6703 (AstraZeneca), JX-401 (Jexys Pharmaceuticals), BMS-2 (Bristol-Myers Squibb), a p38 MAP kinase inhibitor from Novartis, a p38-alpha MAP kinase inhibitor from Amphora, Pharmaprojects No. 5704 (Pharmacopeia), RPR-200765A (Sanofi-Aventis), SD-282 (Johnson & Johnson), TAK-715 (Takeda), and an analogue or derivative thereof.
19043. The method of item 18969 wherein the anti- fibrotic agent is a palmitoyl-protein thioesterase inhibitor.
19044. The method of item 18969 wherein the anti- fibrotic agent is a PDGF receptor kinase inhibitor selected from the group consisting of AAL-993, AMN-107, or ABP-309 (Novartis), AMG-706 (Amgen), BAY-57-9352 (Bayer), CDP-860 (UCB)1 E-7080 (Eisai), imatinib (CAS No. 152459-95-5) (Novartis), OSI-930 (OSI Pharmaceuticals), RPR- 127963E (Sanofi-Aventis), RWJ-540973 (Johnson & Johnson), sorafenib tosylate (Bayer), SU-11657 (Pfizer), tandutinib (CAS No. 387867-13-2) (Millennium Pharmaceuticals), vatalanib (Novartis), ZK-CDK (Schering AG), and an analogue or derivative thereof.
19045. The method of item 18969 wherein the anti- fibrotic agent is (-)-halofenate (Metabolex), AMG-131 (Amgen), antidiabetics from Japan Tobacco, AZD-4619, AZD-8450, AZD-8677 (AstraZeneca), DRF-10945, balaglitazone (Dr Reddy's), CS-00088, CS-00098 (Chipscreen Biosciences), E-3030 (Eisai), etalocib (CAS No. 161172-51-6) (EIi Lilly), GSK-641597 (Ligand), GSK-677954 (GlaxoSmithKline), GW-409544 (Ligand), GW-590735 (GlaxoSmithKline), K-111 (Hoffmann-La Roche), LY- 518674 (EIi Lilly), LY-674 (Ligand), LY-929 (Ligand), MC-3001 , MC-3002 (MaxoCore Pharmaceuticals), metformin HCI + pioglitazone (CAS No. 1115- 70-4 and 112529-15-4), ACTOPLUS MET from Andrx), muraglitazar (CAS No. 331741-94-7) (Bristol-Myers Squibb), naveglitazar (Ligand), oleoylethanolamide (Kadmus Pharmaceuticals), ONO-5129, pioglitazone hydrochloride (CAS No. 111025-46-8 and 112529-15-4) (Takeda), PLX-204 (Plexxikon), PPAR agonists from Genfit, PPAR delta agonists from EIi Lilly, PPAR-alpha agonists from CrystalGenomics, PPAR-gamma modulators and PPAR-β modulators from CareX, rosiglitazone maleate (CAS No. 122324- 73-4 or 155141-29-0) (GlaxoSmithKline), rosiglitazone maleate/glimepir (CAS No. 155141-29-0 and 93479-97-1), AVANDARYL, rosiglitazone maleate/metformin extend (CAS No. 155141-29-0 and 657-24-9), AVANDAMET, rosiglitazone maleate+metformin, AVANDAMET (GlaxoSmithKline), tesaglitazar (AstraZeneca), and an analogue or derivative thereof. 19046. The method of item 18969 wherein the anti- fibrotic agent is a phosphatase inhibitor.
19047. The method of item 18969 wherein the anti- fibrotic agent is a phosphodiesterase (PDE) inhibitor selected from the group consisting of avanafil (Tanabe Seiyaku), dasantafil (CAS No. 569351- 91-3) (Schering-Plough), A-906119 (CAS No. 134072-58-5), DL-850 (Sanofi-Aventis), GRC-3015, GRC-3566, GRC-3886 (Glenmark), HWA-153 (CAS No. 56395-66-5) (Sanofi-Aventis), hydroxypumafentrine (Altana), IBFB-130011, IBFB-14-016, IBFB-140301, IBFB-150007, IBFB-211913 (IBFB Pharma), L-826141 (Merck & Co), medorinone (CAS No. 88296-61-1) (Sanofi-Aventis), MEM-1917 (Memory Pharmaceuticals), ND-1251 (Neuro3d), PDE inhibitors from ICOS, PDE IV inhibitors from Memory Pharmaceuticals and CrystalGenomics, Pharmaprojects No. 2742 and 6141 (Sanofi-Aventis), QAD-171 (Novartis), RHC-2963 (CAS No. 76993-12-9 and 76993-14-1), RPR-117658, RPR-122818 derivatives, SR-24870 , and RPR- 132294 (Sanofi-Aventis), SK-350 (ln2Gen), stroke therapy agents from deCODE Genetics, TAS-203 (Taiho), tofimilast (CAS No. 185954-27-2) (Pfizer), UK-371800 (Pfizer), WIN-65579 (CAS No. 158020-82-7) (Sanofi- Aventis), IBFB-130020 (IBFB Pharma), OPC-6535 (CAS No. 145739-56-6) (Otsuka), a phosphodiesterase III inhibitor, enoximone, a phosphodiesterase IV inhibitor, fosfosal, Atopik (Barrier Therapeutics), triflusal, a phosphodiesterase V inhibitor, and an analogue or derivative thereof.
19048. The method of item 18969 wherein the anti- fibrotic agent is a PKC inhibitor.
19049. The method of item 18969 wherein the anti- fibrotic agent is a platelet activating factor antagonist. 19050. The method of item 18969 wherein the anti- fibrotic agent is a platelet-derived growth factor receptor kinase inhibitor.
19051. The method of item 18969 wherein the anti- fibrotic agent is a prolyl hydroxylase inhibitor.
19052. The method of item 18969 wherein the anti- fibrotic agent is a polymorphonuclear neutrophil inhibitor.
19053. The method of item 18969 wherein the anti- fibrotic agent is a protein kinase B inhibitor.
19054. The method of item 18969 wherein the anti- fibrotic agent is a protein kinase C stimulant.
19055. The method of item 18969 wherein the anti- fibrotic agent is a purine nucleoside analogue.
19056. The method of item 18969 wherein the anti- fibrotic agent is a purinoreceptor P2X antagonist.
19057. The method of item 18969 wherein the anti- fibrotic agent is a Raf kinase inhibitor.
19058. The method of item 18969 wherein the anti- fibrotic agent is a reversible inhibitor of ErbB1 and ErbB2.
19059. The method of item 18969 wherein the anti- fibrotic agent is a ribonucleoside triphosphate reductase inhibitor. 19060. The method of item 18969 wherein the anti- fibrotic agent is an SDF-1 antagonist.
19061. The method of item 18969 wherein the anti- fibrotic agent is a sheddase inhibitor.
19062. The method of item 18969 wherein the anti- fibrotic agent is an SRC inhibitor.
19063. The method of item 18969 wherein the anti- fibrotic agent is a stromelysin inhibitor.
19064. The method of item 18969 wherein the anti- fibrotic agent is an Syk kinase inhibitor.
- , 19065. The method of item 18969 wherein the anti- fibrotic agent is a telomerase inhibitor.
19066. The method of item 18969 wherein the anti- fibrotic agent is a TGF beta inhibitor selected from the group consisting of pirfenidone (CAS No. 53179-13-8) (MARNAC)1 tranilast (CAS No. 53902- 12-8) (Kissei), IN-1130 (ln2Gen), mannose-6-phosphate (BTG), TGF-β antagonists from Inflazyme (Pharmaprojects No. 6075), TGF-β antagonists from Sydney, non-industrial source), TGF-β I receptor kinase inhibitors from EIi Lilly, TGF-β receptor inhibitors from Johnson & Johnson, and an analogue or derivative thereof.
19067. The method of item 18969 wherein the anti- fibrotic agent is a TNFα antagonist or TACE inhibitor selected from the group consisting of adalimumab (CAS No. 331731-18-1) (Cambridge Antibody Technology), AGIX-4207 (AtheroGenics), AGT-1 (Advanced Biotherapy), an anti-inflammatory from Borean Pharma, Cellzome, or Paradigm Therapeutics, anti-inflammatory vaccine (TNF-alpha kinoid) from Neovacs, humanized anti-TNF antibody or an anti-TNF MAb (CB0006) Celltech (UCB)1 apratastat (CAS No. 287445-51-0) (Wyeth), BMS-561392 (Bristol-Myers Squibb), BN-006 (Bone), certolizumab pegol (CAS No. 428863-50-7 or CH-138 (UCB), cilomilast (CAS No. 153259-65-5) (GlaxoSmithKline), CR-1 (Nuada Pharmaceuticals), CRx-119 (CombinatoRx), D-5410 (UCB), dacopafant (CAS No. 125372-33-0) (Sanofi- Aventis), dersalazine (CAS No. 188913-57-7/188913-58-8) (Uriach), etanercept (CAS No. 185243-69-0) (Amgen), ethyl pyruvate (Critical (Critical Therapeutics), golimumab (CAS No. 476181-74-5) (Johnson & Johnson), hormono-immunotherapy from Ipsen, CDP571 (e.g., Humicade from UCB), IC-485 (ICOS), infliximab (CAS No. 170277-31-3) (Johnson & Johnson), IP- 751 (Manhattan Pharmaceuticals), ISIS-104838 (CAS No. 250755-32-9) (ISIS Pharmaceuticals), lenalidomide (CAS No. 191732-72-6) (Celgene), lentinan (CAS No. 37339-90-5) (Ajjnomoto), MDL-201112 (CAS No. 142130-73-2) (Sanofi-Aventis), medroxyprogesterone (CAS No. 520-85-4) (InKine Pharmaceutical), N-acetylcysteine (CAS No. 616-91-1) (Zambon), NBE-P2 (DIREVO Biotech), nerelimomab (CAS No. 162774-06-3) (Chiron), OM-294DP (OM PHARMA), onercept (CAS No. 199685-57-9) (Yeda), PASSTN F-alpha (Verigen), pentoxifylline or oxypentifylline (Sanofi-Aventis), Pharmaprojects No. 4091 , 4241 , 4295, or 4488 (Sanofi-Aventis), Pharmaprojects No. 5480 (Amgen), Pharmaprojects No. 6749 (Cengent), pirfenidone (CAS No. 53179-13-8) (MARNAC), RPR-132294 (Sanofi- Aventis), S5 (F002) (Fulcrum Pharmaceuticals), simvastatin (CAS No. 79902-63-9) (Merck & Co), STA-6292 (Synta Pharmaceuticals), tacrolimus (CAS No. 104987-11-3) (Fujisawa LifeCycle Pharma), talactoferrin alfa (CAS No. 308240-58-6) (Agennix), thalidomide (CAS No. 50-35-1) (Celgene), TNF antagonists form ProStrakan, and Synergen, TNF inhibitors (Amgen), TNF-alpha antagonists from Dynavax Technologies and Jerina AG (Germany), TNF-alpha inhibitors from IBFB Pharma and Xencor (Xencor), torbafylline (CAS No. 105102-21-4) (Sanofi-Aventis), UR-1505 (Uriach), VT-346 (Viron Therapeutics), YSIL6 (Y1S Therapeutics), YSTH2 (Y's Therapeutics), NPI-1302a-3 (Nereus Pharmaceuticals, a TNF antagonist from Jerina AG (Germany), dersalazine, and an analogue or derivative thereof.
19068. The method of item 18969 wherein the anti- fibrotic agent is a Toll receptor inhibitor.
19069. The method of item 18969 wherein the anti- fibrotic agent is a tubulin antagonist.
19070. The method of item 18969 wherein the anti- fibrotic agent is a tyrosine kinase inhibitor selected from the group consisting of SU-011248, SUTENT from Pfizer Inc. (New York, NY), BMS-354825, PN- 355 (Paracelsian Pharmaceuticals), AGN-199659 (Allergan), AAL-993 or ABP-309 (Novartis), adaphostin (NIH), AEE-788 (Novartis), AG-013736 (OSI Pharmaceuticals), AG-13736 (Pfizer), ALT-110 (Alteris Therapeutics), AMG-706 (Amgen), anticancer MAbs from Xencor, anti-EGFrvlll MAbs from Abgenix, anti-HER2 MAb from Abiogen, AZD-2171 or AZD-9935 (AstraZeneca), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), CEP-5214 (Cephalon), CEP-7055 (Cephalon), cetuximab (ImClone Systems), CHIR-200131 and CHIR-258 (Chiron), CP-547632 (OSI Pharmaceuticals), CP-724714 (Pfizer), CT-301 (Creabilis Therapeutics), D- 69491 (Baxter International), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EGFR/ErbB2 inhibitors from Array BioPharma, erlotinib (CAS No. 183319- 69-9) (OSI Pharmaceuticals), EXEL-2880 (Exelixis), FK-778 (Sanofi- Aventis) , gefitinib (CAS No. 184475-35-2) (AstraZeneca), GW-2286 or GW- 654652 (GlaxoSmithKline), her2/neu antigen from AlphaVax, HER-2/neu inhibitor from Generex, Herzyme (Medipad) (Sirna Therapeutics), HKI-272 (Wyeth), HuMax-EGFr (Genmab), idronoxil (CAS No. 81267-65-4) (Novogen), IGF-1 inhibitors from Ontogen, IMC-11F8 (ImCIone Systems), kahalalide F (CAS No. 149204-42-2) (PharmaMar), KDR inhibitor from LG Life Sciences, KDR inhibitors from Abbott Laboratories, KDR kinase inhibitors (UCB), Kdr kinase inhibitors from Merck & Co, KRN-633 and KRN- 951 (Kirin Brewery), KSB-102 (Xenova), lapatinib ditosylate (CAS No. 388082-78-8) (GlaxoSmithKline), matuzumab (Merck KGaA), MDX-214 (Medarex), ME-103 (Pharmexa), MED-A300 (Gerolymatos), MNAC-13 (Lay Line Genomics), nimotuzumab (Center of Molecular Immunology), NSC- 330507 or NSC-707545 (NIH), NV-50 (Novogen), OSI-930 (OSI Pharmaceuticals), panitumumab (Abgenix), pelitinib (CAS No. 287933-82-7) (Wyeth), pertuzumab (CAS No. 380610-27-5) (Genentech), Pharmaprojects No. 3985 (Sanofi-Aventis), prostate cancer therapeutics from Sequenom (SQPC35, SQPC36, SQPC90), removab and remoxab (Trion Pharma), RG- 13022 (CAS No. 136831-48-6), RG-13291 (CAS No. 138989-50-1), or RG- 14620 (CAS No. 136831-49-7) (Sanofi-Aventis), RM-6427 (Romark), RNAi breast cancer therapy from Benitec, RP 53801 (CAS No. 125882-88-4) (Sanofi-Aventis), sorafenib tosylate (Bayer), SU-11657 (Pfizer), Tie-2 antagonists from Semaia (Hybrigenics), Tie-2 inhibitors from Ontogen, trastuzumab (CAS No. 180288-69-1) (Genentech), tyrosine kinase inhibitors from Sanofi-Aventis, U3-1287, U3-1565, U3-1784, U3-1800 (U3 Pharma), vatalanib (Novartis), VEGFR-2 kinase inhibitor from Bristol-Myers Squibb, XL-647 (Exelixis), ZD-6474 (AstraZeneca), ZK-CDK (Schering AG), an EGFR tyrosine kinase inhibitor, EKB-569 (Wyeth), and an analogue or derivative thereof.
19071. The method of item 18969 wherein the anti- fibrotic agent is a VEGF inhibitor.
19072. The method of item 18969 wherein the anti- fibrotic agent is a vitamin D receptor agonist. 19073. The method of item 18969 wherein the anti- fibrotic agent is ZD-6474 (an angiogenesis inhibitor).
19074. The method of item 18969 wherein the anti- fibrotic agent is AP-23573 (an mTOR inhibitor).
19075. The method of item 18969 wherein the anti- fibrotic agent is synthadotin (a tubulin antagonist).
19076. The method of item 18969 wherein the anti- fibrotic agent is S-0885 (a collagenase inhibitor).
19077. The method of item 18969 wherein the anti- fibrotic agent is aplidine (an elongation factor-1 alpha inhibitor).
19078. The method of item 18969 wherein the anti- fibrotic agent is ixabepilone (an epithilone).
19079. The method of item 18969 wherein the anti- fibrotic agent is IDN-5390 (an angiogenesis inhibitor).
19080. The method of item 18969 wherein the anti- fibrotic agent is SB-2723005 (an angiogenesis inhibitor).
19081. The method of item 18969 wherein the anti- fibrotic agent is ABT-518 (an angiogenesis inhibitor).
19082. The method of item 18969 wherein the anti- fibrotic agent is combretastatin (an angiogenesis inhibitor). 19083. The method of item 18969 wherein the anti- fibrotic agent is anecortave acetate (an angiogenesis inhibitor).
19084. The method of item 18969 wherein the anti- fibrotic agent is SB-715992 (a kinesin antagonist).
19085. The method of item 18969 wherein the anti- fibrotic agent is temsirolimus (an mTOR inhibitor).
19086. The method of item 18969 wherein the anti- fibrotic agent is adalimumab (a TNFα antagonist).
19087. The method of item 18969 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with an anti-fibrotic agent, and (b) implanting the medical device into the host.
19088. The method of item 18969 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with an anti-infective agent, and (b) implanting the medical device into the host.
19089. The method of item 18969 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with a polymer; and (b) implanting the medical device into the host.
19090. The method of item 18969 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with a composition comprising an anti-fibrotic agent and a polymer, and (b) implanting the medical device into the host. 19091. The method of item 18969 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with a composition comprising an anti-infective agent and a polymer, and (b) implanting the medical device into the host.
19092. The method of item 18969 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with a composition comprising an anti-fibrotic agent, an anti- infective agent and a polymer, and (b) implanting the medical device into the host.
19093. The method of item 18969 wherein the anti- infective agent is an anthracycline.
19094. The method of item 18969 wherein the anti- infective agent is doxorubicin.
19095. The method of item 18969 wherein the anti- infective agent is is mitoxantrone.
19096. The method of item 18969 wherein the anti- infective agent is a fluoropyrimidine.
19097. The method of item 18969 wherein the anti- infective agent is 5-fluorouracil (5-FU).
19098. The method of item 18969 wherein the anti- infective agent is a folic acid antagonist.
19099. The method of item 18969 wherein the anti- infective agent is methotrexate. 19100. The method of item 18969 wherein the anti- infective agent is a podophylotoxin.
19101. The method of item 18969 wherein the anti- infective agent is etoposide.
19102. The method of item 18969 wherein the anti- infective agent is camptothecin.
19103. The method of item 18969 wherein the anti- infective agent is a hydroxyurea.
19104. The method of item 18969 wherein the anti- infective agent is a platinum complex.
19105. The method of item 18969 wherein the anti- infective agent is cisplatin.
19106. The method of item 18969 wherein the composition comprises an anti-thrombotic agent.
19107. The method of item 18969 wherein the polymer is formed from reactants comprising a naturally occurring polymer.
19108. The method of item 18969 wherein the polymer is formed from reactants comprising protein.
19109. The method of item 18969 wherein the polymer is formed from reactants comprising carbohydrate. 19110. The method of item 18969 wherein the polymer reactants comprising biodegradable polymer.
19111. The method of item 18969 wherein the polymer reactants comprising nonbiodegradable polymer.
19112. The method of item 18969 wherein the polymer reactants comprising collagen.
19113. The method of item 18969 wherein the polymer reactants comprising methylated collagen.
19114. The method of item 18969 wherein the polymer reactants comprising fibrinogen.
19115. The method of item 18969 wherein the polymer reactants comprising thrombin.
19116. The method of item 18969 wherein the polymer reactants comprising blood plasma.
19117. The method of item 18969 wherein the polymer reactants comprising calcium salt.
19118. The method of item 18969 wherein the polymer reactants comprising an antifibronolytic agent.
19119. The method of item 18969 wherein the polymer reactants comprising fibrinogen analog. 19120. The method of item 18969 wherein the polymer reactants comprising albumin.
19121. The method of item 18969 wherein the polymer reactants comprising plasminogen.
19122. The method of item 18969 wherein the polymer reactants comprising von Willebrands factor.
19123. The method of item 18969 wherein the polymer reactants comprising Factor VIII.
19124. The method of item 18969 wherein the polymer reactants comprising hypoallergenic collagen.
19125. The method of item 18969 wherein the polymer reactants comprising atelopeptidic collagen.
19126. The method of item 18969 wherein the polymer reactants comprising telopeptide collagen.
19127. The method of item 18969 wherein the polymer reactants comprising crosslinked collagen.
19128. The method of item 18969 wherein the polymer reactants comprising aprotinin.
19129. The method of item 18969 wherein the polymer reactants comprising epsilon-amino-n-caproic acid. 19130. The method of item 18969 wherein the polymer is formed from reactants comprising gelatin.
19131. The method of item 18969 wherein the polymer is formed from reactants comprising protein conjugates.
19132. The method of item 18969 wherein the polymer is formed from reactants comprising gelatin conjugates.
19133. The method of item 18969 wherein the polymer is formed from reactants comprising a synthetic polymer.
19134. The method of item 18969 wherein the polymer is formed from reactants comprising a synthetic isocyanate-containing compound.
19135. The method of item 18969 wherein the polymer is formed from reactants comprising a synthetic thiol-containing compound.
19136. The method of item 18969 wherein the polymer is formed from reactants comprising a synthetic compound containing at least two thiol groups.
19137. The method of item 18969 wherein the polymer is formed from reactants comprising a synthetic compound containing at least three thiol groups.
19138. The method of item 18969 wherein the polymer is formed from reactants comprising a synthetic compound containing at least four thiol groups. 19139. The method of item 18969 wherein the polymer is formed from reactants comprising a synthetic amino-containing compound.
19140. The method of item 18969 wherein the polymer is formed from reactants comprising a synthetic compound containing at least two amino groups.
19141. The method of item 18969 wherein the polymer is formed from reactants comprising a synthetic compound containing at least three amino groups.
19142. The method of item 18969 wherein the polymer is formed from reactants comprising a synthetic compound containing at least four amino groups.
19143. The method of item 18969 wherein the polymer is formed from reactants comprising a synthetic compound comprising a carbonyl-oxygen-succinimidyl group.
19144. The method of item 18969 wherein the polymer is formed from reactants comprising a synthetic compound comprising at least two carbonyl-oxygen-succinimidyl groups.
19145. The method of item 18969 wherein the polymer is formed from reactants comprising a synthetic compound comprising at least three carbonyl-oxygen-succinimidyl groups.
19146. The method of item 18969 wherein the polymer is formed from reactants comprising a synthetic compound comprising at least four carbonyl-oxygen-succinimidyl groups. 19147. The method of item 18969 wherein the polymer is formed from reactants comprising a synthetic polyalkylene oxide- containing compound.
19148. The method of item 18969 wherein the polymer is formed from reactants comprising a synthetic compound comprising both polyalkylene oxide and biodegradable polyester blocks.
19149. The method of item 18969 wherein the polymer is formed from reactants comprising a synthetic polyalkylene oxide- containing compound having reactive amino groups.
19150. The method of item 18969 wherein the polymer is formed from reactants comprising a synthetic polyalkylene oxide- containing compound having reactive thiol groups.
19151. The method of item 18969 wherein the polymer is formed from reactants comprising a synthetic polyalkylene oxide- containing compound having reactive carbonyl-oxygen-succinimidyl groups.
19152. The method of item 18969 wherein the polymer is formed from reactants comprising a synthetic compound comprising a biodegradable polyester block.
19153. The method of item 18969 wherein the polymer is formed from reactants comprising a synthetic polymer formed in whole or part from lactic acid or lactide.
19154. The method of item 18969 wherein the polymer is formed from reactants comprising a synthetic polymer formed in whole or part from glycolic acid or glycolide. 19155. The method of item 18969 wherein the polymer is formed from reactants comprising polylysine.
19156. The method of item 18969 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound comprising a polyalkylene oxide portion.
19157. The method of item 18969 wherein the polymer is formed from reactants comprising (a) protein and (b) polylysine.
19158. The method of item 18969 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound having at least four thiol groups.
19159. The method of item 18969 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound having at least four amino groups.
19160. The method of item 18969 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound having at least four carbonyl-oxygen-succinimide groups.
19161. The method of item 18969 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound having a biodegradable region formed from reactants selected from lactic acid, lactide, glycolic acid, glycolide, and epison-caprolactone.
19162. The method of item 18969 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound comprising a polyalkylene oxide portion. 19163. The method of item 18969 wherein the polymer is formed from reactants comprising (a) collagen and (b) polylysine.
19164. The method of item 18969 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound having at least four thiol groups.
19165. The method of item 18969 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound having at least four amino groups.
19166. The method of item 18969 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound having at least four carbonyl-oxygen-succinimide groups.
19167. The method of item 18969 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound having a biodegradable region formed from reactants selected from lactic acid, lactide, glycolic acid, glycolide, and epison-caprolactone.
19168. The method of item 18969 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound comprising a polyalkylene oxide portion.
19169. The method of item 18969 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) polylysine.
19170. The method of item 18969 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound having at least four thiol groups. 19171. The method of item 18969 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound having at least four amino groups.
19172. The method of item 18969 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound having at least four carbonyl-oxygen-succinimide groups.
19173. The method of item 18969 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound having a biodegradable region formed from reactants selected from lactic acid, lactide, glycolic acid, glycolide, and epison-caprolactone.
19174. The method of item 18969 wherein the polymer is formed from reactants comprising hyaluronic acid.
19175. The method of item 18969 wherein the polymer is formed from reactants comprising a hyaluronic acid derivative.
19176. The method of item 18969 wherein the polymer is formed from reactants comprising pentaerythritol poly(ethylene glycol)ether tetra-sulfhydryl of number average molecular weight between 3,000 and 30,000.
19177. The method of item 18969 wherein the polymer is formed from reactants comprising pentaerythritol poly(ethylene glycol)ether tetra-amino of number average molecular weight between 3,000 and 30,000.
19178. The method of item 18969 wherein the polymer is formed from reactants comprising (a) a synthetic compound having a number average molecular weight between 3,000 and 30,000 and comprising a polyalkylene oxide region and multiple nucleophilic groups, and (b) a synthetic compound having a number average molecular weight between 3,000 and 30,000 and comprising a polyalkylene oxide region and multiple electrophilic groups.
19179. The method of item 18969 wherein the composition comprises a colorant.
19180. The method of item 18969 wherein the composition is sterile.
19181. A method as in any one of items 18969-19180, wherein the device is an intraocular lens device for preventing lens opacification.
19182. A method as in any one of items 18969-19180, wherein the device is a polymethylmethacrylate intraocular lens.
19183. A method as in any one of items 18969-19180, wherein the device is a silicone intraocular lens.
19184. A method as in any one of items 18969-19180, wherein the device is an achromatic lens.
19185. A method as in any one of items 18969-19180, wherein the device is a pseudophako.
19186. A method as in any one of items 18969-19180, wherein the device is a phakic lens. 19187. A method as in any one of items 18969-19180, wherein the device is a aphakic lens.
19188. A method as in any one of items 18969-19180, wherein the device is a multi-focal intraocular lens.
19189. A method as in any one of items 18969-19180, wherein the device is a hydrophilic and hydrophobic acrylic intraocular lens.
19190. A method as in any one of items 18969-19180, wherein the device is an intraocular implant.
19191. A method as in any one of items 18969-19180, wherein the device is an optic lens.
19192. A method as in any one of items 18969-19180, wherein the device is a rigid gas permeable lens.
19193. A method as in any one of items 18969-19180, wherein the device is a foldable intraocular lens.
19194. A method as in any one of items 18969-19180, wherein the device is a rigid intraocular lens.
19195. A method as in any one of items 18969-19180, wherein the device is a corrective implant for vision impairment.
19196. A method as in any one of items 18969-19180, wherein the device is an intraocular implant adapted for being used in conjunction with a transplant for the cornea. 19197. A method as in any one of items 18969-19180, wherein the device is an intraocular implant adapted for being used in conjunction with treatment of secondary cataract after extracapsular cataract extraction.
19198. A method for implanting a medical device comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with i) an anti-fibrotic agent, ii) an anti-infective agent, iii) a polymer, iv) a composition comprising an anti-fibrotic agent and a polymer, v) a composition comprising an anti-infective agent and a polymer, or vi) a composition comprising an anti-fibrotic agent, an anti- infective agent and a polymer, and (b) implanting the medical device into the host, wherein the medical device is a medical device for treating hypertropic scar or keloid.
19199. The method of item 19198 wherein the anti- fibrotic agent is an adensosine A2A receptor antagonist.
19200. The method of item 19198 wherein the anti- fibrotic agent is an AKT inhibitor.
19201. The method of item 19198 wherein the anti- fibrotic agent is an alpha 2 integrin antagonist, wherein the alpha 2 integrin antagonist is Pharmaprojects No. 5754 (Merck KgaA).
19202. The method of item 19198 wherein the anti- fibrotic agent is an alpha 4 integrin antagonist.
19203. The method of item 19198 wherein the anti- fibrotic agent is an alpha 7 nicotinic receptor agonist. 19204. The method of item 19198 wherein the anti- fibrotic agent is an angiogenesis inhibitor selected from the group consisting of AG-12,958 (Pfizer), ATN-161 (Attention LLC), neovastat, an angiogenesis inhibitor from Jerina AG (Germany), NM-3 (Mercian), VGA-1155 (Taisho), FCE-26644 (Pfizer), FCE-26950 (Pfizer), FPMA (Meiji Daries), FFM 11142 (Fujisawa), GGTI-298, GM-1306 (Ligand), GPA-1734 (Novartis), NNC-47- 0011 (Novo Nordisk), herbamycin (Nippon Kayaku), lenalidomide (Celegene), IP-10 (NIH), ABT-828 (Abbott), KIN-841 (Tokushima University, Japan), SF-1126 (Semafore Pharmaceuticals), laminin technology (NIH), CHIR-258 (Chiron), NVP-AEW541 (Novartis), NVP-AEW541 (Novartis), Vt16907 (Alchemia), OXl-8007 (Oxigene), EG-3306 (Ark Therapeutics), Maspin (Arriva), ABT-567 (Abbott), PPI-2458 (Praecis Pharmaceuticals), CC-5079, CC-4089 (Celgene), HIF-1 alpha inhibitors (Xenova), S-247 (Pfizer), AP-23573 (Ariad), AZD-9935 (Astra Zeneca), mebendazole (Introgen Therapeutics), MetAP-2 inhibitors (GlaxoSmithKline), AG-615 (Angiogene Pharmaceuticals), Tie-2 antagonists (Hybrigenics), NC-381, CYC-381 , NC-169, NC-219, NC-383, NC-384, NC-407 (Lorus Therapeutics), ATN-224 (Attenuon), ON-01370 (Onconova), Vitronectin antagonists (Amgen), SDX-103 (Salmedix), Vitronectin antagonists (Shire), CHP (Riemser), TEK (Amgen), Anecortave acetate (Alcon), T46.2 (Matrix Therapeutics), HG-2 (Heptagen), TEM antagonists (Genzyme), Oxi-4500 (Oxigene), ATN-161 (Attenuon), WX-293 (Wilex), M-2025 (Metris Therapeutics), Alphastatin (BioActa), YH-16 (Yantai Rongchang), BIBF- 1120 (Boehringer Ingelheim), BAY-57-9352 (Bayer), AS-1404 (Cancer Research Technology), SC-77964 (Pfizer), glycomimetics (BioTie Therapies), TIE-2 Inhibitors (Ontogen), DIMI, Octamer (Octamer), ABR- 215050 (Active Biotech), ABT-518 (Abbott), KDR inhibitors (Abbott), BSF- 466895 (Abbott), SCH-221153 (Schering-Plough), DAC:antiangiogenic (ConjuChem), TFPI (EntreMed), AZD-2171 (Astra-Zenaca), CDC-394 (Celgene), LY290293 (EIi Lilly), IDN-5390 (Indena), Kdr Kinase Inhibitors (Merck), CT-113020, CT-116433, CT-116563, CT-31890, CT-32228) (Cell Therapeutics), A-299620 (Abbott), TWEAK Inhibitor (Amgen), VEGF modulators (Johnson and Johnson), Tum-N53, tumstatin (Genzyme), Thios- 1 , Thios-2 (Thios Pharmaceuticals), MV-6401 (Miravant Medical Technologies), Spisulosine (PharmaMar), CEP-7055 (Cephalon), AUV-201 (Auvation), LM-609 (EIi Lilly), SKF-106615 (AnorMED), Oglufanide disodium (Cytran), BW-114 (Pharminox), Calreticulin (NIH), WX-678 (Wilex), SD-7784 (Pfizer), WX-UK1 (Wilex), SH-268 (Schering AG), 2-Me-PGA (Celgene), S- 137 (Pfizer), ZD-6126 (Angiogene Pharmaceuticals), SG-292 (SignalGen), Benefin (Lane Labs), A6, A36 (Angstrom), SB-2723005 (GlaxoSmithKline), SC-7 (Cell Therapeutics), ZEN-014 (AEterna Zentaris), 2-methoxyestradiol (EntreMed), NK-130119 (Nippon Kayaku), CC-10004 (Celgene), AVE- 8062A (Ajinomoto), Tacedinaline (Pfizer), Actinonin (Tokyo Metropolitan Institute of Medical Science), Lenalidomide (Celgene), VGA-1155, BTO-956 (SRI International), ER-68203-00 (Eisai), CT-6685 (UCB), JKC-362 (Phoenix Pharmaceuticals), DMI-3798 (DMI Biosciences, Angiomate (Ipsen), ZD-6474 (AstraZeneca), CEP-5214 (Cephalon), Canstatin (Genzyme), NM-3 (Mercian), Oridigm (MediQuest Therapeutics), Exherin (Adherex), BLS-0597 (Boston Life Sciences), PTC-299 (PTC Therapeutics), NPI-2358 (Nereus Pharmaceuticals), CGP-79787 (Novartis), AEE-788 (Novartis), CKD-732 (Chong Kun Dang), CP-564959 (OSI Pharmaceuticals), CM-101 (CarboMed), CT-2584, CT3501 (Cell Therapeutics), combretastatin and analogues and derivatives thereof (Oxigene), Rebimastat (Bristol-Meyers Squibb), Dextrin 2-sulfate (ML Laboratories), Cilengitide (Merk KGaA), NSC-706704 (Pharminox), KRN- 951 (Kirin Brewery), Ukrain, NSC-631570 (Nowicky Pharma), Tecogalan sodium (Daiichi Pharmaceutical), Tz-93 (Tsumura), TBC-1635 (Encysive Pharmaceuticals), TAN-1120 (Takeda), Semaxanib (Pfizer), BDI-7800 (Biopharmacopae), SD-186, SD-983 (Bristol-Meyers Squibb), SB-223245 (GlaxoSmithKline), SC-236 (Pfizer), RWJ-590973 (Johnson and Johnson), ILX-1850 (Genzyme), SC-68488, S-836 (Pfizer), CG-55069-11 (CuraGen), Ki-23057 (Kirin Brewery), CCX-700 (Chemoentryx), Pegaptanib octasodium (Giled Sciences), ANGIOCOL (available from Biostratum Inc.), or an analogue or derivative thereof.
19205. The method of item 19198 wherein the anti- fibrotic agent is an apoptosis antagonist.
19206. The method of item 19198 wherein the anti- fibrotic agent is an apoptosis activator.
19207. The method of item 19198 wherein the anti- fibrotic agent is a beta 1 integrin antagonist.
19208. The method of item 19198 wherein the anti- fibrotic agent is a beta tubulin inhibitor.
19209. The method of item 19198 wherein the anti- fibrotic agent is a blocker of enzyme production in Hepatitis C.
19210. The method of item 19198 wherein the anti- fibrotic agent is a Bruton's tyrosine kinase inhibitor.
19211. The method of item 19198 wherein the anti- fibrotic agent is a calcineurin inhibitor.
19212. The method of item 19198 wherein the anti- fibrotic agent is a caspase 3 inhibitor.
19213. The method of item 19198 wherein the anti- fibrotic agent is a CC chemokine receptor antagonist. 19214. The method of item 19198 wherein the anti- fibrotic agent is a cell cycle inhibitor selected from the group consisting of SNS-595 (Sunesis), synthadotin, KRX-0403, and an analogue or derivative thereof.
19215. the method of item 19198 wherein the anti- fibrotic agent is a cathepsin B inhibitor.
19216. The method of item 19198 wherein the anti- fibrotic agent is a cathepsin K inhibitor, wherein the cathepsin K inhibitor is 462795 (GlaxoSmithKline), INPL-022-D6 (Amura Therapeutics), or an analogue or derivative thereof.
19217. The method of item 19198 wherein the anti- fibrotic agent is a cathepsin L inhibitor.
19218. The method of item 19198 wherein the anti- fibrotic agent is a CD40 antagonist.
19219. The method of item 19198 wherein the anti- fibrotic agent is a chemokine receptor agonist.
19220. The method of item 19198 wherein the anti- fibrotic agent is a chymase inhibitor.
19221. The method of item 19198 wherein the anti- fibrotic agent is a collagenase antagonist.
19222. The method of item 19198 wherein the anti- fibrotic agent is a CXCR antagonist. 19223. The method of item 19198 wherein the anti- fibrotic agent is a cyclin dependent kinase inhibitor selected from the group consisting of a CDK-1 inhibitor, a CDK-2 inhibitor, a CDK- 4 inhibitor, a CDK-6 inhibitor, a CAK1 inhibitor from GPC Biotech and Bristol-Myers Squibb, RGB-286199 (GPC Biotech), an anticancer agent from Astex Technology, a CAK1 inhibitor from GPC Biotech, a CDK inhibitor from Sanofi-Aventis, a CDK1/CDK2 inhibitor from Amgen, a CDK2 inhibitor from SUGEN-2 (Pfizer), a hearing loss therapy agent (Sound Pharmaceuticals), PD-0332991 (Pfizer), RGB-286199 (GPC Biotech), Ro-0505124 (Hoffmann- La Roche), a Ser/Thr kinase inhibitor from Lilly (EIi Lilly), CVT-2584 (CAS No. 199986-75-9) (CV Therapeutics), and an analogue or derivative thereof.
19224. The method of item 19198 wherein the anti- fibrotic agent is a cyclooxygenase 1 inhibitor.
19225. The method of item 19198 wherein the anti- fibrotic agent is a DHFR inhibitor.
19226. The method of item 19198 wherein the anti- fibrotic agent is a dual integrin inhibitor.
19227. The method of item 19198 wherein the anti- fibrotic agent is an elastase inhibitor.
19228. The method of item 19198 wherein the anti- fibrotic agent is an elongation factor-1 alpha inhibitor.
19229. The method of item 19198 wherein the anti- fibrotic agent is an endothelial growth factor antagonist. 19230. The method of item 19198 wherein the anti- fibrotic agent is an endothelial growth factor receptor kinase inhibitor selected from the group consisting of sorafenib tosylate (Bayer), AAL-993 (Novartis), ABP-309 (Novartis), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EXEL- 2880 (Exelixis), GW-654652 (GlaxoSmithKline), a KDR inhibitor from LG Life Sciences, CT-6685 and CT-6729 (UCB), KRN-633 and KRN-951 (Kirin Brewery), OSI-930 (OSI Pharmaceuticals), SP-5.2 (Supratek Pharma), SU- 11657 (Pfizer), a Tie-2 antagonist (Hybrigenics), a VEGFR-2 kinase inhibitor (Bristol-Myers Squibb), XL-647 (Exelixis), a KDR inhibitor from Abbott Laboratories, sorafenib tosylate, and an analogue or derivative thereof.
19231. The method of item 19198 wherein the anti- fibrotic agent is an endotoxin antagonist.
19232. The method of item 19198 wherein the anti- fibrotic agent is an epothilone and tubulin binder.
19233. The method of item 19198 wherein the anti- fibrotic agent is an estrogen receptor antagonist.
19234. The method of item 19198 wherein the anti- fibrotic agent is an FGF inhibitor.
19235. The method of item 19198 wherein the anti- fibrotic agent is a famexyl transferase inhibitor.
19236. The method of item 19198 wherein the anti- fibrotic agent is farnesyltransferase inhibitor selected from the group of A- 197574 (Abbott), a farnesyltransferase inhibitor from Servier, FPTIII (Strathclyde Institute for Drug R), LB-42908 (LG Life Sciences), Pharmaprojects No. 5063 (Genzyme), Pharmaprojects No. 5597 (Ipsen), Yissum Project No. B-1055 (Yissum), and an analogue or derivative thereof.
19237. The method of item 19198 wherein the anti- fibrotic agent is an FLT-3 kinase inhibitor.
19238. The method of item 19198 wherein the anti- fibrotic agent is an FGF receptor kinase inhibitor.
19239. The method of item 19198 wherein the anti- fibrotic agent is a fibrinogen antagonist selected from the group consisting of AUV-201 (Auvation), MG-13926 (Sanofi-Aventis), plasminogen activator (CAS No. 105913-11-9) (from Sanofi-Aventis or UCB), plasminogen activator-2 (tPA-2) (Sanofi-Aventis), pro-urokinase (CAS No. 82657-92-9) (Sanofi-Aventis), and an analogue or derivative thereof.
19240. The method of item 19198 wherein the anti- fibrotic agent is a heat shock protein 90 antagonist selected from the group consisting of SRN-005 (Sirenade), geldanamycin, NSC-33050 (17- allylaminogeldanamycin; 17-AAG), 17-dimethylaminoethylamino-17- demethoxy-geldanamycin (17-DMAG), rifabutin (rifamycin XIV, 1',4- didehydro-1-deoxy-1 ,4-dihydro-5'-(2-methylpropyl)-1-oxo-), and an analogue or derivative thereof.
19241. The method of item 19198 wherein the anti- fibrotic agent is a histone deacetylase inhibitor.
19242. The method of item 19198 wherein the anti- fibrotic agent is an HMGCoA reductase inhibitor selected from the group consisting of an atherosclerosis therapeutic from Lipid Sciences, ATM 6000 (ARYx Therapeutics), KS-01-019 (Kos Pharmaceuticals), Pharmaprojects No. 2197 (Sanofi-Aventi), RP 61969 (Sanofi-Aventis), and an analogue or derivative thereof.
19243. The method of item 19198 wherein the anti- fibrotic agent is an ICAM inhibitor.
19244. The method of item 19198 wherein the anti- fibrotic agent is an IL, ICE and IRAK antagonist, wherein the antagonist is a CJ-14877, CP-424174 (Pfizer), NF-61 (Negma-Lerads), and an analogue or derivative thereof.
19245. The method of item 19198 wherein the anti- fibrotic agent is an IL-2 inhibitor.
19246. The method of item 19198 wherein the anti- fibrotic agent is an immunosuppressant selected from the group consisting of teriflunomide (Sanofi Aventis), chlorsulfaquinoxalone (NSC-339004), chlorsulfaquinoxalone sulfate, CS-712 (Sankyo), ismomultin alfa (CAS No. 457913-93-8) (Akzo Nobel), antiallergics from GenPat77, antiinflammatories or AT-005 (Androclus Therapeutics), autoimmune disease therapeutics from EpiVax, BN-007 (Bone), budesonide (CAS No. 51333-22- 3) (MAP Pharmaceuticals), CO-14 (Genzyme), edratide (CAS No. 433922- 67-9) (Teva), EP-314 (Enanta), eprovafen (CAS No. 101335-99-3) (Sanofi- Aventis), HWA-131 (CAS No. 118788-41-3) (Sanofi-Aventis), immunomodulators from MerLion Pharmaceuticals, immunosuppressives from Alchemia, IPL-12 (Inflazyme), MDL-9563 (CAS No. 27086-86-8) (Sanofi-Aventis), Pharmaprojects No. 2330 (Sanofi-Aventis), Pharmaprojects No. 6426 (Abgenix), PXS-25 (Pharmaxis), rosmarinic acid (CAS No. 20283-92-5) (Sanofi-Aventis), RP 42927 or RP 54745 (CAS No. 135330-08-4) (Sanofi-Aventis), SGN-35 (Seattle Genetics), ST-1959 (Sigma-Tau), type I diabetes therapy from SYNX Pharma, UNIL-88 (Debiopharm), VP-025 (Vasogen), VR-694 (Vectura), PRTX-001 (Protalex), and an analogue or derivative thereof.
19247. The method of item 19198 wherein the anti- fibrotic agent is an IMPDH (inosine monophosphate).
19248. The method of item 19198 wherein the anti- fibrotic agent is an integrin antagonist.
19249. The method of item 19198 wherein the anti- fibrotic agent is an interleukin antagonist.
19250. The method of item 19198 wherein the anti- fibrotic agent is an inhibitor of type III receptor tyrosine kinase.
1925-1. The method of item 19198 wherein the anti- fibrotic agent is an irreversible inhibitor of enzyme methionine aminopeptidase type 2.
19252. The method of item 19198 wherein the anti- fibrotic agent is an isozyme selective delta protein kinase C inhibitor.
19253. The method of item 19198 wherein the anti- fibrotic agent a JAK3 enzyme inhibitor.
19254. The method of item 19198 wherein the anti- fibrotic agent is a JNK inhibitor.
19255. The method of item 19198 wherein the anti- fibrotic agent is a kinase inhibitor. 19256. The method of item 19198 wherein the anti- fibrotic agent is kinesin antagonist.
19257. The method of item 19198 wherein the anti- fibrotic agent is a kinesin antagonist.
19258. The method of item 19198 wherein the anti- fibrotic agent is a leukotriene inhibitor and antagonist selected from the group consisting of ambicromil (CAS No. 58805-38-2) (Sanofi-Aventis), amelubant (CAS No. 346735-24-8) (Boehringer Ingelheim), DW-1141 (Dong Wha), ebselen (Daiichi Pharmaceutical), ibudilast (Kyorin), leucotriene inhibitors from Sanofi-Aventis, lymphotoxin-beta receptor (LT-D) from Biogen Idee, Pharmaprojects No. 1535 and 2728 (CAS No. 119340-33-9) (Sanofi-Aventis), R-112 (Rigel), Rev-5367 (CAS No. 92532-05-3) (Sanofi- Aventis), RG-14893 (CAS No. 141835-49-6) (Sanofi-Aventis), RG-5901-A (CAS No. 101910-24-1), 92532-23-5, RP 66153 (CAS No. 142422-79-5), RP 66364 (CAS No.186916-92-5), RP 69698 (CAS No. 141748-00-7) (Sanofi-Aventis), SC-411930 (Pfizer), SC-41930 (CAS No. 120072-59-5) (Pfizer), SC-50605 (CAS No. 138828-39-4) (Pfizer), SC-51146 (CAS No. 141059-52-1), SC-53228 (CAS No. 153633-01-3) (Pfizer), spaglumic acid (ZY-15106) (CAS No.3106-85-2), 80619-64-3 (Novartis), tipredane (CAS No. 85197-77-9) (Bristol-Myers Squibb), U-75302 (CAS No. 119477-85-9) (Pfizer), and analogue or derivative thereof.
19259. The method of item 19198 wherein the anti- fibrotic agent is an MAP kinase inhibitor.
19260. The method of item 19198 wherein the anti- fibrotic agent is a matrix metalloproteinase inhibitor. 19261. The method of item 19198 wherein the anti- fibrotic agent is an MCP-CCR2 inhibitor.
19262. The method of item 19198 wherein the anti- fibrotic agent is an mTOR inhibitor.
19263. The method of item 19198 wherein the anti- fibrotic agent is an mTOR kinase inhibitor.
19264. The method of item 19198 wherein the anti- fibrotic agent is a microtubule inhibitor selected from the group consisting of antibody-maytansinoid conjugates from Biogen Idee, colchicines (MantiCore Pharmaceuticals), anticancer immunoconjugates from Johnson & Johnson, DIME from Octamer, gni-1f (GNI), huC242-DM4, huMy9-6-DM1 (immunoGen), IDN-5404 (Indena), IMO-098, IMOderm (Imotep), mebendazole (Introgen Therapeutics), microtubule poisons from Cambridge Enterprise, paclitaxel such as LOTAX from Aphios (CAS No. 33069-62-4), Genexol-PM from Samyang, Pharmaprojects No. 6383 (Tapestry Pharmaceuticals), RPR-112378 (Sanofi-Aventis), SGN-75 (Seattle Genetics), SPL-7435 (Starpharma), SSR-250411 (Sanofi-Aventis), trastuzumab-DM1 (Genentech), vinorelbine, and an analogue or derivative thereof.
19265. The method of item 19198 wherein the anti- fibrotic agent is an MIF inhibitor.
19266. The method of item 19198 wherein the anti- fibrotic agent is an MMP inhibitor.
19267. The method of item 19198 wherein the anti- fibrotic agent is a neurokinin (NK) antagonist selected from the group consisting of anthrotainin (CAS No. 148084-40-6) (Sanofi-Aventis), an IBS thereapeutic from ArQuIe, MDL-105212A (CAS No. 167261-60-1) (Ssanofi- Aventis), Pharmaprojects No. 2744, 3258 (CAS No. 139167-47-8) 4006, 4201, or 5986 (Sanofi-Aventis), RP 67580 (CAS No. 135911-02-3), SR- 144190 (CAS No. 201152-86-5), SSR-240600, SSR-241586 (Sanofi- Aventis), TKA-457 (Novartis), vestipitant mesylate (CAS No. 334476-64-1) (GlaxoSmithKline), Win-64821 (Sanofi-Aventis), PRX-96026 (Predix Pharmaceuticals), and an analogue or derivative thereof.
19268. The method of item 19198 wherein the anti- fibrotic agent is an NF kappa B inhibitor selected from the group consisting of AVE-0545 or AVE-0547 (Sanofi-Aventis), bortezomib (CAS No. 179324- 69-7) (Millennium Pharmaceuticals), dexanabinol (CAS No. 112924-45-5) (Pharmos), dexlipotam (Viatris), Pharmaprojects No. 6283 (INDRA) (OXiGENE), IPL-576092 (CAS No. 137571-30-3) (Inflazyme), NFKB decoy (Corgentech), NFKB decoy oligo (AnGes MG), NFKB's from Ariad, osteoporosis treatments or S5 (F005) from Fulcrum Pharmaceuticals, P61 (Phytopharm), R-flurbiprofen (CAS No. 5104-49-4) (Encore Pharmaceuticals), and an analogue or derivative thereof.
19269. The method of item 19198 wherein the anti- fibrotic agent is a nitric oxide agonist.
19270. The method of item 19198 wherein the anti- fibrotic agent is an ornithine decarboxylase inhibitor.
19271. The method of item 19198 wherein the anti- fibrotic agent is a p38 MAP kinase inhibitor selected from the group consisting of AZD-6703 (AstraZeneca), JX-401 (Jexys Pharmaceuticals), BMS-2 (Bristol-Myers Squibb), a p38 MAP kinase inhibitor from Novartis, a p38-alpha MAP kinase inhibitor from Amphora, Pharmaprojects No. 5704 (Pharmacopeia), RPR-200765A (Sanofi-Aventis), SD-282 (Johnson & Johnson), TAK-715 (Takeda), and an analogue or derivative thereof.
19272. The method of item 19198 wherein the anti- fibrotic agent is a palmitoyl-protein thioesterase inhibitor.
19273. The method of item 19198 wherein the anti- fibrotic agent is a PDGF receptor kinase inhibitor selected from the group consisting of AAL-993, AMN-107, or ABP-309 (Novartis), AMG-706 (Amgen), BAY-57-9352 (Bayer), CDP-860 (UCB), E-7080 (Eisai), imatinib (CAS No. 152459-95-5) (Novartis), OSI-930 (OSI Pharmaceuticals), RPR- 127963E (Sanofi-Aventis), RWJ-540973 (Johnson & Johnson), sorafenib tosylate (Bayer), SU-11657 (Pfizer), tandutinib (CAS No. 387867-13-2) (Millennium Pharmaceuticals), vatalanib (Novartis), ZK-CDK (Schering AG), and an analogue or derivative thereof.
19274. The method of item 19198 wherein the anti- fibrotic agent is (-)-halofenate (Metabolex), AMG-131 (Amgen), antidiabetics from Japan Tobacco, AZD-4619, AZD-8450, AZD-8677 (AstraZeneca), DRF-10945, balaglitazone (Dr Reddy's), CS-00088, CS-00098 (Chipscreen Biosciences), E-3030 (Eisai), etalocib (CAS No. 161172-51-6) (EIi Lilly), GSK-641597 (Ligand), GSK-677954 (GlaxoSmithKline), GW-409544 (Ligand), GW-590735 (GlaxoSmithKline), K-111 (Hoffmann-La Roche), LY- 518674 (EIi Lilly), LY-674 (Ligand), LY-929 (Ligand), MC-3001, MC-3002 (MaxoCore Pharmaceuticals), metformin HCI + pioglitazone (CAS No. 1115- 70-4 and 112529-15-4), ACTOPLUS MET from Andrx), muraglitazar (CAS No. 331741-94-7) (Bristol-Myers Squibb), naveglitazar (Ligand), oleoylethanolamide (Kadmus Pharmaceuticals), ONO-5129, pioglitazone hydrochloride (CAS No. 111025-46-8 and 112529-15-4) (Takeda), PLX-204 (Plexxikon), PPAR agonists from Genfit, PPAR delta agonists from EIi Lilly, PPAR-alpha agonists from CrystalGenomics, PPAR-gamma modulators and PPAR-β modulators from CareX, rosiglitazone maleate (CAS No. 122324- 73-4 or 155141-29-0) (GlaxoSmithKline), rosiglitazone maleate/glimepir (CAS No. 155141-29-0 and 93479-97-1), AVANDARYL, rosiglitazone maleate/metformin extend (CAS No. 155141-29-0 and 657-24-9), AVANDAMET, rosiglitazone maleate+metformin, AVANDAMET (GlaxoSmithKline), tesaglitazar (AstraZeneca), and an analogue or derivative thereof.
19275. The method of item 19198 wherein the anti- fibrotic agent is a phosphatase inhibitor.
19276. The method of item 19198 wherein the anti- fibrotic agent is a phosphodiesterase (PDE) inhibitor selected from the group consisting of avanafil (Tanabe Seiyaku), dasantafil (CAS No. 569351- 91-3) (Schering-Plough), A-906119 (CAS No. 134072-58-5), DL-850 (Sanofi-Aventis), GRC-3015, GRC-3566, GRC-3886 (Glenmark), HWA-153 (CAS No. 56395-66-5) (Sanofi-Aventis), hydroxypumafentrine (Altana), IBFB-130011 , IBFB-14-016, IBFB-140301 , IBFB-150007, IBFB-211913 (IBFB Pharma), L-826141 (Merck & Co), medorinone (CAS No. 88296-61-1) (Sanofi-Aventis), MEM-1917 (Memory Pharmaceuticals), ND-1251 (Neuro3d), PDE inhibitors from ICOS, PDE IV inhibitors from Memory Pharmaceuticals and CrystalGenomics, Pharmaprojects No. 2742 and 6141 (Sanofi-Aventis), QAD-171 (Novartis), RHC-2963 (CAS No. 76993-12-9 and 76993-14-1), RPR-117658, RPR-122818 derivatives, SR-24870 , and RPR- 132294 (Sanofi-Aventis), SK-350 (ln2Gen), stroke therapy agents from deCODE Genetics, TAS-203 (Taiho), tofimilast (CAS No. 185954-27-2) (Pfizer), UK-371800 (Pfizer), WlN-65579 (CAS No. 158020-82-7) (Sanofi- Aventis), IBFB-130020 (IBFB Pharma), OPC-6535 (CAS No. 145739-56-6) (Otsuka), a phosphodiesterase III inhibitor, enoximone, a phosphodiesterase IV inhibitor, fosfosal, Atopik (Barrier Therapeutics), triflusal, a phosphodiesterase V inhibitor, and an analogue or derivative thereof.
19277. The method of item 19198 wherein the anti- fibrotic agent is a PKC inhibitor.
19278. The method of item 19198 wherein the anti- fibrotic agent is a platelet activating factor antagonist.
19279. The method of item 19198 wherein the anti- fibrotic agent is a platelet-derived growth factor receptor kinase inhibitor.
19280. The method of item 19198 wherein the anti- fibrotic agent is a prolyl hydroxylase inhibitor.
19281. The method of item 19198 wherein the anti- fibrotic agent is a polymorphonuclear neutrophil inhibitor.
19282. The method of item 19198 wherein the anti- fibrotic agent is a protein kinase B inhibitor.
19283. The method of item 19198 wherein the anti- fibrotic agent is a protein kinase C stimulant.
19284. The method of item 19198 wherein the anti- fibrotic agent is a purine nucleoside analogue.
19285. The method of item 19198 wherein the anti- fibrotic agent is a purinoreceptor P2X antagonist. 19286. The method of item 19198 wherein the anti- fibrotic agent is a Raf kinase inhibitor.
19287. The method of item 19198 wherein the anti- fibrotic agent is a reversible inhibitor of ErbB1 and ErbB2.
19288. The method of item 19198 wherein the anti- fibrotic agent is a ribonucleoside triphosphate reductase inhibitor.
19289. The method of item 19198 wherein the anti- fibrotic agent is an SDF-1 antagonist.
19290. The method of item 19198 wherein the anti- fibrotic agent is a sheddase inhibitor.
19291. The method of item 19198 wherein the anti- fibrotic agent is an SRC inhibitor.
19292. The method of item 19198 wherein the anti- fibrotic agent is a stromelysin inhibitor.
19293. The method of item 19198 wherein the anti- fibrotic agent is an Syk kinase inhibitor.
19294. The method of item 19198 wherein the anti- fibrotic agent is a telomerase inhibitor.
19295. The method of item 19198 wherein the anti- fibrotic agent is a TGF beta inhibitor selected from the group consisting of pirfenidone (CAS No. 53179-13-8) (MARNAC), tranilast (CAS No. 53902- 12-8) (Kissei), IN-1130 (ln2Gen), mannose-6-phosphate (BTG), TGF-β antagonists from Inflazyme (Pharmaprojects No. 6075), TGF-β antagonists from Sydney, non-industrial source), TGF-β I receptor kinase inhibitors from EIi Lilly, TGF-β receptor inhibitors from Johnson & Johnson, and an analogue or derivative thereof.
19296. The method of item 19198 wherein the anti- fibrotic agent is a TNFα antagonist or TACE inhibitor selected from the group consisting of adalimumab (CAS No. 331731-18-1) (Cambridge Antibody Technology), AGIX-4207 (AtheroGenics), AGT-1 (Advanced Biotherapy), an anti-inflammatory from Borean Pharma, Cellzome, or Paradigm Therapeutics, anti-inflammatory vaccine (TNF-alpha kinoid) from Neovacs, humanized anti-TNF antibody or an anti-TNF MAb (CB0006) Celltech (UCB), apratastat (CAS No. 287445-51-0) (Wyeth), BMS-561392 (Bristol-Myers Squibb), BN-006 (Bone), certolizumab pegol (CAS No. 428863-50-7 or CH-138 (UCB), cilomilast (CAS No. 153259-65-5) (GlaxoSmithKline), CR-1 (Nuada Pharmaceuticals), CRx-119 (CombinatoRx), D-5410 (UCB), dacopafant (CAS No. 125372-33-0) (Sanofi- Aventis), dersalazine (CAS No. 188913-57-7/188913-58-8) (Uriach), etanercept (CAS No. 185243-69-0) (Amgen), ethyl pyruvate (Critical (Critical Therapeutics), golimumab (CAS No. 476181-74-5) (Johnson & Johnson), hormono-immunotherapy from Ipsen, CDP571 (e.g., Humicade from UCB), IC-485 (ICOS), infliximab (CAS No. 170277-31-3) (Johnson & Johnson), IP- 751 (Manhattan Pharmaceuticals), ISIS-104838 (CAS No. 250755-32-9) (ISIS Pharmaceuticals), lenalidomide (CAS No. 191732-72-6) (Celgene), lentinan (CAS No. 37339-90-5) (Ajinomoto), MDL-201112 (CAS No. 142130-73-2) (Sanofi-Aventis), medroxyprogesterone (CAS No. 520-85-4) (InKine Pharmaceutical), N-acetylcysteine (CAS No. 616-91-1) (Zambon), NBE-P2 (DIREVO Biotech), nerelimomab (CAS No. 162774-06-3) (Chiron), OM-294DP (OM PHARMA), onercept (CAS No. 199685-57-9) (Yeda), PASSTNF-alpha (Verigen), pentoxifylline or oxypentifylline (Sanofi-Aventis), Pharmaprojects No. 4091 , 4241 , 4295, or 4488 (Sanofi-Aventis), Pharmaprojects No. 5480 (Amgen), Pharmaprojects No. 6749 (Cengent), pirfenidone (CAS No. 53179-13-8) (MARNAC), RPR-132294 (Sanofi- Aventis), S5 (F002) (Fulcrum Pharmaceuticals), simvastatin (CAS No. 79902-63-9) (Merck & Co), STA-6292 (Synta Pharmaceuticals), tacrolimus (CAS No. 104987-11-3) (Fujisawa LifeCycle Pharma), talactoferrin alfa (CAS No. 308240-58-6) (Agennix), thalidomide (CAS No. 50-35-1) (Celgene), TNF antagonists form ProStrakan, and Synergen, TNF inhibitors (Amgen), TNF-alpha antagonists from Dynavax Technologies and Jerina AG (Germany), TNF-alpha inhibitors from IBFB Pharma and Xencor (Xencor), torbafylline (CAS No. 105102-21-4) (Sanofi-Aventis), UR-1505 (Uriach), VT-346 (Viron Therapeutics), YSIL6 (Y's Therapeutics), YSTH2 (Y's Therapeutics), NPI-1302a-3 (Nereus Pharmaceuticals, a TNF antagonist from Jerina AG (Germany), dersalazine, and an analogue or derivative thereof.
19297. The method of item 19198 wherein the anti- fibrotic agent is a Toll receptor inhibitor.
19298. The method of item 19198 wherein the anti- fibrotic agent is a tubulin antagonist.
19299. The method of item 19198 wherein the anti- fibrotic agent is a tyrosine kinase inhibitor selected from the group consisting of SU-011248, SUTENT from Pfizer Inc. (New York, NY), BMS-354825, PN- 355 (Paracelsian Pharmaceuticals), AGN-199659 (Allergan), AAL-993 or ABP-309 (Novartis), adaphostin (NIH), AEE-788 (Novartis), AG-013736 (OSI Pharmaceuticals), AG-13736 (Pfizer), ALT-110 (Alteris Therapeutics), AMG-706 (Amgen), anticancer MAbs from Xencor, anti-EGFrvlll MAbs from Abgenix, anti-HER2 MAb from Abiogen, AZD-2171 or AZD-9935 (AstraZeneca), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), CEP-5214 (Cephalon), CEP-7055 (Cephalon), cetuximab (ImClone Systems), CHIR-200131 and CHIR-258 (Chiron), CP-547632 (OSI Pharmaceuticals), CP-724714 (Pfizer), CT-301 (Creabilis Therapeutics), D- 69491 (Baxter International), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EGFR/ErbB2 inhibitors from Array BioPharma, erlotinib (CAS No. 183319- 69-9) (OSI Pharmaceuticals), EXEL-2880 (Exelixis), FK-778 (Sanofi- Aventis) , gefitinib (CAS No. 184475-35-2) (AstraZeneca), GW-2286 or GW- 654652 (GlaxoSmithKline), her2/neu antigen from AlphaVax, HER-2/neu inhibitor from Generex, Herzyme (Medipad) (Sirna Therapeutics), HKI-272 (Wyeth), HuMax-EGFr (Genmab), idronoxil (CAS No. 81267-65-4) (Novogen), IGF-1 inhibitors from Ontogen, IMC-11F8 (ImClone Systems), kahalalide F (CAS No. 149204-42-2) (PharmaMar), KDR inhibitor from LG Life Sciences, KDR inhibitors from Abbott Laboratories, KDR kinase inhibitors (UCB), Kdr kinase inhibitors from Merck & Co, KRN-633 and KRN- 951 (Kirin Brewery), KSB-102 (Xenova), lapatinib ditosylate (CAS No. 388082-78-8) (GlaxoSmithKline), matuzumab (Merck KGaA), MDX-214 (Medarex), ME-103 (Pharmexa), MED-A300 (Gerolymatos), MNAC-13 (Lay Line Genomics), nimotuzumab (Center of Molecular Immunology), NSC- 330507 or NSC-707545 (NIH), NV-50 (Novogen), OSI-930 (OSI Pharmaceuticals), panitumumab (Abgenix), pelitinib (CAS No. 287933-82-7) (Wyeth), pertuzumab (CAS No. 380610-27-5) (Genentech), Pharmaprojects No. 3985 (Sanofi-Aventis), prostate cancer therapeutics from Sequenom (SQPC35, SQPC36, SQPC90), removab and remoxab (Trion Pharma), RG- 13022 (CAS No. 136831-48-6), RG-13291 (CAS No. 138989-50-1), or RG- 14620 (CAS No. 136831-49-7) (Sanofi-Aventis), RM-6427 (Romark), RNAi breast cancer therapy from Benitec, RP 53801 (CAS No. 125882-88-4) (Sanofi-Aventis), sorafenib tosylate (Bayer), SU-11657 (Pfizer), Tie-2 antagonists from Semaia (Hybrigenics), Tie-2 inhibitors from Ontogen, trastuzumab (CAS No. 180288-69-1) (Genentech), tyrosine kinase inhibitors from Sanofi-Aventis, U3-1287, U3-1565, U3-1784, U3-1800 (U3 Pharma), vatalanib (Novartis), VEGFR-2 kinase inhibitor from Bristol-Myers Squibb, XL-647 (Exelixis), ZD-6474 (AstraZeneca), ZK-CDK (Schering AG), an EGFR tyrosine kinase inhibitor, EKB-569 (Wyeth), and an analogue or derivative thereof.
19300. The method of item 19198 wherein the anti- fibrotic agent is a VEGF inhibitor.
19301. The method of item 19198 wherein the anti- fibrotic agent is a vitamin D receptor agonist.
19302. The method of item 19198 wherein the anti- fibrotic agent is ZD-6474 (an angiogenesis inhibitor).
19303. The method of item 19198 wherein the anti- fibrotic agent is AP-23573 (an mTOR inhibitor).
19304. The method of item 19198 wherein the anti- fibrotic agent is synthadotin (a tubulin antagonist).
19305. The method of item 19198 wherein the anti- fibrotic agent is S-0885 (a collagenase inhibitor).
19306. The method of item 19198 wherein the anti- fibrotic agent is aplidine (an elongation factor-1 alpha inhibitor).
19307. The method of item 19198 wherein the anti- fibrotic agent is ixabepilone (an epithilone).
19308. The method of item 19198 wherein the anti- fibrotic agent is IDN-5390 (an angiogenesis inhibitor). 19309. The method of item 19198 wherein the anti- fibrotic agent is SB-2723005 (an angiogenesis inhibitor).
19310. The method of item 19198 wherein the anti- fibrotic agent is ABT-518 (an angiogenesis inhibitor).
19311. The method of item 19198 wherein the anti- fibrotic agent is combretastatin (an angiogenesis inhibitor).
19312. The method of item 19198 wherein the anti- fibrotic agent is anecortave acetate (an angiogenesis inhibitor).
19313. The method of item 19198 wherein the anti- fibrotic agent is SB-715992 (a kinesin antagonist).
19314. The method of item 19198 wherein the anti- fibrotic agent is temsirolimus (an mTOR inhibitor).
19315. The method of item 19198 wherein the anti- fibrotic agent is adalimumab (a TNFα antagonist).
19316. The method of item 19198 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with an anti-fibrotic agent, and (b) implanting the medical device into the host.
19317. The method of item 19198 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with an anti-infective agent, and (b) implanting the medical device into the host. 19318. The method of item 19198 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with a polymer; and (b) implanting the medical device into the host.
19319. The method of item 19198 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with a composition comprising an anti-fibrotic agent and a polymer, and (b) implanting the medical device into the host.
19320. The method of item 19198 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with a composition comprising an anti-infective agent and a polymer, and (b) implanting the medical device into the host.
19321. The method of item 19198 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with a composition comprising an anti-fibrotic agent, an anti- infective agent and a polymer, and (b) implanting the medical device into the host.
19322. The method of item 19198 wherein the anti- infective agent is an anthracycline.
19323. The method of item 19198 wherein the anti- infective agent is doxorubicin.
19324. The method of item 19198 wherein the anti- infective agent is is mitoxantrone. 19325. The method of item 19198 wherein the anti- infective agent is a fluoropyrimidine.
19326. The method of item 19198 wherein the anti- infective agent is 5-fluorouracil (5-FU).
19327. The method of item 19198 wherein the anti- infective agent is a folic acid antagonist.
19328. The method of item 19198 wherein the anti- infective agent is methotrexate.
19329. The method of item 19198 wherein the anti- infective agent is a podophylotoxin.
19330. The method of item 19198 wherein the anti- infective agent is etoposide.
19331. The method of item 19198 wherein the anti- infective agent is camptothecin.
19332. The method of item 19198 wherein the anti- infective agent is a hydroxyurea.
19333. The method of item 19198 wherein the anti- infective agent is a platinum complex.
19334. The method of item 19198 wherein the anti- infective agent is cisplatin. 19335. The method of item 19198 wherein the composition comprises an antithrombotic agent.
19336. The method of item 19198 wherein the polymer is formed from reactants comprising a naturally occurring polymer.
19337. The method of item 19198 wherein the polymer is formed from reactants comprising protein.
19338. The method of item 19198 wherein the polymer is formed from reactants comprising carbohydrate.
19339. The method of item 19198 wherein the polymer is formed from reactants comprising biodegradable polymer.
19340. The method of item 19198 wherein the polymer is formed from reactants comprising nonbiodegradable polymer.
19341. The method of item 19198 wherein the polymer is formed from reactants comprising collagen.
19342. The method of item 19198 wherein the polymer is formed from reactants comprising methylated collagen.
19343. The method of item 19198 wherein the polymer is formed from reactants comprising fibrinogen.
19344. The method of item 19198 wherein the polymer is formed from reactants comprising thrombin. 19345. The method of item 19198 wherein the polymer reactants comprising blood plasma.
19346. The method of item 19198 wherein the polymer reactants comprising calcium salt.
19347. The method of item 19198 wherein the polymer reactants comprising an antifibronolytic agent.
19348. The method of item 19198 wherein the polymer reactants comprising fibrinogen analog.
19349. The method of item 19198 wherein the polymer reactants comprising albumin.
19350. The method of item 19198 wherein the polymer reactants comprising plasminogen.
19351. The method of item 19198 wherein the polymer reactants comprising von Willebrands factor.
19352. The method of item 19198 wherein the polymer reactants comprising Factor VIII.
19353. The method of item 19198 wherein the polymer reactants comprising hypoallergenic collagen.
19354. The method of item 19198 wherein the polymer reactants comprising atelopeptidic collagen. 19355. The method of item 19198 wherein the polymer is formed from reactants comprising telopeptide collagen.
19356. The method of item 19198 wherein the polymer is formed from reactants comprising crosslinked collagen.
19357. The method of item 19198 wherein the polymer is formed from reactants comprising aprotinin.
19358. The method of item 19198 wherein the polymer is formed from reactants comprising epsilon-amino-n-caproic acid.
19359. The method of item 19198 wherein the polymer is formed from reactants comprising gelatin.
19360. The method of item 19198 wherein the polymer is formed from reactants comprising protein conjugates.
19361. The method of item 19198 wherein the polymer is formed from reactants comprising gelatin conjugates.
19362. The method of item 19198 wherein the polymer is formed from reactants comprising a synthetic polymer.
19363. The method of item 19198 wherein the polymer is formed from reactants comprising a synthetic isocyanate-containing compound.
19364. The method of item 19198 wherein the polymer is formed from reactants comprising a synthetic thiol-containing compound. 19365. The method of item 19198 wherein the polymer is formed from reactants comprising a synthetic compound containing at least two thiol groups.
19366. The method of item 19198 wherein the polymer is formed from reactants comprising a synthetic compound containing at least three thiol groups.
19367. The method of item 19198 wherein the polymer is formed from reactants comprising a synthetic compound containing at least four thiol groups.
19368. The method of item 19198 wherein the polymer is formed from reactants comprising a synthetic amino-containing compound.
19369. The method of item 19198 wherein the polymer is formed from reactants comprising a synthetic compound containing at least two amino groups.
19370. The method of item 19198 wherein the polymer is formed from reactants comprising a synthetic compound containing at least three amino groups.
19371. The method of item 19198 wherein the polymer is formed from reactants comprising a synthetic compound containing at least four amino groups.
19372. The method of item 19198 wherein the polymer is formed from reactants comprising a synthetic compound comprising a carbonyl-oxygen-succinimidyl group. 19373. The method of item 19198 wherein the polymer is formed from reactants comprising a synthetic compound comprising at least two carbonyl-oxygen-succinimidyl groups.
19374. The method of item 19198 wherein the polymer is formed from reactants comprising a synthetic compound comprising at least three carbonyl-oxygen-succinimidyl groups.
19375. The method of item 19198 wherein the polymer is formed from reactants comprising a synthetic compound comprising at least four carbonyl-oxygen-succinimidyl groups.
19376. The method of item 19198 wherein the polymer is formed from reactants comprising a synthetic polyalkylene oxide- containing compound.
19377. The method of item 19198 wherein the polymer is formed from reactants comprising a synthetic compound comprising both polyalkylene oxide and biodegradable polyester blocks.
19378. The method of item 19198 wherein the polymer is formed from reactants comprising a synthetic polyalkylene oxide- containing compound having reactive amino groups.
19379. The method of item 19198 wherein the polymer is formed from reactants comprising a synthetic polyalkylene oxide- containing compound having reactive thiol groups.
19380. The method of item 19198 wherein the polymer is formed from reactants comprising a synthetic polyalkylene oxide- containing compound having reactive carbonyl-oxygen-succinimidyl groups. 19381. The method of item 19198 wherein the polymer is formed from reactants comprising a synthetic compound comprising a biodegradable polyester block.
19382. The method of item 19198 wherein the polymer is formed from reactants comprising a synthetic polymer formed in whole or part from lactic acid or lactide.
19383. The method of item 19198 wherein the polymer is formed from reactants comprising a synthetic polymer formed in whole or part from glycolic acid or glycolide.
19384. The method of item 19198 wherein the polymer is formed from reactants comprising polylysine.
19385. The method of item 19198 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound comprising a polyalkylene oxide portion.
19386. The method of item 19198 wherein the polymer is formed from reactants comprising (a) protein and (b) polylysine.
19387. The method of item 19198 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound having at least four thiol groups.
19388. The method of item 19198 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound having at least four amino groups. 19389. The method of item 19198 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound having at least four carbonyl-oxygen-succinimide groups.
19390. The method of item 19198 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound having a biodegradable region formed from reactants selected from lactic acid, lactide, glycolic acid, glycolide, and epison-caprolactone.
19391. The method of item 19198 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound comprising a polyalkylene oxide portion.
19392. The method of item 19198 wherein the polymer is formed from reactants comprising (a) collagen and (b) polylysine.
19393. The method of item 19198 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound having at least four thiol groups.
19394. The method of item 19198 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound having at least four amino groups.
19395. The method of item 19198 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound having at least four carbonyl-oxygen-succinimide groups.
19396. The method of item 19198 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound having a biodegradable region formed from reactants selected from lactic acid, lactide, glycolic acid, glycolide, and epison-caprolactone.
19397. The method of item 19198 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound comprising a polyalkylene oxide portion.
19398. The method of item 19198 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) polylysine.
19399. The method of item 19198 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound having at least four thiol groups.
19400. The method of item 19198 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound having at least four amino groups.
19401. The method of item 19198 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound having at least four carbonyl-oxygen-succinimide groups.
19402. The method of item 19198 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound having a biodegradable region formed from reactants selected from lactic acid, lactide, glycolic acid, glycolide, and epison-caprolactone.
19403. The method of item 19198 wherein the polymer is formed from reactants comprising hyaluronic acid. 19404. The method of item 19198 wherein the polymer is formed from reactants comprising a hyaluronic acid derivative.
19405. The method of item 19198 wherein the polymer is formed from reactants comprising pentaerythritol poly(ethylene glycol)ether tetra-sulfhydryl of number average molecular weight between 3,000 and 30,000.
19406. The method of item 19198 wherein the polymer is formed from reactants comprising pentaerythritol poly(ethylene glycol)ether tetra-amino of number average molecular weight between 3,000 and 30,000.
19407. The method of item 19198 wherein the polymer is formed from reactants comprising (a) a synthetic compound having a number average molecular weight between 3,000 and 30,000 and comprising a polyalkylene oxide region and multiple nucleophilic groups, and (b) a synthetic compound having a number average molecular weight between 3,000 and 30,000 and comprising a polyalkylene oxide region and multiple electrophilic groups.
19408. The method of item 19198 wherein the composition comprises a colorant.
19409. The method of item 19198 wherein the composition is sterile.
19410. A method as in any one of items 19198-19409, wherein the device is a device for treating hypertropic scar or keloid that comprises an external tissue expansion device. 19411. A method as in any one of items 19198-19409, wherein the device is a device for treating hypertropic scar or keloid that comprises a masking element, and wherein the masking element may be pressed onto the scar tissue.
19412. A method as in any one of items 19198-19409, wherein the device is a device for treating hypertropic scar or keloid that comprises a locking element and a grasping structure so that the dermal and epidermal layers of a skin wound can be pushed together.
19413. A method for implanting a medical device comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with i) an anti-fibrotic agent, ii) an anti-infective agent, iii) a polymer, iv) a composition comprising an anti-fibrotic agent and a polymer, v) a composition comprising an anti-infective agent and a polymer, or vi) a composition comprising an anti-fibrotic agent, an anti- infective agent and a polymer, and (b) implanting the medical device into the host, wherein the medical device is a vascular graft.
19414. The method of item 19413 wherein the anti- fibrotic agent is an adensosine A2A receptor antagonist.
19415. The method of item 19413 wherein the anti- fibrotic agent is an AKT inhibitor.
19416. The method of item 19413 wherein the anti- fibrotic agent is an alpha 2 integrin antagonist, wherein the alpha 2 integrin antagonist is Pharmaprojects No. 5754 (Merck KgaA).
19417. The method of item 19413 wherein the anti- fibrotic agent is an alpha 4 integrin antagonist. 19418. The method of item 19413 wherein the anti- fibrotic agent is an alpha 7 nicotinic receptor agonist.
19419. The method of item 19413 wherein the anti- fibrotic agent is an angiogenesis inhibitor selected from the group consisting of AG-12,958 (Pfizer), ATN-161 (Attenuon LLC), neovastat, an angiogenesis inhibitor from Jerina AG (Germany), NM-3 (Mercian), VGA-1155 (Taisho), FCE-26644 (Pfizer), FCE-26950 (Pfizer), FPMA (Meiji Daries), FR-111142 (Fujisawa), GGTI-298, GM-1306 (Ligand), GPA-1734 (Novartis), NNC-47- 0011 (Novo Nordisk), herbamycin (Nippon Kayaku), lenalidomide (Celegene), IP-10 (NIH), ABT-828 (Abbott), KIN-841 (Tokushima University, Japan), SF-1126 (Semafore Pharmaceuticals), laminin technology (NIH), CHIR-258 (Chiron), NVP-AEW541 (Novartis), NVP-AEW541 (Novartis), Vt16907 (Alchemia), OXI-8007 (Oxigene), EG-3306 (Ark Therapeutics), Maspin (Arriva), ABT-567 (Abbott), PPI-2458 (Praecis Pharmaceuticals), CC-5079, CC-4089 (Celgene), HIF-1 alpha inhibitors (Xenova), S-247 (Pfizer), AP-23573 (Ariad), AZD-9935 (Astra Zeneca), mebendazole (Introgen Therapeutics), MetAP-2 inhibitors (GlaxoSmithKline), AG-615 (Angiogene Pharmaceuticals), Tie-2 antagonists (Hybrigenics), NC-381, CYC-381 , NC-169, NC-219, NC-383, NC-384, NC-407 (Lorus Therapeutics), ATN-224 (Attenuon), ON-01370 (Onconova), Vitronectin antagonists (Amgen), SDX-103 (Salmedix), Vitronectin antagonists (Shire), CHP (Riemser), TEK (Amgen), Anecortave acetate (Alcon), T46.2 (Matrix Therapeutics), HG-2 (Heptagen), TEM antagonists (Genzyme), Oxi-4500 (Oxigene), ATN-161 (Attenuon), WX-293 (Wilex), M-2025 (Metris Therapeutics), Alphastatin (BioActa), YH-16 (Yantai Rongchang), BIBF- 1120 (Boehringer Ingelheim), BAY-57-9352 (Bayer), AS-1404 (Cancer Research Technology), SC-77964 (Pfizer), glycomimetics (BioTie Therapies), TIE-2 Inhibitors (Ontogen), DIMI, Octamer (Octamer), ABR- 215050 (Active Biotech), ABT-518 (Abbott), KDR inhibitors (Abbott), BSF- 466895 (Abbott), SCH-221153 (Schering-Plough), DAC:antiangiogenic (ConjuChem), TFPI (EntreMed), AZD-2171 (Astra-Zenaca), CDC-394 (Celgene), LY290293 (EIi Lilly), IDN-5390 (Indena), Kdr Kinase Inhibitors (Merck), CT-113020, CT-116433, CT-116563, CT-31890, CT-32228) (Cell Therapeutics), A-299620 (Abbott), TWEAK Inhibitor (Amgen), VEGF modulators (Johnson and Johnson), Tum~N53, tumstatin (Genzyme), Thios- 1 , Thios-2 (Thios Pharmaceuticals), MV-6401 (Miravant Medical Technologies), Spisulosine (PharmaMar), CEP-7055 (Cephalon), AUV-201 (Auvation), LM-609 (EIi Lilly), SKF-106615 (AnorMED), Oglufanide disodium (Cytran), BW-114 (Pharminox), Calreticulin (NIH)1 WX-678 (Wilex), SD-7784 (Pfizer), WX-UK1 (Wilex), SH-268 (Schering AG), 2-Me-PGA (Celgene), S- 137 (Pfizer), ZD-6126 (Angiogene Pharmaceuticals), SG-292 (SignalGen), Benefin (Lane Labs), A6, A36 (Angstrom), SB-2723005 (GlaxoSmithKline), SC-7 (Cell Therapeutics), ZEN-014 (AEterna Zentaris), 2-methoxyestradiol (EntreMed), NK-130119 (Nippon Kayaku), CC-10004 (Celgene), AVE- 8062A (Ajinomoto), Tacedinaline (Pfizer), Actinonin (Tokyo Metropolitan Institute of Medical Science), Lenalidomide (Celgene), VGA-1155, BTO-956 (SRI International), ER-68203-00 (Eisai), CT-6685 (UCB), JKC-362 (Phoenix Pharmaceuticals), DMI-3798 (DMI Biosciences, Angiomate (Ipsen), ZD-6474 (AstraZeneca), CEP-5214 (Cephalon), Canstatin (Genzyme), NM-3 (Mercian), Oridigm (MediQuest Therapeutics), Exherin (Adherex), BLS-0597 (Boston Life Sciences), PTC-299 (PTC Therapeutics), NPI-2358 (Nereus Pharmaceuticals), CGP-79787 (Novartis), AEE-788 (Novartis), CKD-732 (Chong Kun Dang), CP-564959 (OSI Pharmaceuticals), CM-101 (CarboMed), CT-2584, CT3501 (Cell Therapeutics), combretastatin and analogues and derivatives thereof (Oxigene), Rebimastat (Bristol-Meyers Squibb), Dextrin 2-sulfate (ML Laboratories), Cilengitide (Merk KGaA), NSC-706704 (Pharminox), KRN- 951 (Kirin Brewery), Ukrain, NSC-631570 (Nowicky Pharma), Tecogalan sodium (Daiichi Pharmaceutical), Tz-93 (Tsumura), TBC-1635 (Encysive Pharmaceuticals), TAN-1120 (Takeda), Semaxanib (Pfizer), BDI-7800 (Biopharmacopae), SD-186, SD-983 (Bristol-Meyers Squibb), SB-223245 (GlaxoSmithKline), SC-236 (Pfizer), RWJ-590973 (Johnson and Johnson), ILX-1850 (Genzyme), SC-68488, S-836 (Pfizer), CG-55069-11 (CuraGen), Ki-23057 (Kirin Brewery), CCX-700 (Chemoentryx), Pegaptanib octasodium (Giled Sciences), ANGIOCOL (available from Biostratum Inc.), or an analogue or derivative thereof.
19420. The method of item 19413 wherein the anti- fibrotic agent is an apoptosis antagonist.
19421. The method of item 19413 wherein the anti- fibrotic agent is an apoptosis activator.
19422. The method of item 19413 wherein the anti- fibrotic agent is a beta 1 integrin antagonist.
19423. The method of item 19413 wherein the anti- fibrotic agent is a beta tubulin inhibitor.
19424. The method of item 19413 wherein the anti- fibrotic agent is a blocker of enzyme production in Hepatitis C.
19425. The method of item 19413 wherein the anti- fibrotic agent is a Bruton's tyrosine kinase inhibitor.
19426. The method of item 19413 wherein the anti- fibrotic agent is a calcineurin inhibitor.
19427. The method of item 19413 wherein the anti- fibrotic agent is a caspase 3 inhibitor. 19428. The method of item 19413 wherein the anti- fibrotic agent is a CC chemokine receptor antagonist.
19429. The method of item 19413 wherein the anti- fibrotic agent is a cell cycle inhibitor selected from the group consisting of SNS-595 (Sunesis), synthadotin, KRX-0403, and an analogue or derivative thereof.
19430. The method of item 19413 wherein the anti- fibrotic agent is a cathepsin B inhibitor.
19431. The method of item 19413 wherein the anti- fibrotic agent is a cathepsin K inhibitor, wherein the cathepsin K inhibitor is 462795 (GlaxoSmithKline), INPL-022-D6 (Amura Therapeutics), or an analogue or derivative thereof.
19432. The method of item 19413 wherein the anti- fibrotic agent is a cathepsin L inhibitor.
19433. The method of item 19413 wherein the anti- fibrotic agent is a CD40 antagonist.
19434. The method of item 19413 wherein the anti- fibrotic agent is a chemokine receptor agonist.
19435. The method of item 19413 wherein the anti- fibrotic agent is a chymase inhibitor.
19436. The method of item 19413 wherein the anti- fibrotic agent is a collagenase antagonist. 19437. The method of item 19413 wherein the anti- fibrotic agent is a CXCR antagonist.
19438. The method of item 19413 wherein the anti- fibrotic agent is a cyclin dependent kinase inhibitor selected from the group consisting of a CDK-1 inhibitor, a CDK-2 inhibitor, a CDK- 4 inhibitor, a CDK-6 inhibitor, a CAK1 inhibitor from GPC Biotech and Bristol-Myers Squibb, RGB-286199 (GPC Biotech), an anticancer agent from Astex Technology, a CAK1 inhibitor from GPC Biotech, a CDK inhibitor from Sanofi-Aventis, a CDK1/CDK2 inhibitor from Amgen, a CDK2 inhibitor from SUGEN-2 (Pfizer), a hearing loss therapy agent (Sound Pharmaceuticals), PD-0332991 (Pfizer), RGB-286199 (GPC Biotech), Ro-0505124 (Hoffmann- La Roche), a Ser/Thr kinase inhibitor from Lilly (EIi Lilly), CVT-2584 (CAS No. 199986-75-9) (CV Therapeutics), and an analogue or derivative thereof.
19439. The method of item 19413 wherein the anti- fibrotic agent is a cyclooxygenase 1 inhibitor.
19440. The method of item 19413 wherein the anti- fibrotic agent is a DHFR inhibitor.
19441. The method of item 19413 wherein the anti- fibrotic agent is a dual integrin inhibitor.
19442. The method of item 19413 wherein the anti- fibrotic agent is an elastase inhibitor.
19443. The method of item 19413 wherein the anti- fibrotic agent is an elongation factor-1 alpha inhibitor. 19444. The method of item 19413 wherein the anti- fibrotic agent is an endothelial growth factor antagonist.
19445. The method of item 19413 wherein the anti- fibrotic agent is an endothelial growth factor receptor kinase inhibitor selected from the group consisting of sorafenib tosylate (Bayer), AAL-993 (Novartis), ABP-309 (Novartis), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EXEL- 2880 (Exelixis), GW-654652 (GlaxoSmithKline), a KDR inhibitor from LG Life Sciences, CT-6685 and CT-6729 (UCB), KRN-633 and KRN-951 (Kirin Brewery), OSI-930 (OSI Pharmaceuticals), SP-5.2 (Supratek Pharma), SU- 11657 (Pfizer), a Tie-2 antagonist (Hybrigenics), a VEGFR-2 kinase inhibitor (Bristol-Myers Squibb), XL-647 (Exelixis), a KDR inhibitor from Abbott Laboratories, sorafenib tosylate, and an analogue or derivative thereof.
19446. The method of item 19413 wherein the anti- fibrotic agent is an endotoxin antagonist.
19447. The method of item 19413 wherein the anti- fibrotic agent is an epothilone and tubulin binder.
19448. The method of item 19413 wherein the anti- fibrotic agent is an estrogen receptor antagonist.
19449. The method of item 19413 wherein the anti- fibrotic agent is an FGF inhibitor.
19450. The method of item 19413 wherein the anti- fibrotic agent is a farnexyl transferase inhibitor. 19451. The method of item 19413 wherein the anti- fibrotic agent is farnesyltransferase inhibitor selected from the group of A- 197574 (Abbott), a farnesyltransferase inhibitor from Servier, FPTIlI (Strathclyde Institute for Drug R), LB-42908 (LG Life Sciences), Pharmaprojects No. 5063 (Genzyme), Pharmaprojects No. 5597 (Ipsen), Yissum Project No. B-1055 (Yissum), and an analogue or derivative thereof.
19452. The method of item 19413 wherein the anti- fibrotic agent is an FLT-3 kinase inhibitor.
19453. The method of item 19413 wherein the anti- fibrotic agent is an FGF receptor kinase inhibitor.
19454. The method of item 19413 wherein the anti- fibrotic agent is a fibrinogen antagonist selected from the group consisting of AUV-201 (Auvation), MG-13926 (Sanofi-Aventis), plasminogen activator (CAS No. 105913-11-9) (from Sanofi-Aventis or UCB), plasminogen activator-2 (tPA-2) (Sanofi-Aventis), pro-urokinase (CAS No. 82657-92-9) (Sanofi-Aventis), and an analogue or derivative thereof.
19455. The method of item 19413 wherein the anti- fibrotic agent is a heat shock protein 90 antagonist selected from the group consisting of SRN-005 (Sirenade), geldanamycin, NSC-33050 (17- allylaminogeldanamycin; 17-AAG), 17-dimethylaminoethylamino-17- demethoxy-geldanamycin (17-DMAG), rifabutin (rifamycin XIV, 1',4- didehydro-1-deoxy-1,4-dihydro-5'-(2-methylpropyl)-1-oxo-), and an analogue or derivative thereof.
19456. The method of item 19413 wherein the anti- fibrotic agent is a histone deacetylase inhibitor. 19457. The method of item 19413 wherein the anti- fibrotic agent is an HMGCoA reductase inhibitor selected from the group consisting of an atherosclerosis therapeutic from Lipid Sciences, ATI-16000 (ARYx Therapeutics), KS-01-019 (Kos Pharmaceuticals), Pharmaprojects No. 2197 (Sanofi-Aventi), RP 61969 (Sanofi-Aventis), and an analogue or derivative thereof.
19458. The method of item 19413 wherein the anti- fibrotic agent is an ICAM inhibitor.
19459. The method of item 19413 wherein the anti- fibrotic agent is an IL, ICE and IRAK antagonist, wherein the antagonist is a CJ-14877, CP-424174 (Pfizer), NF-61 (Negma-Lerads), and an analogue or derivative thereof.
19460. The method of item 19413 wherein the anti- fibrotic agent is an IL-2 inhibitor.
19461. The method of item 19413 wherein the anti- fibrotic agent is an immunosuppressant selected from the group consisting of teriflunomide (Sanofi Aventis), chlorsulfaquinoxalone (NSC-339004), chlorsulfaquinoxalone sulfate, CS-712 (Sankyo), ismomultin alfa (CAS No. 457913-93-8) (Akzo Nobel), antiallergics from GenPat77, antiinflammatories or AT-005 (Androclus Therapeutics), autoimmune disease therapeutics from EpiVax, BN-007 (Bone), budesonide (CAS No. 51333-22- 3) (MAP Pharmaceuticals), CO-14 (Genzyme), edratide (CAS No. 433922- 67-9) (Teva), EP-314 (Enanta), eprovafen (CAS No. 101335-99-3) (Sanofi- Aventis), HWA-131 (CAS No. 118788-41-3) (Sanofi-Aventis), immunomodulators from MerLion Pharmaceuticals, immunosuppressives from Alchemia, IPL-12 (Inflazyme), MDL-9563 (CAS No. 27086-86-8) (Sanofi-Aventis), Pharmaprojects No. 2330 (Sanofi-Aventis), Pharmaprojects No. 6426 (Abgenix), PXS-25 (Pharmaxis), rosmarinic acid (CAS No. 20283-92-5) (Sanofi-Aventis), RP 42927 or RP 54745 (CAS No. 135330-08-4) (Sanofi-Aventis), SGN-35 (Seattle Genetics), ST-1959 (Sigma-Tau), type I diabetes therapy from SYNX Pharma, UNIL-88 (Debiopharm), VP-025 (Vasogen), VR-694 (Vectura), PRTX-001 (Protalex), and an analogue or derivative thereof.
19462. The method of item 19413 wherein the anti- fibrotic agent is an IMPDH (inosine monophosphate).
19463. The method of item 19413 wherein the anti- fibrotic agent is an integrin antagonist.
19464. The method of item 19413 wherein the anti- fibrotic agent is an interleukin antagonist.
19465. The method of item 19413 wherein the anti- fibrotic agent is an inhibitor of type III receptor tyrosine kinase.
19466. The method of item 19413 wherein the anti- fibrotic agent is an irreversible inhibitor of enzyme methionine aminopeptidase type 2.
19467. The method of item 19413 wherein the anti- fibrotic agent is an isozyme selective delta protein kinase C inhibitor.
19468. The method of item 19413 wherein the anti- fibrotic agent a JAK3 enzyme inhibitor.
19469. The method of item 19413 wherein the anti- fibrotic agent is a JNK inhibitor. 19470. The method of item 19413 wherein the anti- fibrotic agent is a kinase inhibitor.
19471. The method of item 19413 wherein the anti- fibrotic agent is kinesin antagonist.
19472. The method of item 19413 wherein the anti- fibrotic agent is a kinesin antagonist.
19473. The method of item 19413 wherein the anti- fibrotic agent is a leukotriene inhibitor and antagonist selected from the group consisting of ambicromil (CAS No. 58805-38-2) (Sanofi-Aventis), amelubant (CAS No. 346735-24-8) (Boehringer Ingelheim), DW-1141 (Dong Wha), ebselen (Daiichi Pharmaceutical), ibudilast (Kyorin), leucotriene inhibitors from Sanofi-Aventis, iymphotoxin-beta receptor (LT-D) from Biogen Idee, Pharmaprojects No. 1535 and 2728 (CAS No. 119340-33-9) (Sanofi-Aventis), R-112 (Rigel), Rev-5367 (CAS No. 92532-05-3) (Sanofi- Aventis), RG-14893 (CAS No. 141835-49-6) (Sanofi-Aventis), RG-5901-A (CAS No. 101910-24-1), 92532-23-5, RP 66153 (CAS No. 142422-79-5), RP 66364 (CAS No.186916-92-5), RP 69698 (CAS No. 141748-00-7) (Sanofi-Aventis), SC-411930 (Pfizer), SC-41930 (CAS No. 120072-59-5) (Pfizer), SC-50605 (CAS No. 138828-39-4) (Pfizer), SC-51146 (CAS No. 141059-52-1), SC-53228 (CAS No. 153633-01-3) (Pfizer), spaglumic acid (ZY-15106) (CAS No.3106-85-2), 80619-64-3 (Novartis), tipredane (CAS No. 85197-77-9) (Bristol-Myers Squibb), U-75302 (CAS No. 119477-85-9) (Pfizer), and analogue or derivative thereof.
19474. The method of item 19413 wherein the anti- fibrotic agent is an MAP kinase inhibitor. 19475. The method of item 19413 wherein the anti- fibrotic agent is a matrix metalioproteinase inhibitor.
19476. The method of item 19413 wherein the anti- fibrotic agent is an MCP-CCR2 inhibitor.
19477. The method of item 19413 wherein the anti- fibrotic agent is an mTOR inhibitor.
19478. The method of item 19413 wherein the anti- fibrotic agent is an mTOR kinase inhibitor.
19479. The method of item 19413 wherein the anti- fibrotic agent is a microtubule inhibitor selected from the group consisting of antibody-maytansinoid conjugates from Biogen Idee, colchicines (MantiCore Pharmaceuticals), anticancer immunoconjugates from Johnson & Johnson, DIME from Octamer, gni-1f (GNI), huC242-DM4, huMy9-6-DM1 (ImmunoGen), IDN-5404 (Indena), IMO-098, IMOderm (Imotep), mebendazole (Introgen Therapeutics), microtubule poisons from Cambridge Enterprise, paclitaxel such as LOTAX from Aphios (CAS No. 33069-62-4), Genexol-PM from Samyang, Pharmaprojects No. 6383 (Tapestry Pharmaceuticals), RPR-112378 (Sanofi-Aventis), SGN-75 (Seattle Genetics), SPL-7435 (Starpharma), SSR-250411 (Sanofi-Aventis), trastuzumab-DM1 (Genentech), vinorelbine, and an analogue or derivative thereof.
19480. The method of item 19413 wherein the anti- fibrotic agent is an Ml F inhibitor.
19481. The method of item 19413 wherein the anti- fibrotic agent is an MMP inhibitor. 19482. The method of item 19413 wherein the anti- fibrotic agent is a neurokinin (NK) antagonist selected from the group consisting of anthrotainin (CAS No. 148084-40-6) (Sanofi-Aventis), an IBS thereapeutic from ArQuIe, MDL-105212A (CAS No. 167261-60-1) (Ssanofi- Aventis), Pharmaprojects No. 2744, 3258 (CAS No. 139167-47-8) 4006, 4201 , or 5986 (Sanofi-Aventis), RP 67580 (CAS No. 135911-02-3), SR- 144190 (CAS No. 201152-86-5), SSR-240600, SSR-241586 (Sanofi- Aventis), TKA-457 (Novartis), vestipitant mesylate (CAS No. 334476-64-1) (GlaxoSmithKline), Win-64821 (Sanofi-Aventis), PRX-96026 (Predix Pharmaceuticals), and an analogue or derivative thereof.
19483. The method of item 19413 wherein the anti- fibrotic agent is an NF kappa B inhibitor selected from the group consisting of AVE-0545 or AVE-0547 (Sanofi-Aventis), bortezomib (CAS No. 179324- 69-7) (Millennium Pharmaceuticals), dexanabinol (CAS No. 112924-45-5) (Pharmos), dexlipotam (Viatris), Pharmaprojects No. 6283 (INDRA) (OXiGENE), IPL-576092 (CAS No. 137571-30-3) (Inflazyme), NFKB decoy (Corgentech), NFKB decoy oligo (AnGes MG), NFKB's from Ariad, osteoporosis treatments or S5 (F005) from Fulcrum Pharmaceuticals, P61 (Phytopharm), R-flurbiprofen (CAS No. 5104-49-4) (Encore Pharmaceuticals), and an analogue or derivative thereof.
19484. The method of item 19413 wherein the anti- fibrotic agent is a nitric oxide agonist.
19485. The method of item 19413 wherein the anti- fibrotic agent is an ornithine decarboxylase inhibitor.
19486. The method of item 19413 wherein the anti- fibrotic agent is a p38 MAP kinase inhibitor selected from the group consisting of AZD-6703 (AstraZeneca), JX-401 (Jexys Pharmaceuticals), BMS-2 (Bristol-Myers Squibb), a p38 MAP kinase inhibitor from Novartis, a p38-alpha MAP kinase inhibitor from Amphora, Pharmaprojects No. 5704 (Pharmacopeia), RPR-200765A (Sanofi-Aventis), SD-282 (Johnson & Johnson), TAK-715 (Takeda), and an analogue or derivative thereof.
19487. The method of item 19413 wherein the anti- fibrotic agent is a palmitoyl-protein thioesterase inhibitor.
19488. The method of item 19413 wherein the anti- fibrotic agent is a PDGF receptor kinase inhibitor selected from the group consisting of AAL-993, AMN-107, or ABP-309 (Novartis), AMG-706 (Amgen), BAY-57-9352 (Bayer), CDP-860 (UCB), E-7080 (Eisai), imatinib (CAS No. 152459-95-5) (Novartis), OSI-930 (OSI Pharmaceuticals), RPR- 127963E (Sanofi-Aventis), RWJ-540973 (Johnson & Johnson), sorafenib tosylate (Bayer), SU-11657 (Pfizer), tandutinib (CAS No. 387867-13-2) (Millennium Pharmaceuticals), vatalanib (Novartis), ZK-CDK (Schering AG), and an analogue or derivative thereof.
19489. The method of item 19413 wherein the anti- fibrotic agent is (-)-halofenate (Metabolex), AMG-131 (Amgen), antidiabetics from Japan Tobacco, AZD-4619, AZD-8450, AZD-8677 (AstraZeneca), DRF-10945, balaglitazone (Dr Reddy's), CS-00088, CS-00098 (Chipscreen Biosciences), E-3030 (Eisai), etalocib (CAS No. 161172-51-6) (EIi Lilly), GSK-641597 (Ligand), GSK-677954 (GlaxoSmithKline), GW-409544 (Ligand), GW-590735 (GlaxoSmithKline), K-111 (Hoffmann-La Roche), LY- 518674 (EIi Lilly), LY-674 (Ligand), LY-929 (Ligand), MC-3001, MC-3002 (MaxoCore Pharmaceuticals), metformin HCI + pioglitazone (CAS No. 1115- 70-4 and 112529-15-4), ACTOPLUS MET from Andrx), muraglitazar (CAS No. 331741-94-7) (Bristol-Myers Squibb), naveglitazar (Ligand), oleoylethanolamide (Kadmus Pharmaceuticals), ONO-5129, pioglitazone hydrochloride (CAS No. 111025-46-8 and 112529-15-4) (Takeda), PLX-204 (Plexxikon), PPAR agonists from Genfit, PPAR delta agonists from EIi Lilly, PPAR-alpha agonists from CrystalGenomics, PPAR-gamma modulators and PPAR-β modulators from CareX, rosiglitazone maleate (CAS No. 122324- 73-4 or 155141-29-0) (GlaxoSmithKline), rosiglitazone maleate/glimepir (CAS No. 155141-29-0 and 93479-97-1), AVANDARYL, rosiglitazone maleate/metformin extend (CAS No. 155141-29-0 and 657-24-9), AVANDAMET, rosiglitazone maleate+metformin, AVANDAMET (GlaxoSmithKline), tesaglitazar (AstraZeneca), and an analogue or derivative thereof.
19490. The method of item 19413 wherein the anti- fibrotic agent is a phosphatase inhibitor.
19491. The method of item 19413 wherein the anti- fibrotic agent is a phosphodiesterase (PDE) inhibitor selected from the group consisting of avanafil (Tanabe Seiyaku), dasantafil (CAS No. 569351- 91-3) (Schering-Plough), A-906119 (CAS No. 134072-58-5), DL-850 (Sanofi-Aventis), GRC-3015, GRC-3566, GRC-3886 (Glenmark), HWA-153 (CAS No. 56395-66-5) (Sanofi-Aventis), hydroxypumafentrine (Altana), IBFB-130011 , IBFB-14-016, IBFB-140301 , IBFB-150007, IBFB-211913 (IBFB Pharma), L-826141 (Merck & Co), medorinone (CAS No. 88296-61-1) (Sanofi-Aventis), MEM-1917 (Memory Pharmaceuticals), ND-1251 (Neuro3d), PDE inhibitors from ICOS, PDE IV inhibitors from Memory Pharmaceuticals and CrystalGenomics, Pharmaprojects No. 2742 and 6141 (Sanofi-Aventis), QAD-171 (Novartis), RHC-2963 (CAS No. 76993-12-9 and 76993-14-1), RPR-117658, RPR-122818 derivatives, SR-24870 , and RPR- 132294 (Sanofi-Aventis), SK-350 (ln2Gen), stroke therapy agents from deCODE Genetics, TAS-203 (Taiho), tofimilast (CAS No. 185954-27-2) (Pfizer), UK-371800 (Pfizer), WIN-65579 (CAS No. 158020-82-7) (Sanofi- Aventis), IBFB-130020 (IBFB Pharma), OPC-6535 (CAS No. 145739-56-6) (Otsuka), a phosphodiesterase III inhibitor, enoximone, a phosphodiesterase IV inhibitor, fosfosal, Atopik (Barrier Therapeutics), triflusal, a phosphodiesterase V inhibitor, and an analogue or derivative thereof.
19492. The method of item 19413 wherein the anti- fibrotic agent is a PKC inhibitor.
19493. The method of item 19413 wherein the anti- fibrotic agent is a platelet activating factor antagonist.
19494. The method of item 19413 wherein the anti- fibrotic agent is a platelet-derived growth factor receptor kinase inhibitor.
19495. The method of item 19413 wherein the anti- fibrotic agent is a prolyl hydroxylase inhibitor.
19496. The method of item 19413 wherein the anti- fibrotic agent is a polymorphonuclear neutrophil inhibitor.
19497. The method of item 19413 wherein the anti- fibrotic agent is a protein kinase B inhibitor.
19498. The method of item 19413 wherein the anti- fibrotic agent is a protein kinase C stimulant.
19499. The method of item 19413 wherein the anti- fibrotic agent is a purine nucleoside analogue.
19500. The method of item 19413 wherein the anti- fibrotic agent is a purinoreceptor P2X antagonist. 19501. The method of item 19413 wherein the anti- fibrotic agent is a Raf kinase inhibitor.
19502. The method of item 19413 wherein the anti- fibrotic agent is a reversible inhibitor of ErbB1 and ErbB2.
19503. The method of item 19413 wherein the anti- fibrotic agent is a ribonucleoside triphosphate reductase inhibitor.
19504. The method of item 19413 wherein the anti- fibrotic agent is an SDF-1 antagonist.
19505. The method of item 19413 wherein the anti- fibrotic agent is a sheddase inhibitor.
19506. The method of item 19413 wherein the anti- fibrotic agent is an SRC inhibitor.
19507. The method of item 19413 wherein the anti- fibrotic agent is a stromelysin inhibitor.
19508. The method of item 19413 wherein the anti- fibrotic agent is an Syk kinase inhibitor.
19509. The method of item 19413 wherein the anti- fibrotic agent is a telomerase inhibitor.
19510. The method of item 19413 wherein the anti- fibrotic agent is a TGF beta inhibitor selected from the group consisting of pirfenidone (CAS No. 53179-13-8) (MARNAC), tranilast (CAS No. 53902- 12-8) (Kissei), IN-1130 (ln2Gen), mannose-6-phosphate (BTG)1 TGF-β antagonists from Inflazyme (Pharmaprojects No. 6075), TGF-β antagonists from Sydney, non-industrial source), TGF-β I receptor kinase inhibitors from EIi Lilly, TGF-β receptor inhibitors from Johnson & Johnson, and an analogue or derivative thereof.
19511. The method of item 19413 wherein the anti- fibrotic agent is a TNFα antagonist or TACE inhibitor selected from the group consisting of adalimumab (CAS No. 331731-18-1) (Cambridge Antibody Technology), AGIX-4207 (AtheroGenics), AGT-1 (Advanced Biotherapy), an anti-inflammatory from Borean Pharma, Cellzome, or Paradigm Therapeutics, anti-inflammatory vaccine (TNF-alpha kinoid) from Neovacs, humanized anti-TNF antibody or an anti-TNF MAb (CB0006) Celltech (UCB), apratastat (CAS No. 287445-51-0) (Wyeth), BMS-561392 (Bristol-Myers Squibb), BN-006 (Bone), certolizumab pegol (CAS No. 428863-50-7 or CH-138 (UCB), cilomilast (CAS No. 153259-65-5) (GlaxoSmithKline), CR-1 (Nuada Pharmaceuticals), CRx-119 (CombinatoRx), D-5410 (UCB), dacopafant (CAS No. 125372-33-0) (Sanofi- Aventis), dersalazine (CAS No. 188913-57-7/188913-58-8) (Uriach), etanercept (CAS No. 185243-69-0) (Amgen), ethyl pyruvate (Critical (Critical Therapeutics), golimumab (CAS No. 476181-74-5) (Johnson & Johnson), hormono-immunotherapy from Ipsen, CDP571 (e.g., Humicade from UCB), IC-485 (ICOS), infliximab (CAS No. 170277-31-3) (Johnson & Johnson), IP- 751 (Manhattan Pharmaceuticals), ISIS-104838 (CAS No. 250755-32-9) (ISIS Pharmaceuticals), lenalidomide (CAS No. 191732-72-6) (Celgene), lentinan (CAS No. 37339-90-5) (Ajinomoto), MDL-201112 (CAS No. 142130-73-2) (Sanofi-Aventis), medroxyprogesterone (CAS No. 520-85-4) (InKine Pharmaceutical), N-acetylcysteine (CAS No. 616-91-1) (Zambon), NBE-P2 (DIREVO Biotech), nerelimomab (CAS No. 162774-06-3) (Chiron), OM-294DP (OM PHARMA), onercept (CAS No. 199685-57-9) (Yeda), PASSTNF-alpha (Verigen), pentoxifylline or oxypentifylline (Sanofi-Aventis), Pharmaprojects No. 4091, 4241, 4295, or 4488 (Sanofi-Aventis), Pharmaprojects No. 5480 (Amgen), Pharmaprojects No. 6749 (Cengent), pirfenidone (CAS No. 53179-13-8) (MARNAC), RPR-132294 (Sanofi- Aventis), S5 (F002) (Fulcrum Pharmaceuticals), simvastatin (CAS No. 79902-63-9) (Merck & Co), STA-6292 (Synta Pharmaceuticals), tacrolimus (CAS No. 104987-11-3) (Fujisawa LifeCycle Pharma), talactoferrin alfa (CAS No. 308240-58-6) (Agennix), thalidomide (CAS No. 50-35-1) (Celgene), TNF antagonists form ProStrakan, and Synergen, TNF inhibitors (Amgen), TNF-alpha antagonists from Dynavax Technologies and Jerina AG (Germany), TNF-alpha inhibitors from IBFB Pharma and Xencor (Xencor), torbafylline (CAS No. 105102-21-4) (Sanofi-Aventis), UR-1505 (Uriach), VT-346 (Viron Therapeutics), YSIL6 (Y's Therapeutics), YSTH2 (Y's Therapeutics), NPI-1302a-3 (Nereus Pharmaceuticals, a TNF antagonist from Jerina AG (Germany), dersalazine, and an analogue or derivative thereof.
19512. The method of item 19413 wherein the anti- fibrotic agent is a Toll receptor inhibitor.
19513. The method of item 19413 wherein the anti- fibrotic agent is a tubulin antagonist.
19514. The method of item 19413 wherein the anti- fibrotic agent is a tyrosine kinase inhibitor selected from the group consisting of SU-011248, SUTENT from Pfizer Inc. (New York, NY), BMS-354825, PN- 355 (Paracelsian Pharmaceuticals), AGN-199659 (Allergan), AAL-993 or ABP-309 (Novartis), adaphostin (NIH), AEE-788 (Novartis), AG-013736 (OSI Pharmaceuticals), AG-13736 (Pfizer), ALT-110 (Alteris Therapeutics), AMG-706 (Amgen), anticancer MAbs from Xencor, anti-EGFrvlll MAbs from Abgenix, anti-HER2 MAb from Abiogen, AZD-2171 or AZD-9935 (AstraZeneca), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), CEP-5214 (Cephalon), CEP-7055 (Cephalon), cetuximab (ImClone Systems), CHIR-200131 and CHIR-258 (Chiron), CP-547632 (OSI Pharmaceuticals), CP-724714 (Pfizer), CT-301 (Creabilis Therapeutics), D- 69491 (Baxter International), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EGFR/ErbB2 inhibitors from Array BioPharma, erlotinib (CAS No. 183319- 69-9) (OSI Pharmaceuticals), EXEL-2880 (Exelixis), FK-778 (Sanofi- Aventis) , gefitinib (CAS No. 184475-35-2) (AstraZeneca), GW-2286 or GW- 654652 (GlaxoSmithKline), her2/neu antigen from AlphaVax, HER-2/neu inhibitor from Generex, Herzyme (Medipad) (Sirna Therapeutics), HKI-272 (Wyeth), HuMax-EGFr (Genmab), idronoxil (CAS No. 81267-65-4) (Novogen), IGF-1 inhibitors from Ontogen, IMC-11F8 (ImClone Systems), kahalalide F (CAS No. 149204-42-2) (PharmaMar), KDR inhibitor from LG Life Sciences, KDR inhibitors from Abbott Laboratories, KDR kinase inhibitors (UCB), Kdr kinase inhibitors from Merck & Co, KRN-633 and KRN- 951 (Kirin Brewery), KSB-102 (Xenova), lapatinib ditosylate (CAS No. 388082-78-8) (GlaxoSmithKline), matuzumab (Merck KGaA), MDX-214 (Medarex), ME-103 (Pharmexa), MED-A300 (Gerolymatos), MNAC-13 (Lay Line Genomics), nimotuzumab (Center of Molecular Immunology), NSC- 330507 or NSC-707545 (NIH), NV-50 (Novogen), OSI-930 (OSI Pharmaceuticals), panitumumab (Abgenix), pelitinib (CAS No. 287933-82-7) (Wyeth), pertuzumab (CAS No. 380610-27-5) (Genentech), Pharmaprojects No. 3985 (Sanofi-Aventis), prostate cancer therapeutics from Sequenom (SQPC35, SQPC36, SQPC90), removab and remoxab (Trion Pharma), RG- 13022 (CAS No. 136831-48-6), RG-13291 (CAS No. 138989-50-1), or RG- 14620 (CAS No. 136831-49-7) (Sanofi-Aventis), RM-6427 (Romark), RNAi breast cancer therapy from Benitec, RP 53801 (CAS No. 125882-88-4) (Sanofi-Aventis), sorafenib tosylate (Bayer), SU-11657 (Pfizer), Tie-2 antagonists from Semaia (Hybrigenics), Tie-2 inhibitors from Ontogen, trastuzumab (CAS No. 180288-69-1) (Genentech), tyrosine kinase inhibitors from Sanofi-Aventis, U3-1287, U3-1565, U3-1784, U3-1800 (U3 Pharma), vatalanib (Novartis), VEGFR-2 kinase inhibitor from Bristol-Myers Squibb, XL-647 (Exelixis), ZD-6474 (AstraZeneca), ZK-CDK (Schering AG), an EGFR tyrosine kinase inhibitor, EKB-569 (Wyeth), and an analogue or derivative thereof.
19515. The method of item 19413 wherein the anti- fibrotic agent is a VEGF inhibitor.
19516. The method of item 19413 wherein the anti- fibrotic agent is a vitamin D receptor agonist.
19517. The method of item 19413 wherein the anti- fibrotic agent is ZD-6474 (an angiogenesis inhibitor).
19518. The method of item 19413 wherein the anti- fibrotic agent is AP-23573 (an mTOR inhibitor).
19519. The method of item 19413 wherein the anti- fibrotic agent is synthadotin (a tubulin antagonist).
19520. The method of item 19413 wherein the anti- fibrotic agent is S-0885 (a collagenase inhibitor).
19521. The method of item 19413 wherein the anti- fibrotic agent is aplidine (an elongation factor-1 alpha inhibitor).
19522. The method of item 19413 wherein the anti- fibrotic agent is ixabepilone (an epithilone).
19523. The method of item 19413 wherein the anti- fibrotic agent is IDN-5390 (an angiogenesis inhibitor). 19524. The method of item 19413 wherein the anti- fibrotic agent is SB-2723005 (an angiogenesis inhibitor).
19525. The method of item 19413 wherein the anti- fibrotic agent is ABT-518 (an angiogenesis inhibitor).
19526. The method of item 19413 wherein the anti- fibrotic agent is combretastatin (an angiogenesis inhibitor).
19527. The method of item 19413 wherein the anti- fibrotic agent is anecortave acetate (an angiogenesis inhibitor).
19528. The method of item 19413 wherein the anti- fibrotic agent is SB-715992 (a kinesin antagonist).
19529. The method of item 19413 wherein the anti- fibrotic agent is temsirolimus (an mTOR inhibitor).
19530. The method of item 19413 wherein the anti- fibrotic agent is adalimumab (a TNFα antagonist).
19531. The method of item 19413 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with an anti-fibrotic agent, and (b) implanting the medical device into the host.
19532. The method of item 19413 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with an anti-infective agent, and (b) implanting the medical device into the host. 19533. The method of item 19413 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with a polymer; and (b) implanting the medical device into the host.
19534. The method of item 19413 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with a composition comprising an anti-fibrotic agent and a polymer, and (b) implanting the medical device into the host.
19535. The method of item 19413 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with a composition comprising an anti-infective agent and a polymer, and (b) implanting the medical device into the host.
19536. The method of item 19413 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with a composition comprising an anti-fibrotic agent, an anti- infective agent and a polymer, and (b) implanting the medical device into the host.
19537. The method of item 19413 wherein the anti- infective agent is an anthracycline.
19538. The method of item 19413 wherein the anti- infective agent is doxorubicin.
19539. The method of item 19413 wherein the anti- infective agent is is mitoxantrone. 19540. The method of item 19413 wherein the anti- infective agent is a fluoropyrimidine.
19541. The method of item 19413 wherein the anti- infective agent is 5-fluorouracil (5-FU).
19542. The method of item 19413 wherein the anti- infective agent is a folic acid antagonist.
19543. The method of item 19413 wherein the anti- infective agent is methotrexate.
19544. The method of item 19413 wherein the anti- infective agent is a podophylotoxin.
19545. The method of item 19413 wherein the anti- infective agent is etoposide.
19546. The method of item 19413 wherein the anti- infective agent is camptothecin.
19547. The method of item 19413 wherein the anti- infective agent is a hydroxyurea.
19548. The method of item 19413 wherein the anti- infective agent is a platinum complex.
19549. The method of item 19413 wherein the anti- infective agent is cisplatin. 19550. The method of item 19413 wherein the composition comprises an anti-thrombotic agent.
19551. The method of item 19413 wherein the polymer is formed from reactants comprising a naturally occurring polymer.
19552. The method of item 19413 wherein the polymer is formed from reactants comprising protein.
19553. The method of item 19413 wherein the polymer is formed from reactants comprising carbohydrate.
19554. The method of item 19413 wherein the polymer is formed from reactants comprising biodegradable polymer.
19555. The method of item 19413 wherein the polymer is formed from reactants comprising nonbiodegradable polymer.
19556. The method of item 19413 wherein the polymer is formed from reactants comprising collagen.
19557. The method of item 19413 wherein the polymer is formed from reactants comprising methylated collagen.
19558. The method of item 19413 wherein the polymer is formed from reactants comprising fibrinogen.
19559. The method of item 19413 wherein the polymer is formed from reactants comprising thrombin. 19560. The method of item 19413 wherein the polymer reactants comprising blood plasma.
19561. The method of item 19413 wherein the polymer reactants comprising calcium salt.
19562. The method of item 19413 wherein the polymer reactants comprising an antifibronolytic agent.
19563. The method of item 19413 wherein the polymer reactants comprising fibrinogen analog.
19564. The method of item 19413 wherein the polymer reactants comprising albumin.
19565. The method of item 19413 wherein the polymer reactants comprising plasminogen.
19566. The method of item 19413 wherein the polymer reactants comprising von Willebrands factor.
19567. The method of item 19413 wherein the polymer reactants comprising Factor VIII.
19568. The method of item 19413 wherein the polymer reactants comprising hypoallergenic collagen.
19569. The method of item 19413 wherein the polymer reactants comprising atelopeptidic collagen. 19570. The method of item 19413 wherein the polymer is formed from reactants comprising telopeptide collagen.
19571. The method of item 19413 wherein the polymer is formed from reactants comprising crosslinked collagen.
19572. The method of item 19413 wherein the polymer is formed from reactants comprising aprotinin.
19573. The method of item 19413 wherein the polymer is formed from reactants comprising epsilon-amino-n-caproic acid.
19574. The method of item 19413 wherein the polymer is formed from reactants comprising gelatin.
19575. The method of item 19413 wherein the polymer is formed from reactants comprising protein conjugates.
19576. The method of item 19413 wherein the polymer is formed from reactants comprising gelatin conjugates.
19577. The method of item 19413 wherein the polymer is formed from reactants comprising a synthetic polymer.
19578. The method of item 19413 wherein the polymer is formed from reactants comprising a synthetic isocyanate-containing compound.
19579. The method of item 19413 wherein the polymer is formed from reactants comprising a synthetic thiol-containing compound. 19580. The method of item 19413 wherein the polymer is formed from reactants comprising a synthetic compound containing at least two thiol groups.
19581. The method of item 19413 wherein the polymer is formed from reactants comprising a synthetic compound containing at least three thiol groups.
19582. The method of item 19413 wherein the polymer is formed from reactants comprising a synthetic compound containing at least four thiol groups.
19583. The method of item 19413 wherein the polymer is formed from reactants comprising a synthetic amino-containing compound.
19584. The method of item 19413 wherein the polymer is formed from reactants comprising a synthetic compound containing at least two amino groups.
19585. The method of item 19413 wherein the polymer is formed from reactants comprising a synthetic compound containing at least three amino groups.
19586. The method of item 19413 wherein the polymer is formed from reactants comprising a synthetic compound containing at least four amino groups.
19587. The method of item 19413 wherein the polymer is formed from reactants comprising a synthetic compound comprising a carbonyl-oxygen-succinimidyl group. 19588. The method of item 19413 wherein the polymer is formed from reactants comprising a synthetic compound comprising at least two carbonyl-oxygen-succinimidyl groups.
19589. The method of item 19413 wherein the polymer is formed from reactants comprising a synthetic compound comprising at least three carbonyl-oxygen-succinimidyl groups.
19590. The method of item 19413 wherein the polymer is formed from reactants comprising a synthetic compound comprising at least four carbonyl-oxygen-succinimidyl groups.
19591. The method of item 19413 wherein the polymer is formed from reactants comprising a synthetic polyalkylene oxide- containing compound.
19592. The method of item 19413 wherein the polymer is formed from reactants comprising a synthetic compound comprising both polyalkylene oxide and biodegradable polyester blocks.
19593. The method of item 19413 wherein the polymer is formed from reactants comprising a synthetic polyalkylene oxide- containing compound having reactive amino groups.
19594. The method of item 19413 wherein the polymer is formed from reactants comprising a synthetic polyalkylene oxide- containing compound having reactive thiol groups.
19595. The method of item 19413 wherein the polymer is formed from reactants comprising a synthetic polyalkylene oxide- containing compound having reactive carbonyl-oxygen-succinimidyl groups. 19596. The method of item 19413 wherein the polymer is formed from reactants comprising a synthetic compound comprising a biodegradable polyester block.
19597. The method of item 19413 wherein the polymer is formed from reactants comprising a synthetic polymer formed in whole or part from lactic acid or lactide.
19598. The method of item 19413 wherein the polymer is formed from reactants comprising a synthetic polymer formed in whole or part from glycolic acid or glycolide.
19599. The method of item 19413 wherein the polymer is formed from reactants comprising polylysine.
19600. The method of item 19413 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound comprising a polyalkylene oxide portion.
19601. The method of item 19413 wherein the polymer is formed from reactants comprising (a) protein and (b) polylysine.
19602. The method of item 19413 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound having at least four thiol groups.
19603. The method of item 19413 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound having at least four amino groups. 19604. The method of item 19413 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound having at least four carbonyl-oxygen-succinimide groups.
19605. The method of item 19413 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound having a biodegradable region formed from reactants selected from lactic acid, lactide, glycolic acid, glycolide, and epison-caprolactone.
19606. The method of item 19413 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound comprising a polyalkylene oxide portion.
19607. The method of item 19413 wherein the polymer is formed from reactants comprising (a) collagen and (b) polylysine.
19608. The method of item 19413 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound having at least four thiol groups.
19609. The method of item 19413 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound having at least four amino groups.
19610. The method of item 19413 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound having at least four carbonyl-oxygen-succinimide groups.
19611. The method of item 19413 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound having a biodegradable region formed from reactants selected from lactic acid, lactide, glycolic acid, glycolide, and epison-caprolactone.
19612. The method of item 19413 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound comprising a polyalkylene oxide portion.
19613. The method of item 19413 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) polylysine.
19614. The method of item 19413 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound having at least four thiol groups.
19615. The method of item 19413 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound having at least four amino groups.
19616. The method of item 19413 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound having at least four carbonyl-oxygen-succinimide groups.
19617. The method of item 19413 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound having a biodegradable region formed from reactants selected from lactic acid, lactide, glycolic acid, glycolide, and epison-caprolactone.
19618. The method of item 19413 wherein the polymer is formed from reactants comprising hyaluronic acid. 19619. The method of item 19413 wherein the polymer is formed from reactants comprising a hyaluronic acid derivative.
19620. The method of item 19413 wherein the polymer is formed from reactants comprising pentaerythritol poly(ethylene glycol)ether tetra-sulfhydryl of number average molecular weight between 3,000 and 30,000.
19621. The method of item 19413 wherein the polymer is formed from reactants comprising pentaerythritol poly(ethylene glycol)ether tetra-amino of number average molecular weight between 3,000 and 30,000.
19622. The method of item 19413 wherein the polymer is formed from reactants comprising (a) a synthetic compound having a number average molecular weight between 3,000 and 30,000 and comprising a polyalkylene oxide region and multiple nucleophilic groups, and (b) a synthetic compound having a number average molecular weight between 3,000 and 30,000 and comprising a polyalkylene oxide region and multiple electrophilic groups.
19623. The method of item 19413 wherein the composition comprises a colorant.
19624. The method of item 19413 wherein the composition is sterile.
19625. A method as in any one of items 19413-19624, wherein the device is an extravascular graft. 19626. A method as in any one of items 19413-19624, wherein the device is an intravascular graft.
19627. A method as in any one of items 19413-19624, wherein the device is a vascular graft adapted for replacing a blood vessel damaged by aneurysm.
19628. A method as in any one of items 19413-19624, wherein the device is a vascular graft adapted for replacing a blood vessel damaged by intimal hyperplasia.
19629. A method as in any one of items 19413-19624, wherein the device is a vascular graft adapted for replacing a blood vessel damaged by thrombosis.
19630. A method as in any one of items 19413-19624, . _ . - wherein the device is a vascular graft adapted for providing access to blood vessel.
19631. A method as in any one of items 19413-19624, wherein the device is a vascular graft adapted for providing an alternative conduit for blood flow through a damaged or diseased area in a vein.
19632. A method as in any one of items 19413-19624, wherein the device is a vascular graft adapted for providing an alternative conduit for blood flow through a damaged or diseased area in an artery.
19633. A method as in any one of items 19413-19624, wherein the device is a synthetic bypass graft. 19634. A method as in any one of items 19413-19624, wherein the device is a femoral-popliteal bypass graft.
19635. A method as in any one of items 19413-19624, wherein the device is a femoral-femoral bypass graft.
19636. A method as in any one of items 19413-19624, wherein the device is an axillary-femoral bypass graft.
19637. A method as in any one of items 19413-19624, wherein the device is a vein graft.
19638. A method as in any one of items 19413-19624, wherein the device is a peripheral vein graft.
19639. A method as in any one of items 19413-19624, wherein the device is a coronary vein graft.
19640. A method as in any one of items 19413-19624, wherein the device is an internal mammary graft.
19641. A method as in any one of items 19413-19624, wherein the device is a bifurcated vascular graft.
19642. A method as in any one of items 19413-19624, wherein the device is an intraluminal graft.
19643. A method as in any one of items 19413-19624, wherein the device is a prosthetic vascular graft. 19644. A method for implanting a medical device comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with i) an anti-fibrotic agent, H) an anti-infective agent, iii) a polymer, iv) a composition comprising an anti-fibrotic agent and a polymer v) a composition comprising an anti-infective agent and a polymer, or vi) a composition comprising an anti-fibrotic agent, an anti- infective agent and a polymer, and (b) implanting the medical device into the host, wherein the medical device is a hemodialysis access device.
19645. The method of item 19644 wherein the anti- fibrotic agent is an adensosine A2A receptor antagonist.
19646. The method of item 19644 wherein the anti- fibrotic agent is an AKT inhibitor.
19647. The method of item 19644 wherein the anti- fibrotic agent is an alpha 2 integrin antagonist, wherein the alpha 2 integrin antagonist is Pharmaprojects No. 5754 (Merck KgaA).
19648. The method of item 19644 wherein the anti- fibrotic agent is an alpha 4 integrin antagonist.
19649. The method of item 19644 wherein the anti- fibrotic agent is an alpha 7 nicotinic receptor agonist.
19650. The method of item 19644 wherein the anti- fibrotic agent is an angiogenesis inhibitor selected from the group consisting of AG-12,958 (Pfizer), ATN-161 (Attenuon LLC), neovastat, an angiogenesis inhibitor from Jerina AG (Germany), NM-3 (Mercian), VGA-1155 (Taisho), FCE-26644 (Pfizer), FCE-26950 (Pfizer), FPMA (Meiji Daries), FR-111142 (Fujisawa), GGTI-298, GM-1306 (Ligand), GPA-1734 (Novartis), NNC-47- 0011 (Novo Nordisk), herbamycin (Nippon Kayaku), lenalidomide (Celegene), IP-10 (NIH), ABT-828 (Abbott), KIN-841 (Tokushima University, Japan), SF-1126 (Semafore Pharmaceuticals), laminin technology (NIH), CHIR-258 (Chiron), NVP-AEW541 (Novartis), NVP-AEW541 (Novartis), Vt16907 (Alchemia), OXI-8007 (Oxigene), EG-3306 (Ark Therapeutics), Maspin (Arriva), ABT-567 (Abbott), PPI-2458 (Praecis Pharmaceuticals), CC-5079, CC-4089 (Celgene), HIF-1 alpha inhibitors (Xenova), S-247 (Pfizer), AP-23573 (Ariad), AZD-9935 (Astra Zeneca), mebendazole (Introgen Therapeutics), MetAP-2 inhibitors (GlaxoSmithKline), AG-615 (Angiogene Pharmaceuticals), Tie-2 antagonists (Hybrigenics), NC-381 , CYC-381, NC-169, NC-219, NC-383, NC-384, NC-407 (Lorus Therapeutics), ATN-224 (Attenuon), ON-01370 (Onconova), Vitronectin antagonists (Amgen), SDX-103 (Salmedix), Vitronectin antagonists (Shire), CHP (Riemser), TEK (Amgen), Anecortave acetate (Alcon), T46.2 (Matrix Therapeutics), HG-2 (Heptagen), TEM antagonists (Genzyme), Oxi-4500 (Oxigene), ATN-161 (Attenuon), WX-293 (Wilex), M-2025 (Metris Therapeutics), Alphastatin (BioActa), YH-16 (Yantai Rongchang), BIBF- 1120 (Boehringer Ingelheim), BAY-57-9352 (Bayer), AS-1404 (Cancer Research Technology), SC-77964 (Pfizer), glycomimetics (BioTie Therapies), TIE-2 Inhibitors (Ontogen), DIMI, Octamer (Octamer), ABR- 215050 (Active Biotech), ABT-518 (Abbott), KDR inhibitors (Abbott), BSF- 466895 (Abbott), SCH-221153 (Schering-Plough), DAC:antiangiogenic (ConjuChem), TFPI (EntreMed), AZD-2171 (Astra-Zenaca), CDC-394 (Celgene), LY290293 (EIi Lilly), IDN-5390 (Indena), Kdr Kinase Inhibitors (Merck), CT-113020, CT-116433, CT-116563, CT-31890, CT-32228) (Cell Therapeutics), A-299620 (Abbott), TWEAK Inhibitor (Amgen), VEGF modulators (Johnson and Johnson), Tum-N53, tumstatin (Genzyme), Thios- 1 , Thios-2 (Thios Pharmaceuticals), MV-6401 (Miravant Medical Technologies), Spisulosine (PharmaMar), CEP-7055 (Cephalon), AUV-201 (Auvation), LM-609 (EIi Lilly), SKF-106615 (AnorMED), Oglufanide disodium (Cytran), BW-114 (Pharminox), Calreticulin (NIH), WX-678 (Wilex), SD-7784 (Pfizer), WX-UK1 (Wilex), SH-268 (Schering AG), 2-Me-PGA (Celgene), S- 137 (Pfizer), ZD-6126 (Angiogene Pharmaceuticals), SG-292 (SignalGen), Benefin (Lane Labs), A6, A36 (Angstrom), SB-2723005 (GlaxoSmithKline), SC-7 (Cell Therapeutics), ZEN-014 (AEterna Zentaris), 2-methoxyestradiol (EntreMed), NK-130119 (Nippon Kayaku), CC-10004 (Celgene), AVE- 8062A (Ajinomoto), Tacedinaline (Pfizer), Actinonin (Tokyo Metropolitan Institute of Medical Science), Lenalidomide (Celgene), VGA-1155, BTO-956 (SRI International), ER-68203-00 (Eisai), CT-6685 (UCB), JKC-362 (Phoenix Pharmaceuticals), DMI-3798 (DMI Biosciences, Angiomate (Ipsen), ZD-6474 (AstraZeneca), CEP-5214 (Cephalon), Canstatin (Genzyme), NM-3 (Mercian), Oridigm (MediQuest Therapeutics), Exherin (Adherex), BLS-0597 (Boston Life Sciences), PTC-299 (PTC Therapeutics), NPI-2358 (Nereus Pharmaceuticals), CGP-79787 (Novartis), AEE-788 (Novartis), CKD-732 (Chong Kun Dang), CP-564959 (OSI Pharmaceuticals), CM-101 (CarboMed), CT-2584, CT3501 (Cell Therapeutics), combretastatin and analogues and derivatives thereof (Oxigene), Rebimastat (Bristol-Meyers Squibb), Dextrin 2-sulfate (ML Laboratories), Cilengitide (Merk KGaA), NSC-706704 (Pharminox), KRN- 951 (Kirin Brewery), Ukrain, NSC-631570 (Nowicky Pharma), Tecogalan sodium (Daiichi Pharmaceutical), Tz-93 (Tsumura), TBC-1635 (Encysive Pharmaceuticals), TAN-1120 (Takeda), Semaxanib (Pfizer), BDI-7800 (Biopharmacopae), SD-186, SD-983 (Bristol-Meyers Squibb), SB-223245 (GlaxoSmithKline), SC-236 (Pfizer), RWJ-590973 (Johnson and Johnson), ILX-1850 (Genzyme), SC-68488, S-836 (Pfizer), CG-55069-11 (CuraGen), Ki-23057 (Kirin Brewery), CCX-700 (Chemoentryx), Pegaptanib octasodium (Giled Sciences), ANGIOCOL (available from Biostratum Inc.), or an analogue or derivative thereof.
19651. The method of item 19644 wherein the anti- fibrotic agent is an apoptosis antagonist. 19652. The method of item 19644 wherein the anti- fibrotic agent is an apoptosis activator.
19653. The method of item 19644 wherein the anti- fibrotic agent is a beta 1 integrin antagonist.
19654. The method of item 19644 wherein the anti- fibrotic agent is a beta tubulin inhibitor.
19655. The method of item 19644 wherein the anti- fibrotic agent is a blocker of enzyme production in Hepatitis C.
19656. The method of item 19644 wherein the anti- fibrotic agent is a Bruton's tyrosine kinase inhibitor.
19657. The method of item 19644 wherein the anti- fibrotic agent is a calcineurin inhibitor.
19658. The method of item 19644 wherein the anti- fibrotic agent is a caspase 3 inhibitor.
19659. The method of item 19644 wherein the anti- fibrotic agent is a CC chemokine receptor antagonist.
19660. The method of item 19644 wherein the anti- fibrotic agent is a cell cycle inhibitor selected from the group consisting of SNS-595 (Sunesis), synthadotin, KRX-0403, and an analogue or derivative thereof.
19661. The method of item 19644 wherein the anti- fibrotic agent is a cathepsin B inhibitor. 19662. The method of item 19644 wherein the anti- fibrotic agent is a cathepsin K inhibitor, wherein the cathepsin K inhibitor is 462795 (GlaxoSmithKline), INPL-022-D6 (Amura Therapeutics), or an analogue or derivative thereof.
19663. The method of item 19644 wherein the anti- fibrotic agent is a cathepsin L inhibitor.
19664. The method of item 19644 wherein the anti- fibrotic agent is a CD40 antagonist.
19665. The method of item 19644 wherein the anti- fibrotic agent is a chemokine receptor agonist.
19666. The method of item 19644 wherein the anti- fibrotic agent is a chymase inhibitor.
19667. The method of item 19644 wherein the anti- fibrotic agent is a collagenase antagonist.
19668. The method of item 19644 wherein the anti- fibrotic agent is a CXCR antagonist.
19669. The method of item 19644 wherein the anti- fibrotic agent is a cyclin dependent kinase inhibitor selected from the group consisting of a CDK-1 inhibitor, a CDK-2 inhibitor, a CDK- 4 inhibitor, a CDK-6 inhibitor, a CAK1 inhibitor from GPC Biotech and Bristol-Myers Squibb, RGB-286199 (GPC Biotech), an anticancer agent from Astex Technology, a CAK1 inhibitor from GPC Biotech, a CDK inhibitor from Sanofi-Aventis, a CDK1/CDK2 inhibitor from Amgen, a CDK2 inhibitor from SUGEN-2 (Pfizer), a hearing loss therapy agent (Sound Pharmaceuticals), PD-0332991 (Pfizer), RGB-286199 (GPC Biotech), Ro-0505124 (Hoffmann- La Roche), a Ser/Thr kinase inhibitor from Lilly (EIi Lilly), CVT-2584 (CAS No. 199986-75-9) (CV Therapeutics), and an analogue or derivative thereof.
19670. The method of item 19644 wherein the anti- fibrotic agent is a cyclooxygenase 1 inhibitor.
19671. The method of item 19644 wherein the anti- fibrotic agent is a DHFR inhibitor.
19672. The method of item 19644 wherein the anti- fibrotic agent is a dual integrin inhibitor.
19673. The method of item 19644 wherein the anti- fibrotic agent is an elastase inhibitor.
19674. The method of item 19644 wherein the anti- fibrotic agent is an elongation factor-1 alpha inhibitor.
19675. The method of item 19644 wherein the anti- fibrotic agent is an endothelial growth factor antagonist.
19676. The method of item 19644 wherein the anti- fibrotic agent is an endothelial growth factor receptor kinase inhibitor selected from the group consisting of sorafenib tosylate (Bayer), AAL-993 (Novartis), ABP-309 (Novartis), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EXEL- 2880 (Exelixis), GW-654652 (GlaxoSmithKline), a KDR inhibitor from LG Life Sciences, CT-6685 and CT-6729 (UCB), KRN-633 and KRN-951 (Kirin Brewery), OSI-930 (OSI Pharmaceuticals), SP-5.2 (Supratek Pharma), SU-
11657 (Pfizer), a Tie-2 antagonist (Hybrigenics), a VEGFR-2 kinase inhibitor (Bristol-Myers Squibb), XL-647 (Exelixis), a KDR inhibitor from Abbott Laboratories, sorafenib tosylate, and an analogue or derivative thereof.
19677. The method of item 19644 wherein the anti- fibrotic agent is an endotoxin antagonist.
19678. The method of item 19644 wherein the anti- fibrotic agent is an epothilone and tubulin binder.
19679. The method of item 19644 wherein the anti- fibrotic agent is an estrogen receptor antagonist.
19680. The method of item 19644 wherein the anti- fibrotic agent is an FGF inhibitor.
19681. The method of item 19644 wherein the anti- fibrotic agent is a famexyl transferase inhibitor.
19682. The method of item 19644 wherein the anti- fibrotic agent is farnesyltransferase inhibitor selected from the group of A- 197574 (Abbott), a farnesyltransferase inhibitor from Servier, FPTIII (Strathclyde Institute for Drug R), LB-42908 (LG Life Sciences), Pharmaprojects No. 5063 (Genzyme), Pharmaprojects No. 5597 (Ipsen), Yissum Project No. B-1055 (Yissum), and an analogue or derivative thereof.
19683. The method of item 19644 wherein the anti- fibrotic agent is an FLT-3 kinase inhibitor.
19684. The method of item 19644 wherein the anti- fibrotic agent is an FGF receptor kinase inhibitor. 19685. The method of item 19644 wherein the anti- fibrotic agent is a fibrinogen antagonist selected from the group consisting of AUV-201 (Auvation), MG-13926 (Sanofi-Aventis), plasminogen activator (CAS No. 105913-11-9) (from Sanofi-Aventis or UCB), plasminogen activator-2 (tPA-2) (Sanofi-Aventis), pro-urokinase (CAS No. 82657-92-9) (Sanofi-Aventis), and an analogue or derivative thereof.
19686. The method of item 19644 wherein the anti- fibrotic agent is a heat shock protein 90 antagonist selected from the group consisting of SRN-005 (Sirenade), geldanamycin, NSC-33050 (17- allylaminogeldanamycin; 17-AAG), 17-dimethylaminoethylamino-17- demethoxy-geldanamycin (17-DMAG), rifabutin (rifamycin XIV, 1',4- didehydro-1-deoxy-1 ,4-dihydro-5'-(2-methylpropyl)-1-oxo-), and an analogue or derivative thereof.
19687. The method of item 19644 wherein the anti- fibrotic agent is a histone deacetylase inhibitor.
19688. The method of item 19644 wherein the anti- fibrotic agent is an HMGCoA reductase inhibitor selected from the group consisting of an atherosclerosis therapeutic from Lipid Sciences, ATI-16000 (ARYx Therapeutics), KS-01-019 (Kos Pharmaceuticals), Pharmaprojects No. 2197 (Sanofi-Aventi), RP 61969 (Sanofi-Aventis), and an analogue or derivative thereof.
19689. The method of item 19644 wherein the anti- fibrotic agent is an ICAM inhibitor.
19690. The method of item 19644 wherein the anti- fibrotic agent is an IL, ICE and IRAK antagonist, wherein the antagonist is a CJ-14877, CP-424174 (Pfizer), NF-61 (Negma-Lerads), and an analogue or derivative thereof.
19691. The method of item 19644 wherein the anti- fibrotic agent is an IL-2 inhibitor.
19692. The method of item 19644 wherein the anti- fibrotic agent is an immunosuppressant selected from the group consisting of teriflunomide (Sanofi Aventis), chlorsulfaquinoxalone (NSC-339004), chlorsulfaquinoxalone sulfate, CS-712 (Sankyo), ismomultin alfa (CAS No. 457913-93-8) (Akzo Nobel), antiallergics from GenPat77, antiinflammatories or AT-005 (Androclus Therapeutics), autoimmune disease therapeutics from EpiVax, BN-007 (Bone), budesonide (CAS No. 51333-22- 3) (MAP Pharmaceuticals), CO-14 (Genzyme), edratide (CAS No. 433922- 67-9) (Teva), EP-314 (Enanta), eprovafen (CAS No. 101335-99-3) (Sanofi- Aventis), HWA-131 (CAS No. 118788-41-3) (Sanofi-Aventis), immunomodulators from MerLion Pharmaceuticals, immunosuppressives from Alchemia, IPL-12 (Inflazyme), MDL-9563 (CAS No. 27086-86-8) (Sanofi-Aventis), Pharmaprojects No. 2330 (Sanofi-Aventis), Pharmaprojects No. 6426 (Abgenix), PXS-25 (Pharmaxis), rosmarinic acid (CAS No. 20283-92-5) (Sanofi-Aventis), RP 42927 or RP 54745 (CAS No. 135330-08-4) (Sanofi-Aventis), SGN-35 (Seattle Genetics), ST-1959 (Sigma-Tau), type I diabetes therapy from SYNX Pharma, UNIL-88 (Debiopharm), VP-025 (Vasogen), VR-694 (Vectura), PRTX-001 (Protalex), and an analogue or derivative thereof.
19693. The method of item 19644 wherein the anti- fibrotic agent is an IMPDH (inosine monophosphate).
19694. The method of item 19644 wherein the anti- fibrotic agent is an integrin antagonist. 19695. The method of item 19644 wherein the anti- fibrotic agent is an interleukin antagonist.
19696. The method of item 19644 wherein the anti- fibrotic agent is an inhibitor of type III receptor tyrosine kinase.
19697. The method of item.19644 wherein the anti- fibrotic agent is an irreversible inhibitor of enzyme methionine aminopeptidase type 2.
19698. The method of item 19644 wherein the anti- fibrotic agent is an isozyme selective delta protein kinase C inhibitor.
19699. The method of item 19644 wherein the anti- fibrotic agent a JAK3 enzyme inhibitor.
19700. The method of item 19644 wherein the anti- fibrotic agent is a JNK inhibitor.
19701. The method of item 19644 wherein the anti- fibrotic agent is a kinase inhibitor.
19702. The method of item 19644 wherein the anti- fibrotic agent is kinesin antagonist.
19703. The method of item 19644 wherein the anti- fibrotic agent is a kinesin antagonist.
19704. The method of item 19644 wherein the anti- fibrotic agent is a leukotriene inhibitor and antagonist selected from the group consisting of ambicromil (CAS No. 58805-38-2) (Sanofi-Aventis), amelubant (CAS No. 346735-24-8) (Boehringer Ingelheim), DW-1141 (Dong Wha), ebselen (Daiichi Pharmaceutical), ibudilast (Kyorin), leucotriene inhibitors from Sanofi-Aventis, lymphotoxin-beta receptor (LT-D) from Biogen Idee, Pharmaprojects No. 1535 and 2728 (CAS No. 119340-33-9) (Sanofi-Aventis), R-112 (Rigel), Rev-5367 (CAS No. 92532-05-3) (Sanofi- Aventis), RG-14893 (CAS No. 141835-49-6) (Sanofi-Aventis), RG-5901-A (CAS No. 101910-24-1), 92532-23-5, RP 66153 (CAS No. 142422-79-5), RP 66364 (CAS No.186916-92-5), RP 69698 (CAS No. 141748-00-7) (Sanofi-Aventis), SC-411930 (Pfizer), SC-41930 (CAS No. 120072-59-5) (Pfizer), SC-50605 (CAS No. 138828-39-4) (Pfizer), SC-51146 (CAS No. 141059-52-1), SC-53228 (CAS No. 153633-01-3) (Pfizer), spaglumic acid (ZY-15106) (CAS No.3106-85-2), 80619-64-3 (Novartis), tipredane (CAS No. 85197-77-9) (Bristol-Myers Squibb), U-75302 (CAS No. 119477-85-9) (Pfizer), and analogue or derivative thereof.
19705. The method of item 19644 wherein the anti- fibrotic agent is an MAP kinase inhibitor.
19706. The method of item 19644 wherein the anti- fibrotic agent is a matrix metalloproteinase inhibitor.
19707. The method of item 19644 wherein the anti- fibrotic agent is an MCP-CCR2 inhibitor.
19708. The method of item 19644 wherein the anti- fibrotic agent is an mTOR inhibitor.
19709. The method of item 19644 wherein the anti- fibrotic agent is an mTOR kinase inhibitor. 19710. The method of item 19644 wherein the anti- fibrotic agent is a microtubule inhibitor selected from the group consisting of antibody-maytansinoid conjugates from Biogen Idee, colchicines (MantiCore Pharmaceuticals), anticancer immunoconjugates from Johnson & Johnson, DIME from Octamer, gni-1f (GNI), huC242-DM4, huMy9-6-DM1 (ImmunoGen), IDN-5404 (Indena), IMO-098, IMOderm (Imotep), mebendazole (Introgen Therapeutics), microtubule poisons from Cambridge Enterprise, paclitaxel such as LOTAX from Aphios (CAS No. 33069-62-4), Genexol-PM from Samyang, Pharmaprojects No. 6383 (Tapestry Pharmaceuticals), RPR-112378 (Sanofi-Aventis), SGN-75 (Seattle Genetics), SPL-7435 (Starpharma), SSR-250411 (Sanofi-Aventis), trastuzumab-DM1 (Genentech), vinorelbine, and an analogue or derivative thereof.
19711. The method of item 19644 wherein the anti- fibrotic agent is an MIF inhibitor.
19712. The method of item 19644 wherein the anti- fibrotic agent is an MMP inhibitor.
19713. The method of item 19644 wherein the anti- fibrotic agent is a neurokinin (NK) antagonist selected from the group consisting of anthrotainin (CAS No. 148084-40-6) (Sanofi-Aventis), an IBS thereapeutic from ArQuIe, MDL-105212A (CAS No. 167261-60-1) (Ssanofi- Aventis), Pharmaprojects No. 2744, 3258 (CAS No. 139167-47-8) 4006, 4201 , or 5986 (Sanofi-Aventis), RP 67580 (CAS No. 135911-02-3), SR- 144190 (CAS No. 201152-86-5), SSR-240600, SSR-241586 (Sanofi- Aventis), TKA-457 (Novartis), vestipitant mesylate (CAS No. 334476-64-1) (GlaxoSmithKline), Win-64821 (Sanofi-Aventis), PRX-96026 (Predix Pharmaceuticals), and an analogue or derivative thereof. 19714. The method of item 19644 wherein the anti- fibrotic agent is an NF kappa B inhibitor selected from the group consisting of AVE-0545 or AVE-0547 (Sanofi-Aventis), bortezomib (CAS No. 179324- 69-7) (Millennium Pharmaceuticals), dexanabinol (CAS No. 112924-45-5) (Pharmos), dexlipotam (Viatris), Pharmaprojects No. 6283 (INDRA) (OXiGENE), IPL-576092 (CAS No. 137571-30-3) (Inflazyme), NFKB decoy (Corgentech), NFKB decoy oligo (AnGes MG), NFKB's from Ariad, osteoporosis treatments or S5 (F005) from Fulcrum Pharmaceuticals, P61 (Phytopharm), R-flurbiprofen (CAS No. 5104-49-4) (Encore Pharmaceuticals), and an analogue or derivative thereof.
19715. The method of item 19644 wherein the anti- fibrotic agent is a nitric oxide agonist.
19716. The method of item 19644 wherein the anti- fibrotic agent is an ornithine decarboxylase inhibitor.
19717. The method of item 19644 wherein the anti- fibrotic agent is a p38 MAP kinase inhibitor selected from the group consisting of AZD-6703 (AstraZeneca), JX-401 (Jexys Pharmaceuticals), BMS-2 (Bristol-Myers Squibb), a p38 MAP kinase inhibitor from Novartis, a p38-alpha MAP kinase inhibitor from Amphora, Pharmaprojects No. 5704 (Pharmacopeia), RPR-200765A (Sanofi-Aventis), SD-282 (Johnson & Johnson), TAK-715 (Takeda), and an analogue or derivative thereof.
19718. The method of item 19644 wherein the anti- fibrotic agent is a palmitoyl-protein thioesterase inhibitor.
19719. The method of item 19644 wherein the anti- fibrotic agent is a PDGF receptor kinase inhibitor selected from the group consisting of AAL-993, AMN-107, or ABP-309 (Novartis), AMG-706 (Amgen), BAY-57-9352 (Bayer), CDP-860 (UCB), E-7080 (Eisai), imatinib (CAS No. 152459-95-5) (Novartis), OSI-930 (OSI Pharmaceuticals), RPR- 127963E (Sanofi-Aventis), RWJ-540973 (Johnson & Johnson), sorafenib tosylate (Bayer), SU-11657 (Pfizer), tandutinib (CAS No. 387867-13-2) (Millennium Pharmaceuticals), vatalanib (Novartis), ZK-CDK (Schering AG), and an analogue or derivative thereof.
19720. The method of item 19644 wherein the anti- fibrotic agent is (-)-halofenate (Metabolex), AMG-131 (Amgen), antidiabetics from Japan Tobacco, AZD-4619, AZD-8450, AZD-8677 (AstraZeneca), DRF-10945, balaglitazone (Dr Reddy's), CS-00088, CS-00098 (Chipscreen Biosciences), E-3030 (Eisai), etalocib (CAS No. 161172-51-6) (EIi Lilly), GSK-641597 (Ligand), GSK-677954 (GlaxoSmithKline), GW-409544 (Ligand), GW-590735 (GlaxoSmithKline), K-111 (Hoffmann-La Roche), LY- 518674 (EIi Lilly), LY-674 (Ligand), LY-929 (Ligand), MC-3001, MC-3002 (MaxoCore Pharmaceuticals), metformin HCI + pioglitazone (CAS No. 1115- 70-4 and 112529-15-4), ACTOPLUS MET from Andrx), muraglitazar (CAS No. 331741-94-7) (Bristol-Myers Squibb), naveglitazar (Ligand), oleoylethanolamide (Kadmus Pharmaceuticals), ONO-5129, pioglitazone hydrochloride (CAS No. 111025-46-8 and 112529-15-4) (Takeda), PLX-204 (Plexxikon), PPAR agonists from Genfit, PPAR delta agonists from EIi Lilly, PPAR-alpha agonists from CrystalGenomics, PPAR-gamma modulators and PPAR-β modulators from CareX, rosiglitazone maleate (CAS No. 122324- 73-4 or 155141-29-0) (GlaxoSmithKline), rosiglitazone maleate/glimepir (CAS No. 155141-29-0 and 93479-97-1), AVANDARYL, rosiglitazone maleate/metformin extend (CAS No. 155141-29-0 and 657-24-9), AVANDAMET, rosiglitazone maleate+metformin, AVANDAMET (GlaxoSmithKline), tesaglitazar (AstraZeneca), and an analogue or derivative thereof. 19721. The method of item 19644 wherein the anti- fibrotic agent is a phosphatase inhibitor.
19722. The method of item 19644 wherein the anti- fibrotic agent is a phosphodiesterase (PDE) inhibitor selected from the group consisting of avanafil (Tanabe Seiyaku), dasantafil (CAS No. 569351- 91-3) (Schering-Plough), A-906119 (CAS No. 134072-58-5), DL-850 (Sanofi-Aventis), GRC-3015, GRC-3566, GRC-3886 (Glenmark), HWA-153 (CAS No. 56395-66-5) (Sanofi-Aventis), hydroxypumafentrine (Altana), IBFB-130011, IBFB-14-016, IBFB-140301, IBFB-150007, IBFB-211913 (IBFB Pharma), L-826141 (Merck & Co), medorinone (CAS No. 88296-61-1) (Sanofi-Aventis), MEM-1917 (Memory Pharmaceuticals), ND-1251 (Neuro3d), PDE inhibitors from ICOS, PDE IV inhibitors from Memory Pharmaceuticals and CrystalGenomics, Pharmaprojects No. 2742 and 6141 (Sanofi-Aventis), QAD-171 (Novartis), RHC-2963 (CAS No. 76993-12-9 and 76993-14-1), RPR-117658, RPR-122818 derivatives, SR-2487Q , and RPR- - 132294 (Sanofi-Aventis), SK-350 (ln2Gen), stroke therapy agents from deCODE Genetics, TAS-203 (Taiho), tofimilast (CAS No. 185954-27-2) (Pfizer), UK-371800 (Pfizer), WIN-65579 (CAS No. 158020-82-7) (Sanofi- Aventis), IBFB-130020 (IBFB Pharma), OPC-6535 (CAS No. 145739-56-6) (Otsuka), a phosphodiesterase III inhibitor, enoximone, a phosphodiesterase IV inhibitor, fosfosal, Atopik (Barrier Therapeutics), triflusal, a phosphodiesterase V inhibitor, and an analogue or derivative thereof.
19723. The method of item 19644 wherein the anti- fibrotic agent is a PKC inhibitor.
19724. The method of item 19644 wherein the anti- fibrotic agent is a platelet activating factor antagonist. 19725. The method of item 19644 wherein the anti- fibrotic agent is a platelet-derived growth factor receptor kinase inhibitor.
19726. The method of item 19644 wherein the anti- fibrotic agent is a prolyl hydroxylase inhibitor.
19727. The method of item 19644 wherein the anti- fibrotic agent is a polymorphonuclear neutrophil inhibitor.
19728. The method of item 19644 wherein the anti- fibrotic agent is a protein kinase B inhibitor.
19729. The method of item 19644 wherein the anti- fibrotic agent is a protein kinase C stimulant.
19730. The method of item 19644 wherein the anti- fibrotic agent is a purine nucleoside analogue.
19731. The method of item 19644 wherein the anti- fibrotic agent is a purinoreceptor P2X antagonist.
19732. The method of item 19644 wherein the anti- fibrotic agent is a Raf kinase inhibitor.
19733. The method of item 19644 wherein the anti- fibrotic agent is a reversible inhibitor of ErbB1 and ErbB2.
19734. The method of item 19644 wherein the anti- fibrotic agent is a ribonucleoside triphosphate reductase inhibitor. 19735. The method of item 19644 wherein the anti- fibrotic agent is an SDF-1 antagonist.
19736. The method of item 19644 wherein the anti- fibrotic agent is a sheddase inhibitor.
19737. The method of item 19644 wherein the anti- fibrotic agent is an SRC inhibitor.
19738. The method of item 19644 wherein the anti- fibrotic agent is a stromelysin inhibitor,
19739. The method of item 19644 wherein the anti- fibrotic agent is an Syk kinase inhibitor.
19740. The method of item 19644 wherein the anti- fibrotic agent is a telomerase inhibitor.
19741. The method of item 19644 wherein the anti- fibrotic agent is a TGF beta inhibitor selected from the group consisting of pirfenidone (CAS No. 53179-13-8) (MARNAC), tranilast (CAS No. 53902- 12-8) (Kissei), IN-1130 (ln2Gen), mannose-6-phosphate (BTG), TGF-β antagonists from Inflazyme (Pharmaprojects No. 6075), TGF-β antagonists from Sydney, non-industrial source), TGF-β I receptor kinase inhibitors from EIi Lilly, TGF-β receptor inhibitors from Johnson & Johnson, and an analogue or derivative thereof.
19742. The method of item 19644 wherein the anti- fibrotic agent is a TNFα antagonist or TACE inhibitor selected from the group consisting of adalimumab (CAS No. 331731-18-1) (Cambridge Antibody Technology), AGIX-4207 (AtheroGenics), AGT-1 (Advanced Biotherapy), an anti-inflammatory from Borean Pharma, Cellzome, or Paradigm Therapeutics, anti-inflammatory vaccine (TNF-alpha kinoid) from Neovacs, humanized anti-TNF antibody or an anti-TNF MAb (CB0006) Celltech (UCB), apratastat (CAS No. 287445-51-0) (Wyeth), BMS-561392 (Bristol-Myers Squibb), BN-006 (Bone), certolizumab pegol (CAS No. 428863-50-7 or CH-138 (UCB), cilomilast (CAS No. 153259-65-5) (GlaxoSmithKline), CR-1 (Nuada Pharmaceuticals), CRx-119 (CombinatoRx), D-5410 (UCB)1 dacopafant (CAS No. 125372-33-0) (Sanofi- Aventis), dersalazine (CAS No. 188913-57-7/188913-58-8) (Uriach), etanercept (CAS No. 185243-69-0) (Amgen), ethyl pyruvate (Critical (Critical Therapeutics), golimumab (CAS No. 476181-74-5) (Johnson & Johnson), hormono-immunotherapy from Ipsen, CDP571 (e.g., Humicade from UCB), IC-485 (ICOS), infliximab (CAS No. 170277-31-3) (Johnson & Johnson), IP- 751 (Manhattan Pharmaceuticals), ISIS-104838 (CAS No. 250755-32-9) (ISIS Pharmaceuticals), lenalidomide (CAS No. 191732-72-6) (Celgene), lentinan (CAS No. 37339-90-5) (Ajinomoto), MDL-201112 (CAS No. 142130-73-2) (Sanofi-Aventis), medroxyprogesterone (CAS No. 520-85-4) (InKine Pharmaceutical), N-acetylcysteine (CAS No. 616-91-1) (Zambon), NBE-P2 (DIREVO Biotech), nerelimomab (CAS No. 162774-06-3) (Chiron), OM-294DP (OM PHARMA), onercept (CAS No. 199685-57-9) (Yeda), PASSTNF-alpha (Verigen), pentoxifylline or oxypentifylline (Sanofi-Aventis), Pharmaprojects No. 4091, 4241, 4295, or 4488 (Sanofi-Aventis), Pharmaprojects No. 5480 (Amgen), Pharmaprojects No. 6749 (Cengent), pirfenidone (CAS No. 53179-13-8) (MARNAC), RPR-132294 (Sanofi- Aventis), S5 (F002) (Fulcrum Pharmaceuticals), simvastatin (CAS No. 79902-63-9) (Merck & Co), STA-6292 (Synta Pharmaceuticals), tacrolimus (CAS No. 104987-11-3) (Fujisawa LifeCycle Pharma), talactoferrin alfa (CAS No. 308240-58-6) (Agennix), thalidomide (CAS No. 50-35-1) (Celgene), TNF antagonists form ProStrakan, and Synergen, TNF inhibitors (Amgen), TNF-alpha antagonists from Dynavax Technologies and Jerina AG (Germany), TNF-alpha inhibitors from IBFB Pharma and Xencor (Xencor), torbafylline (CAS No. 105102-21-4) (Sanofi-Aventis), UR-1505 (Uriach), VT-346 (Viron Therapeutics), YSIL6 (Y's Therapeutics), YSTH2 (Y's Therapeutics), NPI~1302a-3 (Nereus Pharmaceuticals, a TNF antagonist from Jerina AG (Germany), dersalazine, and an analogue or derivative thereof.
19743. The method of item 19644 wherein the anti- fibrotic agent is a Toll receptor inhibitor.
19744. The method of item 19644 wherein the anti- fibrotic agent is a tubulin antagonist.
19745. The method of item 19644 wherein the anti- fibrotic agent is a tyrosine kinase inhibitor selected from the group consisting of SU-011248, SUTENT from Pfizer Inc. (New York, NY), BMS-354825, PN- 355 (Paracelsian Pharmaceuticals), AGN-199659 (Allergan), AAL-993 or ABP-309 (Novartis), adaphostin (NIH), AEE-788 (Novartis), AG-013736 (OSI Pharmaceuticals), AG-13736 (Pfizer), ALT-110 (Alteris Therapeutics), AMG-706 (Amgen), anticancer MAbs from Xencor, anti-EGFrvlll MAbs from Abgenix, anti-HER2 MAb from Abiogen, AZD-2171 or AZD-9935 (AstraZeneca), BAY-57-9352 (Bayer), B1BF-1120 (Boehringer Ingelheim), CEP-5214 (Cephalon), CEP-7055 (Cephalon), cetuximab (ImClone Systems), CHIR-200131 and CHIR-258 (Chiron), CP-547632 (OSI Pharmaceuticals), CP-724714 (Pfizer), CT-301 (Creabilis Therapeutics), D- 69491 (Baxter International), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EGFR/ErbB2 inhibitors from Array BioPharma, erlotinib (CAS No. 183319- 69-9) (OSI Pharmaceuticals), EXEL-2880 (Exelixis), FK-778 (Sanofi- Aventis) , gefitinib (CAS No. 184475-35-2) (AstraZeneca), GW-2286 or GW- 654652 (GlaxoSmithKline), her2/neu antigen from AlphaVax, HER-2/neu inhibitor from Generex, Herzyme (Medipad) (Sirna Therapeutics), HKI-272 (Wyeth), HuMax-EGFr (Genmab), idronoxil (CAS No. 81267-65-4) (Novogen), IGF-1 inhibitors from Ontogen, IMC-11F8 (ImCIone Systems), kahalalide F (CAS No. 149204-42-2) (PharmaMar), KDR inhibitor from LG Life Sciences, KDR inhibitors from Abbott Laboratories, KDR kinase inhibitors (UCB), Kdr kinase inhibitors from Merck & Co, KRN-633 and KRN- 951 (Kirin Brewery), KSB-102 (Xenova), lapatinib ditosylate (CAS No. 388082-78-8) (GlaxoSmithKline), matuzumab (Merck KGaA), MDX-214 (Medarex), ME-103 (Pharmexa), MED-A300 (Gerolymatos), MNAC-13 (Lay Line Genomics), nimotuzumab (Center of Molecular Immunology), NSC- 330507 or NSC-707545 (NIH), NV-50 (Novogen), OSI-930 (OSI Pharmaceuticals), panitumumab (Abgenix), pelitinib (CAS No. 287933-82-7) (Wyeth), pertuzumab (CAS No. 380610-27-5) (Genentech), Pharmaprojects No. 3985 (Sanofi-Aventis), prostate cancer therapeutics from Sequenom (SQPC35, SQPC36, SQPC90), removab and remoxab (Trion Pharma), RG- 13022 (CAS No. 136831-48-6), RG-13291 (CAS No. 138989-50-1), or RG- 14620 (CAS No. 136831-49-7) (Sanofi-Aventis), RM-6427 (Romark), RNAi breast cancer therapy from Benitec, RP 53801 (CAS No. 125882-88-4) (Sanofi-Aventis), sorafenib tosylate (Bayer), SU-11657 (Pfizer), Tie-2 antagonists from Semaia (Hybrigenics), Tie-2 inhibitors from Ontogen, trastuzumab (CAS No. 180288-69-1) (Genentech), tyrosine kinase inhibitors from Sanofi-Aventis, U3-1287, U3-1565, U3-1784, U3-1800 (U3 Pharma), vatalanib (Novartis), VEGFR-2 kinase inhibitor from Bristol-Myers Squibb, XL-647 (Exelixis), ZD-6474 (AstraZeneca), ZK-CDK (Schering AG), an EGFR tyrosine kinase inhibitor, EKB-569 (Wyeth), and an analogue or derivative thereof.
19746. The method of item 19644 wherein the anti- fibrotic agent is a VEGF inhibitor.
19747. The method of item 19644 wherein the anti- fibrotic agent is a vitamin D receptor agonist. 19748. The method of item 19644 wherein the anti- fibrotic agent is ZD-6474 (an angiogenesis inhibitor).
19749. The method of item 19644 wherein the anti- fibrotic agent is AP-23573 (an mTOR inhibitor).
19750. The method of item 19644 wherein the anti- fibrotic agent is synthadotin (a tubulin antagonist).
19751. The method of item 19644 wherein the anti- fibrotic agent is S-0885 (a collagenase inhibitor).
19752. The method of item 19644 wherein the anti- fibrotic agent is aplidine (an elongation factor-1 alpha inhibitor).
19753. The method of item 19644 wherein the anti- _ fibrotic agent is ixabepilone (an epithilone).
19754. The method of item 19644 wherein the anti- fibrotic agent is IDN-5390 (an angiogenesis inhibitor).
19755. The method of item 19644 wherein the anti- fibrotic agent is SB-2723005 (an angiogenesis inhibitor).
19756. The method of item 19644 wherein the anti- fibrotic agent is ABT-518 (an angiogenesis inhibitor).
19757. The method of item 19644 wherein the anti- fibrotic agent is combretastatin (an angiogenesis inhibitor). 19758. The method of item 19644 wherein the anti- fibrotic agent is anecortave acetate (an angiogenesis inhibitor).
19759. The method of item 19644 wherein the anti- fibrotic agent is SB-715992 (a kinesin antagonist).
19760. The method of item 19644 wherein the anti- fibrotic agent is temsirolimus (an mTOR inhibitor).
19761. The method of item 19644 wherein the anti- fibrotic agent is adalimumab (a TNFα antagonist).
19762. The method of item 19644 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with an anti-fibrotic agent, and (b) implanting the medical device into the host.
19763. The method of item 19644 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with an anti-infective agent, and (b) implanting the medical device into the host.
19764. The method of item 19644 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with a polymer; and (b) implanting the medical device into the host.
19765. The method of item 19644 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with a composition comprising an anti-fibrotic agent and a polymer, and (b) implanting the medical device into the host. 19766. The method of item 19644 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with a composition comprising an anti-infective agent and a polymer, and (b) implanting the medical device into the host.
19767. The method of item 19644 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with a composition comprising an anti-fibrotic agent, an anti- infective agent and a polymer, and (b) implanting the medical device into the host.
19768. The method of item 19644 wherein the anti- infective agent is an anthracycline.
19769. The method of item 19644 wherein the anti- infective agent is doxorubicin.
19770. The method of item 19644 wherein the anti- infective agent is is mitoxantrone.
19771. The method of item 19644 wherein the anti- infective agent is a fluoropyrimidine.
19772. The method of item 19644 wherein the anti- infective agent is 5-fluorouracil (5-FU).
19773. The method of item 19644 wherein the anti- infective agent is a folic acid antagonist.
19774. The method of item 19644 wherein the anti- infective agent is methotrexate. 19775. The method of item 19644 wherein the anti- infective agent is a podophylotoxin.
19776. The method of item 19644 wherein the anti- infective agent is etoposide.
19777. The method of item 19644 wherein the anti- infective agent is camptothecin.
19778. The method of item 19644 wherein the anti- infective agent is a hydroxyurea.
19779. The method of item 19644 wherein the anti- infective agent is a platinum complex.
19780. The method of item 19644 wherein the anti- infective agent is cisplatin.
19781. The method of item 19644 wherein the composition comprises an antithrombotic agent.
19782. The method of item 19644 wherein the polymer is formed from reactants comprising a naturally occurring polymer.
19783. The method of item 19644 wherein the polymer is formed from reactants comprising protein.
19784. The method of item 19644 wherein the polymer is formed from reactants comprising carbohydrate. 19785. The method of item 19644 wherein the polymer reactants comprising biodegradable polymer.
19786. The method of item 19644 wherein the polymer reactants comprising nonbiodegradable polymer.
19787. The method of item 19644 wherein the polymer reactants comprising collagen.
19788. The method of item 19644 wherein the polymer reactants comprising methylated collagen.
19789. The method of item 19644 wherein the polymer reactants comprising fibrinogen.
19790. The method of item 19644 wherein the polymer . reactants comprising thrombin.
19791. The method of item 19644 wherein the polymer reactants comprising blood plasma.
19792. The method of item 19644 wherein the polymer reactants comprising calcium salt.
19793. The method of item 19644 wherein the polymer reactants comprising an antifibronolytic agent.
19794. The method of item 19644 wherein the polymer reactants comprising fibrinogen analog. 19795. The method of item 19644 wherein the polymer reactants comprising albumin.
19796. The method of item 19644 wherein the polymer reactants comprising plasminogen.
19797. The method of item 19644 wherein the polymer reactants comprising von Willebrands factor.
19798. The method of item 19644 wherein the polymer reactants comprising Factor VIII.
19799. The method of item 19644 wherein the polymer reactants comprising hypoallergenic collagen.
19800. The method of item 19644 wherein the polymer reactants comprising atelopeptidic collagen.
19801. The method of item 19644 wherein the polymer reactants comprising telopeptide collagen.
19802. The method of item 19644 wherein the polymer reactants comprising crosslinked collagen.
19803. The method of item 19644 wherein the polymer reactants comprising aprotinin.
19804. The method of item 19644 wherein the polymer reactants comprising epsilon-amino-n-caproic acid. 19805. The method of item 19644 wherein the polymer is formed from reactants comprising gelatin.
19806. The method of item 19644 wherein the polymer is formed from reactants comprising protein conjugates.
19807. The method of item 19644 wherein the polymer is formed from reactants comprising gelatin conjugates.
19808. The method of item 19644 wherein the polymer is formed from reactants comprising a synthetic polymer.
19809. The method of item 19644 wherein the polymer is formed from reactants comprising a synthetic isocyanate-containing compound.
19810. The method of item 19644 wherein the polymer is formed from reactants comprising a synthetic thiol-containing compound.
19811. The method of item 19644 wherein the polymer is formed from reactants comprising a synthetic compound containing at least two thiol groups.
19812. The method of item 19644 wherein the polymer is formed from reactants comprising a synthetic compound containing at least three thiol groups.
19813. The method of item 19644 wherein the polymer is formed from reactants comprising a synthetic compound containing at least four thiol groups. 19814. The method of item 19644 wherein the polymer is formed from reactants comprising a synthetic amino-containing compound.
19815. The method of item 19644 wherein the polymer is formed from reactants comprising a synthetic compound containing at least two amino groups.
19816. The method of item 19644 wherein the polymer is formed from reactants comprising a synthetic compound containing at least three amino groups.
19817. The method of item 19644 wherein the polymer is formed from reactants comprising a synthetic compound containing at least four amino groups.
19818. The method of item 19644 wherein the polymer is formed from reactants comprising a synthetic compound comprising a carbonyl-oxygen-succinimidyl group.
19819. The method of item 19644 wherein the polymer is formed from reactants comprising a synthetic compound comprising at least two carbonyl-oxygen-succinimidyl groups.
19820. The method of item 19644 wherein the polymer is formed from reactants comprising a synthetic compound comprising at least three carbonyl-oxygen-succinimidyl groups.
19821. The method of item 19644 wherein the polymer is formed from reactants comprising a synthetic compound comprising at least four carbonyl-oxygen-succinimidyl groups. 19822. The method of item 19644 wherein the polymer is formed from reactants comprising a synthetic polyalkylene oxide- containing compound.
19823. The method of item 19644 wherein the polymer is formed from reactants comprising a synthetic compound comprising both polyalkylene oxide and biodegradable polyester blocks.
19824. The method of item 19644 wherein the polymer is formed from reactants comprising a synthetic polyalkylene oxide- containing compound having reactive amino groups.
19825. The method of item 19644 wherein the polymer is formed from reactants comprising a synthetic polyalkylene oxide- containing compound having reactive thiol groups.
19826. The method of item 19644 wherein the polymer is formed from reactants comprising a synthetic polyalkylene oxide- containing compound having reactive carbonyl-oxygen-succinimidyl groups.
19827. The method of item 19644 wherein the polymer is formed from reactants comprising a synthetic compound comprising a biodegradable polyester block.
19828. The method of item 19644 wherein the polymer is formed from reactants comprising a synthetic polymer formed in whole or part from lactic acid or lactide.
19829. The method of item 19644 wherein the polymer is formed from reactants comprising a synthetic polymer formed in whole or part from glycolic acid or glycolide. 19830. The method of item 19644 wherein the polymer is formed from reactants comprising polylysine.
19831. The method of item 19644 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound comprising a polyalkylene oxide portion.
19832. The method of item 19644 wherein the polymer is formed from reactants comprising (a) protein and (b) polylysine.
19833. The method of item 19644 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound having at least four thiol groups.
19834. The method of item 19644 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound having at least four amino groups.
19835. The method of item 19644 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound having at least four carbonyl-oxygen-succinimide groups.
19836. The method of item 19644 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound having a biodegradable region formed from reactants selected from lactic acid, lactide, glycolic acid, glycolide, and epison-caprolactone.
19837. The method of item 19644 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound comprising a polyalkylene oxide portion. 19838. The method of item 19644 wherein the polymer is formed from reactants comprising (a) collagen and (b) polylysine.
19839. The method of item 19644 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound having at least four thiol groups.
19840. The method of item 19644 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound having at least four amino groups.
19841. The method of item 19644 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound having at least four carbonyl-oxygen-succinimide groups.
19842. The method of item 19644 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound having a biodegradable region formed from reactants selected from lactic acid, lactide, glycolic acid, glycolide, and epison-caprolactone.
19843. The method of item 19644 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound comprising a polyalkylene oxide portion.
19844. The method of item 19644 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) polylysine.
19845. The method of item 19644 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound having at least four thiol groups. 19846. The method of item 19644 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound having at least four amino groups.
19847. The method of item 19644 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound having at least four carbonyl-oxygen-succinimide groups.
19848. The method of item 19644 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound having a biodegradable region formed from reactants selected from lactic acid, lactide, glycolic acid, glycolide, and epison-caprolactone.
19849. The method of item 19644 wherein the polymer is formed from reactants comprising hyaluronic acid.
19850. The method of item 19644 wherein the polymer is formed from reactants comprising a hyaluronic acid derivative.
19851. The method of item 19644 wherein the polymer is formed from reactants comprising pentaerythritol poly(ethylene glycol)ether tetra-sulfhydryl of number average molecular weight between 3,000 and 30,000.
19852. The method of item 19644 wherein the polymer is formed from reactants comprising pentaerythritol poly(ethylene glycol)ether tetra-amino of number average molecular weight between 3,000 and 30,000.
19853. The method of item 19644 wherein the polymer is formed from reactants comprising (a) a synthetic compound having a number average molecular weight between 3,000 and 30,000 and comprising a polyalkylene oxide region and multiple nucleophilic groups, and (b) a synthetic compound having a number average molecular weight between 3,000 and 30,000 and comprising a polyalkylene oxide region and multiple electrophilic groups.
19854. The method of item 19644 wherein the composition comprises a colorant.
19855. The method of item 19644 wherein the composition is sterile.
19856. A method as in any one of items 19644-19855, wherein the device is an AV fistula graft.
19857. A method as in any one of items 19644-19855, wherein the device is an AV access graft.
19858. A method as in any one of items 19644-19855, wherein the device is a venous catheter.
19859. A method as in any one of items 19644-19855, wherein the device is a vascular graft.
19860. A method as in any one of items 19644-19855, wherein the device is an implantable port.
19861. A method as in any one of items 19644-19855, wherein the device is an AV shunt. 19862. A method for implanting a medical device comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with i) an anti-fibrotic agent, ii) an anti-infective agent, iii) a polymer, iv) a composition comprising an anti-fibrotic agent and a polymer, v) a composition comprising an anti-infectin agent and a polymer, or vi) a composition comprising an anti-fibrotic agent, an anti-infective agent and a polymer, and (b) implanting the medical device into the host, wherein the medical device is a medical device that comprises a film or a mesh.
19863. The method of item 19862 wherein the anti- fibrotic agent is an adensosine A2A receptor antagonist.
19864. The method of item 19862 wherein the anti- fibrotic agent is an AKT inhibitor.
19865. The method of item 19862 wherein the anti- fibrotic agent is an alpha 2 integrin antagonist, wherein the alpha 2 integrin antagonist is Pharmaprojects No. 5754 (Merck KgaA).
19866. The method of item 19862 wherein the anti- fibrotic agent is an alpha 4 integrin antagonist.
19867. The method of item 19862 wherein the anti- fibrotic agent is an alpha 7 nicotinic receptor agonist.
19868. The method of item 19862 wherein the anti- fibrotic agent is an angiogenesis inhibitor selected from the group consisting of AG-12,958 (Pfizer), ATN-161 (Attenuon LLC), neovastat, an angiogenesis inhibitor from Jerina AG (Germany), NM-3 (Mercian), VGA-1155 (Taisho), FCE-26644 (Pfizer), FCE-26950 (Pfizer), FPMA (Meiji Daries), FR-111142 (Fujisawa), GGTI-298, GM-1306 (Ligand), GPA-1734 (Novartis), NNC-47- 0011 (Novo Nordisk), herbamycin (Nippon Kayaku), lenalidomide (Celegene), IP-10 (NIH), ABT-828 (Abbott), KIN-841 (Tokushima University, Japan), SF-1126 (Semafore Pharmaceuticals), laminin technology (NIH), CHIR-258 (Chiron), NVP-AEW541 (Novartis), NVP-AEW541 (Novartis), Vt16907 (Alchemia), OXI-8007 (Oxigene), EG-3306 (Ark Therapeutics), Maspin (Arriva), ABT-567 (Abbott), PPI-2458 (Praecis Pharmaceuticals), CC-5079, CC-4089 (Celgene), HIF-1 alpha inhibitors (Xenova), S-247 (Pfizer), AP-23573 (Ariad), AZD-9935 (Astra Zeneca), mebendazole (Introgen Therapeutics), MetAP-2 inhibitors (GlaxoSmithKline), AG-615 (Angiogene Pharmaceuticals), Tie-2 antagonists (Hybrigenics), NC-381 , CYC-381 , NC-169, NC-219, NC-383, NC-384, NC-407 (Lorus Therapeutics), ATN-224 (Attenuon), ON-01370 (Onconova), Vitronectin antagonists (Amgen), SDX-103 (Salmedix), Vitronectin antagonists (Shire), CHP (Riemser), TEK (Amgen), Anecortave acetate (Alcon), T46.2 (Matrix Therapeutics), HG-2 (Heptagen), TEM antagonists (Genzyme), Oxi-4500 (Oxigene), ATN-161 (Attenuon), WX-293 (Wilex), M-2025 (Metris Therapeutics), Alphastatin (BioActa), YH-16 (Yantai Rongchang), BIBF- 1120 (Boehringer Ingelheim), BAY-57-9352 (Bayer), AS-1404 (Cancer Research Technology), SC-77964 (Pfizer), glycomimetics (BioTie Therapies), TIE-2 Inhibitors (Ontogen), DIMI, Octamer (Octamer), ABR- 215050 (Active Biotech), ABT-518 (Abbott), KDR inhibitors (Abbott), BSF- 466895 (Abbott), SCH-221153 (Schering-Plough), DAC:antiangiogenic (ConjuChem), TFPI (EntreMed), AZD-2171 (Astra-Zenaca), CDC-394 (Celgene), LY290293 (EIi Lilly), IDN-5390 (Indena), Kdr Kinase Inhibitors (Merck), CT-113020, CT-116433, CT-116563, CT-31890, CT-32228) (Cell Therapeutics), A-299620 (Abbott), TWEAK Inhibitor (Amgen), VEGF modulators (Johnson and Johnson), Tum-N53, tumstatin (Genzyme), Thios- 1 , Thios-2 (Thios Pharmaceuticals), MV-6401 (Miravant Medical Technologies), Spisulosine (PharmaMar), CEP-7055 (Cephalon), AUV-201 (Auvation), LM-609 (EIi Lilly), SKF-106615 (AnorMED), Oglufanide disodium (Cytran), BW-114 (Pharminox), Calreticulin (NIH), WX-678 (Wilex), SD-7784 (Pfizer), WX-UK1 (Wilex), SH-268 (Schering AG), 2-Me-PGA (Celgene), S- 137 (Pfizer), ZD-6126 (Angiogene Pharmaceuticals), SG-292 (SignalGen), Benefin (Lane Labs), A6, A36 (Angstrom), SB-2723005 (GlaxoSmithKline), SC-7 (Cell Therapeutics), ZEN-014 (AEterna Zentaris), 2-methoxyestradiol (EntreMed), NK-130119 (Nippon Kayaku), CC-10004 (Celgene), AVE- 8062A (Ajinomoto), Tacedinaline (Pfizer), Actinonin (Tokyo Metropolitan Institute of Medical Science), Lenalidomide (Celgene), VGA-1155, BTO-956 (SRI International), ER-68203-00 (Eisai), CT-6685 (UCB), JKC-362 (Phoenix Pharmaceuticals), DMI-3798 (DMI Biosciences, Angiomate (Ipsen), ZD-6474 (AstraZeneca), CEP-5214 (Cephalon), Canstatin (Genzyme), NM-3 (Mercian), Oridigm (MediQuest Therapeutics), Exherin (Adherex), BLS-0597 (Boston Life Sciences), PTC-299 (PTC Therapeutics), NPI-2358 (Nereus Pharmaceuticals), CGP-79787 (Novartis), AEE-788 (Novartis), CKD-732 (Chong Kun Dang), CP-564959 (OSI Pharmaceuticals), CM-101 (CarboMed), CT-2584, CT3501 (Cell Therapeutics), combretastatin and analogues and derivatives thereof (Oxigene), Rebimastat (Bristol-Meyers Squibb), Dextrin 2-sulfate (ML Laboratories), Cilengitide (Merk KGaA), NSC-706704 (Pharminox), KRN- 951 (Kirin Brewery), Ukrain, NSC-631570 (Nowicky Pharma), Tecogalan sodium (Daiichi Pharmaceutical), Tz-93 (Tsumura), TBC-1635 (Encysive Pharmaceuticals), TAN-1120 (Takeda), Semaxanib (Pfizer), BDI-7800 (Biopharmacopae), SD-186, SD-983 (Bristol-Meyers Squibb), SB-223245 (GlaxoSmithKline), SC-236 (Pfizer), RWJ-590973 (Johnson and Johnson), ILX-1850 (Genzyme), SC-68488, S-836 (Pfizer), CG-55069-11 (CuraGen), Ki-23057 (Kirin Brewery), CCX-700 (Chemoentryx), Pegaptanib octasodium (Giled Sciences), ANGIOCOL (available from Biostratum Inc.), or an analogue or derivative thereof.
19869. The method of item 19862 wherein the anti- fibrotic agent is an apoptosis antagonist. 19870. The method of item 19862 wherein the anti- fibrotic agent is an apoptosis activator.
19871. The method of item 19862 wherein the anti- fibrotic agent is a beta 1 integrin antagonist.
19872. The method of item 19862 wherein the anti- fibrotic agent is a beta tubulin inhibitor.
19873. The method of item 19862 wherein the anti- fibrotic agent is a blocker of enzyme production in Hepatitis C.
19874. The method of item 19862 wherein the anti- fibrotic agent is a Bruton's tyrosine kinase inhibitor.
. . 19875. The method of item 19862 wherein the anti- fibrotic agent is a calcineurin inhibitor.
19876. The method of item 19862 wherein the anti- fibrotic agent is a caspase 3 inhibitor.
19877. The method of item 19862 wherein the anti- fibrotic agent is a CC chemokine receptor antagonist.
19878. The method of item 19862 wherein the anti- fibrotic agent is a cell cycle inhibitor selected from the group consisting of SNS-595 (Sunesis), synthadotin, KRX-0403, and an analogue or derivative thereof.
19879. The method of item 19862 wherein the anti- fibrotic agent is a cathepsin B inhibitor. 19880. The method of item 19862 wherein the anti- fibrotic agent is a cathepsin K inhibitor, wherein the cathepsin K inhibitor is 462795 (GlaxoSmithKline), INPL-022-D6 (Amura Therapeutics), or an analogue or derivative thereof.
19881. The method of item 19862 wherein the anti- fibrotic agent is a cathepsin L inhibitor.
19882. The method of item 19862 wherein the anti- fibrotic agent is a CD40 antagonist.
19883. The method of item 19862 wherein the anti- fibrotic agent is a chemokine receptor agonist.
19884. The method of item 19862 wherein the anti- fibrotic agent is a chymase inhibitor.
19885. The method of item 19862 wherein the anti- fibrotic agent is a collagenase antagonist.
19886. The method of item 19862 wherein the anti- fibrotic agent is a CXCR antagonist.
19887. The method of item 19862 wherein the anti- fibrotic agent is a cyclin dependent kinase inhibitor selected from the group consisting of a CDK-1 inhibitor, a CDK-2 inhibitor, a CDK- 4 inhibitor, a CDK-6 inhibitor, a CAK1 inhibitor from GPC Biotech and Bristol-Myers Squibb, RGB-286199 (GPC Biotech), an anticancer agent from Astex Technology, a CAK1 inhibitor from GPC Biotech, a CDK inhibitor from Sanofi-Aventis, a CDK1/CDK2 inhibitor from Amgen, a CDK2 inhibitor from SUGEN-2 (Pfizer), a hearing loss therapy agent (Sound Pharmaceuticals), PD-0332991 (Pfizer), RGB-286199 (GPC Biotech), Ro-0505124 (Hoffmann- La Roche), a Ser/Thr kinase inhibitor from Lilly (EIi Lilly), CVT-2584 (CAS No. 199986-75-9) (CV Therapeutics), and an analogue or derivative thereof.
19888. The method of item 19862 wherein the anti- fibrotic agent is a cyclooxygenase 1 inhibitor.
19889. The method of item 19862 wherein the anti- fibrotic agent is a DHFR inhibitor.
19890. The method of item 19862 wherein the anti- fibrotic agent is a dual integrin inhibitor.
19891. The method of item 19862 wherein the anti- fibrotic agent is an elastase inhibitor.
19892. The method of item 19862 wherein the anti- fibrotic agent is an elongation factor-1 alpha inhibitor.
19893. The method of item 19862 wherein the anti- fibrotic agent is an endothelial growth factor antagonist.
19894. The method of item 19862 wherein the anti- fibrotic agent is an endothelial growth factor receptor kinase inhibitor selected from the group consisting of sorafenib tosylate (Bayer), AAL-993 (Novartis), ABP-309 (Novartis), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EXEL- 2880 (Exelixis), GW-654652 (GlaxoSmithKline), a KDR inhibitor from LG Life Sciences, CT-6685 and CT-6729 (UCB), KRN-633 and KRN-951 (Kirin Brewery), OSI-930 (OSI Pharmaceuticals), SP-5.2 (Supratek Pharma), SU-
11657 (Pfizer), a Tie-2 antagonist (Hybrigenics), a VEGFR-2 kinase inhibitor (Bristol-Myers Squibb), XL-647 (Exelixis), a KDR inhibitor from Abbott Laboratories, sorafenib tosylate, and an analogue or derivative thereof.
19895. The method of item 19862 wherein the anti- fibrotic agent is an endotoxin antagonist.
19896. The method of item 19862 wherein the anti- fibrotic agent is an epothilone and tubulin binder.
19897. The method of item 19862 wherein the anti- fibrotic agent is an estrogen receptor antagonist.
19898. The method of item 19862 wherein the anti- fibrotic agent is an FGF inhibitor.
19899. The method of item 19862 wherein the anti- fibrotic agent is a famexyl transferase inhibitor.
19900. The method of item 19862 wherein the anti- fibrotic agent is farnesyltransferase inhibitor selected from the group of A- 197574 (Abbott), a farnesyltransferase inhibitor from Servier, FPTIII (Strathclyde Institute for Drug R), LB-42908 (LG Life Sciences), Pharmaprojects No. 5063 (Genzyme), Pharmaprojects No. 5597 (Ipsen), Yissum Project No. B-1055 (Yissum), and an analogue or derivative thereof.
19901. The method of item 19862 wherein the anti- fibrotic agent is an FLT-3 kinase inhibitor.
19902. The method of item 19862 wherein the anti- fibrotic agent is an FGF receptor kinase inhibitor. 19903. The method of item 19862 wherein the anti- fibrotic agent is a fibrinogen antagonist selected from the group consisting of AUV-201 (Auvation), MG-13926 (Sanofi-Aventis), plasminogen activator (CAS No. 105913-11-9) (from Sanofi-Aventis or UCB)1 plasminogen activator-2 (tPA-2) (Sanofi-Aventis), pro-urokinase (CAS No. 82657-92-9) (Sanofi-Aventis), and an analogue or derivative thereof.
19904. The method of item 19862 wherein the anti- fibrotic agent is a heat shock protein 90 antagonist selected from the group consisting of SRN-005 (Sirenade), geldanamycin, NSC-33050 (17- allylaminogeldanamycin; 17-AAG), 17-dimethylaminoethylamino-17- demethoxy-geldanamycin (17-DMAG), rifabutin (rifamycin XIV, 1',4- didehydro-1-deoxy-1 ,4-dihydro-5'-(2-methylpropyl)-1-oxo-), and an analogue or derivative thereof.
19905. The method of item 19862 wherein the anti- fibrotic agent is a histone deacetylase inhibitor.
19906. The method of item 19862 wherein the anti- fibrotic agent is an HMGCoA reductase inhibitor selected from the group consisting of an atherosclerosis therapeutic from Lipid Sciences, ATI-16000 (ARYx Therapeutics), KS-01-019 (Kos Pharmaceuticals), Pharmaprojects No. 2197 (Sanofi-Aventi), RP 61969 (Sanofi-Aventis), and an analogue or derivative thereof.
19907. The method of item 19862 wherein the anti- fibrotic agent is an ICAM inhibitor.
19908. The method of item 19862 wherein the anti- fibrotic agent is an IL, ICE and IRAK antagonist, wherein the antagonist is a CJ-14877, CP-424174 (Pfizer), NF-61 (Negma-Lerads), and an analogue or derivative thereof.
19909. The method of item 19862 wherein the anti- fibrotic agent is an IL-2 inhibitor.
19910. The method of item 19862 wherein the anti- fibrotic agent is an immunosuppressant selected from the group consisting of teriflunomide (Sanofi Aventis), chlorsulfaquinoxalone (NSC-339004), chlorsulfaquinoxalone sulfate, CS-712 (Sankyo), ismomultin alfa (CAS No. 457913-93-8) (Akzo Nobel), antiallergics from GenPat77, antiinflammatories or AT-005 (Androclus Therapeutics), autoimmune disease therapeutics from EpiVax, BN-007 (Bone), budesonide (CAS No. 51333-22- 3) (MAP Pharmaceuticals), CO-14 (Genzyme), edratide (CAS No. 433922- 67-9) (Teva), EP-314 (Enanta), eprovafen (CAS No. 101335-99-3) (Sanofi- Aventis), HWA-131 (CAS No. 118788-41-3) (Sanofi-Aventis), immunomodulators from MerLion Pharmaceuticals, immunosuppressives from Alchemia, IPL-12 (Inflazyme), MDL-9563 (CAS No. 27086-86-8) (Sanofi-Aventis), Pharmaprojects No. 2330 (Sanofi-Aventis), Pharmaprojects No. 6426 (Abgenix), PXS-25 (Pharmaxis), rosmarinic acid (CAS No. 20283-92-5) (Sanofi-Aventis), RP 42927 or RP 54745 (CAS No. 135330-08-4) (Sanofi-Aventis), SGN-35 (Seattle Genetics), ST-1959 (Sigma-Tau), type I diabetes therapy from SYNX Pharma, UNIL-88 (Debiopharm), VP-025 (Vasogen), VR-694 (Vectura), PRTX-001 (Protalex), and an analogue or derivative thereof.
19911. The method of item 19862 wherein the anti- fibrotic agent is an IMPDH (inosine monophosphate).
19912. The method of item 19862 wherein the anti- fibrotic agent is an integrin antagonist. 19913. The method of item 19862 wherein the anti- fibrotic agent is an interleukin antagonist.
19914. The method of item 19862 wherein the anti- fibrotic agent is an inhibitor of type III receptor tyrosine kinase.
19915. The method of item 19862 wherein the anti- fibrotic agent is an irreversible inhibitor of enzyme methionine aminopeptidase type 2.
19916. The method of item 19862 wherein the anti- fibrotic agent is an isozyme selective delta protein kinase C inhibitor.
19917. The method of item 19862 wherein the anti- fibrotic agent a JAK3 enzyme inhibitor.
19918. The method of item 19862 wherein the anti- fibrotic agent is a JNK inhibitor.
19919. The method of item 19862 wherein the anti- fibrotic agent is a kinase inhibitor.
19920. The method of item 19862 wherein the anti- fibrotic agent is kinesin antagonist.
19921. The method of item 19862 wherein the anti- fibrotic agent is a kinesin antagonist.
19922. The method of item 19862 wherein the anti- fibrotic agent is a leukotriene inhibitor and antagonist selected from the group consisting of ambicromil (CAS No. 58805-38-2) (Sanofi-Aventis), amelubant (CAS No. 346735-24-8) (Boehringer Ingelheim), DW-1141 (Dong Wha), ebselen (Daiichi Pharmaceutical), ibudilast (Kyorin), leucotriene inhibitors from Sanofi-Aventis, lymphotoxin-beta receptor (LT-D) from Biogen Idee, Pharmaprojects No. 1535 and 2728 (CAS No. 119340-33-9) (Sanofi-Aventis), R-112 (Rigel), Rev-5367 (CAS No. 92532-05-3) (Sanofi- Aventis), RG-14893 (CAS No. 141835-49-6) (Sanofi-Aventis), RG-5901-A (CAS No. 101910-24-1), 92532-23-5, RP 66153 (CAS No. 142422-79-5), RP 66364 (CAS No.186916-92-5), RP 69698 (CAS No. 141748-00-7) (Sanofi-Aventis), SC-411930 (Pfizer), SC-41930 (CAS No. 120072-59-5) (Pfizer), SC-50605 (CAS No. 138828^39-4) (Pfizer), SC-51146 (CAS No. 141059-52-1), SC-53228 (CAS No. 153633-01-3) (Pfizer), spaglumic acid (ZY-15106) (CAS No.3106-85-2), 80619-64-3 (Novartis), tipredane (CAS No. 85197-77-9) (Bristol-Myers Squibb), U-75302 (CAS No. 119477-85-9) (Pfizer), and analogue or derivative thereof.
19923. The method of item 19862 wherein the anti- fibrotic agent is an MAP kinase inhibitor.
19924. The method of item 19862 wherein the anti- fibrotic agent is a matrix metalloproteinase inhibitor.
19925. The method of item 19862 wherein the anti- fibrotic agent is an MCP-CCR2 inhibitor.
19926. The method of item 19862 wherein the anti- fibrotic agent is an mTOR inhibitor.
19927. The method of item 19862 wherein the anti- fibrotic agent is an mTOR kinase inhibitor. 19928. The method of item 19862 wherein the anti- fibrotic agent is a microtubule inhibitor selected from the group consisting of antibody-maytansinoid conjugates from Biogen Idee, colchicines (MantiCore Pharmaceuticals), anticancer immunoconjugates from Johnson & Johnson, DIME from Octamer, gni-1f (GNI), huC242-DM4, huMy9-6-DM1 (ImmunoGen), IDN-5404 (Indena), IMO-098, IMOderm (Imotep), mebendazole (Introgen Therapeutics), microtubule poisons from Cambridge Enterprise, paclitaxel such as LOTAX from Aphios (CAS No. 33069-62-4), Genexol-PM from Samyang, Pharmaprojects No. 6383 (Tapestry Pharmaceuticals), RPR-112378 (Sanofi-Aventis), SGN-75 (Seattle Genetics), SPL-7435 (Starpharma), SSR-250411 (Sanofi-Aventis), trastuzumab-DM1 (Genentech), vinorelbine, and an analogue or derivative thereof.
19929. The method of item 19862 wherein the anti- fibrotic agent is an MIF inhibitor.
19930. The method of item 19862 wherein the anti- fibrotic agent is an MMP inhibitor.
19931. The method of item 19862 wherein the anti- fibrotic agent is a neurokinin (NK) antagonist selected from the group consisting of anthrotainin (CAS No. 148084-40-6) (Sanofi-Aventis), an IBS thereapeutic from ArQuIe, MDL-105212A (CAS No. 167261-60-1) (Ssanofi- Aventis), Pharmaprojects No. 2744, 3258 (CAS No. 139167-47-8) 4006, 4201, or 5986 (Sanofi-Aventis), RP 67580 (CAS No. 135911-02-3), SR- 144190 (CAS No. 201152-86-5), SSR-240600, SSR-241586 (Sanofi- Aventis), TKA-457 (Novartis), vestipitant mesylate (CAS No. 334476-64-1) (GlaxoSmithKline), Win-64821 (Sanofi-Aventis), PRX-96026 (Predix Pharmaceuticals), and an analogue or derivative thereof. 19932. The method of item 19862 wherein the anti- fibrotic agent is an NF kappa B inhibitor selected from the group consisting of AVE-0545 or AVE-0547 (Sanofi-Aventis), bortezomib (CAS No. 179324- 69-7) (Millennium Pharmaceuticals), dexanabinol (CAS No. 112924-45-5) (Pharmos), dexlipotam (Viatris), Pharmaprojects No. 6283 (INDRA) (OXiGENE), IPL-576092 (CAS No. 137571-30-3) (Inflazyme), NFKB decoy (Corgentech), NFKB decoy oligo (AnGes MG), NFKB's from Ariad, osteoporosis treatments or S5 (F005) from Fulcrum Pharmaceuticals, P61 (Phytopharm), R-flurbiprofen (CAS No. 5104-49-4) (Encore Pharmaceuticals), and an analogue or derivative thereof.
19933. The method of item 19862 wherein the anti- fibrotic agent is a nitric oxide agonist.
19934. The method of item 19862 wherein the anti- fibrotic agent is an ornithine decarboxylase inhibitor.
19935. The method of item 19862 wherein the anti- fibrotic agent is a p38 MAP kinase inhibitor selected from the group consisting of AZD-6703 (AstraZeneca), JX-401 (Jexys Pharmaceuticals), BMS-2 (Bristol-Myers Squibb), a p38 MAP kinase inhibitor from Novartis, a p38-alpha MAP kinase inhibitor from Amphora, Pharmaprojects No. 5704 (Pharmacopeia), RPR-200765A (Sanofi-Aventis), SD-282 (Johnson & Johnson), TAK-715 (Takeda), and an analogue or derivative thereof.
19936. The method of item 19862 wherein the anti- fibrotic agent is a palmitoyl-protein thioesterase inhibitor.
19937. The method of item 19862 wherein the anti- fibrotic agent is a PDGF receptor kinase inhibitor selected from the group consisting of AAL-993, AMN-107, or ABP-309 (Novartis), AMG-706 (Amgen), BAY-57-9352 (Bayer), CDP-860 (UCB), E-7080 (Eisai), imatinib (CAS No. 152459-95-5) (Novartis), OSI-930 (OSI Pharmaceuticals), RPR- 127963E (Sanofi-Aventis), RWJ-540973 (Johnson & Johnson), sorafenib tosylate (Bayer), SlM 1657 (Pfizer), tandutinib (CAS No. 387867-13-2) (Millennium Pharmaceuticals), vatalanib (Novartis), ZK-CDK (Schering AG), and an analogue or derivative thereof.
19938. The method of item 19862 wherein the anti- fibrotic agent is (-)-halofenate (Metabolex), AMG-131 (Amgen), antidiabetics from Japan Tobacco, AZD-4619, AZD-8450, AZD-8677 (AstraZeneca), DRF-10945, balaglitazone (Dr Reddy's), CS-00088, CS-00098 (Chipscreen Biosciences), E-3030 (Eisai), etalocib (CAS No. 161172-51-6) (EIi Lilly), GSK-641597 (Ligand), GSK-677954 (GlaxoSmithKline), GW-409544 (Ligand), GW-590735 (GlaxoSmithKline), K-111 (Hoffmann-La Roche), LY- 518674 (EIi Lilly), LY-674 (Ligand), LY-929 (Ligand), MC-3001, MC-3002 (MaxoCore Pharmaceuticals), metformin HCI + pioglitazone (CAS No. 1115- 70-4 and 112529-15-4), ACTOPLUS MET from Andrx), muraglitazar (CAS No. 331741-94-7) (Bristol-Myers Squibb), naveglitazar (Ligand), oleoylethanolamide (Kadmus Pharmaceuticals), ONO-5129, pioglitazone hydrochloride (CAS No. 111025-46-8 and 112529-15-4) (Takeda), PLX-204 (Plexxikon), PPAR agonists from Genfit, PPAR delta agonists from EIi Lilly, PPAR-alpha agonists from CrystalGenomics, PPAR-gamma modulators and PPAR-β modulators from CareX, rosiglitazone maleate (CAS No. 122324- 73-4 or 155141-29-0) (GlaxoSmithKline), rosiglitazone maleate/glimepir (CAS No. 155141-29-0 and 93479-97-1), AVANDARYL, rosiglitazone maleate/metformin extend (CAS No. 155141-29-0 and 657-24-9), AVANDAMET, rosiglitazone maleate+metformin, AVANDAMET (GlaxoSmithKline), tesaglitazar (AstraZeneca), and an analogue or derivative thereof. 19939. The method of item 19862 wherein the anti- fibrotic agent is a phosphatase inhibitor.
19940. The method of item 19862 wherein the anti- fibrotic agent is a phosphodiesterase (PDE) inhibitor selected from the group consisting of avanafil (Tanabe Seiyaku), dasantafil (CAS No. 569351- 91-3) (Schering-Plough), A-906119 (CAS No. 134072-58-5), DL-850 (Sanofi-Aventis), GRC-3015, GRC-3566, GRC-3886 (Glenmark), HWA-153 (CAS No. 56395-66-5) (Sanofi-Aventis), hydroxypumafentrine (Altana), IBFB-130011 , IBFB-14-016, IBFB-140301 , IBFB-150007, IBFB-211913 (IBFB Pharma), L-826141 (Merck & Co), medorinone (CAS No. 88296-61-1) (Sanofi-Aventis), MEM-1917 (Memory Pharmaceuticals), ND-1251 (Neuro3d), PDE inhibitors from ICOS, PDE IV inhibitors from Memory Pharmaceuticals and CrystalGenomics, Pharmaprojects No. 2742 and 6141 (Sanofi-Aventis), QAD-171 (Novartis), RHC-2963 (CAS No. 76993-12-9 and 76993-14-1), RPR-117658, RPR-122818 derivatives, SR-24870 , and RPR- 132294 (Sanofi-Aventis), SK-350 (ln2Gen), stroke therapy agents from deCODE Genetics, TAS-203 (Taiho), tofimilast (CAS No. 185954-27-2) (Pfizer), UK-371800 (Pfizer), WIN-65579 (CAS No. 158020-82-7) (Sanofi- Aventis), IBFB-130020 (IBFB Pharma), OPC-6535 (CAS No. 145739-56-6) (Otsuka), a phosphodiesterase III inhibitor, enoximone, a phosphodiesterase IV inhibitor, fosfosal, Atopik (Barrier Therapeutics), triflusal, a phosphodiesterase V inhibitor, and an analogue or derivative thereof.
19941. The method of item 19862 wherein the anti- fibrotic agent is a PKC inhibitor.
19942. The method of item 19862 wherein the anti- fibrotic agent is a platelet activating factor antagonist. 19943. The method of item 19862 wherein the anti- fibrotic agent is a platelet-derived growth factor receptor kinase inhibitor.
19944. The method of item 19862 wherein the anti- fibrotic agent is a prolyl hydroxylase inhibitor.
19945. The method of item 19862 wherein the anti- fibrotic agent is a polymorphonuclear neutrophil inhibitor.
19946. The method of item 19862 wherein the anti- fibrotic agent is a protein kinase B inhibitor.
19947. The method of item 19862 wherein the anti- fibrotic agent is a protein kinase C stimulant.
19948. The method of item 19862 wherein the anti- fibrotic agent is a purine nucleoside analogue.
19949. The method of item 19862 wherein the anti- fibrotic agent is a purinoreceptor P2X antagonist.
19950. The method of item 19862 wherein the anti- fibrotic agent is a Raf kinase inhibitor.
19951. The method of item 19862 wherein the anti- fibrotic agent is a reversible inhibitor of ErbB1 and ErbB2.
19952. The method of item 19862 wherein the anti- fibrotic agent is a ribonucleoside triphosphate reductase inhibitor. 19953. The method of item 19862 wherein the anti- fibrotic agent is an SDF-1 antagonist.
19954. The method of item 19862 wherein the anti- fibrotic agent is a sheddase inhibitor.
19955. The method of item 19862 wherein the anti- fibrotic agent is an SRC inhibitor.
19956. The method of item 19862 wherein the anti- fibrotic agent is a stromelysin inhibitor.
19957. The method of item 19862 wherein the anti- fibrotic agent is an Syk kinase inhibitor.
19958. The method of item 19862 wherein the anti- fibrotic agent is a telomerase inhibitor.
19959. The method of item 19862 wherein the anti- fibrotic agent is a TGF beta inhibitor selected from the group consisting of pirfenidone (CAS No. 53179-13-8) (MARNAC)1 tranilast (CAS No. 53902- 12-8) (Kissei), IN-1130 (ln2Gen), mannose-6-phosphate (BTG), TGF-β antagonists from Inflazyme (Pharmaprojects No. 6075), TGF-β antagonists from Sydney, non-industrial source), TGF-β I receptor kinase inhibitors from EIi Lilly, TGF-β receptor inhibitors from Johnson & Johnson, and an analogue or derivative thereof.
19960. The method of item 19862 wherein the anti- fibrotic agent is a TN Fa antagonist or TACE inhibitor selected from the group consisting of adalimumab (CAS No. 331731-18-1) (Cambridge Antibody Technology), AGIX-4207 (AtheroGenics), AGT-1 (Advanced Biotherapy), an anti-inflammatory from Borean Pharma, Cellzome, or Paradigm Therapeutics, anti-inflammatory vaccine (TNF-alpha kinoid) from Neovacs, humanized anti-TNF antibody or an anti-TNF MAb (CB0006) Celltech (UCB), apratastat (CAS No. 287445-51-0) (Wyeth), BMS-561392 (Bristol-Myers Squibb), BN-006 (Bone), certolizumab pegol (CAS No. 428863-50-7 or CH-138 (UCB), cilomilast (CAS No. 153259-65-5) (GlaxoSmithKline), CR-1 (Nuada Pharmaceuticals), CRx-119 (CombinatoRx), D-5410 (UCB), dacopafant (CAS No. 125372-33-0) (Sanofi- Aventis), dersalazine (CAS No. 188913-57-7/188913-58-8) (Uriach), etanercept (CAS No. 185243-69-0) (Amgen), ethyl pyruvate (Critical (Critical Therapeutics), golimumab (CAS No. 476181-74-5) (Johnson & Johnson), hormono-immunotherapy from Ipsen, CDP571 (e.g., Humicade from UCB), IC-485 (ICOS), infliximab (CAS No. 170277-31-3) (Johnson & Johnson), IP- 751 (Manhattan Pharmaceuticals), ISIS-104838 (CAS No. 250755-32-9) (ISIS Pharmaceuticals), lenalidomide (CAS No. 191732-72-6) (Celgene), lentinan (CAS No. 37339-90-5) (Ajinomoto), MDL-201112 (CAS No. 142130-73-2) (Sanofi-Aventis), medroxyprogesterone (CAS No. 520-85-4) (InKine Pharmaceutical), N-acetylcysteine (CAS No. 616-91-1) (Zambon), NBE-P2 (DIREVO Biotech), nerelimomab (CAS No. 162774-06-3) (Chiron), OM-294DP (OM PHARMA), onercept (CAS No. 199685-57-9) (Yeda), PASSTNF-alpha (Verigen), pentoxifylline or oxypentifylline (Sanofi-Aventis), Pharmaprojects No. 4091 , 4241 , 4295, or 4488 (Sanofi-Aventis), Pharmaprojects No. 5480 (Amgen), Pharmaprojects No. 6749 (Cengent), pirfenidone (CAS No. 53179-13-8) (MARNAC), RPR-132294 (Sanofi- Aventis), S5 (F002) (Fulcrum Pharmaceuticals), simvastatin (CAS No. 79902-63-9) (Merck & Co), STA-6292 (Synta Pharmaceuticals), tacrolimus (CAS No. 104987-11-3) (Fujisawa LifeCycle Pharma), talactoferrin alfa (CAS No. 308240-58-6) (Agennix), thalidomide (CAS No. 50-35-1) (Celgene), TNF antagonists form ProStrakan, and Synergen, TNF inhibitors (Amgen), TNF-alpha antagonists from Dynavax Technologies and Jerina AG (Germany), TNF-alpha inhibitors from IBFB Pharma and Xencor (Xencor), torbafylline (CAS No. 105102-21-4) (Sanofi-Aventis), UR-1505 (Uriach), VT-346 (Viron Therapeutics), YSIL6 (Y's Therapeutics), YSTH2 (Y's Therapeutics), NPI-1302a-3 (Nereus Pharmaceuticals, a TNF antagonist from Jerina AG (Germany), dersalazine, and an analogue or derivative thereof.
19961. The method of item 19862 wherein the anti- fibrotic agent is a Toll receptor inhibitor.
19962. The method of item 19862 wherein the anti- fibrotic agent is a tubulin antagonist.
19963. The method of item 19862 wherein the anti- fibrotic agent is a tyrosine kinase inhibitor selected from the group consisting of SU-011248, SUTENT from Pfizer Inc. (New York, NY), BMS-354825, PN- 355 (Paracelsian Pharmaceuticals), AGN-199659 (Allergan), AAL-993 or ABP-309 (Novartis), adaphostin (NIH), AEE-788 (Novartis), AG-013736 (OSI Pharmaceuticals), AG-13736 (Pfizer), ALT-110 (Alteris Therapeutics), AMG-706 (Amgen), anticancer MAbs from Xencor, anti-EGFrvlll MAbs from Abgenix, anti-HER2 MAb from Abiogen, AZD-2171 or AZD-9935 (AstraZeneca), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), CEP-5214 (Cephalon), CEP-7055 (Cephalon), cetuximab (ImClone Systems), CHIR-200131 and CHIR-258 (Chiron), CP-547632 (OSI Pharmaceuticals), CP-724714 (Pfizer), CT-301 (Creabilis Therapeutics), D- 69491 (Baxter International), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EGFR/ErbB2 inhibitors from Array BioPharma, erlotinib (CAS No. 183319- 69-9) (OSI Pharmaceuticals), EXEL-2880 (Exelixis), FK-778 (Sanofi- Aventis) , gefitinib (CAS No. 184475-35-2) (AstraZeneca), GW-2286 or GW- 654652 (GlaxoSmithKline), her2/neu antigen from AlphaVax, HER-2/neu inhibitor from Generex, Herzyme (Medipad) (Sirna Therapeutics), HKI-272 (Wyeth), HuMax-EGFr (Genmab), idronoxil (CAS No. 81267-65-4) (Novogen), IGF-1 inhibitors from Ontogen, IMC-11F8 (ImCIone Systems), kahalalide F (CAS No. 149204-42-2) (PharmaMar), KDR inhibitor from LG Life Sciences, KDR inhibitors from Abbott Laboratories, KDR kinase inhibitors (UCB), Kdr kinase inhibitors from Merck & Co, KRN-633 and KRN- 951 (Kirin Brewery), KSB-102 (Xenova), lapatinib ditosylate (CAS No. 388082-78-8) (GlaxoSmithKline), matuzumab (Merck KGaA), MDX-214 (Medarex), ME-103 (Pharmexa), MED-A300 (Gerolymatos), MNAC-13 (Lay Line Genomics), nimotuzumab (Center of Molecular Immunology), NSC- 330507 or NSC-707545 (NIH), NV-50 (Novogen), OSI-930 (OSI Pharmaceuticals), panitumumab (Abgenix), pelitinib (CAS No. 287933-82-7) (Wyeth), pertuzumab (CAS No. 380610-27-5) (Genentech), Pharmaprojects No. 3985 (Sanofi-Aventis), prostate cancer therapeutics from Sequenom (SQPC35, SQPC36, SQPC90), removab and remoxab (Trion Pharma), RG- 13022 (CAS No. 136831-48-6), RG-13291 (CAS No. 138989-50-1), or RG- 14620 (CAS No. 136831-49-7) (Sanofi-Aventis), RM-6427 (Romark), RNAi breast cancer therapy from Benitec, RP 53801 (CAS No. 125882-88-4) (Sanofi-Aventis), sorafenib tosylate (Bayer), SU-11657 (Pfizer), Tie-2 antagonists from Semaia (Hybrigenics), Tie-2 inhibitors from Ontogen, trastuzumab (CAS No. 180288-69-1) (Genentech), tyrosine kinase inhibitors from Sanofi-Aventis, U3-1287, U3-1565, U3-1784, U3-1800 (U3 Pharma), vatalanib (Novartis), VEGFR-2 kinase inhibitor from Bristol-Myers Squibb, XL-647 (Exelixis), ZD-6474 (AstraZeneca), ZK-CDK (Schering AG), an EGFR tyrosine kinase inhibitor, EKB-569 (Wyeth), and an analogue or derivative thereof.
19964. The method of item 19862 wherein the anti- fibrotic agent is a VEGF inhibitor.
19965. The method of item 19862 wherein the anti- fibrotic agent is a vitamin D receptor agonist. 19966. The method of item 19862 wherein the anti- fibrotic agent is ZD-6474 (an angiogenesis inhibitor).
19967. The method of item 19862 wherein the anti- fibrotic agent is AP-23573 (an mTOR inhibitor).
19968. The method of item 19862 wherein the anti- fibrotic agent is synthadotin (a tubulin antagonist).
19969. The method of item 19862 wherein the anti- fibrotic agent is S-0885 (a collagenase inhibitor).
19970. The method of item 19862 wherein the anti- fibrotic agent is aplidine (an elongation factor-1 alpha inhibitor).
19971. The method of item 19862 wherein the anti- fibrotic agent is ixabepilone (an epithilone).
19972. The method of item 19862 wherein the anti- fibrotic agent is IDN-5390 (an angiogenesis inhibitor).
19973. The method of item 19862 wherein the anti- fibrotic agent is SB-2723005 (an angiogenesis inhibitor).
19974. The method of item 19862 wherein the anti- fibrotic agent is ABT-518 (an angiogenesis inhibitor).
19975. The method of item 19862 wherein the anti- fibrotic agent is combretastatin (an angiogenesis inhibitor). 19976. The method of item 19862 wherein the anti- fibrotic agent is anecortave acetate (an angiogenesis inhibitor).
19977. The method of item 19862 wherein the anti- fibrotic agent is SB-715992 (a kinesin antagonist).
19978. The method of item 19862 wherein the anti- fibrotic agent is temsirolimus (an mTOR inhibitor).
19979. The method of item 19862 wherein the anti- fibrotic agent is adalimumab (a TNFα antagonist).
19980. The method of item 19862 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with an anti-fibrotic agent, and (b) implanting the medical device into the host.
19981. The method of item 19862 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with an anti-infective agent, and (b) implanting the medical device into the host.
19982. The method of item 19862 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with a polymer; and (b) implanting the medical device into the host.
19983. The method of item 19862 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with a composition comprising an anti-fibrotic agent and a polymer, and (b) implanting the medical device into the host. 19984. The method of item 19862 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with a composition comprising an anti-infective agent and a polymer, and (b) implanting the medical device into the host.
19985. The method of item 19862 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with a composition comprising an anti-fibrotic agent, an anti- infective agent and a polymer, and (b) implanting the medical device into the host.
19986. The method of item 19862 wherein the anti- infective agent is an anthracycline.
19987. The method of item 19862 wherein the anti- infective agent is doxorubicin.
19988. The method of item 19862 wherein the anti- infective agent is is mitoxantrone.
19989. The method of item 19862 wherein the anti- infective agent is a fluoropyrimidine.
19990. The method of item 19862 wherein the anti- infective agent is 5-fluorouracil (5-FU).
19991. The method of item 19862 wherein the anti- infective agent is a folic acid antagonist.
19992. The method of item 19862 wherein the anti- infective agent is methotrexate. 19993. The method of item 19862 wherein the anti- infective agent is a podophylotoxin.
19994. The method of item 19862 wherein the anti- infective agent is etoposide.
19995. The method of item 19862 wherein the anti- infective agent is camptothecin.
19996. The method of item 19862 wherein the anti- infective agent is a hydroxyurea.
19997. The method of item 19862 wherein the anti- infective agent is a platinum complex.
19998. The method of item 19862 wherein the anti- infective agent is cisplatin.
19999. The method of item 19862 wherein the composition comprises an antithrombotic agent.
20000. The method of item 19862 wherein the polymer is formed from reactants comprising a naturally occurring polymer.
20001. The method of item 19862 wherein the polymer is formed from reactants comprising protein.
20002. The method of item 19862 wherein the polymer is formed from reactants comprising carbohydrate. 20003. The method of item 19862 wherein the polymer reactants comprising biodegradable polymer.
20004. The method of item 19862 wherein the polymer reactants comprising nonbiodegradable polymer.
20005. The method of item 19862 wherein the polymer reactants comprising collagen.
20006. The method of item 19862 wherein the polymer reactants comprising methylated collagen.
20007. The method of item 19862 wherein the polymer reactants comprising fibrinogen.
20008. The method of item 19862 wherein the polymer reactants comprising thrombin.
20009. The method of item 19862 wherein the polymer reactants comprising blood plasma.
20010. The method of item 19862 wherein the polymer reactants comprising calcium salt.
20011. The method of item 19862 wherein the polymer reactants comprising an antifibronolytic agent.
20012. The method of item 19862 wherein the polymer reactants comprising fibrinogen analog. 20013. The method of item 19862 wherein the polymer reactants comprising albumin.
20014. The method of item 19862 wherein the polymer reactants comprising plasminogen.
20015. The method of item 19862 wherein the polymer reactants comprising von Willebrands factor.
20016. The method of item 19862 wherein the polymer reactants comprising Factor VIII.
20017. The method of item 19862 wherein the polymer reactants comprising hypoallergenic collagen.
20018. The method of item 19862 wherein the polymer reactants comprising atelopeptidic collagen.
20019. The method of item 19862 wherein the polymer reactants comprising telopeptide collagen.
20020. The method of item 19862 wherein the polymer reactants comprising crosslinked collagen.
20021. The method of item 19862 wherein the polymer reactants comprising aprotinin.
20022. The method of item 19862 wherein the polymer reactants comprising epsilon-amino-n-caproic acid. 20023. The method of item 19862 wherein the polymer is formed from reactants comprising gelatin.
20024. The method of item 19862 wherein the polymer is formed from reactants comprising protein conjugates.
20025. The method of item 19862 wherein the polymer is formed from reactants comprising gelatin conjugates.
20026. The method of item 19862 wherein the polymer is formed from reactants comprising a synthetic polymer.
20027. The method of item 19862 wherein the polymer is formed from reactants comprising a synthetic isocyanate-containing compound.
20028. The method of item 19862 wherein the polymer is formed from reactants comprising a synthetic thiol-containing compound.
20029. The method of item 19862 wherein the polymer is formed from reactants comprising a synthetic compound containing at least two thiol groups.
20030. The method of item 19862 wherein the polymer is formed from reactants comprising a synthetic compound containing at least three thiol groups.
20031. The method of item 19862 wherein the polymer is formed from reactants comprising a synthetic compound containing at least four thiol groups. 20032. The method of item 19862 wherein the polymer is formed from reactants comprising a synthetic amino-containing compound.
20033. The method of item 19862 wherein the polymer is formed from reactants comprising a synthetic compound containing at least two amino groups.
20034. The method of item 19862 wherein the polymer is formed from reactants comprising a synthetic compound containing at least three amino groups.
20035. The method of item 19862 wherein the polymer is formed from reactants comprising a synthetic compound containing at least four amino groups.
20036. The method of item 19862 wherein the polymer is formed from reactants comprising a synthetic compound comprising a carbonyl-oxygen-succinimidyl group.
20037. The method of item 19862 wherein the polymer is formed from reactants comprising a synthetic compound comprising at least two carbonyl-oxygen-succinimidyl groups.
20038. The method of item 19862 wherein the polymer is formed from reactants comprising a synthetic compound comprising at least three carbonyl-oxygen-succinimidyl groups.
20039. The method of item 19862 wherein the polymer is formed from reactants comprising a synthetic compound comprising at least four carbonyl-oxygen-succinimidyl groups. 20040. The method of item 19862 wherein the polymer is formed from reactants comprising a synthetic polyalkylene oxide- containing compound.
20041. The method of item 19862 wherein the polymer is formed from reactants comprising a synthetic compound comprising both polyalkylene oxide and biodegradable polyester blocks.
20042. The method of item 19862 wherein the polymer is formed from reactants comprising a synthetic polyalkylene oxide- containing compound having reactive amino groups.
20043. The method of item 19862 wherein the polymer is formed from reactants comprising a synthetic polyalkylene oxide- containing compound having reactive thiol groups.
20044. The method of item 19862 wherein the polymer is formed from reactants comprising a synthetic polyalkylene oxide- containing compound having reactive carbonyl-oxygen-succinimidyl groups.
20045. The method of item 19862 wherein the polymer is formed from reactants comprising a synthetic compound comprising a biodegradable polyester block.
20046. The method of item 19862 wherein the polymer is formed from reactants comprising a synthetic polymer formed in whole or part from lactic acid or lactide.
20047. The method of item 19862 wherein the polymer is formed from reactants comprising a synthetic polymer formed in whole or part from glycolic acid or glycolide. 20048. The method of item 19862 wherein the polymer is formed from reactants comprising polylysine.
20049. The method of item 19862 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound comprising a polyalkylene oxide portion.
20050. The method of item 19862 wherein the polymer is formed from reactants comprising (a) protein and (b) polylysine.
20051. The method of item 19862 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound having at least four thiol groups.
20052. The method of item 19862 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound having at least four amino groups.
20053. The method of item 19862 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound having at least four carbonyl-oxygen-succinimide groups.
20054. The method of item 19862 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound having a biodegradable region formed from reactants selected from lactic acid, lactide, glycolic acid, glycolide, and epison-caprolactone.
20055. The method of item 19862 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound comprising a polyalkylene oxide portion. ' 20056. The method of item 19862 wherein the polymer is formed from reactants comprising (a) collagen and (b) polylysine.
20057. The method of item 19862 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound having at least four thiol groups.
20058. The method of item 19862 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound having at least four amino groups.
20059. The method of item 19862 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound having at least four carbonyl-oxygen-succinimide groups.
20060. The method of item 19862 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound having a biodegradable region formed from reactants selected from lactic acid, lactide, glycolic acid, glycolide, and epison-caprolactone.
20061. The method of item 19862 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound comprising a polyalkylene oxide portion.
20062. The method of item 19862 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) polylysine.
20063. The method of item 19862 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound having at least four thiol groups. 20064. The method of item 19862 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound having at least four amino groups.
20065. The method of item 19862 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound having at least four carbonyl-oxygen-succinimide groups.
20066. The method of item 19862 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound having a biodegradable region formed from reactants selected from lactic acid, lactide, glycolic acid, glycolide, and epison-caprolactone.
20067. The method of item 19862 wherein the polymer is formed from reactants comprising hyaluronic acid.
20068. The method of item 19862 wherein the polymer is formed from reactants comprising a hyaluronic acid derivative.
20069. The method of item 19862 wherein the polymer is formed from reactants comprising pentaerythritol poly(ethylene glycol)ether tetra-sulfhydryl of number average molecular weight between 3,000 and 30,000.
20070. The method of item 19862 wherein the polymer is formed from reactants comprising pentaerythritol poly(ethylene glycol)ether tetra-amino of number average molecular weight between 3,000 and 30,000.
20071. The method of item 19862 wherein the polymer is formed from reactants comprising (a) a synthetic compound having a number average molecular weight between 3,000 and 30,000 and comprising a polyalkylene oxide region and multiple nucleophilic groups, and (b) a synthetic compound having a number average molecular weight between 3,000 and 30,000 and comprising a polyalkylene oxide region and multiple electrophilic groups.
20072. The method of item 19862 wherein the composition comprises a colorant.
20073. The method of item 19862 wherein the composition is sterile.
20074. A method as in any one of items 19862-20073, wherein the device is a surgical barrier.
20075. A method as in any one of items 19862-20073, wherein the device is a surgical adhesion barrier.
20076. A method as in any one of items 19862-20073, wherein the device is a surgical sheet.
20077. A method as in any one of items 19862-20073, wherein the device is a surgical patch.
20078. A method as in any one of items 19862-20073, wherein the device is a surgical wrap.
20079. A method as in any one of items 19862-20073, wherein the device is a vascular wrap. 20080. A method as in any one of items 19862-20073, wherein the device is a perivascular wrap.
20081. A method as in any one of items 19862-20073, wherein the device is a adventitial wrap.
20082. A method as in any one of items 19862-20073, wherein the device is a periadventitial wrap.
20083. A method as in any one of items 19862-20073, wherein the device is an adventitial sheet.
20084. A method as in any one of items 19862-20073, wherein the device is a perivascular mesh.
20085. A method as in any one of items 19862-20073, wherein the device is a bandage.
20086. A method as in any one of items 19862-20073, wherein the device is a liquid bandage.
20087. A method as in any one of items 19862-20073, wherein the device is a surgical dressing.
20088. A method as in any one of items 19862-20073, wherein the device is a gauze.
20089. A method as in any one of items 19862-20073, wherein the device is a fabric. 20090. A method as in any one of items 19862-20073, wherein the device is a tape.
20091. A method as in any one of items 19862-20073, wherein the device is a surgical membrane.
20092. A method as in any one of items 19862-20073, wherein the device is a polymer matrix.
20093. A method as in any one of items 19862-20073, wherein the device is a tissue covering.
20094. A method as in any one of items 19862-20073, wherein the device is a surgical matrix.
20095. A method as in any one of items 19862-20073, wherein the device is an envelope.
20096. A method as in any one of items 19862-20073, wherein the device is a tissue covering.
20097. A method for implanting a medical device comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with i) an anti-fibrotic agent, ii) an anti-infective agent, iii) a polymer, iv) a composition comprising an anti-fibrotic agent and a polymer, v) a composition comprising an anti-infective agent and a polymer, or vi) a composition comprising an anti-fibrotic agent, an anti- infective agent and a polymer, and (b) implanting the medical device into the host, wherein the medical device is a glaucoma drainage device. 20098. The method of item 20097 wherein the anti- fibrotic agent is an adensosine A2A receptor antagonist.
20099. The method of item 20097 wherein the anti- fibrotic agent is an AKT inhibitor.
20100. The method of item 20097 wherein the anti- fibrotic agent is an alpha 2 integrin antagonist, wherein the alpha 2 integrin antagonist is Pharmaprojects No. 5754 (Merck KgaA).
20101. The method of item 20097 wherein the anti- fibrotic agent is an alpha 4 integrin antagonist.
20102. The method of item 20097 wherein the anti- fibrotic agent is an alpha 7 nicotinic receptor agonist.
20103. The method of item 20097 wherein the anti- fibrotic agent is an angiogenesis inhibitor selected from the group consisting of AG-12,958 (Pfizer), ATN-161 (Attention LLC), neovastat, an angiogenesis inhibitor from Jerina AG (Germany), NM-3 (Mercian), VGA-1155 (Taisho), FCE-26644 (Pfizer), FCE-26950 (Pfizer), FPMA (Meiji Daries), FR-111142 (Fujisawa), GGTI-298, GM-1306 (Ligand), GPA-1734 (Novartis), NNC-47- 0011 (Novo Nordisk), herbamycin (Nippon Kayaku), lenalidomide (Celegene), IP-10 (NIH), ABT-828 (Abbott), KIN-841 (Tokushima University, Japan), SF-1126 (Semafore Pharmaceuticals), laminin technology (NIH), CHIR-258 (Chiron), NVP-AEW541 (Novartis), NVP-AEW541 (Novartis), Vt16907 (Alchemia), OXI-8007 (Oxigene), EG-3306 (Ark Therapeutics), Maspin (Arriva), ABT-567 (Abbott), PPI-2458 (Praecis Pharmaceuticals), CC-5079, CC-4089 (Celgene), HIF-1 alpha inhibitors (Xenova), S-247 (Pfizer), AP-23573 (Ariad), AZD-9935 (Astra Zeneca), mebendazole (Introgen Therapeutics), MetAP-2 inhibitors (GlaxoSmithKline), AG-615 (Angiogene Pharmaceuticals), Tie-2 antagonists (Hybrigenics), NC-381 , CYC-381, NC-169, NC-219, NC-383, NC-384, NC-407 (Lorus Therapeutics), ATN-224 (Attenuon), ON-01370 (Onconova), Vitronectin antagonists (Amgen), SDX-103 (Salmedix), Vitronectin antagonists (Shire), CHP (Riemser), TEK (Amgen), Anecortave acetate (Alcon), T46.2 (Matrix Therapeutics), HG-2 (Heptagen), TEM antagonists (Genzyme), Oxi-4500 (Oxigene), ATN-161 (Attenuon), WX-293 (Wilex), M-2025 (Metris Therapeutics), Alphastatin (BioActa), YH-16 (Yantai Rongchang), BIBF- 1120 (Boehringer Ingelheim), BAY-57-9352 (Bayer), AS-1404 (Cancer Research Technology), SC-77964 (Pfizer), glycomimetics (BioTie Therapies), TIE-2 Inhibitors (Ontogen), DIMI, Octamer (Octamer), ABR- 215050 (Active Biotech), ABT-518 (Abbott), KDR inhibitors (Abbott), BSF- 466895 (Abbott), SCH-221153 (Schering-Plough), DAC.antiangiogenic (ConjuChem), TFPI (EntreMed), AZD-2171 (Astra-Zenaca), CDC-394 (Celgene), LY290293 (EIi Lilly), IDN-5390 (Indena), Kdr Kinase Inhibitors (Merck), CT-113020, CT-116433, CT-116563, CT-31890, CT-32228) (CeII- Therapeutics), A-299620 (Abbott), TWEAK Inhibitor (Amgen), VEGF modulators (Johnson and Johnson), Tum-N53, tumstatin (Genzyme), Thios- 1 , Thios-2 (Thios Pharmaceuticals), MV-6401 (Miravant Medical Technologies), Spisulosine (PharmaMar), CEP-7055 (Cephalon), AUV-201 (Auvation), LM-609 (EIi Lilly), SKF-106615 (AnorMED), Oglufanide disodium (Cytran), BW-114 (Pharminox), Calreticulin (NIH), WX-678 (Wilex), SD-7784 (Pfizer), WX-UK1 (Wilex), SH-268 (Schering AG), 2-Me-PGA (Celgene), S- 137 (Pfizer), ZD-6126 (Angiogene Pharmaceuticals), SG-292 (SignalGen), Benefin (Lane Labs), A6, A36 (Angstrom), SB-2723005 (GlaxoSmithKline), SC-7 (Cell Therapeutics), ZEN-014 (AEterna Zentaris), 2-methoxyestradiol (EntreMed), NK-130119 (Nippon Kayaku), CC-10004 (Celgene), AVE- 8062A (Ajinomoto), Tacedinaline (Pfizer), Actinonin (Tokyo Metropolitan Institute of Medical Science), Lenalidomide (Celgene), VGA-1155, BTO-956 (SRI International), ER-68203-00 (Eisai), CT-6685 (UCB), JKC-362 (Phoenix Pharmaceuticals), DMI-3798 (DMI Biosciences, Angiomate (Ipsen), ZD-6474 (AstraZeneca), CEP-5214 (Cephalon), Canstatin (Genzyme), NM-3 (Mercian), Oridigm (MediQuest Therapeutics), Exherin (Adherex), BLS-0597 (Boston Life Sciences), PTC-299 (PTC Therapeutics), NPI-2358 (Nereus Pharmaceuticals), CGP-79787 (Novartis), AEE-788 (Novartis), CKD-732 (Chong Kun Dang), CP-564959 (OSI Pharmaceuticals), CM-101 (CarboMed), CT-2584, CT3501 (Cell Therapeutics), combretastatin and analogues and derivatives thereof (Oxigene), Rebimastat (Bristol-Meyers Squibb), Dextrin 2-sulfate (ML Laboratories), Cilengitide (Merk KGaA), NSC-706704 (Pharminox), KRN- 951 (Kirin Brewery), Ukrain, NSC-631570 (Nowicky Pharma), Tecogalan sodium (Daiichi Pharmaceutical), Tz-93 (Tsumura), TBC-1635 (Encysive Pharmaceuticals), TAN-1120 (Takeda), Semaxanib (Pfizer), BDI-7800 (Biopharmacopae), SD-186, SD-983 (Bristol-Meyers Squibb), SB-223245 (GlaxoSmithKline), SC-236 (Pfizer), RWJ-590973 (Johnson and Johnson), ILX-1850 (Genzyme), SC-68488, S-836 (Pfizer), CG-55069-11 (CuraGen), Ki-23057 (Kirin Brewery), CCX-700 (Chemoentryx), Pegaptanib octasodium (Giled Sciences), ANGIOCOL (available from Biostratum Inc.), or an analogue or derivative thereof.
20104. The method of item 20097 wherein the anti- fibrotic agent is an apoptosis antagonist.
20105. The method of item 20097 wherein the anti- fibrotic agent is an apoptosis activator.
20106. The method of item 20097 wherein the anti- fibrotic agent is a beta 1 integrin antagonist.
20107. The method of item 20097 wherein the anti- fibrotic agent is a beta tubulin inhibitor. 20108. The method of item 20097 wherein the anti- fibrotic agent is a blocker of enzyme production in Hepatitis C.
20109. The method of item 20097 wherein the anti- fibrotic agent is a Brυton's tyrosine kinase inhibitor.
20110. The method of item 20097 wherein the anti- fibrotic agent is a calcineurin inhibitor.
20111. The method of item 20097 wherein the anti- fibrotic agent is a caspase 3 inhibitor.
20112. The method of item 20097 wherein the anti- fibrotic agent is a CC chemokine receptor antagonist.
20113. The method of item 20097 wherein the anti- fibrotic agent is a cell cycle inhibitor selected from the group consisting of SNS-595 (Sunesis), synthadotin, KRX-0403, and an analogue or derivative thereof.
20114. The method of item 20097 wherein the anti- fibrotic agent is a cathepsin B inhibitor.
20115. The method of item 20097 wherein the anti- fibrotic agent is a cathepsin K inhibitor, wherein the cathepsin K inhibitor is 462795 (GlaxoSmithKline), INPL-022-D6 (Amura Therapeutics), or an analogue or derivative thereof.
20116. The method of item 20097 wherein the anti- fibrotic agent is a cathepsin L inhibitor. 20117. The method of item 20097 wherein the anti- fibrotic agent is a CD40 antagonist.
20118. The method of item 20097 wherein the anti- fibrotic agent is a chemokine receptor agonist.
20119. The method of item 20097 wherein the anti- fibrotic agent is a chymase inhibitor.
20120. The method of item 20097 wherein the anti- fibrotic agent is a collagenase antagonist.
20121. The method of item 20097 wherein the anti- fibrotic agent is a CXCR antagonist.
20122. The method of item 20097 wherein the anti- fibrotic agent is a cyclin dependent kinase inhibitor selected from the group consisting of a CDK-1 inhibitor, a CDK-2 inhibitor, a CDK- 4 inhibitor, a CDK-6 inhibitor, a CAK1 inhibitor from GPC Biotech and Bristol-Myers Squibb, RGB-286199 (GPC Biotech), an anticancer agent from Astex Technology, a CAK1 inhibitor from GPC Biotech, a CDK inhibitor from Sanofi-Aventis, a CDK1/CDK2 inhibitor from Amgen, a CDK2 inhibitor from SUGEN-2 (Pfizer), a hearing loss therapy agent (Sound Pharmaceuticals), PD-0332991 (Pfizer), RGB-286199 (GPC Biotech), Ro-0505124 (Hoffmann- La Roche), a Ser/Thr kinase inhibitor from Lilly (EIi Lilly), CVT-2584 (CAS No. 199986-75-9) (CV Therapeutics), and an analogue or derivative thereof,
20123. The method of item 20097 wherein the anti- fibrotic agent is a cyclooxygenase 1 inhibitor. 20124. The method of item 20097 wherein the anti- fibrotic agent is a DHFR inhibitor.
20125. The method of item 20097 wherein the anti- fibrotic agent is a dual integrin inhibitor.
20126. The method of item 20097 wherein the anti- fibrotic agent is an elastase inhibitor.
20127. The method of item 20097 wherein the anti- fibrotic agent is an elongation factor-1 alpha inhibitor.
20128. The method of item 20097 wherein the anti- fibrotic agent is an endothelial growth factor antagonist.
20129. The method of item 20097 wherein the anti- fibrotic agent is an endothelial growth factor receptor kinase inhibitor selected from the group consisting of sorafenib tosylate (Bayer), AAL-993 (Novartis), ABP-309 (Novartis), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EXEL- 2880 (Exelixis), GW-654652 (GlaxoSmithKline), a KDR inhibitor from LG Life Sciences, CT-6685 and CT-6729 (UCB), KRN-633 and KRN-951 (Kirin Brewery), OSI-930 (OSI Pharmaceuticals), SP-5.2 (Supratek Pharma), SU- 11657 (Pfizer), a Tie-2 antagonist (Hybrigenics), a VEGFR-2 kinase inhibitor (Bristol-Myers Squibb), XL-647 (Exelixis), a KDR inhibitor from Abbott Laboratories, sorafenib tosylate, and an analogue or derivative thereof.
20130. The method of item 20097 wherein the anti- fibrotic agent is an endotoxin antagonist. 20131. The method of item 20097 wherein the anti- fibrotic agent is an epothilone and tubulin binder.
20132. The method of item 20097 wherein the anti- fibrotic agent is an estrogen receptor antagonist.
20133. The method of item 20097 wherein the anti- fibrotic agent is an FGF inhibitor.
20134. The method of item 20097 wherein the anti- fibrotic agent is a farnexyl transferase inhibitor.
20135. The method of item 20097 wherein the anti- fibrotic agent is farnesyltransferase inhibitor selected from the group of A- 197574 (Abbott), a farnesyltransferase inhibitor from Servier, FPTIII (Strathclyde Institute for Drug R), LB-42908 (LG Life Sciences), Pharmaprojects No. 5063 (Genzyme), Pharmaprojects No. 5597 (Ipsen), Yissum Project No. B-1055 (Yissum), and an analogue or derivative thereof.
20136. The method of item 20097 wherein the anti- fibrotic agent is an FLT-3 kinase inhibitor.
20137. The method of item 20097 wherein the anti- fibrotic agent is an FGF receptor kinase inhibitor.
20138. The method of item 20097 wherein the anti- fibrotic agent is a fibrinogen antagonist selected from the group consisting of AUV-201 (Auvation), MG-13926 (Sanofi-Aventis), plasminogen activator (CAS No. 105913-11-9) (from Sanofi-Aventis or UCB), plasminogen activator-2 (tPA-2) (Sanofi-Aventis), pro-urokinase (CAS No. 82657-92-9) (Sanofi-Aventis), and an analogue or derivative thereof. 20139. The method of item 20097 wherein the anti- fibrotic agent is a heat shock protein 90 antagonist selected from the group consisting of SRN-005 (Sirenade), geldanamycin, NSC-33050 (17- allylaminogeldanamycin; 17-AAG), 17-dimethylaminoethylamino-17- demethoxy-geldanamycin (17-DMAG), rifabutin (rifamycin XIV, 1',4- didehydro-1-deoxy-1 ,4-dihydro-5'-(2-methylpropyl)-1-oxo-), and an analogue or derivative thereof.
20140. The method of item 20097 wherein the anti- fibrotic agent is a histone deacetylase inhibitor.
20141. The method of item 20097 wherein the anti- fibrotic agent is an HMGCoA reductase inhibitor selected from the group consisting of an atherosclerosis therapeutic from Lipid Sciences, ATM 6000 (ARYx Therapeutics), KS-01-019 (Kos Pharmaceuticals), Pharmaprojects No. 2197 (Sanofi-Aventi), RP 61969 (Sanofi-Aventis), and an analogue or derivative thereof.
20142. The method of item 20097 wherein the anti- fibrotic agent is an ICAM inhibitor.
20143. The method of item 20097 wherein the anti- fibrotic agent is an IL, ICE and IRAK antagonist, wherein the antagonist is a CJ-14877, CP-424174 (Pfizer), NF-61 (Negma-Lerads), and an analogue or derivative thereof.
20144. The method of item 20097 wherein the anti- fibrotic agent is an IL-2 inhibitor.
20145. The method of item 20097 wherein the anti- fibrotic agent is an immunosuppressant selected from the group consisting of teriflunomide (Sanofi Aventis), chlorsulfaquinoxalone (NSC-339004), chlorsulfaquinoxalone sulfate, CS-712 (Sankyo), ismomultin alfa (CAS No. 457913-93-8) (Akzo Nobel), antiallergics from GenPat77, antiinflammatories or AT-005 (Androclus Therapeutics), autoimmune disease therapeutics from EpiVax, BN-007 (Bone), budesonide (CAS No. 51333-22- 3) (MAP Pharmaceuticals), CO-14 (Genzyme), edratide (CAS No. 433922- 67-9) (Teva), EP-314 (Enanta), eprovafen (CAS No. 101335-99-3) (Sanofi- Aventis), HWA-131 (CAS No. 118788-41-3) (Sanofi-Aventis), immunomodulators from MerLion Pharmaceuticals, immunosuppressives from Alchemia, IPL-12 (Inflazyme), MDL-9563 (CAS No. 27086-86-8) (Sanofi-Aventis), Pharmaprojects No. 2330 (Sanofi-Aventis), Pharmaprojects No. 6426 (Abgenix), PXS-25 (Pharmaxis), rosmarinic acid (CAS No. 20283-92-5) (Sanofi-Aventis), RP 42927 or RP 54745 (CAS No. 135330-08-4) (Sanofi-Aventis), SGN-35 (Seattle Genetics), ST-1959 (Sigma-Tau), type I diabetes therapy from SYNX Pharma, UNIL-88 (Debiopharm), VP-025 (Vasogen), VR-694 (Vectura), PRTX-001 (Protalex), and an analogue or derivative thereof.
20146. The method of item 20097 wherein the anti- fibrotic agent is an IMPDH (inosine monophosphate).
20147. The method of item 20097 wherein the anti- fibrotic agent is an integrin antagonist.
20148. The method of item 20097 wherein the anti- fibrotic agent is an interleukin antagonist.
20149. The method of item 20097 wherein the anti- fibrotic agent is an inhibitor of type III receptor tyrosine kinase. 20150. The method of item 20097 wherein the anti- fibrotic agent is an irreversible inhibitor of enzyme methionine aminopeptidase type 2.
20151. The method of item 20097 wherein the anti- fibrotic agent is an isozyme selective delta protein kinase C inhibitor.
20152. The method of item 20097 wherein the anti- fibrotic agent a JAK3 enzyme inhibitor.
20153. The method of item 20097 wherein the anti- fibrotic agent is a JNK inhibitor.
20154. The method of item 20097 wherein the anti- fibrotic agent is a kinase inhibitor.
20155. The method of item 20097 wherein the anti- fibrotic agent is kinesin antagonist.
20156. The method of item 20097 wherein the anti- fibrotic agent is a kinesin antagonist.
20157. The method of item 20097 wherein the anti- fibrotic agent is a leukotriene inhibitor and antagonist selected from the group consisting of ambicromil (CAS No. 58805-38-2) (Sanofi-Aventis), amelubant (CAS No. 346735-24-8) (Boehringer Ingelheim), DW-1141 (Dong Wha), ebselen (Daiichi Pharmaceutical), ibudilast (Kyorin), leucotriene inhibitors from Sanofi-Aventis, lymphotoxin-beta receptor (LT-D) from Biogen Idee, Pharmaprojects No. 1535 and 2728 (CAS No. 119340-33-9) (Sanofi-Aventis), R-112 (Rigel), Rev-5367 (CAS No. 92532-05-3) (Sanofi- Aventis), RG-14893 (CAS No. 141835-49-6) (Sanofi-Aventis), RG-5901-A (CAS No. 101910-24-1), 92532-23-5, RP 66153 (CAS No. 142422-79-5), RP 66364 (CAS No.186916-92-5), RP 69698 (CAS No. 141748-00-7) (Sanofi-Aventis), SC-411930 (Pfizer), SC-41930 (CAS No. 120072-59-5) (Pfizer), SC-50605 (CAS No. 138828-39-4) (Pfizer), SC-51146 (CAS No. 141059-52-1), SC-53228 (CAS No. 153633-01-3) (Pfizer), spaglumic acid (ZY-15106) (CAS No.3106-85-2), 80619-64-3 (Novartis), tipredane (CAS No. 85197-77-9) (Bristol-Myers Squibb), U-75302 (CAS No. 119477-85-9) (Pfizer), and analogue or derivative thereof.
20158. The method of item 20097 wherein the anti- fibrotic agent is an MAP kinase inhibitor.
20159. The method of item 20097 wherein the anti- fibrotic agent is a matrix metalloproteinase inhibitor.
20160. The method of item 20097 wherein the anti- fibrotic agent is an MCP-CCR2 inhibitor.
20161. The method of item 20097 wherein the anti- fibrotic agent is an mTOR inhibitor.
20162. The method of item 20097 wherein the anti- fibrotic agent is an mTOR kinase inhibitor.
20163. The method of item 20097 wherein the anti- fibrotic agent is a microtubule inhibitor selected from the group consisting of antibody-maytansinoid conjugates from Biogen Idee, colchicines (MantiCore Pharmaceuticals), anticancer immunoconjugates from Johnson & Johnson, DIME from Octamer, gni-1f (GNI), huC242-DM4, huMy9-6-DM1 (ImmunoGen), IDN-5404 (Indena), IMO-098, IMOderm (Imotep), mebendazole (Introgen Therapeutics), microtubule poisons from Cambridge Enterprise, paclitaxel such as LOTAX from Aphios (CAS No. 33069-62-4), Genexol-PM from Samyang, Pharmaprojects No. 6383 (Tapestry Pharmaceuticals), RPR-112378 (Sanofi-Aventis), SGN-75 (Seattle Genetics), SPL-7435 (Starpharma), SSR-250411 (Sanofi-Aventis), trastuzumab-DM1 (Genentech), vinorelbine, and an analogue or derivative thereof.
20164. The method of item 20097 wherein the anti- fibrotic agent is an MIF inhibitor.
20165. The method of item 20097 wherein the anti- fibrotic agent is an MMP inhibitor.
20166. The method of item 20097 wherein the anti- fibrotic agent is a neurokinin (NK) antagonist selected from the group consisting of anthrotainin (CAS No. 148084-40-6) (Sanofi-Aventis), an IBS thereapeutic from ArQuIe, MDL-105212A (CAS No. 167261-60-1) (Ssanofi- Aventis), Pharmaprojects No. 2744, 3258 (CAS No. 139167-47-8) 4006, 4201 , or 5986 (Sanofi-Aventis), RP 67580 (CAS No. 135911-02-3), SR- 144190 (CAS No. 201152-86-5), SSR-240600, SSR-241586 (Sanofi- Aventis), TKA-457 (Novartis), vestipitant mesylate (CAS No. 334476-64-1) (GlaxoSmithKline), Win-64821 (Sanofi-Aventis), PRX-96026 (Predix Pharmaceuticals), and an analogue or derivative thereof.
20167. The method of item 20097 wherein the anti- fibrotic agent is an NF kappa B inhibitor selected from the group consisting of AVE-0545 or AVE-0547 (Sanofi-Aventis), bortezomib (CAS No. 179324- 69-7) (Millennium Pharmaceuticals), dexanabinol (CAS No. 112924-45-5) (Pharmos), dexlipotam (Viatris), Pharmaprojects No. 6283 (INDRA) (OXiGENE), IPL-576092 (CAS No. 137571-30-3) (Inflazyme), NFKB decoy (Corgentech), NFKB decoy oligo (AnGes MG), NFKB's from Ariad, osteoporosis treatments or S5 (F005) from Fulcrum Pharmaceuticals, P61 (Phytopharm), R-flurbiprofen (CAS No. 5104-49-4) (Encore Pharmaceuticals), and an analogue or derivative thereof.
20168. The method of item 20097 wherein the anti- fibrotic agent is a nitric oxide agonist.
20169. The method of item 20097 wherein the anti- fibrotic agent is an ornithine decarboxylase inhibitor.
20170. The method of item 20097 wherein the anti- fibrotic agent is a p38 MAP kinase inhibitor selected from the group consisting of AZD-6703 (AstraZeneca), JX-401 (Jexys Pharmaceuticals), BMS-2 (Bristol-Myers Squibb), a p38 MAP kinase inhibitor from Novartis, a p38-alpha MAP kinase inhibitor from Amphora, Pharmaprojects No. 5704 (Pharmacopeia), RPR-200765A (Sanofi-Aventis), SD-282 (Johnson & Johnson), TAK-715 (Takeda), and an analogue or derivative thereof.
20171. The method of item 20097 wherein the anti- fibrotic agent is a palmitoyl-protein thioesterase inhibitor.
20172. The method of item 20097 wherein the anti- fibrotic agent is a PDGF receptor kinase inhibitor selected from the group consisting of AAL-993, AMN-107, or ABP-309 (Novartis), AMG-706 (Amgen), BAY-57-9352 (Bayer), CDP-860 (UCB), E-7080 (Eisai), imatinib (CAS No. 152459-95-5) (Novartis), OSI-930 (OSI Pharmaceuticals), RPR- 127963E (Sanofi-Aventis), RWJ-540973 (Johnson & Johnson), sorafenib tosylate (Bayer), SU-11657 (Pfizer), tandutinib (CAS No. 387867-13-2) (Millennium Pharmaceuticals), vatalanib (Novartis), ZK-CDK (Schering AG), and an analogue or derivative thereof. 20173. The method of item 20097 wherein the anti- fibrotic agent is (-)-halofenate (Metabolex), AMG-131 (Amgen), antidiabetics from Japan Tobacco, AZD-4619, AZD-8450, AZD-8677 (AstraZeneca), DRF-10945, balaglitazone (Dr Reddy's), CS-00088, CS-00098 (Chipscreen Biosciences), E-3030 (Eisai), etalocib (CAS No. 161172-51-6) (EIi Lilly), GSK-641597 (Ligand), GSK-677954 (GlaxoSmithKline), GW-409544 (Ligand), GW-590735 (GlaxoSmithKline), K-111 (Hoffmann-La Roche), LY- 518674 (EIi Lilly), LY-674 (Ligand), LY-929 (Ligand), MC-3001 , MC-3002 (MaxoCore Pharmaceuticals), metformin HCI + pioglitazone (CAS No. 1115- 70-4 and 112529-15-4), ACTOPLUS MET from Andrx), muraglitazar (CAS No. 331741-94-7) (Bristol-Myers Squibb), naveglitazar (Ligand), oleoylethanolamide (Kadmus Pharmaceuticals), ONO-5129, pioglitazone hydrochloride (CAS No. 111025-46-8 and 112529-15-4) (Takeda), PLX-204 (Plexxikon), PPAR agonists from Genfit, PPAR delta agonists from EIi Lilly, PPAR-alpha agonists from CrystalGenomics, PPAR-gamma modulators and PPAR-β modulators from CareX, rosiglitazone maleate (CAS No. 122324- 73-4 or 155141-29-0) (GlaxoSmithKline), rosiglitazone maleate/glimepir (CAS No. 155141-29-0 and 93479-97-1), AVANDARYL, rosiglitazone maleate/metformin extend (CAS No. 155141-29-0 and 657-24-9), AVANDAMET, rosiglitazone maleate+metformin, AVANDAMET (GlaxoSmithKline), tesaglitazar (AstraZeneca), and an analogue or derivative thereof.
20174. The method of item 20097 wherein the anti- fibrotic agent is a phosphatase inhibitor.
20175. The method of item 20097 wherein the anti- fibrotic agent is a phosphodiesterase (PDE) inhibitor selected from the group consisting of avanafil (Tanabe Seiyaku), dasantafil (CAS No. 569351- 91-3) (Schering-Plough), A-906119 (CAS No. 134072-58-5), DL-850 (Sanofi-Aventis), GRC-3015, GRC-3566, GRC-3886 (Glenmark), HWA-153 (CAS No. 56395-66-5) (Sanofi-Aventis), hydroxypumafentrine (Altana), IBFB-130011 , IBFB-14-016, IBFB-140301 , IBFB-150007, IBFB-211913 (IBFB Pharma), L-826141 (Merck & Co), medorinone (CAS No. 88296-61-1) (Sanofi-Aventis), MEM-1917 (Memory Pharmaceuticals), ND-1251 (Neuro3d), PDE inhibitors from ICOS, PDE IV inhibitors from Memory Pharmaceuticals and CrystalGenomics, Pharmaprojects No. 2742 and 6141 (Sanofi-Aventis), QAD-171 (Novartis), RHC-2963 (CAS No. 76993-12-9 and 76993-14-1), RPR-117658, RPR-122818 derivatives, SR-24870 , and RPR- 132294 (Sanofi-Aventis), SK-350 (ln2Gen), stroke therapy agents from deCODE Genetics, TAS-203 (Taiho), tofimilast (CAS No. 185954-27-2) (Pfizer), UK-371800 (Pfizer), WIN-65579 (CAS No. 158020-82-7) (Sanofi- Aventis), IBFB-130020 (IBFB Pharma), OPC-6535 (CAS No. 145739-56-6) (Otsuka), a phosphodiesterase III inhibitor, enoximone, a phosphodiesterase IV inhibitor, fosfosal, Atopik (Barrier Therapeutics), triflusal, a phosphodiesterase V inhibitor, and an analogue or derivative thereof.
20176. The method of item 20097 wherein the anti- fibrotic agent is a PKC inhibitor.
20177. The method of item 20097 wherein the anti- fibrotic agent is a platelet activating factor antagonist.
20178. The method of item 20097 wherein the anti- fibrotic agent is a platelet-derived growth factor receptor kinase inhibitor.
20179. The method of item 20097 wherein the anti- fibrotic agent is a prolyl hydroxylase inhibitor.
20180. The method of item 20097 wherein the anti- fibrotic agent is a polymorphonuclear neutrophil inhibitor. 20181. The method of item 20097 wherein the anti- fibrotic agent is a protein kinase B inhibitor.
20182. The method of item 20097 wherein the anti- fibrotic agent is a protein kinase C stimulant.
20183. The method of item 20097 wherein the anti- fibrotic agent is a purine nucleoside analogue.
20184. The method of item 20097 wherein the anti- fibrotic agent is a purinoreceptor P2X antagonist.
20185. The method of item 20097 wherein the anti- fibrotic agent is a Raf kinase inhibitor.
20186. The method of item 20097 wherein the anti- fibrotic agent is a reversible inhibitor of ErbB1 and ErbB2.
20187. The method of item 20097 wherein the anti- fibrotic agent is a ribonucleoside triphosphate reductase inhibitor.
20188. The method of item 20097 wherein the anti- fibrotic agent is an SDF-1 antagonist.
20189. The method of item 20097 wherein the anti- fibrotic agent is a sheddase inhibitor.
20190. The method of item 20097 wherein the anti- fibrotic agent is an SRC inhibitor. 20191. The method of item 20097 wherein the anti- fibrotic agent is a stromelysin inhibitor.
20192. The method of item 20097 wherein the anti- fibrotic agent is an Syk kinase inhibitor.
20193. The method of item 20097 wherein the anti- fibrotic agent is a telomerase inhibitor.
20194. The method of item 20097 wherein the anti- fibrotic agent is a TGF beta inhibitor selected from the group consisting of pirfenidone (CAS No. 53179-13-8) (MARNAC), tranilast (CAS No. 53902- 12-8) (Kissei), IN-1130 (ln2Gen), mannose-6-phosphate (BTG), TGF-β antagonists from Inflazyme (Pharmaprojects No. 6075), TGF-β antagonists from Sydney, non-industrial source), TGF-β I receptor kinase inhibitors from EIi Lilly, TGF-β receptor inhibitors from Johnson & Johnson, and an analogue or derivative thereof.
20195. The method of item 20097 wherein the anti- fibrotic agent is a TNFα antagonist or TACE inhibitor selected from the group consisting of adalimumab (CAS No. 331731-18-1) (Cambridge Antibody Technology), AGIX-4207 (AtheroGenics), AGT-1 (Advanced Biotherapy), an anti-inflammatory from Borean Pharma, Cellzome, or Paradigm Therapeutics, anti-inflammatory vaccine (TNF-alpha kinoid) from Neovacs, humanized anti-TNF antibody or an anti-TNF MAb (CB0006) Celltech (UCB), apratastat (CAS No. 287445-51-0) (Wyeth), BMS-561392 (Bristol-Myers Squibb), BN-006 (Bone), certolizumab pegol (CAS No. 428863-50-7 or CH-138 (UCB), cilomilast (CAS No. 153259-65-5) (GlaxoSmithKline), CR-1 (Nuada Pharmaceuticals), CRx-119 (CombinatoRx), D-5410 (UCB), dacopafant (CAS No. 125372-33-0) (Sanofi- Aventis), dersalazine (CAS No. 188913-57-7/188913-58-8) (Uriach), etanercept (CAS No. 185243-69-0) (Amgen), ethyl pyruvate (Critical (Critical Therapeutics), golimumab (CAS No. 476181-74-5) (Johnson & Johnson), hormono-immunotherapy from Ipsen, CDP571 (e.g., Humicade from UCB), IC-485 (ICOS), infliximab (CAS No. 170277-31-3) (Johnson & Johnson), IP- 751 (Manhattan Pharmaceuticals), ISIS-104838 (CAS No. 250755-32-9) (ISIS Pharmaceuticals), lenalidomide (CAS No. 191732-72-6) (Celgene), lentinan (CAS No. 37339-90-5) (Ajinomoto), MDL-201112 (CAS No. 142130-73-2) (Sanofi-Aventis), medroxyprogesterone (CAS No. 520-85-4) (InKine Pharmaceutical), N-acetylcysteine (CAS No. 616-91-1) (Zambon), NBE-P2 (DIREVO Biotech), nerelimomab (CAS No. 162774-06-3) (Chiron), OM-294DP (OM PHARMA), onercept (CAS No. 199685-57-9) (Yeda), PASSTNF-alpha (Verigen), pentoxifylline or oxypentifylline (Sanofi-Aventis), Pharmaprojects No. 4091 , 4241 , 4295, or 4488 (Sanofi-Aventis), Pharmaprojects No. 5480 (Amgen), Pharmaprojects No. 6749 (Cengent), pirfenidone (CAS No. 53179-13-8) (MARNAC), RPR-132294 (Sanofi- Aventis), S5 (F002) (Fulcrum Pharmaceuticals), simvastatin (CAS No. 79902-63-9) (Merck & Co), STA-6292 (Synta Pharmaceuticals), tacrolimus (CAS No. 104987-11-3) (Fujisawa LifeCycle Pharma), talactoferrin alfa (CAS No. 308240-58-6) (Agennix), thalidomide (CAS No. 50-35-1) (Celgene), TNF antagonists form ProStrakan, and Synergen, TNF inhibitors (Amgen), TNF-alpha antagonists from Dynavax Technologies and Jerina AG (Germany), TNF-alpha inhibitors from IBFB Pharma and Xencor (Xencor), torbafylline (CAS No. 105102-21-4) (Sanofi-Aventis), UR-1505 (Uriach), VT-346 (Viron Therapeutics), YSIL6 (Y's Therapeutics), YSTH2 (Y's Therapeutics), NPI-1302a-3 (Nereus Pharmaceuticals, a TNF antagonist from Jerina AG (Germany), dersalazine, and an analogue or derivative thereof.
20196. The method of item 20097 wherein the anti- fibrotic agent is a Toll receptor inhibitor. 20197. The method of item 20097 wherein the anti- fibrotic agent is a tubulin antagonist.
20198. The method of item 20097 wherein the anti- fibrotic agent is a tyrosine kinase inhibitor selected from the group consisting of SU-011248, SUTENT from Pfizer Inc. (New York, NY), BMS-354825, PN- 355 (Paracelsian Pharmaceuticals), AGN-199659 (Allergan), AAL-993 or ABP-309 (Novartis), adaphostin (NIH), AEE-788 (Novartis), AG-013736 (OSI Pharmaceuticals), AG-13736 (Pfizer), ALT-110 (Alteris Therapeutics), AMG-706 (Amgen), anticancer MAbs from Xencor, anti-EGFrvlll MAbs from Abgenix, anti-HER2 MAb from Abiogen, AZD-2171 or AZD-9935 (AstraZeneca), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), CEP-5214 (Cephalon), CEP-7055 (Cephalon), cetuximab (ImClone Systems), CHIR-200131 and CHIR-258 (Chiron), CP-547632 (OSI Pharmaceuticals), CP-724714 (Pfizer), CT-301 (Creabilis Therapeutics), D- 69491 (Baxter International), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EGFR/ErbB2 inhibitors from Array BioPharma, erlotinib (CAS No. 183319- 69-9) (OSI Pharmaceuticals), EXEL-2880 (Exelixis), FK-778 (Sanofi- Aventis) , gefitinib (CAS No. 184475-35-2) (AstraZeneca), GW-2286 or GW- 654652 (GlaxoSmithKline), her2/neu antigen from AlphaVax, HER-2/neu inhibitor from Generex, Herzyme (Medipad) (Sirna Therapeutics), HKI-272 (Wyeth), HuMax-EGFr (Genmab), idronoxil (CAS No. 81267-65-4) (Novogen), IGF-1 inhibitors from Ontogen, IMC-11F8 (ImClone Systems), kahalalide F (CAS No. 149204-42-2) (PharmaMar), KDR inhibitor from LG Life Sciences, KDR inhibitors from Abbott Laboratories, KDR kinase inhibitors (UCB), Kdr kinase inhibitors from Merck & Co, KRN-633 and KRN- 951 (Kirin Brewery), KSB-102 (Xenova), lapatinib ditosylate (CAS No. 388082-78-8) (GlaxoSmithKline), matuzumab (Merck KGaA), MDX-214 (Medarex), ME-103 (Pharmexa), MED-A300 (Gerolymatos), MNAC-13 (Lay Line Genomics), nimotuzumab (Center of Molecular Immunology), NSC- 330507 or NSC-707545 (NIH), NV-50 (Novogen), OSI-930 (OSI Pharmaceuticals), panitumumab (Abgenix), pelitinib (CAS No. 287933-82-7) (Wyeth), pertuzumab (CAS No. 380610-27-5) (Genentech), Pharmaprojects No. 3985 (Sanofi-Aventis), prostate cancer therapeutics from Sequenom (SQPC35, SQPC36, SQPC90), removab and remoxab (Trion Pharma), RG- 13022 (CAS No. 136831-48-6), RG-13291 (CAS No. 138989-50-1), or RG- 14620 (CAS No. 136831-49-7) (Sanofi-Aventis), RM-6427 (Romark), RNAi breast cancer therapy from Benitec, RP 53801 (CAS No. 125882-88-4) (Sanofi-Aventis), sorafenib tosylate (Bayer), SU-11657 (Pfizer), Tie-2 antagonists from Semaia (Hybrigenics), Tie-2 inhibitors from Ontogen, trastuzumab (CAS No. 180288-69-1) (Genentech), tyrosine kinase inhibitors from Sanofi-Aventis, U3-1287, U3-1565, U3-1784, U3-1800 (U3 Pharma), vatalanib (Novartis), VEGFR-2 kinase inhibitor from Bristol-Myers Squibb, XL-647 (Exelixis), ZD-6474 (AstraZeneca), ZK-CDK (Schering AG), an EGFR tyrosine kinase inhibitor, EKB-569 (Wyeth), and an analogue or derivative thereof.
20199. The method of item 20097 wherein the anti- fibrotic agent is a VEGF inhibitor.
20200. The method of item 20097 wherein the anti- fibrotic agent is a vitamin D receptor agonist.
20201. The method of item 20097 wherein the anti- fibrotic agent is ZD-6474 (an angiogenesis inhibitor).
20202. The method of item 20097 wherein the anti- fibrotic agent is AP-23573 (an mTOR inhibitor).
20203. The method of item 20097 wherein the anti- fibrotic agent is synthadotin (a tubulin antagonist). 20204. The method of item 20097 wherein the anti- fibrotic agent is S-0885 (a collagenase inhibitor).
20205. The method of item 20097 wherein the anti- fibrotic agent is aplidine (an elongation factor-1 alpha inhibitor).
20206. The method of item 20097 wherein the anti- fibrotic agent is ixabepilone (an epithilone).
20207. The method of item 20097 wherein the anti- fibrotic agent is IDN-5390 (an angiogenesis inhibitor).
20208. The method of item 20097 wherein the anti- fibrotic agent is SB-2723005 (an angiogenesis inhibitor).
20209. The method of item 20097 wherein the anti- fibrotic agent is ABT-518 (an angiogenesis inhibitor).
20210. The method of item 20097 wherein the anti- fibrotic agent is combretastatin (an angiogenesis inhibitor).
20211. The method of item 20097 wherein the anti- fibrotic agent is anecortave acetate (an angiogenesis inhibitor).
20212. The method of item 20097 wherein the anti- fibrotic agent is SB-715992 (a kinesin antagonist).
20213. The method of item 20097 wherein the anti- fibrotic agent is temsirolimus (an mTOR inhibitor). 20214. The method of item 20097 wherein the anti- fibrotic agent is adalimumab (a TNFα antagonist).
20215. The method of item 20097 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with an anti-fibrotic agent, and (b) implanting the medical device into the host.
20216. The method of item 20097 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with an anti-infective agent, and (b) implanting the medical device into the host.
20217. The method of item 20097 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with a polymer; and (b) implanting the medical device into the host.
20218. The method of item 20097 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with a composition comprising an anti-fibrotic agent and a polymer, and (b) implanting the medical device into the host.
20219. The method of item 20097 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with a composition comprising an anti-infective agent and a polymer, and (b) implanting the medical device into the host.
20220. The method of item 20097 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with a composition comprising an anti-fibrotic agent, an anti- infective agent and a polymer, and (b) implanting the medical device into the host.
20221. The method of item 20097 wherein the anti- infective agent is an anthracycline.
20222. The method of item 20097 wherein the anti- infective agent is doxorubicin.
20223. The method of item 20097 wherein the anti- infective agent is is mitoxantrone.
20224. The method of item 20097 wherein the anti- infective agent is a fluoropyrimidine.
20225. The method of item 20097 wherein the anti- infective agent is 5-fluorouracil (5-FU).
20226. The method of item 20097 wherein the anti- infective agent is a folic acid antagonist.
20227. The method of item 20097 wherein the anti- infective agent is methotrexate.
20228. The method of item 20097 wherein the anti- infective agent is a podophylotoxin.
20229. The method of item 20097 wherein the anti- infective agent is etoposide. 20230. The method of item 20097 wherein the anti- infective agent is camptothecin.
20231. The method of item 20097 wherein the anti- infective agent is a hydroxyurea.
20232. The method of item 20097 wherein the anti- infective agent is a platinum complex.
20233. The method of item 20097 wherein the anti- infective agent is cisplatin.
20234. The method of item 20097 wherein the composition comprises an anti-thrombotic agent.
20235. The method of item 20097 wherein the polymer is formed from reactants comprising a naturally occurring polymer.
20236. The method of item 20097 wherein the polymer is formed from reactants comprising protein.
20237. The method of item 20097 wherein the polymer is formed from reactants comprising carbohydrate.
20238. The method of item 20097 wherein the polymer is formed from reactants comprising biodegradable polymer.
20239. The method of item 20097 wherein the polymer is formed from reactants comprising nonbiodegradable polymer. 20240. The method of item 20097 wherein the polymer reactants comprising collagen.
20241. The method of item 20097 wherein the polymer reactants comprising methylated collagen.
20242. The method of item 20097 wherein the polymer reactants comprising fibrinogen.
20243. The method of item 20097 wherein the polymer reactants comprising thrombin.
20244. The method of item 20097 wherein the polymer reactants comprising blood plasma.
20245. The method of item 20097 wherein the polymer reactants comprising calcium salt.
20246. The method of item 20097 wherein the polymer reactants comprising an antifibronolytic agent.
20247. The method of item 20097 wherein the polymer reactants comprising fibrinogen analog.
20248. The method of item 20097 wherein the polymer reactants comprising albumin.
20249. The method of item 20097 wherein the polymer reactants comprising plasminogen. 20250. The method of item 20097 wherein the polymer reactants comprising von Willebrands factor.
20251. The method of item 20097 wherein the polymer reactants comprising Factor VIII.
20252. The method of item 20097 wherein the polymer reactants comprising hypoallergenic collagen.
20253. The method of item 20097 wherein the polymer reactants comprising atelopeptidic collagen.
20254. The method of item 20097 wherein the polymer reactants comprising telopeptide collagen.
20255. The method of item 20097 wherein the polymer reactants comprising crosslinked collagen.
20256. The method of item 20097 wherein the polymer reactants comprising aprotinin.
20257. The method of item 20097 wherein the polymer reactants comprising epsilon-amino-n-caproic acid.
20258. The method of item 20097 wherein the polymer reactants comprising gelatin.
20259. The method of item 20097 wherein the polymer reactants comprising protein conjugates. 20260. The method of item 20097 wherein the polymer is formed from reactants comprising gelatin conjugates.
20261. The method of item 20097 wherein the polymer is formed from reactants comprising a synthetic polymer.
20262. The method of item 20097 wherein the polymer is formed from reactants comprising a synthetic isocyanate-containing compound.
20263. The method of item 20097 wherein the polymer is formed from reactants comprising a synthetic thiol-containing compound.
20264. The method of item 20097 wherein the polymer is formed from reactants comprising a synthetic compound containing at least two thiol groups.
20265. The method of item 20097 wherein the polymer is formed from reactants comprising a synthetic compound containing at least three thiol groups.
20266. The method of item 20097 wherein the polymer is formed from reactants comprising a synthetic compound containing at least four thiol groups.
20267. The method of item 20097 wherein the polymer is formed from reactants comprising a synthetic amino-containing compound. 20268. The method of item 20097 wherein the polymer is formed from reactants comprising a synthetic compound containing at least two amino groups.
20269. The method of item 20097 wherein the polymer is formed from reactants comprising a synthetic compound containing at least three amino groups.
20270. The method of item 20097 wherein the polymer is formed from reactants comprising a synthetic compound containing at least four amino groups.
20271. The method of item 20097 wherein the polymer is formed from reactants comprising a synthetic compound comprising a carbonyl-oxygen-succinimidyl group.
20272. The method of item 20097 wherein the polymer is formed from reactants comprising a synthetic compound comprising at least two carbonyl-oxygen-succinimidyl groups.
20273. The method of item 20097 wherein the polymer is formed from reactants comprising a synthetic compound comprising at least three carbonyl-oxygen-succinimidyl groups.
20274. The method of item 20097 wherein the polymer is formed from reactants comprising a synthetic compound comprising at least four carbonyl-oxygen-succinimidyl groups.
20275. The method of item 20097 wherein the polymer is formed from reactants comprising a synthetic polyalkylene oxide- containing compound. 20276. The method of item 20097 wherein the polymer is formed from reactants comprising a synthetic compound comprising both poiyalkylene oxide and biodegradable polyester blocks.
20277. The method of item 20097 wherein the polymer is formed from reactants comprising a synthetic poiyalkylene oxide- containing compound having reactive amino groups.
20278. The method of item 20097 wherein the polymer is formed from reactants comprising a synthetic poiyalkylene oxide- containing compound having reactive thiol groups.
20279. The method of item 20097 wherein the polymer is formed from reactants comprising a synthetic poiyalkylene oxide- containing compound having reactive carbonyl-oxygen-succinimidyl groups.
20280. The method of item 20097 wherein the polymer is formed from reactants comprising a synthetic compound comprising a biodegradable polyester block.
20281. The method of item 20097 wherein the polymer is formed from reactants comprising a synthetic polymer formed in whole or part from lactic acid or lactide.
20282. The method of item 20097 wherein the polymer is formed from reactants comprising a synthetic polymer formed in whole or part from glycolic acid or glycolide.
20283. The method of item 20097 wherein the polymer is formed from reactants comprising polylysine. 20284. The method of item 20097 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound comprising a polyalkylene oxide portion.
20285. The method of item 20097 wherein the polymer is formed from reactants comprising (a) protein and (b) polylysine.
20286. The method of item 20097 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound having at least four thiol groups.
20287. The method of item 20097 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound having at least four amino groups.
20288. The method of item 20097 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound having at least four carbonyl-oxygen-succinimide groups.
20289. The method of item 20097 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound having a biodegradable region formed from reactants selected from lactic acid, lactide, glycolic acid, glycolide, and epison-caprolactone.
20290. The method of item 20097 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound comprising a polyalkylene oxide portion.
20291. The method of item 20097 wherein the polymer is formed from reactants comprising (a) collagen and (b) polylysine. 20292. The method of item 20097 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound having at least four thiol groups.
20293. The method of item 20097 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound having at least four amino groups.
20294. The method of item 20097 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound having at least four carbonyl-oxygen-succinimide groups.
20295. The method of item 20097 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound having a biodegradable region formed from reactants selected from lactic acid, lactide, glycolic acid, glycolide, and epison-caprolactone.
20296. The method of item 20097 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound comprising a polyalkylene oxide portion.
20297. The method of item 20097 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) polylysine.
20298. The method of item 20097 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound having at least four thiol groups. 20299. The method of item 20097 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound having at least four amino groups.
20300. The method of item 20097 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound having at least four carbonyl-oxygen-succinimide groups.
20301. The method of item 20097 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound having a biodegradable region formed from reactants selected from lactic acid, lactide, glycolic acid, glycolide, and epison-caprolactone.
20302. The method of item 20097 wherein the polymer is formed from reactants comprising hyaluronic acid.
20303. The method of item 20097 wherein the polymer is formed from reactants comprising a hyaluronic acid derivative.
20304. The method of item 20097 wherein the polymer is formed from reactants comprising pentaerythritol poly(ethylene glycol)ether tetra-sulfhydryl of number average molecular weight between 3,000 and 30,000.
20305. The method of item 20097 wherein the polymer is formed from reactants comprising pentaerythritol poly(ethylene glycol)ether tetra-amino of number average molecular weight between 3,000 and 30,000.
20306. The method of item 20097 wherein the polymer is formed from reactants comprising (a) a synthetic compound having a number average molecular weight between 3,000 and 30,000 and comprising a polyalkylene oxide region and multiple nucleophilic groups, and (b) a synthetic compound having a number average molecular weight between 3,000 and 30,000 and comprising a polyalkylene oxide region and multiple electrophilic groups.
20307. The method of item 20097 wherein the composition comprises a colorant.
20308. The method of item 20097 wherein the composition is sterile.
20309. A method as in any one of items 20097-20308, wherein the device is a glaucoma drainage device comprising a plate and a tube.
20310. A method for implanting a medical device comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with i) an anti-fibrotic agent, ii) an anti-infective agent, iii) a polymer, iv) a composition comprising an anti-fibrotic agent and a polymer, v) a composition comprising an anti-infective agent and a polymer, or vi) a composition comprising an anti-fibrotic agent, an anti- infective agent and a polymer, and (b) implanting the medical device into the host, wherein the medical device is a prosthetic heart valve or a component thereof.
20311. The method of item 20310 wherein the anti- fibrotic agent is an adensosine A2A receptor antagonist.
20312. The method of item 20310 wherein the anti- fibrotic agent is an AKT inhibitor. 20313. The method of item 20310 wherein the anti- fibrotic agent is an alpha 2 integrin antagonist, wherein the alpha 2 integrin antagonist is Pharmaprojects No. 5754 (Merck KgaA).
20314. The method of item 20310 wherein the anti- fibrotic agent is an alpha 4 integrin antagonist.
20315. The method of item 20310 wherein the anti- fibrotic agent is an alpha 7 nicotinic receptor agonist.
20316. The method of item 20310 wherein the anti- fibrotic agent is an angiogenesis inhibitor selected from the group consisting of AG-12,958 (Pfizer), ATN-161 (Attenuon LLC), neovastat, an angiogenesis inhibitor from Jerina AG (Germany), NM-3 (Mercian), VGA-1155 (Taisho), FCE-26644 (Pfizer), FCE-26950 (Pfizer), FPMA (Meiji Daries), FR-111142 (Fujisawa), GGTI-298, GM-1306 (Ligand), GPA-1734 (Novartis), NNC-47- 0011 (Novo Nordisk), herbamycin (Nippon Kayaku), lenalidomide (Celegene), IP-10 (NIH), ABT-828 (Abbott), KIN-841 (Tokushima University, Japan), SF-1126 (Semafore Pharmaceuticals), laminin technology (NIH), CHIR-258 (Chiron), NVP-AEW541 (Novartis), NVP-AEW541 (Novartis), Vt16907 (Alchemia), OXI-8007 (Oxigene), EG-3306 (Ark Therapeutics), Maspin (Arriva), ABT-567 (Abbott), PPI-2458 (Praecis Pharmaceuticals), CC-5079, CC-4089 (Celgene), HIF-1 alpha inhibitors (Xenova), S-247 (Pfizer), AP-23573 (Ariad), AZD-9935 (Astra Zeneca), mebendazole (Introgen Therapeutics), MetAP-2 inhibitors (GlaxoSmithKline), AG-615 (Angiogene Pharmaceuticals), Tie-2 antagonists (Hybrigenics), NC-381 , CYC-381 , NC-169, NC-219, NC-383, NC-384, NC-407 (Lorus Therapeutics), ATN-224 (Attenuon), ON-01370 (Onconova), Vitronectin antagonists (Amgen), SDX-103 (Salmedix), Vitronectin antagonists (Shire), CHP (Riemser), TEK (Amgen), Anecortave acetate (Alcon), T46.2 (Matrix Therapeutics), HG-2 (Heptagen), TEM antagonists (Genzyme), Oxi-4500 (Oxigene), ATN-161 (Attention), WX-293 (Wilex), M-2025 (Metris Therapeutics), Alphastatin (BioActa), YH-16 (Yantai Rongchang), BIBF- 1120 (Boehringer Ingelheim), BAY-57-9352 (Bayer), AS-1404 (Cancer Research Technology), SC-77964 (Pfizer), glycomimetics (BioTie Therapies), TIE-2 Inhibitors (Ontogen), DIMI, Octamer (Octamer), ABR- 215050 (Active Biotech), ABT-518 (Abbott), KDR inhibitors (Abbott), BSF- 466895 (Abbott), SCH-221153 (Schering-Plough), DAC:antiangiogenic (ConjuChem), TFPI (EntreMed), AZD-2171 (Astra-Zenaca), CDC-394 (Celgene), LY290293 (EIi Lilly), IDN-5390 (Indena), Kdr Kinase Inhibitors (Merck), CT-113020, CT-116433, CT-116563, CT-31890, CT-32228) (Cell Therapeutics), A-299620 (Abbott), TWEAK Inhibitor (Amgen), VEGF modulators (Johnson and Johnson), Tum-N53, tumstatin (Genzyme), Thios- 1 , Thios-2 (Thios Pharmaceuticals), MV-6401 (Miravant Medical Technologies), Spisulosine (PharmaMar), CEP-7055 (Cephalon), AUV-201 (Auvation), LM-609 (EIi Lilly), SKF-106615 (AnorMED), Oglufanide disodium (Cytran), BW-114 (Pharminox), Calreticulin (NIH), WX-678 (Wilex), SD-7784 (Pfizer), WX-UK1 (Wilex), SH-268 (Schering AG), 2-Me-PGA (Celgene), S- 137 (Pfizer), ZD-6126 (Angiogene Pharmaceuticals), SG-292 (SignalGen), Benefin (Lane Labs), A6, A36 (Angstrom), SB-2723005 (GlaxoSmithKline), SC-7 (Cell Therapeutics), ZEN-014 (AEtema Zentaris), 2-methoxyestradiol (EntreMed), NK-130119 (Nippon Kayaku), CC-10004 (Celgene), AVE- 8062A (Ajinomoto), Tacedinaline (Pfizer), Actinonin (Tokyo Metropolitan Institute of Medical Science), Lenalidomide (Celgene), VGA-1155, BTO-956 (SRI International), ER-68203-00 (Eisai), CT-6685 (UCB), JKC-362 (Phoenix Pharmaceuticals), DMI-3798 (DMI Biosciences, Angiomate (Ipsen), ZD-6474 (AstraZeneca), CEP-5214 (Cephalon), Canstatin (Genzyme), NM-3 (Mercian), Oridigm (MediQuest Therapeutics), Exherin (Adherex), BLS-0597 (Boston Life Sciences), PTC-299 (PTC Therapeutics), NPI-2358 (Nereus Pharmaceuticals), CGP-79787 (Novartis), AEE-788 (Novartis), CKD-732 (Chong Kun Dang), CP-564959 (OSl Pharmaceuticals), CM-101 (CarboMed), CT-2584, CT3501 (Cell Therapeutics), combretastatin and analogues and derivatives thereof (Oxigene), Rebimastat (Bristol-Meyers Squibb), Dextrin 2-sulfate (ML Laboratories), Cilengitide (Merk KGaA), NSC-706704 (Pharminox), KRN- 951 (Kirin Brewery), Ukrain, NSC-631570 (Nowicky Pharma), Tecogalan sodium (Daiichi Pharmaceutical), Tz-93 (Tsumura), TBC-1635 (Encysive Pharmaceuticals), TAN-1120 (Takeda), Semaxanib (Pfizer), BDI-7800 (Biopharmacopae), SD-186, SD-983 (Bristol-Meyers Squibb), SB-223245 (GlaxoSmithKline), SC-236 (Pfizer), RWJ-590973 (Johnson and Johnson), ILX-1850 (Genzyme), SC-68488, S-836 (Pfizer), CG-55069-11 (CuraGen), Ki-23057 (Kirin Brewery), CCX-700 (Chemoentryx), Pegaptanib octasodium (Giled Sciences), ANGIOCOL (available from Biostratum Inc.), or an analogue or derivative thereof.
20317. The method of item 20310 wherein the anti- fibrotic agent is an apoptosis antagonist.
20318. The method of item 20310 wherein the anti- fibrotic agent is an apoptosis activator.
20319. The method of item 20310 wherein the anti- fibrotic agent is a beta 1 integrin antagonist.
20320. The method of item 20310 wherein the anti- fibrotic agent is a beta tubulin inhibitor.
20321. The method of item 20310 wherein the anti- fibrotic agent is a blocker of enzyme production in Hepatitis C.
20322. The method of item 20310 wherein the anti- fibrotic agent is a Bruton's tyrosine kinase inhibitor. 20323. The method of item 20310 wherein the anti- fibrotic agent is a calcineυrin inhibitor.
20324. The method of item 20310 wherein the anti- fibrotic agent is a caspase 3 inhibitor.
20325. The method of item 20310 wherein the anti- fibrotic agent is a CC chemokine receptor antagonist.
20326. The method of item 20310 wherein the anti- fibrotic agent is a cell cycle inhibitor selected from the group consisting of SNS-595 (Sunesis), synthadotin, KRX-0403, and an analogue or derivative thereof.
20327. The method of item 20310 wherein the anti- fibrotic agent is a cathepsin B inhibitor.
20328. The method of item 20310 wherein the anti- fibrotic agent is a cathepsin K inhibitor, wherein the cathepsin K inhibitor is 462795 (GlaxoSmithKline), INPL-022-D6 (Amura Therapeutics), or an analogue or derivative thereof.
20329. The method of item 20310 wherein the anti- fibrotic agent is a cathepsin L inhibitor.
20330. The method of item 20310 wherein the anti- fibrotic agent is a CD40 antagonist.
20331. The method of item 20310 wherein the anti- fibrotic agent is a chemokine receptor agonist. 20332. The method of item 20310 wherein the anti- fibrotic agent is a chymase inhibitor.
20333. The method of item 20310 wherein the anti- fibrotic agent is a collagenase antagonist.
20334. The method of item 20310 wherein the anti- fibrotic agent is a CXCR antagonist.
20335. The method of item 20310 wherein the anti- fibrotic agent is a cyclin dependent kinase inhibitor selected from the group consisting of a CDK-1 inhibitor, a CDK-2 inhibitor, a CDK- 4 inhibitor, a CDK-6 inhibitor, a CAK1 inhibitor from GPC Biotech and Bristol-Myers Squibb, RGB-286199 (GPC Biotech), an anticancer agent from Astex Technology, a CAK1 inhibitor from GPC Biotech, a CDK inhibitor from Sanofi-Aventis, a CDK1/CDK2 inhibitor from Amgen, a CDK2 inhibitor from SUGEN-2 (Pfizer), a hearing loss therapy agent (Sound Pharmaceuticals), PD-0332991 (Pfizer), RGB-286199 (GPC Biotech), Ro-0505124 (Hoffmann- La Roche), a Ser/Thr kinase inhibitor from Lilly (EIi Lilly), CVT-2584 (CAS No. 199986-75-9) (CV Therapeutics), and an analogue or derivative thereof.
20336. The method of item 20310 wherein the anti- fibrotic agent is a cyclooxygenase 1 inhibitor.
20337. The method of item 20310 wherein the anti- fibrotic agent is a DHFR inhibitor.
20338. The method of item 20310 wherein the anti- fibrotic agent is a dual integrin inhibitor. 20339. The method of item 20310 wherein the anti- fibrotic agent is an elastase inhibitor.
20340. The method of item 20310 wherein the anti- fibrotic agent is an elongation factor-1 alpha inhibitor.
20341. The method of item 20310 wherein the anti- fibrotic agent is an endothelial growth factor antagonist.
20342. The method of item 20310 wherein the anti- fibrotic agent is an endothelial growth factor receptor kinase inhibitor selected from the group consisting of sorafenib tosylate (Bayer), AAL-993 (Novartis), ABP-309 (Novartis), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EXEL- 2880 (Exelixis), GW-654652 (GlaxoSmithKline), a KDR inhibitor from LG Life Sciences, CT-6685 and CT-6729 (UCB), KRN-633 and KRN-951 (Kirin Brewery), OSI-930 (OSI Pharmaceuticals), SP-5.2 (Supratek Pharma), SU-
11657 (Pfizer), a Tie-2 antagonist (Hybrigenics), a VEGFR-2 kinase inhibitor (Bristol-Myers Squibb), XL-647 (Exelixis), a KDR inhibitor from Abbott Laboratories, sorafenib tosylate, and an analogue or derivative thereof.
20343. The method of item 20310 wherein the anti- fibrotic agent is an endotoxin antagonist.
20344. The method of item 20310 wherein the anti- fibrotic agent is an epothilone and tubulin binder.
20345. The method of item 20310 wherein the anti- fibrotic agent is an estrogen receptor antagonist. 20346. The method of item 20310 wherein the anti- fibrotic agent is an FGF inhibitor.
20347. The method of item 20310 wherein the anti- fibrotic agent is a famexyl transferase inhibitor.
20348. The method of item 20310 wherein the anti- fibrotic agent is farnesyltransferase inhibitor selected from the group of A- 197574 (Abbott), a farnesyltransferase inhibitor from Servier, FPTIII (Strathclyde Institute for Drug R), LB-42908 (LG Life Sciences), Pharmaprojects No. 5063 (Genzyme), Pharmaprojects No. 5597 (Ipsen), Yissum Project No. B-1055 (Yissum), and an analogue or derivative thereof.
20349. The method of item 20310 wherein the anti- fibrotic agent is an FLT-3 kinase inhibitor.
20350. The method of item 20310 wherein the anti- fibrotic agent is an FGF receptor kinase inhibitor.
20351. The method of item 20310 wherein the anti- fibrotic agent is a fibrinogen antagonist selected from the group consisting of AUV-201 (Auvation), MG-13926 (Sanofi-Aventis), plasminogen activator (CAS No. 105913-11-9) (from Sanofi-Aventis or UCB), plasminogen activator-2 (tPA-2) (Sanofi-Aventis), pro-urokinase (CAS No. 82657-92-9) (Sanofi-Aventis), and an analogue or derivative thereof.
20352. The method of item 20310 wherein the anti- fibrotic agent is a heat shock protein 90 antagonist selected from the group consisting of SRN-005 (Sirenade), geldanamycin, NSC-33050 (17- allylaminogeldanamycin; 17-AAG), 17-dimethylaminoethylamino-17- demethoxy-geldanamycin (17-DMAG), rifabutin (rifamycin XIV, 1',4- didehydro-1-deoxy-1,4-dihydro-5'-(2-methylpropyl)-1-oxo-), and an analogue or derivative thereof.
20353. The method of item 20310 wherein the anti- fibrotic agent is a histone deacetylase inhibitor.
20354. The method of item 20310 wherein the anti- fibrotic agent is an HMGCoA reductase inhibitor selected from the group consisting of an atherosclerosis therapeutic from Lipid Sciences, ATM 6000 (ARYx Therapeutics), KS-01-019 (Kos Pharmaceuticals), Pharmaprojects No. 2197 (Sanofi-Aventi), RP 61969 (Sanofi-Aventis), and an analogue or derivative thereof.
20355. The method of item 20310 wherein the anti- fibrotic agent is an ICAM inhibitor.
20356. The method of item 20310 wherein the anti- fibrotic agent is an IL, ICE and IRAK antagonist, wherein the antagonist is a CJ-14877, CP-424174 (Pfizer), NF-61 (Negma-Lerads), and an analogue or derivative thereof.
20357. The method of item 20310 wherein the anti- fibrotic agent is an IL-2 inhibitor.
20358. The method of item 20310 wherein the anti- fibrotic agent is an immunosuppressant selected from the group consisting of teriflunomide (Sanofi Aventis), chlorsulfaquinoxalone (NSC-339004), chlorsulfaquinoxalone sulfate, CS-712 (Sankyo), ismomultin alfa (CAS No. 457913-93-8) (Akzo Nobel), antiallergics from GenPat77, antiinflammatories or AT-005 (Androclus Therapeutics), autoimmune disease therapeutics from EpiVax, BN-007 (Bone), budesonide (CAS No. 51333-22- 3) (MAP Pharmaceuticals), CO-14 (Genzyme), edratide (CAS No. 433922- 67-9) (Teva), EP-314 (Enanta), eprovafen (CAS No. 101335-99-3) (Sanofi- Aventis), HWA-131 (CAS No. 118788-41-3) (Sanofi-Aventis), immunomodulators from MerLion Pharmaceuticals, immunosuppressives from Alchemia, IPL-12 (Inflazyme), MDL-9563 (CAS No. 27086-86-8) (Sanofi-Aventis), Pharmaprojects No. 2330 (Sanofi-Aventis), Pharmaprojects No. 6426 (Abgenix), PXS-25 (Pharmaxis), rosmarinic acid (CAS No. 20283-92-5) (Sanofi-Aventis), RP 42927 or RP 54745 (CAS No. 135330-08-4) (Sanofi-Aventis), SGN-35 (Seattle Genetics), ST-1959 (Sigma-Tau), type I diabetes therapy from SYNX Pharma, UNIL-88 (Debiopharm), VP-025 (Vasogen), VR-694 (Vectura), PRTX-001 (Protalex), and an analogue or derivative thereof.
20359. The method of item 20310 wherein the anti- fibrotic agent is an IMPDH (inosine monophosphate).
20360. The method of item 20310 wherein the anti- fibrotic agent is an integrin antagonist.
20361. The method of item 20310 wherein the anti- fibrotic agent is an interleukin antagonist.
20362. The method of item 20310 wherein the anti- fibrotic agent is an inhibitor of type III receptor tyrosine kinase.
20363. The method of item 20310 wherein the anti- fibrotic agent is an irreversible inhibitor of enzyme methionine aminopeptidase type 2.
20364. The method of item 20310 wherein the anti- fibrotic agent is an isozyme selective delta protein kinase C inhibitor. 20365. The method of item 20310 wherein the anti- fibrotic agent a JAK3 enzyme inhibitor.
20366. The method of item 20310 wherein the anti- fibrotic agent is a JNK inhibitor.
20367. The method of item 20310 wherein the anti- fibrotic agent is a kinase inhibitor.
20368. The method of item 20310 wherein the anti- fibrotic agent is kinesin antagonist.
20369. The method of item 20310 wherein the anti- fibrotic agent is a kinesin antagonist.
20370. The method of item 20310 wherein the anti- fibrotic agent is a leukotriene inhibitor and antagonist selected from the group consisting of ambicromil (CAS No. 58805-38-2) (Sanofi-Aventis), amelubant (CAS No. 346735-24-8) (Boehringer Ingelheim), DW-1141 (Dong Wha), ebselen (Daiichi Pharmaceutical), ibudilast (Kyorin), leucotriene inhibitors from Sanofi-Aventis, lymphotoxin-beta receptor (LT-D) from Biogen Idee, Pharmaprojects No. 1535 and 2728 (CAS No. 119340-33-9) (Sanofi-Aventis), R-112 (Rigel), Rev-5367 (CAS No. 92532-05-3) (Sanofi- Aventis), RG-14893 (CAS No. 141835-49-6) (Sanofi-Aventis), RG-5901-A (CAS No. 101910-24-1), 92532-23-5, RP 66153 (CAS No. 142422-79-5), RP 66364 (CAS No.186916-92-5), RP 69698 (CAS No. 141748-00-7) (Sanofi-Aventis), SC-411930 (Pfizer), SC-41930 (CAS No. 120072-59-5) (Pfizer), SC-50605 (CAS No. 138828-39-4) (Pfizer), SC-51146 (CAS No. 141059-52-1), SC-53228 (CAS No. 153633-01-3) (Pfizer), spaglumic acid (ZY-15106) (CAS No.3106-85-2), 80619-64-3 (Novartis), tipredane (CAS No. 85197-77-9) (Bristol-Myers Squibb), U-75302 (CAS No. 119477-85-9) (Pfizer), and analogue or derivative thereof.
20371. The method of item 20310 wherein the anti- fibrotic agent is an MAP kinase inhibitor.
20372. The method of item 20310 wherein the anti- fibrotic agent is a matrix metalloproteinase inhibitor.
20373. The method of item 20310 wherein the anti- fibrotic agent is an MCP-CCR2 inhibitor.
20374. The method of item 20310 wherein the anti- fibrotic agent is an mTOR inhibitor.
20375. The method of item 20310 wherein the anti- fibrotic agent is an mTOR kinase inhibitor.
20376. The method of item 20310 wherein the anti- fibrotic agent is a microtubule inhibitor selected from the group consisting of antibody-maytansinoid conjugates from Biogen Idee, colchicines (MantiCore Pharmaceuticals), anticancer immunoconjugates from Johnson & Johnson, DIME from Octamer, gni-1f (GNI), huC242-DM4, huMy9-6-DM1 (ImmunoGen), IDN-5404 (Indena), IMO-098, IMOderm (Imotep), mebendazole (Introgen Therapeutics), microtubule poisons from Cambridge Enterprise, paclitaxel such as LOTAX from Aphios (CAS No. 33069-62-4), Genexol-PM from Samyang, Pharmaprojects No. 6383 (Tapestry Pharmaceuticals), RPR-112378 (Sanofi-Aventis), SGN-75 (Seattle Genetics), SPL-7435 (Starpharma), SSR-250411 (Sanofi-Aventis), trastuzumab-DM1 (Genentech), vinorelbine, and an analogue or derivative thereof. 20377. The method of item 20310 wherein the anti- fibrotic agent is an MIF inhibitor.
20378. The method of item 20310 wherein the anti- fibrotic agent is an MMP inhibitor.
20379. The method of item 20310 wherein the anti- fibrotic agent is a neurokinin (NK) antagonist selected from the group consisting of anthrotainin (CAS No. 148084-40-6) (Sanofi-Aventis), an IBS thereapeutic from ArQuIe1 MDL-105212A (CAS No. 167261-60-1) (Ssanofi- Aventis), Pharmaprojects No. 2744, 3258 (CAS No. 139167-47-8) 4006, 4201, or 5986 (Sanofi-Aventis), RP 67580 (CAS No. 135911-02-3), SR- 144190 (CAS No. 201152-86-5), SSR-240600, SSR-241586 (Sanofi- Aventis), TKA-457 (Novartis), vestipitant mesylate (CAS No. 334476-64-1) (GlaxoSmithKline), Win-64821 (Sanofi-Aventis), PRX-96026 (Predix Pharmaceuticals), and an analogue or derivative thereof.
20380. The method of item 20310 wherein the anti- fibrotic agent is an NF kappa B inhibitor selected from the group consisting of AVE-0545 or AVE-0547 (Sanofi-Aventis), bortezomib (CAS No. 179324- 69-7) (Millennium Pharmaceuticals), dexanabinol (CAS No. 112924-45-5) (Pharmos), dexlipotam (Viatris), Pharmaprojects No. 6283 (INDRA) (OXiGENE), IPL-576092 (CAS No. 137571-30-3) (Inflazyme), NFKB decoy (Corgentech), NFKB decoy oligo (AnGes MG), NFKB's from Ariad, osteoporosis treatments or S5 (F005) from Fulcrum Pharmaceuticals, P61 (Phytopharm), R-flurbiprofen (CAS No. 5104-49-4) (Encore Pharmaceuticals), and an analogue or derivative thereof.
20381. The method of item 20310 wherein the anti- fibrotic agent is a nitric oxide agonist. 20382. The method of item 20310 wherein the anti- fibrotic agent is an ornithine decarboxylase inhibitor.
20383. The method of item 20310 wherein the anti- fibrotic agent is a p38 MAP kinase inhibitor selected from the group consisting of AZD-6703 (AstraZeneca), JX-401 (Jexys Pharmaceuticals), BMS-2 (Bristol-Myers Squibb), a p38 MAP kinase inhibitor from Novartis, a p38-alpha MAP kinase inhibitor from Amphora, Pharmaprojects No. 5704 (Pharmacopeia), RPR-200765A (Sanofi-Aventis), SD-282 (Johnson & Johnson), TAK-715 (Takeda), and an analogue or derivative thereof.
20384. The method of item 20310 wherein the anti- fibrotic agent is a palmitoyl-protein thioesterase inhibitor.
20385. The method of item 20310 wherein the anti- fibrotic agent is a PDGF receptor kinase inhibitor selected from the group consisting of AAL-993, AMN-107, or ABP-309 (Novartis), AMG-706 (Amgen), BAY-57-9352 (Bayer), CDP-860 (UCB), E-7080 (Eisai), imatinib (CAS No. 152459-95-5) (Novartis), OSI-930 (OSI Pharmaceuticals), RPR- 127963E (Sanofi-Aventis), RWJ-540973 (Johnson & Johnson), sorafenib tosylate (Bayer), SU-11657 (Pfizer), tandutinib (CAS No. 387867-13-2) (Millennium Pharmaceuticals), vatalanib (Novartis), ZK-CDK (Schering AG), and an analogue or derivative thereof.
20386. The method of item 20310 wherein the anti- fibrotic agent is (-)-halofenate (Metabolex), AMG-131 (Amgen), antidiabetics from Japan Tobacco, AZD-4619, AZD-8450, AZD-8677 (AstraZeneca), DRF-10945, balaglitazone (Dr Reddy's), CS-00088, CS-00098 (Chipscreen Biosciences), E-3030 (Eisai), etalocib (CAS No. 161172-51-6) (EIi Lilly), GSK-641597 (Ligand), GSK-677954 (GlaxoSmithKline), GW-409544 (Ligand), GW-590735 (GlaxoSmithKline), K-111 (Hoffmann-La Roche), LY- 518674 (EIi Lilly), LY-674 (Ligand), LY-929 (Ligand), MC-3001, MC-3002 (MaxoCore Pharmaceuticals), metformin HCI + pioglitazone (CAS No. 1115- 70-4 and 112529-15-4), ACTOPLUS MET from Andrx), muraglitazar (CAS No. 331741-94-7) (Bristol-Myers Squibb), naveglitazar (Ligand), oleoylethanolamide (Kadmus Pharmaceuticals), ONO-5129, pioglitazone hydrochloride (CAS No. 111025-46-8 and 112529-15-4) (Takeda), PLX-204 (Plexxikon), PPAR agonists from Genfit, PPAR delta agonists from EIi Lilly, PPAR-alpha agonists from CrystalGenomics, PPAR-gamma modulators and PPAR-β modulators from CareX, rosiglitazone maleate (CAS No. 122324- 73-4 or 155141-29-0) (GlaxoSmithKline), rosiglitazone maleate/glimepir (CAS No. 155141-29-0 and 93479-97-1), AVANDARYL, rosiglitazone maleate/metformin extend (CAS No. 155141-29-0 and 657-24-9), AVANDAMET, rosiglitazone maleate+metformin, AVANDAMET (GlaxoSmithKline), tesaglitazar (AstraZeneca), and an analogue or derivative thereof.
20387. The method of item 20310 wherein the anti- fibrotic agent is a phosphatase inhibitor.
20388. The method of item 20310 wherein the anti- fibrotic agent is a phosphodiesterase (PDE) inhibitor selected from the group consisting of avanafil (Tanabe Seiyaku), dasantafil (CAS No. 569351- 91-3) (Schering-Plough), A-906119 (CAS No. 134072-58-5), DL-850 (Sanofi-Aventis), GRC-3015, GRC-3566, GRC-3886 (Glenmark), HWA-153 (CAS No. 56395-66-5) (Sanofi-Aventis), hydroxypumafentrine (Altana), IBFB-130011 , IBFB-14-016, IBFB-140301 , IBFB-150007, IBFB-211913 (IBFB Pharma), L-826141 (Merck & Co), medorinone (CAS No. 88296-61-1) (Sanofi-Aventis), MEM-1917 (Memory Pharmaceuticals), ND-1251 (Neuro3d), PDE inhibitors from ICOS, PDE IV inhibitors from Memory Pharmaceuticals and CrystalGenomics, Pharmaprojects No. 2742 and 6141 (Sanofi-Aventis), QAD-171 (Novartis), RHC-2963 (CAS No. 76993-12-9 and 76993-14-1), RPR-117658, RPR-122818 derivatives, SR-24870 , and RPR- 132294 (Sanofi-Aventis), SK-350 (ln2Gen), stroke therapy agents from deCODE Genetics, TAS-203 (Taiho), tofimilast (CAS No. 185954-27-2) (Pfizer), UK-371800 (Pfizer), WIN-65579 (CAS No. 158020-82-7) (Sanofi- Aventis), IBFB-130020 (IBFB Pharma), OPC-6535 (CAS No. 145739-56-6) (Otsuka), a phosphodiesterase III inhibitor, enoximone, a phosphodiesterase IV inhibitor, fosfosal, Atopik (Barrier Therapeutics), triflusal, a phosphodiesterase V inhibitor, and an analogue or derivative thereof.
20389. The method of item 20310 wherein the anti- fibrotic agent is a PKC inhibitor.
20390. The method of item 20310 wherein the anti- fibrotic agent is a platelet activating factor antagonist.
20391. The method of item 20310 wherein the anti- fibrotic agent is a platelet-derived growth factor receptor kinase inhibitor.
20392. The method of item 20310 wherein the anti- fibrotic agent is a prolyl hydroxylase inhibitor.
20393. The method of item 20310 wherein the anti- fibrotic agent is a polymorphonuclear neutrophil inhibitor.
20394. The method of item 20310 wherein the anti- fibrotic agent is a protein kinase B inhibitor.
20395. The method of item 20310 wherein the anti- fibrotic agent is a protein kinase C stimulant. 20396. The method of item 20310 wherein the anti- fibrotic agent is a purine nucleoside analogue.
20397. The method of item 20310 wherein the anti- fibrotic agent is a purinoreceptor P2X antagonist.
20398. The method of item 20310 wherein the anti- fibrotic agent is a Raf kinase inhibitor.
20399. The method of item 20310 wherein the anti- fibrotic agent is a reversible inhibitor of ErbB1 and ErbB2.
20400. The method of item 20310 wherein the anti- fibrotic agent is a ribonucleoside triphosphate reductase inhibitor.
20401. The method of item 20310 wherein the anti- fibrotic agent is an SDF-1 antagonist.
20402. The method of item 20310 wherein the anti- fibrotic agent is a sheddase inhibitor.
20403. The method of item 20310 wherein the anti- fibrotic agent is an SRC inhibitor.
20404. The method of item 20310 wherein the anti- fibrotic agent is a stromelysin inhibitor.
20405. The method of item 20310 wherein the anti- fibrotic agent is an Syk kinase inhibitor. 20406. The method of item 20310 wherein the anti- fibrotic agent is a telomerase inhibitor.
20407. The method of item 20310 wherein the anti- fibrotic agent is a TGF beta inhibitor selected from the group consisting of pirfenidone (CAS No. 53179-13-8) (MARNAC), tranilast (CAS No. 53902- 12-8) (Kissei), IN-1130 (ln2Gen), mannose-6-phosphate (BTG), TGF-β antagonists from Inflazyme (Pharmaprojects No. 6075), TGF-β antagonists from Sydney, non-industrial source), TGF-β I receptor kinase inhibitors from EIi Lilly, TGF-β receptor inhibitors from Johnson & Johnson, and an analogue or derivative thereof.
20408. The method of item 20310 wherein the anti- fibrotic agent is a TNFα antagonist or TACE inhibitor selected from the group consisting of adalimumab (CAS No. 331731-18-1) (Cambridge Antibody Technology), AGIX-4207 (AtheroGenics), AGT-1 (Advanced Biotherapy), an anti-inflammatory from Borean Pharma, Cellzome, or Paradigm Therapeutics, anti-inflammatory vaccine (TNF-alpha kinoid) from Neovacs, humanized anti-TNF antibody or an anti-TNF MAb (CB0006) Celltech (UCB), apratastat (CAS No. 287445-51-0) (Wyeth), BMS-561392 (Bristol-Myers Squibb), BN-006 (Bone), certolizumab pegol (CAS No. 428863-50-7 or CH-138 (UCB), cilomilast (CAS No. 153259-65-5) (GlaxoSmithKline), CR-1 (Nuada Pharmaceuticals), CRx-119 (CombinatoRx), D-5410 (UCB), dacopafant (CAS No. 125372-33-0) (Sanofi- Aventis), dersalazine (CAS No. 188913-57-7/188913-58-8) (Uriach), etanercept (CAS No. 185243-69-0) (Amgen), ethyl pyruvate (Critical (Critical Therapeutics), golimumab (CAS No. 476181-74-5) (Johnson & Johnson), hormono-immunotherapy from Ipsen, CDP571 (e.g., Humicade from UCB), IC-485 (ICOS), infliximab (CAS No. 170277-31-3) (Johnson & Johnson), IP- 751 (Manhattan Pharmaceuticals), ISIS-104838 (CAS No. 250755-32-9) (ISIS Pharmaceuticals), lenalidomide (CAS No. 191732-72-6) (Celgene), lentinan (CAS No. 37339-90-5) (Ajinomoto), MDL-201112 (CAS No. 142130-73-2) (Sanofi-Aventis), medroxyprogesterone (CAS No. 520-85-4) (InKine Pharmaceutical), N-acetylcysteine (CAS No. 616-91-1) (Zambon), NBE-P2 (DIREVO Biotech), nerelimomab (CAS No. 162774-06-3) (Chiron), OM-294DP (OM PHARMA)1 onercept (CAS No. 199685-57-9) (Yeda), PASSTNF-alpha (Verigen), pentoxifylline or oxypentifylline (Sanofi-Aventis), Pharmaprojects No. 4091 , 4241 , 4295, or 4488 (Sanofi-Aventis), Pharmaprojects No. 5480 (Amgen), Pharmaprojects No. 6749 (Cengent), pirfenidone (CAS No. 53179-13-8) (MARNAC), RPR-132294 (Sanofi- Aventis), S5 (F002) (Fulcrum Pharmaceuticals), simvastatin (CAS No. 79902-63-9) (Merck & Co), STA-6292 (Synta Pharmaceuticals), tacrolimus (CAS No. 104987-11-3) (Fujisawa LifeCycle Pharma), talactoferrin alfa (CAS No. 308240-58-6) (Agennix), thalidomide (CAS No. 50-35-1) (Celgene), TNF antagonists form ProStrakan, and Synergen, TNF inhibitors (Amgen), TNF-alpha antagonists from Dynavax Technologies and Jerina AG (Germany), TNF-alpha inhibitors from IBFB Pharma and Xencor (Xencor), torbafylline (CAS No. 105102-21-4) (Sanofi-Aventis), UR-1505 (Uriach), VT-346 (Viron Therapeutics), YSIL6 (Y's Therapeutics), YSTH2 (Y's Therapeutics), NPI-1302a-3 (Nereus Pharmaceuticals, a TNF antagonist from Jerina AG (Germany), dersalazine, and an analogue or derivative thereof.
20409. The method of item 20310 wherein the anti- fibrotic agent is a Toll receptor inhibitor.
20410. The method of item 20310 wherein the anti- fibrotic agent is a tubulin antagonist.
20411. The method of item 20310 wherein the anti- fibrotic agent is a tyrosine kinase inhibitor selected from the group consisting of SU-011248, SUTENT from Pfizer Inc. (New York, NY), BMS-354825, PN- 355 (Paracelsian Pharmaceuticals), AGN-199659 (Allergan), AAL-993 or ABP-309 (Novartis), adaphostin (NIH), AEE-788 (Novartis), AG-013736 (OSI Pharmaceuticals), AG-13736 (Pfizer), ALT-110 (Alteris Therapeutics), AMG-706 (Amgen), anticancer MAbs from Xencor, anti-EGFrvlll MAbs from Abgenix, anti-HER2 MAb from Abiogen, AZD-2171 or AZD-9935 (AstraZeneca), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), CEP-5214 (Cephalon), CEP-7055 (Cephalon), cetuximab (ImClone Systems), CHIR-200131 and CHIR-258 (Chiron), CP-547632 (OSI Pharmaceuticals), CP-724714 (Pfizer), CT-301 (Creabilis Therapeutics), D- 69491 (Baxter International), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EGFR/ErbB2 inhibitors from Array BioPharma, erlotinib (CAS No. 183319- 69-9) (OSI Pharmaceuticals), EXEL-2880 (Exelixis), FK-778 (Sanofi- Aventis) , gefitinib (CAS No. 184475-35-2) (AstraZeneca), GW-2286 or GW- 654652 (GlaxoSmithKline), her2/neu antigen from AlphaVax, HER-2/neu inhibitor from Generex, Herzyme (Medipad) (Sirna Therapeutics), HKI-272 (Wyeth), HuMax-EGFr (Genmab), idronoxil (CAS No. 81267-65-4) (Novogen), IGF-1 inhibitors from Ontogen, IMC-11F8 (ImClone Systems), kahalalide F (CAS No. 149204-42-2) (PharmaMar), KDR inhibitor from LG Life Sciences, KDR inhibitors from Abbott Laboratories, KDR kinase inhibitors (UCB), Kdr kinase inhibitors from Merck & Co, KRN-633 and KRN- 951 (Kirin Brewery), KSB-102 (Xenova), lapatinib ditosylate (CAS No. 388082-78-8) (GlaxoSmithKline), matuzumab (Merck KGaA), MDX-214 (Medarex), ME-103 (Pharmexa), MED-A300 (Gerolymatos), MNAC-13 (Lay Line Genomics), nimotuzumab (Center of Molecular Immunology), NSC- 330507 or NSC-707545 (NIH), NV-50 (Novogen), OSI-930 (OSI Pharmaceuticals), panitumumab (Abgenix), pelitinib (CAS No. 287933-82-7) (Wyeth), pertuzumab (CAS No. 380610-27-5) (Genentech), Pharmaprojects No. 3985 (Sanofi-Aventis), prostate cancer therapeutics from Sequenom (SQPC35, SQPC36, SQPC90), removab and remoxab (Trion Pharma), RG- 13022 (CAS No. 136831-48-6), RG-13291 (CAS No. 138989-50-1), or RG- 14620 (CAS No. 136831-49-7) (Sanofi-Aventis), RM-6427 (Romark), RNAi breast cancer therapy from Benitec, RP 53801 (CAS No. 125882-88-4) (Sanofi-Aventis), sorafenib tosylate (Bayer), SU-11657 (Pfizer), Tie-2 antagonists from Semaia (Hybrigenics), Tie-2 inhibitors from Ontogen, trastuzumab (CAS No. 180288-69-1) (Genentech), tyrosine kinase inhibitors from Sanofi-Aventis, U3-1287, U3-1565, U3-1784, U3-1800 (U3 Pharma), vatalanib (Novartis), VEGFR-2 kinase inhibitor from Bristol-Myers Squibb, XL-647 (Exelixis), ZD-6474 (AstraZeneca), ZK-CDK (Schering AG), an EGFR tyrosine kinase inhibitor, EKB-569 (Wyeth), and an analogue or derivative thereof.
20412. The method of item 20310 wherein the anti- fibrotic agent is a VEGF inhibitor.
20413. The method of item 20310 wherein the anti- fibrotic agent is a vitamin D receptor agonist.
20414. The method of item 20310 wherein the anti- fibrotic agent is ZD-6474 (an angiogenesis inhibitor).
20415. The method of item 20310 wherein the anti- fibrotic agent is AP-23573 (an mTOR inhibitor).
20416. The method of item 20310 wherein the anti- fibrotic agent is synthadotin (a tubulin antagonist).
20417. The method of item 20310 wherein the anti- fibrotic agent is S-0885 (a collagenase inhibitor).
20418. The method of item 20310 wherein the anti- fibrotic agent is aplidine (an elongation factor-1 alpha inhibitor). 20419. The method of item 20310 wherein the anti- fibrotic agent is ixabepilone (an epithilone).
20420. The method of item 20310 wherein the anti- fibrotic agent is IDN-5390 (an angiogenesis inhibitor).
20421. The method of item 20310 wherein the anti- fibrotic agent is SB-2723005 (an angiogenesis inhibitor).
20422. The method of item 20310 wherein the anti- fibrotic agent is ABT-518 (an angiogenesis inhibitor).
20423. The method of item 20310 wherein the anti- fibrotic agent is combretastatin (an angiogenesis inhibitor).
20424. The method of item 20310 wherein the anti- fibrotic agent is anecortave acetate (an angiogenesis inhibitor).
20425. The method of item 20310 wherein the anti- fibrotic agent is SB-715992 (a kinesin antagonist).
20426. The method of item 20310 wherein the anti- fibrotic agent is temsirolimus (an mTOR inhibitor).
20427. The method of item 20310 wherein the anti- fibrotic agent is adalimumab (a TNFα antagonist).
20428. The method of item 20310 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with an anti-fibrotic agent, and (b) implanting the medical device into the host. 20429. The method of item 20310 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with an anti-infective agent, and (b) implanting the medical device into the host.
20430. The method of item 20310 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with a polymer; and (b) implanting the medical device into the host.
20431. The method of item 20310 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with a composition comprising an anti-fibrotic agent and a polymer, and (b) implanting the medical device into the host.
20432. The method of item 20310 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with a composition comprising an anti-infective agent and a polymer, and (b) implanting the medical device into the host.
20433. The method of item 20310 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with a composition comprising an anti-fibrotic agent, an anti- infective agent and a polymer, and (b) implanting the medical device into the host.
20434. The method of item 20310 wherein the anti- infective agent is an anthracycline.
20435. The method of item 20310 wherein the anti- infective agent is doxorubicin. 20436. The method of item 20310 wherein the anti- infective agent is is mitoxantrone.
20437. The method of item 20310 wherein the anti- infective agent is a fluoropyrimidine.
20438. The method of item 20310 wherein the anti- infective agent is 5-fluorouracil (5-FU).
20439. The method of item 20310 wherein the anti- infective agent is a folic acid antagonist.
20440. The method of item 20310 wherein the anti- infective agent is methotrexate.
20441. The method of item 20310 wherein the anti- infective agent is a podophylotoxin.
20442. The method of item 20310 wherein the anti- infective agent is etoposide.
20443. The method of item 20310 wherein the anti- infective agent is camptothecin.
20444. The method of item 20310 wherein the anti- infective agent is a hydroxyurea.
20445. The method of item 20310 wherein the anti- infective agent is a platinum complex. 20446. The method of item 20310 wherein the anti- infective agent is cisplatin.
20447. The method of item 20310 wherein the composition comprises an anti-thrombotic agent.
20448. The method of item 20310 wherein the polymer is formed from reactants comprising a naturally occurring polymer.
20449. The method of item 20310 wherein the polymer is formed from reactants comprising protein.
20450. The method of item 20310 wherein the polymer is formed from reactants comprising carbohydrate.
20451. The method of item 20310 wherein the polymer is formed from reactants comprising biodegradable polymer.
20452. The method of item 20310 wherein the polymer is formed from reactants comprising nonbiodegradable polymer.
20453. The method of item 20310 wherein the polymer is formed from reactants comprising collagen.
20454. The method of item 20310 wherein the polymer is formed from reactants comprising methylated collagen.
20455. The method of item 20310 wherein the polymer is formed from reactants comprising fibrinogen. 20456. The method of item 20310 wherein the polymer reactants comprising thrombin.
20457. The method of item 20310 wherein the polymer reactants comprising blood plasma.
20458. The method of item 20310 wherein the polymer reactants comprising calcium salt.
20459. The method of item 20310 wherein the polymer reactants comprising an antifibronolytic agent.
20460. The method of item 20310 wherein the polymer reactants comprising fibrinogen analog.
20461. The method of item 20310 wherein the polymer reactants comprising albumin.
20462. The method of item 20310 wherein the polymer reactants comprising plasminogen.
20463. The method of item 20310 wherein the polymer reactants comprising von Willebrands factor.
20464. The method of item 20310 wherein the polymer reactants comprising Factor VIII.
20465. The method of item 20310 wherein the polymer reactants comprising hypoallergenic collagen. 20466. The method of item 20310 wherein the polymer is formed from reactants comprising atelopeptidic collagen.
20467. The method of item 20310 wherein the polymer is formed from reactants comprising telopeptide collagen.
20468. The method of item 20310 wherein the polymer is formed from reactants comprising crosslinked collagen.
20469. The method of item 20310 wherein the polymer is formed from reactants comprising aprotinin.
20470. The method of item 20310 wherein the polymer is formed from reactants comprising epsilon-amino-n-caproic acid.
20471. The method of item 20310 wherein the polymer is formed from reactants comprising gelatin.
20472. The method of item 20310 wherein the polymer is formed from reactants comprising protein conjugates.
20473. The method of item 20310 wherein the polymer is formed from reactants comprising gelatin conjugates.
20474. The method of item 20310 wherein the polymer is formed from reactants comprising a synthetic polymer.
20475. The method of item 20310 wherein the polymer is formed from reactants comprising a synthetic isocyanate-containing compound. 20476. The method of item 20310 wherein the polymer is formed from reactants comprising a synthetic thiol-containing compound.
20477. The method of item 20310 wherein the polymer is formed from reactants comprising a synthetic compound containing at least two thiol groups.
20478. The method of item 20310 wherein the polymer is formed from reactants comprising a synthetic compound containing at least three thiol groups.
20479. Tr^e method of item 20310 wherein the polymer is formed from reactants comprising a synthetic compound containing at least four thiol groups.
20480. The method of item 20310 wherein the polymer is formed from reactants comprising a synthetic amino-containing compound.
20481. The method of item 20310 wherein the polymer is formed from reactants comprising a synthetic compound containing at least two amino groups.
20482. The method of item 20310 wherein the polymer is formed from reactants comprising a synthetic compound containing at least three amino groups.
20483. The method of item 20310 wherein the polymer is formed from reactants comprising a synthetic compound containing at least four amino groups. 20484. The method of item 20310 wherein the polymer is formed from reactants comprising a synthetic compound comprising a carbonyl-oxygen-succinimidyl group.
20485. The method of item 20310 wherein the polymer is formed from reactants comprising a synthetic compound comprising at least two carbonyl-oxygen-succinimidyl groups.
20486. The method of item 20310 wherein the polymer is formed from reactants comprising a synthetic compound comprising at least three carbonyl-oxygen-succinimidyl groups.
20487. The method of item 20310 wherein the polymer is formed from reactants comprising a synthetic compound comprising at least four carbonyl-oxygen-succinimidyl groups.
20488. The method of item 20310 wherein the polymer is formed from reactants comprising a synthetic polyalkylene oxide- containing compound.
20489. The method of item 20310 wherein the polymer is formed from reactants comprising a synthetic compound comprising both polyalkylene oxide and biodegradable polyester blocks.
20490. The method of item 20310 wherein the polymer is formed from reactants comprising a synthetic polyalkylene oxide- containing compound having reactive amino groups.
20491. The method of item 20310 wherein the polymer is formed from reactants comprising a synthetic polyalkylene oxide- containing compound having reactive thiol groups. 20492. The method of item 20310 wherein the polymer is formed from reactants comprising a synthetic polyalkylene oxide- containing compound having reactive carbonyl-oxygen-succinimidyl groups.
20493. The method of item 20310 wherein the polymer is formed from reactants comprising a synthetic compound comprising a biodegradable polyester block.
20494. The method of item 20310 wherein the polymer is formed from reactants comprising a synthetic polymer formed in whole or part from lactic acid or lactide.
20495. The method of item 20310 wherein the polymer is formed from reactants comprising a synthetic polymer formed in whole or part from glycolic acid or glycolide.
20496. The method of item 20310 wherein the polymer is formed from reactants comprising polylysine.
20497. The method of item 20310 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound comprising a polyalkylene oxide portion.
20498. The method of item 20310 wherein the polymer is formed from reactants comprising (a) protein and (b) polylysine.
20499. The method of item 20310 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound having at least four thiol groups. 20500. The method of item 20310 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound having at least four amino groups.
20501. The method of item 20310 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound having at least four carbonyl-oxygen-succinimide groups.
20502. The method of item 20310 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound having a biodegradable region formed from reactants selected from lactic acid, lactide, glycolic acid, glycolide, and epison-caprolactone.
20503. The method of item 20310 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound comprising a polyalkylene oxide portion.
20504. The method of item 20310 wherein the polymer is formed from reactants comprising (a) collagen and (b) polylysine.
20505. The method of item 20310 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound having at least four thiol groups.
20506. The method of item 20310 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound having at least four amino groups.
20507. The method of item 20310 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound having at least four carbonyl-oxygen-succinimide groups. 20508. The method of item 20310 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound having a biodegradable region formed from reactants selected from lactic acid, lactide, glycolic acid, glycolide, and epison-caprolactone.
20509. The method of item 20310 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound comprising a polyalkylene oxide portion.
20510. The method of item 20310 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) polylysine.
20511. The method of item 20310 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound having at least four thiol groups.
20512. The method of item 20310 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound having at least four amino groups.
20513. The method of item 20310 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound having at least four carbonyl-oxygen-succinimide groups.
20514. The method of item 20310 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound having a biodegradable region formed from reactants selected from lactic acid, lactide, glycolic acid, glycolide, and epison-caprolactone. 20515. The method of item 20310 wherein the polymer is formed from reactants comprising hyaluronic acid.
20516. The method of item 20310 wherein the polymer is formed from reactants comprising a hyaluronic acid derivative.
20517. The method of item 20310 wherein the polymer is formed from reactants comprising pentaerythritol poly(ethylene glycol)ether tetra-sulfhydryl of number average molecular weight between 3,000 and 30,000.
20518. The method of item 20310 wherein the polymer is formed from reactants comprising pentaerythritol poly(ethylene glycol)ether tetra-amino of number average molecular weight between 3,000 and 30,000.
20519. The method of item 20310 wherein the polymer is formed from reactants comprising (a) a synthetic compound having a number average molecular weight between 3,000 and 30,000 and comprising a polyalkylene oxide region and multiple nucleophilic groups, and (b) a synthetic compound having a number average molecular weight between 3,000 and 30,000 and comprising a polyalkylene oxide region and multiple electrophilic groups.
20520. The method of item 20310 wherein the composition comprises a colorant.
20521. The method of item 20310 wherein the composition is sterile. 20522. A method as in any one of items 20310-20521 , wherein the device is a mechanical prosthetic heart valve.
20523. A method as in any one of items 20310-20521 , wherein the device is a bioprosthetic heart valve.
20524. A method as in any one of items 20310-20521 , wherein the device is an implantable annular ring for receiving a prosthetic heart valve.
20525. A method as in any one of items 20310-20521 , wherein the device is a suture ring having an outer peripheral tapered thread for attaching a heart valve prosthesis.
20526. A method as in any one of items 20310-20521 , wherein the device is a suture ring for a mechanical heart valve.
20527. A method for implanting a medical device comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with i) an anti-fibrotic agent, ii) an anti-infective agent, iii) a polymer, iv) a composition comprising an anti-fibrotic agent and a polymer, v) a composition comprising an anti-infective agent and a polymer, or vi) a composition comprising an anti-fibrotic agent, an anti- infective agent and a polymer, and (b) implanting the medical device into the host, wherein the medical device is a penile implant.
20528. The method of item 20527 wherein the anti- fibrotic agent is an adensosine A2A receptor antagonist.
20529. The method of item 20527 wherein the anti- fibrotic agent is an AKT inhibitor. 20530. The method of item 20527 wherein the anti- fibrotic agent is an alpha 2 integrin antagonist, wherein the alpha 2 integrin antagonist is Pharmaprojects No. 5754 (Merck KgaA).
20531. The method of item 20527 wherein the anti- fibrotic agent is an alpha 4 integrin antagonist.
20532. The method of item 20527 wherein the anti- fibrotic agent is an alpha 7 nicotinic receptor agonist.
20533. The method of item 20527 wherein the anti- fibrotic agent is an angiogenesis inhibitor selected from the group consisting of AG-12,958 (Pfizer), ATN-161 (Attenuon LLC), neovastat, an angiogenesis inhibitor from Jerina AG (Germany), NM-3 (Mercian), VGA-1155 (Taisho), FCE-26644 (Pfizer), FCE-26950 (Pfizer), FPMA (Meiji Daries), FR-111142 (Fujisawa), GGTI-298, GM-1306 (Ligand), GPA-1734 (Novartis), NNC-47- 0011 (Novo Nordisk), herbamycin (Nippon Kayaku), lenalidomide (Celegene), IP-10 (NIH), ABT-828 (Abbott), KIN-841 (Tokushima University, Japan), SF-1126 (Semafore Pharmaceuticals), laminin technology (NIH), CHIR-258 (Chiron), NVP-AEW541 (Novartis), NVP-AEW541 (Novartis), Vt16907 (Alchemia), OXI-8007 (Oxigene), EG-3306 (Ark Therapeutics), Maspin (Arriva), ABT-567 (Abbott), PPI-2458 (Praecis Pharmaceuticals), CC-5079, CC-4089 (Celgene), HIF-1 alpha inhibitors (Xenova), S-247 (Pfizer), AP-23573 (Ariad), AZD-9935 (Astra Zeneca), mebendazole (Introgen Therapeutics), MetAP-2 inhibitors (GlaxoSmithKline), AG-615 (Angiogene Pharmaceuticals), Tie-2 antagonists (Hybrigenics), NC-381, CYC-381 , NC-169, NC-219, NC-383, NC-384, NC-407 (Lorus Therapeutics), ATN-224 (Attenuon), ON-01370 (Onconova), Vitronectin antagonists (Amgen), SDX-103 (Salmedix), Vitronectin antagonists (Shire), CHP (Riemser), TEK (Amgen), Anecortave acetate (Alcon), T46.2 (Matrix Therapeutics), HG-2 (Heptagen), TEM antagonists (Genzyme), Oxi-4500 (Oxigene), ATN-161 (Attention), WX-293 (Wilex), M-2025 (Metris Therapeutics), Alphastatin (BioActa), YH-16 (Yantai Rongchang), BIBF- 1120 (Boehringer Ingelheim), BAY-57-9352 (Bayer), AS-1404 (Cancer Research Technology), SC-77964 (Pfizer), glycomimetics (BioTie Therapies), TIE-2 Inhibitors (Ontogen), DIMI, Octamer (Octamer), ABR- 215050 (Active Biotech), ABT-518 (Abbott), KDR inhibitors (Abbott), BSF- 466895 (Abbott), SCH-221153 (Schering-Plough), DAC:antiangiogenic (ConjuChem), TFPI (EntreMed), AZD-2171 (Astra-Zenaca), CDC-394 (Celgene), LY290293 (EIi Lilly), IDN-5390 (Indena), Kdr Kinase Inhibitors (Merck), CT-113020, CT-116433, CT-116563, CT-31890, CT-32228) (Cell Therapeutics), A-299620 (Abbott), TWEAK Inhibitor (Amgen), VEGF modulators (Johnson and Johnson), Tum-N53, tumstatin (Genzyme), Thios- 1, Thios-2 (Thios Pharmaceuticals), MV-6401 (Miravant Medical Technologies), Spisulosine (PharmaMar), CEP-7055 (Cephalon), AUV-201 (Auvation), LM-609 (EIi Lilly), SKF-106615 (AnorMED), Oglufanide disodium (Cytran), BW-114 (Pharminox), Calreticulin (NIH), WX-678 (Wilex), SD-7784 (Pfizer), WX-UK1 (Wilex), SH-268 (Schering AG), 2-Me-PGA (Celgene), S- 137 (Pfizer), ZD-6126 (Angiogene Pharmaceuticals), SG-292 (SignalGen), Benefin (Lane Labs), A6, A36 (Angstrom), SB-2723005 (GlaxoSmithKline), SC-7 (Cell Therapeutics), ZEN-014 (AEterna Zentaris), 2-methoxyestradiol (EntreMed), NK-130119 (Nippon Kayaku), CC-10004 (Celgene), AVE- 8062A (Ajinomoto), Tacedinaline (Pfizer), Actinonin (Tokyo Metropolitan Institute of Medical Science), Lenalidomide (Celgene), VGA-1155, BTO-956 (SRI International), ER-68203-00 (Eisai), CT-6685 (UCB), JKC-362 (Phoenix Pharmaceuticals), DMI-3798 (DMI Biosciences, Angiomate (Ipsen), ZD-6474 (AstraZeneca), CEP-5214 (Cephalon), Canstatin (Genzyme), NM-3 (Mercian), Oridigm (MediQuest Therapeutics), Exherin (Adherex), BLS-0597 (Boston Life Sciences), PTC-299 (PTC Therapeutics), NPI-2358 (Nereus Pharmaceuticals), CGP-79787 (Novartis), AEE-788 (Novartis), CKD-732 (Chong Kun Dang), CP-564959 (OSI Pharmaceuticals), CM-101 (CarboMed), CT-2584, CT3501 (Cell Therapeutics), combretastatin and analogues and derivatives thereof (Oxigene), Rebimastat (Bristol-Meyers Squibb), Dextrin 2-sulfate (ML Laboratories), Cilengitide (Merk KGaA), NSC-706704 (Pharminox), KRN- 951 (Kirin Brewery), Ukrain, NSC-631570 (Nowicky Pharma), Tecogalan sodium (Daiichi Pharmaceutical), Tz-93 (Tsumura), TBC-1635 (Encysive Pharmaceuticals), TAN-1120 (Takeda), Semaxanib (Pfizer), BDI-7800 (Biopharmacopae), SD-186, SD-983 (Bristol-Meyers Squibb), SB-223245 (GlaxoSmithKline), SC-236 (Pfizer), RWJ-590973 (Johnson and Johnson), ILX-1850 (Genzyme), SC-68488, S-836 (Pfizer), CG-55069-11 (CuraGen), Ki-23057 (Kirin Brewery), CCX-700 (Chemoentryx), Pegaptanib octasodium (Giled Sciences), ANGIOCOL (available from Biostratum Inc.), or an analogue or derivative thereof.
20534. The method of item 20527 wherein the anti- fibrotic agent is an apoptosis antagonist.
20535. The method of item 20527 wherein the anti- fibrotic agent is an apoptosis activator.
20536. The method of item 20527 wherein the anti- fibrotic agent is a beta 1 integrin antagonist.
20537. The method of item 20527 wherein the anti- fibrotic agent is a beta tubulin inhibitor.
20538. The method of item 20527 wherein the anti- fibrotic agent is a blocker of enzyme production in Hepatitis C.
20539. The method of item 20527 wherein the anti- fibrotic agent is a Bruton's tyrosine kinase inhibitor. 20540. The method of item 20527 wherein the anti- fibrotic agent is a calcineurin inhibitor.
20541. The method of item 20527 wherein the anti- fibrotic agent is a caspase 3 inhibitor.
20542. The method of item 20527 wherein the anti- fibrotic agent is a CC chemokine receptor antagonist.
20543. The method of item 20527 wherein the anti- fibrotic agent is a cell cycle inhibitor selected from the group consisting of SNS-595 (Sunesis), synthadotin, KRX-0403, and an analogue or derivative thereof.
20544. The method of item 20527 wherein the anti- fibrotic agent is a cathepsin B inhibitor.
20545. The method of item 20527 wherein the anti- fibrotic agent is a cathepsin K inhibitor, wherein the cathepsin K inhibitor is 462795 (GlaxoSmithKline), INPL-022-D6 (Amura Therapeutics), or an analogue or derivative thereof.
20546. The method of item 20527 wherein the anti- fibrotic agent is a cathepsin L inhibitor.
20547. The method of item 20527 wherein the anti- fibrotic agent is a CD40 antagonist.
20548. The method of item 20527 wherein the anti- fibrotic agent is a chemokine receptor agonist. 20549. The method of item 20527 wherein the anti- fibrotic agent is a chymase inhibitor.
20550. The method of item 20527 wherein the anti- fibrotic agent is a collagenase antagonist.
20551. The method of item 20527 wherein the anti- fibrotic agent is a CXCR antagonist.
20552. The method of item 20527 wherein the anti- fibrotic agent is a cyclin dependent kinase inhibitor selected from the group consisting of a CDK-1 inhibitor, a CDK-2 inhibitor, a CDK- 4 inhibitor, a CDK-6 inhibitor, a CAK1 inhibitor from GPC Biotech and Bristol-Myers Squibb, RGB-286199 (GPC Biotech), an anticancer agent from Astex Technology, a CAK1 inhibitor from GPC Biotech, a CDK inhibitor from Sanofi-Aventis, a CDK1/CDK2 inhibitor from Amgen, a CDK2 inhibitor from SUGEN-2 (Pfizer), a hearing loss therapy agent (Sound Pharmaceuticals), PD-0332991 (Pfizer), RGB-286199 (GPC Biotech), Ro-0505124 (Hoffmann- La Roche), a Ser/Thr kinase inhibitor from Lilly (EIi Lilly), CVT-2584 (CAS No. 199986-75-9) (CV Therapeutics), and an analogue or derivative thereof.
20553. The method of item 20527 wherein the anti- fibrotic agent is a cyclooxygenase 1 inhibitor.
20554. The method of item 20527 wherein the anti- fibrotic agent is a DHFR inhibitor.
20555. The method of item 20527 wherein the anti- fibrotic agent is a dual integrin inhibitor. 20556. The method of item 20527 wherein the anti- fibrotic agent is an elastase inhibitor.
20557. The method of item 20527 wherein the anti- fibrotic agent is an elongation factor-1 alpha inhibitor.
20558. The method of item 20527 wherein the anti- fibrotic agent is an endothelial growth factor antagonist.
20559. The method of item 20527 wherein the anti- fibrotic agent is an endothelial growth factor receptor kinase inhibitor selected from the group consisting of sorafenib tosylate (Bayer), AAL-993 (Novartis), ABP-309 (Novartis), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EXEL- 2880 (Exelixis), GW-654652 (GlaxoSmithKline), a KDR inhibitor from LG Life Sciences, CT-6685 and CT-6729 (UCB), KRN-633 and KRN-951 (Kirin Brewery), OSI-930 (OSI Pharmaceuticals), SP-5.2 (Supratek Pharma), SU- 11657 (Pfizer), a Tie-2 antagonist (Hybrigenics), a VEGFR-2 kinase inhibitor (Bristol-Myers Squibb), XL-647 (Exelixis), a KDR inhibitor from Abbott Laboratories, sorafenib tosylate, and an analogue or derivative thereof.
20560. The method of item 20527 wherein the anti- fibrotic agent is an endotoxin antagonist.
20561. The method of item 20527 wherein the anti- fibrotic agent is an epothilone and tubulin binder.
20562. The method of item 20527 wherein the anti- fibrotic agent is an estrogen receptor antagonist. 20563. The method of item 20527 wherein the anti- fibrotic agent is an FGF inhibitor.
20564. The method of item 20527 wherein the anti- fibrotic agent is a farnexyl transferase inhibitor.
20565. The method of item 20527 wherein the anti- fibrotic agent is farnesyltransferase inhibitor selected from the group of A- 197574 (Abbott), a farnesyltransferase inhibitor from Servier, FPTIII (Strathclyde Institute for Drug R), LB-42908 (LG Life Sciences), Pharmaprojects No. 5063 (Genzyme), Pharmaprojects No. 5597 (Ipsen), Yissum Project No. B-1055 (Yissum), and an analogue or derivative thereof.
20566. The method of item 20527 wherein the anti- fibrotic agent is an FLT-3 kinase inhibitor.
20567. The method of item 20527 wherein the anti- fibrotic agent is an FGF receptor kinase inhibitor.
20568. The method of item 20527 wherein the anti- fibrotic agent is a fibrinogen antagonist selected from the group consisting of AUV-201 (Auvation), MG-13926 (Sanofi-Aventis), plasminogen activator (CAS No. 105913-11-9) (from Sanofi-Aventis or UCB), plasminogen activator-2 (tPA-2) (Sanofi-Aventis), pro-urokinase (CAS No. 82657-92-9) (Sanofi-Aventis), and an analogue or derivative thereof.
20569. The method of item 20527 wherein the anti- fibrotic agent is a heat shock protein 90 antagonist selected from the group consisting of SRN-005 (Sirenade), geldanamycin, NSC-33050 (17- allylaminogeldanamycin; 17-AAG), 17-dimethy!aminoethylamino-17- demethoxy-geldanamycin (17-DMAG), rifabutin (rifamycin XIV, 1',4- didehydro-1-deoxy-1,4-dihydro-542-methylpropyl)~1-oxo-), and an analogue or derivative thereof.
20570. The method of item 20527 wherein the anti- fibrotic agent is a histone deacetylase inhibitor.
20571. The method of item 20527 wherein the anti- fibrotic agent is an HMGCoA reductase inhibitor selected from the group consisting of an atherosclerosis therapeutic from Lipid Sciences, ATI-16000 (ARYx Therapeutics), KS-01-019 (Kos Pharmaceuticals), Pharmaprojects No. 2197 (Sanofi-Aventi), RP 61969 (Sanofi-Aventis), and an analogue or derivative thereof.
20572. The method of item 20527 wherein the anti- fibrotic agent is an ICAM inhibitor.
20573. The method of item 20527 wherein the anti- fibrotic agent is an IL, ICE and IRAK antagonist, wherein the antagonist is a CJ-14877, CP-424174 (Pfizer), NF-61 (Negma-Lerads), and an analogue or derivative thereof.
20574. The method of item 20527 wherein the anti- fibrotic agent is an IL-2 inhibitor.
20575. The method of item 20527 wherein the anti- fibrotic agent is an immunosuppressant selected from the group consisting of teriflunomide (Sanofi Aventis), chlorsulfaquinoxalone (NSC-339004), chlorsulfaquinoxalone sulfate, CS-712 (Sankyo), ismomultin alfa (CAS No. 457913-93-8) (Akzo Nobel), antiallergics from GenPat77, antiinflammatories or AT-005 (Androclus Therapeutics), autoimmune disease therapeutics from EpiVax, BN-007 (Bone), budesonide (CAS No. 51333-22- 3) (MAP Pharmaceuticals), CO-14 (Genzyme), edratide (CAS No. 433922- 67-9) (Teva), EP-314 (Enanta), eprovafen (CAS No. 101335-99-3) (Sanofi- Aventis), HWA-131 (CAS No. 118788-41-3) (Sanofi-Aventis), immunomodulators from MerLion Pharmaceuticals, immunosuppressives from Alchemia, IPL-12 (Inflazyme), MDL-9563 (CAS No. 27086-86-8) (Sanofi-Aventis), Pharmaprojects No. 2330 (Sanofi-Aventis), Pharmaprojects No. 6426 (Abgenix), PXS-25 (Pharmaxis), rosmarinic acid (CAS No. 20283-92-5) (Sanofi-Aventis), RP 42927 or RP 54745 (CAS No. 135330-08-4) (Sanofi-Aventis), SGN-35 (Seattle Genetics), ST-1959 (Sigma-Tau), type I diabetes therapy from SYNX Pharma, UNIL-88 (Debiopharm), VP-025 (Vasogen), VR-694 (Vectura), PRTX-001 (Protalex), and an analogue or derivative thereof.
20576. The method of item 20527 wherein the anti- fibrotic agent is an IMPDH (inosine monophosphate).
20577. The method of item 20527 wherein the anti- fibrotic agent is an integrin antagonist.
20578. The method of item 20527 wherein the anti- fibrotic agent is an interleukin antagonist.
20579. The method of item 20527 wherein the anti- fibrotic agent is an inhibitor of type III receptor tyrosine kinase.
20580. The method of item 20527 wherein the anti- fibrotic agent is an irreversible inhibitor of enzyme methionine aminopeptidase type 2.
20581. The method of item 20527 wherein the anti- fibrotic agent is an isozyme selective delta protein kinase C inhibitor. 20582. The method of item 20527 wherein the anti- fibrotic agent a JAK3 enzyme inhibitor.
20583. The method of item 20527 wherein the anti- fibrotic agent is a JNK inhibitor.
20584. The method of item 20527 wherein the anti- fibrotic agent is a kinase inhibitor.
20585. The method of item 20527 wherein the anti- fibrotic agent is kinesin antagonist.
20586. The method of item 20527 wherein the anti- fibrotic agent is a kinesin antagonist.
20587. The method of item 20527 wherein the anti- fibrotic agent is a leukotriene inhibitor and antagonist selected from the group consisting of ambicromil (CAS No. 58805-38-2) (Sanofi-Aventis), amelubant (CAS No. 346735-24-8) (Boehringer Ingelheim), DW-1141 (Dong Wha), ebselen (Daiichi Pharmaceutical), ibudilast (Kyorin), leucotriene inhibitors from Sanofi-Aventis, lymphotoxin-beta receptor (LT-D) from Biogen Idee, Pharmaprojects No. 1535 and 2728 (CAS No. 119340-33-9) (Sanofi-Aventis), R-112 (Rigel), Rev-5367 (CAS No. 92532-05-3) (Sanofi- Aventis), RG-14893 (CAS No. 141835-49-6) (Sanofi-Aventis), RG-5901-A (CAS No. 101910-24-1), 92532-23-5, RP 66153 (CAS No. 142422-79-5), RP 66364 (CAS No.186916-92-5), RP 69698 (CAS No. 141748-00-7) (Sanofi-Aventis), SC-411930 (Pfizer), SC-41930 (CAS No. 120072-59-5) (Pfizer), SC-50605 (CAS No. 138828-39-4) (Pfizer), SC-51146 (CAS No. 141059-52-1), SC-53228 (CAS No. 153633-01-3) (Pfizer), spaglumic acid (ZY-15106) (CAS No.3106-85-2), 80619-64-3 (Novartis), tipredane (CAS No. 85197-77-9) (Bristol-Myers Squibb), U-75302 (CAS No. 119477-85-9) (Pfizer), and analogue or derivative thereof.
20588. The method of item 20527 wherein the anti- fibrotic agent is an MAP kinase inhibitor.
20589. The method of item 20527 wherein the anti- fibrotic agent is a matrix metalloproteinase inhibitor.
20590. The method of item 20527 wherein the anti- fibrotic agent is an MCP-CCR2 inhibitor.
20591. The method of item 20527 wherein the anti- fibrotic agent is an mTOR inhibitor.
20592. The method of item 20527 wherein the anti- fibrotic agent is an mTOR kinase inhibitor.
20593. The method of item 20527 wherein the anti- fibrotic agent is a microtubule inhibitor selected from the group consisting of antibody-maytansinoid conjugates from Biogen Idee, colchicines (MantiCore Pharmaceuticals), anticancer immunoconjugates from Johnson & Johnson, DIME from Octamer, gni-1f (GNI)1 huC242-DM4, huMy9-6-DM1 (ImmunoGen), IDN-5404 (Indena), IMO-098, IMOderm (Imotep), mebendazole (Introgen Therapeutics), microtubule poisons from Cambridge Enterprise, paclitaxel such as LOTAX from Aphios (CAS No. 33069-62-4), Genexol-PM from Samyang, Pharmaprojects No. 6383 (Tapestry Pharmaceuticals), RPR-112378 (Sanofi-Aventis), SGN-75 (Seattle Genetics), SPL-7435 (Starpharma), SSR-250411 (Sanofi-Aventis), trastuzumab-DM1 (Genentech), vinorelbine, and an analogue or derivative thereof. 20594. The method of item 20527 wherein the anti- fibrotic agent is an MIF inhibitor.
20595. The method of item 20527 wherein the anti- fibrotic agent is an MMP inhibitor.
20596. The method of item 20527 wherein the anti- fibrotic agent is a neurokinin (NK) antagonist selected from the group consisting of anthrotainin (CAS No. 148084-40-6) (Sanofi-Aventis), an IBS thereapeutic from ArQuIe, MDL-105212A (CAS No. 167261-60-1) (Ssanofi- Aventis), Pharmaprojects No. 2744, 3258 (CAS No. 139167-47-8) 4006, 4201 , or 5986 (Sanofi-Aventis), RP 67580 (CAS No. 135911-02-3), SR- 144190 (CAS No. 201152-86-5), SSR-240600, SSR-241586 (Sanofi- Aventis), TKA-457 (Novartis), vestipitant mesylate (CAS No. 334476-64-1) (GlaxoSmithKline), Win-64821 (Sanofi-Aventis), PRX-96026 (Predix Pharmaceuticals), and an analogue or derivative thereof.
20597. The method of item 20527 wherein the anti- fibrotic agent is an NF kappa B inhibitor selected from the group consisting of AVE-0545 or AVE-0547 (Sanofi-Aventis), bortezomib (CAS No. 179324- 69-7) (Millennium Pharmaceuticals), dexanabinol (CAS No. 112924-45-5) (Pharmos), dexlipotam (Viatris), Pharmaprojects No. 6283 (INDRA) (OXiGENE), IPL-576092 (CAS No. 137571-30-3) (Inflazyme), NFKB decoy (Corgentech), NFKB decoy oligo (AnGes MG), NFKB's from Ariad, osteoporosis treatments or S5 (F005) from Fulcrum Pharmaceuticals, P61 (Phytopharm), R-flurbiprofen (CAS No. 5104-49-4) (Encore Pharmaceuticals), and an analogue or derivative thereof.
20598. The method of item 20527 wherein the anti- fibrotic agent is a nitric oxide agonist. 20599. The method of item 20527 wherein the anti- fibrotic agent is an ornithine decarboxylase inhibitor.
20600. The method of item 20527 wherein the anti- fibrotic agent is a p38 MAP kinase inhibitor selected from the group consisting of AZD-6703 (AstraZeneca), JX-401 (Jexys Pharmaceuticals), BMS-2 (Bristol-Myers Squibb), a p38 MAP kinase inhibitor from Novartis, a p38-alpha MAP kinase inhibitor from Amphora, Pharmaprojects No. 5704 (Pharmacopeia), RPR-200765A (Sanofi-Aventis), SD-282 (Johnson & Johnson), TAK-715 (Takeda), and an analogue or derivative thereof.
20601. The method of item 20527 wherein the anti- fibrotic agent is a palmitoyl-protein thioesterase inhibitor.
20602. The method of item 20527 wherein the anti- fibrotic agent is a PDGF receptor kinase inhibitor selected from the group consisting of AAL-993, AMN-107, or ABP-309 (Novartis), AMG-706 (Amgen), BAY-57-9352 (Bayer), CDP-860 (UCB), E-7080 (Eisai), imatinib (CAS No. 152459-95-5) (Novartis), OSI-930 (OSI Pharmaceuticals), RPR- 127963E (Sanofi-Aventis), RWJ-540973 (Johnson & Johnson), sorafenib tosylate (Bayer), SlM 1657 (Pfizer), tandutinib (CAS No. 387867-13-2) (Millennium Pharmaceuticals), vatalanib (Novartis), ZK-CDK (Schering AG), and an analogue or derivative thereof.
20603. The method of item 20527 wherein the anti- fibrotic agent is (-)-halofenate (Metabolex), AMG-131 (Amgen), antidiabetics from Japan Tobacco, AZD-4619, AZD-8450, AZD-8677 (AstraZeneca), DRF-10945, balaglitazone (Dr Reddy's), CS-00088, CS-00098 (Chipscreen Biosciences), E-3030 (Eisai), etalocib (CAS No. 161172-51-6) (EIi Lilly), GSK-641597 (Ligand), GSK-677954 (GlaxoSmithKline), GW-409544 (Ligand), GW-590735 (GlaxoSmithKline), K-111 (Hoffmann-La Roche), LY- 518674 (EIi Lilly), LY-674 (Ligand), LY-929 (Ligand), MC-3001 , MC-3002 (MaxoCore Pharmaceuticals), metformin HCI + pioglitazone (CAS No. 1115- 70-4 and 112529-15-4), ACTOPLUS MET from Andrx), muraglitazar (CAS No. 331741-94-7) (Bristol-Myers Squibb), naveglitazar (Ligand), oleoylethanolamide (Kadmus Pharmaceuticals), ONO-5129, pioglitazone hydrochloride (CAS No. 111025-46-8 and 112529-15-4) (Takeda), PLX-204 (Plexxikon), PPAR agonists from Genfit, PPAR delta agonists from EIi Lilly, PPAR-alpha agonists from CrystalGenomics, PPAR-gamma modulators and PPAR-β modulators from CareX, rosiglitazone maleate (CAS No. 122324- 73-4 or 155141-29-0) (GlaxoSmithKline), rosiglitazone maleate/glimepir (CAS No. 155141-29-0 and 93479-97-1), AVANDARYL, rosiglitazone maleate/metformin extend (CAS No. 155141-29-0 and 657-24-9), AVANDAMET, rosiglitazone maleate+metformin, AVANDAMET (GlaxoSmithKline), tesaglitazar (AstraZeneca), and an analogue or derivative thereof.
20604. The method of item 20527 wherein the anti- fibrotic agent is a phosphatase inhibitor.
20605. The method of item 20527 wherein the anti- fibrotic agent is a phosphodiesterase (PDE) inhibitor selected from the group consisting of avanafil (Tanabe Seiyaku), dasantafil (CAS No. 569351- 91-3) (Schering-Plough), A-906119 (CAS No. 134072-58-5), DL-850 (Sanofi-Aventis), GRC-3015, GRC-3566, GRC-3886 (Glenmark), HWA-153 (CAS No. 56395-66-5) (Sanofi-Aventis), hydroxypumafentrine (Altana), IBFB-130011 , IBFB-14-016, IBFB-140301 , IBFB-150007, IBFB-211913 (IBFB Pharma), L-826141 (Merck & Co), medorinone (CAS No. 88296-61-1) (Sanofi-Aventis), MEM-1917 (Memory Pharmaceuticals), ND-1251 (Neuro3d), PDE inhibitors from ICOS, PDE IV inhibitors from Memory Pharmaceuticals and CrystalGenomics, Pharmaprojects No. 2742 and 6141 (Sanofi-Aventis), QAD-171 (Novartis), RHC-2963 (CAS No. 76993-12-9 and 76993-14-1), RPR-117658, RPR-122818 derivatives, SR-24870 , and RPR- 132294 (Sanofi-Aventis), SK-350 (ln2Gen), stroke therapy agents from deCODE Genetics, TAS-203 (Taiho), tofimilast (CAS No. 185954-27-2) (Pfizer), UK-371800 (Pfizer), WIN-65579 (CAS No. 158020-82-7) (Sanofi- Aventis), IBFB-130020 (IBFB Pharma), OPC-6535 (CAS No. 145739-56-6) (Otsuka), a phosphodiesterase III inhibitor, enoximone, a phosphodiesterase IV inhibitor, fosfosal, Atopik (Barrier Therapeutics), triflusal, a phosphodiesterase V inhibitor, and an analogue or derivative thereof.
20606. The method of item 20527 wherein the anti- fibrotic agent is a PKC inhibitor.
20607. The method of item 20527 wherein the anti- fibrotic agent is a platelet activating factor antagonist.
20608. The method of item 20527 wherein the anti- fibrotic agent is a platelet-derived growth factor receptor kinase inhibitor.
20609. The method of item 20527 wherein the anti- fibrotic agent is a prolyl hydroxylase inhibitor.
20610. The method of item 20527 wherein the anti- fibrotic agent is a polymorphonuclear neutrophil inhibitor.
20611. The method of item 20527 wherein the anti- fibrotic agent is a protein kinase B inhibitor.
20612. The method of item 20527 wherein the anti- fibrotic agent is a protein kinase C stimulant. 20613. The method of item 20527 wherein the anti- fibrotic agent is a purine nucleoside analogue.
20614. The method of item 20527 wherein the anti- fibrotic agent is a purinoreceptor P2X antagonist.
20615. The method of item 20527 wherein the anti- fibrotic agent is a Raf kinase inhibitor.
20616. The method of item 20527 wherein the anti- fibrotic agent is a reversible inhibitor of ErbB1 and ErbB2.
20617. The method of item 20527 wherein the anti- fibrotic agent is a ribonucleoside triphosphate reductase inhibitor.
20618. The method of item 20527 wherein the anti- fibrotic agent is an SDF-1 antagonist.
20619. The method of item 20527 wherein the anti- fibrotic agent is a sheddase inhibitor.
20620. The method of item 20527 wherein the anti- fibrotic agent is an SRC inhibitor.
20621. The method of item 20527 wherein the anti- fibrotic agent is a stromelysin inhibitor.
20622. The method of item 20527 wherein the anti- fibrotic agent is an Syk kinase inhibitor. 20623. The method of item 20527 wherein the anti- fibrotic agent is a telomerase inhibitor.
20624. The method of item 20527 wherein the anti- fibrotic agent is a TGF beta inhibitor selected from the group consisting of pirfenidone (CAS No. 53179-13-8) (MARNAC), tranilast (CAS No. 53902- 12-8) (Kissei), IN-1130 (ln2Gen), mannose-6-phosphate (BTG), TGF-β antagonists from Inflazyme (Pharmaprojects No. 6075), TGF-β antagonists from Sydney, non-industrial source), TGF-β I receptor kinase inhibitors from EIi Lilly, TGF-β receptor inhibitors from Johnson & Johnson, and an analogue or derivative thereof.
20625. The method of item 20527 wherein the anti- fibrotic agent is a TNFα antagonist or TACE inhibitor selected from the group consisting of adalimumab (CAS No. 331731-18-1) (Cambridge Antibody Technology), AGIX-4207 (AtheroGenics), AGT-1 (Advanced Biotherapy), an anti-inflammatory from Borean Pharma, Cellzome, or Paradigm Therapeutics, anti-inflammatory vaccine (TNF-alpha kinoid) from Neovacs, humanized anti-TNF antibody or an anti-TNF MAb (CB0006) Celltech (UCB), apratastat (CAS No. 287445-51-0) (Wyeth), BMS-561392 (Bristol-Myers Squibb), BN-006 (Bone), certolizumab pegol (CAS No. 428863-50-7 or CH-138 (UCB), cilomilast (CAS No. 153259-65-5) (GlaxoSmithKline), CR-1 (Nuada Pharmaceuticals), CRx-119 (CombinatoRx), D-5410 (UCB), dacopafant (CAS No. 125372-33-0) (Sanofi- Aventis), dersalazine (CAS No. 188913-57-7/188913-58-8) (Uriach), etanercept (CAS No. 185243-69-0) (Amgen), ethyl pyruvate (Critical (Critical Therapeutics), golimumab (CAS No. 476181-74-5) (Johnson & Johnson), hormono-immunotherapy from Ipsen, CDP571 (e.g., Humicade from UCB), IC-485 (ICOS), infliximab (CAS No. 170277-31-3) (Johnson & Johnson), IP- 751 (Manhattan Pharmaceuticals), ISIS-104838 (CAS No. 250755-32-9) (ISIS Pharmaceuticals), lenalidomide (CAS No. 191732-72-6) (Celgene), lentinan (CAS No. 37339-90-5) (Ajinomoto), MDL-201112 (CAS No. 142130-73-2) (Sanofi-Aventis), medroxyprogesterone (CAS No. 520-85-4) (InKine Pharmaceutical), N-acetylcysteine (CAS No. 616-91-1) (Zambon), NBE-P2 (DIREVO Biotech), nerelimomab (CAS No. 162774-06-3) (Chiron), OM-294DP (OM PHARMA), onercept (CAS No. 199685-57-9) (Yeda), PASSTNF-alpha (Verigen), pentoxifylline or oxypentifylline (Sanofi-Aventis), Pharmaprojects No. 4091 , 4241 , 4295, or 4488 (Sanofi-Aventis), Pharmaprojects No. 5480 (Amgen), Pharmaprojects No. 6749 (Cengent), pirfenidone (CAS No. 53179-13-8) (MARNAC), RPR-132294 (Sanofi- Aventis), S5 (F002) (Fulcrum Pharmaceuticals), simvastatin (CAS No. 79902-63-9) (Merck & Co), STA-6292 (Synta Pharmaceuticals), tacrolimus (CAS No. 104987-11-3) (Fujisawa LifeCycle Pharma), talactoferrin alfa (CAS No. 308240-58-6) (Agennix), thalidomide (CAS No. 50-35-1) (Celgene), TNF antagonists form ProStrakan, and Synergen, TNF inhibitors (Amgen), TNF-alpha antagonists from Dynavax Technologies and Jerina AG (Germany), TNF-alpha inhibitors from IBFB Pharma and Xencor (Xencor), torbafylline (CAS No. 105102-21-4) (Sanofi-Aventis), UR-1505 (Uriach), VT-346 (Viron Therapeutics), YSIL6 (Y's Therapeutics), YSTH2 (Y's Therapeutics), NPI-1302a-3 (Nereus Pharmaceuticals, a TNF antagonist from Jerina AG (Germany), dersalazine, and an analogue or derivative thereof.
20626. The method of item 20527 wherein the anti- fibrotic agent is a Toll receptor inhibitor.
20627. The method of item 20527 wherein the anti- fibrotic agent is a tubulin antagonist.
20628. The method of item 20527 wherein the anti- fibrotic agent is a tyrosine kinase inhibitor selected from the group consisting of SU-011248, SUTENT from Pfizer Inc. (New York, NY), BMS-354825, PN- 355 (Paracelsian Pharmaceuticals), AGN-199659 (Allergan), AAL-993 or ABP-309 (Novartis), adaphostin (NIH), AEE-788 (Novartis), AG-013736 (OSI Pharmaceuticals), AG-13736 (Pfizer), ALT-110 (Alteris Therapeutics), AMG-706 (Amgen), anticancer MAbs from Xencor, anti-EGFrvlll MAbs from Abgenix, anti-HER2 MAb from Abiogen, AZD-2171 or AZD-9935 (AstraZeneca), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), CEP-5214 (Cephalon), CEP-7055 (Cephalon), cetuximab (ImCIone Systems), CHIR-200131 and CHIR-258 (Chiron), CP-547632 (OSI Pharmaceuticals), CP-724714 (Pfizer), CT-301 (Creabilis Therapeutics), D- 69491 (Baxter International), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EGFR/ErbB2 inhibitors from Array BioPharma, erlotinib (CAS No. 183319- 69-9) (OSI Pharmaceuticals), EXEL-2880 (Exelixis), FK-778 (Sanofi- Aventis) , gefitinib (CAS No. 184475-35-2) (AstraZeneca), GW-2286 or GW- 654652 (GlaxoSmithKline), her2/neu antigen from AlphaVax, HER-2/neu inhibitor from Generex, Herzyme (Medipad) (Sirna Therapeutics), HKI-272 (Wyeth), HuMax-EGFr (Genmab), idronoxil (CAS No. 81267-65-4) (Novogen), IGF-1 inhibitors from Ontogen, IMC-11F8 (ImCIone Systems), kahalalide F (CAS No. 149204-42-2) (PharmaMar), KDR inhibitor from LG Life Sciences, KDR inhibitors from Abbott Laboratories, KDR kinase inhibitors (UCB), Kdr kinase inhibitors from Merck & Co, KRN-633 and KRN- 951 (Kirin Brewery), KSB-102 (Xenova), lapatinib ditosylate (CAS No. 388082-78-8) (GlaxoSmithKline), matuzumab (Merck KGaA), MDX-214 (Medarex), ME-103 (Pharmexa), MED-A300 (Gerolymatos), MNAC-13 (Lay Line Genomics), nimotuzumab (Center of Molecular Immunology), NSC- 330507 or NSC-707545 (NIH), NV-50 (Novogen), OSI-930 (OSI Pharmaceuticals), panitumumab (Abgenix), pelitinib (CAS No. 287933-82-7) (Wyeth), pertuzumab (CAS No. 380610-27-5) (Genentech), Pharmaprojects No. 3985 (Sanofi-Aventis), prostate cancer therapeutics from Sequenom (SQPC35, SQPC36, SQPC90), removab and remoxab (Trion Pharma), RG- 13022 (CAS No. 136831-48-6), RG-13291 (CAS No. 138989-50-1), or RG- 14620 (CAS No. 136831-49-7) (Sanofi-Aventis), RM-6427 (Romark), RNAi breast cancer therapy from Benitec, RP 53801 (CAS No. 125882-88-4) (Sanofi-Aventis), sorafenib tosylate (Bayer), SU-11657 (Pfizer), Tie-2 antagonists from Semaia (Hybrigenics), Tie-2 inhibitors from Ontogen, trastuzumab (CAS No. 180288-69-1) (Genentech), tyrosine kinase inhibitors from Sanofi-Aventis, U3-1287, U3-1565, U3-1784, U3-1800 (U3 Pharma), vatalanib (Novartis), VEGFR-2 kinase inhibitor from Bristol-Myers Squibb, XL-647 (Exelixis), ZD-6474 (AstraZeneca), ZK-CDK (Schering AG), an EGFR tyrosine kinase inhibitor, EKB-569 (Wyeth), and an analogue or derivative thereof.
20629. The method of item 20527 wherein the anti- fibrotic agent is a VEGF inhibitor.
20630. The method of item 20527 wherein the anti- fibrotic agent is a vitamin D receptor agonist.
20631. The method of item 20527 wherein the anti- fibrotic agent is ZD-6474 (an angiogenesis inhibitor).
20632. The method of item 20527 wherein the anti- fibrotic agent is AP-23573 (an mTOR inhibitor).
20633. The method of item 20527 wherein the anti- fibrotic agent is synthadotin (a tubulin antagonist).
20634. The method of item 20527 wherein the anti- fibrotic agent is S-0885 (a collagenase inhibitor).
20635. The method of item 20527 wherein the anti- fibrotic agent is aplidine (an elongation factor-1 alpha inhibitor). 20636. The method of item 20527 wherein the anti- fibrotic agent is ixabepilone (an epithilone).
20637. The method of item 20527 wherein the anti- fibrotic agent is IDN-5390 (an angiogenesis inhibitor).
20638. The method of item 20527 wherein the anti- fibrotic agent is SB-2723005 (an angiogenesis inhibitor).
20639. The method of item 20527 wherein the anti- fibrotic agent is ABT-518 (an angiogenesis inhibitor).
20640. The method of item 20527 wherein the anti- fibrotic agent is combretastatin (an angiogenesis inhibitor).
. . . 20641. The method of item 20527 wherein the anti- fibrotic agent is anecortave acetate (an angiogenesis inhibitor).
20642. The method of item 20527 wherein the anti- fibrotic agent is SB-715992 (a kinesin antagonist).
20643. The method of item 20527 wherein the anti- fibrotic agent is temsirolimus (an mTOR inhibitor).
20644. The method of item 20527 wherein the anti- fibrotic agent is adalimumab (a TNFα antagonist).
20645. The method of item 20527 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with an anti-fibrotic agent, and (b) implanting the medical device into the host. 20646. The method of item 20527 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with an anti-infective agent, and (b) implanting the medical device into the host.
20647. The method of item 20527 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with a polymer; and (b) implanting the medical device into the host.
20648. The method of item 20527 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with a composition comprising an anti-fibrotic agent and a polymer, and (b) implanting the medical device into the host.
20649. The method of item 20527 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with a composition comprising an anti-infective agent and a polymer, and (b) implanting the medical device into the host.
20650. The method of item 20527 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with a composition comprising an anti-fibrotic agent, an anti- infective agent and a polymer, and (b) implanting the medical device into the host.
20651. The method of item 20527 wherein the anti- infective agent is an anthracycline.
20652. The method of item 20527 wherein the anti- infective agent is doxorubicin. 20653. The method of item 20527 wherein the anti- infective agent is is mitoxantrone.
20654. The method of item 20527 wherein the anti- infective agent is a fluoropyrimidine.
20655. The method of item 20527 wherein the anti- infective agent is 5-fluorouracil (5-FU).
20656. The method of item 20527 wherein the anti- infective agent is a folic acid antagonist.
20657. The method of item 20527 wherein the anti- infective agent is methotrexate.
20658. The method of item 20527 wherein the anti- infective agent is a podophylotoxin.
20659. The method of item 20527 wherein the anti- infective agent is etoposide.
20660. The method of item 20527 wherein the anti- infective agent is camptothecin.
20661. The method of item 20527 wherein the anti- infective agent is a hydroxyurea.
20662. The method of item 20527 wherein the anti- infective agent is a platinum complex. 20663. The method of item 20527 wherein the anti- infective agent is cisplatin.
20664. The method of item 20527 wherein the composition comprises an anti-thrombotic agent.
20665. The method of item 20527 wherein the polymer is formed from reactants comprising a naturally occurring polymer.
20666. The method of item 20527 wherein the polymer is formed from reactants comprising protein.
20667. The method of item 20527 wherein the polymer is formed from reactants comprising carbohydrate.
20668. The method of item 20527 wherein the polymer is formed from reactants comprising biodegradable polymer.
20669. The method of item 20527 wherein the polymer is formed from reactants comprising nonbiodegradable polymer.
20670. The method of item 20527 wherein the polymer is formed from reactants comprising collagen.
20671. The method of item 20527 wherein the polymer is formed from reactants comprising methylated collagen.
20672. The method of item 20527 wherein the polymer is formed from reactants comprising fibrinogen. 20673. The method of item 20527 wherein the polymer reactants comprising thrombin.
20674. The method of item 20527 wherein the polymer reactants comprising blood plasma.
20675. The method of item 20527 wherein the polymer reactants comprising calcium salt.
20676. The method of item 20527 wherein the polymer reactants comprising an antifibronolytic agent.
20677. The method of item 20527 wherein the polymer reactants comprising fibrinogen analog.
20678. The method of item 20527 wherein the polymer reactants comprising albumin.
20679. The method of item 20527 wherein the polymer reactants comprising plasminogen.
20680. The method of item 20527 wherein the polymer reactants comprising von Willebrands factor.
20681. The method of item 20527 wherein the polymer reactants comprising Factor VIlI.
20682. The method of item 20527 wherein the polymer reactants comprising hypoallergenic collagen. 20683. The method of item 20527 wherein the polymer is formed from reactants comprising atelopeptidic collagen.
20684. The method of item 20527 wherein the polymer is formed from reactants comprising telopeptide collagen.
20685. The method of item 20527 wherein the polymer is formed from reactants comprising crosslinked collagen.
20686. The method of item 20527 wherein the polymer is formed from reactants comprising aprotinin.
20687. The method of item 20527 wherein the polymer is formed from reactants comprising epsilon-amino-n-caproic acid.
- . 20688. . The method of item 20527 wherein the polymer is formed from reactants comprising gelatin.
20689. The method of item 20527 wherein the polymer is formed from reactants comprising protein conjugates.
20690. The method of item 20527 wherein the polymer is formed from reactants comprising gelatin conjugates.
20691. The method of item 20527 wherein the polymer is formed from reactants comprising a synthetic polymer.
20692. The method of item 20527 wherein the polymer is formed from reactants comprising a synthetic isocyanate-containing compound. 20693. The method of item 20527 wherein the polymer is formed from reactants comprising a synthetic thiol-containing compound.
20694. The method of item 20527 wherein the polymer is formed from reactants comprising a synthetic compound containing at least two thiol groups.
20695. The method of item 20527 wherein the polymer is formed from reactants comprising a synthetic compound containing at least three thiol groups.
20696. The method of item 20527 wherein the polymer is formed from reactants comprising a synthetic compound containing at least four thiol groups.
20697. The method of item 20527 wherein the polymer is formed from reactants comprising a synthetic amino-containing compound.
20698. The method of item 20527 wherein the polymer is formed from reactants comprising a synthetic compound containing at least two amino groups.
20699. The method of item 20527 wherein the polymer is formed from reactants comprising a synthetic compound containing at least three amino groups.
20700. The method of item 20527 wherein the polymer is formed from reactants comprising a synthetic compound containing at least four amino groups. 20701. The method of item 20527 wherein the polymer is formed from reactants comprising a synthetic compound comprising a carbonyl-oxygen-succinimidy! group.
20702. The method of item 20527 wherein the polymer is formed from reactants comprising a synthetic compound comprising at least two carbonyl-oxygen-succinimidyl groups.
20703. The method of item 20527 wherein the polymer is formed from reactants comprising a synthetic compound comprising at least three carbonyl-oxygen-succinimidyl groups.
20704. The method of item 20527 wherein the polymer is formed from reactants comprising a synthetic compound comprising at least four carbonyl-oxygen-succinimidyl groups.
20705. The method of item 20527 wherein the polymer is formed from reactants comprising a synthetic polyalkylene oxide- containing compound.
20706. The method of item 20527 wherein the polymer is formed from reactants comprising a synthetic compound comprising both polyalkylene oxide and biodegradable polyester blocks.
20707. The method of item 20527 wherein the polymer is formed from reactants comprising a synthetic polyalkylene oxide- containing compound having reactive amino groups.
20708. The method of item 20527 wherein the polymer is formed from reactants comprising a synthetic polyalkylene oxide- containing compound having reactive thiol groups. 20709. The method of item 20527 wherein the polymer is formed from reactants comprising a synthetic polyalkylene oxide- containing compound having reactive carbonyl-oxygen-succinimidyl groups.
20710. The method of item 20527 wherein the polymer is formed from reactants comprising a synthetic compound comprising a biodegradable polyester block.
20711. The method of item 20527 wherein the polymer is formed from reactants comprising a synthetic polymer formed in whole or part from lactic acid or lactide.
20712. The method of item 20527 wherein the polymer is formed from reactants comprising a synthetic polymer formed in whole or part from glycolic acid or glycolide.
20713. The method of item 20527 wherein the polymer is formed from reactants comprising polylysine.
20714. The method of item 20527 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound comprising a polyalkylene oxide portion.
20715. The method of item 20527 wherein the polymer is formed from reactants comprising (a) protein and (b) polylysine.
20716. The method of item 20527 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound having at least four thiol groups. 20717. The method of item 20527 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound having at least four amino groups.
20718. The method of item 20527 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound having at least four carbonyl-oxygen-succinimide groups.
20719. The method of item 20527 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound having a biodegradable region formed from reactants selected from lactic acid, lactide, glycolic acid, glycolide, and epison-caprolactone.
20720. The method of item 20527 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound comprising a polyalkylene oxide portion.
20721. The method of item 20527 wherein the polymer is formed from reactants comprising (a) collagen and (b) polylysine.
20722. The method of item 20527 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound having at least four thiol groups.
20723. The method of item 20527 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound having at least four amino groups.
20724. The method of item 20527 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound having at least four carbonyl-oxygen-succinimide groups. 20725. The method of item 20527 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound having a biodegradable region formed from reactants selected from lactic acid, lactide, glycolic acid, glycolide, and epison-caprolactone.
20726. The method of item 20527 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound comprising a polyalkylene oxide portion.
20727. The method of item 20527 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) polylysine.
20728. The method of item 20527 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound having at least four thiol groups.
20729. The method of item 20527 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound having at least four amino groups.
20730. The method of item 20527 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound having at least four carbonyl-oxygen-succinimide groups.
20731. The method of item 20527 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound having a biodegradable region formed from reactants selected from lactic acid, lactide, glycolic acid, glycolide, and epison-caprolactone. 20732. The method of item 20527 wherein the polymer is formed from reactants comprising hyaluronic acid.
20733. The method of item 20527 wherein the polymer is formed from reactants comprising a hyaluronic acid derivative.
20734. The method of item 20527 wherein the polymer is formed from reactants comprising pentaerythritol poly(ethylene glycol)ether tetra-sulfhydryl of number average molecular weight between 3,000 and 30,000.
20735. The method of item 20527 wherein the polymer is formed from reactants comprising pentaerythritol poly(ethylene glycol)ether tetra-amino of number average molecular weight between 3,000 and 30,000.
20736. The method of item 20527 wherein the polymer is formed from reactants comprising (a) a synthetic compound having a number average molecular weight between 3,000 and 30,000 and comprising a polyalkylene oxide region and multiple nucleophilic groups, and (b) a synthetic compound having a number average molecular weight between 3,000 and 30,000 and comprising a polyalkylene oxide region and multiple electrophilic groups.
20737. The method of item 20527 wherein the composition comprises a colorant.
20738. The method of item 20527 wherein the composition is sterile. 20739. A method as in any one of items 20527-20738, wherein the device is a penile implant that is a flexible rod.
20740. A method as in any one of items 20527-20738, wherein the device is a penile implant that is a hinged rod.
20741. A method as in any one of items 20527-20738, wherein the device is a penile implant that is an inflatable device with a pump.
20742. A method for implanting a medical device comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with i) an anti-fibrotic agent, ii) an anti-infective agent, iii) a polymer, iv) a composition comprising an anti-fibrotic agent and a polymer, v) a composition comprising an anti-infective agent and a polymer, or vi) a composition comprising an anti-fibrotic agent, an anti- infective agent and a polymer, and (b) implanting the medical device into the host, wherein the medical device is an endotracheal or tracheostomy tube.
20743. The method of item 20742 wherein the anti- fibrotic agent is an adensosine A2A receptor antagonist.
20744. The method of item 20742 wherein the anti- fibrotic agent is an AKT inhibitor.
20745. The method of item 20742 wherein the anti- fibrotic agent is an alpha 2 integrin antagonist, wherein the alpha 2 integrin antagonist is Pharmaprojects No. 5754 (Merck KgaA).
20746. The method of item 20742 wherein the anti- fibrotic agent is an alpha 4 integrin antagonist. 20747. The method of item 20742 wherein the anti- fibrotic agent is an alpha 7 nicotinic receptor agonist.
20748. The method of item 20742 wherein the anti- fibrotic agent is an angiogenesis inhibitor selected from the group consisting of AG-12,958 (Pfizer), ATN-161 (Attenuon LLC), neovastat, an angiogenesis inhibitor from Jerina AG (Germany), NM-3 (Mercian), VGA-1155 (Taisho), FCE-26644 (Pfizer), FCE-26950 (Pfizer), FPMA (Meiji Daries), FR-111142 (Fujisawa), GGTI-298, GM-1306 (Ligand), GPA-1734 (Novartis), NNC-47- 0011 (Novo Nordisk), herbamycin (Nippon Kayaku), lenalidomide (Celegene), IP-10 (NIH), ABT-828 (Abbott), KIN-841 (Tokushima University, Japan), SF-1126 (Semafore Pharmaceuticals), laminin technology (NIH), CHIR-258 (Chiron), NVP-AEW541 (Novartis), NVP-AEW541 (Novartis), Vt16907 (Alchemia), OXI-8007 (Oxigene), EG-3306 (Ark Therapeutics), Maspin (Arriva), ABT-567 (Abbott), PPI-2458 (Praecis Pharmaceuticals), CC-5079, CC-4089 (Celgene), HIF-1 alpha inhibitors (Xenova), S-247 (Pfizer), AP-23573 (Ariad), AZD-9935 (Astra Zeneca), mebendazole (Introgen Therapeutics), MetAP-2 inhibitors (GlaxoSmithKline), AG-615 (Angiogene Pharmaceuticals), Tie-2 antagonists (Hybrigenics), NC-381 , CYC-381, NC-169, NC-219, NC-383, NC-384, NC-407 (Lorus Therapeutics), ATN-224 (Attenuon), ON-01370 (Onconova), Vitronectin antagonists (Amgen), SDX-103 (Salmedix), Vitronectin antagonists (Shire), CHP (Riemser), TEK (Amgen), Anecortave acetate (Alcon), T46.2 (Matrix Therapeutics), HG-2 (Heptagen), TEM antagonists (Genzyme), Oxi-4500 (Oxigene), ATN-161 (Attenuon), WX-293 (Wilex), M-2025 (Metris Therapeutics), Alphastatin (BioActa), YH-16 (Yantai Rongchang), BIBF- 1120 (Boehringer Ingelheim), BAY-57-9352 (Bayer), AS-1404 (Cancer Research Technology), SC-77964 (Pfizer), glycomimetics (BioTie Therapies), TIE-2 Inhibitors (Ontogen), DIMI, Octamer (Octamer), ABR- 215050 (Active Biotech), ABT-518 (Abbott), KDR inhibitors (Abbott), BSF- 466895 (Abbott), SCH-221153 (Schering-Plough), DAC.antiangiogenic (ConjuChem), TFPI (EntreMed), AZD-2171 (Astra-Zenaca), CDC-394 (Celgene), LY290293 (EIi Lilly), IDN-5390 (Indena), Kdr Kinase Inhibitors (Merck), CT-113020, CT-116433, CT-116563, CT-31890, CT-32228) (Cell Therapeutics), A-299620 (Abbott), TWEAK Inhibitor (Amgen), VEGF modulators (Johnson and Johnson), Tum-N53, tumstatin (Genzyme), Thios- 1, Thios-2 (Thios Pharmaceuticals), MV-6401 (Miravant Medical Technologies), Spisulosine (PharmaMar), CEP-7055 (Cephalon), AUV-201 (Auvation), LM-609 (EIi Lilly), SKF-106615 (AnorMED), Oglufanide disodium (Cytran), BW-114 (Pharminox), Calreticulin (NIH), WX-678 (Wilex), SD-7784 (Pfizer), WX-UK1 (Wilex), SH-268 (Schering AG), 2-Me-PGA (Celgene), S- 137 (Pfizer), ZD-6126 (Angiogene Pharmaceuticals), SG-292 (SignalGen), Benefin (Lane Labs), A6, A36 (Angstrom), SB-2723005 (GlaxoSmithKline), SC-7 (Cell Therapeutics), ZEN-014 (AEterna Zentaris), 2-methoxyestradiol (EntreMed), NK-130119 (Nippon Kayaku), CC-10004 (Celgene), AVE- 8062A (Ajinomoto), Tacedinaline (Pfizer), Actinonin (Tokyo Metropolitan Institute of Medical Science), Lenalidomide (Celgene), VGA-1155, BTO-956 (SRI International), ER-68203-00 (Eisai), CT-6685 (UCB), JKC-362 (Phoenix Pharmaceuticals), DMI-3798 (DMI Biosciences, Angiomate (Ipsen), ZD-6474 (AstraZeneca), CEP-5214 (Cephalon), Canstatin (Genzyme), NM-3 (Mercian), Oridigm (MediQuest Therapeutics), Exherin (Adherex), BLS-0597 (Boston Life Sciences), PTC-299 (PTC Therapeutics), NPI-2358 (Nereus Pharmaceuticals), CGP-79787 (Novartis), AEE-788 (Novartis), CKD-732 (Chong Kun Dang), CP-564959 (OSI Pharmaceuticals), CM-101 (CarboMed), CT-2584, CT3501 (Cell Therapeutics), combretastatin and analogues and derivatives thereof (Oxigene), Rebimastat (Bristol-Meyers Squibb), Dextrin 2-sulfate (ML Laboratories), Cilengitide (Merk KGaA), NSC-706704 (Pharminox), KRN- 951 (Kirin Brewery), Ukrain, NSC-631570 (Nowicky Pharma), Tecogalan sodium (Daiichi Pharmaceutical), Tz-93 (Tsumura), TBC-1635 (Encysive Pharmaceuticals), TAN-1120 (Takeda), Semaxanib (Pfizer), BDI-7800 (Biopharmacopae), SD-186, SD-983 (Bristol-Meyers Squibb), SB-223245 (GlaxoSmithKline), SC-236 (Pfizer), RWJ-590973 (Johnson and Johnson), ILX-1850 (Genzyme), SC-68488, S-836 (Pfizer), CG-55069-11 (CuraGen), Ki-23057 (Kirin Brewery), CCX-700 (Chemoentryx), Pegaptanib octasodium (Giled Sciences), ANGIOCOL (available from Biostratum Inc.), or an analogue or derivative thereof.
20749. The method of item 20742 wherein the anti- fibrotic agent is an apoptosis antagonist.
20750. The method of item 20742 wherein the anti- fibrotic agent is an apoptosis activator.
20751. The method of item 20742 wherein the anti- fibrotic agent is a beta 1 integrin antagonist.
20752. The method of item 20742 wherein the anti- fibrotic agent is a beta tubulin inhibitor.
20753. The method of item 20742 wherein the anti- fibrotic agent is a blocker of enzyme production in Hepatitis C.
20754. The method of item 20742 wherein the anti- fibrotic agent is a Bruton's tyrosine kinase inhibitor.
20755. The method of item 20742 wherein the anti- fibrotic agent is a calcineurin inhibitor.
20756. The method of item 20742 wherein the anti- fibrotic agent is a caspase 3 inhibitor. 20757. The method of item 20742 wherein the anti- fibrotic agent is a CC chemokine receptor antagonist.
20758. The method of item 20742 wherein the anti- fibrotic agent is a cell cycle inhibitor selected from the group consisting of SNS-595 (Sunesis), synthadotin, KRX-0403, and an analogue or derivative thereof.
20759. The method of item 20742 wherein the anti- fibrotic agent is a cathepsin B inhibitor.
20760. The method of item 20742 wherein the anti- fibrotic agent is a cathepsin K inhibitor, wherein the cathepsin K inhibitor is 462795 (GlaxoSmithKline), INPL-022-D6 (Amura Therapeutics), or an analogue or derivative thereof.
20761. The method of item 20742 wherein the anti- fibrotic agent is a cathepsin L inhibitor.
20762. The method of item 20742 wherein the anti- fibrotic agent is a CD40 antagonist.
20763. The method of item 20742 wherein the anti- fibrotic agent is a chemokine receptor agonist.
20764. The method of item 20742 wherein the anti- fibrotic agent is a chymase inhibitor.
20765. The method of item 20742 wherein the anti- fibrotic agent is a collagenase antagonist. 20766. The method of item 20742 wherein the anti- fibrotic agent is a CXCR antagonist.
20767. The method of item 20742 wherein the anti- fibrotic agent is a cyclin dependent kinase inhibitor selected from the group consisting of a CDK-1 inhibitor, a CDK-2 inhibitor, a CDK- 4 inhibitor, a CDK-6 inhibitor, a CAK1 inhibitor from GPC Biotech and Bristol-Myers Squibb, RGB-286199 (GPC Biotech), an anticancer agent from Astex Technology, a CAK1 inhibitor from GPC Biotech, a CDK inhibitor from Sanofi-Aventis, a CDK1/CDK2 inhibitor from Amgen, a CDK2 inhibitor from SUGEN-2 (Pfizer), a hearing loss therapy agent (Sound Pharmaceuticals), PD-0332991 (Pfizer), RGB-286199 (GPC Biotech), Ro-0505124 (Hoffmann- La Roche), a Ser/Thr kinase inhibitor from Lilly (EIi Lilly), CVT-2584 (CAS No. 199986-75-9) (CV Therapeutics), and an analogue or derivative thereof.
20768. The method of item 20742 wherein the anti- fibrotic agent is a cyclooxygenase 1 inhibitor.
20769. The method of item 20742 wherein the anti- fibrotic agent is a DHFR inhibitor.
20770. The method of item 20742 wherein the anti- fibrotic agent is a dual integrin inhibitor.
20771. The method of item 20742 wherein the anti- fibrotic agent is an elastase inhibitor.
20772. The method of item 20742 wherein the anti- fibrotic agent is an elongation factor-1 alpha inhibitor. 20773. The method of item 20742 wherein the anti- fibrotic agent is an endothelial growth factor antagonist.
20774. The method of item 20742 wherein the anti- fibrotic agent is an endothelial growth factor receptor kinase inhibitor selected from the group consisting of sorafenib tosylate (Bayer), AAL-993 (Novartis), ABP-309 (Novartis), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EXEL- 2880 (Exelixis), GW-654652 (GlaxoSmithKline), a KDR inhibitor from LG Life Sciences, CT-6685 and CT-6729 (UCB), KRN-633 and KRN-951 (Kirin Brewery), OSI-930 (OSI Pharmaceuticals), SP-5.2 (Supratek Pharma), SU-
11657 (Pfizer), a Tie-2 antagonist (Hybrigenics), a VEGFR-2 kinase inhibitor (Bristol-Myers Squibb), XL-647 (Exelixis), a KDR inhibitor from Abbott Laboratories, sorafenib tosylate, and an analogue or derivative thereof.
20775. The method of item 20742 wherein the anti- fibrotic agent is an endotoxin antagonist.
20776. The method of item 20742 wherein the anti- fibrotic agent is an epothilone and tubulin binder.
20777. The method of item 20742 wherein the anti- fibrotic agent is an estrogen receptor antagonist.
20778. The method of item 20742 wherein the anti- fibrotic agent is an FGF inhibitor.
20779. The method of item 20742 wherein the anti- fibrotic agent is a farnexyl transferase inhibitor. 20780. The method of item 20742 wherein the anti- fibrotic agent is famesyltransferase inhibitor selected from the group of A- 197574 (Abbott), a famesyltransferase inhibitor from Servier, FPTIII (Strathclyde Institute for Drug R), LB-42908 (LG Life Sciences), Pharmaprojects No. 5063 (Genzyme), Pharmaprojects No. 5597 (Ipsen), Yissum Project No. B-1055 (Yissum), and an analogue or derivative thereof.
20781. The method of item 20742 wherein the anti- fibrotic agent is an FLT-3 kinase inhibitor.
20782. The method of item 20742 wherein the anti- fibrotic agent is an FGF receptor kinase inhibitor.
20783. The method of item 20742 wherein the anti- fibrotic agent is a fibrinogen antagonist selected from the group consisting of AUV-201 (Auvation), MG-13926 (Sanofi-Aventis), plasminogen activator (CAS No! 105913-11-9) (from Sanofi-Aventis or UCB), plasminogen activator-2 (tPA-2) (Sanofi-Aventis), pro-urokinase (CAS No. 82657-92-9) (Sanofi-Aventis), and an analogue or derivative thereof.
20784. The method of item 20742 wherein the anti- fibrotic agent is a heat shock protein 90 antagonist selected from the group consisting of SRN-005 (Sirenade), geldanamycin, NSC-33050 (17- allylaminogeldanamycin; 17-AAG), 17-dimethylaminoethylamino-17- demethoxy-geldanamycin (17-DMAG), rifabutin (rifamycin XIV, 1',4- didehydro-1-deoxy-1 ,4-dihydro-5'-(2-methylpropyl)-1-oxo-), and an analogue or derivative thereof.
20785. The method of item 20742 wherein the anti- fibrotic agent is a histone deacetylase inhibitor. 20786. The method of item 20742 wherein the anti- fibrotic agent is an HMGCoA reductase inhibitor selected from the group consisting of an atherosclerosis therapeutic from Lipid Sciences, ATM 6000 (ARYx Therapeutics), KS-01-019 (Kos Pharmaceuticals), Pharmaprojects No. 2197 (Sanofi-Aventi), RP 61969 (Sanofi-Aventis), and an analogue or derivative thereof.
20787. The method of item 20742 wherein the anti- fibrotic agent is an ICAM inhibitor.
20788. The method of item 20742 wherein the anti- fibrotic agent is an IL, ICE and IRAK antagonist, wherein the antagonist is a CJ-14877, CP-424174 (Pfizer), NF-61 (Negma-Lerads), and an analogue or derivative thereof.
20789. The method of item 20742 wherein the anti- fibrotic agent is an IL-2 inhibitor.
20790. The method of item 20742 wherein the anti- fibrotic agent is an immunosuppressant selected from the group consisting of teriflunomide (Sanofi Aventis), chlorsulfaquinoxalone (NSC-339004), chlorsulfaquinoxalone sulfate, CS-712 (Sankyo), ismomultin alfa (CAS No. 457913-93-8) (Akzo Nobel), antiallergics from GenPat77, antiinflammatories or AT-005 (Androclus Therapeutics), autoimmune disease therapeutics from EpiVax, BN-007 (Bone), budesonide (CAS No. 51333-22- 3) (MAP Pharmaceuticals), CO-14 (Genzyme), edratide (CAS No. 433922- 67-9) (Teva), EP-314 (Enanta), eprovafen (CAS No. 101335-99-3) (Sanofi- Aventis), HWA-131 (CAS No. 118788-41-3) (Sanofi-Aventis), immunomodulators from MerLion Pharmaceuticals, immunosuppressives from Alchemia, IPL-12 (Inflazyme), MDL-9563 (CAS No. 27086-86-8) (Sanofi-Aventis), Pharmaprojects No. 2330 (Sanofi-Aventis), Pharmaprojects No. 6426 (Abgenix), PXS-25 (Pharmaxis), rosmarinic acid (CAS No. 20283-92-5) (Sanofi-Aventis), RP 42927 or RP 54745 (CAS No. 135330-08-4) (Sanofi-Aventis), SGN-35 (Seattle Genetics), ST-1959 (Sigma-Tau), type I diabetes therapy from SYNX Pharma, UNIL-88 (Debiopharm), VP-025 (Vasogen), VR-694 (Vectura), PRTX-001 (Protalex), and an analogue or derivative thereof.
20791. The method of item 20742 wherein the anti- fibrotic agent is an IMPDH (inosine monophosphate).
20792. The method of item 20742 wherein the anti- fibrotic agent is an integrin antagonist.
20793. The method of item 20742 wherein the anti- fibrotic agent is an interleukin antagonist.
20794. The method of item 20742 wherein the anti- fibrotic agent is an inhibitor of type III receptor tyrosine kinase.
20795. The method of item 20742 wherein the anti- fibrotic agent is an irreversible inhibitor of enzyme methionine aminopeptidase type 2.
20796. The method of item 20742 wherein the anti- fibrotic agent is an isozyme selective delta protein kinase C inhibitor.
20797. The method of item 20742 wherein the anti- fibrotic agent a JAK3 enzyme inhibitor.
20798. The method of item 20742 wherein the anti- fibrotic agent is a JNK inhibitor. 20799. The method of item 20742 wherein the anti- fibrotic agent is a kinase inhibitor.
20800. The method of item 20742 wherein the anti- fibrotic agent is kinesin antagonist.
20801. The method of item 20742 wherein the anti- fibrotic agent is a kinesin antagonist.
20802. The method of item 20742 wherein the anti- fibrotic agent is a leukotriene inhibitor and antagonist selected from the group consisting of ambicromil (CAS No. 58805-38-2) (Sanofi-Aventis), amelubant (CAS No. 346735-24-8) (Boehringer Ingelheim), DW-1141 (Dong Wha), ebselen (Daiichi Pharmaceutical), ibudilast (Kyorin), leucotriene inhibitors from Sanofi-Aventis, lymphotoxin-beta receptor (LT-D) from Biogen Idee, Pharmaprojects No. 1535 and 2728 (CAS No. 119340-33-9) (Sanofi-Aventis), R-112 (Rigel), Rev-5367 (CAS No. 92532-05-3) (Sanofi- Aventis), RG-14893 (CAS No. 141835-49-6) (Sanofi-Aventis), RG-5901-A (CAS No. 101910-24-1), 92532-23-5, RP 66153 (CAS No. 142422-79-5), RP 66364 (CAS No.186916-92-5), RP 69698 (CAS No. 141748-00-7) (Sanofi-Aventis), SC-411930 (Pfizer), SC-41930 (CAS No. 120072-59-5) (Pfizer), SC-50605 (CAS No. 138828-39-4) (Pfizer), SC-51146 (CAS No. 141059-52-1), SC-53228 (CAS No. 153633-01-3) (Pfizer), spaglumic acid (ZY-15106) (CAS No.3106-85-2), 80619-64-3 (Novartis), tipredane (CAS No. 85197-77-9) (Bristol-Myers Squibb), U-75302 (CAS No. 119477-85-9) (Pfizer), and analogue or derivative thereof.
20803. The method of item 20742 wherein the anti- fibrotic agent is an MAP kinase inhibitor. 20804. The method of item 20742 wherein the anti- fibrotic agent is a matrix metalloproteinase inhibitor.
20805. The method of item 20742 wherein the anti- fibrotic agent is an MCP-CCR2 inhibitor.
20806. The method of item 20742 wherein the anti- fibrotic agent is an mTOR inhibitor.
20807. . The method of item 20742 wherein the anti- fibrotic agent is an mTOR kinase inhibitor.
20808. The method of item 20742 wherein the anti- fibrotic agent is a microtubule inhibitor selected from the group consisting of antibody-maytansinoid conjugates from Biogen Idee, colchicines (MantiCore Pharmaceuticals), anticancer immunoconjugates from Johnson & Johnson, DIME from Octamer, gni-1f (GNI), huC242-DM4, huMy9-6-DM1 (ImmunoGen), IDN-5404 (Indena), IMO-098, IMOderm (Imotep), mebendazole (Introgen Therapeutics), microtubule poisons from Cambridge Enterprise, paclitaxel such as LOTAX from Aphios (CAS No. 33069-62-4), Genexol-PM from Samyang, Pharmaprojects No. 6383 (Tapestry Pharmaceuticals), RPR-112378 (Sanofi-Aventis), SGN-75 (Seattle Genetics), SPL-7435 (Starpharma), SSR-250411 (Sanofi-Aventis), trastuzumab-DM1 (Genentech), vinorelbine, and an analogue or derivative thereof.
20809. The method of item 20742 wherein the anti- fibrotic agent is an MIF inhibitor.
20810. The method of item 20742 wherein the anti- fibrotic agent is an MMP inhibitor. 20811. The method of item 20742 wherein the anti- fibrotic agent is a neurokinin (NK) antagonist selected from the group consisting of anthrotainin (CAS No. 148084-40-6) (Sanofi-Aventis), an IBS thereapeutic from ArQuIe, MDL-105212A (CAS No. 167261-60-1) (Ssanofi- Aventis), Pharmaprojects No. 2744, 3258 (CAS No. 139167-47-8) 4006, 4201 , or 5986 (Sanofi-Aventis), RP 67580 (CAS No. 135911-02-3), SR- 144190 (CAS No. 201152-86-5), SSR-240600, SSR-241586 (Sanofi- Aventis), TKA-457 (Novartis), vestipitant mesylate (CAS No. 334476-64-1) (GlaxoSmithKline), Win-64821 (Sanofi-Aventis), PRX-96026 (Predix Pharmaceuticals), and an analogue or derivative thereof.
20812. The method of item 20742 wherein the anti- fibrotic agent is an NF kappa B inhibitor selected from the group consisting of AVE-0545 or AVE-0547 (Sanofi-Aventis), bortezomib (CAS No. 179324- 69-7) (Millennium Pharmaceuticals), dexanabinol (CAS No. 112924-45-5) (Pharmos), dexlipotam (Viatris), Pharmaprojects No. 6283 (INDRA) (OXiGENE), IPL-576092 (CAS No. 137571-30-3) (Inflazyme), NFKB decoy (Corgentech), NFKB decoy oligo (AnGes MG), NFKB's from Ariad, osteoporosis treatments or S5 (F005) from Fulcrum Pharmaceuticals, P61 (Phytopharm), R-flurbiprofen (CAS No. 5104-49-4) (Encore Pharmaceuticals), and an analogue or derivative thereof.
20813. The method of item 20742 wherein the anti- fibrotic agent is a nitric oxide agonist.
20814. The method of item 20742 wherein the anti- fibrotic agent is an ornithine decarboxylase inhibitor.
20815. The method of item 20742 wherein the anti- fibrotic agent is a p38 MAP kinase inhibitor selected from the group consisting of AZD-6703 (AstraZeneca), JX-401 (Jexys Pharmaceuticals), BMS-2 (Bristol-Myers Squibb), a p38 MAP kinase inhibitor from Novartis, a p38-alpha MAP kinase inhibitor from Amphora, Pharmaprojects No. 5704 (Pharmacopeia), RPR-200765A (Sanofi-Aventis), SD-282 (Johnson & Johnson), TAK-715 (Takeda), and an analogue or derivative thereof.
20816. The method of item 20742 wherein the anti- fibrotic agent is a palmitoyl-protein thioesterase inhibitor.
20817. The method of item 20742 wherein the anti- fibrotic agent is a PDGF receptor kinase inhibitor selected from the group consisting of AAL-993, AMN-107, or ABP-309 (Novartis), AMG-706 (Amgen), BAY-57-9352 (Bayer), CDP-860 (UCB), E-7080 (Eisai), imatinib (CAS No. 152459-95-5) (Novartis), OSI-930 (OSI Pharmaceuticals), RPR- 127963E (Sanofi-Aventis), RWJ-540973 (Johnson & Johnson), sorafenib tosylate (Bayer), SU-11657 (Pfizer), tandutinib (CAS No. 387867-13-2) (Millennium Pharmaceuticals), vatalanib (Novartis), ZK-CDK (Schering AG), and an analogue or derivative thereof.
20818. The method of item 20742 wherein the anti- fibrotic agent is (-)-halofenate (Metabolex), AMG-131 (Amgen), antidiabetics from Japan Tobacco, AZD-4619, AZD-8450, AZD-8677 (AstraZeneca), DRF-10945, balaglitazone (Dr Reddy's), CS-00088, CS-00098 (Chipscreen Biosciences), E-3030 (Eisai), etalocib (CAS No. 161172-51-6) (EIi Lilly), GSK-641597 (Ligand), GSK-677954 (GlaxoSmithKline), GW-409544 (Ligand), GW-590735 (GlaxoSmithKline), K-111 (Hoffmann-La Roche), LY- 518674 (EIi Lilly), LY-674 (Ligand), LY-929 (Ligand), MC-3001 , MC-3002 (MaxoCore Pharmaceuticals), metformin HCI + pioglitazone (CAS No. 1115- 70-4 and 112529-15-4), ACTOPLUS MET from Andrx), muraglitazar (CAS No. 331741-94-7) (Bristol-Myers Squibb), naveglitazar (Ligand), oieoylethanolamide (Kadmus Pharmaceuticals), ONO-5129, pioglitazone hydrochloride (CAS No. 111025-46-8 and 112529-15-4) (Takeda), PLX-204 (Plexxikon), PPAR agonists from Genfit, PPAR delta agonists from EIi Lilly, PPAR-alpha agonists from CrystalGenomics, PPAR-gamma modulators and PPAR-β modulators from CareX, rosiglitazone maleate (CAS No. 122324- 73-4 or 155141-29-0) (GlaxoSmithKline), rosiglitazone maleate/glimepir (CAS No. 155141-29-0 and 93479-97-1), AVANDARYL, rosiglitazone maleate/metformin extend (CAS No. 155141-29-0 and 657-24-9), AVANDAMET, rosiglitazone maleate+metformin, AVANDAMET (GlaxoSmithKline), tesaglitazar (AstraZeneca), and an analogue or derivative thereof.
20819. The method of item 20742 wherein the anti- fibrotic agent is a phosphatase inhibitor.
20820. The method of item 20742 wherein the anti- fibrotic agent is a phosphodiesterase (PDE) inhibitor selected from the group consisting of avanafil (Tanabe Seiyaku), dasantafil (CAS No. 569351- 91-3) (Schering-Plough), A-906119 (CAS No. 134072-58-5), DL-850 (Sanofi-Aventis), GRC-3015, GRC-3566, GRC-3886 (Glenmark), HWA-153 (CAS No. 56395-66-5) (Sanofi-Aventis), hydroxypumafentrine (Altana), IBFB-130011 , IBFB-14-016, IBFB-140301 , IBFB-150007, IBFB-211913 (IBFB Pharma), L-826141 (Merck & Co), medorinone (CAS No. 88296-61-1) (Sanofi-Aventis), MEM-1917 (Memory Pharmaceuticals), ND-1251 (Neuro3d), PDE inhibitors from ICOS, PDE IV inhibitors from Memory Pharmaceuticals and CrystalGenomics, Pharmaprojects No. 2742 and 6141 (Sanofi-Aventis), QAD-171 (Novartis), RHC-2963 (CAS No. 76993-12-9 and 76993-14-1), RPR-117658, RPR-122818 derivatives, SR-24870 , and RPR- 132294 (Sanofi-Aventis), SK-350 (ln2Gen), stroke therapy agents from deCODE Genetics, TAS-203 (Taiho), tofimilast (CAS No. 185954-27-2) (Pfizer), UK-371800 (Pfizer), WIN-65579 (CAS No. 158020-82-7) (Sanofi- Aventis), IBFB-130020 (IBFB Pharma), OPC-6535 (CAS No. 145739-56-6) (Otsuka), a phosphodiesterase III inhibitor, enoximone, a phosphodiesterase IV inhibitor, fosfosal, Atopik (Barrier Therapeutics), triflusal, a phosphodiesterase V inhibitor, and an analogue or derivative thereof.
20821. The method of item 20742 wherein the anti- fibrotic agent is a PKC inhibitor.
20822. The method of item 20742 wherein the anti- fibrotic agent is a platelet activating factor antagonist.
20823. The method of item 20742 wherein the anti- fibrotic agent is a platelet-derived growth factor receptor kinase inhibitor.
20824. The method of item 20742 wherein the anti- fibrotic agent is a prolyl hydroxylase inhibitor.
20825. The method of item 20742 wherein the anti- fibrotic agent is a polymorphonuclear neutrophil inhibitor.
20826. The method of item 20742 wherein the anti- fibrotic agent is a protein kinase B inhibitor.
20827. The method of item 20742 wherein the anti- fibrotic agent is a protein kinase C stimulant.
20828. The method of item 20742 wherein the anti- fibrotic agent is a purine nucleoside analogue.
20829. The method of item 20742 wherein the anti- fibrotic agent is a purinoreceptor P2X antagonist. 20830. The method of item 20742 wherein the anti- fibrotic agent is a Raf kinase inhibitor.
20831. The method of item 20742 wherein the anti- fibrotic agent is a reversible inhibitor of ErbB1 and ErbB2.
20832. The method of item 20742 wherein the anti- fibrotic agent is a ribonucleoside triphosphate reductase inhibitor.
20833. The method of item 20742 wherein the anti- fibrotic agent is an SDF-1 antagonist.
20834. The method of item 20742 wherein the anti- fibrotic agent is a sheddase inhibitor.
20835. The method of item 20742 wherein the anti- fibrotic agent is an SRC inhibitor.
20836. The method of item 20742 wherein the anti- fibrotic agent is a stromelysin inhibitor.
20837. The method of item 20742 wherein the anti- fibrotic agent is an Syk kinase inhibitor.
20838. The method of item 20742 wherein the anti- fibrotic agent is a telomerase inhibitor.
20839. The method of item 20742 wherein the anti- fibrotic agent is a TGF beta inhibitor selected from the group consisting of pirfenidone (CAS No. 53179-13-8) (MARNAC), tranilast (CAS No. 53902- 12-8) (Kissei), IN-1130 (ln2Gen), mannose-6-phosphate (BTG), TGF-β antagonists from Inflazyme (Pharmaprojects No. 6075), TGF-β antagonists from Sydney, non-industrial source), TGF-β I receptor kinase inhibitors from EIi Lilly, TGF-β receptor inhibitors from Johnson & Johnson, and an analogue or derivative thereof.
20840. The method of item 20742 wherein the anti- fibrotic agent is a TNFα antagonist or TACE inhibitor selected from the group consisting of adalimumab (CAS No. 331731-18-1) (Cambridge Antibody Technology), AGIX-4207 (AtheroGenics), AGT-1 (Advanced Biotherapy), an anti-inflammatory from Borean Pharma, Cellzome, or Paradigm Therapeutics, anti-inflammatory vaccine (TNF-alpha kinoid) from Neovacs, humanized anti-TNF antibody or an anti-TNF MAb (CB0006) Celltech (UCB), apratastat (CAS No. 287445-51-0) (Wyeth), BMS-561392 (Bristol-Myers Squibb), BN-006 (Bone), certolizumab pegol (CAS No. 428863-50-7 or CH-138 (UCB), cilomilast (CAS No. 153259-65-5) (GlaxoSmithKline), CR-1 (Nuada Pharmaceuticals), CRx-119 (CombinatoRx), D-5410 (UCB), dacopafant (CAS No. 125372-33-0) (Sanofi- Aventis), dersalazine (CAS No. 188913-57-7/188913-58-8) (Uriach), etanercept (CAS No. 185243-69-0) (Amgen), ethyl pyruvate (Critical (Critical Therapeutics), golimumab (CAS No. 476181-74-5) (Johnson & Johnson), hormono-immunotherapy from Ipsen, CDP571 (e.g., Humicade from UCB), IC-485 (ICOS), infliximab (CAS No. 170277-31-3) (Johnson & Johnson), IP- 751 (Manhattan Pharmaceuticals), ISIS-104838 (CAS No. 250755-32-9) (ISIS Pharmaceuticals), lenalidomide (CAS No. 191732-72-6) (Celgene), lentinan (CAS No. 37339-90-5) (Ajinomoto), MDL-201112 (CAS No. 142130-73-2) (Sanofi-Aventis), medroxyprogesterone (CAS No. 520-85-4) (InKine Pharmaceutical), N-acetylcysteine (CAS No. 616-91-1) (Zambon), NBE-P2 (DIREVO Biotech), nerelimomab (CAS No. 162774-06-3) (Chiron), OM-294DP (OM PHARMA), onercept (CAS No. 199685-57-9) (Yeda), PASSTNF-alpha (Verigen), pentoxifylline or oxypentifylline (Sanofi-Aventis), Pharmaprojects No. 4091, 4241 , 4295, or 4488 (Sanofi-Aventis), Pharmaprojects No. 5480 (Amgen), Pharmaprojects No. 6749 (Cengent), pirfenidone (CAS No. 53179-13-8) (MARNAC), RPR-132294 (Sanofi- Aventis), S5 (F002) (Fulcrum Pharmaceuticals), simvastatin (CAS No. 79902-63-9) (Merck & Co), STA-6292 (Synta Pharmaceuticals), tacrolimus (CAS No. 104987-11-3) (Fujisawa LifeCycle Pharma), talactoferrin alfa (CAS No. 308240-58-6) (Agennix), thalidomide (CAS No. 50-35-1) (Celgene), TNF antagonists form ProStrakan, and Synergen, TNF inhibitors (Amgen), TNF-alpha antagonists from Dynavax Technologies and Jerina AG (Germany), TNF-alpha inhibitors from IBFB Pharma and Xencor (Xencor), torbafylline (CAS No. 105102-21-4) (Sanofi-Aventis), UR-1505 (Uriach), VT-346 (Viron Therapeutics), YSIL6 (Y's Therapeutics), YSTH2 (Y's Therapeutics), NPI-1302a-3 (Nereus Pharmaceuticals, a TNF antagonist from Jerina AG (Germany), dersalazine, and an analogue or derivative thereof.
20841. The method of item 20742 wherein the anti- fibrotic agent is a Toll receptor inhibitor.
20842. The method of item 20742 wherein the anti- fibrotic agent is a tubulin antagonist.
20843. The method of item 20742 wherein the anti- fibrotic agent is a tyrosine kinase inhibitor selected from the group consisting of SU-011248, SUTENT from Pfizer Inc. (New York, NY), BMS-354825, PN- 355 (Paracelsian Pharmaceuticals), AGN-199659 (Allergan), AAL-993 or ABP-309 (Novartis), adaphostin (NIH), AEE-788 (Novartis), AG-013736 (OSI Pharmaceuticals), AG-13736 (Pfizer), ALT-110 (Alteris Therapeutics), AMG-706 (Amgen), anticancer MAbs from Xencor, anti-EGFrvlll MAbs from Abgenix, anti-HER2 MAb from Abiogen, AZD-2171 or AZD-9935 (AstraZeneca), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), CEP-5214 (Cephalon), CEP-7055 (Cephalon), cetuximab (lmClone Systems), CHIR-200131 and CHIR-258 (Chiron), CP-547632 (OSI Pharmaceuticals), CP-724714 (Pfizer), CT-301 (Creabilis Therapeutics), D- 69491 (Baxter International), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EGFR/ErbB2 inhibitors from Array BioPharma, erlotinib (CAS No. 183319- 69-9) (OSI Pharmaceuticals), EXEL-2880 (Exelixis), FK-778 (Sanofi- Aventis) , gefitinib (CAS No. 184475-35-2) (AstraZeneca), GW-2286 or GW- 654652 (GlaxoSmithKline), her2/neu antigen from AlphaVax, HER-2/neu inhibitor from Generex, Herzyme (Medipad) (Sirna Therapeutics), HKI-272 (Wyeth), HuMax-EGFr (Genmab), idronoxil (CAS No. 81267-65-4) (Novogen), IGF-1 inhibitors from Ontogen, IMC-11F8 (ImClone Systems), kahalalide F (CAS No. 149204-42-2) (PharmaMar), KDR inhibitor from LG Life Sciences, KDR inhibitors from Abbott Laboratories, KDR kinase inhibitors (UCB), Kdr kinase inhibitors from Merck & Co, KRN-633 and KRN- 951 (Kirin Brewery), KSB-102 (Xenova), lapatinib ditosylate (CAS No. 388082-78-8) (GlaxoSmithKline), matuzumab (Merck KGaA), MDX-214 (Medarex), ME-103 (Pharmexa), MED-A300 (Gerolymatos), MNAC-13 (Lay Line Genomics), nimotuzumab (Center of Molecular Immunology), NSC- 330507 or NSC-707545 (NIH), NV-50 (Novogen), OSI-930 (OSI Pharmaceuticals), panitumumab (Abgenix), pelitinib (CAS No. 287933-82-7) (Wyeth), pertuzumab (CAS No. 380610-27-5) (Genentech), Pharmaprojects No. 3985 (Sanofi-Aventis), prostate cancer therapeutics from Sequenom (SQPC35, SQPC36, SQPC90), removab and remoxab (Trion Pharma), RG- 13022 (CAS No. 136831-48-6), RG-13291 (CAS No. 138989-50-1), or RG- 14620 (CAS No. 136831-49-7) (Sanofi-Aventis), RM-6427 (Romark), RNAi breast cancer therapy from Benitec, RP 53801 (CAS No. 125882-88-4) (Sanofi-Aventis), sorafenib tosylate (Bayer), SU-11657 (Pfizer), Tie-2 antagonists from Semaia (Hybrigenics), Tie-2 inhibitors from Ontogen, trastuzumab (CAS No. 180288-69-1) (Genentech), tyrosine kinase inhibitors from Sanofi-Aventis, U3-1287, U3-1565, U3-1784, U3-1800 (U3 Pharma), vatalanib (Novartis), VEGFR-2 kinase inhibitor from Bristol-Myers Squibb, XL-647 (Exelixis), ZD-6474 (AstraZeneca), ZK-CDK (Schering AG), an EGFR tyrosine kinase inhibitor, EKB-569 (Wyeth), and an analogue or derivative thereof.
20844. The method of item 20742 wherein the anti- fibrotic agent is a VEGF inhibitor.
20845. The method of item 20742 wherein the anti- fibrotic agent is a vitamin D receptor agonist.
20846. The method of item 20742 wherein the anti- fibrotic agent is ZD-6474 (an angiogenesis inhibitor).
20847. The method of item 20742 wherein the anti- fibrotic agent is AP-23573 (an mTOR inhibitor).
20848. The method of item 20742 wherein the anti- fibrotic agent is synthadotin (a tubulin antagonist).
20849. The method of item 20742 wherein the anti- fibrotic agent is S-0885 (a collagenase inhibitor).
20850. The method of item 20742 wherein the anti- fibrotic agent is aplidine (an elongation factor-1 alpha inhibitor).
20851. The method of item 20742 wherein the anti- fibrotic agent is ixabepilone (an epithilone).
20852. The method of item 20742 wherein the anti- fibrotic agent is IDN-5390 (an angiogenesis inhibitor). 20853. The method of item 20742 wherein the anti- fibrotic agent is SB-2723005 (an angiogenesis inhibitor).
20854. The method of item 20742 wherein the anti- fibrotic agent is ABT-518 (an angiogenesis inhibitor).
20855. The method of item 20742 wherein the anti- fibrotic agent is combretastatin (an angiogenesis inhibitor).
20856. The method of item 20742 wherein the anti- fibrotic agent is anecortave acetate (an angiogenesis inhibitor).
20857. The method of item 20742 wherein the anti- fibrotic agent is SB-715992 (a kinesin antagonist).
20858. The method of item 20742 wherein the anti- fibrotic agent is temsirolimus (an mTOR inhibitor).
20859. The method of item 20742 wherein the anti- fibrotic agent is adalimumab (a TNFα antagonist).
20860. The method of item 20742 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with an anti-fibrotic agent, and (b) implanting the medical device into the host.
20861. The method of item 20742 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with an anti-infective agent, and (b) implanting the medical device into the host. 20862. The method of item 20742 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with a polymer; and (b) implanting the medical device into the host.
20863. The method of item 20742 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with a composition comprising an anti-fibrotic agent and a polymer, and (b) implanting the medical device into the host.
20864. The method of item 20742 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with a composition comprising an anti-infective agent and a polymer, and (b) implanting the medical device into the host.
20865. The method of item 20742 comprising: (a) Infiltrating a tissue of a host where the medical device is to be, or has been, implanted with a composition comprising an anti-fibrotic agent, an anti- infective agent and a polymer, and (b) implanting the medical device into the host.
20866. The method of item 20742 wherein the anti- infective agent is an anthracycline.
20867. The method of item 20742 wherein the anti- infective agent is doxorubicin.
20868. The method of item 20742 wherein the anti- infective agent is is mitoxantrone. 20869. The method of item 20742 wherein the anti- infective agent is a fluoropyrimidine.
20870. The method of item 20742 wherein the anti- infective agent is 5-fluorouracil (5-FU).
20871. The method of item 20742 wherein the anti- infective agent is a folic acid antagonist.
20872. The method of item 20742 wherein the anti- infective agent is methotrexate.
20873. The method of item 20742 wherein the anti- infective agent is a podophylotoxin.
20874. The method of item 20742 wherein the anti- infective agent is etoposide.
20875. The method of item 20742 wherein the anti- infective agent is camptothecin.
20876. The method of item 20742 wherein the anti- infective agent is a hydroxyurea.
20877. The method of item 20742 wherein the anti- infective agent is a platinum complex.
20878. The method of item 20742 wherein the anti- infective agent is cisplatin. 20879. The method of item 20742 wherein the composition comprises an antithrombotic agent.
20880. The method of item 20742 wherein the polymer is formed from reactants comprising a naturally occurring polymer.
20881. The method of item 20742 wherein the polymer is formed from reactants comprising protein.
20882. The method of item 20742 wherein the polymer is formed from reactants comprising carbohydrate.
20883. The method of item 20742 wherein the polymer is formed from reactants comprising biodegradable polymer.
20884. The method of item 20742 wherein the polymer is formed from reactants comprising nonbiodegradable polymer.
20885. The method of item 20742 wherein the polymer is formed from reactants comprising collagen.
20886. The method of item 20742 wherein the polymer is formed from reactants comprising methylated collagen.
20887. The method of item 20742 wherein the polymer is formed from reactants comprising fibrinogen.
20888. The method of item 20742 wherein the polymer is formed from reactants comprising thrombin. 20889. The method of item 20742 wherein the polymer reactants comprising blood plasma.
20890. The method of item 20742 wherein the polymer reactants comprising calcium salt.
20891. The method of item 20742 wherein the polymer reactants comprising an antifibronolytic agent.
20892. The method of item 20742 wherein the polymer reactants comprising fibrinogen analog.
20893. The method of item 20742 wherein the polymer reactants comprising albumin.
20894. The method of item 20742 wherein the polymer reactants comprising plasminogen.
20895. The method of item 20742 wherein the polymer reactants comprising von Willebrands factor.
20896. The method of item 20742 wherein the polymer reactants comprising Factor VIII.
20897. The method of item 20742 wherein the polymer reactants comprising hypoallergenic collagen.
20898. The method of item 20742 wherein the polymer reactants comprising atelopeptidic collagen. 20899. The method of item 20742 wherein the polymer is formed from reactants comprising telopeptide collagen.
20900. The method of item 20742 wherein the polymer is formed from reactants comprising crosslinked collagen.
20901. The method of item 20742 wherein the polymer is formed from reactants comprising aprotinin.
20902. The method of item 20742 wherein the polymer is formed from reactants comprising epsilon-amino-n-caproic acid.
20903. The method of item 20742 wherein the polymer is formed from reactants comprising gelatin.
20904. The method of item 20742 wherein the polymer is formed from reactants comprising protein conjugates.
20905. The method of item 20742 wherein the polymer is formed from reactants comprising gelatin conjugates.
20906. The method of item 20742 wherein the polymer is formed from reactants comprising a synthetic polymer.
20907. The method of item 20742 wherein the polymer is formed from reactants comprising a synthetic isocyanate-containing compound.
20908. The method of item 20742 wherein the polymer is formed from reactants comprising a synthetic thiol-containing compound. 20909. The method of item 20742 wherein the polymer is formed from reactants comprising a synthetic compound containing at least two thiol groups.
20910. The method of item 20742 wherein the polymer is formed from reactants comprising a synthetic compound containing at least three thiol groups.
20911. The method of item 20742 wherein the polymer is formed from reactants comprising a synthetic compound containing at least four thiol groups.
20912. The method of item 20742 wherein the polymer is formed from reactants comprising a synthetic amino-containing compound.
20913. The method of item 20742 wherein the polymer is formed from reactants comprising a synthetic compound containing at least two amino groups.
20914. The method of item 20742 wherein the polymer is formed from reactants comprising a synthetic compound containing at least three amino groups.
20915. The method of item 20742 wherein the polymer is formed from reactants comprising a synthetic compound containing at least four amino groups.
20916. The method of item 20742 wherein the polymer is formed from reactants comprising a synthetic compound comprising a carbonyl-oxygen-succinimidyl group. 20917. The method of item 20742 wherein the polymer is formed from reactants comprising a synthetic compound comprising at least two carbonyl-oxygen-succinimidyl groups.
20918. The method of item 20742 wherein the polymer is formed from reactants comprising a synthetic compound comprising at least three carbonyl-oxygen-succinimidyl groups.
20919. The method of item 20742 wherein the polymer is formed from reactants comprising a synthetic compound comprising at least four carbonyl-oxygen-succinimidyl groups.
20920. The method of item 20742 wherein the polymer is formed from reactants comprising a synthetic polyalkylene oxide- containing compound.
20921. The method of item 20742 wherein the polymer is formed from reactants comprising a synthetic compound comprising both polyalkylene oxide and biodegradable polyester blocks.
20922. The method of item 20742 wherein the polymer is formed from reactants comprising a synthetic polyalkylene oxide- containing compound having reactive amino groups.
20923. The method of item 20742 wherein the polymer is formed from reactants comprising a synthetic polyalkylene oxide- containing compound having reactive thiol groups.
20924. The method of item 20742 wherein the polymer is formed from reactants comprising a synthetic polyalkylene oxide- containing compound having reactive carbonyl-oxygen-succinimidyl groups. 20925. The method of item 20742 wherein the polymer is formed from reactants comprising a synthetic compound comprising a biodegradable polyester block.
20926. The method of item 20742 wherein the polymer is formed from reactants comprising a synthetic polymer formed in whole or part from lactic acid or lactide.
20927. The method of item 20742 wherein the polymer is formed from reactants comprising a synthetic polymer formed in whole or part from glycolic acid or glycolide.
20928. The method of item 20742 wherein the polymer is formed from reactants comprising polylysine.
20929. The method of item 20742 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound comprising a polyalkylene oxide portion.
20930. The method of item 20742 wherein the polymer is formed from reactants comprising (a) protein and (b) polylysine.
20931. The method of item 20742 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound having at least four thiol groups.
20932. The method of item 20742 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound having at least four amino groups. 20933. The method of item 20742 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound having at least four carbonyl-oxygen-succinimide groups.
20934. The method of item 20742 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound having a biodegradable region formed from reactants selected from lactic acid, lactide, glycolic acid, glycolide, and epison-caprolactone.
20935. The method of item 20742 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound comprising a polyalkylene oxide portion.
20936. The method of item 20742 wherein the polymer is formed from reactants comprising (a) collagen and (b) polylysine.
20937. The method of item 20742 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound having at least four thiol groups.
20938. The method of item 20742 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound having at least four amino groups.
20939. The method of item 20742 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound having at least four carbonyl-oxygen-succinimide groups.
20940. The method of item 20742 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound having a biodegradable region formed from reactants selected from lactic acid, lactide, glycolic acid, glycolide, and epison-caprolactone.
20941. The method of item 20742 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound comprising a polyalkylene oxide portion.
20942. The method of item 20742 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) polylysine.
20943. The method of item 20742 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound having at least four thiol groups.
20944. The method of item 20742 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound having at least four amino groups.
20945. The method of item 20742 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound having at least four carbonyl-oxygen-succinimide groups.
20946. The method of item 20742 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound having a biodegradable region formed from reactants selected from lactic acid, lactide, glycolic acid, glycolide, and epison-caprolactone.
20947. The method of item 20742 wherein the polymer is formed from reactants comprising hyaluronic acid. 20948. The method of item 20742 wherein the polymer is formed from reactants comprising a hyaluronic acid derivative.
20949. The method of item 20742 wherein the polymer is formed from reactants comprising pentaerythritol poly(ethylene glycoi)ether tetra-sulfhydryl of number average molecular weight between 3,000 and 30,000.
20950. The method of item 20742 wherein the polymer is formed from reactants comprising pentaerythritol poly(ethylene glycol)ether tetra-amino of number average molecular weight between 3,000 and 30,000.
20951. The method of item 20742 wherein the polymer is formed from reactants comprising (a) a synthetic compound having a number average molecular weight between 3,000 and 30,000 and comprising a polyalkylene oxide region and multiple nucleophilic groups, and (b) a synthetic compound having a number average molecular weight between 3,000 and 30,000 and comprising a polyalkylene oxide region and multiple electrophilic groups.
20952. The method of item 20742 wherein the composition comprises a colorant.
20953. The method of item 20742 wherein the composition is sterile.
20954. A method as in any one of items 20742-20953, wherein the device is an endotracheal tube. 20955. A method as in any one of items 20742-20953, wherein the device is an endotracheal tube with a single lumen.
20956. A method as in any one of items 20742-20953, wherein the device is an endotracheal tube with double lumens.
20957. A method as in any one of items 20742-20953, wherein the device is a tracheostomy tube.
20958. A method for implanting a medical device comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with i) an anti-fibrotic agent, ii) an anti-infective agent, iii) a polymer, iv) a composition comprising an anti-fibrotic agent and a polymer, v) a composition comprising an anti-infective agent and a polymer, or vi) a composition comprising an anti-fibrotic agent, an anti- infective agent and a polymer, and (b) implanting the medical device into the host, wherein the medical device is a peritoneal dialysis catheter.
20959. The method of item 20958 wherein the anti- fibrotic agent is an adensosine A2A receptor antagonist.
20960. The method of item 20958 wherein the anti- fibrotic agent is an AKT inhibitor.
20961. The method of item 20958 wherein the anti- fibrotic agent is an alpha 2 integrin antagonist, wherein the alpha 2 integrin antagonist is Pharmaprojects No. 5754 (Merck KgaA).
20962. The method of item 20958 wherein the anti- fibrotic agent is an alpha 4 integrin antagonist. 20963. The method of item 20958 wherein the anti- fibrotic agent is an alpha 7 nicotinic receptor agonist.
20964. The method of item 20958 wherein the anti- fibrotic agent is an angiogenesis inhibitor selected from the group consisting of AG-12,958 (Pfizer), ATN-161 (Attention LLC), neovastat, an angiogenesis inhibitor from Jerina AG (Germany), NM-3 (Mercian), VGA-1155 (Taisho), FCE-26644 (Pfizer), FCE-26950 (Pfizer), FPMA (Meiji Daries), FR-111142 (Fujisawa), GGTI-298, GM-1306 (Ligand), GPA-1734 (Novartis), NNC-47- 0011 (Novo Nordisk), herbamycin (Nippon Kayaku), lenalidomide (Celegene), IP-10 (NIH), ABT-828 (Abbott), KIN-841 (Tokushima University, Japan), SF-1126 (Semafore Pharmaceuticals), laminin technology (NIH), CHIR-258 (Chiron), NVP-AEW541 (Novartis), NVP-AEW541 (Novartis), Vt16907 (Alchemia), OXI-8007 (Oxigene), EG-3306 (Ark Therapeutics), Maspin (Arriva), ABT-567 (Abbott), PPI-2458 (Praecis Pharmaceuticals), CC-5079, CC-4089 (Celgene), HIF-1 alpha inhibitors (Xenova), S-247 (Pfizer), AP-23573 (Ariad), AZD-9935 (Astra Zeneca), mebendazole (Introgen Therapeutics), MetAP-2 inhibitors (GlaxoSmithKline), AG-615 (Angiogene Pharmaceuticals), Tie-2 antagonists (Hybrigenics), NC-381 , CYC-381, NC-169, NC-219, NC-383, NC-384, NC-407 (Lorus Therapeutics), ATN-224 (Attenuon), ON-01370 (Onconova), Vitronectin antagonists (Amgen), SDX-103 (Salmedix), Vitronectin antagonists (Shire), CHP (Riemser), TEK (Amgen), Anecortave acetate (Alcon), T46.2 (Matrix Therapeutics), HG-2 (Heptagen), TEM antagonists (Genzyme), Oxi-4500 (Oxigene), ATN-161 (Attenuon), WX-293 (Wilex), M-2025 (Metris Therapeutics), Alphastatin (BioActa), YH-16 (Yantai Rongchang), BIBF- 1120 (Boehringer Ingelheim), BAY-57-9352 (Bayer), AS-1404 (Cancer Research Technology), SC-77964 (Pfizer), glycomimetics (BioTie Therapies), TIE-2 Inhibitors (Ontogen), DIMI, Octamer (Octamer), ABR- 215050 (Active Biotech), ABT-518 (Abbott), KDR inhibitors (Abbott), BSF- 466895 (Abbott), SCH-221153 (Schering-Plough), DAC:antiangiogenic (ConjuChem), TFPI (EntreMed), AZD-2171 (Astra-Zenaca), CDC-394 (Celgene), LY290293 (EIi Lilly), IDN-5390 (Indena), Kdr Kinase Inhibitors (Merck), CT-113020, CT-116433, CT-116563, CT-31890, CT-32228) (Cell Therapeutics), A-299620 (Abbott), TWEAK Inhibitor (Amgen), VEGF modulators (Johnson and Johnson), Tum-N53, tumstatin (Genzyme), Thios- 1 , Thios-2 (Thios Pharmaceuticals), MV-6401 (Miravant Medical Technologies), Spisulosine (PharmaMar), CEP-7055 (Cephalon), AUV-201 (Auvation), LM-609 (EIi Lilly), SKF-106615 (AnorMED), Oglufanide disodium (Cytran), BW-114 (Pharminox), Calreticulin (NIH), WX-678 (Wilex), SD-7784 (Pfizer), WX-UK1 (Wilex), SH-268 (Schering AG), 2-Me-PGA (Celgene), S- 137 (Pfizer), ZD-6126 (Angiogene Pharmaceuticals), SG-292 (SignalGen), Benefin (Lane Labs), A6, A36 (Angstrom), SB-2723005 (GlaxoSmithKline), SC-7 (Cell Therapeutics), ZEN-014 (AEterna Zentaris), 2-methoxyestradiol (EntreMed), NK-130119 (Nippon Kayaku), CC-10004 (Celgene), AVE- 8062A (Ajinomoto), Tacedinaline (Pfizer), Actinonin (Tokyo Metropolitan Institute of Medical Science), Lenalidomide (Celgene), VGA-1155, BTO-956 (SRI International), ER-68203-00 (Eisai), CT-6685 (UCB), JKC-362 (Phoenix Pharmaceuticals), DMI-3798 (DMI Biosciences, Angiomate (Ipsen), ZD-6474 (AstraZeneca), CEP-5214 (Cephalon), Canstatin (Genzyme), NM-3 (Mercian), Oridigm (MediQuest Therapeutics), Exherin (Adherex), BLS-0597 (Boston Life Sciences), PTC-299 (PTC Therapeutics), NPI-2358 (Nereus Pharmaceuticals), CGP-79787 (Novartis), AEE-788 (Novartis), CKD-732 (Chong Kun Dang), CP-564959 (OSI Pharmaceuticals), CM-101 (CarboMed), CT-2584, CT3501 (Cell Therapeutics), combretastatin and analogues and derivatives thereof (Oxigene), Rebimastat (Bristol-Meyers Squibb), Dextrin 2-sulfate (ML Laboratories), Cilengitide (Merk KGaA), NSC-706704 (Pharminox), KRN- 951 (Kirin Brewery), Ukrain, NSC-631570 (Nowicky Pharma), Tecogalan sodium (Daiichi Pharmaceutical), Tz-93 (Tsumura), TBC-1635 (Encysive Pharmaceuticals), TAN-1120 (Takeda), Semaxanib (Pfizer), BDI-7800 (Biopharmacopae), SD-186, SD-983 (Bristol-Meyers Squibb), SB-223245 (GlaxoSmithKline), SC-236 (Pfizer), RWJ-590973 (Johnson and Johnson), ILX-1850 (Genzyme), SC-68488, S-836 (Pfizer), CG-55069-11 (CuraGen), Ki-23057 (Kirin Brewery), CCX-700 (Chemoentryx), Pegaptanib octasodium (Giled Sciences), ANGIOCOL (available from Biostratum Inc.), or an analogue or derivative thereof.
20965. The method of item 20958 wherein the anti- fibrotic agent is an apoptosis antagonist.
20966. The method of item 20958 wherein the anti- fibrotic agent is an apoptosis activator.
20967. The method of item 20958 wherein the anti- fibrotic agent is a beta 1 integrin antagonist.
20968. The method of item 20958 wherein the anti- fibrotic agent is a beta tubulin inhibitor.
20969. The method of item 20958 wherein the anti- fibrotic agent is a blocker of enzyme production in Hepatitis C.
20970. The method of item 20958 wherein the anti- fibrotic agent is a Bruton's tyrosine kinase inhibitor.
20971. The method of item 20958 wherein the anti- fibrotic agent is a calcineurin inhibitor.
20972. The method of item 20958 wherein the anti- fibrotic agent is a caspase 3 inhibitor. 20973. The method of item 20958 wherein the anti- fibrotic agent is a CC chemokine receptor antagonist.
20974. The method of item 20958 wherein the anti- fibrotic agent is a cell cycle inhibitor selected from the group consisting of SNS-595 (Sunesis), synthadotin, KRX-0403, and an analogue or derivative thereof.
20975. The method of item 20958 wherein the anti- fibrotic agent is a cathepsin B inhibitor.
20976. The method of item 20958 wherein the anti- fibrotic agent is a cathepsin K inhibitor, wherein the cathepsin K inhibitor is 462795 (GlaxoSmithKline), INPL-022-D6 (Amura Therapeutics), or an analogue or derivative thereof.
20977. The method of item 20958 wherein the anti- fibrotic agent is a cathepsin L inhibitor.
20978. The method of item 20958 wherein the anti- fibrotic agent is a CD40 antagonist.
20979. The method of item 20958 wherein the anti- fibrotic agent is a chemokine receptor agonist.
20980. The method of item 20958 wherein the anti- fibrotic agent is a chymase inhibitor.
20981. The method of item 20958 wherein the anti- fibrotic agent is a collagenase antagonist. 20982. The method of item 20958 wherein the anti- fibrotic agent is a CXCR antagonist.
20983. The method of item 20958 wherein the anti- fibrotic agent is a cyclin dependent kinase inhibitor selected from the group consisting of a CDK-1 inhibitor, a CDK-2 inhibitor, a CDK- 4 inhibitor, a CDK-6 inhibitor, a CAK1 inhibitor from GPC Biotech and Bristol-Myers Squibb, RGB-286199 (GPC Biotech), an anticancer agent from Astex Technology, a CAK1 inhibitor from GPC Biotech, a CDK inhibitor from Sanofi-Aventis, a CDK1/CDK2 inhibitor from Amgen, a CDK2 inhibitor from SUGEN-2 (Pfizer), a hearing loss therapy agent (Sound Pharmaceuticals), PD-0332991 (Pfizer), RGB-286199 (GPC Biotech), Ro-0505124 (Hoffmann- La Roche), a Ser/Thr kinase inhibitor from Lilly (EIi Lilly), CVT-2584 (CAS No. 199986-75-9) (CV Therapeutics), and an analogue or derivative thereof.
20984. The method of item 20958 wherein the anti- fibrotic agent is a cyclooxygenase 1 inhibitor.
20985. The method of item 20958 wherein the anti- fibrotic agent is a DHFR inhibitor.
20986. The method of item 20958 wherein the anti- fibrotic agent is a dual integrin inhibitor.
20987. The method of item 20958 wherein the anti- fibrotic agent is an elastase inhibitor.
20988. The method of item 20958 wherein the anti- fibrotic agent is an elongation factor-1 alpha inhibitor. 20989. The method of item 20958 wherein the anti- fibrotic agent is an endothelial growth factor antagonist.
20990. The method of item 20958 wherein the anti- fibrotic agent is an endothelial growth factor receptor kinase inhibitor selected from the group consisting of sorafenib tosylate (Bayer), AAL-993 (Novartis), ABP-309 (Novartis), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EXEL- 2880 (Exelixis), GW-654652 (GlaxoSmithKline), a KDR inhibitor from LG Life Sciences, CT-6685 and CT-6729 (UCB), KRN-633 and KRN-951 (Kirin Brewery), OSI-930 (OSI Pharmaceuticals), SP-5.2 (Supratek Pharma), SU- 11657 (Pfizer), a Tie-2 antagonist (Hybrigenics), a VEGFR-2 kinase inhibitor (Bristol-Myers Squibb), XL-647 (Exelixis), a KDR inhibitor from Abbott Laboratories, sorafenib tosylate, and an analogue or derivative thereof.
20991. The method of item 20958 wherein the anti- fibrotic agent is an endotoxin antagonist.
20992. The method of item 20958 wherein the anti- fibrotic agent is an epothilone and tubulin binder.
20993. The method of item 20958 wherein the anti- fibrotic agent is an estrogen receptor antagonist.
20994. The method of item 20958 wherein the anti- fibrotic agent is an FGF inhibitor.
20995. The method of item 20958 wherein the anti- fibrotic agent is a famexyl transferase inhibitor. 20996. The method of item 20958 wherein the anti- fibrotic agent is famesyltransferase inhibitor selected from the group of A- 197574 (Abbott), a famesyltransferase inhibitor from Servier, FPTIII (Strathclyde Institute for Drug R), LB-42908 (LG Life Sciences), Pharmaprojects No. 5063 (Genzyme), Pharmaprojects No. 5597 (Ipsen), Yissum Project No. B-1055 (Yissum), and an analogue or derivative thereof.
20997. The method of item 20958 wherein the anti- fibrotic agent is an FLT-3 kinase inhibitor.
20998. The method of item 20958 wherein the anti- fibrotic agent is an FGF receptor kinase inhibitor.
20999. The method of item 20958 wherein the anti- fibrotic agent is a fibrinogen antagonist selected from the group consisting of AUV-201 (Auvation), MG-13926 (Sanofi-Aventis), plasminogen activator (CAS No. 105913-11-9) (from Sanofi-Aventis or UCB), plasminogen activator-2 (tPA-2) (Sanofi-Aventis), pro-urokinase (CAS No. 82657-92-9) (Sanofi-Aventis), and an analogue or derivative thereof.
21000. The method of item 20958 wherein the anti- fibrotic agent is a heat shock protein 90 antagonist selected from the group consisting of SRN-005 (Sirenade), geldanamycin, NSC-33050 (17- allylaminogeldanamycin; 17-AAG), 17-dimethylaminoethylamino-17- demethoxy-geldanamycin (17-DMAG), rifabutin (rifamycin XIV, 1',4- didehydro-1-deoxy-1 ,4-dihydro-5'-(2-methylpropyl)-1-oxo-), and an analogue or derivative thereof.
21001. The method of item 20958 wherein the anti- fibrotic agent is a histone deacetylase inhibitor. 21002. The method of item 20958 wherein the anti- fibrotic agent is an HMGCoA reductase inhibitor selected from the group consisting of an atherosclerosis therapeutic from Lipid Sciences, ATI-16000 (ARYx Therapeutics), KS-01-019 (Kos Pharmaceuticals), Pharmaprojects No. 2197 (Sanofi-Aventi), RP 61969 (Sanofi-Aventis), and an analogue or derivative thereof.
21003. The method of item 20958 wherein the anti- fibrotic agent is an ICAM inhibitor.
21004. The method of item 20958 wherein the anti- fibrotic agent is an IL, ICE and IRAK antagonist, wherein the antagonist is a CJ-14877, CP-424174 (Pfizer), NF-61 (Negma-Lerads), and an analogue or derivative thereof.
21005. The method of item 20958 wherein the anti- fibrotic agent is an IL-2 inhibitor.
21006. The method of item 20958 wherein the anti- fibrotic agent is an immunosuppressant selected from the group consisting of teriflunomide (Sanofi Aventis), chlorsulfaquinoxalone (NSC-339004), chlorsulfaquinoxalone sulfate, CS-712 (Sankyo), ismomultin alfa (CAS No. 457913-93-8) (Akzo Nobel), antiallergics from GenPat77, antiinflammatories or AT-005 (Androclus Therapeutics), autoimmune disease therapeutics from EpiVax, BN-007 (Bone), budesonide (CAS No. 51333-22- 3) (MAP Pharmaceuticals), CO-14 (Genzyme), edratide (CAS No. 433922- 67-9) (Teva), EP-314 (Enanta), eprovafen (CAS No. 101335-99-3) (Sanofi- Aventis), HWA-131 (CAS No. 118788-41-3) (Sanofi-Aventis), immunomodulators from MerLion Pharmaceuticals, immunosuppressives from Alchemia, IPL-12 (Inflazyme), MDL-9563 (CAS No. 27086-86-8) (Sanofi-Aventis), Pharmaprojects No. 2330 (Sanofi-Aventis), Pharmaprojects No. 6426 (Abgenix), PXS-25 (Pharmaxis), rosmarinic acid (CAS No. 20283-92-5) (Sanofi-Aventis), RP 42927 or RP 54745 (CAS No. 135330-08-4) (Sanofi-Aventis), SGN-35 (Seattle Genetics), ST-1959 (Sigma-Tau), type I diabetes therapy from SYNX Pharma, UNIL-88 (Debiopharm), VP-025 (Vasogen), VR-694 (Vectura), PRTX-001 (Protaiex), and an analogue or derivative thereof.
21007. The method of item 20958 wherein the anti- fibrotic agent is an IMPDH (inosine monophosphate).
21008. The method of item 20958 wherein the anti- fibrotic agent is an integrin antagonist.
21009. The method of item 20958 wherein the anti- fibrotic agent is an interleukin antagonist.
21010. The method of item 20958 wherein the anti- fibrotic agent is an inhibitor of type III receptor tyrosine kinase.
21011. The method of item 20958 wherein the anti- fibrotic agent is an irreversible inhibitor of enzyme methionine aminopeptidase type 2.
21012. The method of item 20958 wherein the anti- fibrotic agent is an isozyme selective delta protein kinase C inhibitor.
21013. The method of item 20958 wherein the anti- fibrotic agent a JAK3 enzyme inhibitor.
21014. The method of item 20958 wherein the anti- fibrotic agent is a JNK inhibitor. 21015. The method of item 20958 wherein the anti- fibrotic agent is a kinase inhibitor.
21016. The method of item 20958 wherein the anti- fibrotic agent is kinesin antagonist.
21017. The method of item 20958 wherein the anti- fibrotic agent is a kinesin antagonist.
21018. The method of item 20958 wherein the anti- fibrotic agent is a leukotriene inhibitor and antagonist selected from the group consisting of ambicromil (CAS No. 58805-38-2) (Sanofi-Aventis), amelubant (CAS No. 346735-24-8) (Boehringer Ingelheim), DW-1141 (Dong Wha), ebselen (Daiichi Pharmaceutical), ibudilast (Kyorin), leucotriene inhibitors from Sanofi-Aventis, lymphotoxin-beta receptor (LT-D) from Biogen Idee, Pharmaprojects No. 1535 and 2728 (CAS No. 119340-33-9) (Sanofi-Aventis), R-112 (Rigeϊ), Rev-5367 (CAS No. 92532-05-3) (Sanofi- Aventis), RG-14893 (CAS No. 141835-49-6) (Sanofi-Aventis), RG-5901-A (CAS No. 101910-24-1), 92532-23-5, RP 66153 (CAS No. 142422-79-5), RP 66364 (CAS No.186916-92-5), RP 69698 (CAS No. 141748-00-7) (Sanofi-Aventis), SC-411930 (Pfizer), SC-41930 (CAS No. 120072-59-5) (Pfizer), SC-50605 (CAS No. 138828-39-4) (Pfizer), SC-51146 (CAS No. 141059-52-1), SC-53228 (CAS No. 153633-01-3) (Pfizer), spaglumic acid (ZY-15106) (CAS No.3106-85-2), 80619-64-3 (Novartis), tipredane (CAS No. 85197-77-9) (Bristol-Myers Squibb), U-75302 (CAS No. 119477-85-9) (Pfizer), and analogue or derivative thereof.
21019. The method of item 20958 wherein the anti- fibrotic agent is an MAP kinase inhibitor. 21020. The method of item 20958 wherein the anti- fibrotic agent is a matrix metalloproteinase inhibitor.
21021. The method of item 20958 wherein the anti- fibrotic agent is an MCP-CCR2 inhibitor.
21022. The method of item 20958 wherein the anti- fibrotic agent is an mTOR inhibitor.
21023. The method of item 20958 wherein the anti- fibrotic agent is an mTOR kinase inhibitor.
21024. The method of item 20958 wherein the anti- fibrotic agent is a microtubule inhibitor selected from the group consisting of antibody-maytansinoid conjugates from Biogen Idee, colchicines (MantiCore Pharmaceuticals), anticancer immunoconjugates from Johnson & Johnson, DIME from Octamer, gni-1f (GNI), huC242-DM4, huMy9-6-DM1 (ImmunoGen), IDN-5404 (Indena), IMO-098, IMOderm (Imotep), mebendazole (Introgen Therapeutics), microtubule poisons from Cambridge Enterprise, paclitaxel such as LOTAX from Aphios (CAS No. 33069-62-4), Genexol-PM from Samyang, Pharmaprojects No. 6383 (Tapestry Pharmaceuticals), RPR-112378 (Sanofi-Aventis), SGN-75 (Seattle Genetics), SPL-7435 (Starpharma), SSR-250411 (Sanofi-Aventis), trastuzumab-DM1 (Genentech), vinorelbine, and an analogue or derivative thereof.
21025. The method of item 20958 wherein the anti- fibrotic agent is an MlF inhibitor.
21026. The method of item 20958 wherein the anti- fibrotic agent is an MMP inhibitor. 21027. The method of item 20958 wherein the anti- fibrotic agent is a neurokinin (NK) antagonist selected from the group consisting of anthrotainin (CAS No. 148084-40-6) (Sanofi-Aventis), an IBS thereapeutic from ArQuIe, MDL-105212A (CAS No. 167261-60-1) (Ssanofi- Aventis), Pharmaprojects No. 2744, 3258 (CAS No. 139167-47-8) 4006, 4201 , or 5986 (Sanofi-Aventis), RP 67580 (CAS No. 135911-02-3), SR- 144190 (CAS No. 201152-86-5), SSR-240600, SSR-241586 (Sanofi- Aventis), TKA-457 (Novartis), vestipitant mesylate (CAS No. 334476-64-1) (GlaxoSmithKline), Win-64821 (Sanofi-Aventis), PRX-96026 (Predix Pharmaceuticals), and an analogue or derivative thereof.
21028. The method of item 20958 wherein the anti- fibrotic agent is an NF kappa B inhibitor selected from the group consisting of AVE-0545 or AVE-0547 (Sanofi-Aventis), bortezomib (CAS No. 179324- 69-7) (Millennium Pharmaceuticals), dexanabinol (CAS No. 112924-45-5) (Pharmos), dexlipotam (Viatris), Pharmaprojects No. 6283 (INDRA) (OXiGENE), IPL-576092 (CAS No. 137571-30-3) (Inflazyme), NFKB decoy (Corgentech), NFKB decoy oligo (AnGes MG), NFKB's from Ariad, osteoporosis treatments or S5 (F005) from Fulcrum Pharmaceuticals, P61 (Phytopharm), R-flurbiprofen (CAS No. 5104-49-4) (Encore Pharmaceuticals), and an analogue or derivative thereof.
21029. The method of item 20958 wherein the anti- fibrotic agent is a nitric oxide agonist.
21030. The method of item 20958 wherein the anti- fibrotic agent is an ornithine decarboxylase inhibitor.
21031. The method of item 20958 wherein the anti- fibrotic agent is a p38 MAP kinase inhibitor selected from the group consisting of AZD-6703 (AstraZeneca), JX-401 (Jexys Pharmaceuticals), BMS-2 (Bristol-Myers Squibb), a p38 MAP kinase inhibitor from Novartis, a p38-alpha MAP kinase inhibitor from Amphora, Pharmaprojects No. 5704 (Pharmacopeia), RPR-200765A (Sanofi-Aventis), SD-282 (Johnson & Johnson), TAK-715 (Takeda), and an analogue or derivative thereof.
21032. The method of item 20958 wherein the anti- fibrotic agent is a palmitoyl-protein thioesterase inhibitor.
21033. The method of item 20958 wherein the anti- fibrotic agent is a PDGF receptor kinase inhibitor selected from the group consisting of AAL-993, AMN-107, or ABP-309 (Novartis), AMG-706 (Amgen), BAY-57-9352 (Bayer), CDP-860 (UCB)1 E-7080 (Eisai), imatinib (CAS No. 152459-95-5) (Novartis), OSI-930 (OSI Pharmaceuticals), RPR- 127963E (Sanofi-Aventis), RWJ-540973 (Johnson & Johnson), sorafenib tosylate (Bayer), SU-11657 (Pfizer), tandutinib (CAS No. 387867-13-2) (Millennium Pharmaceuticals), vatalanib (Novartis), ZK-CDK (Schering AG), and an analogue or derivative thereof.
21034. The method of item 20958 wherein the anti- fibrotic agent is (-)-halofenate (Metabolex), AMG-131 (Amgen), antidiabetics from Japan Tobacco, AZD-4619, AZD-8450, AZD-8677 (AstraZeneca), DRF-10945, balaglitazone (Dr Reddy's), CS-00088, CS-00098 (Chipscreen Biosciences), E-3030 (Eisai), etalocib (CAS No. 161172-51-6) (EIi Lilly), GSK-641597 (Ligand), GSK-677954 (GlaxoSmithKline), GW-409544 (Ligand), GW-590735 (GlaxoSmithKline), K-111 (Hoffmann-La Roche), LY- 518674 (EIi Lilly), LY-674 (Ligand), LY-929 (Ligand), MC-3001, MC-3002 (MaxoCore Pharmaceuticals), metformin HCI + pioglitazone (CAS No. 1115- 70-4 and 112529-15-4), ACTOPLUS MET from Andrx), muraglitazar (CAS No. 331741-94-7) (Bristol-Myers Squibb), naveglitazar (Ligand), oleoylethanolamide (Kadmus Pharmaceuticals), ONO-5129, pioglitazone hydrochloride (CAS No. 111025-46-8 and 112529-15-4) (Takeda), PLX-204 (Plexxikon), PPAR agonists from Genfit, PPAR delta agonists from EIi Lilly, PPAR-alpha agonists from CrystalGenomics, PPAR-gamma modulators and PPAR-β modulators from CareX, rosiglitazone maleate (CAS No. 122324- 73-4 or 155141-29-0) (GlaxoSrnithKline), rosiglitazone maleate/glimepir (CAS No. 155141-29-0 and 93479-97-1), AVANDARYL1 rosiglitazone maleate/metformin extend (CAS No. 155141-29-0 and 657-24-9), AVANDAMET, rosiglitazone maleate+metformin, AVANDAMET (GlaxoSmithKline), tesaglitazar (AstraZeneca), and an analogue or derivative thereof.
21035. The method of item 20958 wherein the anti- fibrotic agent is a phosphatase inhibitor.
21036. The method of item 20958 wherein the anti- fibrotic agent is a phosphodiesterase (PDE) inhibitor selected from the group consisting of avanafil (Tanabe Seiyaku), dasantafil (CAS No. 569351- 91-3) (Schering-Plough), A-906119 (CAS No. 134072-58-5), DL-850 (Sanofi-Aventis), GRC-3015, GRC-3566, GRC-3886 (Glenmark), HWA-153 (CAS No. 56395-66-5) (Sanofi-Aventis), hydroxypumafentrine (Altana), IBFB-130011 , IBFB-14-016, IBFB-140301 , IBFB-150007, IBFB-211913 (IBFB Pharma), L-826141 (Merck & Co), medorinone (CAS No. 88296-61-1) (Sanofi-Aventis), MEM-1917 (Memory Pharmaceuticals), ND-1251 (Neuro3d), PDE inhibitors from ICOS, PDE IV inhibitors from Memory Pharmaceuticals and CrystalGenomics, Pharmaprojects No. 2742 and 6141 (Sanofi-Aventis), QAD-171 (Novartis), RHC-2963 (CAS No. 76993-12-9 and 76993-14-1), RPR-117658, RPR-122818 derivatives, SR-24870 , and RPR- 132294 (Sanofi-Aventis), SK-350 (ln2Gen), stroke therapy agents from deCODE Genetics, TAS-203 (Taiho), tofimilast (CAS No. 185954-27-2) (Pfizer), UK-371800 (Pfizer), WIN-65579 (CAS No. 158020-82-7) (Sanofi- Aventis), IBFB-130020 (IBFB Pharma), OPC-6535 (CAS No. 145739-56-6) (Otsuka), a phosphodiesterase III inhibitor, enoximone, a phosphodiesterase IV inhibitor, fosfosal, Atopik (Barrier Therapeutics), triflusal, a phosphodiesterase V inhibitor, and an analogue or derivative thereof.
21037. The method of item 20958 wherein the anti- fibrotic agent is a PKC inhibitor.
21038. The method of item 20958 wherein the anti- fibrotic agent is a platelet activating factor antagonist.
21039. The method of item 20958 wherein the anti- fibrotic agent is a platelet-derived growth factor receptor kinase inhibitor.
21040. The method of item 20958 wherein the anti- fibrotic agent is a prolyl hydroxylase inhibitor.
21041. The method of item 20958 wherein the anti- fibrotic agent is a polymorphonuclear neutrophil inhibitor.
21042. The method of item 20958 wherein the anti- fibrotic agent is a protein kinase B inhibitor.
21043. The method of item 20958 wherein the anti- fibrotic agent is a protein kinase C stimulant.
21044. The method of item 20958 wherein the anti- fibrotic agent is a purine nucleoside analogue.
21045. The method of item 20958 wherein the anti- fibrotic agent is a purinoreceptor P2X antagonist. 21046. The method of item 20958 wherein the anti- fibrotic agent is a Raf kinase inhibitor.
21047. The method of item 20958 wherein the anti- fibrotic agent is a reversible inhibitor of ErbB1 and ErbB2.
21048. The method of item 20958 wherein the anti- fibrotic agent is a ribonucleoside triphosphate reductase inhibitor.
21049. The method of item 20958 wherein the anti- fibrotic agent is an SDF-1 antagonist.
21050. The method of item 20958 wherein the anti- fibrotic agent is a sheddase inhibitor.
21051. The method of item 20958 wherein the anti- fibrotic agent is an SRC inhibitor.
21052. The method of item 20958 wherein the anti- fibrotic agent is a stromelysin inhibitor.
21053. The method of item 20958 wherein the anti- fibrotic agent is an Syk kinase inhibitor.
21054. The method of item 20958 wherein the anti- fibrotic agent is a telomerase inhibitor.
21055. The method of item 20958 wherein the anti- fibrotic agent is a TGF beta inhibitor selected from the group consisting of pirfenidone (CAS No. 53179-13-8) (MARNAC), tranilast (CAS No. 53902- 12-8) (Kissei), IN-1130 (ln2Gen), mannose-6-phosphate (BTG), TGF-β antagonists from Inflazyme (Pharmaprojects No. 6075), TGF-β antagonists from Sydney, non-industrial source), TGF-β I receptor kinase inhibitors from EIi Lilly, TGF-β receptor inhibitors from Johnson & Johnson, and an analogue or derivative thereof.
21056. The method of item 20958 wherein the anti- fibrotic agent is a TNFα antagonist or TACE inhibitor selected from the group consisting of adalimumab (CAS No. 331731-18-1) (Cambridge Antibody Technology), AGIX-4207 (AtheroGenics), AGT-1 (Advanced Biotherapy), an anti-inflammatory from Borean Pharma, Cellzome, or Paradigm Therapeutics, anti-inflammatory vaccine (TNF-alpha kinoid) from Neovacs, humanized anti-TNF antibody or an anti-TNF MAb (CB0006) Celltech (UCB), apratastat (CAS No. 287445-51-0) (Wyeth), BMS-561392 (Bristol-Myers Squibb), BN-006 (Bone), certolizumab pegol (CAS No. 428863-50-7 or CH-138 (UCB), cilomilast (CAS No. 153259-65-5) (GlaxoSmith Kline), CR-1 (Nuada Pharmaceuticals), CRx-119 (CombinatoRx), D-5410 (UCB), dacopafant (CAS No. 125372-33-0) (Sanofi- Aventis), dersalazine (CAS No. 188913-57-7/188913-58-8) (Uriach), etanercept (CAS No. 185243-69-0) (Amgen), ethyl pyruvate (Critical (Critical Therapeutics), golimumab (CAS No. 476181-74-5) (Johnson & Johnson), hormono-immunotherapy from Ipsen, CDP571 (e.g., Humicade from UCB), IC-485 (ICOS), infliximab (CAS No. 170277-31-3) (Johnson & Johnson), IP- 751 (Manhattan Pharmaceuticals), ISIS-104838 (CAS No. 250755-32-9) (ISIS Pharmaceuticals), lenalidomide (CAS No. 191732-72-6) (Celgene), lentinan (CAS No. 37339-90-5) (Ajinomoto), MDL-201112 (CAS No. 142130-73-2) (Sanofi-Aventis), medroxyprogesterone (CAS No. 520-85-4) (InKine Pharmaceutical), N-acetylcysteine (CAS No. 616-91-1) (Zambon), NBE-P2 (DIREVO Biotech), nerelimomab (CAS No. 162774-06-3) (Chiron), OM-294DP (OM PHARMA), onercept (CAS No. 199685-57-9) (Yeda), PASSTNF-alpha (Verigen), pentoxifylline or oxypentifylline (Sanofi-Aventis), Pharmaprojects No. 4091 , 4241 , 4295, or 4488 (Sanofi-Aventis), Pharmaprojects No. 5480 (Amgen), Pharmaprojects No. 6749 (Cengent), pirfenϊdone (CAS No. 53179-13-8) (MARNAC), RPR-132294 (Sanofi- Aventis), S5 (F002) (Fulcrum Pharmaceuticals), simvastatin (CAS No. 79902-63-9) (Merck & Co), STA-6292 (Synta Pharmaceuticals), tacrolimus (CAS No. 104987-11-3) (Fujisawa LifeCycle Pharma), talactoferrin alfa (CAS No. 308240-58-6) (Agennix), thalidomide (CAS No. 50-35-1) (Celgene), TNF antagonists form ProStrakan, and Synergen, TNF inhibitors (Amgen), TNF-alpha antagonists from Dynavax Technologies and Jerina AG (Germany), TNF-alpha inhibitors from IBFB Pharma and Xencor (Xencor), torbafylline (CAS No. 105102-21-4) (Sanofi-Aventis), UR-1505 (Uriach), VT-346 (Viron Therapeutics), YSIL6 (Y1S Therapeutics), YSTH2 (Y's Therapeutics), NPI-1302a-3 (Nereus Pharmaceuticals, a TNF antagonist from Jerina AG (Germany), dersalazine, and an analogue or derivative thereof.
21057. The method of item 20958 wherein the anti- fibrotic agent is a Toll receptor inhibitor.
21058. The method of item 20958 wherein the anti- fibrotic agent is a tubulin antagonist
21059. The method of item 20958 wherein the anti- fibrotic agent is a tyrosine kinase inhibitor selected from the group consisting of SU-011248, SUTENT from Pfizer Inc. (New York, NY), BMS-354825, PN- 355 (Paracelsian Pharmaceuticals), AGN-199659 (Allergan), AAL-993 or ABP-309 (Novartis), adaphostin (NIH), AEE-788 (Novartis), AG-013736 (OSI Pharmaceuticals), AG-13736 (Pfizer), ALT-110 (Alteris Therapeutics), AMG-706 (Amgen), anticancer MAbs from Xencor, anti-EGFrvlll MAbs from Abgenix, anti~HER2 MAb from Abiogen, AZD-2171 or AZD-9935 (AstraZeneca), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), CEP-5214 (Cephalon), CEP-7055 (Cephalon), cetuximab (ImClone Systems), CHIR-200131 and CHIR-258 (Chiron), CP-547632 (OSI Pharmaceuticals), CP-724714 (Pfizer), CT-301 (Creabilis Therapeutics), D- 69491 (Baxter International), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EGFR/ErbB2 inhibitors from Array BioPharma, erlotinib (CAS No. 183319- 69-9) (OSI Pharmaceuticals), EXEL-2880 (Exelixis), FK-778 (Sanofi- Aventis) , gefitinib (CAS No. 184475-35-2) (AstraZeneca), GW-2286 or GW- 654652 (GlaxoSmithKline), her2/neu antigen from AlphaVax, HER-2/neu inhibitor from Generex, Herzyme (Medipad) (Sima Therapeutics), HKI-272 (Wyeth), HuMax-EGFr (Genmab), idronoxil (CAS No. 81267-65-4) (Novogen), IGF-1 inhibitors from Ontogen, IMC-11F8 (ImClone Systems), kahalalide F (CAS No. 149204-42-2) (PharmaMar), KDR inhibitor from LG Life Sciences, KDR inhibitors from Abbott Laboratories, KDR kinase inhibitors (UCB), Kdr kinase inhibitors from Merck & Co, KRN-633 and KRN- 951 (Kirin Brewery), KSB-102 (Xenova), lapatinib ditosylate (CAS No. 388082-78-8) (GlaxoSmithKline), matuzumab (Merck KGaA), MDX-214 (Medarex), ME-103 (Pharmexa), MED-A300 (Gerolymatos), MNAC-13 (Lay Line Genomics), nimotuzumab (Center of Molecular Immunology), NSC- 330507 or NSC-707545 (NIH), NV-50 (Novogen), OSI-930 (OSI Pharmaceuticals), panitumumab (Abgenix), pelitinib (CAS No. 287933-82-7) (Wyeth), pertuzumab (CAS No. 380610-27-5) (Genentech), Pharmaprojects No. 3985 (Sanofi-Aventis), prostate cancer therapeutics from Sequenom (SQPC35, SQPC36, SQPC90), removab and remoxab (Trion Pharma), RG- 13022 (CAS No. 136831-48-6), RG-13291 (CAS No. 138989-50-1), or RG- 14620 (CAS No. 136831-49-7) (Sanofi-Aventis), RM-6427 (Romark), RNAi breast cancer therapy from Benitec, RP 53801 (CAS No. 125882-88-4) (Sanofi-Aventis), sorafenib tosylate (Bayer), SU-11657 (Pfizer), Tie-2 antagonists from Semaia (Hybrigenics), Tie-2 inhibitors from Ontogen, trastuzumab (CAS No. 180288-69-1 ) (Genentech), tyrosine kinase inhibitors from Sanofi-Aventis, U3-1287, U3-1565, U3-1784, U3-1800 (U3 Pharma), vatalanib (Novartis), VEGFR-2 kinase inhibitor from Bristol-Myers Squibb, XL-647 (Exelixis), ZD-6474 (AstraZeneca), ZK-CDK (Schering AG), an EGFR tyrosine kinase inhibitor, EKB-569 (Wyeth), and an analogue or derivative thereof.
21060. The method of item 20958 wherein the anti- fibrotic agent is a VEGF inhibitor.
21061. The method of item 20958 wherein the anti- fibrotic agent is a vitamin D receptor agonist.
21062. The method of item 20958 wherein the anti- fibrotic agent is ZD-6474 (an angiogenesis inhibitor).
21063. The method of item 20958 wherein the anti- fibrotic agent is AP-23573 (an mTOR inhibitor).
21064. The method of item 20958 wherein the anti- fibrotic agent is synthadotin (a tubulin antagonist).
21065. The method of item 20958 wherein the anti- fibrotic agent is S-0885 (a collagenase inhibitor).
21066. The method of item 20958 wherein the anti- fibrotic agent is aplidine (an elongation factor-1 alpha inhibitor).
21067. The method of item 20958 wherein the anti- fibrotic agent is ixabepilone (an epithilone).
21068. The method of item 20958 wherein the anti- fibrotic agent is IDN-5390 (an angiogenesis inhibitor). 21069. The method of item 20958 wherein the anti- fibrotic agent is SB-2723005 (an angiogenesis inhibitor).
21070. The method of item 20958 wherein the anti- fibrotic agent is ABT-518 (an angiogenesis inhibitor).
21071. The method of item 20958 wherein the anti- fibrotic agent is combretastatin (an angiogenesis inhibitor).
21072. The method of item 20958 wherein the anti- fibrotic agent is anecortave acetate (an angiogenesis inhibitor).
21073. The method of item 20958 wherein the anti- fibrotic agent is SB-715992 (a kinesin antagonist).
21074. The method of item 20958 wherein the anti- fibrotic agent is temsirolimus (an mTOR inhibitor).
21075. The method of item 20958 wherein the anti- fibrotic agent is adalimumab (a TNFα antagonist).
21076. The method of item 20958 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with an anti-fibrotic agent, and (b) implanting the medical device into the host.
21077. The method of item 20958 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with an anti-infective agent, and (b) implanting the medical device into the host. 21078. The method of item 20958 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with a polymer; and (b) implanting the medical device into the host.
21079. The method of item 20958 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with a composition comprising an anti-fibrotic agent and a polymer, and (b) implanting the medical device into the host.
21080. The method of item 20958 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with a composition comprising an anti-infective agent and a polymer, and (b) implanting the medical device into the host.
21081. The method of item 20958 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with a composition comprising an anti-fibrotic agent, an anti- infective agent and a polymer, and (b) implanting the medical device into the host.
21082. The method of item 20958 wherein the anti- infective agent is an anthracycline.
21083. The method of item 20958 wherein the anti- infective agent is doxorubicin.
21084. The method of item 20958 wherein the anti- infective agent is is mitoxantrone. 21085. The method of item 20958 wherein the anti- infective agent is a fluoropyrimidine.
21086. The method of item 20958 wherein the anti- infective agent is 5-fluorouracil (5-FU).
21087. The method of item 20958 wherein the anti- infective agent is a folic acid antagonist.
21088. The method of item 20958 wherein the anti- infective agent is methotrexate.
21089. The method of item 20958 wherein the anti- infective agent is a podophylotoxin.
21090. The method of item 20958 wherein the anti- infective agent is etoposide.
21091. The method of item 20958 wherein the anti- infective agent is camptothecin.
21092. The method of item 20958 wherein the anti- infective agent is a hydroxyurea.
21093. The method of item 20958 wherein the anti- infective agent is a platinum complex.
21094. The method of item 20958 wherein the anti- infective agent is cisplatin. 21095. The method of item 20958 wherein the composition comprises an anti-thrombotic agent.
21096. The method of item 20958 wherein the polymer is formed from reactants comprising a naturally occurring polymer.
21097. The method of item 20958 wherein the polymer is formed from reactants comprising protein.
21098. The method of item 20958 wherein the polymer is formed from reactants comprising carbohydrate.
21099. The method of item 20958 wherein the polymer is formed from reactants comprising biodegradable polymer.
21100. The method of item 20958 wherein the polymer is formed from reactants comprising nonbiodegradable polymer.
21101. The method of item 20958 wherein the polymer is formed from reactants comprising collagen.
21102. The method of item 20958 wherein the polymer is formed from reactants comprising methylated collagen.
21103. The method of item 20958 wherein the polymer is formed from reactants comprising fibrinogen.
21104. The method of item 20958 wherein the polymer is formed from reactants comprising thrombin. 21105. The method of item 20958 wherein the polymer reactants comprising blood plasma.
21106. The method of item 20958 wherein the polymer reactants comprising calcium salt.
21107. The method of item 20958 wherein the polymer reactants comprising an antifibronolytic agent.
21108. The method of item 20958 wherein the polymer reactants comprising fibrinogen analog.
21109. The method of item 20958 wherein the polymer reactants comprising albumin.
21110. The method of item 20958 wherein the polymer reactants comprising plasminogen.
21111. The method of item 20958 wherein the polymer reactants comprising von Willebrands factor.
21112. The method of item 20958 wherein the polymer reactants comprising Factor VIII.
21113. The method of item 20958 wherein the polymer reactants comprising hypoallergenic collagen.
21114. The method of item 20958 wherein the polymer reactants comprising atelopeptidic collagen. 21115. The method of item 20958 wherein the polymer is formed from reactants comprising teiopeptide collagen.
21116. The method of item 20958 wherein the polymer is formed from reactants comprising crosslinked collagen.
21117. The method of item 20958 wherein the polymer is formed from reactants comprising aprotinin.
21118. The method of item 20958 wherein the polymer is formed from reactants comprising epsilon-amino-n-caproic acid.
21119. The method of item 20958 wherein the polymer is formed from reactants comprising gelatin.
21120. The method of item 20958 wherein the polymer, is formed from reactants comprising protein conjugates.
21121. The method of item 20958 wherein the polymer is formed from reactants comprising gelatin conjugates.
21122. The method of item 20958 wherein the polymer is formed from reactants comprising a synthetic polymer.
21123. The method of item 20958 wherein the polymer is formed from reactants comprising a synthetic isocyanate-containing compound.
21124. The method of item 20958 wherein the polymer is formed from reactants comprising a synthetic thiol-containing compound. 21125. The method of item 20958 wherein the polymer is formed from reactants comprising a synthetic compound containing at least two thiol groups.
21126. The method of item 20958 wherein the polymer is formed from reactants comprising a synthetic compound containing at least three thiol groups.
21127. The method of item 20958 wherein the polymer is formed from reactants comprising a synthetic compound containing at least four thiol groups.
21128. The method of item 20958 wherein the polymer is formed from reactants comprising a synthetic amino-containing compound.
21129. The method of item 20958 wherein the polymer is formed from reactants comprising a synthetic compound containing at least two amino groups.
21130. The method of item 20958 wherein the polymer is formed from reactants comprising a synthetic compound containing at least three amino groups.
21131. The method of item 20958 wherein the polymer is formed from reactants comprising a synthetic compound containing at least four amino groups.
21132. The method of item 20958 wherein the polymer is formed from reactants comprising a synthetic compound comprising a carbonyl-oxygen-succinimidyl group. 21133. The method of item 20958 wherein the polymer is formed from reactants comprising a synthetic compound comprising at least two carbonyl-oxygen-succinimidyl groups.
21134. The method of item 20958 wherein the polymer is formed from reactants comprising a synthetic compound comprising at least three carbonyl-oxygen-succinimidyl groups.
21135. The method of item 20958 wherein the polymer is formed from reactants comprising a synthetic compound comprising at least four carbonyl-oxygen-succinimidyl groups.
21136. The method of item 20958 wherein the polymer is formed from reactants comprising a synthetic polyalkylene oxide- containing compound.
21137. The method of item 20958 wherein the polymer is formed from reactants comprising a synthetic compound comprising both polyalkylene oxide and biodegradable polyester blocks.
21138. The method of item 20958 wherein the polymer is formed from reactants comprising a synthetic polyalkylene oxide- containing compound having reactive amino groups.
21139. The method of item 20958 wherein the polymer is formed from reactants comprising a synthetic polyalkylene oxide- containing compound having reactive thiol groups.
21140. The method of item 20958 wherein the polymer is formed from reactants comprising a synthetic polyalkylene oxide- containing compound having reactive carbonyl-oxygen-succinimidyl groups. 21141. The method of item 20958 wherein the polymer is formed from reactants comprising a synthetic compound comprising a biodegradable polyester block.
21142. The method of item 20958 wherein the polymer is formed from reactants comprising a synthetic polymer formed in whole or part from lactic acid or lactide.
21143. The method of item 20958 wherein the polymer is formed from reactants comprising a synthetic polymer formed in whole or part from glycolic acid or glycolide.
21144. The method of item 20958 wherein the polymer is formed from reactants comprising polylysine.
21145. The method of item 20958 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound comprising a polyalkylene oxide portion.
21146. The method of item 20958 wherein the polymer is formed from reactants comprising (a) protein and (b) polylysine.
21147. The method of item 20958 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound having at least four thiol groups.
21148. The method of item 20958 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound having at least four amino groups. 21149. The method of item 20958 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound having at least four carbonyl-oxygen-succinimide groups.
21150. The method of item 20958 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound having a biodegradable region formed from reactants selected from lactic acid, lactide, glycolic acid, glycolide, and epison-caprolactone.
21151. The method of item 20958 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound comprising a polyalkylene oxide portion.
21152. The method of item 20958 wherein the polymer is formed from reactants comprising (a) collagen and (b) polylysine.
21153. The method of item 20958 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound having at least four thiol groups.
21154. The method of item 20958 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound having at least four amino groups.
21155. The method of item 20958 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound having at least four carbonyl-oxygen-succinimide groups.
21156. The method of item 20958 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound having a biodegradable region formed from reactants selected from lactic acid, lactide, glycolic acid, glycolide, and epison-caprolactone.
21157. The method of item 20958 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound comprising a polyalkylene oxide portion.
21158. The method of item 20958 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) polylysine.
21159. The method of item 20958 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound having at least four thiol groups.
21160. The method of item 20958 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound having at least four amino groups.
21161. The method of item 20958 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound having at least four carbonyl-oxygen-succinimide groups.
21162. The method of item 20958 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound having a biodegradable region formed from reactants selected from lactic acid, lactide, glycolic acid, glycolide, and epison-caprolactone.
21163. The method of item 20958 wherein the polymer is formed from reactants comprising hyaluronic acid. 21164. The method of item 20958 wherein the polymer is formed from reactants comprising a hyaluronic acid derivative.
21165. The method of item 20958 wherein the polymer is formed from reactants comprising pentaerythritol poly(ethylene glycol)ether tetra-sulfhydryl of number average molecular weight between 3,000 and 30,000.
21166. The method of item 20958 wherein the polymer is formed from reactants comprising pentaerythritol poly(ethylene glycol)ether tetra-amino of number average molecular weight between 3,000 and 30,000.
21167. The method of item 20958 wherein the polymer is formed from reactants comprising (a) a synthetic compound having a number average molecular weight between 3,000 and 30,000 and comprising a polyalkylene oxide region and multiple nucleophilic groups, and (b) a synthetic compound having a number average molecular weight between 3,000 and 30,000 and comprising a polyalkylene oxide region and multiple electrophilic groups.
21168. The method of item 20958 wherein the composition comprises a colorant.
21169. The method of item 20958 wherein the composition is sterile.
21170. A method as in any one of items 20958-21169, wherein the device is a peritoneal dialysis catheter adapted for delivering a drug to the peritoneum. 21171. A method for implanting a medical device comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with i) an anti-fibrotic agent, ii) an anti-infective agent, iii) a polymer, iv) a composition comprising an anti-fibrotic agent and a polymer, v) a composition comprising an anti-infective agent and a polymer, or vi) a composition comprising an anti-fibrotic agent, an anti- infective agent and a polymer, and (b) implanting the medical device into the host, wherein the medical device is a central nervous system shunt or a pressure monitoring device.
21172. The method of item 21171 wherein the anti- fibrotic agent is an adensosine A2A receptor antagonist.
21173. The method of item 21171 wherein the anti- fibrotic agent is an AKT inhibitor.
21174. The method of item 21171 wherein the anti- fibrotic agent is an alpha 2 integrin antagonist, wherein the alpha 2 integrin antagonist is Pharmaprojects No. 5754 (Merck KgaA).
21175. The method of item 21171 wherein the anti- fibrotic agent is an alpha 4 integrin antagonist.
21176. The method of item 21171 wherein the anti- fibrotic agent is an alpha 7 nicotinic receptor agonist.
21177. The method of item 21171 wherein the anti- fibrotic agent is an angiogenesis inhibitor selected from the group consisting of AG-12,958 (Pfizer), ATN-161 (Attenuon LLC), neovastat, an angiogenesis inhibitor from Jerina AG (Germany), NM-3 (Mercian), VGA-1155 (Taisho), FCE-26644 (Pfizer), FCE-26950 (Pfizer), FPMA (Meiji Daries), FR-111142 (Fujisawa), GGTI-298, GM-1306 (Ligand), GPA-1734 (Novartis), NNC-47- 0011 (Novo Nordisk), herbamycin (Nippon Kayaku), lenalidomide (Celegene), IP-10 (NIH), ABT-828 (Abbott), KIN-841 (Tokushima University, Japan), SF-1126 (Semafore Pharmaceuticals), laminin technology (NIH), CHIR-258 (Chiron), NVP-AEW541 (Novartis), NVP-AEW541 (Novartis), Vt16907 (Alchemia), OXI-8007 (Oxigene), EG-3306 (Ark Therapeutics), Maspin (Arriva), ABT-567 (Abbott), PPI-2458 (Praecis Pharmaceuticals), CC-5079, CC-4089 (Celgene), HIF-1 alpha inhibitors (Xenova), S-247 (Pfizer), AP-23573 (Ariad), AZD-9935 (Astra Zeneca), mebendazole (Introgen Therapeutics), MetAP-2 inhibitors (GlaxoSmithKline), AG-615 (Angiogene Pharmaceuticals), Tie-2 antagonists (Hybrigenics), NC-381 , CYC-381, NC-169, NC-219, NC-383, NC-384, NC-407 (Lorus Therapeutics), ATN-224 (Attenuon), ON-01370 (Onconova), Vitronectin antagonists (Amgen), SDX-103 (Salmedix), Vitronectin antagonists (Shire), CHP (Riemser), TEK (Amgen), Anecortave acetate (Alcon), T46.2 (Matrix Therapeutics), HG-2 (Heptagen), TEM antagonists (Genzyme), Oxi-4500 (Oxigene), ATN-161 (Attenuon), WX-293 (Wilex), M-2025 (Metris Therapeutics), Alphastatin (BioActa), YH-16 (Yantai Rongchang), BIBF- 1120 (Boehringer Ingelheim), BAY-57-9352 (Bayer), AS-1404 (Cancer Research Technology), SC-77964 (Pfizer), glycomimetics (BioTie Therapies), TIE-2 Inhibitors (Ontogen), DIMI, Octamer (Octamer), ABR- 215050 (Active Biotech), ABT-518 (Abbott), KDR inhibitors (Abbott), BSF- 466895 (Abbott), SCH-221153 (Schering-Plough), DAC:antiangiogenic (ConjuChem), TFPI (EntreMed), AZD-2171 (Astra-Zenaca), CDC-394 (Celgene), LY290293 (EIi Lilly), IDN-5390 (Indena), Kdr Kinase Inhibitors (Merck), CT-113020, CT-116433, CT-116563, CT-31890, CT-32228) (Cell Therapeutics), A-299620 (Abbott), TWEAK Inhibitor (Amgen), VEGF modulators (Johnson and Johnson), Tum-N53, tumstatin (Genzyme), Thios- 1 , Thios-2 (Thios Pharmaceuticals), MV-6401 (Miravant Medical Technologies), Spisulosine (PharmaMar), CEP-7055 (Cephalon), AUV-201 (Auvation), LM-609 (EIi Lilly), SKF-106615 (AnorMED), Oglufanide disodium (Cytran), BW-114 (Pharminox), Calreticulin (NIH), WX-678 (Wilex), SD-7784 (Pfizer), WX-UK1 (Wilex), SH-268 (Schering AG), 2-Me-PGA (Celgene), S- 137 (Pfizer), ZD-6126 (Angiogene Pharmaceuticals), SG-292 (SignalGen), Benefin (Lane Labs), A6, A36 (Angstrom), SB-2723005 (GlaxoSmithKline), SC-7 (Cell Therapeutics), ZEN-014 (AEterna Zentaris), 2-methoxyestradiol (EntreMed), NK-130119 (Nippon Kayaku), CC-10004 (Celgene), AVE- 8062A (Ajinomoto), Tacedinaline (Pfizer), Actinonin (Tokyo Metropolitan Institute of Medical Science), Lenalidomide (Celgene), VGA-1155, BTO-956 (SRI International), ER-68203-00 (Eisai), CT-6685 (UCB), JKC-362 (Phoenix Pharmaceuticals), DMI-3798 (DMI Biosciences, Angiomate (Ipsen), ZD-6474 (AstraZeneca), CEP-5214 (Cephalon), Canstatin (Genzyme), NM-3 (Mercian), Oridigm (MediQuest Therapeutics), Exherin (Adherex), BLS-0597 (Boston Life Sciences), PTC-299 (PTC Therapeutics), NPI-2358 (Nereus Pharmaceuticals), CGP-79787 (Novartis), AEE-788 (Novartis), CKD-732 (Chong Kun Dang), CP-564959 (OSI Pharmaceuticals),. CM-101 (CarboMed), CT-2584, CT3501 (Cell Therapeutics), combretastatin and analogues and derivatives thereof (Oxigene), Rebimastat (Bristol-Meyers Squibb), Dextrin 2-sulfate (ML Laboratories), Cilengitide (Merk KGaA), NSC-706704 (Pharminox), KRN- 951 (Kirin Brewery), Ukrain, NSC-631570 (Nowicky Pharma), Tecogalan sodium (Daiichi Pharmaceutical), Tz-93 (Tsumura), TBC-1635 (Encysive Pharmaceuticals), TAN-1120 (Takeda), Semaxanib (Pfizer), BDI-7800 (Biopharmacopae), SD-186, SD-983 (Bristol-Meyers Squibb), SB-223245 (GlaxoSmithKline), SC-236 (Pfizer), RWJ-590973 (Johnson and Johnson), ILX-1850 (Genzyme), SC-68488, S-836 (Pfizer), CG-55069-11 (CuraGen), Ki-23057 (Kirin Brewery), CCX-700 (Chemoentryx), Pegaptanib octasodium (Giled Sciences), ANGIOCOL (available from Biostratum Inc.), or an analogue or derivative thereof.
21178. The method of item 21171 wherein the anti- fibrotic agent is an apoptosis antagonist. 21179. The method of item 21171 wherein the anti- fibrotic agent is an apoptosis activator.
21180. The method of item 21171 wherein the anti- fibrotic agent is a beta 1 integrin antagonist.
21181. The method of item 21171 wherein the anti- fibrotic agent is a beta tubulin inhibitor.
21182. The method of item 21171 wherein the anti- fibrotic agent is a blocker of enzyme production in Hepatitis C.
21183. The method of item 21171 wherein the anti- fibrotic agent is a Bruton's tyrosine kinase inhibitor.
21184. The method of item 21171 wherein the anti- fibrotic agent is a calcineurin inhibitor.
21185. The method of item 21171 wherein the anti- fibrotic agent is a caspase 3 inhibitor.
21186. The method of item 21171 wherein the anti- fibrotic agent is a CC chemokine receptor antagonist.
21187. The method of item 21171 wherein the anti- fibrotic agent is a cell cycle inhibitor selected from the group consisting of SNS-595 (Sunesis), synthadotin, KRX-0403, and an analogue or derivative thereof.
21188. The method of item 21171 wherein the anti- fibrotic agent Is a cathepsin B inhibitor. 21189. The method of item 21171 wherein the anti- fibrotic agent is a cathepsin K inhibitor, wherein the cathepsin K inhibitor is 462795 (GlaxoSmithKline), INPL-022-D6 (Amura Therapeutics), or an analogue or derivative thereof.
21190. The method of item 21171 wherein the anti- fibrotic agent is a cathepsin L inhibitor.
21191. The method of item 21171 wherein the anti- fibrotic agent is a CD40 antagonist.
21192. The method of item 21171 wherein the anti- fibrotic agent is a chemokine receptor agonist.
21193. The method of item 21171 wherein the anti- fibrotic agent is a chymase inhibitor.
21194. The method of item 21171 wherein the anti- fibrotic agent is a coϋagenase antagonist.
21195. The method of item 21171 wherein the anti- fibrotic agent is a CXCR antagonist.
21196. The method of item 21171 wherein the anti- fibrotic agent is a cyclin dependent kinase inhibitor selected from the group consisting of a CDK-1 inhibitor, a CDK-2 inhibitor, a CDK- 4 inhibitor, a CDK-6 inhibitor, a CAK1 inhibitor from GPC Biotech and Bristol-Myers Squibb, RGB-286199 (GPC Biotech), an anticancer agent from Astex Technology, a CAK1 inhibitor from GPC Biotech, a CDK inhibitor from Sanofi-Aventis, a CDK1/CDK2 inhibitor from Amgen, a CDK2 inhibitor from SUGEN-2 (Pfizer), a hearing loss therapy agent (Sound Pharmaceuticals), PD-0332991 (Pfizer), RGB-286199 (GPC Biotech), Ro-0505124 (Hoffmann- La Roche), a Ser/Thr kinase inhibitor from Lilly (EIi Lilly), CVT-2584 (CAS No. 199986-75-9) (CV Therapeutics), and an analogue or derivative thereof.
21197. The method of item 21171 wherein the anti- fibrotic agent is a cyclooxygenase 1 inhibitor.
21198. The method of item 21171 wherein the anti- fibrotic agent is a DHFR inhibitor.
21199. The method of item 21171 wherein the anti- fibrotic agent is a dual integrin inhibitor.
21200. The method of item 21171 wherein the anti- fibrotic agent is an elastase inhibitor.
21201. The method of item 21171 wherein the anti- fibrotic agent is an elongation factor-1 alpha inhibitor.
21202. The method of item 21171 wherein the anti- fibrotic agent is an endothelial growth factor antagonist.
21203. The method of item 21171 wherein the anti- fibrotic agent is an endothelial growth factor receptor kinase inhibitor selected from the group consisting of sorafenib tosylate (Bayer), AAL-993 (Novartis), ABP-309 (Novartis), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EXEL- 2880 (Exelixis), GW-654652 (GlaxoSmithKline), a KDR inhibitor from LG Life Sciences, CT-6685 and CT-6729 (UCB), KRN-633 and KRN-951 (Kirin Brewery), OSI-930 (OSI Pharmaceuticals), SP-5.2 (Supratek Pharma), SU- 11657 (Pfizer), a Tie-2 antagonist (Hybrigenics), a VEGFR-2 kinase inhibitor (Bristol-Myers Squibb), XL-647 (Exelixis), a KDR inhibitor from Abbott Laboratories, sorafenib tosylate, and an analogue or derivative thereof.
21204. The method of item 21171 wherein the anti- fibrotic agent is an endotoxin antagonist.
21205. The method of item 21171 wherein the anti- fibrotic agent is an epothilone and tubulin binder.
21206. The method of item 21171 wherein the anti- fibrotic agent is an estrogen receptor antagonist.
21207. The method of item 21171 wherein the anti- fibrotic agent is an FGF inhibitor.
21208. The method of item 21171 wherein the anti- fibrotic agent is a farnexyl transferase inhibitor.
21209. The method of item 21171 wherein the anti- fibrotic agent is farnesyltransferase inhibitor selected from the group of A- 197574 (Abbott), a farnesyltransferase inhibitor from Servier, FPTIII (Strathclyde Institute for Drug R), LB-42908 (LG Life Sciences), Pharmaprojects No. 5063 (Genzyme), Pharmaprojects No. 5597 (Ipsen), Yissum Project No. B-1055 (Yissum), and an analogue or derivative thereof.
21210. The method of item 21171 wherein the anti- fibrotic agent is an FLT-3 kinase inhibitor.
21211. The method of item 21171 wherein the anti- fibrotic agent is an FGF receptor kinase inhibitor. 21212. The method of item 21171 wherein the anti- fibrotic agent is a fibrinogen antagonist selected from the group consisting of AUV-201 (Auvation), MG-13926 (Sanofi-Aventis), plasminogen activator (CAS No. 105913-11-9) (from Sanofi-Aventis or UCB), plasminogen activator-2 (tPA-2) (Sanofi-Aventis), pro-urokinase (CAS No. 82657-92-9) (Sanofi-Aventis), and an analogue or derivative thereof.
21213. The method of item 21171 wherein the anti- fibrotic agent is a heat shock protein 90 antagonist selected from the group consisting of SRN-005 (Sirenade), geldanamycin, NSC-33050 (17- allylaminogeldanamycin; 17-AAG), 17-dimethylaminoethylamino-17- demethoxy-geldanamycin (17-DMAG), rifabutin (rifamycin XIV, 1',4- didehydro-i-deoxy-i ^-dihydro-δ'-^-methylpropyO-i-oxo-), and an analogue or derivative thereof.
21214. The method of item 21171 wherein the anti- fibrotic agent is a histone deacetylase inhibitor.
21215. The method of item 21171 wherein the anti- fibrotic agent is an HMGCoA reductase inhibitor selected from the group consisting of an atherosclerosis therapeutic from Lipid Sciences, ATI-16000 (ARYx Therapeutics), KS-01-019 (Kos Pharmaceuticals), Pharmaprojects No. 2197 (Sanofi-Aventi), RP 61969 (Sanofi-Aventis), and an analogue or derivative thereof.
21216. The method of item 21171 wherein the anti- fibrotic agent is an ICAM inhibitor.
21217. The method of item 21171 wherein the anti- fibrotic agent is an IL, ICE and IRAK antagonist, wherein the antagonist is a CJ-14877, CP-424174 (Pfizer), NF-61 (Negma-Lerads), and an analogue or derivative thereof.
21218. The method of item 21171 wherein the anti- fibrotic agent is an IL-2 inhibitor.
21219. The method of item 21171 wherein the anti- fibrotic agent is an immunosuppressant selected from the group consisting of teriflunomide (Sanofi Aventis), chlorsulfaquinoxalone (NSC-339004), chlorsulfaquinoxalone sulfate, CS-712 (Sankyo), ismomultin alfa (CAS No. 457913-93-8) (Akzo Nobel), antiallergics from GenPat77, antiinflammatories or AT-005 (Androclus Therapeutics), autoimmune disease therapeutics from EpiVax, BN-007 (Bone), budesonide (CAS No. 51333-22- 3) (MAP Pharmaceuticals), CO-14 (Genzyme), edratide (CAS No. 433922- 67-9) (Teva), EP-314 (Enanta), eprovafen (CAS No. 101335-99-3) (Sanofi- Aventis), HWA-131 (CAS No. 118788-41-3) (Sanofi-Aventis), immunomodulators from MerLion Pharmaceuticals, immunosuppressives from Alchemia, IPL-12 (Inflazyme), MDL-9563 (CAS No. 27086-86-8) (Sanofi-Aventis), Pharmaprojects No. 2330 (Sanofi-Aventis), Pharmaprojects No. 6426 (Abgenix), PXS-25 (Pharmaxis), rosmarinic acid (CAS No. 20283-92-5) (Sanofi-Aventis), RP 42927 or RP 54745 (CAS No. 135330-08-4) (Sanofi-Aventis), SGN-35 (Seattle Genetics), ST-1959 (Sigma-Tau), type I diabetes therapy from SYNX Pharma, UNIL-88 (Debiopharm), VP-025 (Vasogen), VR-694 (Vectura), PRTX-001 (Protalex), and an analogue or derivative thereof.
21220. The method of item 21171 wherein the anti- fibrotic agent is an IMPDH (inosine monophosphate).
21221. The method of item 21171 wherein the anti- fibrotic agent is an integrin antagonist. 21222. The method of item 21171 wherein the anti- fibrotic agent is an interleukin antagonist.
21223. The method of item 21171 wherein the anti- fibrotic agent is an inhibitor of type 111 receptor tyrosine kinase.
21224. The method of item 21171 wherein the anti- fibrotic agent is an irreversible inhibitor of enzyme methionine aminopeptidase type 2.
21225. The method of item 21171 wherein the anti- fibrotic agent is an isozyme selective delta protein kinase C inhibitor.
21226. The method of item 21171 wherein the anti- fibrotic agent a JAK3 enzyme inhibitor.
21227. The method of item 21171 wherein the anti- fibrotic agent is a JNK inhibitor.
21228. The method of item 21171 wherein the anti- fibrotic agent is a kinase inhibitor.
21229. The method of item 21171 wherein the anti- fibrotic agent is kinesin antagonist.
21230. The method of item 21171 wherein the anti- fibrotic agent is a kinesin antagonist.
21231. The method of item 21171 wherein the anti- fibrotic agent is a leukotriene inhibitor and antagonist selected from the group consisting of ambicromil (CAS No. 58805-38-2) (Sanofi-Aventis), amelubant (CAS No. 346735-24-8) (Boehringer Ingelheim), DW-1141 (Dong Wha), ebselen (Daiichi Pharmaceutical), ibudilast (Kyorin), leucotriene inhibitors from Sanofi-Aventis, lymphotoxin-beta receptor (LT-D) from Biogen Idee, Pharmaprojects No. 1535 and 2728 (CAS No. 119340-33-9) (Sanofi-Aventis), R-112 (Rigel), Rev-5367 (CAS No. 92532-05-3) (Sanofi- Aventis), RG-14893 (CAS No. 141835-49-6) (Sanofi-Aventis), RG-5901-A (CAS No. 101910-24-1), 92532-23-5, RP 66153 (CAS No. 142422-79-5), RP 66364 (CAS No.186916-92-5), RP 69698 (CAS No. 141748-00-7) (Sanofi-Aventis), SC-411930 (Pfizer), SC-41930 (CAS No. 120072-59-5) (Pfizer), SC-50605 (CAS No. 138828-39-4) (Pfizer), SC-51146 (CAS No. 141059-52-1), SC-53228 (CAS No. 153633-01-3) (Pfizer), spaglumic acid (ZY-15106) (CAS No.3106-85-2), 80619-64-3 (Novartis), tipredane (CAS No. 85197-77-9) (Bristol-Myers Squibb), U-75302 (CAS No. 119477-85-9) (Pfizer), and analogue or derivative thereof.
21232. The method of item 21171 wherein the anti- fibrotic agent is an MAP kinase inhibitor.
21233. The method of item 21171 wherein the anti- fibrotic agent is a matrix metalloproteinase inhibitor.
21234. The method of item 21171 wherein the anti- fibrotic agent is an MCP-CCR2 inhibitor.
21235. The method of item 21171 wherein the anti- fibrotic agent is an mTOR inhibitor.
21236. The method of item 21171 wherein the anti- fibrotic agent is an mTOR kinase inhibitor. 21237. The method of item 21171 wherein the anti- fibrotic agent is a microtubule inhibitor selected from the group consisting of antibody-maytansinoid conjugates from Biogen Idee, colchicines (MantiCore Pharmaceuticals), anticancer immunoconjugates from Johnson & Johnson, DIME from Octamer, gni-1f (GNI), huC242-DM4, huMy9-6-DM1 (ImmunoGen), IDN-5404 (Indena), IMO-098, IMOderm (Imotep), mebendazole (Introgen Therapeutics), microtubule poisons from Cambridge Enterprise, paclitaxel such as LOTAX from Aphios (CAS No. 33069-62-4), Genexol-PM from Samyang, Pharmaprojects No. 6383 (Tapestry Pharmaceuticals), RPR-112378 (Sanofi-Aventis), SGN-75 (Seattle Genetics), SPL-7435 (Starpharma), SSR-250411 (Sanofi-Aventis), trastuzumab-DM1 (Genentech), vinorelbine, and an analogue or derivative thereof.
21238. The method of item 21171 wherein the anti- fibrotic agent is an MIF inhibitor.
21239. The method of item 21171 wherein the anti- fibrotic agent is an MMP inhibitor.
21240. The method of item 21171 wherein the anti- fibrotic agent is a neurokinin (NK) antagonist selected from the group consisting of anthrotainin (CAS No. 148084-40-6) (Sanofi-Aventis), an IBS thereapeutic from ArQuIe, MDL-105212A (CAS No. 167261-60-1) (Ssanofi- Aventis), Pharmaprojects No. 2744, 3258 (CAS No. 139167-47-8) 4006, 4201 , or 5986 (Sanofi-Aventis), RP 67580 (CAS No. 135911-02-3), SR- 144190 (CAS No. 201152-86-5), SSR-240600, SSR-241586 (Sanofi- Aventis), TKA-457 (Novartis), vestipitant mesylate (CAS No. 334476-64-1) (GlaxoSmithKline), Win-64821 (Sanofi-Aventis), PRX-96026 (Predix Pharmaceuticals), and an analogue or derivative thereof. 21241. The method of item 21171 wherein the anti- fibrotic agent is an NF kappa B inhibitor selected from the group consisting of AVE-0545 or AVE-0547 (Sanofi-Aventis), bortezomib (CAS No. 179324- 69-7) (Millennium Pharmaceuticals), dexanabinol (CAS No. 112924-45-5) (Pharmos), dexlipotam (Viatris), Pharmaprojects No. 6283 (INDRA) (OXiGENE), IPL-576092 (CAS No. 137571-30-3) (Inflazyme), NFKB decoy (Corgentech), NFKB decoy oligo (AnGes MG), NFKB's from Ariad, osteoporosis treatments or S5 (F005) from Fulcrum Pharmaceuticals, P61 (Phytopharm), R-flurbiprofen (CAS No. 5104-49-4) (Encore Pharmaceuticals), and an analogue or derivative thereof.
21242. The method of item 21171 wherein the anti- fibrotic agent is a nitric oxide agonist.
21243. The method of item 21171 wherein the anti- fibrotic agent is an ornithine decarboxylase inhibitor.
21244. The method of item 21171 wherein the anti- fibrotic agent is a p38 MAP kinase inhibitor selected from the group consisting of AZD-6703 (AstraZeneca), JX-401 (Jexys Pharmaceuticals), BMS-2 (Bristol-Myers Squibb), a p38 MAP kinase inhibitor from Novartis, a p38-alpha MAP kinase inhibitor from Amphora, Pharmaprojects No. 5704 (Pharmacopeia), RPR-200765A (Sanofi-Aventis), SD-282 (Johnson & Johnson), TAK-715 (Takeda), and an analogue or derivative thereof.
21245. The method of item 21171 wherein the anti- fibrotic agent is a palmitoyl-protein thioesterase inhibitor.
21246. The method of item 21171 wherein the anti- fibrotic agent is a PDGF receptor kinase inhibitor selected from the group consisting of AAL-993, AMN-107, or ABP-309 (Novartis), AMG-706 (Amgen), BAY-57-9352 (Bayer), CDP-860 (UCB), E-7080 (Eisai), imatinib (CAS No. 152459-95-5) (Novartis), OSI-930 (OSI Pharmaceuticals), RPR- 127963E (Sanofi-Aventis), RWJ-540973 (Johnson & Johnson), sorafenib tosylate (Bayer), SU-11657 (Pfizer), tandutinib (CAS No. 387867-13-2) (Millennium Pharmaceuticals), vatalanib (Novartis), ZK-CDK (Schering AG), and an analogue or derivative thereof.
21247. The method of item 21171 wherein the anti- fibrotic agent is (-)-halofenate (Metabolex), AMG-131 (Amgen), antidiabetics from Japan Tobacco, AZD-4619, AZD-8450, AZD-8677 (AstraZeneca), DRF-10945, balaglitazone (Dr Reddy's), CS-00088, CS-00098 (Chipscreen Biosciences), E-3030 (Eisai), etalocib (CAS No. 161172-51-6) (EIi Lilly), GSK-641597 (Ligand), GSK-677954 (GlaxoSmithKline), GW-409544 (Ligand), GW-590735 (GlaxoSmithKline), K-111 (Hoffmann-La Roche), LY- 518674 (EIi Lilly), LY-674 (Ligand), LY-929 (Ligand), MC-3001, MC-3002 (MaxoCore Pharmaceuticals), metformin HCI + pioglitazone (CAS No. 1115- 70-4 and 112529-15-4), ACTOPLUS MET from Andrx), muraglitazar (CAS No. 331741-94-7) (Bristol-Myers Squibb), naveglitazar (Ligand), oleoylethanolamide (Kadmus Pharmaceuticals), ONO-5129, pioglitazone hydrochloride (CAS No. 111025-46-8 and 112529-15-4) (Takeda), PLX-204 (Plexxikon), PPAR agonists from Genfit, PPAR delta agonists from EIi Lilly, PPAR-alpha agonists from CrystalGenomics, PPAR-gamma modulators and PPAR-β modulators from CareX, rosiglitazone maleate (CAS No. 122324- 73-4 or 155141-29-0) (GlaxoSmithKline), rosiglitazone maleate/glimepir (CAS No. 155141-29-0 and 93479-97-1), AVANDARYL, rosiglitazone maleate/metformin extend (CAS No. 155141-29-0 and 657-24-9), AVANDAMET, rosiglitazone maleate+metformin, AVANDAMET (GlaxoSmithKline), tesaglitazar (AstraZeneca), and an analogue or derivative thereof. 21248. The method of item 21171 wherein the anti- fibrotic agent is a phosphatase inhibitor.
21249. The method of item 21171 wherein the anti- fibrotic agent is a phosphodiesterase (PDE) inhibitor selected from the group consisting of avanafil (Tanabe Seiyaku), dasantafil (CAS No. 569351- 91-3) (Schering-Plough), A-906119 (CAS No. 134072-58-5), DL-850 (Sanofi-Aventis), GRC-3015, GRC-3566, GRC-3886 (Glenmark), HWA-153 (CAS No. 56395-66-5) (Sanofi-Aventis), hydroxypumafentrine (Altana), IBFB-130011 , IBFB-14-016, IBFB-140301, IBFB-150007, IBFB-211913 (IBFB Pharma), L-826141 (Merck & Co), medorinone (CAS No. 88296-61-1) (Sanofi-Aventis), MEM-1917 (Memory Pharmaceuticals), ND-1251 (Neuro3d), PDE inhibitors from ICOS, PDE IV inhibitors from Memory Pharmaceuticals and CrystalGenomics, Pharmaprojects No. 2742 and 6141 (Sanofi-Aventis), QAD-171 (Novartis), RHC-2963 (CAS No. 76993-12-9 and 76993-14-1), RPR-117658, RPR-122818 derivatives, SR-24870 , and RPR- 132294 (Sanofi-Aventis), SK-350 (ln2Gen), stroke therapy agents from deCODE Genetics, TAS-203 (Taiho), tofimilast (CAS No. 185954-27-2) (Pfizer), UK-371800 (Pfizer), WIN-65579 (CAS No. 158020-82-7) (Sanofi- Aventis), IBFB-130020 (IBFB Pharma), OPC-6535 (CAS No. 145739-56-6) (Otsuka), a phosphodiesterase III inhibitor, enoximone, a phosphodiesterase IV inhibitor, fosfosal, Atopik (Barrier Therapeutics), triflusal, a phosphodiesterase V inhibitor, and an analogue or derivative thereof.
21250. The method of item 21171 wherein the anti- fibrotic agent is a PKC inhibitor.
21251. The method of item 21171 wherein the anti- fibrotic agent is a platelet activating factor antagonist. 21252. The method of item 21171 wherein the anti- fibrotic agent is a platelet-derived growth factor receptor kinase inhibitor.
21253. The method of item 21171 wherein the anti- fibrotic agent is a prolyl hydroxylase inhibitor.
21254. The method of item 21171 wherein the anti- fibrotic agent is a polymorphonuclear neutrophil inhibitor.
21255. The method of item 21171 wherein the anti- fibrotic agent is a protein kinase B inhibitor.
21256. The method of item 21171 wherein the anti- fibrotic agent is a protein kinase C stimulant.
21257. The method of item 21171 wherein the anti- fibrotic agent is a purine nucleoside analogue.
21258. The method of item 21171 wherein the anti- fibrotic agent is a purinoreceptor P2X antagonist.
21259. The method of item 21171 wherein the anti- fibrotic agent is a Raf kinase inhibitor.
21260. The method of item 21171 wherein the anti- fibrotic agent is a reversible inhibitor of ErbB1 and ErbB2.
21261. The method of item 21171 wherein the anti- fibrotic agent is a ribonucleoside triphosphate reductase inhibitor. 21262. The method of item 21171 wherein the anti- fibrotic agent is an SDF-1 antagonist.
21263. The method of item 21171 wherein the anti- fibrotic agent is a sheddase inhibitor.
21264. The method of item 21171 wherein the anti- fibrotic agent is an SRC inhibitor.
21265. The method of item 21171 wherein the anti- fibrotic agent is a stromelysin inhibitor.
21266. The method of item 21171 wherein the anti- fibrotic agent is an Syk kinase inhibitor.
21267. The method of item 21171 wherein the anti- fibrotic agent is a telomerase inhibitor.
21268. The method of item 21171 wherein the anti- fibrotic agent is a TGF beta inhibitor selected from the group consisting of pirfenidone (CAS No. 53179-13-8) (MARNAC)1 tranilast (CAS No. 53902- 12-8) (Kissei), IN-1130 (ln2Gen), mannose-6-phosphate (BTG), TGF-B antagonists from Inflazyme (Pharmaprojects No. 6075), TGF-β antagonists from Sydney, non-industrial source), TGF-β I receptor kinase inhibitors from EIi Lilly, TGF-β receptor inhibitors from Johnson & Johnson, and an analogue or derivative thereof.
21269. The method of item 21171 wherein the anti- fibrotic agent is a TN Fa antagonist or TACE inhibitor selected from the group consisting of adalimumab (CAS No. 331731-18-1) (Cambridge Antibody Technology), AGIX-4207 (AtheroGenics), AGT-1 (Advanced Biotherapy), an anti-inflammatory from Borean Pharma, Cellzome, or Paradigm Therapeutics, anti-inflammatory vaccine (TNF-alpha kinoid) from Neovacs, humanized anti-TNF antibody or an anti-TNF MAb (CB0006) Celltech (UCB), apratastat (CAS No. 287445-51-0) (Wyeth), BMS-561392 (Bristol-Myers Squibb), BN-006 (Bone), certoiizumab pegol (CAS No. 428863-50-7 or CH-138 (UCB), cilomilast (CAS No. 153259-65-5) (GlaxoSmithKline), CR-1 (Nuada Pharmaceuticals), CRx-119 (CombinatoRx), D-5410 (UCB), dacopafant (CAS No. 125372-33-0) (Sanofi- Aventis), dersalazine (CAS No. 188913-57-7/188913-58-8) (Uriach), etanercept (CAS No. 185243-69-0) (Amgen), ethyl pyruvate (Critical (Critical Therapeutics), golimumab (CAS No. 476181-74-5) (Johnson & Johnson), hormono-immunotherapy from Ipsen, CDP571 (e.g., Humicade from UCB), IC-485 (ICOS), infliximab (CAS No. 170277-31-3) (Johnson & Johnson), IP- 751 (Manhattan Pharmaceuticals), ISIS-104838 (CAS No. 250755-32-9) (ISIS Pharmaceuticals), lenalidomide (CAS No. 191732-72-6) (Celgene), lentinan (CAS No. 37339-90-5) (Ajinomoto), MDL-201112 (CAS No. 142130-73-2) (Sanofi-Aventis), medroxyprogesterone (CAS No. 520-85-4) (InKine Pharmaceutical), N-acetylcysteine (CAS No. 616-91-1) (Zambon), NBE-P2 (DIREVO Biotech), nerelimomab (CAS No. 162774-06-3) (Chiron), OM-294DP (OM PHARMA), onercept (CAS No. 199685-57-9) (Yeda), PASSTNF-alpha (Verigen), pentoxifylline or oxypentifylline (Sanofi-Aventis), Pharmaprojects No. 4091, 4241 , 4295, or 4488 (Sanofi-Aventis), Pharmaprojects No. 5480 (Amgen), Pharmaprojects No. 6749 (Cengent), pirfenidone (CAS No. 53179-13-8) (MARNAC), RPR-132294 (Sanofi- Aventis), S5 (F002) (Fulcrum Pharmaceuticals), simvastatin (CAS No. 79902-63-9) (Merck & Co), STA-6292 (Synta Pharmaceuticals), tacrolimus (CAS No. 104987-11-3) (Fujisawa LifeCycle Pharma), talactoferrin alfa (CAS No. 308240-58-6) (Agennix), thalidomide (CAS No. 50-35-1) (Celgene), TNF antagonists form ProStrakan, and Synergen, TNF inhibitors (Amgen), TNF-alpha antagonists from Dynavax Technologies and Jerina AG (Germany), TNF-alpha inhibitors from IBFB Pharma and Xencor (Xencor), torbafylline (CAS No. 105102-21-4) (Sanofi-Aventis), UR-1505 (Uriach), VT-346 (Viron Therapeutics), YSIL6 (Y's Therapeutics), YSTH2 (Y's Therapeutics), NPI-1302a-3 (Nereus Pharmaceuticals, a TNF antagonist from Jerina AG (Germany), dersalazine, and an analogue or derivative thereof.
21270. The method of item 21171 wherein the anti- fibrotic agent is a Toll receptor inhibitor.
21271. The method of item 21171 wherein the anti- fibrotic agent is a tubulin antagonist.
21272. The method of item 21171 wherein the anti- fibrotic agent is a tyrosine kinase inhibitor selected from the group consisting of SU-011248, SUTENT from Pfizer Inc. (New York, NY), BMS-354825, PN- 355 (Paracelsian Pharmaceuticals), AGN-199659 (Allergan), AAL-993 or ABP-309 (Novartis), adaphostin (NIH), AEE-788 (Novartis), AG-013736 (OSI Pharmaceuticals), AG-13736 (Pfizer), ALT-110 (Alteris Therapeutics), AMG-706 (Amgen), anticancer MAbs from Xencor, anti-EGFrvlll MAbs from Abgenix, anti-HER2 MAb from Abiogen, AZD-2171 or AZD-9935 (AstraZeneca), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), CEP-5214 (Cephalon), CEP-7055 (Cephalon), cetuximab (ImClone Systems), CHIR-200131 and CHIR-258 (Chiron), CP-547632 (OSI Pharmaceuticals), CP-724714 (Pfizer), CT-301 (Creabilis Therapeutics), D- 69491 (Baxter International), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EGFR/ErbB2 inhibitors from Array BioPharma, erlotinib (CAS No. 183319- 69-9) (OSI Pharmaceuticals), EXEL-2880 (Exelixis), FK-778 (Sanofi- Aventis) , gefitinib (CAS No. 184475-35-2) (AstraZeneca), GW-2286 or GW- 654652 (GlaxoSmithKline), her2/neu antigen from AlphaVax, HER-2/neu inhibitor from Generex, Herzyme (Medipad) (Sirna Therapeutics), HKI-272 (Wyeth), HuMax-EGFr (Genmab), idronoxil (CAS No. 81267-65-4) (Novogen), IGF-1 inhibitors from Ontogen, IMC-11F8 (ImCIone Systems), kahalalide F (CAS No. 149204-42-2) (PharmaMar), KDR inhibitor from LG Life Sciences, KDR inhibitors from Abbott Laboratories, KDR kinase inhibitors (UCB), Kdr kinase inhibitors from Merck & Co, KRN-633 and KRN- 951 (Kirin Brewery), KSB-102 (Xenova), lapatinib ditosylate (CAS No. 388082-78-8) (GlaxoSmithKline), matuzumab (Merck KGaA), MDX-214 (Medarex), ME-103 (Pharmexa), MED-A300 (Gerolymatos), MNAC-13 (Lay Line Genomics), nimotuzumab (Center of Molecular Immunology), NSC- 330507 or NSC-707545 (NIH), NV-50 (Novogen), OSI-930 (OSI Pharmaceuticals), panitumumab (Abgenix), pelitinib (CAS No. 287933-82-7) (Wyeth), pertuzumab (CAS No. 380610-27-5) (Genentech), Pharmaprojects No. 3985 (Sanofi-Aventis), prostate cancer therapeutics from Sequenom (SQPC35, SQPC36, SQPC90), removab and remoxab (Trion Pharma), RG- 13022 (CAS No. 136831-48-6), RG-13291 (CAS No. 138989-50-1), or RG- 14620 (CAS No. 136831-49-7) (Sanofi-Aventis), RM-6427 (Romark), RNAi breast cancer therapy from Benitec, RP 53801 (CAS No. 125882-88-4) (Sanofi-Aventis), sorafenib tosylate (Bayer), SU-11657 (Pfizer), Tie-2 antagonists from Semaia (Hybrigenics), Tie-2 inhibitors from Ontogen, trastuzumab (CAS No. 180288-69-1) (Genentech), tyrosine kinase inhibitors from Sanofi-Aventis, U3-1287, U3-1565, U3-1784, U3-1800 (U3 Pharma), vatalanib (Novartis), VEGFR-2 kinase inhibitor from Bristol-Myers Squibb, XL-647 (Exelixis), ZD-6474 (AstraZeneca), ZK-CDK (Schering AG), an EGFR tyrosine kinase inhibitor, EKB-569 (Wyeth), and an analogue or derivative thereof.
21273. The method of item 21171 wherein the anti- fibrotic agent is a VEGF inhibitor.
21274. The method of item 21171 wherein the anti- fibrotic agent is a vitamin D receptor agonist. 21275. The method of item 21171 wherein the anti- fibrotic agent is ZD-6474 (an angiogenesis inhibitor).
21276. The method of item 21171 wherein the anti- fibrotic agent is AP-23573 (an mTOR inhibitor).
21277. The method of item 21171 wherein the anti- fibrotic agent is synthadotin (a tubulin antagonist).
21278. The method of item 21171 wherein the anti- fibrotic agent is S-0885 (a collagenase inhibitor).
21279. The method of item 21171 wherein the anti- fibrotic agent is aplidine (an elongation factor-1 alpha inhibitor).
21280. The method of item 21171 wherein the anti- fibrotic agent is ixabepilone (an epithilone).
21281. The method of item 21171 wherein the anti- fibrotic agent is IDN-5390 (an angiogenesis inhibitor).
21282. The method of item 21171 wherein the anti- fibrotic agent is SB-2723005 (an angiogenesis inhibitor).
21283. The method of item 21171 wherein the anti- fibrotic agent is ABT-518 (an angiogenesis inhibitor).
21284. The method of item 21171 wherein the anti- fibrotic agent is combretastatin (an angiogenesis inhibitor). 21285. The method of item 21171 wherein the anti- fibrotic agent is anecortave acetate (an angiogenesis inhibitor).
21286. The method of item 21171 wherein the anti- fibrotic agent is SB-715992 (a kinesin antagonist).
21287. The method of item 21171 wherein the anti- fibrotic agent is temsirolimus (an mTOR inhibitor).
21288. The method of item 21171 wherein the anti- fibrotic agent is adalimumab (a TNFα antagonist).
21289. The method of item 21171 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with an anti-fibrotic agent, and (b) implanting the medical device into the host.
21290. The method of item 21171 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with an anti-infective agent, and (b) implanting the medical device into the host.
21291. The method of item 21171 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with a polymer; and (b) implanting the medical device into the host.
21292. The method of item 21171 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with a composition comprising an anti-fibrotic agent and a polymer, and (b) implanting the medical device into the host. 21293. The method of item 21171 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with a composition comprising an anti-infective agent and a polymer, and (b) implanting the medical device into the host.
21294. The method of item 21171 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with a composition comprising an anti-fibrotic agent, an anti- infective agent and a polymer, and (b) implanting the medical device into the host.
21295. The method of item 21171 wherein the anti- infective agent is an anthracycline.
21296. The method of item 21171 wherein the anti- infective agent is doxorubicin.
21297. The method of item 21171 wherein the anti- infective agent is is mitoxantrone.
21298. The method of item 21171 wherein the anti- infective agent is a fluoropyrimidine.
21299. The method of item 21171 wherein the anti- infective agent is 5-fluorouracil (5-FU).
21300. The method of item 21171 wherein the anti- infective agent is a folic acid antagonist.
21301. The method of item 21171 wherein the anti- infective agent is methotrexate. 21302. The method of item 21171 wherein the anti- infective agent is a podophylotoxin.
21303. The method of item 21171 wherein the anti- infective agent is etoposide.
21304. The method of item 21171 wherein the anti- infective agent is camptothecin.
21305. The method of item 21171 wherein the anti- infective agent is a hydroxyurea.
21306. The method of item 21171 wherein the anti- infective agent is a platinum complex.
21307. The method of item 21171 wherein the anti- infective agent is cisplatin.
21308. The method of item 21171 wherein the composition comprises an antithrombotic agent.
21309. The method of item 21171 wherein the polymer is formed from reactants comprising a naturally occurring polymer.
21310. The method of item 21171 wherein the polymer is formed from reactants comprising protein.
21311. The method of item 21171 wherein the polymer is formed from reactants comprising carbohydrate. 21312. The method of item 21171 wherein the polymer reactants comprising biodegradable polymer.
21313. The method of item 21171 wherein the polymer reactants comprising nonbiodegradable polymer.
21314. The method of item 21171 wherein the polymer reactants comprising collagen.
21315. The method of item 21171 wherein the polymer reactants comprising methylated collagen.
21316. The method of item 21171 wherein the polymer reactants comprising fibrinogen.
21317. The method of item 21171 wherein the polymer reactants comprising thrombin.
21318. The method of item 21171 wherein the polymer reactants comprising blood plasma.
21319. The method of item 21171 wherein the polymer reactants comprising calcium salt.
21320. The method of item 21171 wherein the polymer reactants comprising an antifibronolytic agent.
21321. The method of item 21171 wherein the polymer reactants comprising fibrinogen analog. 21322. The method of item 21171 wherein the polymer reactants comprising albumin.
21323. The method of item 21171 wherein the polymer reactants comprising plasminogen.
21324. The method of item 21171 wherein the polymer reactants comprising von Willebrands factor.
21325. The method of item 21171 wherein the polymer reactants comprising Factor VIII.
21326. The method of item 21171 wherein the polymer reactants comprising hypoallergenic collagen.
21327. The method of item 21171 wherein the polymer reactants comprising atelopeptidic collagen.
21328. The method of item 21171 wherein the polymer reactants comprising telopeptide collagen.
21329. The method of item 21171 wherein the polymer reactants comprising crosslinked collagen.
21330. The method of item 21171 wherein the polymer reactants comprising aprotinin.
21331. The method of item 21171 wherein the polymer reactants comprising epsilon-amino-n-caproic acid. 21332. The method of item 21171 wherein the polymer is formed from reactants comprising gelatin.
21333. The method of item 21171 wherein the polymer is formed from reactants comprising protein conjugates.
21334. The method of item 21171 wherein the polymer is formed from reactants comprising gelatin conjugates.
21335. The method of item 21171 wherein the polymer is formed from reactants comprising a synthetic polymer.
21336. The method of item 21171 wherein the polymer is formed from reactants comprising a synthetic isocyanate-containing compound.
21337. The method of item 21171 wherein the polymer is formed from reactants comprising a synthetic thiol-containing compound.
21338. The method of item 21171 wherein the polymer is formed from reactants comprising a synthetic compound containing at least two thiol groups.
21339. The method of item 21171 wherein the polymer is formed from reactants comprising a synthetic compound containing at least three thiol groups.
21340. The method of item 21171 wherein the polymer is formed from reactants comprising a synthetic compound containing at least four thiol groups. 21341. The method of item 21171 wherein the polymer is formed from reactants comprising a synthetic amino-containing compound.
21342. The method of item 21171 wherein the polymer is formed from reactants comprising a synthetic compound containing at least two amino groups.
21343. The method of item 21171 wherein the polymer is formed from reactants comprising a synthetic compound containing at least three amino groups.
21344. The method of item 21171 wherein the polymer is formed from reactants comprising a synthetic compound containing at least four amino groups.
21345. The method of item 21171 wherein the polymer is formed from reactants comprising a synthetic compound comprising a carbonyl-oxygen-succinimidyl group.
21346. The method of item 21171 wherein the polymer is formed from reactants comprising a synthetic compound comprising at least two carbonyl-oxygen-succinimidyl groups.
21347. The method of item 21171 wherein the polymer is formed from reactants comprising a synthetic compound comprising at least three carbonyl-oxygen-succinimidyl groups.
21348. The method of item 21171 wherein the polymer is formed from reactants comprising a synthetic compound comprising at least four carbonyl-oxygen-succinimidyl groups. 21349. The method of item 21171 wherein the polymer is formed from reactants comprising a synthetic polyalkylene oxide- containing compound.
21350. The method of item 21171 wherein the polymer is formed from reactants comprising a synthetic compound comprising both polyalkylene oxide and biodegradable polyester blocks.
21351. The method of item 21171 wherein the polymer is formed from reactants comprising a synthetic polyalkylene oxide- containing compound having reactive amino groups.
21352. The method of item 21171 wherein the polymer is formed from reactants comprising a synthetic polyalkylene oxide- containing compound having reactive thiol groups.
21353. The method of item 21171 wherein the polymer is formed from reactants comprising a synthetic polyalkylene oxide- containing compound having reactive carbonyl-oxygen-succinimidyl groups.
21354. The method of item 21171 wherein the polymer is formed from reactants comprising a synthetic compound comprising a biodegradable polyester block.
21355. The method of item 21171 wherein the polymer is formed from reactants comprising a synthetic polymer formed in whole or part from lactic acid or lactide.
21356. The method of item 21171 wherein the polymer is formed from reactants comprising a synthetic polymer formed in whole or part from glycolic acid or glycolide. 21357. The method of item 21171 wherein the polymer is formed from reactants comprising polylysine.
21358. The method of item 21171 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound comprising a polyalkylene oxide portion.
21359. The method of item 21171 wherein the polymer is formed from reactants comprising (a) protein and (b) polylysine.
21360. The method of item 21171 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound having at least four thiol groups.
21361. The method of item 21171 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound having at least four amino groups.
21362. The method of item 21171 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound having at least four carbonyl-oxygen-succinimide groups.
21363. The method of item 21171 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound having a biodegradable region formed from reactants selected from lactic acid, lactide, glycolic acid, glycolide, and epison-caprolactone.
21364. The method of item 21171 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound comprising a polyalkylene oxide portion. 21365. The method of item 21171 wherein the polymer is formed from reactants comprising (a) collagen and (b) polylysine.
21366. The method of item 21171 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound having at least four thiol groups.
21367. The method of item 21171 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound having at least four amino groups.
21368. The method of item 21171 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound having at least four carbonyl-oxygen-succinimide groups.
21369. The method of item 21171 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound having a biodegradable region formed from reactants selected from lactic acid, lactide, glycolic acid, glycolide, and epison-caprolactone.
21370. The method of item 21171 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound comprising a polyalkylene oxide portion.
21371. The method of item 21171 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) polylysine.
21372. The method of item 21171 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound having at least four thiol groups. 21373. The method of item 21171 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound having at least four amino groups.
21374. The method of item 21171 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound having at least four carbonyl-oxygen-succinimide groups.
21375. The method of item 21171 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound having a biodegradable region formed from reactants selected from lactic acid, lactide, glycolic acid, glycolide, and epison-caprolactone.
21376. The method of item 21171 wherein the polymer is formed from reactants comprising hyaluronic acid.
21377. The method of item 21171 wherein the polymer is formed from reactants comprising a hyaluronic acid derivative.
21378. The method of item 21171 wherein the polymer is formed from reactants comprising pentaerythritol poly(ethylene glycol)ether tetra-sulfhydryl of number average molecular weight between 3,000 and 30,000.
21379. The method of item 21171 wherein the polymer is formed from reactants comprising pentaerythritol poly(ethylene glycol)ether tetra-amino of number average molecular weight between 3,000 and 30,000.
21380. The method of item 21171 wherein the polymer is formed from reactants comprising (a) a synthetic compound having a number average molecular weight between 3,000 and 30,000 and comprising a polyalkylene oxide region and multiple nucleophilic groups, and (b) a synthetic compound having a number average molecular weight between 3,000 and 30,000 and comprising a polyalkylene oxide region and multiple electrophilic groups.
21381. The method of item 21171 wherein the composition comprises a colorant.
21382. The method of item 21171 wherein the composition is sterile.
21383. A method as in any one of items 21171-21382, wherein the device is a ventriculopleural shunt.
21384. A method as in any one of items 21171-21382, wherein the device is a jugular vein shunt.
21385. A method as in any one of items 21171-21382, wherein the device is a vena cava shunt.
21386. A method as in any one of items 21171-21382, wherein the device is a ventriculoperitoneal shunt.
21387. A method as in any one of items 21171-21382, wherein the device is a gallbladder shunt.
21388. A method as in any one of items 21171-21382, wherein the device is a peritoneum shunt. 21389. A method as in any one of items 21171-21382, wherein the device is an external ventricular drainage device.
21390. A method as in any one of items 21171-21382, wherein the device is an intracranial pressure monitoring device.
21391. A method as in any one of items 21171-21382, wherein the device is a dural patch.
21392. A method as in any one of items 21171 -21382, , wherein the device is an implant to prevent epidural fibrosis post- laminectomy.
21393. A method as in any one of items 21171-21382, wherein the device is a device for continuous subarachnoid infusion.
21394. A method as in any one of items 21171-21382, wherein the device is a drainage shunt useful for draining fluids in the brain.
21395. A method as in any one of items 21171-21382, wherein the device is a pressure monitoring device.
21396. A method for implanting a medical device comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with i) an anti-fibrotic agent, ii) an anti-infective agent, iii) a polymer, iv) a composition comprising an anti-fibrotic agent and a polymer, v) a composition comprising an anti-infective agent and a polymer, or vi) a composition comprising an anti-fibrotic agent, an anti- infective agent and a polymer, and (b) implanting the medical device into the host, wherein the medical device is an inferior vena cava filter. 21397. The method of item 21396 wherein the anti- fibrotic agent is an adensosine A2A receptor antagonist.
21398. The method of item 21396 wherein the anti- fibrotic agent is an AKT inhibitor.
21399. The method of item 21396 wherein the anti- fibrotic agent is an alpha 2 integrin antagonist, wherein the alpha 2 integrin antagonist is Pharmaprojects No. 5754 (Merck KgaA).
21400. The method of item 21396 wherein the anti- fibrotic agent is an alpha 4 integrin antagonist.
21401. The method of item 21396 wherein the anti- fibrotic agent is an alpha 7 nicotinic receptor agonist.
21402. The method of item 21396 wherein the anti- fibrotic agent is an angiogenesis inhibitor selected from the group consisting of AG-12,958 (Pfizer), ATN-161 (Attenuon LLC), neovastat, an angiogenesis inhibitor from Jerina AG (Germany), NM-3 (Mercian), VGA-1155 (Taisho), FCE-26644 (Pfizer), FCE-26950 (Pfizer), FPMA (Meiji Daries), FFM 11142 (Fujisawa), GGTI-298, GM-1306 (Ligand), GPA-1734 (Novartis), NNC-47- 0011 (Novo Nordisk), herbamycin (Nippon Kayaku), lenalidomide (Celegene), IP-10 (NIH), ABT-828 (Abbott), KIN-841 (Tokushima University, Japan), SF-1126 (Semafore Pharmaceuticals), laminin technology (NIH), CHIR-258 (Chiron), NVP-AEW541 (Novartis), NVP-AEW541 (Novartis), Vt16907 (Alchemia), OXI-8007 (Oxigene), EG-3306 (Ark Therapeutics), Maspin (Arriva), ABT-567 (Abbott), PPI-2458 (Praecis Pharmaceuticals), CC-5079, CC-4089 (Celgene), HIF-1 alpha inhibitors (Xenova), S-247 (Pfizer), AP-23573 (Ariad), AZD-9935 (Astra Zeneca), mebendazole (Introgen Therapeutics), MetAP-2 inhibitors (GlaxoSmithKline), AG-615 (Angiogene Pharmaceuticals), Tie-2 antagonists (Hybrigenics), NC-381 , CYC-381 , NC-169, NC-219, NC-383, NC-384, NC-407 (Lorus Therapeutics), ATN-224 (Attenuon), ON-01370 (Onconova), Vitronectin antagonists (Amgen), SDX-103 (Saimedix), Vitronectin antagonists (Shire), CHP (Riemser), TEK (Amgen), Anecortave acetate (Alcon), T46.2 (Matrix Therapeutics), HG-2 (Heptagen), TEM antagonists (Genzyme), Oxi-4500 (Oxigene), ATN-161 (Attenuon), WX-293 (Wilex), M-2025 (Metris Therapeutics), Alphastatin (BioActa), YH-16 (Yantai Rongchang), BIBF- 1120 (Boehringer Ingelheim), BAY-57-9352 (Bayer), AS-1404 (Cancer Research Technology), SC-77964 (Pfizer), glycomimetics (BioTie Therapies), TIE-2 Inhibitors (Ontogen), DIMI, Octamer (Octamer), ABR- 215050 (Active Biotech), ABT-518 (Abbott), KDR inhibitors (Abbott), BSF- 466895 (Abbott), SCH-221153 (Schering-Plough), DAC.antiangiogenic (ConjuChem), TFPI (EntreMed), AZD-2171 (Astra-Zenaca), CDC-394 (Celgene), LY290293 (EIi Lilly), IDN-5390 (Indena), Kdr Kinase Inhibitors (Merck), CT-113020, CT-116433, CT-116563, CT-31890, CT-32228) (Cell Therapeutics), A-299620 (Abbott), TWEAK Inhibitor (Amgen), VEGF modulators (Johnson and Johnson), Tum-N53, tumstatin (Genzyme), Thios- 1 , Thios-2 (Thios Pharmaceuticals), MV-6401 (Miravant Medical Technologies), Spisulosine (PharmaMar), CEP-7055 (Cephalon), AUV-201 (Auvation), LM-609 (EIi Lilly), SKF-106615 (AnorMED), Oglufanide disodium (Cytran), BW-114 (Pharminox), Calreticulin (NIH), WX-678 (Wilex), SD-7784 (Pfizer), WX-UK1 (Wilex), SH-268 (Schering AG), 2-Me-PGA (Celgene), S- 137 (Pfizer), ZD-6126 (Angiogene Pharmaceuticals), SG-292 (SignalGen), Benefin (Lane Labs), A6, A36 (Angstrom), SB-2723005 (GlaxoSmithKline), SC-7 (Cell Therapeutics), ZEN-014 (AEterna Zentaris), 2-methoxyestradiol (EntreMed), NK-130119 (Nippon Kayaku), CC-10004 (Celgene), AVE- 8062A (Ajinomoto), Tacedinaline (Pfizer), Actinonin (Tokyo Metropolitan Institute of Medical Science), Lenalidomide (Celgene), VGA-1155, BTO-956 (SRI International), ER-68203-00 (Eisai), CT-6685 (UCB), JKC-362 (Phoenix Pharmaceuticals), DMI-3798 (DMI Biosciences, Angiomate (Ipsen), ZD-6474 (AstraZeneca), CEP-5214 (Cephalon), Canstatin (Genzyme), NM-3 (Mercian), Oridigm (MediQuest Therapeutics), Exherin (Adherex), BLS-0597 (Boston Life Sciences), PTC-299 (PTC Therapeutics), NPI-2358 (Nereus Pharmaceuticals), CGP-79787 (Novartis), AEE-788 (Novartis), CKD-732 (Chong Kun Dang), CP-564959 (OSI Pharmaceuticals), CM-101 (CarboMed), CT-2584, CT3501 (Cell Therapeutics), combretastatin and analogues and derivatives thereof (Oxigene), Rebimastat (Bristol-Meyers Squibb), Dextrin 2-sulfate (ML Laboratories), Cilengitide (Merk KGaA), NSC-706704 (Pharminox), KRN- 951 (Kirin Brewery), Ukrain, NSC-631570 (Nowicky Pharma), Tecogalan sodium (Daiichi Pharmaceutical), Tz-93 (Tsumura), TBC-1635 (Encysive Pharmaceuticals), TAN-1120 (Takeda), Semaxanib (Pfizer), BDI-7800 (Biopharmacopae), SD-186, SD-983 (Bristol-Meyers Squibb), SB-223245 (GlaxoSmithKline), SC-236 (Pfizer), RWJ-590973 (Johnson and Johnson), ILX-1850 (Genzyme), SC-68488, S-836 (Pfizer), CG-55069-11 (CuraGen), Ki-23057 (Kirin Brewery), CCX-700 (Chemoentryx), Pegaptanib octasodium (Giled Sciences), ANGIOCOL (available from Biostratum Inc.), or an analogue or derivative thereof.
21403. The method of item 21396 wherein the anti- fibrotic agent is an apoptosis antagonist.
21404. The method of item 21396 wherein the anti- fibrotic agent is an apoptosis activator.
21405. The method of item 21396 wherein the anti- fibrotic agent is a beta 1 integrin antagonist.
21406. The method of item 21396 wherein the anti- fibrotic agent is a beta tubulin inhibitor. 21407. The method of item 21396 wherein the anti- fibrotic agent is a blocker of enzyme production in Hepatitis C.
21408. The method of item 21396 wherein the anti- fibrotic agent is a Bruton's tyrosine kinase inhibitor.
21409. The method of item 21396 wherein the anti- fibrotic agent is a calcineurin inhibitor.
21410. The method of item 21396 wherein the anti- fibrotic agent is a caspase 3 inhibitor.
21411. The method of item 21396 wherein the anti- fibrotic agent is a CC chemokine receptor antagonist.
21412. The method of item 21396 wherein the anti- fibrotic agent is a cell cycle inhibitor selected from the group consisting of SNS-595 (Sunesis), synthadotin, KRX-0403, and an analogue or derivative thereof.
21413. The method of item 21396 wherein the anti- fibrotic agent is a cathepsin B inhibitor.
21414. The method of item 21396 wherein the anti- fibrotic agent is a cathepsin K inhibitor, wherein the cathepsin K inhibitor is 462795 (GlaxoSmithKline), INPL-022-D6 (Amura Therapeutics), or an analogue or derivative thereof.
21415. The method of item 21396 wherein the anti- fibrotic agent is a cathepsin L inhibitor. 21416. The method of item 21396 wherein the anti- fibrotic agent is a CD40 antagonist.
21417. The method of item 21396 wherein the anti- fibrotic agent is a chemokine receptor agonist.
21418. The method of item 21396 wherein the anti- fibrotic agent is a chymase inhibitor.
21419. The method of item 21396 wherein the anti- fibrotic agent is a collagenase antagonist.
21420. The method of item 21396 wherein the anti- fibrotic agent is a CXCR antagonist.
21421. The method of item 21396 wherein the anti- fibrotic agent is a cyclin dependent kinase inhibitor selected from the group consisting of a CDK-1 inhibitor, a CDK-2 inhibitor, a CDK- 4 inhibitor, a CDK-6 inhibitor, a CAK1 inhibitor from GPC Biotech and Bristol-Myers Squibb, RGB-286199 (GPC Biotech), an anticancer agent from Astex Technology, a CAK1 inhibitor from GPC Biotech, a CDK inhibitor from Sanofi-Aventis, a CDK1/CDK2 inhibitor from Amgen, a CDK2 inhibitor from SUGEN-2 (Pfizer), a hearing loss therapy agent (Sound Pharmaceuticals), PD-0332991 (Pfizer), RGB-286199 (GPC Biotech), Ro-0505124 (Hoffmann- La Roche), a Ser/Thr kinase inhibitor from Lilly (EIi Lilly), CVT-2584 (CAS No. 199986-75-9) (CV Therapeutics), and an analogue or derivative thereof.
21422. The method of item 21396 wherein the anti- fibrotic agent is a cyclooxygenase 1 inhibitor. 21423. The method of item 21396 wherein the anti- fibrotic agent is a DHFR inhibitor.
21424. The method of item 21396 wherein the anti- fibrotic agent is a dual integrin inhibitor.
21425. The method of item 21396 wherein the anti- fibrotic agent is an elastase inhibitor.
21426. The method of item 21396 wherein the anti- fibrotic agent is an elongation factor-1 alpha inhibitor.
21427. The method of item 21396 wherein the anti- fibrotic agent is an endothelial growth factor antagonist.
21428. The method of item 21396 wherein the anti- . fibrotic agent is an endothelial growth factor receptor kinase inhibitor selected from the group consisting of sorafenib tosylate (Bayer), AAL-993 (Novartis), ABP-309 (Novartis), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EXEL- 2880 (Exelixis), GW-654652 (GlaxoSmithKline), a KDR inhibitor from LG Life Sciences, CT-6685 and CT-6729 (UCB), KRN-633 and KRN-951 (Kirin Brewery), OSI-930 (OSI Pharmaceuticals), SP-5.2 (Supratek Pharma), SU- 11657 (Pfizer), a Tie-2 antagonist (Hybrigenics), a VEGFR-2 kinase inhibitor (Bristol-Myers Squibb), XL-647 (Exelixis), a KDR inhibitor from Abbott Laboratories, sorafenib tosylate, and an analogue or derivative thereof.
21429. The method of item 21396 wherein the anti- fibrotic agent is an endotoxin antagonist. 21430. The method of item 21396 wherein the anti- fibrotic agent is an epothilone and tubulin binder.
21431. The method of item 21396 wherein the anti- fibrotic agent is an estrogen receptor antagonist.
21432. The method of item 21396 wherein the anti- fibrotic agent is an FGF inhibitor.
21433. The method of item 21396 wherein the anti- fibrotic agent is a farnexyl transferase inhibitor.
21434. The method of item 21396 wherein the anti- fibrotic agent is famesyltransferase inhibitor selected from the group of A- 197574 (Abbott), a famesyltransferase inhibitor from Servier, FPTIII (Strathclyde Institute for Drug R), LB-42908 (LG Life Sciences), Pharmaprojects No. 5063 (Genzyme), Pharmaprojects No. 5597 (Ipsen), Yissum Project No. B-1055 (Yissum), and an analogue or derivative thereof.
21435. The method of item 21396 wherein the anti- fibrotic agent is an FLT-3 kinase inhibitor.
21436. The method of item 21396 wherein the anti- fibrotic agent is an FGF receptor kinase inhibitor.
21437. The method of item 21396 wherein the anti- fibrotic agent is a fibrinogen antagonist selected from the group consisting of AUV-201 (Auvation), MG-13926 (Sanofi-Aventis), plasminogen activator (CAS No. 105913-11-9) (from Sanofi-Aventis or UCB), plasminogen activator-2 (tPA-2) (Sanofi-Aventis), pro-urokinase (CAS No. 82657-92-9) (Sanofi-Aventis), and an analogue or derivative thereof. 21438. The method of item 21396 wherein the anti- fibrotic agent is a heat shock protein 90 antagonist selected from the group consisting of SRN-005 (Sirenade), geldanamycin, NSC-33050 (17- allylaminogeldanamycin; 17-AAG), 17-dimethylaminoethylamino-17- demethoxy-geldanamycin (17-DMAG), rifabutin (rifamycin XIV, 1',4- didehydro-1-deoxy-1 ,4-dihydro-5'-(2-methylpropyl)-1-oxo-), and an analogue or derivative thereof.
21439. The method of item 21396 wherein the anti- fibrotic agent is a histone deacetylase inhibitor.
21440. The method of item 21396 wherein the anti- fibrotic agent is an HMGCoA reductase inhibitor selected from the group consisting of an atherosclerosis therapeutic from Lipid Sciences, ATM 6000 (ARYx Therapeutics), KS-01-019 (Kos Pharmaceuticals), Pharmaprojects No. 2197 (Sanofi-Aventi), RP 61969 (Sanofi-Aventis), and an analogue or derivative thereof.
21441. The method of item 21396 wherein the anti- fibrotic agent is an ICAM inhibitor.
21442. The method of item 21396 wherein the anti- fibrotic agent is an IL, ICE and IRAK antagonist, wherein the antagonist is a CJ-14877, CP-424174 (Pfizer), NF-61 (Negma-Lerads), and an analogue or derivative thereof.
21443. The method of item 21396 wherein the anti- fibrotic agent is an IL-2 inhibitor.
21444. The method of item 21396 wherein the anti- fibrotic agent is an immunosuppressant selected from the group consisting of teriflunomide (Sanofi Aventis), chlorsulfaquinoxalone (NSC-339004), chlorsulfaquinoxalone sulfate, CS-712 (Sankyo), ismomultin alfa (CAS No. 457913-93-8) (Akzo Nobel), antiallergics from GenPat77, antiinflammatories or AT-005 (Androclus Therapeutics), autoimmune disease therapeutics from EpiVax, BN-007 (Bone), budesonide (CAS No. 51333-22- 3) (MAP Pharmaceuticals), CO-14 (Genzyme), edratide (CAS No. 433922- 67-9) (Teva), EP-314 (Enanta), eprovafen (CAS No. 101335-99-3) (Sanofi- Aventis), HWA-131 (CAS No. 118788-41-3) (Sanofi-Aventis), immunomodulators from MerLion Pharmaceuticals, immunosuppressives from Alchemia, IPL-12 (Inflazyme), MDL-9563 (CAS No. 27086-86-8) (Sanofi-Aventis), Pharmaprojects No. 2330 (Sanofi-Aventis), Pharmaprojects No. 6426 (Abgenix), PXS-25 (Pharmaxis), rosmarinic acid (CAS No. 20283-92-5) (Sanofi-Aventis), RP 42927 or RP 54745 (CAS No. 135330-08-4) (Sanofi-Aventis), SGN-35 (Seattle Genetics), ST-1959 (Sigma-Tau), type I diabetes therapy from SYNX Pharma, UNIL-88 (Debiopharm), VP-025 (Vasogen), VR-694 (Vectura), PRTX-001 (Protalex), and an analogue or derivative thereof.
21445. The method of item 21396 wherein the anti- fibrotic agent is an IMPDH (inosine monophosphate).
21446. The method of item 21396 wherein the anti- fibrotic agent is an integrin antagonist.
21447. The method of item 21396 wherein the anti- fibrotic agent is an interleukin antagonist.
21448. The method of item 21396 wherein the anti- fibrotic agent is an inhibitor of type ill receptor tyrosine kinase. 21449. The method of item 21396 wherein the anti- fibrotic agent is an irreversible inhibitor of enzyme methionine aminopeptidase type 2.
21450. The method of item 21396 wherein the anti- fibrotic agent is an isozyme selective delta protein kinase C inhibitor.
21451. The method of item 21396 wherein the anti- fibrotic agent a JAK3 enzyme inhibitor.
21452. The method of item 21396 wherein the anti- fibrotic agent is a JNK inhibitor.
21453. The method of item 21396 wherein the anti- fibrotic agent is a kinase inhibitor.
21454. The method of item 21396 wherein the anti- fibrotic agent is kinesin antagonist.
21455. The method of item 21396 wherein the anti- fibrotic agent is a kinesin antagonist.
21456. The method of item 21396 wherein the anti- fibrotic agent is a leukotriene inhibitor and antagonist selected from the group consisting of ambicromil (CAS No. 58805-38-2) (Sanofi-Aventis), amelubant (CAS No. 346735-24-8) (Boehringer Ingeiheim), DW-1141 (Dong Wha), ebselen (Daiichi Pharmaceutical), ibudilast (Kyorin), leucotriene inhibitors from Sanofi-Aventis, lymphotoxin-beta receptor (LT-(H) from Biogen Idee, Pharmaprojects No. 1535 and 2728 (CAS No. 119340-33-9) (Sanofi-Aventis), R-112 (Rigel), Rev-5367 (CAS No. 92532-05-3) (Sanofi- Aventis), RG-14893 (CAS No. 141835-49-6) (Sanofi-Aventis), RG-5901-A (CAS No. 101910-24-1), 92532-23-5, RP 66153 (CAS No. 142422-79-5), RP 66364 (CAS No.186916-92-5), RP 69698 (CAS No. 141748-00-7) (Sanofi-Aventis), SC-411930 (Pfizer), SC-41930 (CAS No. 120072-59-5) (Pfizer), SC-50605 (CAS No. 138828-39-4) (Pfizer), SC-51146 (CAS No. 141059-52-1), SC-53228 (CAS No. 153633-01-3) (Pfizer), spaglumic acid (ZY-15106) (CAS No.3106-85-2), 80619-64-3 (Novartis), tipredane (CAS No. 85197-77-9) (Bristol-Myers Squibb), U-75302 (CAS No. 119477-85-9) (Pfizer), and analogue or derivative thereof.
21457. The method of item 21396 wherein the anti- fibrotic agent is an MAP kinase inhibitor.
21458. The method of item 21396 wherein the anti- fibrotic agent is a matrix metalloproteinase inhibitor.
21459. The method of item 21396 wherein the anti- fibrotic agent is an MCP-CCR2 inhibitor.
21460. The method of item 21396 wherein the anti- fibrotic agent is an mTOR inhibitor.
21461. The method of item 21396 wherein the anti- fibrotic agent is an mTOR kinase inhibitor.
21462. The method of item 21396 wherein the anti- fibrotic agent is a microtubule inhibitor selected from the group consisting of antibody-maytansinoid conjugates from Biogen Idee, colchicines (MantiCore Pharmaceuticals), anticancer immunoconjugates from Johnson & Johnson, DIME from Octamer, gni-1f (GNI), huC242-DM4, huMy9-6-DM1 (ImmunoGen), IDN-5404 (Indena), IMO-098, IMOderm (Imotep), mebendazole (Introgen Therapeutics), microtubule poisons from Cambridge Enterprise, paclitaxel such as LOTAX from Aphios (CAS No. 33069-62-4), Genexol-PM from Samyang, Pharmaprojects No. 6383 (Tapestry Pharmaceuticals), RPR-112378 (Sanofi-Aventis), SGN-75 (Seattle Genetics), SPL-7435 (Starpharma), SSR-250411 (Sanofi-Aventis), trastuzυmab-DM1 (Genentech), vinorelbine, and an analogue or derivative thereof.
21463. The method of item 21396 wherein the anti- fibrotic agent is an MIF inhibitor.
21464. The method of item 21396 wherein the anti- fibrotic agent is an MMP inhibitor.
21465. The method of item 21396 wherein the anti- fibrotic agent is a neurokinin (NK) antagonist selected from the group consisting of anthrotainin (CAS No. 148084-40-6) (Sanofi-Aventis), an IBS thereapeutic from ArQuIe, MDL-105212A (CAS No. 167261-60-1) (Ssanofi- Aventis), Pharmaprojects No. 2744, 3258 (CAS No. 139167-47-8) 4006, 4201, or 5986 (Sanofi-Aventis), RP 67580 (CAS No. 135911-02-3), SR- 144190 (CAS No. 201152-86-5), SSR-240600, SSR-241586 (Sanofi- Aventis), TKA-457 (Novartis), vestipitant mesylate (CAS No. 334476-64-1) (GlaxoSmithKline), Win-64821 (Sanofi-Aventis), PRX-96026 (Predix Pharmaceuticals), and an analogue or derivative thereof.
21466. The method of item 21396 wherein the anti- fibrotic agent is an NF kappa B inhibitor selected from the group consisting of AVE-0545 or AVE-0547 (Sanofi-Aventis), bortezomib (CAS No. 179324- 69-7) (Millennium Pharmaceuticals), dexanabinol (CAS No. 112924-45-5) (Pharmos), dexlipotam (Viatris), Pharmaprojects No. 6283 (INDRA) (OXiGENE), IPL-576092 (CAS No. 137571-30-3) (Inflazyme), NFKB decoy (Corgentech), NFKB decoy oligo (AnGes MG), NFKB's from Ariad, osteoporosis treatments or S5 (F005) from Fulcrum Pharmaceuticals, P61 (Phytopharm), R-flurbiprofen (CAS No. 5104-49-4) (Encore Pharmaceuticals), and an analogue or derivative thereof.
21467. The method of item 21396 wherein the anti- fibrotic agent is a nitric oxide agonist.
21468. The method of item 21396 wherein the anti- fibrotic agent is an ornithine decarboxylase inhibitor.
21469. The method of item 21396 wherein the anti- fibrotic agent is a p38 MAP kinase inhibitor selected from the group consisting of AZD-6703 (AstraZeneca), JX-401 (Jexys Pharmaceuticals), BMS-2 (Bristol-Myers Squibb), a p38 MAP kinase inhibitor from Novartis, a p38-alpha MAP kinase inhibitor from Amphora, Pharmaprojects No. 5704 (Pharmacopeia), RPR-200765A (Sanofi-Aventis), SD-282 (Johnson & Johnson), TAK-715 (Takeda), and an analogue or derivative thereof.
21470. The method of item 21396 wherein the anti- fibrotic agent is a palmitoyl-protein thioesterase inhibitor.
21471. The method of item 21396 wherein the anti- fibrotic agent is a PDGF receptor kinase inhibitor selected from the group consisting of AAL-993, AMN-107, or ABP-309 (Novartis), AMG-706 (Amgen), BAY-57-9352 (Bayer), CDP-860 (UCB), E-7080 (Eisai), imatinib (CAS No. 152459-95-5) (Novartis), OSI-930 (OSI Pharmaceuticals), RPR- 127963E (Sanofi-Aventis), RWJ-540973 (Johnson & Johnson), sorafenib tosylate (Bayer), SU-11657 (Pfizer), tandutinib (CAS No. 387867-13-2) (Millennium Pharmaceuticals), vatalanib (Novartis), ZK-CDK (Schering AG), and an analogue or derivative thereof. 21472. The method of item 21396 wherein the anti- fibrotic agent is (-)-halofenate (Metabolex), AMG-131 (Amgen), antidiabetics from Japan Tobacco, AZD-4619, AZD-8450, AZD-8677 (AstraZeneca), DRF-10945, balaglitazone (Dr Reddy's), CS-00088, CS-00098 (Chipscreen Biosciences), E-3030 (Eisai), etalocib (CAS No. 161172-51-6) (Eli Lilly), GSK-641597 (Ligand), GSK-677954 (GlaxoSmithKline), GW-409544 (Ligand), GW-590735 (GlaxoSmithKline), K-111 (Hoffmann-La Roche), LY- 518674 (EIi Lilly), LY-674 (Ligand), LY-929 (Ligand), MC-3001, MC-3002 (MaxoCore Pharmaceuticals), metformin HCl + pioglitazone (CAS No. 1115- 70-4 and 112529-15-4), ACTOPLUS MET from Andrx), muraglitazar (CAS No. 331741-94-7) (Bristol-Myers Squibb), naveglitazar (Ligand), oleoylethanolamide (Kadmus Pharmaceuticals), ONO-5129, pioglitazone hydrochloride (CAS No. 111025-46-8 and 112529-15-4) (Takeda), PLX-204 (Plexxikon), PPAR agonists from Genfit, PPAR delta agonists from EIi Lilly, PPAR-alpha agonists from CrystalGenomics, PPAR-gamma modulators and PPAR-β modulators from CareX, rosiglitazone maleate (CAS No. 122324- 73-4 or 155141-29-0) (GlaxoSmithKline), rosiglitazone maleate/glimepir (CAS No. 155141-29-0 and 93479-97-1), AVANDARYL, rosiglitazone maleate/metformin extend (CAS No. 155141-29-0 and 657-24-9), AVANDAMET, rosiglitazone maleate+metformin, AVANDAMET (GlaxoSmithKline), tesaglitazar (AstraZeneca), and an analogue or derivative thereof.
21473. The method of item 21396 wherein the anti- fibrotic agent is a phosphatase inhibitor.
21474. The method of item 21396 wherein the anti- fibrotic agent is a phosphodiesterase (PDE) inhibitor selected from the group consisting of avanafil (Tanabe Seiyaku), dasantafil (CAS No. 569351- 91-3) (Schering-Plough), A-906119 (CAS No. 134072-58-5), DL-850 (Sanofi-Aventis), GRC-3015, GRC-3566, GRC-3886 (Glenmark), HWA-153 (CAS No. 56395-66-5) (Sanofi-Aventis), hydroxypumafentrine (Altana), 1BFB-130011 , IBFB-14-016, IBFB-140301 , IBFB-150007, IBFB-211913 (IBFB Pharma), L-826141 (Merck & Co), medorinone (CAS No. 88296-61-1) (Sanofi-Aventis), MEM-1917 (Memory Pharmaceuticals), ND-1251 (Neuro3d), PDE inhibitors from ICOS, PDE IV inhibitors from Memory Pharmaceuticals and CrystalGenomics, Pharmaprojects No. 2742 and 6141 (Sanofi-Aventis), QAD-171 (Novartis), RHC-2963 (CAS No. 76993-12-9 and 76993-14-1), RPR-117658, RPR-122818 derivatives, SR-24870 , and RPR- 132294 (Sanofi-Aventis), SK-350 (ln2Gen), stroke therapy agents from deCODE Genetics, TAS-203 (Taiho), tofimilast (CAS No. 185954-27-2) (Pfizer), UK-371800 (Pfizer), WIN-65579 (CAS No. 158020-82-7) (Sanofi- Aventis), IBFB-130020 (IBFB Pharma), OPC-6535 (CAS No. 145739-56-6) (Otsuka), a phosphodiesterase III inhibitor, enoximone, a phosphodiesterase IV inhibitor, fosfosal, Atopik (Barrier Therapeutics), triflusal, a phosphodiesterase V inhibitor, and an analogue or derivative thereof.
21475. The method of item 21396 wherein the anti- fibrotic agent is a PKC inhibitor.
21476. The method of item 21396 wherein the anti- fibrotic agent is a platelet activating factor antagonist.
21477. The method of item 21396 wherein the anti- fibrotic agent is a platelet-derived growth factor receptor kinase inhibitor.
21478. The method of item 21396 wherein the anti- fibrotic agent is a prolyl hydroxylase inhibitor.
21479. The method of item 21396 wherein the anti- fibrotic agent is a polymorphonuclear neutrophil inhibitor. 21480. The method of item 21396 wherein the anti- fibrotic agent is a protein kinase B inhibitor.
21481. The method of item 21396 wherein the anti- fibrotic agent is a protein kinase C stimulant.
21482. The method of item 21396 wherein the anti- fibrotic agent is a purine nucleoside analogue.
21483. The method of item 21396 wherein the anti- fibrotic agent is a purinoreceptor P2X antagonist.
21484. The method of item 21396 wherein the anti- fibrotic agent is a Raf kinase inhibitor.
21485. The method of item 21396 wherein the anti- - - fibrotic agent is a reversible inhibitor of ErbB1 and ErbB2.
21486. The method of item 21396 wherein the anti- fibrotic agent is a ribonucleoside triphosphate reductase inhibitor.
21487. The method of item 21396 wherein the anti- fibrotic agent is an SDF-1 antagonist.
21488. The method of item 21396 wherein the anti- fibrotic agent is a sheddase inhibitor.
21489. The method of item 21396 wherein the anti- fibrotic agent is an SRC inhibitor. 21490. The method of item 21396 wherein the anti- fibrotic agent is a stromelysin inhibitor.
21491. The method of item 21396 wherein the anti- fibrotic agent is an Syk kinase inhibitor.
21492. The method of item 21396 wherein the anti- fibrotic agent is a telomerase inhibitor.
21493. The method of item 21396 wherein the anti- fibrotic agent is a TGF beta inhibitor selected from the group consisting of pirfenidone (CAS No. 53179-13-8) (MARNAC), tranilast (CAS No. 53902- 12-8) (Kissei), IN-1130 (ln2Gen), mannose-6-phosphate (BTG), TGF-β antagonists from Inflazyme (Pharmaprojects No. 6075), TGF-β antagonists from Sydney, non-industrial source), TGF-β I receptor kinase inhibitors from EIi Lilly, TGF-β receptor inhibitors from Johnson & Johnson, and an analogue or derivative thereof.
21494. The method of item 21396 wherein the anti- fibrotic agent is a TNFα antagonist or TACE inhibitor selected from the group consisting of adalimumab (CAS No. 331731-18-1) (Cambridge Antibody Technology), AGIX-4207 (AtheroGenics), AGT-1 (Advanced Biotherapy), an anti-inflammatory from Borean Pharma, Cellzome, or Paradigm Therapeutics, anti-inflammatory vaccine (TNF-alpha kinoid) from Neovacs, humanized anti-TNF antibody or an anti-TNF MAb (CB0006) Celltech (UCB), apratastat (CAS No. 287445-51-0) (Wyeth), BMS-561392 (Bristol-Myers Squibb), BN-006 (Bone), certolizumab pegol (CAS No. 428863-50-7 or CH-138 (UCB), cilomilast (CAS No. 153259-65-5) (GlaxoSmithKline), CR-1 (Nuada Pharmaceuticals), CRx-119 (CombinatoRx), D-5410 (UCB), dacopafant (CAS No. 125372-33-0) (Sanofi- Aventis), dersalazine (CAS No. 188913-57-7/188913-58-8) (Uriach), etanercept (CAS No. 185243-69-0) (Amgen), ethyl pyruvate (Critical (Critical Therapeutics), golimumab (CAS No. 476181-74-5) (Johnson & Johnson), hormono-immunotherapy from Ipsen, CDP571 (e.g., Humicade from UCB), IC-485 (ICOS), infliximab (CAS No. 170277-31-3) (Johnson & Johnson), IP- 751 (Manhattan Pharmaceuticals), ISIS-104838 (CAS No. 250755-32-9) (ISIS Pharmaceuticals), lenalidomide (CAS No. 191732-72-6) (Celgene), lentinan (CAS No. 37339-90-5) (Ajinomoto), MDL-201112 (CAS No. 142130-73-2) (Sanofi-Aventis), medroxyprogesterone (CAS No. 520-85-4) (InKine Pharmaceutical), N-acetylcysteine (CAS No. 616-91-1) (Zambon), NBE-P2 (DIREVO Biotech), nerelimomab (CAS No. 162774-06-3) (Chiron), OM-294DP (OM PHARMA), onercept (CAS No. 199685-57-9) (Yeda), PASSTNF-alpha (Verigen), pentoxifylline or oxypentifylline (Sanofi-Aventis), Pharmaprojects No. 4091, 4241, 4295, or 4488 (Sanofi-Aventis), Pharmaprojects No. 5480 (Amgen), Pharmaprojects No. 6749 (Cengent), pirfenidone (CAS No. 53179-13-8) (MARNAC), RPR-132294 (Sanofi- Aventis), S5 (F002) (Fulcrum Pharmaceuticals), simvastatin (CAS No. 79902-63-9) (Merck & Co), STA-6292 (Synta Pharmaceuticals), tacrolimus (CAS No. 104987-11-3) (Fujisawa LifeCycle Pharma), talactoferrin alfa (CAS No. 308240-58-6) (Agennix), thalidomide (CAS No. 50-35-1) (Celgene), TNF antagonists form ProStrakan, and Synergen, TNF inhibitors (Amgen), TNF-alpha antagonists from Dynavax Technologies and Jerina AG (Germany), TNF-alpha inhibitors from IBFB Pharma and Xencor (Xencor), torbafylline (CAS No. 105102-21-4) (Sanofi-Aventis), UR-1505 (Uriach), VT-346 (Viron Therapeutics), YSIL6 (Y's Therapeutics), YSTH2 (Y's Therapeutics), NPI-1302a-3 (Nereus Pharmaceuticals, a TNF antagonist from Jerina AG (Germany), dersalazine, and an analogue or derivative thereof.
21495. The method of item 21396 wherein the anti- fibrotic agent is a Toll receptor inhibitor. 21496. The method of item 21396 wherein the anti- fibrotic agent is a tubulin antagonist.
21497. The method of item 21396 wherein the anti- fibrotic agent is a tyrosine kinase inhibitor selected from the group consisting of SU-011248, SUTENT from Pfizer Inc. (New York, NY), BMS-354825, PN- 355 (Paracelsian Pharmaceuticals), AGN-199659 (Allergan), AAL-993 or ABP-309 (Novartis), adaphostin (NIH), AEE-788 (Novartis), AG-013736 (OSI Pharmaceuticals), AG-13736 (Pfizer), ALT-110 (Alteris Therapeutics), AMG-706 (Amgen), anticancer MAbs from Xencor, anti-EGFrvlll MAbs from Abgenix, anti-HER2 MAb from Abiogen, AZD-2171 or AZD-9935 (AstraZeneca), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), CEP-5214 (Cephalon), CEP-7055 (Cephalon), cetuximab (ImClone Systems), CHIR-200131 and CHIR-258 (Chiron), CP-547632 (OSI Pharmaceuticals), CP-724714 (Pfizer), CT-301 (Creabilis Therapeutics), D- 69491 (Baxter International), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EGFR/ErbB2 inhibitors from Array BioPharma, erlotinib (CAS No. 183319- 69-9) (OSI Pharmaceuticals), EXEL-2880 (Exelixis), FK-778 (Sanofi- Aventis) , gefitinib (CAS No. 184475-35-2) (AstraZeneca), GW-2286 or GW- 654652 (GlaxoSmithKline), her2/neu antigen from AlphaVax, HER-2/neu inhibitor from Generex, Herzyme (Medipad) (Sirna Therapeutics), HKI-272 (Wyeth), HuMax-EGFr (Genmab), idronoxil (CAS No. 81267-65-4) (Novogen), IGF-1 inhibitors from Ontogen, IMC-11F8 (ImClone Systems), kahalalide F (CAS No. 149204-42-2) (PharmaMar), KDR inhibitor from LG Life Sciences, KDR inhibitors from Abbott Laboratories, KDR kinase inhibitors (UCB), Kdr kinase inhibitors from Merck & Co, KRN-633 and KRN- 951 (Kirin Brewery), KSB-102 (Xenova), lapatinib ditosylate (CAS No. 388082-78-8) (GlaxoSmithKline), matuzumab (Merck KGaA), MDX-214 (Medarex), ME-103 (Pharmexa), MED-A300 (Gerolymatos), MNAC-13 (Lay Line Genomics), nimotuzumab (Center of Molecular Immunology), NSC- 330507 or NSC-707545 (NIH), NV-50 (Novogen), OSI-930 (OSI Pharmaceuticals), panitumumab (Abgenix), pelitinib (CAS No. 287933-82-7) (Wyeth), pertuzumab (CAS No. 380610-27-5) (Genentech), Pharmaprojects No. 3985 (Sanofi-Aventis), prostate cancer therapeutics from Sequenom (SQPC35, SQPC36, SQPC90), removab and remoxab (Trion Pharma), RG- 13022 (CAS No. 136831-48-6), RG-13291 (CAS No. 138989-50-1), or RG- 14620 (CAS No. 136831-49-7) (Sanofi-Aventis), RM-6427 (Romark), RNAi breast cancer therapy from Benitec, RP 53801 (CAS No. 125882-88-4) (Sanofi-Aventis), sorafenib tosylate (Bayer), SU-11657 (Pfizer), Tie-2 antagonists from Semaia (Hybrigenics), Tie-2 inhibitors from Ontogen, trastuzumab (CAS No. 180288-69-1) (Genentech), tyrosine kinase inhibitors from Sanofi-Aventis, U3-1287, U3-1565, U3-1784, U3-1800 (U3 Pharma), vatalanib (Novartis), VEGFR-2 kinase inhibitor from Bristol-Myers Squibb, XL-647 (Exelixis), ZD-6474 (AstraZeneca), ZK-CDK (Schering AG), an EGFR tyrosine kinase inhibitor, EKB-569 (Wyeth), and an analogue or derivative thereof.
21498. The method of item 21396 wherein the anti- fibrotic agent is a VEGF inhibitor.
21499. The method of item 21396 wherein the anti- fibrotic agent is a vitamin D receptor agonist.
21500. The method of item 21396 wherein the anti- fibrotic agent is ZD-6474 (an angiogenesis inhibitor).
21501. The method of item 21396 wherein the anti- fibrotic agent is AP-23573 (an mTOR inhibitor).
21502. The method of item 21396 wherein the anti- fibrotic agent is synthadotin (a tubulin antagonist). 21503. The method of item 21396 wherein the anti- fibrotic agent is S-0885 (a collagenase inhibitor).
21504. The method of item 21396 wherein the anti- fibrotic agent is aplidine (an elongation factor-1 alpha inhibitor).
21505. The method of item 21396 wherein the anti- fibrotic agent is ixabepilone (an epithilone).
21506. The method of item 21396 wherein the anti- fibrotic agent is IDN-5390 (an angiogenesis inhibitor).
21507. The method of item 21396 wherein the anti- fibrotic agent is SB-2723005 (an angiogenesis inhibitor).
21508. The method of item 21396 wherein the anti- fibrotic agent is ABT-518 (an angiogenesis inhibitor).
21509. The method of item 21396 wherein the anti- fibrotic agent is combretastatin (an angiogenesis inhibitor).
21510. The method of item 21396 wherein the anti- fibrotic agent is anecortave acetate (an angiogenesis inhibitor).
21511. The method of item 21396 wherein the anti- fibrotic agent is SB-715992 (a kinesin antagonist).
21512. The method of item 21396 wherein the anti- fibrotic agent is temsirolimus (an mTOR inhibitor). 21513. The method of item 21396 wherein the anti- fibrotic agent is adalimumab (a TNFα antagonist).
21514. The method of item 21396 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with an anti-fibrotic agent, and (b) implanting the medical device into the host.
21515. The method of item 21396 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with an anti-infective agent, and (b) implanting the medical device into the host.
21516. The method of item 21396 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with a polymer; and (b) implanting the medical device into the host.
21517. The method of item 21396 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with a composition comprising an anti-fibrotic agent and a polymer, and (b) implanting the medical device into the host.
21518. The method of item 21396 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with a composition comprising an anti-infective agent and a polymer, and (b) implanting the medical device into the host.
21519. The method of item 21396 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with a composition comprising an anti-fibrotic agent, an anti- infective agent and a polymer, and (b) implanting the medical device into the host.
21520. The method of item 21396 wherein the anti- infective agent is an anthracycline.
21521. The method of item 21396 wherein the anti- infective agent is doxorubicin.
21522. The method of item 21396 wherein the anti- infective agent is is mitoxantrone.
21523. The method of item 21396 wherein the anti- infective agent is a fluoropyrimidine.
21524. The method of item 21396 wherein the anti- infective agent is 5-fluorouracil (5-FU).
21525. The method of item 21396 wherein the anti- infective agent is a folic acid antagonist.
21526. The method of item 21396 wherein the anti- infective agent is methotrexate.
21527. The method of item 21396 wherein the anti- infective agent is a podophylotoxin.
21528. The method of item 21396 wherein the anti- infective agent is etoposide. 21529. The method of item 21396 wherein the anti- infective agent is camptothecin.
21530. The method of item 21396 wherein the anti- infective agent is a hydroxyurea.
21531. The method of item 21396 wherein the anti- infective agent is a platinum complex.
21532. The method of item 21396 wherein the anti- infective agent is cisplatin.
21533. The method of item 21396 wherein the composition comprises an anti-thrombotic agent.
21534. The method of item 21396 wherein the polymer is formed from reactants comprising a naturally occurring polymer.
21535. The method of item 21396 wherein the polymer is formed from reactants comprising protein.
21536. The method of item 21396 wherein the polymer is formed from reactants comprising carbohydrate.
21537. The method of item 21396 wherein the polymer is formed from reactants comprising biodegradable polymer.
21538. The method of item 21396 wherein the polymer is formed from reactants comprising nonbiodegradable polymer. 21539. The method of item 21396 wherein the polymer reactants comprising collagen.
21540. The method of item 21396 wherein the polymer reactants comprising methylated collagen.
21541. The method of item 21396 wherein the polymer reactants comprising fibrinogen.
21542. The method of item 21396 wherein the polymer reactants comprising thrombin.
21543. The method of item 21396 wherein the polymer reactants comprising blood plasma.
21544. The method of item 21396 wherein the polymer reactants comprising calcium salt.
21545. The method of item 21396 wherein the polymer reactants comprising an antifibronolytic agent.
21546. The method of item 21396 wherein the polymer reactants comprising fibrinogen analog.
21547. The method of item 21396 wherein the polymer reactants comprising albumin.
21548. The method of item 21396 wherein the polymer reactants comprising plasminogen. 21549. The method of item 21396 wherein the polymer reactants comprising von Willebrands factor.
21550. The method of item 21396 wherein the polymer reactants comprising Factor VIlI.
21551. The method of item 21396 wherein the polymer reactants comprising hypoallergenic collagen.
21552. The method of item 21396 wherein the polymer reactants comprising atelopeptidic collagen.
21553. The method of item 21396 wherein the polymer reactants comprising telopeptide collagen.
21554. The method of item 21396 wherein the polymer reactants comprising crosslinked collagen.
21555. The method of item 21396 wherein the polymer reactants comprising aprotinin.
21556. The method of item 21396 wherein the polymer reactants comprising epsilon-amino-n-caproic acid.
21557. The method of item 21396 wherein the polymer reactants comprising gelatin.
21558. The method of item 21396 wherein the polymer reactants comprising protein conjugates. 21559. The method of item 21396 wherein the polymer is formed from reactants comprising gelatin conjugates.
21560. The method of item 21396 wherein the polymer is formed from reactants comprising a synthetic polymer.
21561. The method of item 21396 wherein the polymer is formed from reactants comprising a synthetic isocyanate-containing compound.
21562. The method of item 21396 wherein the polymer is formed from reactants comprising a synthetic thiol-containing compound.
21563. The method of item 21396 wherein the polymer is formed from reactants comprising a synthetic compound containing at least two thiol groups.
21564. The method of item 21396 wherein the polymer is formed from reactants comprising a synthetic compound containing at least three thiol groups.
21565. The method of item 21396 wherein the polymer is formed from reactants comprising a synthetic compound containing at least four thiol groups.
21566. The method of item 21396 wherein the polymer is formed from reactants comprising a synthetic amino-containing compound. 21567. The method of item 21396 wherein the polymer is formed from reactants comprising a synthetic compound containing at least two amino groups.
21568. The method of item 21396 wherein the polymer is formed from reactants comprising a synthetic compound containing at least three amino groups.
21569. The method of item 21396 wherein the polymer is formed from reactants comprising a synthetic compound containing at least four amino groups.
21570. The method of item 21396 wherein the polymer is formed from reactants comprising a synthetic compound comprising a carbonyl-oxygen-succinimidyl group.
21571. The method of item 21396 wherein the polymer is formed from reactants comprising a synthetic compound comprising at least two carbonyl-oxygen-succinimidyl groups.
21572. The method of item 21396 wherein the polymer is formed from reactants comprising a synthetic compound comprising at least three carbonyl-oxygen-succinimidyl groups.
21573. The method of item 21396 wherein the polymer is formed from reactants comprising a synthetic compound comprising at least four carbonyl-oxygen-succinimidyl groups.
21574. The method of item 21396 wherein the polymer is formed from reactants comprising a synthetic polyalkylene oxide- containing compound. 21575. The method of item 21396 wherein the polymer is formed from reactants comprising a synthetic compound comprising both polyalkylene oxide and biodegradable polyester blocks.
21576. The method of item 21396 wherein the polymer is formed from reactants comprising a synthetic polyalkylene oxide- containing compound having reactive amino groups.
21577. The method of item 21396 wherein the polymer is formed from reactants comprising a synthetic polyalkylene oxide- containing compound having reactive thiol groups.
21578. The method of item 21396 wherein the polymer is formed from reactants comprising a synthetic polyalkylene oxide- containing compound having reactive carbonyl-oxygen-succinimidyl groups.
21579. The method of item 21396 wherein the polymer is formed from reactants comprising a synthetic compound comprising a biodegradable polyester block.
21580. The method of item 21396 wherein the polymer is formed from reactants comprising a synthetic polymer formed in whole or part from lactic acid or lactide.
21581. The method of item 21396 wherein the polymer is formed from reactants comprising a synthetic polymer formed in whole or part from glycolic acid or glycolide.
21582. The method of item 21396 wherein the polymer is formed from reactants comprising polylysine. 21583. The method of item 21396 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound comprising a polyaikylene oxide portion.
21584. The method of item 21396 wherein the polymer is formed from reactants comprising (a) protein and (b) polylysine.
21585. The method of item 21396 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound having at least four thiol groups.
21586. The method of item 21396 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound having at least four amino groups.
21587. The method of item 21396 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound having at least four carbonyl-oxygen-succinimide groups.
21588. The method of item 21396 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound having a biodegradable region formed from reactants selected from lactic acid, lactide, glycolic acid, glycolide, and epison-caprolactone.
21589. The method of item 21396 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound comprising a polyaikylene oxide portion.
21590. The method of item 21396 wherein the polymer is formed from reactants comprising (a) collagen and (b) polylysine. 21591. The method of item 21396 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound having at least four thiol groups.
21592. The method of item 21396 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound having at least four amino groups.
21593. The method of item 21396 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound having at least four carbonyl-oxygen-succinimide groups.
21594. The method of item 21396 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound having a biodegradable region formed from reactants selected from lactic acid, lactide, glycolic acid, glycolide, and epison-caprolactone.
21595. The method of item 21396 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound comprising a polyalkylene oxide portion.
21596. The method of item 21396 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) polylysine.
21597. The method of item 21396 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound having at least four thiol groups. 21598. The method of item 21396 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound having at least four amino groups.
21599. The method of item 21396 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound having at least four carbonyl-oxygen-succinimide groups.
21600. The method of item 21396 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound having a biodegradable region formed from reactants selected from lactic acid, lactide, glycolic acid, glycolide, and epison-caprolactone.
21601. The method of item 21396 wherein the polymer is formed from reactants comprising hyaluronic acid.
21602. The method of item 21396 wherein the polymer is formed from reactants comprising a hyaluronic acid derivative.
21603. The method of item 21396 wherein the polymer is formed from reactants comprising pentaerythritol poly(ethylene glycol)ether tetra-sulfhydryl of number average molecular weight between 3,000 and 30,000.
21604. The method of item 21396 wherein the polymer is formed from reactants comprising pentaerythritol poly(ethylene glycol)ether tetra-amino of number average molecular weight between 3,000 and 30,000.
21605. The method of item 21396 wherein the polymer is formed from reactants comprising (a) a synthetic compound having a number average molecular weight between 3,000 and 30,000 and comprising a polyalkylene oxide region and multiple nucleophilic groups, and (b) a synthetic compound having a number average molecular weight between 3,000 and 30,000 and comprising a polyalkylene oxide region and multiple electrophilic groups.
21606. The method of item 21396 wherein the composition comprises a colorant.
21607. The method of item 21396 wherein the composition is sterile.
21608. A method as in any one of items 21396-21607, wherein the device is a vascular filter.
21609. A method as in any one of items 21396-21607, wherein the device is a blood filter.
21610. A method as in any one of items 21396-21607, wherein the device is a caval filter.
21611. A method as in any one of items 21396-21607, wherein the device is a vena cava filter.
21612. A method as in any one of items 21396-21607, wherein the device is a thrombus filter.
21613. A method as in any one of items 21396-21607, wherein the device is an antimigration filter. 21614. A method as in any one of items 21396-21607, wherein the device is a percutaneous filter system.
21615. A method as in any one of items 21396-21607, wherein the device is an intravascular trap.
21616. A method as in any one of items 21396-21607, wherein the device is an intravascular filter.
21617. A method as in any one of items 21396-21607, wherein the device is a clot filter.
21618. A method as in any one of items 21396-21607, wherein the device is a vein filter.
21619. A method as in any one of items 21396-21607, wherein the device is a body vessel filter.
21620. A method for implanting a medical device comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with i) an anti-fibrotic agent, ii) an anti-infective agent, iii) a polymer, iv) a composition comprising an anti-fibrotic agent and a polymer, v) a composition comprising an anti-infective agent and a polymer, or vi) a composition comprising an anti-fibrotic agent, an anti- infective agent and a polymer, and (b) implanting the medical device into the host, wherein the medical device is a gastrointestinal device.
21621. The method of item 21620 wherein the anti- fibrotic agent is an adensosine A2A receptor antagonist. 21622. The method of item 21620 wherein the anti- fibrotic agent is an AKT inhibitor.
21623. The method of item 21620 wherein the anti- fibrotic agent is an alpha 2 integrin antagonist, wherein the alpha 2 integrin antagonist is Pharmaprojects No. 5754 (Merck KgaA).
21624. The method of item 21620 wherein the anti- fibrotic agent is an alpha 4 integrin antagonist.
21625. The method of item 21620 wherein the anti- fibrotic agent is an alpha 7 nicotinic receptor agonist.
21626. The method of item 21620 wherein the anti- fibrotic agent is an angiogenesis inhibitor selected from the group consisting of AG-12,958 (Pfizer), ATN-161 (Attenuon LLC), neovastat, an angiogenesis inhibitor from Jerina AG (Germany), NM-3 (Mercian), VGA-1155 (Taisho), FCE-26644 (Pfizer), FCE-26950 (Pfizer), FPMA (Meiji Daries), FR-111142 (Fujisawa), GGTI-298, GM-1306 (Ligand), GPA-1734 (Novartis), NNC-47- 0011 (Novo Nordisk), herbamycin (Nippon Kayaku), lenalidomide (Celegene), IP-10 (NIH), ABT-828 (Abbott), KIN-841 (Tokushima University, Japan), SF-1126 (Semafore Pharmaceuticals), laminin technology (NIH), CHIR-258 (Chiron), NVP-AEW541 (Novartis), NVP-AEW541 (Novartis), Vt16907 (Alchemia), OXI-8007 (Oxigene), EG-3306 (Ark Therapeutics), Maspin (Arriva), ABT-567 (Abbott), PPI-2458 (Praecis Pharmaceuticals), CC-5079, CC-4089 (Celgene), HIF-1 alpha inhibitors (Xenova), S-247 (Pfizer), AP-23573 (Ariad), AZD-9935 (Astra Zeneca), mebendazole (Introgen Therapeutics), MetAP-2 inhibitors (GlaxoSmithKline), AG-615 (Angiogene Pharmaceuticals), Tie-2 antagonists (Hybrigenics), NC-381 , CYC-381, NC-169, NC-219, NC-383, NC-384, NC-407 (Lorus Therapeutics), ATN-224 (Attenuon), ON-01370 (Onconova), Vitronectin antagonists (Amgen), SDX-103 (Salmedix), Vitronectin antagonists (Shire), CHP (Riemser), TEK (Amgen), Anecortave acetate (Alcon), T46.2 (Matrix Therapeutics), HG-2 (Heptagen), TEM antagonists (Genzyme), Oxi-4500 (Oxigene), ATN-161 (Attenuon), WX-293 (Wiiex), M-2025 (Metris Therapeutics), Alphastatin (BioActa), YH-16 (Yantai Rongchang), BIBF- 1120 (Boehringer Ingeiheim), BAY-57-9352 (Bayer), AS-1404 (Cancer Research Technology), SC-77964 (Pfizer), glycomimetics (BioTie Therapies), TIE-2 Inhibitors (Ontogen), DIMI, Octamer (Octamer), ABR- 215050 (Active Biotech), ABT-518 (Abbott), KDR inhibitors (Abbott), BSF- 466895 (Abbott), SCH-221153 (Schering-Plough), DAC:antiangiogenic (ConjuChem), TFPI (EntreMed), AZD-2171 (Astra-Zenaca), CDC-394 (Celgene), LY290293 (EIi Lilly), IDN-5390 (Indena), Kdr Kinase Inhibitors (Merck), CT-113020, CT-116433, GT-116563, CT-31890, CT-32228) (Cell Therapeutics), A-299620 (Abbott), TWEAK Inhibitor (Amgen), VEGF modulators (Johnson and Johnson), Tum~N53, tumstatin (Genzyme), Thios- 1 , Thios-2 (Thios Pharmaceuticals), MV-6401 (Miravant Medical Technologies), Spisulosine (PharmaMar), CEP-7055 (Cephalon), AUV-201 (Auvation), LM-609 (EIi Lilly), SKF-106615 (AnorMED), Oglufanide disodium (Cytran), BW-114 (Pharminox), Calreticulin (NIH), WX-678 (Wilex), SD-7784 (Pfizer), WX-UK1 (Wilex), SH-268 (Schering AG), 2-Me-PGA (Celgene), S- 137 (Pfizer), ZD-6126 (Angiogene Pharmaceuticals), SG-292 (SignalGen), Benefin (Lane Labs), A6, A36 (Angstrom), SB-2723005 (GlaxoSmithKline), SC-7 (Cell Therapeutics), ZEN-014 (AEterna Zentaris), 2-methoxyestradiol (EntreMed), NK-130119 (Nippon Kayaku), CC-10004 (Celgene), AVE- 8062A (Ajinomoto), Tacedinaline (Pfizer), Actinonin (Tokyo Metropolitan Institute of Medical Science), Lenalidomide (Celgene), VGA-1155, BTO-956 (SRI International), ER-68203-00 (Eisai), CT-6685 (UCB), JKC-362 (Phoenix Pharmaceuticals), DMI-3798 (DMI Biosciences, Angiomate (Ipsen), ZD-6474 (AstraZeneca), CEP-5214 (Cephalon), Canstatin (Genzyme), NM-3 (Mercian), Oridigm (MediQuest Therapeutics), Exherin (Adherex), BLS-0597 (Boston Life Sciences), PTC-299 (PTC Therapeutics), NPI-2358 (Nereus Pharmaceuticals), CGP-79787 (Novartis), AEE-788 (Novartis), CKD-732 (Chong Kυn Dang), CP-564959 (OSl Pharmaceuticals), CM-101 (CarboMed), CT-2584, CT3501 (Cell Therapeutics), combretastatin and analogues and derivatives thereof (Oxigene), Rebimastat (Bristol-Meyers Squibb), Dextrin 2-sulfate (ML Laboratories), Cilengitide (Merk KGaA), NSC-706704 (Pharminox), KRN- 951 (Kirin Brewery), Ukrain, NSC-631570 (Nowicky Pharma), Tecogalan sodium (Daiichi Pharmaceutical), Tz-93 (Tsumura), TBC-1635 (Encysive Pharmaceuticals), TAN-1120 (Takeda), Semaxanib (Pfizer), BDI-7800 (Biopharmacopae), SD-186, SD-983 (Bristol-Meyers Squibb), SB-223245 (GlaxoSmithKline), SC-236 (Pfizer), RWJ-590973 (Johnson and Johnson), ILX-1850 (Genzyme), SC-68488, S-836 (Pfizer), CG-55069-11 (CuraGen), Ki-23057 (Kirin Brewery), CCX-700 (Chemoentryx), Pegaptanib octasodium (Giled Sciences), ANGIOCOL (available from Biostratum Inc.), or an analogue or derivative thereof.
21627. The method of item 21620 wherein the anti- fibrotic agent is an apoptosis antagonist.
21628. The method of item 21620 wherein the anti- fibrotic agent is an apoptosis activator.
21629. The method of item 21620 wherein the anti- fibrotic agent is a beta 1 integrin antagonist.
21630. The method of item 21620 wherein the anti- fibrotic agent is a beta tubulin inhibitor.
21631. The method of item 21620 wherein the anti- fibrotic agent is a blocker of enzyme production in Hepatitis C. 21632. The method of item 21620 wherein the anti- fibrotic agent is a Bruton's tyrosine kinase inhibitor.
21633. The method of item 21620 wherein the anti- fibrotic agent is a calcineurin inhibitor.
21634. The method of item 21620 wherein the anti- fibrotic agent is a caspase 3 inhibitor.
21635. The method of item 21620 wherein the anti- fibrotic agent is a CC chemokine receptor antagonist.
21636. The method of item 21620 wherein the anti- fibrotic agent is a cell cycle inhibitor selected from the group consisting of SNS-595 (Sunesis), synthadotin, KRX-0403, and an analogue or derivative thereof.
21637. The method of item 21620 wherein the anti- fibrotic agent is a cathepsin B inhibitor.
21638. The method of item 21620 wherein the anti- fibrotic agent is a cathepsin K inhibitor, wherein the cathepsin K inhibitor is 462795 (GlaxoSmithKline), INPL-022-D6 (Amura Therapeutics), or an analogue or derivative thereof.
21639. The method of item 21620 wherein the anti- fibrotic agent is a cathepsin L inhibitor.
21640. The method of item 21620 wherein the anti- fibrotic agent is a CD40 antagonist. 21641. The method of item 21620 wherein the anti- fibrotic agent is a chemokine receptor agonist.
21642. The method of item 21620 wherein the anti- fibrotic agent is a chymase inhibitor. „
21643. The method of item 21620 wherein the anti- fibrotic agent is a collagenase antagonist.
21644. The method of item 21620 wherein the anti- fibrotic agent is a CXCR antagonist.
21645. The method of item 21620 wherein the anti- fibrotic agent is a cyclin dependent kinase inhibitor selected from the group consisting of a CDK-1 inhibitor, a CDK-2 inhibitor, a CDK- 4 inhibitor, a CDK-6 inhibitor, a CAK1 inhibitor from GPC Biotech and Bristol-Myers - Squibb, RGB-286199 (GPC Biotech), an anticancer agent from Astex Technology, a CAK1 inhibitor from GPC Biotech, a CDK inhibitor from Sanofi-Aventis, a CDK1/CDK2 inhibitor from Amgen, a CDK2 inhibitor from SUGEN-2 (Pfizer), a hearing loss therapy agent (Sound Pharmaceuticals), PD-0332991 (Pfizer), RGB-286199 (GPC Biotech), Ro-0505124 (Hoffmann- La Roche), a Ser/Thr kinase inhibitor from Lilly (EIi Lilly), CVT-2584 (CAS No. 199986-75-9) (CV Therapeutics), and an analogue or derivative thereof.
21646. The method of item 21620 wherein the anti- fibrotic agent is a cyclooxygenase 1 inhibitor.
21647. The method of item 21620 wherein the anti- fibrotic agent is a DHFR inhibitor. 21648. The method of item 21620 wherein the anti- fibrotic agent is a dual integrin inhibitor.
21649. The method of item 21620 wherein the anti- fibrotic agent is an elastase inhibitor.
21650. The method of item 21620 wherein the anti- fibrotic agent is an elongation factor-1 alpha inhibitor.
21651. The method of item 21620 wherein the anti- fibrotic agent is an endothelial growth factor antagonist.
21652. The method of item 21620 wherein the anti- fibrotic agent is an endothelial growth factor receptor kinase inhibitor selected from the group consisting of sorafenib tosylate (Bayer), AAL-993 (Novartis), ABP-309 (Novartis), BAY-57-9352 (Bayer), BIBF-1120 - (Boehringer Ingelheim), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EXEL- 2880 (Exelixis), GW-654652 (GlaxoSmithKline), a KDR inhibitor from LG Life Sciences, CT-6685 and CT-6729 (UCB), KRN-633 and KRN-951 (Kirin Brewery), OSI-930 (OSI Pharmaceuticals), SP-5.2 (Supratek Pharma), SU-
11657 (Pfizer), a Tie-2 antagonist (Hybrigenics), a VEGFR-2 kinase inhibitor (Bristol-Myers Squibb), XL-647 (Exelixis), a KDR inhibitor from Abbott Laboratories, sorafenib tosylate, and an analogue or derivative thereof.
21653. The method of item 21620 wherein the anti- fibrotic agent is an endotoxin antagonist.
21654. The method of item 21620 wherein the anti- fibrotic agent is an epothilone and tubulin binder. 21655. The method of item 21620 wherein the anti- fibrotic agent is an estrogen receptor antagonist.
21656. The method of item 21620 wherein the anti- fibrotic agent is an FGF inhibitor.
21657. The method of item 21620 wherein the anti- fibrotic agent is a famexyl transferase inhibitor.
21658. The method of item 21620 wherein the anti- fibrotic agent is farnesyltransferase inhibitor selected from the group of A- 197574 (Abbott), a farnesyltransferase inhibitor from Servier, FPTIII (Strathclyde Institute for Drug R), LB-42908 (LG Life Sciences), Pharmaprojects No. 5063 (Genzyme), Pharmaprojects No. 5597 (Ipsen), Yissum Project No. B-1055 (Yissum), and an analogue or derivative thereof.
21659. The method of item 21620 wherein the anti- fibrotic agent is an FLT-3 kinase inhibitor.
21660. The method of item 21620 wherein the anti- fibrotic agent is an FGF receptor kinase inhibitor.
21661. The method of item 21620 wherein the anti- fibrotic agent is a fibrinogen antagonist selected from the group consisting of AUV-201 (Auvation), MG-13926 (Sanofi-Aventis), plasminogen activator (CAS No. 105913-11-9) (from Sanofi-Aventis or UCB), plasminogen activator-2 (tPA-2) (Sanofi-Aventis), pro-urokinase (CAS No. 82657-92-9) (Sanofi-Aventis), and an analogue or derivative thereof.
21662. The method of item 21620 wherein the anti- fibrotic agent is a heat shock protein 90 antagonist selected from the group consisting of SRN-005 (Sirenade), getdanamycin, NSC-33050 (17- allylaminogeldanamycin; 17-AAG), 17-dimethylaminoethylamino-17- demethoxy-geldanamycin (17-DMAG), rifabutin (rifamycin XIV, 1',4- didehydro-1-deoxy-1,4-dihydro-5'-(2-methylpropyl)-1-oxo-), and an analogue or derivative thereof.
21663. The method of item 21620 wherein the anti- fibrotic agent is a histone deacetylase inhibitor.
21664. The method of item 21620 wherein the anti- fibrotic agent is an HMGCoA reductase inhibitor selected from the group consisting of an atherosclerosis therapeutic from Lipid Sciences, ATI-16000 (ARYx Therapeutics), KS-01-019 (Kos Pharmaceuticals), Pharmaprojects No. 2197 (Sanofi-Aventi), RP 61969 (Sanofi-Aventis), and an analogue or derivative thereof.
21665. The method of item 21620 wherein the anti- fibrotic agent is an ICAM inhibitor.
21666. The method of item 21620 wherein the anti- fibrotic agent is an IL, ICE and IRAK antagonist, wherein the antagonist is a CJ-14877, CP-424174 (Pfizer), NF-61 (Negma-Lerads), and an analogue or derivative thereof.
21667. The method of item 21620 wherein the anti- fibrotic agent is an IL-2 inhibitor.
21668. The method of item 21620 wherein the anti- fibrotic agent is an immunosuppressant selected from the group consisting of teriflunomide (Sanofi Aventis), chlorsulfaquinoxalone (NSC-339004), chlorsulfaquinoxalone sulfate, CS-712 (Sankyo), ismomultin alfa (CAS No. 457913-93-8) (Akzo Nobel), antiallergics from GenPat77, antiinflammatories or AT-005 (Androclυs Therapeutics), autoimmune disease therapeutics from EpiVax, BN-007 (Bone), budesonide (CAS No. 51333-22- 3) (MAP Pharmaceuticals), CO-14 (Genzyme), edratide (CAS No. 433922- 67-9) (Teva), EP-314 (Enanta), eprovafen (CAS No. 101335-99-3) (Sanofi- Aventis), HWA-131 (CAS No. 118788-41-3) (Sanofi-Aventis), immunomodulators from MerLion Pharmaceuticals, immunosuppressives from Alchemia, IPL-12 (Inflazyme), MDL-9563 (CAS No. 27086-86-8) (Sanofi-Aventis), Pharmaprojects No. 2330 (Sanofi-Aventis), Pharmaprojects No. 6426 (Abgenix), PXS-25 (Pharmaxis), rosmarinic acid (CAS No. 20283-92-5) (Sanofi-Aventis), RP 42927 or RP 54745 (CAS No. 135330-08-4) (Sanofi-Aventis), SGN-35 (Seattle Genetics), ST-1959 (Sigma-Tau), type I diabetes therapy from SYNX Pharma, UNlL-88 (Debiopharm), VP-025 (Vasogen), VR-694 (Vectura), PRTX-001 (Protalex), and an analogue or derivative thereof.
21669. The method of item 21620 wherein the anti- fibrotic agent is an IMPDH (inosine monophosphate).
21670. The method of item 21620 wherein the anti- fibrotic agent is an integrin antagonist.
21671. The method of item 21620 wherein the anti- fibrotic agent is an interleukin antagonist.
21672. The method of item 21620 wherein the anti- fibrotic agent is an inhibitor of type III receptor tyrosine kinase.
21673. The method of item 21620 wherein the anti- fibrotic agent is an irreversible inhibitor of enzyme methionine aminopeptidase type 2. 21674. The method of item 21620 wherein the anti- fibrotic agent is an isozyme selective delta protein kinase C inhibitor.
21675. The method of item 21620 wherein the anti- fibrotic agent a JAK3 enzyme inhibitor.
21676. The method of item 21620 wherein the anti- fibrotic agent is a JNK inhibitor.
21677. The method of item 21620 wherein the anti- fibrotic agent is a kinase inhibitor.
21678. The method of item 21620 wherein the anti- fibrotic agent is kinesin antagonist.
21679. The method of item 21620 wherein the anti- fibrotic agent is a kinesin antagonist.
21680. The method of item 21620 wherein the anti- fibrotic agent is a leukotriene inhibitor and antagonist selected from the group consisting of ambicromil (CAS No. 58805-38-2) (Sanofi-Aventis), amelubant (CAS No. 346735-24-8) (Boehringer Ingelheim), DW-1141 (Dong Wha), ebselen (Daiichi Pharmaceutical), ibudilast (Kyorin), leucotriene inhibitors from Sanofi-Aventis, lymphotoxin-beta receptor (LT-D) from Biogen Idee, Pharmaprojects No. 1535 and 2728 (CAS No. 119340-33-9) (Sanofi-Aventis), R-112 (Rigel), Rev-5367 (CAS No. 92532-05-3) (Sanofi- Aventis), RG-14893 (CAS No. 141835-49-6) (Sanofi-Aventis), RG-5901-A (CAS No. 101910-24-1), 92532-23-5, RP 66153 (CAS No. 142422-79-5), RP 66364 (CAS No.186916-92-5), RP 69698 (CAS No. 141748-00-7) (Sanofi-Aventis), SC-411930 (Pfizer), SC-41930 (CAS No. 120072-59-5) (Pfizer), SC-50605 (CAS No. 138828-39-4) (Pfizer), SC-51146 (CAS No. 141059-52-1), SC-53228 (CAS No. 153633-01-3) (Pfizer), spaglumic acid (ZY-15106) (CAS No.3106-85-2), 80619-64-3 (Novartis), tipredane (CAS No. 85197-77-9) (Bristol-Myers Squibb), U-75302 (CAS No. 119477-85-9) (Pfizer), and analogue or derivative thereof.
21681. The method of item 21620 wherein the anti- fibrotic agent is an MAP kinase inhibitor.
21682. The method of item 21620 wherein the anti- fibrotic agent is a matrix metailoproteinase inhibitor.
21683. The method of item 21620 wherein the anti- fibrotic agent is an MCP-CCR2 inhibitor.
21684. The method of item 21620 wherein the anti- fibrotic agent is an mTOR inhibitor.
21685. The method of item 21620 wherein the anti- fibrotic agent is an mTOR kinase inhibitor.
21686. The method of item 21620 wherein the anti- fibrotic agent is a microtubule inhibitor selected from the group consisting of antibody-maytansinoid conjugates from Biogen Idee, colchicines (MantiCore Pharmaceuticals), anticancer immunoconjugates from Johnson & Johnson, DIME from Octamer, gni-1f (GNI), huC242-DM4, huMy9-6-DM1 (ImmunoGen), IDN-5404 (Indena), IMO-098, IMOderm (Imotep), mebendazole (Introgen Therapeutics), microtubule poisons from Cambridge Enterprise, paclitaxel such as LOTAX from Aphios (CAS No. 33069-62-4), Genexol-PM from Samyang, Pharmaprojects No. 6383 (Tapestry Pharmaceuticals), RPR-112378 (Sanofi-Aventis), SGN-75 (Seattle Genetics), SPL-7435 (Starpharma), SSR-250411 (Sanofi-Aventis), trastuzumab-DM1 (Genentech), vinorelbine, and an analogue or derivative thereof.
21687. The method of item 21620 wherein the anti- fibrotic agent is an MIF inhibitor.
21688. The method of item 21620 wherein the anti- fibrotic agent is an MMP inhibitor.
21689. The method of item 21620 wherein the anti- fibrotic agent is a neurokinin (NK) antagonist selected from the group consisting of anthrotainin (CAS No. 148084-40-6) (Sanofi-Aventis), an IBS thereapeutic from ArQuIe, MDL-105212A (CAS No. 167261-60-1) (Ssanofi- Aventis), Pharmaprojects No. 2744, 3258 (CAS No. 139167-47-8) 4006, 4201, or 5986 (Sanofi-Aventis), RP 67580 (CAS No. 135911-02-3), SR- 144190 (CAS No. 201152-86-5), SSR-240600, SSR-241586 (Sanofi- Aventis), TKA-457 (Novartis), vestipitant mesylate (CAS No. 334476-64-1) (GlaxoSmithKline), Win-64821 (Sanofi-Aventis), PRX-96026 (Predix Pharmaceuticals), and an analogue or derivative thereof.
21690. The method of item 21620 wherein the anti- fibrotic agent is an NF kappa B inhibitor selected from the group consisting of AVE-0545 or AVE-0547 (Sanofi-Aventis), bortezomib (CAS No. 179324- 69-7) (Millennium Pharmaceuticals), dexanabinol (CAS No. 112924-45-5) (Pharmos), dexlipotam (Viatris), Pharmaprojects No. 6283 (INDRA) (OXiGENE), IPL-576092 (CAS No. 137571-30-3) (Inflazyme), NFKB decoy (Corgentech), NFKB decoy oligo (AnGes MG), NFKB's from Ariad, osteoporosis treatments or S5 (F005) from Fulcrum Pharmaceuticals, P61 (Phytopharm), R-flurbiprofen (CAS No. 5104-49-4) (Encore Pharmaceuticals), and an analogue or derivative thereof. 21691. The method of item 21620 wherein the anti- fibrotic agent is a nitric oxide agonist.
21692. The method of item 21620 wherein the anti- fibrotic agent is an ornithine decarboxylase inhibitor.
21693. The method of item 21620 wherein the anti- fibrotic agent is a p38 MAP kinase inhibitor selected from the group consisting of AZD-6703 (AstraZeneca), JX-401 (Jexys Pharmaceuticals), BMS-2 (Bristol-Myers Squibb), a p38 MAP kinase inhibitor from Novartis, a p38-alpha MAP kinase inhibitor from Amphora, Pharmaprojects No. 5704 (Pharmacopeia), RPR-200765A (Sanofi-Aventis), SD-282 (Johnson & Johnson), TAK-715 (Takeda), and an analogue or derivative thereof.
21694. The method of item 21620 wherein the anti- fibrotic agent is a palmitoyl-protein thioesterase inhibitor.
21695. The method of item 21620 wherein the anti- fibrotic agent is a PDGF receptor kinase inhibitor selected from the group consisting of AAL-993, AMN-107, or ABP-309 (Novartis), AMG-706 (Amgen), BAY-57-9352 (Bayer), CDP-860 (UCB), E-7080 (Eisai), imatinib (CAS No. 152459-95-5) (Novartis), OSI-930 (OSI Pharmaceuticals), RPR- 127963E (Sanofi-Aventis), RWJ-540973 (Johnson & Johnson), sorafenib tosylate (Bayer), SU-11657 (Pfizer), tandutinib (CAS No. 387867-13-2) (Millennium Pharmaceuticals), vatalanib (Novartis), ZK-CDK (Schering AG), and an analogue or derivative thereof.
21696. The method of item 21620 wherein the anti- fibrotic agent is (-)-halofenate (Metabolex), AMG-131 (Amgen), antidiabetics from Japan Tobacco, AZD-4619, AZD-8450, AZD-8677 (AstraZeneca), DRF-10945, balaglitazone (Dr Reddy's), CS-00088, CS-00098 (Chipscreen Biosciences), E-3030 (Eisai), etalocib (CAS No. 161172-51-6) (EIi Lilly), GSK-641597 (Ligand), GSK-677954 (GlaxoSmithKline), GW-409544 (Ligand), GW-590735 (GlaxoSmithKline), K-111 (Hoffmann-La Roche), LY- 518674 (EIi Lilly), LY-674 (Ligand), LY-929 (Ligand), MC-3001, MC-3002 (MaxoCore Pharmaceuticals), metformin HCI + pioglitazone (CAS No. 1115- 70-4 and 112529-15-4), ACTOPLUS MET from Andrx), muraglitazar (CAS No. 331741-94-7) (Bristol-Myers Squibb), naveglitazar (Ligand), oleoylethanolamide (Kadmus Pharmaceuticals), ONO-5129, pioglitazone hydrochloride (CAS No. 111025-46-8 and 112529-15-4) (Takeda), PLX-204 (Plexxikon), PPAR agonists from Genfit, PPAR delta agonists from EIi Lilly, PPAR-alpha agonists from CrystalGenomics, PPAR-gamma modulators and PPAR-β modulators from CareX, rosiglitazone maleate (CAS No. 122324- 73-4 or 155141-29-0) (GlaxoSmithKline), rosiglitazone maleate/glimepir (CAS No. 155141-29-0 and 93479-97-1), AVANDARYL, rosiglitazone maieate/metformin extend (CAS No. 155141-29-0 and 657-24-9), AVANDAMET, rosiglitazone maieate+metformin, AVANDAMET (GlaxoSmithKline), tesaglitazar (AstraZeneca), and an analogue or derivative thereof.
21697. The method of item 21620 wherein the anti- fibrotic agent is a phosphatase inhibitor.
21698. The method of item 21620 wherein the anti- fibrotic agent is a phosphodiesterase (PDE) inhibitor selected from the group consisting of avanafil (Tanabe Seiyaku), dasantafil (CAS No. 569351- 91-3) (Schering-Plough), A-906119 (CAS No. 134072-58-5), DL-850 (Sanofi-Aventis), GRG-3015, GRC-3566, GRC-3886 (Glenmark), HWA-153 (CAS No. 56395-66-5) (Sanofi-Aventis), hydroxypumafentrine (Altana), IBFB-130011, IBFB-14-016, IBFB-140301, IBFB-150007, IBFB-211913 (IBFB Pharma), L-826141 (Merck & Co), medorinone (CAS No. 88296-61-1) (Sanofi-Aventis), MEM-1917 (Memory Pharmaceuticals), ND-1251 (Neuro3d), PDE inhibitors from ICOS, PDE IV inhibitors from Memory Pharmaceuticals and CrystalGenomics, Pharmaprojects No. 2742 and 6141 (Sanofi-Aventis), QAD-171 (Novartis), RHC-2963 (CAS No. 76993-12-9 and 76993-14-1), RPR-117658, RPR-122818 derivatives, SR-24870 , and RPR- 132294 (Sanofi-Aventis), SK-350 (ln2Gen), stroke therapy agents from deCODE Genetics, TAS-203 (Taiho), tofimilast (CAS No. 185954-27-2) (Pfizer), UK-371800 (Pfizer), WIN-65579 (CAS No. 158020-82-7) (Sanofi- Aventis), IBFB-130020 (IBFB Pharma), OPC-6535 (CAS No. 145739-56-6) (Otsuka), a phosphodiesterase ill inhibitor, enoximone, a phosphodiesterase IV inhibitor, fosfosal, Atopik (Barrier Therapeutics), triflusal, a phosphodiesterase V inhibitor, and an analogue or derivative thereof.
21699. The method of item 21620 wherein the anti- fibrotic agent is a PKC inhibitor.
21700. The method of item 21620 wherein the anti- fibrotic agent is a platelet activating factor antagonist.
21701. The method of item 21620 wherein the anti- fibrotic agent is a platelet-derived growth factor receptor kinase inhibitor.
21702. The method of item 21620 wherein the anti- fibrotic agent is a prolyl hydroxylase inhibitor.
21703. The method of item 21620 wherein the anti- fibrotic agent is a polymorphonuclear neutrophil inhibitor.
21704. The method of item 21620 wherein the anti- fibrotic agent is a protein kinase B inhibitor. 21705. The method of item 21620 wherein the anti- fibrotic agent is a protein kinase C stimulant.
21706. The method of item 21620 wherein the anti- fibrotic agent is a purine nucleoside analogue.
21707. The method of item 21620 wherein the anti- fibrotic agent is a purinoreceptor P2X antagonist.
21708. The method of item 21620 wherein the anti- fibrotic agent is a Raf kinase inhibitor.
21709. The method of item 21620 wherein the anti- fibrotic agent is a reversible inhibitor of ErbB1 and ErbB2.
21710. The method of item 21620 wherein the anti- fibrotic agent is a ribonucleoside triphosphate reductase inhibitor.
21711. The method of item 21620 wherein the anti- fibrotic agent is an SDF- 1 antagonist.
21712. The method of item 21620 wherein the anti- fibrotic agent is a sheddase inhibitor.
21713. The method of item 21620 wherein the anti- fibrotic agent is an SRC inhibitor.
21714. The method of item 21620 wherein the anti- fibrotic agent is a stromelysin inhibitor. 21715. The method of item 21620 wherein the anti- fibrotic agent is an Syk kinase inhibitor.
21716. The method of item 21620 wherein the anti- fibrotic agent is a telomerase inhibitor.
21717. The method of item 21620 wherein the anti- fibrotic agent is a TGF beta inhibitor selected from the group consisting of pirfenidone (CAS No. 53179-13-8) (MARNAC), tranilast (CAS No. 53902- 12-8) (Kissei), IN-1130 (ln2Gen), mannose-6-phosphate (BTG), TGF-β antagonists from Inflazyme (Pharmaprojects No. 6075), TGF-β antagonists from Sydney, non-industrial source), TGF-β I receptor kinase inhibitors from Eli Lilly, TGF-β receptor inhibitors from Johnson & Johnson, and an analogue or derivative thereof.
21718. The method of item 21620 wherein the anti- fibrotic agent is a TNFα antagonist or TACE inhibitor selected from the group consisting of adalimumab (CAS No. 331731-18-1) (Cambridge Antibody Technology), AGIX-4207 (AtheroGenics), AGT-1 (Advanced Biotherapy), an anti-inflammatory from Borean Pharma, Cellzome, or Paradigm Therapeutics, anti-inflammatory vaccine (TNF-alpha kinoid) from Neovacs, humanized anti-TNF antibody or an anti-TNF MAb (CB0006) Celltech (UCB), apratastat (CAS No. 287445-51-0) (Wyeth), BMS-561392 (Bristol-Myers Squibb), BN-006 (Bone), certolizumab pegol (CAS No. 428863-50-7 or CH-138 (UCB), cilomilast (CAS No. 153259-65-5) (GlaxoSmithKline), CR-1 (Nuada Pharmaceuticals), CRx-119 (CombinatoRx), D-5410 (UCB), dacopafant (CAS No. 125372-33-0) (Sanofi- Aventis), dersalazine (CAS No. 188913-57-7/188913-58-8) (Uriach), etanercept (CAS No. 185243-69-0) (Amgen), ethyl pyruvate (Critical (Critical Therapeutics), golimumab (CAS No. 476181-74-5) (Johnson & Johnson), hormono-immunotherapy from Ipsen, CDP571 (e.g., Humicade from UCB), IC-485 (ICOS)1 infliximab (CAS No. 170277-31-3) (Johnson & Johnson), IP- 751 (Manhattan Pharmaceuticals), ISIS-104838 (CAS No. 250755-32-9) (ISIS Pharmaceuticals), lenalidomide (CAS No. 191732-72-6) (Celgene), lentinan (CAS No. 37339-90-5) (Ajinomoto), MDL-201112 (CAS No. 142130-73-2) (Sanofi-Aventis), medroxyprogesterone (CAS No. 520-85-4) (InKine Pharmaceutical), N-acetylcysteine (CAS No. 616-91-1) (Zambon), NBE-P2 (DIREVO Biotech), nerelimomab (CAS No. 162774-06-3) (Chiron), OM-294DP (OM PHARMA)1 onercept (CAS No. 199685-57-9) (Yeda), PASSTNF-alpha (Verigen), pentoxifylline or oxypentifylline (Sanofi-Aventis), Pharmaprojects No. 4091 , 4241 , 4295, or 4488 (Sanofi-Aventis), Pharmaprojects No. 5480 (Amgen), Pharmaprojects No. 6749 (Cengent), pirfenidone (CAS No. 53179-13-8) (MARNAC), RPR-132294 (Sanofi- Aventis), S5 (F002) (Fulcrum Pharmaceuticals), simvastatin (CAS No. 79902-63-9) (Merck & Co), STA-6292 (Synta Pharmaceuticals), tacrolimus (CAS No. 104987-11-3) (Fujisawa LifeCycle Pharma), talactoferrin alfa (CAS No. 308240-58-6) (Agennix), thalidomide (CAS No. 50-35-1) (Celgene), TNF antagonists form ProStrakan, and Synergen, TNF inhibitors (Amgen), TNF-alpha antagonists from Dynavax Technologies and Jerina AG (Germany), TNF-alpha inhibitors from IBFB Pharma and Xencor (Xencor), torbafylline (CAS No. 105102-21-4) (Sanofi-Aventis), UR-1505 (Uriach), VT-346 (Viron Therapeutics), YSIL6 (Y's Therapeutics), YSTH2 (Y's Therapeutics), NPI-1302a-3 (Nereus Pharmaceuticals, a TNF antagonist from Jerina AG (Germany), dersalazine, and an analogue or derivative thereof.
21719. The method of item 21620 wherein the anti- fibrotic agent is a Toll receptor inhibitor.
21720. The method of item 21620 wherein the anti- fibrotic agent is a tubulin antagonist. 21721. The method of item 21620 wherein the anti- fibrotic agent is a tyrosine kinase inhibitor selected from the group consisting of SU-011248, SUTENT from Pfizer Inc. (New York, NY)1 BMS-354825, PN- 355 (Paracelsian Pharmaceuticals), AGN-199659 (Allergan), AAL-993 or ABP-309 (Novartis), adaphostin (NIH), AEE-788 (Novartis), AG-013736 (OSI Pharmaceuticals), AG-13736 (Pfizer), ALT-110 (Alteris Therapeutics), AMG-706 (Amgen), anticancer MAbs from Xencor, anti-EGFrvlll MAbs from Abgenix, anti-HER2 MAb from Abiogen, AZD-2171 or AZD-9935 (AstraZeneca), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), CEP-5214 (Cephalon), CEP-7055 (Cephalon), cetuximab (ImClone Systems), CHIR-200131 and CHIR-258 (Chiron), CP-547632 (OSI Pharmaceuticals), CP-724714 (Pfizer), CT-301 (Creabilis Therapeutics), D- 69491 (Baxter International), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EGFR/ErbB2 inhibitors from Array BioPharma, erlotinib (CAS No. 183319- 69-9) (OSI Pharmaceuticals), EXEL-2880 (Exelixis), FK-778 (Sanofi- Aventis) , gefitinib (CAS No. 184475-35-2) (AstraZeneca), GW-2286 or GW- 654652 (GlaxoSmithKline), her2/neu antigen from AlphaVax, HER-2/neu inhibitor from Generex, Herzyme (Medipad) (Sirna Therapeutics), HKI-272 (Wyeth), HuMax-EGFr (Genmab), idronoxil (CAS No. 81267-65-4) (Novogen), IGF-1 inhibitors from Ontogen, IMC-11F8 (ImClone Systems), kahalalide F (CAS No. 149204-42-2) (PharmaMar), KDR inhibitor from LG Life Sciences, KDR inhibitors from Abbott Laboratories, KDR kinase inhibitors (UCB), Kdr kinase inhibitors from Merck & Co, KRN-633 and KRN- 951 (Kirin Brewery), KSB-102 (Xenova), lapatinib ditosylate (CAS No. 388082-78-8) (GlaxoSmithKline), matuzumab (Merck KGaA), MDX-214 (Medarex), ME-103 (Pharmexa), MED-A300 (Gerolymatos), MNAC-13 (Lay Line Genomics), nimotuzumab (Center of Molecular Immunology), NSC- 330507 or NSC-707545 (NIH), NV-50 (Novogen), OSI-930 (OSI Pharmaceuticals), panitumumab (Abgenix), pelitinib (CAS No. 287933-82-7) (Wyeth), pertuzumab (CAS No. 380610-27-5) (Genentech), Pharmaprojects No. 3985 (Sanofi-Aventis), prostate cancer therapeutics from Sequenom (SQPC35, SQPC36, SQPC90), removab and remoxab (Trion Pharma), RG- 13022 (CAS No. 136831-48-6), RG-13291 (CAS No. 138989-50-1), or RG- 14620 (CAS No. 136831-49-7) (Sanofi-Aventis), RM-6427 (Romark), RNAi breast cancer therapy from Benitec, RP 53801 (CAS No. 125882-88-4) (Sanofi-Aventis), sorafenib tosylate (Bayer), SU-11657 (Pfizer), Tie-2 antagonists from Semaia (Hybrigenics), Tie-2 inhibitors from Ontogen, trastuzumab (CAS No. 180288-69-1) (Genentech), tyrosine kinase inhibitors from Sanofi-Aventis, U3-1287, U3-1565, U3-1784, U3-1800 (U3 Pharma), vatalanib (Novartis), VEGFR-2 kinase inhibitor from Bristol-Myers Squibb, XL-647 (Exelixis), ZD-6474 (AstraZeneca), ZK-CDK (Schering AG), an EGFR tyrosine kinase inhibitor, EKB-569 (Wyeth), and an analogue or derivative thereof.
21722. The method of item 21620 wherein the anti- fibrotic agent is a VEGF inhibitor.
21723. The method of item 21620 wherein the anti- fibrotic agent is a vitamin D receptor agonist.
21724. The method of item 21620 wherein the anti- fibrotic agent is ZD-6474 (an angiogenesis inhibitor).
21725. The method of item 21620 wherein the anti- fibrotic agent is AP-23573 (an mTOR inhibitor).
21726. The method of item 21620 wherein the anti- fibrotic agent is synthadotin (a tubulin antagonist).
21727. The method of item 21620 wherein the anti- fibrotic agent is S-0885 (a collagenase inhibitor). 21728. The method of item 21620 wherein the anti- fibrotic agent is aplidine (an elongation factor-1 alpha inhibitor).
21729. The method of item 21620 wherein the anti- fibrotic agent is ixabepilone (an epithilone).
21730. The method of item 21620 wherein the anti- fibrotic agent is IDN-5390 (an angiogenesis inhibitor).
21731. The method of item 21620 wherein the anti- fibrotic agent is SB-2723005 (an angiogenesis inhibitor).
21732. The method of item 21620 wherein the anti- fibrotic agent is ABT-518 (an angiogenesis inhibitor).
21733. The method of item 21620 wherein the anti- fibrotic agent is combretastatin (an angiogenesis inhibitor).
21734. The method of item 21620 wherein the anti- fibrotic agent is anecortave acetate (an angiogenesis inhibitor).
21735. The method of item 21620 wherein the anti- fibrotic agent is SB-715992 (a kinesin antagonist).
21736. The method of item 21620 wherein the anti- fibrotic agent is temsirolimus (an mTOR inhibitor).
21737. The method of item 21620 wherein the anti- fibrotic agent is adalimumab (a TNFα antagonist). 21738. The method of item 21620 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with an anti-fibrotic agent, and (b) implanting the medical device into the host.
21739. The method of item 21620 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with an anti-infective agent, and (b) implanting the medical device into the host.
21740. The method of item 21620 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with a polymer; and (b) implanting the medical device into the host.
21741. The method of item 21620 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with a composition comprising an anti-fibrotic agent and a polymer, and (b) implanting the medical device into the host.
21742. The method of item 21620 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with a composition comprising an anti-infective agent and a polymer, and (b) implanting the medical device into the host.
21743. The method of item 21620 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with a composition comprising an anti-fibrotic agent, an anti- infective agent and a polymer, and (b) implanting the medical device into the host. 21744. The method of item 21620 wherein the anti- infective agent is an anthracycline.
21745. The method of item 21620 wherein the anti- infective agent is doxorubicin.
21746. The method of item 21620 wherein the anti- infective agent is is mitoxantrone.
21747. The method of item 21620 wherein the anti- infective agent is a fluoropyrimidine.
21748. The method of item 21620 wherein the anti- infective agent is 5-fluorouracil (5-FU).
21749. The method of item 21620 wherein the anti- infective agent is a folic acid antagonist.
21750. The method of item 21620 wherein the anti- infective agent is methotrexate.
21751. The method of item 21620 wherein the anti- infective agent is a podophylotoxin.
21752. The method of item 21620 wherein the anti- infective agent is etoposide.
21753. The method of item 21620 wherein the anti- infective agent is camptothecin. 21754. The method of item 21620 wherein the anti- infective agent is a hydroxyurea.
21755. The method of item 21620 wherein the anti- infective agent is a platinum complex.
21756. The method of item 21620 wherein the anti- infective agent is cisplatin.
21757. The method of item 21620 wherein the composition comprises an anti-thrombotic agent.
21758. The method of item 21620 wherein the polymer is formed from reactants comprising a naturally occurring polymer.
21759. The method of item 21620 wherein the polymer is formed from reactants comprising protein.
21760. The method of item 21620 wherein the polymer is formed from reactants comprising carbohydrate.
21761. The method of item 21620 wherein the polymer is formed from reactants comprising biodegradable polymer.
21762. The method of item 21620 wherein the polymer is formed from reactants comprising nonbiodegradable polymer.
21763. The method of item 21620 wherein the polymer is formed from reactants comprising collagen. 21764. The method of item 21620 wherein the polymer reactants comprising methylated collagen.
21765. The method of item 21620 wherein the polymer reactants comprising fibrinogen.
21766. The method of item 21620 wherein the polymer reactants comprising thrombin.
21767. The method of item 21620 wherein the polymer reactants comprising blood plasma.
21768. The method of item 21620 wherein the polymer reactants comprising calcium salt.
21769. The method of item 21620 wherein the polymer reactants comprising an antifibronolytic agent.
21770. The method of item 21620 wherein the polymer reactants comprising fibrinogen analog.
21771. The method of item 21620 wherein the polymer reactants comprising albumin.
21772. The method of item 21620 wherein the polymer reactants comprising plasminogen.
21773. The method of item 21620 wherein the polymer reactants comprising von Willebrands factor. 21774. The method of item 21620 wherein the polymer reactants comprising Factor VIII.
21775. The method of item 21620 wherein the polymer reactants comprising hypoallergenic collagen.
21776. The method of item 21620 wherein the polymer reactants comprising atelopeptidic collagen.
21777. The method of item 21620 wherein the polymer reactants comprising telopeptide collagen.
21778. The method of item 21620 wherein the polymer reactants comprising crosslinked collagen.
21779. The method of item 21620 wherein the polymer reactants comprising aprotinin.
21780. The method of item 21620 wherein the polymer reactants comprising epsilon-amino-n-caproic acid.
21781. The method of item 21620 wherein the polymer reactants comprising gelatin.
21782. The method of item 21620 wherein the polymer reactants comprising protein conjugates.
21783. The method of item 21620 wherein the polymer reactants comprising gelatin conjugates. 21784. The method of item 21620 wherein the polymer is formed from reactants comprising a synthetic polymer.
21785. The method of item 21620 wherein the polymer is formed from reactants comprising a synthetic isocyanate-containing compound.
21786. The method of item 21620 wherein the polymer is formed from reactants comprising a synthetic thiol-containing compound.
21787. The method of item 21620 wherein the polymer is formed from reactants comprising a synthetic compound containing at least two thiol groups.
21788. The method of item 21620 wherein the polymer is formed from reactants comprising a synthetic compound containing at least three thiol groups.
21789. The method of item 21620 wherein the polymer is formed from reactants comprising a synthetic compound containing at least four thiol groups.
21790. The method of item 21620 wherein the polymer is formed from reactants comprising a synthetic amino-containing compound.
21791. The method of item 21620 wherein the polymer is formed from reactants comprising a synthetic compound containing at least two amino groups. 21792. The method of item 21620 wherein the polymer is formed from reactants comprising a synthetic compound containing at least three amino groups.
21793. The method of item 21620 wherein the polymer is formed from reactants comprising a synthetic compound containing at least four amino groups.
21794. The method of item 21620 wherein the polymer is formed from reactants comprising a synthetic compound comprising a carbonyl-oxygen-succinimidyl group.
21795. The method of item 21620 wherein the polymer is formed from reactants comprising a synthetic compound comprising at least two carbonyl-oxygen-succinimidyl groups.
21796. The method of item 21620 wherein the polymer is formed from reactants comprising a synthetic compound comprising at least three carbonyl-oxygen-succinimidyl groups.
21797. The method of item 21620 wherein the polymer is formed from reactants comprising a synthetic compound comprising at least four carbonyl-oxygen-succinimidyl groups.
21798. The method of item 21620 wherein the polymer is formed from reactants comprising a synthetic polyalkylene oxide- containing compound.
21799. The method of item 21620 wherein the polymer is formed from reactants comprising a synthetic compound comprising both polyalkylene oxide and biodegradable polyester blocks. 21800. The method of item 21620 wherein the polymer is formed from reactants comprising a synthetic polyalkyiene oxide- containing compound having reactive amino groups.
21801. The method of item 21620 wherein the polymer is formed from reactants comprising a synthetic polyalkyiene oxide- containing compound having reactive thiol groups.
21802. The method of item 21620 wherein the polymer is formed from reactants comprising a synthetic polyalkyiene oxide- containing compound having reactive carbonyl-oxygen-succinimidyl groups.
21803. The method of item 21620 wherein the polymer is formed from reactants comprising a synthetic compound comprising a biodegradable polyester block.
21804. The method of item 21620 wherein the polymer is formed from reactants comprising a synthetic polymer formed in whole or part from lactic acid or lactide.
21805. The method of item 21620 wherein the polymer is formed from reactants comprising a synthetic polymer formed in whole or part from glycolic acid or glycolide.
21806. The method of item 21620 wherein the polymer is formed from reactants comprising polylysine.
21807. The method of item 21620 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound comprising a polyalkyiene oxide portion. 21808. The method of item 21620 wherein the polymer is formed from reactants comprising (a) protein and (b) polylysine.
21809. The method of item 21620 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound having at least four thiol groups.
21810. The method of item 21620 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound having at least four amino groups.
21811. The method of item 21620 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound having at least four carbonyl-oxygen-succinimide groups.
21812. The method of item 21620 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound having a biodegradable region formed from reactants selected from lactic acid, lactide, glycolic acid, glycolide, and epison-caprolactone.
21813. The method of item 21620 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound comprising a polyalkylene oxide portion.
21814. The method of item 21620 wherein the polymer is formed from reactants comprising (a) collagen and (b) polylysine.
21815. The method of item 21620 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound having at least four thiol groups. 21816. The method of item 21620 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound having at least four amino groups.
21817. The method of item 21620 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound having at least four carbonyl-oxygen-succinimide groups.
21818. The method of item 21620 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound having a biodegradable region formed from reactants selected from lactic acid, lactide, glycolic acid, glycolide, and epison-caprolactone.
21819. The method of item 21620 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound comprising a polyalkylene oxide portion.
21820. The method of item 21620 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) polylysine.
21821. The method of item 21620 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound having at least four thiol groups.
21822. The method of item 21620 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound having at least four amino groups. 21823. The method of item 21620 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound having at least four carbonyl-oxygen-succinimide groups.
21824. The method of item 21620 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound having a biodegradable region formed from reactants selected from lactic acid, lactide, glycolic acid, glycolide, and epison-caprolactone.
21825. The method of item 21620 wherein the polymer is formed from reactants comprising hyaluronic acid.
21826. The method of item 21620 wherein the polymer is formed from reactants comprising a hyaluronic acid derivative.
21827. The method of item 21620 wherein the polymer is formed from reactants comprising pentaerythritol poly(ethylene glycol)ether tetra-sulfhydryl of number average molecular weight between 3,000 and 30,000.
21828. The method of item 21620 wherein the polymer is formed from reactants comprising pentaerythritol poly(ethylene glycol)ether tetra-amino of number average molecular weight between 3,000 and 30,000.
21829. The method of item 21620 wherein the polymer is formed from reactants comprising (a) a synthetic compound having a number average molecular weight between 3,000 and 30,000 and comprising a polyalkylene oxide region and multiple nucleophilic groups, and (b) a synthetic compound having a number average molecular weight between 3,000 and 30,000 and comprising a polyalkylene oxide region and multiple electrophilic groups.
21830. The method of item 21620 wherein the composition comprises a colorant.
21831. The method of item 21620 wherein the composition is sterile.
21832. A method as in any one of items 21620-21831, wherein the device is a drainage tube.
21833. A method as in any one of items 21620-21831 , wherein the device is a feeding tube.
21834. A method as in any one of items 21620-21831, wherein the device is a portosystemic shunt.
21835. A method as in any one of items 21620-21831 , wherein the device is a shunt for ascite.
21836. A method as in any one of items 21620-21831, wherein the device is a nasogastric or nasoenteral tube.
21837. A method as in any one of items 21620-21831 , wherein the device is a gastrostomy or percutaneous feeding tube.
21838. A method as in any one of items 21620-21831 , wherein the device is a jejunostomy endoscopic tube. 21839. A method as in any one of items 21620-21831 , wherein the device is a colostomy device.
21840. A method as in any one of items 21620-21831 , wherein the device is a biliary T-tube.
21841. A method as in any one of items 21620-21831 , wherein the device is a biopsy forceps.
21842. A method as in any one of items 21620-21831 , wherein the device is a biliary stone removal device.
21843. A method as in any one of items 21620-21831 , wherein the device is an endoscopic retrograde cholangiopancretography device.
21844. A method as in any one of items 21620-21831, wherein the device is a dilation balloon.
21845. A method as in any one of items 21620-21831 , wherein the device is an enteral feeding device.
21846. A method as in any one of items 21620-21831 , wherein the device is a stent.
21847. A method as in any one of items 21620-21831 , wherein the device is a low profile device.
21848. A method as in any one of items 21620-21831, wherein the device is a virtual colonoscopy device. 21849. A method as in any one of items 21620-21831 , wherein the device is a capsule endoscope.
21850. A method as in any one of items 21620-21831 , wherein the device is a retrieval device.
21851. A method as in any one of items 21620-21831 , wherein the device is a gastrointestinal device adapted for examining the interior of the gastrointestinal tract.
21852. A method as in any one of items 21620-21831 , wherein the device is a gastrointestinal device adapted for irrigation or aspiration of the gastrointestinal tract.
21853. A method as in any one of items 21620-21831 , wherein the device is a colostomy device.
21854. A method as in any one of items 21620-21831 , wherein the device is a hemostatic device adapted for control of gastrointestinal bleeding.
21855. A method as in any one of items 21620-21831 , wherein the device is a gastrointestinal device adapted for cleaning a blocked gastrointestinal tract.
21856. A method as in any one of items 21620-21831 , wherein the device is a gastrointestinal device for providing communication between two bodily systems.
21857. A method as in any one of items 21620-21831, wherein the device is a portosystemic shunt. 21858. A method as in any one of items 21620-21831, wherein the device is a dilation catheter.
21859. A method for implanting a medical device comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with i) an anti-fibrotic agent, ii) an anti-infective agent, iii) a polymer, iv) a composition comprising an anti-fibrotic agent and a polymer, v) a composition comprising an anti-infective agent and a polymer, or vi) a composition comprising an anti-fibrotic agent, an anti- infective agent and a polymer, and (b) implanting the medical device into the host, wherein the medical device is a central venous catheter.
21860. The method of item 21859 wherein the anti- fibrotic agent is an adensosine A2A receptor antagonist.
21861. The method of item 21859 wherein the anti- fibrotic agent is an AKT inhibitor.
21862. The method of item 21859 wherein the anti- fibrotic agent is an alpha 2 integrin antagonist, wherein the alpha 2 integrin antagonist is Pharmaprojects No. 5754 (Merck KgaA).
21863. The method of item 21859 wherein the anti- fibrotic agent is an alpha 4 integrin antagonist.
21864. The method of item 21859 wherein the anti- fibrotic agent is an alpha 7 nicotinic receptor agonist.
21865. The method of item 21859 wherein the anti- fibrotic agent is an angiogenesis inhibitor selected from the group consisting of AG-12,958 (Pfizer), ATN-161 (Attenuon LLC), neovastat, an angiogenesis inhibitor from Jerina AG (Germany), NM-3 (Mercian), VGA-1155 (Taisho), FCE-26644 (Pfizer), FCE-26950 (Pfizer), FPMA (Meiji Daries), FR-111142 (Fujisawa), GGTI-298, GM-1306 (Ligand), GPA-1734 (Novartis), NNC-47- 0011 (Novo Nordisk), herbamycin (Nippon Kayaku), lenalidomide (Celegene), IP-10 (NIH), ABT-828 (Abbott), KIN-841 (Tokushima University, Japan), SF-1126 (Semafore Pharmaceuticals), laminin technology (NIH), CHIR-258 (Chiron), NVP-AEW541 (Novartis), NVP-AEW541 (Novartis), Vt16907 (Alchemia), OXI-8007 (Oxigene), EG-3306 (Ark Therapeutics), Maspin (Arriva), ABT-567 (Abbott), PPI-2458 (Praecis Pharmaceuticals), CC-5079, CC-4089 (Celgene), HIF-1 alpha inhibitors (Xenova), S-247 (Pfizer), AP-23573 (Ariad), AZD-9935 (Astra Zeneca), mebendazole (Introgen Therapeutics), MetAP-2 inhibitors (GlaxoSmithKline), AG-615 (Angiogene Pharmaceuticals), Tie-2 antagonists (Hybrigenics), NC-381, CYC-381 , NC-169, NC-219, NC-383, NC-384, NC-407 (Lorus Therapeutics), ATN-224 (Attenuon), ON-01370 (Onconova), Vitronectin antagonists (Amgen), SDX-103 (Salmedix), Vitronectin antagonists (Shire), CHP (Riemser), TEK (Amgen), Anecortave acetate (Alcon), T46.2 (Matrix Therapeutics), HG-2 (Heptagen), TEM antagonists (Genzyme), Oxi-4500 (Oxigene), ATN-161 (Attenuon), WX-293 (Wilex), M-2025 (Metris Therapeutics), Alphastatin (BioActa), YH-16 (Yantai Rongchang), BIBF- 1120 (Boehringer Ingelheim), BAY-57-9352 (Bayer), AS-1404 (Cancer Research Technology), SC-77964 (Pfizer), glycomimetics (BioTie Therapies), TIE-2 Inhibitors (Ontogen), DIMI, Octamer (Octamer), ABR- 215050 (Active Biotech), ABT-518 (Abbott), KDR inhibitors (Abbott), BSF- 466895 (Abbott), SCH-221153 (Schering-Plough), DAC:antiangiogenic (ConjuChem), TFPI (EntreMed), AZD-2171 (Astra-Zenaca), CDC-394 (Celgene), LY290293 (EIi Lilly), IDN-5390 (Indena), Kdr Kinase Inhibitors (Merck), CT-113020, CT-116433, CT-116563, CT-31890, CT-32228) (Cell Therapeutics), A-299620 (Abbott), TWEAK Inhibitor (Amgen), VEGF modulators (Johnson and Johnson), Tum-N53, tumstatin (Genzyme), Thios- 1, Thios-2 (Thios Pharmaceuticals), MV-6401 (Miravant Medical Technologies), Spisulosine (PharmaMar), CEP-7055 (Cephalon), AUV-201 (Auvation), LM-609 (EIi Lilly), SKF-106615 (AnorMED), Oglufanide disodium (Cytran), BW-114 (Pharminox), Calreticulin (NIH), WX-678 (Wilex), SD-7784 (Pfizer), WX-UK1 (Wilex), SH-268 (Schering AG), 2-Me-PGA (Celgene), S- 137 (Pfizer), ZD-6126 (Angiogene Pharmaceuticals), SG-292 (SignalGen), Benefin (Lane Labs), A6, A36 (Angstrom), SB-2723005 (GlaxoSmithKline), SC-7 (Cell Therapeutics), ZEN-014 (AEterna Zentaris), 2-methoxyestradiol (EntreMed), NK-130119 (Nippon Kayaku), CC-10004 (Celgene), AVE- 8062A (Ajinomoto), Tacedinaline (Pfizer), Actinonin (Tokyo Metropolitan Institute of Medical Science), Lenalidomide (Celgene), VGA-1155, BTO-956 (SRI International), ER-68203-00 (Eisai), CT-6685 (UCB), JKC-362 (Phoenix Pharmaceuticals), DMI-3798 (DMI Biosciences, Angiomate (Ipsen), ZD-6474 (AstraZeneca), CEP-5214 (Cephalon), Canstatin (Genzyme), NM-3 (Mercian), Oridigm (MediQuest Therapeutics), Exherin (Adherex), BLS-0597 (Boston Life Sciences), PTC-299 (PTC Therapeutics), NPI-2358 (Nereus Pharmaceuticals), CGP-79787 (Novartis), AEE-788 (Novartis), CKD-732 (Chong Kun Dang), CP-564959 (OSI Pharmaceuticals), CM-101 (CarboMed), CT-2584, CT3501 (Cell Therapeutics), combretastatin and analogues and derivatives thereof (Oxigene), Rebimastat (Bristol-Meyers Squibb), Dextrin 2-sulfate (ML Laboratories), Cilengitide (Merk KGaA), NSC-706704 (Pharminox), KRN- 951 (Kirin Brewery), Ukrain, NSC-631570 (Nowicky Pharma), Tecogalan sodium (Daiichi Pharmaceutical), Tz-93 (Tsumura), TBC-1635 (Encysive Pharmaceuticals), TAN-1120 (Takeda), Semaxanib (Pfizer), BDI-7800 (Biopharmacopae), SD-186, SD-983 (Bristol-Meyers Squibb), SB-223245 (GlaxoSmithKline), SC-236 (Pfizer), RWJ-590973 (Johnson and Johnson), 1LX-1850 (Genzyme), SC-68488, S-836 (Pfizer), CG-55069-11 (CuraGen), Ki-23057 (Kirin Brewery), CCX-700 (Chemoentryx), Pegaptanib octasodium (Giled Sciences), ANGIOCOL (available from Biostratum Inc.), or an analogue or derivative thereof. 21866. The method of item 21859 wherein the anti- fibrotic agent is an apoptosis antagonist.
21867. The method of item 21859 wherein the anti- fibrotic agent is an apoptosis activator.
21868. The method of item 21859 wherein the anti- fibrotic agent is a beta 1 integrin antagonist.
21869. The method of item 21859 wherein the anti- fibrotic agent is a beta tubulin inhibitor.
21870. The method of item 21859 wherein the anti- fibrotic agent is a blocker of enzyme production in Hepatitis C.
21871. The method of item 21859 wherein the anti- fibrotic agent is a Bruton's tyrosine kinase inhibitor.
21872. The method of item 21859 wherein the anti- fibrotic agent is a calcineurin inhibitor.
21873. The method of item 21859 wherein the anti- fibrotic agent is a caspase 3 inhibitor.
21874. The method of item 21859 wherein the anti- fibrotic agent is a CC chemokine receptor antagonist.
21875. The method of item 21859 wherein the anti- fibrotic agent is a cell cycle inhibitor selected from the group consisting of SNS-595 (Sunesis), synthadotin, KRX-0403, and an analogue or derivative thereof. 21876. The method of item 21859 wherein the anti- fibrotic agent is a cathepsin B inhibitor.
21877. The method of item 21859 wherein the anti- fibrotic agent is a cathepsin K inhibitor, wherein the cathepsin K inhibitor is 462795 (GlaxoSmithKline), INPL-022-D6 (Amura Therapeutics), or an analogue or derivative thereof.
21878. The method of item 21859 wherein the anti- fibrotic agent is a cathepsin L inhibitor.
21879. The method of item 21859 wherein the anti- fibrotic agent is a CD40 antagonist.
21880. The method of item 21859 wherein the anti- fibrotic agent is a chemokine receptor agonist.
21881. The method of item 21859 wherein the anti- fibrotic agent is a chymase inhibitor.
21882. The method of item 21859 wherein the anti- fibrotic agent is a collagenase antagonist.
21883. The method of item 21859 wherein the anti- fibrotic agent is a CXCR antagonist.
21884. The method of item 21859 wherein the anti- fibrotic agent is a cyclin dependent kinase inhibitor selected from the group consisting of a CDK-1 inhibitor, a CDK-2 inhibitor, a CDK- 4 inhibitor, a CDK-6 inhibitor, a CAK1 inhibitor from GPC Biotech and Bristol-Myers Squibb, RGB-286199 (GPC Biotech), an anticancer agent from Astex Technology, a CAK1 inhibitor from GPC Biotech, a CDK inhibitor from Sanofi-Aventis, a CDK1/CDK2 inhibitor from Amgen, a CDK2 inhibitor from SUGEN-2 (Pfizer), a hearing loss therapy agent (Sound Pharmaceuticals), PD-0332991 (Pfizer), RGB-286199 (GPC Biotech), Ro-0505124 (Hoffmann- La Roche), a Ser/Thr kinase inhibitor from Lilly (EIi Lilly), CVT-2584 (CAS No. 199986-75-9) (CV Therapeutics), and an analogue or derivative thereof.
21885. The method of item 21859 wherein the anti- fibrotic agent is a cyclooxygenase 1 inhibitor.
21886. The method of item 21859 wherein the anti- fibrotic agent is a DHFR inhibitor.
21887. The method of item 21859 wherein the anti- fibrotic agent is a dual integrin inhibitor.
21888. The method of item 21859 wherein the anti- fibrotic agent is an elastase inhibitor.
21889. The method of item 21859 wherein the anti- fibrotic agent is an elongation factor-1 alpha inhibitor.
21890. The method of item 21859 wherein the anti- fibrotic agent is an endothelial growth factor antagonist.
21891. The method of item 21859 wherein the anti- fibrotic agent is an endothelial growth factor receptor kinase inhibitor selected from the group consisting of sorafenib tosylate (Bayer), AAL-993 (Novartis), ABP-309 (Novartis), BAY-57-9352 (Bayer), BlBF-1120 (Boehringer Ingelheim), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EXEL- 2880 (Exelixis), GW-654652 (GlaxoSmithKline), a KDR inhibitor from LG Life Sciences, CT-6685 and CT-6729 (UCB), KRN-633 and KRN-951 (Kirin Brewery), OSI-930 (OSI Pharmaceuticals), SP-5.2 (Supratek Pharma), SU- 11657 (Pfizer), a Tie-2 antagonist (Hybrigenics), a VEGFR-2 kinase inhibitor (Bristol-Myers Squibb), XL-647 (Exelixis), a KDR inhibitor from Abbott Laboratories, sorafenib tosylate, and an analogue or derivative thereof.
21892. The method of item 21859 wherein the anti- fibrotic agent is an endotoxin antagonist.
21893. The method of item 21859 wherein the anti- fibrotic agent is an epothilone and tubulin binder.
21894. The method of item 21859 wherein the anti- fibrotic agent is an estrogen receptor antagonist.
21895. The method of item 21859 wherein the anti- fibrotic agent is an FGF inhibitor.
21896. The method of item 21859 wherein the anti- fibrotic agent is a farnexyl transferase inhibitor.
21897. The method of item 21859 wherein the anti- fibrotic agent is farnesyltransferase inhibitor selected from the group of A- 197574 (Abbott), a farnesyltransferase inhibitor from Servier, FPTIII (Strathclyde Institute for Drug R), LB-42908 (LG Life Sciences), Pharmaprojects No. 5063 (Genzyme), Pharmaprojects No. 5597 (Ipsen), Yissum Project No. B-1055 (Yissum), and an analogue or derivative thereof.
21898. The method of item 21859 wherein the anti- fibrotic agent is an FLT-3 kinase inhibitor. 21899. The method of item 21859 wherein the anti- fibrotic agent is an FGF receptor kinase inhibitor.
21900. The method of item 21859 wherein the anti- fibrotic agent is a fibrinogen antagonist selected from the group consisting of AUV-201 (Auvation), MG-13926 (Sanofi-Aventis), plasminogen activator (CAS No. 105913-11-9) (from Sanofi-Aventis or UCB), plasminogen activator-2 (tPA-2) (Sanofi-Aventis), pro-υrokinase (CAS No. 82657-92-9) (Sanofi-Aventis), and an analogue or derivative thereof.
21901. The method of item 21859 wherein the anti- fibrotic agent is a heat shock protein 90 antagonist selected from the group consisting of SRN-005 (Sirenade), geldanamycin, NSC-33050 (17- allylaminogeldanamycin; 17-AAG), 17-dimethylaminoethylamino-17- demethoxy-geldanamycin (17-DMAG), rifabutin (rifamycin XIV, 1',4- didehydro-1-deoxy-1 ,4-dihydro-5'-(2-methylpropyl)-1-oxo-), and an analogue . or derivative thereof.
21902. The method of item 21859 wherein the anti- fibrotic agent is a histone deacetylase inhibitor.
21903. The method of item 21859 wherein the anti- fibrotic agent is an HMGCoA reductase inhibitor selected from the group consisting of an atherosclerosis therapeutic from Lipid Sciences, ATI-16000 (ARYx Therapeutics), KS-01-019 (Kos Pharmaceuticals), Pharmaprojects No. 2197 (Sanofi-Aventi), RP 61969 (Sanofi-Aventis), and an analogue or derivative thereof.
21904. The method of item 21859 wherein the anti- fibrotic agent is an ICAM inhibitor. 21905. The method of item 21859 wherein the anti- fibrotic agent is an IL, ICE and IRAK antagonist, wherein the antagonist is a CvJ-14877, CP-424174 (Pfizer), NF-61 (Negma-Lerads), and an analogue or derivative thereof.
21906. The method of item 21859 wherein the anti- fibrotic agent is an IL-2 inhibitor.
21907. The method of item 21859 wherein the anti- fibrotic agent is an immunosuppressant selected from the group consisting of teriflunomide (Sanofi Aventis), chlorsulfaquinoxalone (NSC-339004), chlorsulfaquinoxalone sulfate, CS-712 (Sankyo), ismomultin alfa (CAS No. 457913-93-8) (Akzo Nobel), antiallergics from GenPat77, antiinflammatories or AT-005 (Androclus Therapeutics), autoimmune disease therapeutics from EpiVax, BN-007 (Bone), budesonide (CAS No. 51333-22- 3) (MAP Pharmaceuticals), CO-14 (Genzyme), edratide (CAS No. 433922- 67-9) (Teva), EP-314 (Enanta), eprovafen (CAS No. 101335-99-3) (Sanofi- Aventis), HWA-131 (CAS No. 118788-41-3) (Sanofi-Aventis), immunomodulators from MerLion Pharmaceuticals, immunosuppressives from Alchemia, IPL-12 (Inflazyme), MDL-9563 (CAS No. 27086-86-8) (Sanofi-Aventis), Pharmaprojects No. 2330 (Sanofi-Aventis), Pharmaprojects No. 6426 (Abgenix), PXS-25 (Pharmaxis), rosmarinic acid (CAS No. 20283-92-5) (Sanofi-Aventis), RP 42927 or RP 54745 (CAS No. 135330-08-4) (Sanofi-Aventis), SGN-35 (Seattle Genetics), ST-1959 (Sigma-Tau), type I diabetes therapy from SYNX Pharma, UNIL-88 (Debiopharm), VP-025 (Vasogen), VR-694 (Vectura), PRTX-001 (Protalex), and an analogue or derivative thereof.
21908. The method of item 21859 wherein the anti- fibrotic agent is an IMPDH (inosine monophosphate). 21909. The method of item 21859 wherein the anti- fibrotic agent is an integrin antagonist.
21910. The method of item 21859 wherein the anti- fibrotic agent is an interleukin antagonist.
21911. The method of item 21859 wherein the anti- fibrotic agent is an inhibitor of type III receptor tyrosine kinase.
21912. The method of item 21859 wherein the anti- fibrotic agent is an irreversible inhibitor of enzyme methionine aminopeptidase type 2.
21913. The method of item 21859 wherein the anti- fibrotic agent is an isozyme selective delta protein kinase C inhibitor.
21914. The method of item 21859 wherein the anti- fibrotic agent a JAK3 enzyme inhibitor.
21915. The method of item 21859 wherein the anti- fibrotic agent is a JNK inhibitor.
21916. The method of item 21859 wherein the anti- fibrotic agent is a kinase inhibitor.
21917. The method of item 21859 wherein the anti- fibrotic agent is kinesin antagonist.
21918. The method of item 21859 wherein the anti- fibrotic agent is a kinesin antagonist. 21919. The method of item 21859 wherein the anti- fibrotic agent is a leukotriene inhibitor and antagonist selected from the group consisting of ambϊcromil (CAS No. 58805-38-2) (Sanofi-Aventis), amelubant (CAS No. 346735-24-8) (Boehringer Ingelheim), DW-1141 (Dong Wha), ebselen (Daiichi Pharmaceutical), ibudilast (Kyorin), leucotriene inhibitors from Sanofi-Aventis, lymphotoxin-beta receptor (LT-D) from Biogen Idee, Pharmaprojects No. 1535 and 2728 (CAS No. 119340-33-9) (Sanofi-Aventis), R-112 (Rigel), Rev-5367 (CAS No. 92532-05-3) (Sanofi- Aventis), RG-14893 (CAS No. 141835-49-6) (Sanofi-Aventis), RG-5901-A (CAS No. 101910-24-1), 92532-23-5, RP 66153 (CAS No. 142422-79-5), RP 66364 (CAS No.186916-92-5), RP 69698 (CAS No. 141748-00-7) (Sanofi-Aventis), SC-411930 (Pfizer), SC-41930 (CAS No. 120072-59-5) (Pfizer), SC-50605 (CAS No. 138828-39-4) (Pfizer), SC-51146 (CAS No. 141059-52-1), SC-53228 (CAS No. 153633-01-3) (Pfizer), spaglumic acid (ZY-15106) (CAS No.3106-85-2), 80619-64-3 (Novartis), tipredane (CAS No. 85197-77-9) (Bristol-Myers Squibb), U-75302 (CAS No. 119477-85-9) (Pfizer), and analogue or derivative thereof.
21920. The method of item 21859 wherein the anti- fibrotic agent is an MAP kinase inhibitor.
21921. The method of item 21859 wherein the anti- fibrotic agent is a matrix metalloproteinase inhibitor.
21922. The method of item 21859 wherein the anti- fibrotic agent is an MCP-CCR2 inhibitor.
21923. The method of item 21859 wherein the anti- fibrotic agent is an mTOR inhibitor. 21924. The method of item 21859 wherein the anti- fibrotic agent is an mTOR kinase inhibitor.
21925. The method of item 21859 wherein the anti- fibrotic agent is a microtubule inhibitor selected from the group consisting of antibody-maytansinoid conjugates from Biogen Idee, colchicines (MantiCore Pharmaceuticals), anticancer immunoconjugates from Johnson & Johnson, DIME from Octamer, gni-1f (GNI), huC242-DM4, huMy9-6-DM1 (ImmunoGen), IDN-5404 (Indena), IMO-098, IMOderm (Imotep), mebendazole (Introgen Therapeutics), microtubule poisons from Cambridge Enterprise, paclitaxel such as LOTAX from Aphios (CAS No. 33069-62-4), Genexol-PM from Samyang, Pharmaprojects No. 6383 (Tapestry Pharmaceuticals), RPR-112378 (Sanofi-Aventis), SGN-75 (Seattle Genetics), SPL-7435 (Starpharma), SSR-250411 (Sanofi-Aventis), trastuzumab-DM1 (Genentech), vinorelbine, and an analogue or derivative thereof.
21926. The method of item 21859 wherein the anti- fibrotic agent is an MIF inhibitor.
21927. The method of item 21859 wherein the anti- fibrotic agent is an MMP inhibitor.
21928. The method of item 21859 wherein the anti- fibrotic agent is a neurokinin (NK) antagonist selected from the group consisting of anthrotainin (CAS No. 148084-40-6) (Sanofi-Aventis), an IBS thereapeuticfrom ArQuIe, MDL-105212A (CAS No. 167261-60-1) (Ssanofi- Aventis), Pharmaprojects No. 2744, 3258 (CAS No. 139167-47-8) 4006, 4201 , or 5986 (Sanofi-Aventis), RP 67580 (CAS No. 135911-02-3), SR- 144190 (CAS No. 201152-86-5), SSR-240600, SSR-241586 (Sanofi- Aventis), TKA-457 (Novartis), vestipitant mesylate (CAS No. 334476-64-1) (GlaxoSmithKline), Win-64821 (Sanofi-Aventis), PRX-96026 (Predix Pharmaceuticals), and an analogue or derivative thereof.
21929. The method of item 21859 wherein the anti- fibrotic agent is an NF kappa B inhibitor selected from the group consisting of AVE-0545 or AVE-0547 (Sanofi-Aventis), bortezomib (CAS No. 179324- 69-7) (Millennium Pharmaceuticals), dexanabinol (CAS No. 112924-45-5) (Pharmos), dexlipotam (Viatris), Pharmaprojects No. 6283 (INDRA) (OXiGENE), lPL-576092 (CAS No. 137571-30-3) (Inflazyme), NFKB decoy (Corgentech), NFKB decoy oligo (AnGes MG), NFKB's from Ariad, osteoporosis treatments or S5 (F005) from Fulcrum Pharmaceuticals, P61 (Phytopharm), R-flurbiprofen (CAS No. 5104-49-4) (Encore Pharmaceuticals), and an analogue or derivative thereof.
21930. The method of item 21859 wherein the anti- fibrotic agent is a nitric oxide agonist.
21931. The method of item 21859 wherein the anti- fibrotic agent is an ornithine decarboxylase inhibitor.
21932. The method of item 21859 wherein the anti- fibrotic agent is a p38 MAP kinase inhibitor selected from the group consisting of AZD-6703 (AstraZeneca), JX-401 (Jexys Pharmaceuticals), BMS-2 (Bristol-Myers Squibb), a p38 MAP kinase inhibitor from Novartis, a p38-alpha MAP kinase inhibitor from Amphora, Pharmaprojects No. 5704 (Pharmacopeia), RPR-200765A (Sanofi-Aventis), SD-282 (Johnson & Johnson), TAK-715 (Takeda), and an analogue or derivative thereof.
21933. The method of item 21859 wherein the anti- fibrotic agent is a palmitoyl-protein thioesterase inhibitor. 21934. The method of item 21859 wherein the anti- fibrotic agent is a PDGF receptor kinase inhibitor selected from the group consisting of AAL-993, AMN-107, or ABP-309 (Novartis), AMG-706 (Amgen), BAY-57-9352 (Bayer), CDP-860 (UCB), E-7080 (Eisai), imatinib (CAS No. 152459-95-5) (Novartis), OSI-930 (OSI Pharmaceuticals), RPR- 127963E (Sanofi-Aventis), RWJ-540973 (Johnson & Johnson), sorafenib tosylate (Bayer), SU-11657 (Pfizer), tandutinib (CAS No. 387867-13-2) (Millennium Pharmaceuticals), vatalanib (Novartis), ZK-CDK (Schering AG), and an analogue or derivative thereof.
21935. The method of item 21859 wherein the anti- fibrotic agent is (-)-halofenate (Metabolex), AMG-131 (Amgen), antidiabetics from Japan Tobacco, AZD-4619, AZD-8450, AZD-8677 (AstraZeneca), DRF-10945, balaglitazone (Dr Reddy's), CS-00088, CS-00098 (Chipscreen Biosciences), E-3030 (Eisai), etalocib (CAS No. 161172-51-6) (EIi Lilly), GSK-641597 (Ligand), GSK-677954 (GlaxoSmithKline), GW-409544 (Ligand), GW-590735 (GlaxoSmithKline), K-111 (Hoffmann-La Roche), LY- 518674 (EIi Lilly), LY-674 (Ligand), LY-929 (Ligand), MC-3001, MC-3002 (MaxoCore Pharmaceuticals), metformin HCI + pioglitazone (CAS No. 1115- 70-4 and 112529-15-4), ACTOPLUS MET from Andrx), muraglitazar (CAS No. 331741-94-7) (Bristol-Myers Squibb), naveglitazar (Ligand), oleoylethanolamide (Kadmus Pharmaceuticals), ONO-5129, pioglitazone hydrochloride (CAS No. 111025-46-8 and 112529-15r4) (Takeda), PLX-204 (Plexxikon), PPAR agonists from Genfit, PPAR delta agonists from EIi Lilly, PPAR-alpha agonists from CrystalGenomics, PPAR-gamma modulators and PPAR-β modulators from CareX, rosiglitazone maleate (CAS No. 122324- 73-4 or 155141-29-0) (GlaxoSmithKline), rosiglitazone maleate/glimepir (CAS No. 155141-29-0 and 93479-97-1), AVANDARYL, rosiglitazone maleate/metformin extend (CAS No. 155141-29-0 and 657-24-9), AVANDAMET, rosiglitazone maleate+metformin, AVANDAMET (GlaxoSmithKline), tesaglitazar (AstraZeneca), and an analogue or derivative thereof.
21936. The method of item 21859 wherein the anti- fibrotic agent is a phosphatase inhibitor.
21937. The method of item 21859 wherein the anti- fibrotic agent is a phosphodiesterase (PDE) inhibitor selected from the group consisting of avanafil (Tanabe Seiyaku), dasantafil (CAS No. 569351- 91-3) (Schering-Plough), A-906119 (CAS No. 134072-58-5), DL-850 (Sanofi-Aventis), GRC-3015, GRC-3566, GRC-3886 (Glenmark), HWA-153 (CAS No. 56395-66-5) (Sanofi-Aventis), hydroxypumafentrine (Altana), IBFB-130011, IBFB-14-016, IBFB-140301 , IBFB-150007, IBFB-211913 (IBFB Pharma), L-826141 (Merck & Co), medorinone (CAS No. 88296-61-1) (Sanofi-Aventis), MEM-1917 (Memory Pharmaceuticals), ND-1251 (Neuro3d), PDE inhibitors from ICOS1 PDE IV inhibitors from Memory Pharmaceuticals and CrystalGenomics, Pharmaprojects No. 2742 and 6141 (Sanofi-Aventis), QAD-171 (Novartis), RHC-2963 (CAS No. 76993-12-9 and 76993-14-1), RPR-117658, RPR-122818 derivatives, SR-24870 , and RPR- 132294 (Sanofi-Aventis), SK-350 (ln2Gen), stroke therapy agents from deCODE Genetics, TAS-203 (Taiho), tofimilast (CAS No. 185954-27-2) (Pfizer), UK-371800 (Pfizer), WIN-65579 (CAS No. 158020-82-7) (Sanofi- Aventis), IBFB-130020 (IBFB Pharma), OPC-6535 (CAS No. 145739-56-6) (Otsuka), a phosphodiesterase III inhibitor, enoximone, a phosphodiesterase IV inhibitor, fosfosal, Atopik (Barrier Therapeutics), triflusal, a phosphodiesterase V inhibitor, and an analogue or derivative thereof.
21938. The method of item 21859 wherein the anti- fibrotic agent is a PKC inhibitor. 21939. The method of item 21859 wherein the anti- fibrotic agent is a platelet activating factor antagonist.
21940. The method of item 21859 wherein the anti- fibrotic agent is a platelet-derived growth factor receptor kinase inhibitor.
21941. The method of item 21859 wherein the anti- fibrotic agent is a prolyl hydroxylase inhibitor.
21942. The method of item 21859 wherein the anti- fibrotic agent is a polymorphonuclear neutrophil inhibitor.
21943. The method of item 21859 wherein the anti- fibrotic agent is a protein kinase B inhibitor.
21944. The method of item 21859 wherein the anti- fibrotic agent is a protein kinase C stimulant.
21945. The method of item 21859 wherein the anti- fibrotic agent is a purine nucleoside analogue.
21946. The method of item 21859 wherein the anti- fibrotic agent is a purinoreceptor P2X antagonist.
21947. The method of item 21859 wherein the anti- fibrotic agent is a Raf kinase inhibitor.
21948. The method of item 21859 wherein the anti- fibrotic agent is a reversible inhibitor of ErbB1 and ErbB2. 21949. The method of item 21859 wherein the anti- fibrotic agent is a ribonucleoside triphosphate reductase inhibitor.
21950. The method of item 21859 wherein the anti- fibrotic agent is an SDF-1 antagonist.
21951. The method of item 21859 wherein the anti- fibrotic agent is a sheddase inhibitor.
21952. The method of item 21859 wherein the anti- fibrotic agent is an SRC inhibitor.
21953. The method of item 21859 wherein the anti- fibrotic agent is a stromelysin inhibitor.
21954. The method of item 21859 wherein the anti- fibrotic agent is an Syk kinase inhibitor.
21955. The method of item 21859 wherein the anti- fibrotic agent is a telomerase inhibitor.
21956. The method of item 21859 wherein the anti- fibrotic agent is a TGF beta inhibitor selected from the group consisting of pirfenidone (CAS No. 53179-13-8) (MARNAC)1 tranilast (CAS No. 53902- 12-8) (Kissei), IN-1130 (ln2Gen), mannose-6-phosphate (BTG), TGF-β antagonists from Inflazyme (Pharmaprojects No. 6075), TGF-β antagonists from Sydney, non-industrial source), TGF-β I receptor kinase inhibitors from EIi Lilly, TGF-β receptor inhibitors from Johnson & Johnson, and an analogue or derivative thereof. 21957. The method of item 21859 wherein the anti- fibrotic agent is a TNFα antagonist or TACE inhibitor selected from the group consisting of adalimumab (CAS No. 331731-18-1) (Cambridge Antibody Technology), AGIX-4207 (AtheroGenics), AGT-1 (Advanced Biotherapy), an anti-inflammatory from Borean Pharma, Cellzome, or Paradigm Therapeutics, anti-inflammatory vaccine (TNF-alpha kinoid) from Neovacs, humanized anti-TNF antibody or an anti-TNF MAb (CB0006) Celltech (UCB), apratastat (CAS No. 287445-51-0) (Wyeth), BMS-561392 (Bristol-Myers Squibb), BN-006 (Bone), certolizumab pegol (CAS No. 428863-50-7 or CH-138 (UCB), cilomilast (CAS No. 153259-65-5) (GlaxoSmithKline), CR-1 (Nuada Pharmaceuticals), CRx-119 (CombinatoRx), D-5410 (UCB), dacopafant (CAS No. 125372-33-0) (Sanofi- Aventis), dersalazine (CAS No. 188913-57-7/188913-58-8) (Uriach), etanercept (CAS No. 185243-69-0) (Amgen), ethyl pyruvate (Critical (Critical Therapeutics), golimumab (CAS No. 476181-74-5) (Johnson & Johnson), hormono-immunotherapy from Ipsen, CDP571 (e.g., Humicade from UCB), IC-485 (ICOS), infliximab (CAS No. 170277-31-3) (Johnson & Johnson), IP- 751 (Manhattan Pharmaceuticals), ISIS-104838 (CAS No. 250755-32-9) (ISIS Pharmaceuticals), lenalidomide (CAS No. 191732-72-6) (Celgene), lentinan (CAS No. 37339-90-5) (Ajinomoto), MDL-201112 (CAS No. 142130-73-2) (Sanofi-Aventis), medroxyprogesterone (CAS No. 520-85-4) (InKine Pharmaceutical), N-acetylcysteine (CAS No. 616-91-1) (Zambon), NBE-P2 (DIREVO Biotech), nerelimomab (CAS No. 162774-06-3) (Chiron), OM-294DP (OM PHARMA), onercept (CAS No. 199685-57-9) (Yeda), PASSTNF-alpha (Verigen), pentoxifylline or oxypentifylline (Sanofi-Aventis), Pharmaprojects No. 4091 , 4241 , 4295, or 4488 (Sanofi-Aventis), Pharmaprojects No. 5480 (Amgen), Pharmaprojects No. 6749 (Cengent), pirfenidone (CAS No. 53179-13-8) (MARNAC), RPR-132294 (Sanofi- Aventis), S5 (F002) (Fulcrum Pharmaceuticals), simvastatin (CAS No. 79902-63-9) (Merck & Co), STA-6292 (Synta Pharmaceuticals), tacrolimus (CAS No. 104987-11-3) (Fujisawa LifeCycle Pharma), talactoferrin alfa (CAS No. 308240-58-6) (Agennix), thalidomide (CAS No. 50-35-1) (Celgene), TNF antagonists form ProStrakan, and Synergen, TNF inhibitors (Amgen), TNF-alpha antagonists from Dynavax Technologies and Jerina AG (Germany), TNF-alpha inhibitors from IBFB Pharma and Xencor (Xencor), torbafylline (CAS No. 105102-21-4) (Sanofi-Aventis), UR-1505 (Uriach), VT-346 (Viron Therapeutics), YSIL6 (Y1S Therapeutics), YSTH2 (Y's Therapeutics), NPI-1302a-3 (Nereus Pharmaceuticals, a TNF antagonist from Jerina AG (Germany), dersalazine, and an analogue or derivative thereof.
21958. The method of item 21859 wherein the anti- fibrotic agent is a Toll receptor inhibitor.
21959. The method of item 21859 wherein the anti- fibrotic agent is a tubulin antagonist.
21960. The method of item 21859 wherein the anti- fibrotic agent is a tyrosine kinase inhibitor selected from the group consisting of SU-011248, SUTENT from Pfizer Inc. (New York, NY), BMS-354825, PN- 355 (Paracelsian Pharmaceuticals), AGN-199659 (Allergan), AAL-993 or ABP-309 (Novartis), adaphostin (NIH), AEE-788 (Novartis), AG-013736 (OSI Pharmaceuticals), AG-13736 (Pfizer), ALT-110 (Alteris Therapeutics), AMG-706 (Amgen), anticancer MAbs from Xencor, anti-EGFrvlll MAbs from Abgenix, anti-HER2 MAb from Abiogen, AZD-2171 or AZD-9935 (AstraZeneca), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), CEP-5214 (Cephalon), CEP-7055 (Cephalon), cetuximab (ImClone Systems), CHIR-200131 and CHlR-258 (Chiron), CP-547632 (OSI Pharmaceuticals), CP-724714 (Pfizer), CT-301 (Creabilis Therapeutics), D- 69491 (Baxter International), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EGFR/ErbB2 inhibitors from Array BioPharma, erlotinib (CAS No. 183319- 69-9) (OSI Pharmaceuticals), EXEL-2880 (Exelixis), FK-778 (Sanofi- Aventis) , gefitinib (CAS No. 184475-35-2) (AstraZeneca), GW-2286 or GW- 654652 (GlaxoSmithKline), her2/neu antigen from AlphaVax, HER-2/neu inhibitor from Generex, Herzyme (Medipad) (Sima Therapeutics), HKI-272 (Wyeth), HuMax-EGFr (Genmab), idronoxil (CAS No. 81267-65-4) (Novogen), IGF-1 inhibitors from Ontogen, 1MC-11F8 (ImCIone Systems), kahalalide F (CAS No. 149204-42-2) (PharmaMar), KDR inhibitor from LG Life Sciences, KDR inhibitors from Abbott Laboratories, KDR kinase inhibitors (UCB), Kdr kinase inhibitors from Merck & Co, KRN-633 and KRN- 951 (Kirin Brewery), KSB-102 (Xenova), lapatinib ditosylate (CAS No. 388082-78-8) (GlaxoSmithKline), matuzumab (Merck KGaA), MDX-214 (Medarex), ME-103 (Pharmexa), MED-A300 (Gerolymatos), MNAC-13 (Lay Line Genomics), nimotuzumab (Center of Molecular Immunology), NSC- 330507 or NSC-707545 (NIH), NV-50 (Novogen), OSI-930 (OSI Pharmaceuticals), panitumumab (Abgenix), pelitinib (CAS No. 287933-82-7) (Wyeth), pertuzumab (CAS No. 380610-27-5) (Genentech), Pharmaprojects No. 3985 (Sanofi-Aventis), prostate cancer therapeutics from Sequenom (SQPC35, SQPC36, SQPC90), removab and remoxab (Trion Pharma), RG- 13022 (CAS No. 136831-48-6), RG-13291 (CAS No. 138989-50-1), or RG- 14620 (CAS No. 136831-49-7) (Sanofi-Aventis), RM-6427 (Romark), RNAi breast cancer therapy from Benitec, RP 53801 (CAS No. 125882-88-4) (Sanofi-Aventis), sorafenib tosylate (Bayer), SU-11657 (Pfizer), Tie-2 antagonists from Semaia (Hybrigenics), Tie-2 inhibitors from Ontogen, trastuzumab (CAS No. 180288-69-1) (Genentech), tyrosine kinase inhibitors from Sanofi-Aventis, U3-1287, U3-1565, U3-1784, U3-1800 (U3 Pharma), vatalanib (Novartis), VEGFR-2 kinase inhibitor from Bristol-Myers Squibb, XL-647 (Exelixis), ZD-6474 (AstraZeneca), ZK-CDK (Schering AG), an EGFR tyrosine kinase inhibitor, EKB-569 (Wyeth), and an analogue or derivative thereof.
21961. The method of item 21859 wherein the anti- fibrotic agent is a VEGF inhibitor. 21962. The method of item 21859 wherein the anti- fibrotic agent is a vitamin D receptor agonist.
21963. The method of item 21859 wherein the anti- fibrotic agent is ZD-6474 (an angiogenesis inhibitor).
21964. The method of item 21859 wherein the anti- fibrotic agent is AP-23573 (an mTOR inhibitor).
21965. The method of item 21859 wherein the anti- fibrotic agent is synthadotin (a tubulin antagonist).
21966. The method of item 21859 wherein the anti- fibrotic agent is S-0885 (a collagenase inhibitor).
21967. The method of item 21859 wherein the anti- fibrotic agent is aplidine (an elongation factor-1 alpha inhibitor).
21968. The method of item 21859 wherein the anti- fibrotic agent is ixabepilone (an epithilone).
21969. The method of item 21859 wherein the anti- fibrotic agent is IDN-5390 (an angiogenesis inhibitor).
21970. The method of item 21859 wherein the anti- fibrotic agent is SB-2723005 (an angiogenesis inhibitor).
21971. The method of item 21859 wherein the anti- fibrotic agent is ABT-518 (an angiogenesis inhibitor). 21972. The method of item 21859 wherein the anti- fibrotic agent is combretastatin (an angiogenesis inhibitor).
21973. The method of item 21859 wherein the anti- fibrotic agent is anecortave acetate (an angiogenesis inhibitor).
21974. The method of item 21859 wherein the anti- fibrotic agent is SB-715992 (a kinesin antagonist).
21975. The method of item 21859 wherein the anti- fibrotic agent is temsirolimus (an mTOR inhibitor).
21976. The method of item 21859 wherein the anti- fibrotic agent is adalimumab (a TNFα antagonist).
21977. The method of item 21859 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with an anti-fibrotic agent, and (b) implanting the medical device into the host.
21978. The method of item 21859 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with an anti-infective agent, and (b) implanting the medical device into the host.
21979. The method of item 21859 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with a polymer; and (b) implanting the medical device into the host. 21980. The method of item 21859 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with a composition comprising an anti-fibrotic agent and a polymer, and (b) implanting the medical device into the host.
21981. The method of item 21859 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with a composition comprising an anti-infective agent and a polymer, and (b) implanting the medical device into the host.
21982. The method of item 21859 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with a composition comprising an anti-fibrotic agent, an anti- infective agent and a polymer, and (b) implanting the medical device into the host.
21983. The method of item 21859 wherein the anti- infective agent is an anthracycline.
21984. The method of item 21859 wherein the anti- infective agent is doxorubicin.
21985. The method of item 21859 wherein the anti- infective agent is is mitoxantrone.
21986. The method of item 21859 wherein the anti- infective agent is a fluoropyrimidine.
21987. The method of item 21859 wherein the anti- infective agent is 5-fluorouracil (5-FU). 21988. The method of item 21859 wherein the anti- infective agent is a folic acid antagonist.
21989. The method of item 21859 wherein the anti- infective agent is methotrexate.
21990. The method of item 21859 wherein the anti- infective agent is a podophylotoxin.
21991. The method of item 21859 wherein the anti- infective agent is etoposide.
21992. The method of item 21859 wherein the anti- infective agent is camptothecin.
21993. The method of item 21859 wherein the anti- infective agent is a hydroxyurea.
21994. The method of item 21859 wherein the anti- infective agent is a platinum complex.
21995. The method of item 21859 wherein the anti- infective agent is cisplatin.
21996. The method of item 21859 wherein the composition comprises an anti-thrombotic agent.
21997. The method of item 21859 wherein the polymer is formed from reactants comprising a naturally occurring polymer. 21998. The method of item 21859 wherein the polymer reactants comprising protein.
21999. The method of item 21859 wherein the polymer reactants comprising carbohydrate.
22000. The method of item 21859 wherein the polymer reactants comprising biodegradable polymer.
22001. The method of item 21859 wherein the polymer reactants comprising nonbiodegradable polymer.
22002. The method of item 21859 wherein the polymer reactants comprising collagen.
22003. The method of item 21859 wherein the polymer reactants comprising methylated collagen.
22004. The method of item 21859 wherein the polymer reactants comprising fibrinogen.
22005. The method of item 21859 wherein the polymer reactants comprising thrombin.
22006. The method of item 21859 wherein the polymer reactants comprising blood plasma.
22007. The method of item 21859 wherein the polymer reactants comprising calcium salt. 22008. The method of item 21859 wherein the polymer reactants comprising an antifibronolytic agent.
22009. The method of item 21859 wherein the polymer reactants comprising fibrinogen analog.
22010. The method of item 21859 wherein the polymer reactants comprising albumin.
22011. The method of item 21859 wherein the polymer reactants comprising plasminogen.
22012. The method of item 21859 wherein the polymer reactants comprising von Willebrands factor.
22013. The method of item 21859 wherein the polymer reactants comprising Factor VIII.
22014. The method of item 21859 wherein the polymer reactants comprising hypoallergenic collagen.
22015. The method of item 21859 wherein the polymer reactants comprising atelopeptidic collagen.
22016. The method of item 21859 wherein the polymer reactants comprising telopeptide collagen.
22017. The method of item 21859 wherein the polymer reactants comprising crosslinked collagen. 22018. The method of item 21859 wherein the polymer is formed from reactants comprising aprotinin.
22019. The method of item 21859 wherein the polymer is formed from reactants comprising epsilon-amino-n-caproic acid.
22020. The method of item 21859 wherein the polymer is formed from reactants comprising gelatin.
22021. The method of item 21859 wherein the polymer is formed from reactants comprising protein conjugates.
22022. The method of item 21859 wherein the polymer is formed from reactants comprising gelatin conjugates.
22023. The method of item 21859 wherein the polymer is formed from reactants comprising a synthetic polymer.
22024. The method of item 21859 wherein the polymer is formed from reactants comprising a synthetic isocyanate-containing compound.
22025. The method of item 21859 wherein the polymer is formed from reactants comprising a synthetic thiol-containing compound.
22026. The method of item 21859 wherein the polymer is formed from reactants comprising a synthetic compound containing at least two thiol groups. 22027. The method of item 21859 wherein the polymer is formed from reactants comprising a synthetic compound containing at least three thiol groups.
22028. The method of item 21859 wherein the polymer is formed from reactants comprising a synthetic compound containing at least four thiol groups.
22029. The method of item 21859 wherein the polymer is formed from reactants comprising a synthetic amino-containing compound.
22030. The method of item 21859 wherein the polymer is formed from reactants comprising a synthetic compound containing at least two amino groups.
22031. The method of item 21859 wherein the polymer is formed from reactants comprising a synthetic compound containing at least three amino groups.
22032. The method of item 21859 wherein the polymer is formed from reactants comprising a synthetic compound containing at least four amino groups.
22033. The method of item 21859 wherein the polymer is formed from reactants comprising a synthetic compound comprising a carbonyl-oxygen-succinimidyl group.
22034. The method of item 21859 wherein the polymer is formed from reactants comprising a synthetic compound comprising at least two carbonyl-oxygen-succinimidyl groups. 22035. The method of item 21859 wherein the polymer is formed from reactants comprising a synthetic compound comprising at least three carbonyl-oxygen-succinimidy! groups.
22036. The method of item 21859 wherein the polymer is formed from reactants comprising a synthetic compound comprising at least four carbonyl-oxygen-succinimidyl groups.
22037. The method of item 21859 wherein the polymer is formed from reactants comprising a synthetic polyalkylene oxide- containing compound.
22038. The method of item 21859 wherein the polymer is formed from reactants comprising a synthetic compound comprising both polyalkylene oxide and biodegradable polyester blocks.
22039. The method of item 21859 wherein the polymer is formed from reactants comprising a synthetic polyalkylene oxide- containing compound having reactive amino groups.
22040. The method of item 21859 wherein the polymer is formed from reactants comprising a synthetic polyalkylene oxide- containing compound having reactive thiol groups.
22041. The method of item 21859 wherein the polymer is formed from reactants comprising a synthetic polyalkylene oxide- containing compound having reactive carbonyl-oxygen-succinimidyl groups.
22042. The method of item 21859 wherein the polymer is formed from reactants comprising a synthetic compound comprising a biodegradable polyester block. 22043. The method of item 21859 wherein the polymer is formed from reactants comprising a synthetic polymer formed in whole or part from lactic acid or lactide.
22044. The method of item 21859 wherein the polymer is formed from reactants comprising a synthetic polymer formed in whole or part from glycolic acid or glycolide.
22045. The method of item 21859 wherein the polymer is formed from reactants comprising polylysine.
22046. The method of item 21859 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound comprising a polyalkylene oxide portion.
22047. The method of item 21859 wherein the polymer is formed from reactants comprising (a) protein and (b) polylysine.
22048. The method of item 21859 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound having at least four thiol groups.
22049. The method of item 21859 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound having at least four amino groups.
22050. The method of item 21859 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound having at least four carbonyl-oxygen-succinimide groups. 22051. The method of item 21859 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound having a biodegradable region formed from reactants selected from lactic acid, lactide, glycolic acid, glycolide, and epison-caprolactone.
22052. The method of item 21859 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound comprising a polyalkylene oxide portion.
22053. The method of item 21859 wherein the polymer is formed from reactants comprising (a) collagen and (b) polylysine.
22054. The method of item 21859 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound having at least four thiol groups.
22055. The method of item 21859 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound having at least four amino groups.
22056. The method of item 21859 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound having at least four carbonyl-oxygen-succinimide groups.
22057. The method of item 21859 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound having a biodegradable region formed from reactants selected from lactic acid, lactide, glycolic acid, glycolide, and epison-caprolactone. 22058. The method of item 21859 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound comprising a polyalkylene oxide portion.
22059. The method of item 21859 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) polylysine.
22060. The method of item 21859 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound having at least four thiol groups.
22061. The method of item 21859 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound having at least four amino groups.
22062. The method of item 21859 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound having at least four carbonyl-oxygen-succinimide groups.
22063. The method of item 21859 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound having a biodegradable region formed from reactants selected from lactic acid, lactide, glycolic acid, glycolide, and epison-caprolactone.
22064. The method of item 21859 wherein the polymer is formed from reactants comprising hyaluronic acid.
22065. The method of item 21859 wherein the polymer is formed from reactants comprising a hyaluronic acid derivative. 22066. The method of item 21859 wherein the polymer is formed from reactants comprising pentaerythritol poly(ethylene glycol)ether tetra-sulfhydryl of number average molecular weight between 3,000 and 30,000.
22067. The method of item 21859 wherein the polymer is formed from reactants comprising pentaerythritol poly(ethylene glycol)ether tetra-amino of number average molecular weight between 3,000 and 30,000.
22068. The method of item 21859 wherein the polymer is formed from reactants comprising (a) a synthetic compound having a number average molecular weight between 3,000 and 30,000 and comprising a polyalkylene oxide region and multiple nucleophilic groups, and (b) a synthetic compound having a number average molecular weight between 3,000 and 30,000 and comprising a polyalkylene oxide region and multiple electrophilic groups.
22069. The method of item 21859 wherein the composition comprises a colorant.
22070. The method of item 21859 wherein the composition is sterile.
22071. A method as in any one of items 21859-22070, wherein the device is a central venous catheter with a cuff.
22072. A method as in any one of items 21859-22070, wherein the device is a central venous catheter without a cuff. 22073. A method as in any one of items 21859-22070, wherein the device is a central venous catheter with a flange.
22074. A method as in any one of items 21859-22070, wherein the device is a central venous catheter without a flange.
22075. A method as in any one of items 21859-22070, wherein the device is a central venous catheter adapted for providing access to the circulatory system.
22076. A method as in any one of items 21859-22070, wherein the device is a central venous catheter adapted for providing multiple conduits for accessing the circulatory system.
22077. A method as in any one of items 21859-22070, wherein the device is a central venous catheter comprising a means for preventing infection as a result of long term use.
22078. A method as in any one of items 21859-22070, wherein the device is a central venous catheter adaptable for being used with an apparatus that provides a means of controlling the injection or withdrawl of bodily fluids through the central venous catheter.
22079. A method as in any one of items 21859-22070, wherein the device is a parenteral nutrition catheter.
22080. A method as in any one of items 21859-22070, wherein the device is a peripherally inserted central venous catheter. 22081. A method as in any one of items 21859-22070, wherein the device is a flow directed balloon tipped pulmonary artery catheter.
22082. A method as in any one of items 21859-22070, wherein the device is a long term central venous access catheter.
22083. A method for implanting a medical device comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with i) an anti-fibrotic agent, ii) an anti-infective agent, iii) a polymer, iv) a composition comprising an anti-fibrotic agent and a polymer, v) a composition comprising an anti-infective agent and a polymer, or vi) a composition comprising an anti-fibrotic agent, an anti- infective agent and a polymer, and (b) implanting the medical device into the host, wherein the medical device is a ventricular assist device.
22084. The method of item 22083 wherein the anti- fibrotic agent is an adensosine A2A receptor antagonist.
22085. The method of item 22083 wherein the anti- fibrotic agent is an AKT inhibitor.
22086. The method of item 22083 wherein the anti- fibrotic agent is an alpha 2 integrin antagonist, wherein the alpha 2 integrin antagonist is Pharmaprojects No. 5754 (Merck KgaA).
22087. The method of item 22083 wherein the anti- fibrotic agent is an alpha 4 integrin antagonist.
22088. The method of item 22083 wherein the anti- fibrotic agent is an alpha 7 nicotinic receptor agonist. 22089. The method of item 22083 wherein the anti- fibrotic agent is an angiogenesis inhibitor selected from the group consisting of AG-12,958 (Pfizer), ATN-161 (Attenuon LLC), neovastat, an angiogenesis inhibitor from Jerina AG (Germany), NM-3 (Mercian), VGA-1155 (Taisho), FCE-26644 (Pfizer), FCE-26950 (Pfizer), FPMA (Meiji Daries), FR-111142 (Fujisawa), GGTI-298, GM-1306 (Ligand), GPA-1734 (Novartis), NNC-47- 0011 (Novo Nordisk), herbamycin (Nippon Kayaku), lenalidomide (Celegene), IP-10 (NIH), ABT-828 (Abbott), KIN-841 (Tokushima University, Japan), SF-1126 (Semafore Pharmaceuticals), laminin technology (NIH), CHIR-258 (Chiron), NVP-AEW541 (Novartis), NVP-AEW541 (Novartis), Vt16907 (Alchemia), OXI-8007 (Oxigene), EG-3306 (Ark Therapeutics), Maspin (Arriva), ABT-567 (Abbott), PPI-2458 (Praecis Pharmaceuticals), CC-5079, CC-4089 (Celgene), HIF-1 alpha inhibitors (Xenova), S-247 (Pfizer), AP-23573 (Ariad), AZD-9935 (Astra Zeneca), mebendazole (Introgen Therapeutics), MetAP-2 inhibitors (GlaxoSmithKline), AG-615 (Angiogene Pharmaceuticals), Tie-2 antagonists (Hybrigenics), NC-381, - CYC-381 , NC-169, NC-219, NC-383, NC-384, NC-407 (Lorus Therapeutics), ATN-224 (Attenuon), ON-01370 (Onconova), Vitronectin antagonists (Amgen), SDX-103 (Salmedix), Vitronectin antagonists (Shire), CHP (Riemser), TEK (Amgen), Anecortave acetate (Alcon), T46.2 (Matrix Therapeutics), HG-2 (Heptagen), TEM antagonists (Genzyme), Oxi-4500 (Oxigene), ATN-161 (Attenuon), WX-293 (Wilex), M-2025 (Metris Therapeutics), Alphastatin (BioActa), YH-16 (Yantai Rongchang), BIBF- 1120 (Boehringer Ingelheim), BAY-57-9352 (Bayer), AS-1404 (Cancer Research Technology), SC-77964 (Pfizer), glycomimetics (BioTie Therapies), TIE-2 Inhibitors (Ontogen), DIMI, Octamer (Octamer), ABR- 215050 (Active Biotech), ABT-518 (Abbott), KDR inhibitors (Abbott), BSF- 466895 (Abbott), SCH-221153 (Schering-Plough), DAC:antiangiogenic (ConjuChem), TFPI (EntreMed), AZD-2171 (Astra-Zenaca), CDC-394 (Celgene), LY290293 (EIi Lilly), IDN-5390 (Indena), Kdr Kinase Inhibitors (Merck), CT-113020, CT-116433, CT-116563, CT-31890, CT-32228) (Cell Therapeutics), A-299620 (Abbott), TWEAK Inhibitor (Amgen), VEGF modulators (Johnson and Johnson), Tum-N53, tumstatin (Genzyme), Thios- 1 , Thios-2 (Thios Pharmaceuticals), MV-6401 (Miravant Medical Technologies), Spisulosine (PharmaMar), CEP-7055 (Cephalon), AUV-201 (Auvation), LM-609 (EIi Lilly), SKF-106615 (AnorMED), Oglufanide disodium (Cytran), BW-114 (Pharminox), Calreticulin (NIH), WX-678 (Wilex), SD-7784 (Pfizer), WX-UK1 (Wilex), SH-268 (Schering AG), 2-Me-PGA (Celgene), S- 137 (Pfizer), ZD-6126 (Angiogene Pharmaceuticals), SG-292 (SignalGen), Benefin (Lane Labs), A6, A36 (Angstrom), SB-2723005 (GlaxoSmithKline), SC-7 (Cell Therapeutics), ZEN-014 (AEterna Zentaris), 2-methoxyestradiol (EntreMed), NK-130119 (Nippon Kayaku), CC-10004 (Celgene), AVE- 8062A (Ajinomoto), Tacedinaline (Pfizer), Actinonin (Tokyo Metropolitan Institute of Medical Science), Lenalidomide (Celgene), VGA-1155, BTO-956 (SRI International), ER-68203-00 (Eisai), CT-6685 (UCB), JKC-362 (Phoenix Pharmaceuticals), DMI-3798 (DMI Biosciences, Angiomate (Ipsen), ZD-6474 (AstraZeneca), CEP-5214 (Cephalon), Canstatin (Genzyme), NM-3 (Mercian), Oridigm (MediQuest Therapeutics), Exherin (Adherex), BLS-0597 (Boston Life Sciences), PTC-299 (PTC Therapeutics), NPI-2358 (Nereus Pharmaceuticals), CGP-79787 (Novartis), AEE-788 (Novartis), CKD-732 (Chong Kun Dang), CP-564959 (OSI Pharmaceuticals), CM-101 (CarboMed), CT-2584, CT3501 (Cell Therapeutics), combretastatin and analogues and derivatives thereof (Oxigene), Rebimastat (Bristol-Meyers Squibb), Dextrin 2-sulfate (ML Laboratories), Cilengitide (Merk KGaA), NSC-706704 (Pharminox), KRN- 951 (Kirin Brewery), Ukrain, NSC-631570 (Nowicky Pharma), Tecogalan sodium (Daiichi Pharmaceutical), Tz-93 (Tsumura), TBC-1635 (Encysive Pharmaceuticals), TAN-1120 (Takeda), Semaxanib (Pfizer), BDI-7800 (Biopharmacopae), SD-186, SD-983 (Bristol-Meyers Squibb), SB-223245 (GlaxoSmithKline), SC-236 (Pfizer), RWJ-590973 (Johnson and Johnson), ILX-1850 (Genzyme), SC-68488, S-836 (Pfizer), CG-55069-11 (CuraGen), Ki-23057 (Kirin Brewery), CCX-700 (Chemoentryx), Pegaptanib octasodium (Giled Sciences), ANGIOCOL (available from Biostratum Inc.), or an analogue or derivative thereof.
22090. The method of item 22083 wherein the anti- fibrotic agent is an apoptosis antagonist.
22091. The method of item 22083 wherein the anti- fibrotic agent is an apoptosis activator.
22092. The method of item 22083 wherein the anti- fibrotic agent is a beta 1 integrin antagonist.
22093. The method of item 22083 wherein the anti- fibrotic agent is a beta tubulin inhibitor.
22094. The method of item 22083 wherein the anti- fibrotic agent is a blocker of enzyme production in Hepatitis C.
22095. The method of item 22083 wherein the anti- fibrotic agent is a Bruton's tyrosine kinase inhibitor.
22096. The method of item 22083 wherein the anti- fibrotic agent is a calcineurin inhibitor.
22097. The method of item 22083 wherein the anti- fibrotic agent is a caspase 3 inhibitor.
22098. The method of item 22083 wherein the anti- fibrotic agent is a CC chemokine receptor antagonist. 22099. The method of item 22083 wherein the anti- fibrotic agent is a cell cycle inhibitor selected from the group consisting of SNS-595 (Sunesis), synthadotin, KRX-0403, and an analogue or derivative thereof.
22100. The method of item 22083 wherein the anti- fibrotic agent is a cathepsin B inhibitor.
22101. The method of item 22083 wherein the anti- fibrotic agent is a cathepsin K inhibitor, wherein the cathepsin K inhibitor is 462795 (GlaxoSmithKline), INPL-022-D6 (Amura Therapeutics), or an analogue or derivative thereof.
22102. The method of item 22083 wherein the anti- fibrotic agent is a cathepsin L inhibitor.
22103. The method of item 22083 wherein the anti- fibrotic agent is a CD40 antagonist.
22104. The method of item 22083 wherein the anti- fibrotic agent is a chemokine receptor agonist.
22105. The method of item 22083 wherein the anti- fibrotic agent is a chymase inhibitor.
22106. The method of item 22083 wherein the anti- fibrotic agent is a collagenase antagonist.
22107. The method of item 22083 wherein the anti- fibrotic agent is a CXCR antagonist. 22108. The method of item 22083 wherein the anti- fibrotic agent is a cyclin dependent kinase inhibitor selected from the group consisting of a CDK-1 inhibitor, a CDK-2 inhibitor, a CDK- 4 inhibitor, a CDK-6 inhibitor, a CAK1 inhibitor from GPC Biotech and Bristol-Myers Squibb, RGB-286199 (GPC Biotech), an anticancer agent from Astex Technology, a CAK1 inhibitor from GPC Biotech, a CDK inhibitor from Sanofi-Aventis, a CDK1/CDK2 inhibitor from Amgen, a CDK2 inhibitor from SUGEN-2 (Pfizer), a hearing loss therapy agent (Sound Pharmaceuticals), PD-0332991 (Pfizer), RGB-286199 (GPC Biotech), Ro-0505124 (Hoffmann- La Roche), a Ser/Thr kinase inhibitor from Lilly (EIi Lilly), CVT-2584 (CAS No. 199986-75-9) (CV Therapeutics), and an analogue or derivative thereof.
22109. The method of item 22083 wherein the anti- fibrotic agent is a cyclooxygenase 1 inhibitor.
22110. The method of item 22083 wherein the anti- fibrotic agent is a DHFR inhibitor.
22111. The method of item 22083 wherein the anti- fibrotic agent is a dual integrin inhibitor.
22112. The method of item 22083 wherein the anti- fibrotic agent is an elastase inhibitor.
22113. The method of item 22083 wherein the anti- fibrotic agent is an elongation factor-1 alpha inhibitor.
22114. The method of item 22083 wherein the anti- fibrotic agent is an endothelial growth factor antagonist. 22115. The method of item 22083 wherein the anti- fibrotic agent is an endothelial growth factor receptor kinase inhibitor selected from the group consisting of sorafenib tosylate (Bayer), AAL-993 (Novartis), ABP-309 (Novartis), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EXEL- 2880 (Exelixis), GW-654652 (GlaxoSmithKline), a KDR inhibitor from LG Life Sciences, CT-6685 and CT-6729 (UCB), KRN-633 and KRN-951 (Kirin Brewery), OSI-930 (OSI Pharmaceuticals), SP-5.2 (Supratek Pharma), SU- 11657 (Pfizer), a Tie-2 antagonist (Hybrigenics), a VEGFR-2 kinase inhibitor (Bristol-Myers Squibb), XL-647 (Exelixis), a KDR inhibitor from Abbott Laboratories, sorafenib tosylate, and an analogue or derivative thereof.
22116. The method of item 22083 wherein the anti- fibrotic agent is an endotoxin antagonist.
22117. The method of item 22083 wherein the anti- fibrotic agent is an epothilone and tubulin binder.
22118. The method of item 22083 wherein the anti- fibrotic agent is an estrogen receptor antagonist.
22119. The method of item 22083 wherein the anti- fibrotic agent is an FGF inhibitor.
22120. The method of item 22083 wherein the anti- fibrotic agent is a farnexyl transferase inhibitor.
22121. The method of item 22083 wherein the anti- fibrotic agent is famesyltransferase inhibitor selected from the group of A- 197574 (Abbott), a famesyltransferase inhibitor from Servier, FPTIII (Strathclyde Institute for Drug R), LB-42908 (LG Life Sciences), Pharmaprojects No. 5063 (Genzyme), Pharmaprojects No. 5597 (Ipsen), Yissum Project No. B-1055 (Yissum), and an analogue or derivative thereof.
22122. The method of item 22083 wherein the anti- fibrotic agent is an FLT-3 kinase inhibitor.
22123. The method of item 22083 wherein the anti- fibrotic agent is an FGF receptor kinase inhibitor.
22124. The method of item 22083 wherein the anti- fibrotic agent is a fibrinogen antagonist selected from the group consisting of AUV-201 (Auvation), MG-13926 (Sanofi-Aventis), plasminogen activator (CAS No. 105913-11-9) (from Sanofi-Aventis or UCB), plasminogen activator-2 (tPA-2) (Sanofi-Aventis), pro-urokinase (CAS No. 82657-92-9) (Sanofi-Aventis), and an analogue or derivative thereof.
22125. The method of item 22083 wherein the anti- fibrotic agent is a heat shock protein 90 antagonist selected from the group consisting of SRN-005 (Sirenade), geldanamycin, NSC-33050 (17- allylaminogeldanamycin; 17-AAG), 17-dimethylaminoethylamino-17- demethoxy-geldanamycin (17-DMAG), rifabutin (rifamycin XIV, 1',4- didehydro-1-deoxy-1,4-dihydro-5'-(2-methylpropyl)-1-oxo-), and an analogue or derivative thereof.
22126. The method of item 22083 wherein the anti- fibrotic agent is a histone deacetylase inhibitor.
22127. The method of item 22083 wherein the anti- fibrotic agent is an HMGCoA reductase inhibitor selected from the group consisting of an atherosclerosis therapeutic from Lipid Sciences, ATI-16000 (ARYx Therapeutics), KS-01-019 (Kos Pharmaceuticals), Pharmaprojects No. 2197 (Sanofi-Aventi), RP 61969 (Sanofi-Aventis), and an analogue or derivative thereof.
22128. The method of item 22083 wherein the anti- fibrotic agent is an ICAM inhibitor.
22129. The method of item 22083 wherein the anti- fibrotic agent is an IL, ICE and IRAK antagonist, wherein the antagonist is a CJ-14877, CP-424174 (Pfizer), NF-61 (Negma-Lerads), and an analogue or derivative thereof.
22130. The method of item 22083 wherein the anti- fibrotic agent is an IL-2 inhibitor.
22131. The method of item 22083 wherein the anti- fibrotic agent is an immunosuppressant selected from the group consisting of teriflunomide (Sanofi Aventis), chlorsulfaquinoxalone (NSC-339004), chlorsulfaquinoxalone sulfate, CS-712 (Sankyo), ismomultin alfa (CAS No. 457913-93-8) (Akzo Nobel), antiallergics from GenPat77, antiinflammatories or AT-005 (Androclus Therapeutics), autoimmune disease therapeutics from EpiVax, BN-007 (Bone), budesonide (CAS No. 51333-22- 3) (MAP Pharmaceuticals), CO-14 (Genzyme), edratide (CAS No. 433922- 67-9) (Teva), EP-314 (Enanta), eprovafen (CAS No. 101335-99-3) (Sanofi- Aventis), HWA-131 (CAS No. 118788-41-3) (Sanofi-Aventis), immunomodulators from MerLion Pharmaceuticals, immunosuppressives from Alchemia, IPL-12 (Inflazyme), MDL-9563 (CAS No. 27086-86-8) (Sanofi-Aventis), Pharmaprojects No. 2330 (Sanofi-Aventis), Pharmaprojects No. 6426 (Abgenix), PXS-25 (Pharmaxis), rosmarinic acid (CAS No. 20283-92-5) (Sanofi-Aventis), RP 42927 or RP 54745 (CAS No. 135330-08-4) (Sanofi-Aventis), SGN-35 (Seattle Genetics), ST-1959 (Sigma-Tau), type 1 diabetes therapy from SYNX Pharma, UNIL-88 (Debiopharm), VP-025 (Vasogen), VR-694 (Vectura), PRTX-001 (Protalex), and an analogue or derivative thereof.
22132. The method of item 22083 wherein the anti- fibrotic agent is an IMPDH (inosine monophosphate).
22133. The method of item 22083 wherein the anti- fibrotic agent is an integrin antagonist.
22134. The method of item 22083 wherein the anti- fibrotic agent is an interleukin antagonist.
22135. The method of item 22083 wherein the anti- fibrotic agent is an inhibitor of type III receptor tyrosine kinase.
22136. . The method of item 22083 wherein the anti- fibrotic agent is an irreversible inhibitor of enzyme methionine aminopeptidase type 2.
22137. The method of item 22083 wherein the anti- fibrotic agent is an isozyme selective delta protein kinase C inhibitor.
22138. The method of item 22083 wherein the anti- fibrotic agent a JAK3 enzyme inhibitor.
22139. The method of item 22083 wherein the anti- fibrotic agent is a JNK inhibitor.
22140. The method of item 22083 wherein the anti- fibrotic agent is a kinase inhibitor. 22141. The method of item 22083 wherein the anti- fibrotic agent is kinesin antagonist.
22142. The method of item 22083 wherein the anti- fibrotic agent is a kinesin antagonist.
22143. The method of item 22083 wherein the anti- fibrotic agent is a leukotriene inhibitor and antagonist selected from the group consisting of ambicromil (CAS No. 58805-38-2) (Sanofi-Aventis), amelubant (CAS No. 346735-24-8) (Boehringer Ingelheim), DW-1141 (Dong Wha), ebselen (Daiichi Pharmaceutical), ibudilast (Kyorin), leucotriene inhibitors from Sanofi-Aventis, lymphotoxin-beta receptor (LT-O) from Biogen Idee, Pharmaprojects No. 1535 and 2728 (CAS No. 119340-33-9) (Sanofi-Aventis), R-112 (Rigel), Rev-5367 (CAS No. 92532-05-3) (Sanofi- Aventis), RG-14893 (CAS No. 141835-49-6) (Sanofi-Aventis), RG-5901-A (CAS No. 101910-24-1), 92532-23-5, RP 66153 (CAS No. 142422-79-5), RP 66364 (CAS No.186916-92-5), RP 69698 (CAS No. 141748-00-7) (Sanofi-Aventis), SC-411930 (Pfizer), SC-41930 (CAS No. 120072-59-5) (Pfizer), SC-50605 (CAS No. 138828-39-4) (Pfizer), SC-51146 (CAS No. 141059-52-1), SC-53228 (CAS No. 153633-01-3) (Pfizer), spaglumic acid (ZY-15106) (CAS No.3106-85-2), 80619-64-3 (Novartis), tipredane (CAS No. 85197-77-9) (Bristol-Myers Squibb), U-75302 (CAS No. 119477-85-9) (Pfizer), and analogue or derivative thereof.
22144. The method of item 22083 wherein the anti- fibrotic agent is an MAP kinase inhibitor.
22145. The method of item 22083 wherein the anti- fibrotic agent is a matrix metalloproteinase inhibitor. 22146. The method of item 22083 wherein the anti- fibrotic agent is an MCP-CCR2 inhibitor.
22147. The method of item 22083 wherein the anti- fibrotic agent is an mTOR inhibitor.
22148. The method of item 22083 wherein the anti- fibrotic agent is an mTOR kinase inhibitor.
22149. The method of item 22083 wherein the anti- fibrotic agent is a microtubule inhibitor selected from the group consisting of antibody-maytansinoid conjugates from Biogen Idee, colchicines (MantiCore Pharmaceuticals), anticancer immunoconjugates from Johnson & Johnson, DIME from Octamer, gni-1f (GNI), huC242-DM4, huMy9-6-DM1 (ImmunoGen), IDN-5404 (Indena), IMO-098, IMOderm (Imotep), mebendazole (Introgen Therapeutics), microtubule poisons from Cambridge Enterprise, paclitaxel such as LOTAX from Aphios (CAS No. 33069-62-4), Genexol-PM from Samyang, Pharmaprojects No. 6383 (Tapestry Pharmaceuticals), RPR-112378 (Sanofi-Aventis), SGN-75 (Seattle Genetics), SPL-7435 (Starpharma), SSR-250411 (Sanofi-Aventis), trastuzumab-DM1 (Genentech), vinorelbine, and an analogue or derivative thereof.
22150. The method of item 22083 wherein the anti- fibrotic agent is an MIF inhibitor.
22151. The method of item 22083 wherein the anti- fibrotic agent is an MMP inhibitor.
22152. The method of item 22083 wherein the anti- fibrotic agent is a neurokinin (NK) antagonist selected from the group consisting of anthrotainin (CAS No. 148084-40-6) (Sanofi-Aventis), an IBS thereapeutic from ArQuIe, MDL-105212A (CAS No. 167261-60-1) (Ssanofi- Aventis), Pharmaprojects No. 2744, 3258 (CAS No. 139167-47-8) 4006, 4201 , or 5986 (Sanofi-Aventis), RP 67580 (CAS No. 135911-02-3), SR- 144190 (CAS No. 201152-86-5), SSR-240600, SSR-241586 (Sanofi- Aventis), TKA-457 (Novartis), vestipitant mesylate (CAS No. 334476-64-1) (GlaxoSmithKline), Win-64821 (Sanofi-Aventis), PRX-96026 (Predix Pharmaceuticals), and an analogue or derivative thereof.
22153. The method of item 22083 wherein the anti- fibrotic agent is an NF kappa B inhibitor selected from the group consisting of AVE-0545 or AVE-0547 (Sanofi-Aventis), bortezomib (CAS No. 179324- 69-7) (Millennium Pharmaceuticals), dexanabinol (CAS No. 112924-45-5) (Pharmos), dexlipotam (Viatris), Pharmaprojects No. 6283 (INDRA) (OXiGENE), IPL-576092 (CAS No. 137571-30-3) (Inflazyme), NFKB decoy (Corgentech), NFKB decoy oligo (AnGes MG), NFKB's from Ariad, osteoporosis treatments or S5 (F005) from Fulcrum Pharmaceuticals, P61 (Phytopharm), R-flurbiprofen (CAS No. 5104-49-4) (Encore Pharmaceuticals), and an analogue or derivative thereof.
22154. The method of item 22083 wherein the anti- fibrotic agent is a nitric oxide agonist.
22155. The method of item 22083 wherein the anti- fibrotic agent is an ornithine decarboxylase inhibitor.
22156. The method of item 22083 wherein the anti- fibrotic agent is a p38 MAP kinase inhibitor selected from the group consisting of AZD-6703 (AstraZeneca), JX-401 (Jexys Pharmaceuticals), BMS-2 (Bristol-Myers Squibb), a p38 MAP kinase inhibitor from Novartis, a p38-alpha MAP kinase inhibitor from Amphora, Pharmaprojects No. 5704 (Pharmacopeia), RPR-200765A (Sanofi-Aventis), SD-282 (Johnson & Johnson), TAK-715 (Takeda), and an analogue or derivative thereof.
22157. The method of item 22083 wherein the anti- fibrotic agent is a palmitoyl-protein thioesterase inhibitor.
22158. The method of item 22083 wherein the anti- fibrotic agent is a PDGF receptor kinase inhibitor selected from the group consisting of AAL-993, AMN-107, or ABP-309 (Novartis), AMG-706 (Amgen), BAY-57-9352 (Bayer), CDP-860 (UCB), E-7080 (Eisai), imatinib (CAS No. 152459-95-5) (Novartis), OSI-930 (OSI Pharmaceuticals), RPR- 127963E (Sanofi-Aventis), RWJ-540973 (Johnson & Johnson), sorafenib tosylate (Bayer), SU-11657 (Pfizer), tandutinib (CAS No. 387867-13-2) (Millennium Pharmaceuticals), vatalanib (Novartis), ZK-CDK (Schering AG), and an analogue or derivative thereof.
22159. The method of item 22083 wherein the anti- fibrotic agent is (-)-halofenate (Metabolex), AMG-131 (Amgen), antidiabetics from Japan Tobacco, AZD-4619, AZD-8450, AZD-8677 (AstraZeneca), DRF-10945, balaglitazone (Dr Reddy's), CS-00088, CS-00098 (Chipscreen Biosciences), E-3030 (Eisai), etalocib (CAS No. 161172-51-6) (EIi Lilly), GSK-641597 (Ligand), GSK-677954 (GlaxoSmithKline), GW-409544 (Ligand), GW-590735 (GlaxoSmithKline), K-111 (Hoffmann-La Roche), LY- 518674 (EIi Lilly), LY-674 (Ligand), LY-929 (Ligand), MC-3001 , MC-3002 (MaxoCore Pharmaceuticals), metformin HCI + pioglitazone (CAS No. 1115- 70-4 and 112529-15-4), ACTOPLUS MET from Andrx), muraglitazar (CAS No. 331741-94-7) (Bristol-Myers Squibb), naveglitazar (Ligand), oleoylethanolamide (Kadmus Pharmaceuticals), ONO-5129, pioglitazone hydrochloride (CAS No. 111025-46-8 and 112529-15-4) (Takeda), PLX-204 (Plexxikon), PPAR agonists from Genfit, PPAR delta agonists from Eli Lilly, PPAR-alpha agonists from CrystalGenomics, PPAR-gamma modulators and PPAR-β modulators from CareX, rosiglitazone maleate (CAS No. 122324- 73-4 or 155141-29-0) (GlaxoSmithKline), rosiglitazone maleate/glimepir (CAS No. 155141-29-0 and 93479-97-1), AVANDARYL, rosiglitazone maleate/metformin extend (CAS No. 155141-29-0 and 657-24-9), AVANDAMET1 rosiglitazone maleate+metformin, AVANDAMET (GlaxoSmithKline), tesaglitazar (AstraZeneca), and an analogue or derivative thereof.
22160. The method of item 22083 wherein the anti- fibrotic agent is a phosphatase inhibitor.
22161. The method of item 22083 wherein the anti- fibrotic agent is a phosphodiesterase (PDE) inhibitor selected from the group consisting of avanafil (Tanabe Seiyaku), dasantafil (CAS No. 569351- 91-3) (Schering-Plough), A-906119 (CAS No. 134072-58-5), DL-850 (Sanofi-Aventis), GRC-30-15, GRC-3566, GRC-3886 (Glenmark), HWA-153 (CAS No. 56395-66-5) (Sanofi-Aventis), hydroxypumafentrine (Altana), IBFB-130011, IBFB-14-016, IBFB-140301 , IBFB-150007, IBFB-211913 (IBFB Pharma), L-826141 (Merck & Co), medorinone (CAS No. 88296-61-1) (Sanofi-Aventis), MEM-1917 (Memory Pharmaceuticals), ND-1251 (Neuro3d), PDE inhibitors from ICOS, PDE IV inhibitors from Memory Pharmaceuticals and CrystalGenomics, Pharmaprojects No. 2742 and 6141 (Sanofi-Aventis), QAD-171 (Novartis), RHC-2963 (CAS No. 76993-12-9 and 76993-14-1), RPR-117658, RPR-122818 derivatives, SR-24870 , and RPR- 132294 (Sanofi-Aventis), SK-350 (ln2Gen), stroke therapy agents from deCODE Genetics, TAS-203 (Taiho), tofimilast (CAS No. 185954-27-2) (Pfizer), UK-371800 (Pfizer), WIN-65579 (CAS No. 158020-82-7) (Sanofi- Aventis), IBFB-130020 (IBFB Pharma), OPC-6535 (CAS No. 145739-56-6) (Otsuka), a phosphodiesterase III inhibitor, enoximone, a phosphodiesterase IV inhibitor, fosfosal, Atopik (Barrier Therapeutics), triflusal, a phosphodiesterase V inhibitor, and an analogue or derivative thereof.
22162. The method of item 22083 wherein the anti- fibrotic agent is a PKC inhibitor.
22163. The method of item 22083 wherein the anti- fibrotic agent is a platelet activating factor antagonist.
22164. The method of item 22083 wherein the anti- fibrotic agent is a platelet-derived growth factor receptor kinase inhibitor.
22165. The method of item 22083 wherein the anti- fibrotic agent is a prolyl hydroxylase inhibitor.
22166. The method of item 22083 wherein the antk fibrotic agent is a polymorphonuclear neutrophil inhibitor.
22167. The method of item 22083 wherein the anti- fibrotic agent is a protein kinase B inhibitor.
22168. The method of item 22083 wherein the anti- fibrotic agent is a protein kinase C stimulant.
22169. The method of item 22083 wherein the anti- fibrotic agent is a purine nucleoside analogue.
22170. The method of item 22083 wherein the anti- fibrotic agent is a purinoreceptor P2X antagonist. 22171. The method of item 22083 wherein the anti- fibrotic agent is a Raf kinase inhibitor.
22172. The method of item 22083 wherein the anti- fibrotic agent is a reversible inhibitor of ErbB1 and ErbB2.
22173. The method of item 22083 wherein the anti- fibrotic agent is a ribonucleoside triphosphate reductase inhibitor.
. 22174. The method of item 22083 wherein the anti- fibrotic agent is an SDF-1 antagonist.
22175. The method of item 22083 wherein the anti- fibrotic agent is a sheddase inhibitor.
22176. The method of item 22083 wherein the anti- fibrotic agent is an SRC inhibitor.
22177. The method of item 22083 wherein the anti- fibrotic agent is a stromelysin inhibitor.
22178. The method of item 22083 wherein the anti- fibrotic agent is an Syk kinase inhibitor.
22179. The method of item 22083 wherein the anti- fibrotic agent is a telomerase inhibitor.
22180. The method of item 22083 wherein the anti- fibrotic agent is a TGF beta inhibitor selected from the group consisting of pirfenidone (CAS No. 53179-13-8) (MARNAC), tranilast (CAS No. 53902- 12-8) (Kissei), IN-1130 (ln2Gen), mannose-6-phosphate (BTG), TGF-β antagonists from Inflazyme (Pharmaprojects No. 6075), TGF-β antagonists from Sydney, non-industrial source), TGF-β I receptor kinase inhibitors from EIi Lilly, TGF-β receptor inhibitors from Johnson & Johnson, and an analogue or derivative thereof.
22181. The method of item 22083 wherein the anti- fibrotic agent is a TNFα antagonist or TACE inhibitor selected from the group consisting of adalimumab (CAS No. 331731-18-1) (Cambridge Antibody Technology), AGIX-4207 (AtheroGenics), AGT-1 (Advanced Biotherapy), an anti-inflammatory from Borean Pharma, Cellzome, or Paradigm Therapeutics, anti-inflammatory vaccine (TNF-alpha kinoid) from Neovacs, humanized anti-TNF antibody or an anti-TNF MAb (CB0006) Celltech (UCB), apratastat (CAS No. 287445-51-0) (Wyeth), BMS-561392 (Bristol-Myers Squibb), BN-006 (Bone), certolizumab pegol (CAS No. 428863-50-7 or CH-138 (UCB), cilomilast (CAS No. 153259-65-5) (GlaxoSmithKline), CR-1 (Nuada-Pharmaceuticals), CRx-119 (CombinatoRx), D-5410 (UCB), dacopafant (CAS No. 125372-33-0) (Sanofi- Aventis), dersalazine (CAS No. 188913-57-7/188913-58-8) (Uriach), etanercept (CAS No. 185243-69-0) (Amgen), ethyl pyruvate (Critical (Critical Therapeutics), golimumab (CAS No. 476181-74-5) (Johnson & Johnson), hormono-immunotherapy from Ipsen, CDP571 (e.g., Humicade from UCB), IC-485 (ICOS), infliximab (CAS No. 170277-31-3) (Johnson & Johnson), IP- 751 (Manhattan Pharmaceuticals), ISIS-104838 (CAS No. 250755-32-9) (ISIS Pharmaceuticals), lenalidomide (CAS No. 191732-72-6) (Celgene), lentinan (CAS No. 37339-90-5) (Ajinomoto), MDL-201112 (CAS No. 142130-73-2) (Sanofi-Aventis), medroxyprogesterone (CAS No. 520-85-4) (InKine Pharmaceutical), N-acetylcysteine (CAS No. 616-91-1) (Zambon), NBE-P2 (DIREVO Biotech), nerelimomab (CAS No. 162774-06-3) (Chiron), OM-294DP (OM PHARMA), onercept (CAS No. 199685-57-9) (Yeda), PASSTNF-alpha (Verigen), pentoxifylline or oxypentifylline (Sanofi-Aventis), Pharmaprojects No. 4091 , 4241 , 4295, or 4488 (Sanofi-Aventis), Pharmaprojects No. 5480 (Amgen), Pharmaprojects No. 6749 (Cengent), pirfenidone (CAS No. 53179-13-8) (MARNAC), RPR-132294 (Sanofi- Aventis), S5 (F002) (Fulcrum Pharmaceuticals), simvastatin (CAS No. 79902-63-9) (Merck & Co), STA-6292 (Synta Pharmaceuticals), tacrolimus (CAS No. 104987-11-3) (Fujisawa LifeCycle Pharma), talactoferrin alfa (CAS No. 308240-58-6) (Agennix), thalidomide (CAS No. 50-35-1) (Celgene), TNF antagonists form ProStrakan, and Synergen, TNF inhibitors (Amgen), TNF-alpha antagonists from Dynavax Technologies and Jerina AG (Germany), TNF-alpha inhibitors from IBFB Pharma and Xencor (Xencor), torbafylline (CAS No. 105102-21-4) (Sanofi-Aventis), UR-1505 (Uriach), VT-346 (Viron Therapeutics), YSIL6 (Y's Therapeutics), YSTH2 (Y's Therapeutics), NPI-1302a-3 (Nereus Pharmaceuticals, a TNF antagonist from Jerina AG (Germany), dersalazine, and an analogue or derivative thereof.
22182. The method of item 22083 wherein the anti- fibrotic agent is a Toll receptor inhibitor.
22183. The method of item 22083 wherein the anti- fibrotic agent is a tubulin antagonist.
22184. The method of item 22083 wherein the anti- fibrotic agent is a tyrosine kinase inhibitor selected from the group consisting of SU-011248, SUTENT from Pfizer Inc. (New York, NY), BMS-354825, PN- 355 (Paracelsian Pharmaceuticals), AGN-199659 (Allergan), AAL-993 or ABP-309 (Novartis), adaphostin (NIH), AEE-788 (Novartis), AG-013736 (OSI Pharmaceuticals), AG-13736 (Pfizer), ALT-110 (Alteris Therapeutics), AMG-706 (Amgen), anticancer MAbs from Xencor, anti-EGFrvlll MAbs from Abgenix, anti-HER2 MAb from Abiogen, AZD-2171 or AZD-9935 (AstraZeneca), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), CEP-5214 (Cephalon), CEP-7055 (Cephalon), cetuximab (ImClone Systems), CHIR-200131 and CHIR-258 (Chiron), CP-547632 (OSI Pharmaceuticals), CP-724714 (Pfizer), CT-301 (Creabilis Therapeutics), D- 69491 (Baxter International), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EGFR/ErbB2 inhibitors from Array BioPharma, erlotinib (CAS No. 183319- 69-9) (OSI Pharmaceuticals), EXEL-2880 (Exelixis), FK-778 (Sanofi- Aventis) , gefitinib (CAS No. 184475-35-2) (AstraZeneca), GW-2286 or GW- 654652 (GlaxoSmithKline), her2/neu antigen from AlphaVax, HER-2/neu inhibitor from Generex, Herzyme (Medipad) (Sirna Therapeutics), HKI-272 (Wyeth), HuMax-EGFr (Genmab), idronoxil (CAS No. 81267-65-4) (Novogen), IGF-1 inhibitors from Ontogen, IMC-11F8 (ImClone Systems), kahalalide F (CAS No. 149204-42-2) (PharmaMar), KDR inhibitor from LG Life Sciences, KDR inhibitors from Abbott Laboratories, KDR kinase inhibitors (UCB), Kdr kinase inhibitors from Merck & Co, KRN-633 and KRN- 951 (Kirin Brewery), KSB-102 (Xenova), lapatinib ditosylate (CAS No. 388082-78-8) (GlaxoSmithKline), matuzumab (Merck KGaA), MDX-214 (Medarex), ME-103 (Pharmexa), MED-A300 (Gerolymatos), MNAC-13 (Lay Line Genomics), nimotuzumab (Center of Molecular Immunology), NSC- 330507 or NSC-707545 (NIH), NV-50 (Novogen), OSI-930 (OSI Pharmaceuticals), panitumumab (Abgenix), pelitinib (CAS No. 287933-82-7) (Wyeth), pertuzumab (CAS No. 380610-27-5) (Genentech), Pharmaprojects No. 3985 (Sanofi-Aventis), prostate cancer therapeutics from Sequenom (SQPC35, SQPC36, SQPC90), removab and remoxab (Trion Pharma), RG- 13022 (CAS No. 136831-48-6), RG-13291 (CAS No. 138989-50-1), or RG- 14620 (CAS No. 136831-49-7) (Sanofi-Aventis), RM-6427 (Romark), RNAi breast cancer therapy from Benitec, RP 53801 (CAS No. 125882-88-4) (Sanofi-Aventis), sorafenib tosylate (Bayer), SU-11657 (Pfizer), Tie-2 antagonists from Semaia (Hybrigenics), Tie-2 inhibitors from Ontogen, trastuzumab (CAS No. 180288-69-1) (Genentech), tyrosine kinase inhibitors from Sanofi-Aventis, U3-1287, U3-1565, U3-1784, U3-1800 (U3 Pharma), vatalanib (Novartis), VEGFR-2 kinase inhibitor from Bristol-Myers Squibb, XL-647 (Exelixis), ZD-6474 (AstraZeneca), ZK-CDK (Schering AG), an EGFR tyrosine kinase inhibitor, EKB-569 (Wyeth), and an analogue or derivative thereof.
22185. The method of item 22083 wherein the anti- fibrotic agent is a VEGF inhibitor.
22186. The method of item 22083 wherein the anti- fibrotic agent is a vitamin D receptor agonist.
22187. The method of item 22083 wherein the anti- fibrotic agent is ZD-6474 (an angiogenesis inhibitor).
22188. The method of item 22083 wherein the anti- fibrotic agent is AP-23573 (an mTOR inhibitor).
22189. The method of item 22083 wherein the anti- fibrotic agent is synthadotin (a tubulin antagonist).
22190. The method of item 22083 wherein the anti- fibrotic agent is S-0885 (a collagenase inhibitor).
22191. The method of item 22083 wherein the anti- fibrotic agent is aplidine (an elongation factor-1 alpha inhibitor).
22192. The method of item 22083 wherein the anti- fibrotic agent is ixabepilone (an epithilone).
22193. The method of item 22083 wherein the anti- fibrotic agent is lDN-5390 (an angiogenesis inhibitor). 22194. The method of item 22083 wherein the anti- fibrotic agent is SB-2723005 (an angiogenesis inhibitor).
22195. The method of item 22083 wherein the anti- fibrotic agent is ABT-518 (an angiogenesis inhibitor).
22196. The method of item 22083 wherein the anti- fibrotic agent is combretastatin (an angiogenesis inhibitor).
22197. The method of item 22083 wherein the anti- fibrotic agent is anecortave acetate (an angiogenesis inhibitor).
22198. The method of item 22083 wherein the anti- fibrotic agent is SB-715992 (a kinesin antagonist).
22199. The method of item 22083 wherein the anti- fibrotic agent is temsirolimus (an mTOR inhibitor).
22200. The method of item 22083 wherein the anti- fibrotic agent is adalimumab (a TNFα antagonist).
22201. The method of item 22083 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with an anti-fibrotic agent, and (b) implanting the medical device into the host.
22202. The method of item 22083 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with an anti-infective agent, and (b) implanting the medical device into the host. 22203. The method of item 22083 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with a polymer; and (b) implanting the medical device into the host.
22204. The method of item 22083 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with a composition comprising an anti-fibrotic agent and a polymer, and (b) implanting the medical device into the host.
22205. The method of item 22083 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with a composition comprising an anti-infective agent and a polymer, and (b) implanting the medical device into the host.
22206. The method of item 22083 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with a composition comprising an anti-fibrotic agent, an anti- infective agent and a polymer, and (b) implanting the medical device into the host.
22207. The method of item 22083 wherein the anti- infective agent is an anthracycline.
22208. The method of item 22083 wherein the anti- infective agent is doxorubicin.
22209. The method of item 22083 wherein the anti- infective agent is is mitoxantrone. 22210. The method of item 22083 wherein the anti- infective agent is a fluoropyrimidine.
22211. The method of item 22083 wherein the anti- infective agent is 5-fluorouracil (5-FU).
22212. The method of item 22083 wherein the anti- infective agent is a folic acid antagonist.
22213. The method of item 22083 wherein the anti- infective agent is methotrexate.
22214. The method of item 22083 wherein the anti- infective agent is a podophylotoxin.
22215. The method of item 22083 wherein the anti- infective agent is etoposide.
22216. The method of item 22083 wherein the anti- infective agent is camptothecin.
22217. The method of item 22083 wherein the anti- infective agent is a hydroxyurea.
22218. The method of item 22083 wherein the anti- infective agent is a platinum complex.
22219. The method of item 22083 wherein the anti- infective agent is cisplatin. 22220. The method of item 22083 wherein the composition comprises an antithrombotic agent.
22221. The method of item 22083 wherein the polymer is formed from reactants comprising a naturally occurring polymer.
22222. The method of item 22083 wherein the polymer is formed from reactants comprising protein.
22223. The method of item 22083 wherein the polymer is formed from reactants comprising carbohydrate.
22224. The method of item 22083 wherein the polymer is formed from reactants comprising biodegradable polymer.
22225. The method of item 22083 wherein the polymer - is formed from reactants comprising nonbiodegradable polymer.
22226. The method of item 22083 wherein the polymer is formed from reactants comprising collagen.
22227. The method of item 22083 wherein the polymer is formed from reactants comprising methylated collagen.
22228. The method of item 22083 wherein the polymer is formed from reactants comprising fibrinogen.
22229. The method of item 22083 wherein the polymer is formed from reactants comprising thrombin. 22230. The method of item 22083 wherein the polymer reactants comprising blood plasma.
22231. The method of item 22083 wherein the polymer reactants comprising calcium salt.
22232. The method of item 22083 wherein the polymer reactants comprising an antifibronolytic agent.
22233. The method of item 22083 wherein the polymer reactants comprising fibrinogen analog.
22234. The method of item 22083 wherein the polymer reactants comprising albumin.
22235. The method of item 22083 wherein the polymer reactants comprising plasminogen.
22236. The method of item 22083 wherein the polymer reactants comprising von Willebrands factor.
22237. The method of item 22083 wherein the polymer reactants comprising Factor VIII.
22238. The method of item 22083 wherein the polymer reactants comprising hypoallergenic collagen.
22239. The method of item 22083 wherein the polymer reactants comprising atelopeptidic collagen. 22240. The method of item 22083 wherein the polymer is formed from reactants comprising telopeptide collagen.
22241. The method of item 22083 wherein the polymer is formed from reactants comprising crosslinked collagen.
22242. The method of item 22083 wherein the polymer is formed from reactants comprising aprotinin.
22243. The method of item 22083 wherein the polymer is formed from reactants comprising epsilon-amino-n-caproic acid.
22244. The method of item 22083 wherein the polymer is formed from reactants comprising gelatin.
22245. The method of item 22083 wherein the polymer is formed from reactants comprising protein conjugates.
22246. The method of item 22083 wherein the polymer is formed from reactants comprising gelatin conjugates.
22247. The method of item 22083 wherein the polymer is formed from reactants comprising a synthetic polymer.
22248. The method of item 22083 wherein the polymer is formed from reactants comprising a synthetic isocyanate-containing compound.
22249. The method of item 22083 wherein the polymer is formed from reactants comprising a synthetic thiol-containing compound. 22250. The method of item 22083 wherein the polymer is formed from reactants comprising a synthetic compound containing at least two thiol groups.
22251. The method of item 22083 wherein the polymer is formed from reactants comprising a synthetic compound containing at least three thiol groups.
22252. The method of item 22083 wherein the polymer is formed from reactants comprising a synthetic compound containing at least four thiol groups.
22253. The method of item 22083 wherein the polymer is formed from reactants comprising a synthetic amino-containing compound.
22254. The method of item 22083 wherein the polymer is formed from reactants comprising a synthetic compound containing at least two amino groups.
22255. The method of item 22083 wherein the polymer is formed from reactants comprising a synthetic compound containing at least three amino groups.
22256. The method of item 22083 wherein the polymer is formed from reactants comprising a synthetic compound containing at least four amino groups.
22257. The method of item 22083 wherein the polymer is formed from reactants comprising a synthetic compound comprising a carbonyl-oxygen-succinimidyi group. 22258. The method of item 22083 wherein the polymer is formed from reactants comprising a synthetic compound comprising at least two carbonyl-oxygen-succinimidyl groups.
22259. The method of item 22083 wherein the polymer is formed from reactants comprising a synthetic compound comprising at least three carbonyl-oxygen-succinimidyl groups.
22260. The method of item 22083 wherein the polymer is formed from reactants comprising a synthetic compound comprising at least four carbonyl-oxygen-succinimidyl groups.
22261. The method of item 22083 wherein the polymer is formed from reactants comprising a synthetic polyalkylene oxide- containing compound.
22262. The method of item 22083 wherein the polymer is formed from reactants comprising a synthetic compound comprising both polyalkylene oxide and biodegradable polyester blocks.
22263. The method of item 22083 wherein the polymer is formed from reactants comprising a synthetic polyalkylene oxide- containing compound having reactive amino groups.
22264. The method of item 22083 wherein the polymer is formed from reactants comprising a synthetic polyalkylene oxide- containing compound having reactive thiol groups.
22265. The method of item 22083 wherein the polymer is formed from reactants comprising a synthetic polyalkylene oxide- containing compound having reactive carbonyl-oxygen-succinimidyl groups. 22266. The method of item 22083 wherein the polymer is formed from reactants comprising a synthetic compound comprising a biodegradable polyester block.
22267. The method of item 22083 wherein the polymer is formed from reactants comprising a synthetic polymer formed in whole or part from lactic acid or lactide.
22268. The method of item 22083 wherein the polymer is formed from reactants comprising a synthetic polymer formed in whole or part from glycolic acid or glycolide.
22269. The method of item 22083 wherein the polymer is formed from reactants comprising polylysine.
22270. The method of item 22083 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound comprising a polyalkylene oxide portion.
22271. The method of item 22083 wherein the polymer is formed from reactants comprising (a) protein and (b) polylysine.
22272. The method of item 22083 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound having at least four thiol groups.
22273. The method of item 22083 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound having at least four amino groups. 22274. The method of item 22083 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound having at least four carbonyl-oxygen-succinimide groups.
22275. The method of item 22083 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound having a biodegradable region formed from reactants selected from lactic acid, lactide, glycolic acid, glycolide, and epison-caprolactone.
22276. The method of item 22083 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound comprising a polyalkylene oxide portion.
22277. The method of item 22083 wherein the polymer is formed from reactants comprising (a) collagen and (b) polylysine.
22278. The method of item 22083 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound having at least four thiol groups.
22279. The method of item 22083 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound having at least four amino groups.
22280. The method of item 22083 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound having at least four carbonyl-oxygen-succinimide groups.
22281. The method of item 22083 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound having a biodegradable region formed from reactants selected from lactic acid, lactide, glycolic acid, glycolide, and epison-caprolactone.
22282. The method of item 22083 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound comprising a polyalkylene oxide portion.
22283. The method of item 22083 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) polylysine.
22284. The method of item 22083 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound having at least four thiol groups.
22285. The method of item 22083 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound having at least four amino groups.
22286. The method of item 22083 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound having at least four carbonyl-oxygen-succinimide groups.
22287. The method of item 22083 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound having a biodegradable region formed from reactants selected from lactic acid, lactide, glycolic acid, glycolide, and epison-caprolactone.
22288. The method of item 22083 wherein the polymer is formed from reactants comprising hyaluronic acid. 22289. The method of item 22083 wherein the polymer is formed from reactants comprising a hyaluronic acid derivative.
22290. The method of item 22083 wherein the polymer is formed from reactants comprising pentaerythritol poly(ethylene glycol)ether tetra-sulfhydryl of number average molecular weight between 3,000 and 30,000.
22291. The method of item 22083 wherein the polymer is formed from reactants comprising pentaerythritol poly(ethylene glycol)ether tetra-amino of number average molecular weight between 3,000 and 30,000.
22292. The method of item 22083 wherein the polymer is formed from reactants comprising (a) a synthetic compound having a number average molecular weight between 3,000 and 30,000 and comprising a polyalkylene oxide region and multiple nucleophilic groups, and (b) a synthetic compound having a number average molecular weight between 3,000 and 30,000 and comprising a polyalkylene oxide region and multiple electrophilic groups.
22293. The method of item 22083 wherein the composition comprises a colorant.
22294. The method of item 22083 wherein the composition is sterile.
22295. A method as in any one of items 22083-22294, wherein the device is a left ventricular assist device. 22296. A method as in any one of items 22083-22294, wherein the device is a right ventricular assist device.
22297. A method as in any one of items 22083-22294, wherein the device is a biventricular assist device.
22298. A method as in any one of items 22083-22294, wherein the device is a cardiac assist device.
22299. A method as in any one of items 22083-22294, wherein the device is a mechanical assist device.
22300. A method as in any one of items 22083-22294, wherein the device is an artificial cardiac assist device.
22301. A method as in any one of items 22083-22294, wherein the device is an implantable heart assist system.
22302. A method as in any one of items 22083-22294, wherein the device is a heart assist pump.
22303. A method as in any one of items 22083-22294, wherein the device is an intraventricular cardiac assist device.
22304. A method for implanting a medical device comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with i) an anti-fibrotic agent, ii) an anti-infective agent, iii) a polymer, iv) a composition comprising an anti-fibrotic agent and a polymer, v) a composition comprising an anti-infective agent and a polymer, or vi) a composition comprising an anti-fibrotic agent, an anti- infective agent and a polymer, and (b) implanting the medical device into the host, wherein the medical device is a spinal implant.
22305. The method of item 22304 wherein the anti- fibrotic agent is an adensosine A2A receptor antagonist.
22306. The method of item 22304 wherein the anti- fibrotic agent is an AKT inhibitor.
22307. The method of item 22304 wherein the anti- fibrotic agent is an alpha 2 integrin antagonist, wherein the alpha 2 integrin antagonist is Pharmaprojects No. 5754 (Merck KgaA).
22308. The method of item 22304 wherein the anti- fibrotic agent is an alpha 4 integrin antagonist.
22309. The method of item 22304 wherein the anti- fibrotic agent is an alpha 7 nicotinic receptor agonist.
22310. The method of item 22304 wherein the anti- fibrotic agent is an angiogenesis inhibitor selected from the group consisting of AG-12,958 (Pfizer), ATN-161 (Attenuon LLC), neovastat, an angiogenesis inhibitor from Jerina AG (Germany), NM-3 (Mercian), VGA-1155 (Taisho), FCE-26644 (Pfizer), FCE-26950 (Pfizer), FPMA (Meiji Daries), FR-111142 (Fujisawa), GGTI-298, GM-1306 (Ligand), GPA-1734 (Novartis), NNC-47- 0011 (Novo Nordisk), herbamycin (Nippon Kayaku), lenalidomide (Celegene), IP-10 (NIH), ABT-828 (Abbott), KIN-841 (Tokushima University, Japan), SF-1126 (Semafore Pharmaceuticals), laminin technology (NIH), CHIR-258 (Chiron), NVP-AEW541 (Novartis), NVP-AEW541 (Novartis), Vt16907 (Alchemia), OXI-8007 (Oxigene), EG-3306 (Ark Therapeutics), Maspin (Arriva), ABT-567 (Abbott), PPI-2458 (Praecis Pharmaceuticals), CC-5079, CC-4089 (Celgene), HIF-1 alpha inhibitors (Xenova), S-247 (Pfizer), AP-23573 (Ariacl), AZD-9935 (Astra Zeneca), mebendazole (Introgen Therapeutics), MetAP-2 inhibitors (GlaxoSmithKline), AG-615 (Angiogene Pharmaceuticals), Tie-2 antagonists (Hybrigenics), NC-381, CYC-381 , NC-169, NC-219, NC-383, NC-384, NC-407 (Lorus Therapeutics), ATN-224 (Attenuon), ON-01370 (Onconova), Vitronectin antagonists (Amgen), SDX-103 (Salmedix), Vitronectin antagonists (Shire), CHP (Riemser), TEK (Amgen), Anecortave acetate (Alcon), T46.2 (Matrix Therapeutics), HG-2 (Heptagen), TEM antagonists (Genzyme), Oxi-4500 (Oxigene), ATN-161 (Attenuon), WX-293 (Wilex), M-2025 (Metris Therapeutics), Alphastatin (BioActa), YH-16 (Yantai Rongchang), BIBF- 1120 (Boehringer Ingelheim), BAY-57-9352 (Bayer), AS-1404 (Cancer Research Technology), SC-77964 (Pfizer), glycomimetics (BioTie Therapies), TIE-2 Inhibitors (Ontogen), DIMI, Octamer (Octamer), ABR- 215050 (Active Biotech), ABT-518 (Abbott), KDR inhibitors (Abbott), BSF- 466895 (Abbott), SCH-221153 (Schering-Plough), DAC.antiangiogenic (ConjuChem), TFPI (EntreMed), AZD-2171 (Astra-Zenaca), CDC-394 (Celgene), LY290293 (EIi Lilly), IDN-5390 (Indena), Kdr Kinase Inhibitors (Merck), CT-113020, CT-116433, CT-116563, CT-31890, CT-32228) (Cell Therapeutics), A-299620 (Abbott), TWEAK Inhibitor (Amgen), VEGF modulators (Johnson and Johnson), Tum-N53, tumstatin (Genzyme), Thios- 1 , Thios-2 (Thios Pharmaceuticals), MV-6401 (Miravant Medical Technologies), Spisulosine (PharmaMar), CEP-7055 (Cephalon), AUV-201 (Auvation), LM-609 (EIi Lilly), SKF-106615 (AnorMED), Oglufanide disodium (Cytran), BW-114 (Pharminox), Calreticulin (NIH), WX-678 (Wilex), SD-7784 (Pfizer), WX-UK1 (Wilex), SH-268 (Schering AG), 2-Me-PGA (Celgene), S- 137 (Pfizer), ZD-6126 (Angiogene Pharmaceuticals), SG-292 (SignalGen), Benefin (Lane Labs), A6, A36 (Angstrom), SB-2723005 (GlaxoSmithKline), SC-7 (Cell Therapeutics), ZEN-014 (AEterna Zentaris), 2-methoxyestradiol (EntreMed), NK-130119 (Nippon Kayaku), CC-10004 (Celgene), AVE- 8062A (Ajinomoto), Tacedinaline (Pfizer), Actinonin (Tokyo Metropolitan Institute of Medical Science), Lenaϋdomide (Celgene), VGA-1155, BTO-956 (SRI International), ER-68203-00 (Eisai), CT-6685 (UCB), JKC-362 (Phoenix Pharmaceuticals), DMI-3798 (DMI Biosciences, Angiomate (Ipsen), ZD-6474 (AstraZeneca), CEP-5214 (Cephalon), Canstatin (Genzyme), NM-3 (Mercian), Oridigm (MediQuest Therapeutics), Exherin (Adherex), BLS-0597 (Boston Life Sciences), PTC-299 (PTC Therapeutics), NPI-2358 (Nereus Pharmaceuticals), CGP-79787 (Novartis), AEE-788 (Novartis), CKD-732 (Chong Kun Dang), CP-564959 (OSI Pharmaceuticals), CM-101 (CarboMed), CT-2584, CT3501 (Cell Therapeutics), combretastatin and analogues and derivatives thereof (Oxigene), Rebimastat (Bristol-Meyers Squibb), Dextrin 2-sulfate (ML Laboratories), Cilengitide (Merk KGaA), NSC-706704 (Pharminox), KRN- 951 (Kirin Brewery), Ukrain, NSC-631570 (Nowicky Pharma), Tecogalan . sodium (Daiichi Pharmaceutical), Tz-93 (Tsumura), TBC-1635 (Encysive Pharmaceuticals), TAN-1120 (Takeda), Semaxanib (Pfizer), BDI-7800 (Biopharmacopae), SD-186, SD-983 (Bristol-Meyers Squibb), SB-223245 (GlaxoSmithKline), SC-236 (Pfizer), RWJ-590973 (Johnson and Johnson), ILX-1850 (Genzyme), SC-68488, S-836 (Pfizer), CG-55069-11 (CuraGen), Ki-23057 (Kirin Brewery), CCX-700 (Chemoentryx), Pegaptanib octasodium (Giled Sciences), ANGIOCOL (available from Biostratum Inc.), or an analogue or derivative thereof.
22311. The method of item 22304 wherein the anti- fibrotic agent is an apoptosis antagonist.
22312. The method of item 22304 wherein the anti- fibrotic agent is an apoptosis activator.
22313. The method of item 22304 wherein the anti- fibrotic agent is a beta 1 integrin antagonist. 22314. The method of item 22304 wherein the anti- fibrotic agent is a beta tubulin inhibitor.
22315. The method of item 22304 wherein the anti- fibrotic agent is a blocker of enzyme production in Hepatitis C.
22316. The method of item 22304 wherein the anti- fibrotic agent is a Bruton's tyrosine kinase inhibitor.
22317. The method of item 22304 wherein the anti- fibrotic agent is a calcineurin inhibitor.
22318. The method of item 22304 wherein the anti- fibrotic agent is a caspase 3 inhibitor.
22319. The method of item 22304 wherein the anti- fibrotic agent is a CC chemokine receptor antagonist.
22320. The method of item 22304 wherein the anti- fibrotic agent is a cell cycle inhibitor selected from the group consisting of SNS-595 (Sunesis), synthadotin, KRX-0403, and an analogue or derivative thereof.
22321. The method of item 22304 wherein the anti- fibrotic agent is a cathepsin B inhibitor.
22322. The method of item 22304 wherein the anti- fibrotic agent is a cathepsin K inhibitor, wherein the cathepsin K inhibitor is 462795 (GlaxoSmithK)ine), INPL-022-D6 (Amura Therapeutics), or an analogue or derivative thereof. 22323. The method of item 22304 wherein the anti- fibrotic agent is a cathepsin L inhibitor.
22324. The method of item 22304 wherein the anti- fibrotic agent is a CD40 antagonist.
22325. The method of item 22304 wherein the anti- fibrotic agent is a chemokine receptor agonist.
22326. The method of item 22304 wherein the anti- fibrotic agent is a chymase inhibitor.
22327. The method of item 22304 wherein the anti- fibrotic agent is a collagenase antagonist.
22328. The method of item 22304 wherein the anti- fibrotic agent is a CXCR antagonist.
22329. The method of item 22304 wherein the anti- fibrotic agent is a cyclin dependent kinase inhibitor selected from the group consisting of a CDK-1 inhibitor, a CDK-2 inhibitor, a CDK- 4 inhibitor, a CDK-6 inhibitor, a CAK1 inhibitor from GPC Biotech and Bristol-Myers Squibb, RGB-286199 (GPC Biotech), an anticancer agent from Astex Technology, a CAK1 inhibitor from GPC Biotech, a CDK inhibitor from Sanofi-Aventis, a CDK1/CDK2 inhibitor from Amgen, a CDK2 inhibitor from SUGEN-2 (Pfizer), a hearing loss therapy agent (Sound Pharmaceuticals), PD-0332991 (Pfizer), RGB-286199 (GPC Biotech), Ro-0505124 (Hoffmann- La Roche), a Ser/Thr kinase inhibitor from Lilly (EIi Lilly), CVT-2584 (CAS No. 199986-75-9) (CV Therapeutics), and an analogue or derivative thereof. 22330. The method of item 22304 wherein the anti- fibrotic agent is a cyclooxygenase 1 inhibitor.
22331. The method of item 22304 wherein the anti- fibrotic agent is a DHFR inhibitor.
22332. The method of item 22304 wherein the anti- fibrotic agent is a dual integrin inhibitor.
22333. The method of item 22304 wherein the anti- fibrotic agent is an elastase inhibitor.
22334. The method of item 22304 wherein the anti- fibrotic agent is an elongation factor-1 alpha inhibitor.
22335. The method of item 22304 wherein the anti- fibrotic agent is an endothelial growth factor antagonist.
22336. The method of item 22304 wherein the anti- fibrotic agent is an endothelial growth factor receptor kinase inhibitor selected from the group consisting of sorafenib tosylate (Bayer), AAL-993 (Novartis), ABP-309 (Novartis), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EXEL- 2880 (Exelixis), GW-654652 (GlaxoSmithKline), a KDR inhibitor from LG Life Sciences, CT-6685 and CT-6729 (UCB), KRN-633 and KRN-951 (Kirin Brewery), OSI-930 (OSI Pharmaceuticals), SP-5.2 (Supratek Pharma), SU-
11657 (Pfizer), a Tie-2 antagonist (Hybrigenics), a VEGFR-2 kinase inhibitor (Bristol-Myers Squibb), XL-647 (Exelixis), a KDR inhibitor from Abbott Laboratories, sorafenib tosylate, and an analogue or derivative thereof. 22337. The method of item 22304 wherein the anti- fibrotic agent is an endotoxin antagonist.
22338. The method of item 22304 wherein the anti- fibrotic agent is an epothilone and tubulin binder.
22339. The method of item 22304 wherein the anti- fibrotic agent is an estrogen receptor antagonist.
22340. The method of item 22304 wherein the anti- fibrotic agent is an FGF inhibitor. s.
22341. The method of item 22304 wherein the anti- fibrotic agent is a farnexyl transferase inhibitor.
22342. The method of item 22304 wherein the anti- fibrotic agent is farnesyltransferase inhibitor selected from the group of A- 197574 (Abbott), a farnesyltransferase inhibitor from Servier, FPTIII (Strathclyde Institute for Drug R), LB-42908 (LG Life Sciences), Pharmaprojects No. 5063 (Genzyme), Pharmaprojects No. 5597 (Ipsen), Yissum Project No. B-1055 (Yissum), and an analogue or derivative thereof.
22343. The method of item 22304 wherein the anti- fibrotic agent is an FLT-3 kinase inhibitor.
22344. The method of item 22304 wherein the anti- fibrotic agent is an FGF receptor kinase inhibitor.
22345. The method of item 22304 wherein the anti- fibrotic agent is a fibrinogen antagonist selected from the group consisting of AUV-201 (Auvation), MG-13926 (Sanofi-Aventis), plasminogen activator (CAS No. 105913-11-9) (from Sanofi-Aventis or UCB), plasminogen activator-2 (tPA-2) (Sanofi-Aventis), pro-urokinase (CAS No. 82657-92-9) (Sanofi-Aventis), and an analogue or derivative thereof.
22346. The method of item 22304 wherein the anti- fibrotic agent is a heat shock protein 90 antagonist selected from the group consisting of SRN-005 (Sirenade), geldanamycin, NSC-33050 (17- allylaminogeldanamycin; 17-AAG), 17-dimethylaminoethylamino-17- demethoxy-geldanamycin (17-DMAG), rifabutin (rifamycin XIV, 1',4- didehydro-1-deoxy-1,4-dihydro-5'-(2-methylpropyl)-1-oxo-), and an analogue or derivative thereof.
22347. The method of item 22304 wherein the anti- fibrotic agent is a histone deacetylase inhibitor.
22348. The method of item 22304 wherein the anti- fibrotic agent is an HMGCoA reductase inhibitor selected from the group consisting of an atherosclerosis therapeutic from Lipid Sciences, ATI-16000 (ARYx Therapeutics), KS-01-019 (Kos Pharmaceuticals), Pharmaprojects No. 2197 (Sanofi-Aventi), RP 61969 (Sanofi-Aventis), and an analogue or derivative thereof.
22349. The method of item 22304 wherein the anti- fibrotic agent is an ICAM inhibitor.
22350. The method of item 22304 wherein the anti- fibrotic agent is an IL, ICE and IRAK antagonist, wherein the antagonist is a CJ-14877, CP-424174 (Pfizer), NF-61 (Negma-Lerads), and an analogue or derivative thereof. 22351. The method of item 22304 wherein the anti- fibrotic agent is an IL-2 inhibitor.
22352. The method of item 22304 wherein the anti- fibrotic agent is an immunosuppressant selected from the group consisting of teriflunomide (Sanofi Aventis), chlorsulfaquinoxalone (NSC-339004), chlorsulfaquinoxalone sulfate, CS-712 (Sankyo), ismomultin alfa (CAS No. 457913-93-8) (Akzo Nobel), antiallergics from GenPat77, antiinflammatories or AT-005 (Androclus Therapeutics), autoimmune disease therapeutics from EpiVax, BN-007 (Bone), budesonide (CAS No. 51333-22- 3) (MAP Pharmaceuticals), CO-14 (Genzyme), edratide (CAS No. 433922- 67-9) (Teva), EP-314 (Enanta), eprovafen (CAS No. 101335-99-3) (Sanofi- Aventis), HWA-131 (CAS No. 118788-41-3) (Sanofi-Aventis), immunomodulators from MerLion Pharmaceuticals, immunosuppressives from Alchemia, IPL-12 (Inflazyme), MDL-9563 (CAS No. 27086-86-8) (Sanofi-Aventis), Pharmaprojects No. 2330 (Sanofi-Aventis), Pharmaprojects No. 6426 (Abgenix), PXS-25 (Pharmaxis), rosmarinic acid (CAS No. 20283-92-5) (Sanofi-Aventis), RP 42927 or RP 54745 (CAS No. 135330-08-4) (Sanofi-Aventis), SGN-35 (Seattle Genetics), ST-1959 (Sigma-Tau), type I diabetes therapy from SYNX Pharma, UNIL-88 (Debiopharm), VP-025 (Vasogen), VR-694 (Vectura), PRTX-001 (Protalex), and an analogue or derivative thereof.
22353. The method of item 22304 wherein the anti- fibrotic agent is an IMPDH (inosine monophosphate).
22354. The method of item 22304 wherein the anti- fibrotic agent is an integrin antagonist.
22355. The method of item 22304 wherein the anti- fibrotic agent is an interleukin antagonist. 22356. The method of item 22304 wherein the anti- fibrotic agent is an inhibitor of type III receptor tyrosine kinase.
22357. The method of item 22304 wherein the anti- fibrotic agent is an irreversible inhibitor of enzyme methionine aminopeptidase type 2.
22358. The method of item 22304 wherein the anti- fibrotic agent is an isozyme selective delta protein kinase C inhibitor.
22359. The method of item 22304 wherein the anti- fibrotic agent a JAK3 enzyme inhibitor.
22360. The method of item 22304 wherein the anti- fibrotic agent is a JNK inhibitor.
22361. The method of item 22304 wherein the anti- fibrotic agent is a kinase inhibitor.
22362. The method of item 22304 wherein the anti- fibrotic agent is kinesin antagonist.
22363. The method of item 22304 wherein the anti- fibrotic agent is a kinesin antagonist.
22364. The method of item 22304 wherein the anti- fibrotic agent is a leukotriene inhibitor and antagonist selected from the group consisting of ambicromil (CAS No. 58805-38-2) (Sanofi-Aventis), amelubant (CAS No. 346735-24-8) (Boehringer Ingelheim), DW-1141 (Dong Wha), ebselen (Daiichi Pharmaceutical), ibudilast (Kyorin), leucotriene inhibitors from Sanofi-Aventis, lymphotoxin-beta receptor (LT-D) from Biogen Idee, Pharmaprojects No. 1535 and 2728 (CAS No. 119340-33-9) (Sanofi-Aventis), R-112 (Rigel), Rev-5367 (CAS No. 92532-05-3) (Sanofi- Aventis), RG-14893 (CAS No. 141835-49-6) (Sanofi-Aventis), RG-5901-A (CAS No. 101910-24-1), 92532-23-5, RP 66153 (CAS No. 142422-79-5), RP 66364 (CAS No.186916-92-5), RP 69698 (CAS No. 141748-00-7) (Sanofi-Aventis), SC-411930 (Pfizer), SC-41930 (CAS No. 120072-59-5) (Pfizer), SC-50605 (CAS No. 138828-39-4) (Pfizer), SC-51146 (CAS No. 141059-52-1), SC-53228 (CAS No. 153633-01-3) (Pfizer), spaglumic acid (ZY-15106) (CAS No.3106-85-2), 80619-64-3 (Novartis), tipredane (CAS No. 85197-77-9) (Bristol-Myers Squibb), U-75302 (CAS No. 119477-85-9) (Pfizer), and analogue or derivative thereof.
22365. The method of item 22304 wherein the anti- fibrotic agent is an MAP kinase inhibitor.
22366. The method of item 22304 wherein the anti- fibrotic agent is a matrix metalloproteinase inhibitor.
22367. The method of item 22304 wherein the anti- fibrotic agent is an MCP-CCR2 inhibitor.
22368. The method of item 22304 wherein the anti- fibrotic agent is an mTOR inhibitor.
22369. The method of item 22304 wherein the anti- fibrotic agent is an mTOR kinase inhibitor.
22370. The method of item 22304 wherein the anti- fibrotic agent is a microtubule inhibitor selected from the group consisting of antibody-maytansinoid conjugates from Biogen Idee, colchicines (MantiCore Pharmaceuticals), anticancer immunoconjugates from Johnson & Johnson, DIME from Octamer, gni-1f (GNI)1 huC242-DM4, huMy9-6-DM1 (ImmunoGen), IDN-5404 (Indena), IMO-098, IMOderm (Imotep), mebendazole (Introgen Therapeutics), microtubule poisons from Cambridge Enterprise, paclitaxel such as LOTAX from Aphios (CAS No. 33069-62-4), Genexol-PM from Samyang, Pharmaprojects No. 6383 (Tapestry Pharmaceuticals), RPR-112378 (Sanofi-Aventis), SGN-75 (Seattle Genetics), SPL-7435 (Starpharma), SSR-250411 (Sanofi-Aventis), trastuzumab-DM1 (Genentech), vinorelbine, and an analogue or derivative thereof.
22371. The method of item 22304 wherein the anti- fibrotic agent is an MIF inhibitor.
22372. The method of item 22304 wherein the anti- fibrotic agent is an MMP inhibitor.
22373. The method of item 22304 wherein the anti- fibrotic agent is a neurokinin (NK) antagonist selected from the group consisting of anthrotainin (CAS No. 148084-40-6) (Sanofi-Aventis), an IBS thereapeutic from ArQuIe, MDL-105212A (CAS No. 167261-60-1) (Ssanofi- Aventis), Pharmaprojects No. 2744, 3258 (CAS No. 139167-47-8) 4006, 4201 , or 5986 (Sanofi-Aventis), RP 67580 (CAS No. 135911-02-3), SR- 144190 (CAS No. 201152-86-5), SSR-240600, SSR-241586 (Sanofi- Aventis), TKA-457 (Novartis), vestipitant mesylate (CAS No. 334476-64-1) (GlaxoSmithKline), Win-64821 (Sanofi-Aventis), PRX-96026 (Predix Pharmaceuticals), and an analogue or derivative thereof.
22374. The method of item 22304 wherein the anti- fibrotic agent is an NF kappa B inhibitor selected from the group consisting of AVE-0545 or AVE-0547 (Sanofi-Aventis), bortezomib (CAS No. 179324- 69-7) (Millennium Pharmaceuticals), dexanabinol (CAS No. 112924-45-5) (Pharmos), dexlipotam (Viatris), Pharmaprojects No. 6283 (INDRA) (OXiGENE), IPL-576092 (CAS No. 137571-30-3) (Inflazyme), NFKB decoy (Corgentech), NFKB decoy oligo (AnGes MG), NFKB's from Ariad, osteoporosis treatments or S5 (F005) from Fulcrum Pharmaceuticals, P61 (Phytopharm), R-flurbiprofen (CAS No. 5104-49-4) (Encore Pharmaceuticals), and an analogue or derivative thereof.
22375. The method of item 22304 wherein the anti- fibrotic agent is a nitric oxide agonist.
22376. The method of item 22304 wherein the anti- fibrotic agent is an ornithine decarboxylase inhibitor.
22377. The method of item 22304 wherein the anti- fibrotic agent is a p38 MAP kinase inhibitor selected from the group consisting of AZD-6703 (AstraZeneca), JX-401 (Jexys Pharmaceuticals), BMS-2 (Bristol-Myers Squibb), a p38 MAP kinase inhibitor from Novartis, a p38-alpha MAP kinase inhibitor from Amphora, Pharmaprojects No. 5704 (Pharmacopeia), RPR-200765A (Sanofi-Aventis), SD-282 (Johnson & Johnson), TAK-715 (Takeda), and an analogue or derivative thereof.
22378. The method of item 22304 wherein the anti- fibrotic agent is a palmitoyl-protein thioesterase inhibitor.
22379. The method of item 22304 wherein the anti- fibrotic agent is a PDGF receptor kinase inhibitor selected from the group consisting of AAL-993, AMN-107, or ABP-309 (Novartis), AMG-706 (Amgen), BAY-57-9352 (Bayer), CDP-860 (UCB), E-7080 (Eisai), imatinib (CAS No. 152459-95-5) (Novartis), OSI-930 (OSI Pharmaceuticals), RPR- 127963E (Sanofi-Aventis), RWJ-540973 (Johnson & Johnson), sorafenib tosylate (Bayer), SU-11657 (Pfizer), tandutinib (CAS No. 387867-13-2) (Millennium Pharmaceuticals), vatalanib (Novartis), ZK-CDK (Schering AG), and an analogue or derivative thereof.
22380. The method of item 22304 wherein the anti- fibrotic agent is (-)-halofenate (Metabolex), AMG-131 (Amgen), antidiabetics from Japan Tobacco, AZD-4619, AZD-8450, AZD-8677 (AstraZeneca), DRF-10945, balaglitazone (Dr Reddy's), CS-00088, CS-00098 (Chipscreen Biosciences), E-3030 (Eisai), etalocib (CAS No. 161172-51-6) (EIi Lilly), GSK-641597 (Ligand), GSK-677954 (GlaxoSmithKline), GW-409544 (Ligand), GW-590735 (GlaxoSmithKline), K- 111 (Hoffmann-La Roche), LY- 518674 (EIi Lilly), LY-674 (Ligand), LY-929 (Ligand), MC-3001 , MC-3002 (MaxoCore Pharmaceuticals), metformin HCI + pioglitazone (CAS No. 1115- 70-4 and 112529-15-4), ACTOPLUS MET from Andrx), muraglitazar (CAS No. 331741-94-7) (Bristol-Myers Squibb), naveglitazar (Ligand), oleoylethanolamide (Kadmus Pharmaceuticals), ONO-5129, pioglitazone hydrochloride (CAS No. 111025-46-8 and 112529-15-4) (Takeda), PLX-204 (Plexxikon), PPAR agonists from Genfit, PPAR delta agonists from EIi Lilly, PPAR-alpha agonists from CrystalGenomics, PPAR-gamma modulators and PPAR-E modulators from CareX, rosiglitazone maleate (CAS No. 122324- 73-4 or 155141-29-0) (GlaxoSmithKline), rosiglitazone maleate/glimepir (CAS No. 155141-29-0 and 93479-97-1), AVANDARYL, rosiglitazone maleate/metformin extend (CAS No. 155141-29-0 and 657-24-9), AVANDAMET, rosiglitazone maleate+metformin, AVANDAMET (GlaxoSmithKline), tesaglitazar (AstraZeneca), and an analogue or derivative thereof.
22381. The method of item 22304 wherein the anti- fibrotic agent is a phosphatase inhibitor.
22382. The method of item 22304 wherein the anti- fibrotic agent is a phosphodiesterase (PDE) inhibitor selected from the group consisting of avanafil (Tanabe Seiyaku), dasantafil (CAS No. 569351- 91-3) (Schering-Plough), A-906119 (CAS No. 134072-58-5), DL-850 (Sanofi-Aventis), GRC-3015, GRC-3566, GRC-3886 (Glenmark), HWA-153 (CAS No. 56395-66-5) (Sanofi-Aventis), hydroxypumafentrine (Altana), IBFB-130011 , IBFB-14-016, IBFB-140301, IBFB-150007, IBFB-211913 (IBFB Pharma), L-826141 (Merck & Co), medorinone (CAS No. 88296-61-1) (Sanofi-Aventis), MEM-1917 (Memory Pharmaceuticals), ND-1251 (Neuro3d), PDE inhibitors from ICOS, PDE IV inhibitors from Memory Pharmaceuticals and CrystalGenomics, Pharmaprojects No. 2742 and 6141 (Sanofi-Aventis), QAD-171 (Novartis), RHC-2963 (CAS No. 76993-12-9 and 76993-14-1), RPR-117658, RPR-122818 derivatives, SR-24870 , and RPR- 132294 (Sanofi-Aventis), SK-350 (ln2Gen), stroke therapy agents from deCODE Genetics, TAS-203 (Taiho), tofimilast (CAS No. 185954-27-2) (Pfizer), UK-371800 (Pfizer), WIN-65579 (CAS No. 158020-82-7) (Sanofi- Aventis), IBFB-130020 (IBFB Pharma), OPC-6535 (CAS No. 145739-56-6) (Otsuka), a phosphodiesterase III inhibitor, enoximone, a phosphodiesterase IV inhibitor, fosfosal, Atopik (Barrier Therapeutics), triflusal, a phosphodiesterase V inhibitor, and an analogue or derivative thereof.
22383. The method of item 22304 wherein the anti- fibrotic agent is a PKC inhibitor.
22384. The method of item 22304 wherein the anti- fibrotic agent is a platelet activating factor antagonist.
22385. The method of item 22304 wherein the anti- fibrotic agent is a platelet-derived growth factor receptor kinase inhibitor.
22386. The method of item 22304 wherein the anti- fibrotic agent is a prolyl hydroxylase inhibitor. 22387. The method of item 22304 wherein the anti- fibrotic agent is a polymorphonuclear neutrophil inhibitor.
22388. The method of item 22304 wherein the anti- fibrotic agent is a protein kinase B inhibitor.
22389. The method of item 22304 wherein the anti- fibrotic agent is a protein kinase C stimulant.
22390. The method of item 22304 wherein the anti- fibrotic agent is a purine nucleoside analogue.
22391. The method of item 22304 wherein the anti- fibrotic agent is a purinoreceptor P2X antagonist.
22392. The method of item 22304 wherein the anti- fibrotic agent is a Raf kinase inhibitor.
22393. The method of item 22304 wherein the anti- fibrotic agent is a reversible inhibitor of ErbB1 and ErbB2.
22394. The method of item 22304 wherein the anti- fibrotic agent is a ribonucleoside triphosphate reductase inhibitor.
22395. The method of item 22304 wherein the anti- fibrotic agent is an SDF-1 antagonist.
22396. The method of item 22304 wherein the anti- fibrotic agent is a sheddase inhibitor. 22397. The method of item 22304 wherein the anti- fibrotic agent is an SRC inhibitor.
22398. The method of item 22304 wherein the anti- fibrotic agent is a stromelysin inhibitor.
22399. The method of item 22304 wherein the anti- fibrotic agent is an Syk kinase inhibitor.
22400. The method of item 22304 wherein the anti- fibrotic agent is a telomerase inhibitor.
22401. The method of item 22304 wherein the anti- fibrotic agent is a TGF beta inhibitor selected from the group consisting of pirfenidone (CAS No. 53179-13-8) (MARNAC), tranilast (CAS No. 53902- 12-8) (Kissei), IN-1130 (ln2Gen), mannose-6-phosphate (BTG), TGF-β antagonists from Inflazyme (Pharmaprojects No. 6075), TGF-β antagonists from Sydney, non-industrial source), TGF-β I receptor kinase inhibitors from EIi Lilly, TGF-β receptor inhibitors from Johnson & Johnson, and an analogue or derivative thereof.
22402. The method of item 22304 wherein the anti- fibrotic agent is a TNFα antagonist or TACE inhibitor selected from the group consisting of adalimumab (CAS No. 331731-18-1) (Cambridge Antibody Technology), AGIX-4207 (AtheroGenics), AGT-1 (Advanced Biotherapy), an anti-inflammatory from Borean Pharma, Cellzome, or Paradigm Therapeutics, anti-inflammatory vaccine (TNF-alpha kinoid) from Neovacs, humanized anti-TNF antibody or an anti-TNF MAb (CB0006) Celltech (UCB), apratastat (CAS No. 287445-51-0) (Wyeth), BMS-561392 (Bristol-Myers Squibb), BN-006 (Bone), certolizumab pegol (CAS No. 428863-50-7 or CH-138 (UCB), cilomilast (CAS No. 153259-65-5) (GlaxoSmithKline), CR-1 (Nuada Pharmaceuticals), CRx-119 (CombinatoRx), D-5410 (UCB), dacopafant (CAS No. 125372-33-0) (Sanofi- Aventis), dersalazine (CAS No. 188913-57-7/188913-58-8) (Uriach), etanercept (CAS No. 185243-69-0) (Amgen), ethyl pyruvate (Critical (Critical Therapeutics), golimumab (CAS No. 476181-74-5) (Johnson & Johnson), hormono-immunotherapy from Ipsen, CDP571 (e.g., Humicade from UCB), IC-485 (ICOS), infliximab (CAS No. 170277-31-3) (Johnson & Johnson), IP- 751 (Manhattan Pharmaceuticals), ISIS-104838 (CAS No. 250755-32-9) (ISIS Pharmaceuticals), lenalidomide (CAS No. 191732-72-6) (Celgene), lentinan (CAS No. 37339-90-5) (Ajinomoto), MDL-201112 (CAS No. 142130-73-2) (Sanofi-Aventis), medroxyprogesterone (CAS No. 520-85-4) (InKine Pharmaceutical), N-acetylcysteine (CAS No. 616-91-1) (Zambon), NBE-P2 (DIREVO Biotech), nerelimomab (CAS No. 162774-06-3) (Chiron), OM-294DP (OM PHARMA), onercept (CAS No. 199685-57-9) (Yeda), PASSTNF-alpha (Verigen), pentoxifylline or oxypentifylline (Sanofi-Aventis), Pharmaprojects No. 4091 , 4241 , 4295, or 4488 (Sanofi-Aventis), Pharmaprojects No. 5480 (Amgen), Pharmaprojects No. 6749 (Cengent), pirfenidone (CAS No. 53179-13-8) (MARNAC), RPR-132294 (Sanofi- Aventis), S5 (F002) (Fulcrum Pharmaceuticals), simvastatin (CAS No. 79902-63-9) (Merck & Co), STA-6292 (Synta Pharmaceuticals), tacrolimus (CAS No. 104987-11-3) (Fujisawa LifeCycle Pharma), talactoferrin alfa (CAS No. 308240-58-6) (Agennix), thalidomide (CAS No. 50-35-1) (Celgene), TNF antagonists form ProStrakan, and Synergen, TNF inhibitors (Amgen), TNF-alpha antagonists from Dynavax Technologies and Jerina AG (Germany), TNF-alpha inhibitors from IBFB Pharma and Xencor (Xencor), torbafylline (CAS No. 105102-21-4) (Sanofi-Aventis), UR-1505 (Uriach), VT-346 (Viron Therapeutics), YSIL6 (Y1S Therapeutics), YSTH2 (Y's Therapeutics), NPI-1302a-3 (Nereus Pharmaceuticals, a TNF antagonist from Jerina AG (Germany), dersalazine, and an analogue or derivative thereof. 22403. The method of item 22304 wherein the anti- fibrotic agent is a Toll receptor inhibitor.
22404. The method of item 22304 wherein the anti- fibrotic agent is a tubulin antagonist.
22405. The method of item 22304 wherein the anti- fibrotic agent is a tyrosine kinase inhibitor selected from the group consisting of SU-011248, SUTENT from Pfizer Inc. (New York, NY), BMS-354825, PN- 355 (Paracelsian Pharmaceuticals), AGN-199659 (Allergan), AAL-993 or ABP-309 (Novartis), adaphostin (NIH), AEE-788 (Novartis), AG-013736 (OSI Pharmaceuticals), AG-13736 (Pfizer), ALT-110 (Alteris Therapeutics), AMG-706 (Amgen), anticancer MAbs from Xencor, anti-EGFrvlll MAbs from Abgenix, anti-HER2 MAb from Abiogen, AZD-2171 or AZD-9935 (AstraZeneca), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), CEP-5214 (Cephalon), CEP-7055 (Cephalon), cetuximab (ImClone Systems), CHIR-200131 and CHIR-258 (Chiron), CP-547632 (OSI Pharmaceuticals), CP-724714 (Pfizer), CT-301 (Creabilis Therapeutics), D- 69491 (Baxter International), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EGFR/ErbB2 inhibitors from Array BioPharma, erlotinib (CAS No. 183319- 69-9) (OSI Pharmaceuticals), EXEL-2880 (Exelixis), FK-778 (Sanofi- Aventis) , gefitinib (CAS No. 184475-35-2) (AstraZeneca), GW-2286 or GW- 654652 (GlaxoSmithKline), her2/neu antigen from AlphaVax, HER-2/neu inhibitor from Generex, Herzyme (Medipad) (Sirna Therapeutics), HKI-272 (Wyeth), HuMax-EGFr (Genmab), idronoxil (CAS No. 81267-65-4) (Novogen), IGF-1 inhibitors from Ontogen, IMC-11F8 (ImClone Systems), kahalalide F (CAS No. 149204-42-2) (PharmaMar), KDR inhibitor from LG Life Sciences, KDR inhibitors from Abbott Laboratories, KDR kinase inhibitors (UCB), Kdr kinase inhibitors from Merck & Co, KRN-633 and KRN- 951 (Kirin Brewery), KSB-102 (Xenova), lapatinib ditosylate (CAS No. 388082-78-8) (GlaxoSmithKline), matuzumab (Merck KGaA), MDX-214 (Medarex), ME-103 (Pharmexa), MED-A300 (Gerolymatos), MNAC-13 (Lay Line Genomics), nimotuzumab (Center of Molecular Immunology), NSC- 330507 or NSC-707545 (NlH), NV-50 (Novogen), OSI-930 (OSI Pharmaceuticals), panitumumab (Abgenix), pelitinib (CAS No. 287933-82-7) (Wyeth), pertuzumab (CAS No. 380610-27-5) (Genentech), Pharmaprojects No. 3985 (Sanofi-Aventis), prostate cancer therapeutics from Sequenom (SQPC35, SQPC36, SQPC90), removab and remoxab (Trion Pharma), RG- 13022 (CAS No. 136831-48-6), RG-13291 (CAS No. 138989-50-1), or RG- 14620 (CAS No. 136831-49-7) (Sanofi-Aventis), RM-6427 (Romark), RNAi breast cancer therapy from Benitec, RP 53801 (CAS No. 125882-88-4) (Sanofi-Aventis), sorafenib tosylate (Bayer), SU-11657 (Pfizer), Tie-2 antagonists from Semaia (Hybrigenics), Tie-2 inhibitors from Ontogen, trastuzumab (CAS No. 180288-69-1) (Genentech), tyrosine kinase inhibitors from Sanofi-Aventis, U3-1287, U3-1565, U3-1784, U3-1800 (U3 Pharma), vatalanib (Novartis), VEGFR-2 kinase inhibitor from Bristol-Myers Squibb, XL-647 (Exelixis), ZD-6474 (AstraZeneca), ZK-CDK (Schering AG), an EGFR tyrosine kinase inhibitor, EKB-569 (Wyeth), and an analogue or derivative thereof.
22406. The method of item 22304 wherein the anti- fibrotic agent is a VEGF inhibitor.
22407. The method of item 22304 wherein the anti- fibrotic agent is a vitamin D receptor agonist.
22408. The method of item 22304 wherein the anti- fibrotic agent is ZD-6474 (an angiogenesis inhibitor).
22409. The method of item 22304 wherein the anti- fibrotic agent is AP-23573 (an mTOR inhibitor). 22410. The method of item 22304 wherein the anti- fibrotic agent is synthadotin (a tubulin antagonist).
22411. The method of item 22304 wherein the anti- fibrotic agent is S-0885 (a collagenase inhibitor).
22412. The method of item 22304 wherein the anti- fibrotic agent is aplidine (an elongation factor-1 alpha inhibitor).
22413. The method of item 22304 wherein the anti- fibrotic agent is ixabepilone (an epithilone).
22414. The method of item 22304 wherein the anti- fibrotic agent is IDN-5390 (an angiogenesis inhibitor).
22415. The method of item 22304 wherein the anti- fibrotic agent is SB-2723005 (an angiogenesis inhibitor).
22416. The method of item 22304 wherein the anti- fibrotic agent is ABT-518 (an angiogenesis inhibitor).
22417. The method of item 22304 wherein the anti- fibrotic agent is combretastatin (an angiogenesis inhibitor).
22418. The method of item 22304 wherein the anti- fibrotic agent is anecortave acetate (an angiogenesis inhibitor).
22419. The method of item 22304 wherein the anti- fibrotic agent is SB-715992 (a kinesin antagonist). 22420. The method of item 22304 wherein the anti- fibrotic agent is temsirolimus (an mTOR inhibitor).
22421. The method of item 22304 wherein the anti- fibrotic agent is adalimumab (a TNFα antagonist).
22422. The method of item 22304 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with an anti-fibrotic agent, and (b) implanting the medical device into the host.
22423. The method of item 22304 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with an anti-infective agent, and (b) implanting the medical device into the host.
22424. The method of item 22304 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with a polymer; and (b) implanting the medical device into the host.
22425. The method of item 22304 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with a composition comprising an anti-fibrotic agent and a polymer, and (b) implanting the medical device into the host.
22426. The method of item 22304 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with a composition comprising an anti-infective agent and a polymer, and (b) implanting the medical device into the host. 22427. The method of item 22304 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with a composition comprising an anti-fibrotic agent, an anti- infective agent and a polymer, and (b) implanting the medical device into the host.
22428. The method of item 22304 wherein the anti- infective agent is an anthracycline.
22429. The method of item 22304 wherein the anti- infective agent is doxorubicin.
22430. The method of item 22304 wherein the anti- infective agent is is mitoxantrone.
22431. The method of item 22304 wherein the anti- infective agent is a fluoropyrimidine.
22432. The method of item 22304 wherein the anti- infective agent is 5-fluorouracil (5-FU).
22433. The method of item 22304 wherein the anti- infective agent is a folic acid antagonist.
22434. The method of item 22304 wherein the anti- infective agent is methotrexate.
22435. The method of item 22304 wherein the anti- infective agent is a podophylotoxin. 22436. The method of item 22304 wherein the anti- infective agent is etoposide.
22437. The method of item 22304 wherein the anti- infective agent is camptothecin.
22438. The method of item 22304 wherein the anti- infective agent is a hydroxyurea.
22439. The method of item 22304 wherein the anti- infective agent is a platinum complex.
22440. The method of item 22304 wherein the anti- infective agent is cisplatin.
22441. The method of item 22304 wherein the composition comprises an anti-thrombotic agent.
22442. The method of item 22304 wherein the polymer is formed from reactants comprising a naturally occurring polymer.
22443. The method of item 22304 wherein the polymer is formed from reactants comprising protein.
22444. The method of item 22304 wherein the polymer is formed from reactants comprising carbohydrate.
22445. The method of item 22304 wherein the polymer is formed from reactants comprising biodegradable polymer. 22446. The method of item 22304 wherein the polymer reactants comprising nonbiodegradable polymer.
22447. The method of item 22304 wherein the polymer reactants comprising collagen.
22448. The method of item 22304 wherein the polymer reactants comprising methylated collagen.
22449. The method of item 22304 wherein the polymer reactants comprising fibrinogen.
22450. The method of item 22304 wherein the polymer reactants comprising thrombin.
22451. The method of item 22304 wherein the polymer reactants comprising blood plasma.
22452. The method of item 22304 wherein the polymer reactants comprising calcium salt.
22453. The method of item 22304 wherein the polymer reactants comprising an antifibronolytic agent.
22454. The method of item 22304 wherein the polymer reactants comprising fibrinogen analog.
22455. The method of item 22304 wherein the polymer reactants comprising albumin. 22456. The method of item 22304 wherein the polymer reactants comprising plasminogen.
22457. The method of item 22304 wherein the polymer reactants comprising von Willebrands factor.
22458. The method of item 22304 wherein the polymer reactants comprising Factor VIII.
22459. The method of item 22304 wherein the polymer reactants comprising hypoallergenic collagen.
22460. The method of item 22304 wherein the polymer reactants comprising atelopeptidic collagen.
22461. The method of item 22304 wherein the polymer reactants comprising telopeptide collagen.
22462. The method of item 22304 wherein the polymer reactants comprising crosslinked collagen.
22463. The method of item 22304 wherein the polymer reactants comprising aprotinin.
22464. The method of item 22304 wherein the polymer reactants comprising epsilon-amino-n-caproic acid.
22465. The method of item 22304 wherein the polymer reactants comprising gelatin. 22466. The method of item 22304 wherein the polymer is formed from reactants comprising protein conjugates.
22467. The method of item 22304 wherein the polymer is formed from reactants comprising gelatin conjugates.
22468. The method of item 22304 wherein the polymer is formed from reactants comprising a synthetic polymer.
22469. The method of item 22304 wherein the polymer is formed from reactants comprising a synthetic isocyanate-containing compound.
22470. The method of item 22304 wherein the polymer is formed from reactants comprising a synthetic thiol-containing compound.
22471. The method of item 22304 wherein the polymer is formed from reactants comprising a synthetic compound containing at least two thiol groups.
22472. The method of item 22304 wherein the polymer is formed from reactants comprising a synthetic compound containing at least three thiol groups.
22473. The method of item 22304 wherein the polymer is formed from reactants comprising a synthetic compound containing at least four thiol groups.
22474. The method of item 22304 wherein the polymer is formed from reactants comprising a synthetic amino-containing compound. 22475. The method of item 22304 wherein the polymer is formed from reactants comprising a synthetic compound containing at least two amino groups.
22476. The method of item 22304 wherein the polymer is formed from reactants comprising a synthetic compound containing at least three amino groups.
22477. The method of item 22304 wherein the polymer is formed from reactants comprising a synthetic compound containing at least four amino groups.
22478. The method of item 22304 wherein the polymer is formed from reactants comprising a synthetic compound comprising a carbonyl-oxygen-succinimidyl group.
22479. The method of item 22304 wherein the polymer is formed from reactants comprising a synthetic compound comprising at least two carbonyl-oxygen-succinimidyl groups.
22480. The method of item 22304 wherein the polymer is formed from reactants comprising a synthetic compound comprising at least three carbonyl-oxygen-succinimidyl groups.
22481. The method of item 22304 wherein the polymer is formed from reactants comprising a synthetic compound comprising at least four carbonyl-oxygen-succinimidyl groups.
22482. The method of item 22304 wherein the polymer is formed from reactants comprising a synthetic polyalkylene oxide- containing compound. 22483. The method of item 22304 wherein the polymer is formed from reactants comprising a synthetic compound comprising both polyalkylene oxide and biodegradable polyester blocks.
22484. The method of item 22304 wherein the polymer is formed from reactants comprising a synthetic polyalkylene oxide- containing compound having reactive amino groups.
22485. The method of item 22304 wherein the polymer is formed from reactants comprising a synthetic polyalkylene oxide- containing compound having reactive thiol groups.
22486. The method of item 22304 wherein the polymer is formed from reactants comprising a synthetic polyalkylene oxide- containing compound having reactive carbonyl-oxygen-succinimidyl groups.
22487. The method of item 22304 wherein the polymer is formed from reactants comprising a synthetic compound comprising a biodegradable polyester block.
22488. The method of item 22304 wherein the polymer is formed from reactants comprising a synthetic polymer formed in whole or part from lactic acid or lactide.
22489. The method of item 22304 wherein the polymer is formed from reactants comprising a synthetic polymer formed in whole or part from glycolic acid or glycolide.
22490. The method of item 22304 wherein the polymer is formed from reactants comprising polylysine. 22491. The method of item 22304 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound comprising a polyalkylene oxide portion.
22492. The method of item 22304 wherein the polymer is formed from reactants comprising (a) protein and (b) polylysine.
22493. The method of item 22304 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound having at least four thiol groups.
22494. The method of item 22304 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound having at least four amino groups.
22495. The method of item 22304 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound having at least four carbonyl-oxygen-succinimide groups.
22496. The method of item 22304 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound having a biodegradable region formed from reactants selected from lactic acid, lactide, glycolic acid, glycolide, and epison-caprolactone.
22497. The method of item 22304 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound comprising a polyalkylene oxide portion.
22498. The method of item 22304 wherein the polymer is formed from reactants comprising (a) collagen and (b) polylysine. 22499. The method of item 22304 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound having at least four thiol groups.
22500. The method of item 22304 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound having at least four amino groups.
22501. The method of item 22304 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound having at least four carbonyl-oxygen-succinimide groups.
22502. The method of item 22304 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound having a biodegradable region formed from reactants selected from lactic acid, lactide, glycolic acid, glycolide, and epison-caprolactone.
22503. The method of item 22304 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound comprising a polyalkylene oxide portion.
22504. The method of item 22304 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) polylysine.
22505. The method of item 22304 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound having at least four thiol groups. 22506. The method of item 22304 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound having at least four amino groups.
22507. The method of item 22304 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound having at least four carbonyl-oxygen-succinimide groups.
22508. The method of item 22304 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound having a biodegradable region formed from reactants selected from lactic acid, lactide, glycolic acid, glycolide, and epison-caprolactone.
22509. The method of item 22304 wherein the polymer is formed from reactants comprising hyaluronic acid.
22510. The method of item 22304 wherein the polymer is formed from reactants comprising a hyaluronic acid derivative.
22511. The method of item 22304 wherein the polymer is formed from reactants comprising pentaerythritol poly(ethylene glycol)ether tetra-sulfhydryl of number average molecular weight between 3,000 and 30,000.
22512. The method of item 22304 wherein the polymer is formed from reactants comprising pentaerythritol poly(ethylene glycol)ether tetra-amino of number average molecular weight between 3,000 and 30,000.
22513. The method of item 22304 wherein the polymer is formed from reactants comprising (a) a synthetic compound having a number average molecular weight between 3,000 and 30,000 and comprising a polyalkylene oxide region and multiple nucleophilic groups, and (b) a synthetic compound having a number average molecular weight between 3,000 and 30,000 and comprising a polyalkylene oxide region and multiple electrophilic groups.
22514. The method of item 22304 wherein the composition comprises a colorant.
22515. The method of item 22304 wherein the . composition is sterile.
22516. A method as in any one of items 22304-22515, wherein the device is a spinal disc.
22517. A method as in any one of items 22304-22515, wherein the device is a vertebral implant.
22518. A method as in any one of items 22304-22515, wherein the device is a vertebral disc prosthesis.
22519. A method as in any one of items 22304-22515, wherein the device is a lumbar disc implant.
22520. A method as in any one of items 22304-22515, wherein the device is a cervical disc implant.
22521. A method as in any one of items 22304-22515, wherein the device is an intervertebral disc. 22522. A method as in any one of items 22304-22515, wherein the device is a spinal prosthesis.
22523. A method as in any one of items 22304-22515, wherein the device is an artificial disc.
22524. A method as in any one of items 22304-22515, wherein the device is a spinal disc endoprosthesis.
22525. A method as in any one of items 22304-22515, wherein the device is an intervertebral implant.
22526. A method as in any one of items 22304-22515, wherein the device is an implantable spinal graft.
22527. A method as in any one of items 22304-22515, wherein the device is an implantable bone graft.
22528. A method as in any one of items 22304-22515, wherein the device is an artificial lumbar disc.
22529. A method as in any one of items 22304-22515, wherein the device is a spinal nucleus implant.
22530. A method as in any one of items 22304-22515, wherein the device is an intervertebral disc spacer.
22531. A method as in any one of items 22304-22515, wherein the device is a fusion cage. 22532. A method as in any one of items 22304-22515, wherein the device is a fusion basket.
22533. A method as in any one of items 22304-22515, wherein the device is a fusion cage apparatus.
22534. A method as in any one of items 22304-22515, wherein the device is an interbody cage.
22535. A method as in any one of items 22304-22515, wherein the device is an interbody implant.
22536. A method as in any one of items 22304-22515, wherein the device is a fusion cage anchoring device.
22537. A method as in any one of items 22304-22515, wherein the device is a bone fixation apparatus.
22538. A method as in any one of items 22304-22515, wherein the device is a fusion stabilization chamber.
22539. A method as in any one of items 22304-22515, wherein the device is an anchoring bone plate.
22540. A method as in any one of items 22304-22515, wherein the device is a bone screw.
22541. A method for implanting a medical device comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with i) an anti-fibrotic agent, ii) an anti-infective agent, iii) a polymer, iv) a composition comprising an anti-fibrotic agent and a polymer, v) a composition comprising an anti-infective agent and a polymer, or vi) a composition comprising an anti-fibrotic agent, an anti- infective agent and a polymer, and (b) implanting the medical device into the host, wherein the medical device is an implant that provides surgical adhesion barrier.
22542. The method of item 22541 wherein the anti- fibrotic agent is an adensosine A2A receptor antagonist.
22543. The method of item 22541 wherein the anti- fibrotic agent is an AKT inhibitor.
22544. The method of item 22541 wherein the anti- fibrotic agent is an alpha 2 integrin antagonist, wherein the alpha 2 integrin antagonist is Pharmaprojects No. 5754 (Merck KgaA).
22545. The method of item 22541 wherein the anti- fibrotic agent is an alpha 4 integrin antagonist.
22546. The method of item 22541 wherein the anti- fibrotic agent is an alpha 7 nicotinic receptor agonist.
22547. The method of item 22541 wherein the anti- fibrotic agent is an angiogenesis inhibitor selected from the group consisting of AG-12,958 (Pfizer), ATN-161 (Attenuon LLC), neovastat, an angiogenesis inhibitor from Jerina AG (Germany), NM-3 (Mercian), VGA-1155 (Taisho), FCE-26644 (Pfizer), FCE-26950 (Pfizer), FPMA (Meiji Daries), FR-111142 (Fujisawa), GGTI-298, GM-1306 (Ligand), GPA-1734 (Novartis), NNC-47- 0011 (Novo Nordisk), herbamycin (Nippon Kayaku), lenalidomide (Celegene), IP-10 (NIH), ABT-828 (Abbott), KIN-841 (Tokushima University, Japan), SF-1126 (Semafore Pharmaceuticals), laminin technology (NIH), CHIR-258 (Chiron), NVP-AEW541 (Novartis), NVP-AEW541 (Novartis), Vt16907 (Alchemia), OXI-8007 (Oxigene), EG-3306 (Ark Therapeutics), Maspin (Arriva), ABT-567 (Abbott), PPI-2458 (Praecis Pharmaceuticals), CC-5079, CC-4089 (Celgene), HIF-1 alpha inhibitors (Xenova), S-247 (Pfizer), AP-23573 (Ariad), AZD-9935 (Astra Zeneca), mebendazole (Introgen Therapeutics), MetAP-2 inhibitors (GlaxoSmithKline), AG-615 (Angiogene Pharmaceuticals), Tie-2 antagonists (Hybrigenics), NC-381 , CYC-381 , NC-169, NC-219, NC-383, NC-384, NC-407 (Lorus Therapeutics), ATN-224 (Attenuon), ON-01370 (Onconova), Vitronectin antagonists (Amgen), SDX-103 (Salmedix), Vitronectin antagonists (Shire), CHP (Riemser), TEK (Amgen), Anecortave acetate (Alcon), T46.2 (Matrix Therapeutics), HG-2 (Heptagen), TEM antagonists (Genzyme), Oxi-4500 (Oxigene), ATN-161 (Attenuon), WX-293 (Wilex), M-2025 (Metris Therapeutics), Alphastatin (BioActa), YH-16 (Yantai Rongchang), BIBF- 1120 (Boehringer Ingelheim), BAY-57-9352 (Bayer), AS-1404 (Cancer Research Technology), SC-77964 (Pfizer), glycomimetics (BioTie Therapies), TIE-2 Inhibitors (Ontogen), DIMI, Octamer (Octamer), ABR- 215050 (Active Biotech), ABT-518 (Abbott), KDR inhibitors (Abbott), BSF- 466895 (Abbott), SCH-221153 (Schering-Plough), DAC:antiangiogenic (ConjuChem), TFPI (EntreMed), AZD-2171 (Astra-Zenaca), CDC-394 (Celgene), LY290293 (EIi Lilly), IDN-5390 (Indena), Kdr Kinase Inhibitors (Merck), CT-113020, CT-116433, CT-116563, CT-31890, CT-32228) (Cell Therapeutics), A-299620 (Abbott), TWEAK Inhibitor (Amgen), VEGF modulators (Johnson and Johnson), Tum-N53, tumstatin (Genzyme), Thios- 1, Thios-2 (Thios Pharmaceuticals), MV-6401 (Miravant Medical Technologies), Spisulosine (PharmaMar), CEP-7055 (Cephalon), AUV-201 (Auvation), LM-609 (EIi Lilly), SKF-106615 (AnorMED), Oglufanide disodium (Cytran), BW-114 (Pharminox), Calreticulin (NIH), WX-678 (Wilex), SD-7784 (Pfizer), WX-UK1 (Wilex), SH-268 (Schering AG), 2-Me-PGA (Celgene), S- 137 (Pfizer), ZD-6126 (Angiogene Pharmaceuticals), SG-292 (SignalGen), Benefin (Lane Labs), A6, A36 (Angstrom), SB-2723005 (GlaxoSmithKline), SC-7 (Cell Therapeutics), ZEN-014 (AEterna Zentaris), 2-methoxyestradio! (EntreMed), NK-130119 (Nippon Kayaku), CC-10004 (Celgene), AVE- 8062A (Ajinomoto), Tacedinaline (Pfizer), Actinonin (Tokyo Metropolitan Institute of Medical Science), Lenalidomide (Celgene), VGA-1155, BTO-956 (SRI International), ER-68203-00 (Eisai), CT-6685 (UCB), JKC-362 (Phoenix Pharmaceuticals), DMI-3798 (DMI Biosciences, Angiomate (Ipsen), ZD-6474 (AstraZeneca), CEP-5214 (Cephalon), Canstatin (Genzyme), NM-3 (Mercian), Oridigm (MediQuest Therapeutics), Exherin (Adherex), BLS-0597 (Boston Life Sciences), PTC-299 (PTC Therapeutics), NPI-2358 (Nereus Pharmaceuticals), CGP-79787 (Novartis), AEE-788 (Novartis), CKD-732 (Chong Kun Dang), CP-564959 (OSI Pharmaceuticals), CM-101 (CarboMed), CT-2584, CT3501 (Cell Therapeutics), combretastatin and analogues and derivatives thereof (Oxigene), Rebimastat (Bristol-Meyers Squibb), Dextrin 2-sulfate (ML Laboratories), Cilengitide (Merk KGaA), NSC-706704 (Pharminox), KRN- 951 (Kirin Brewery), Ukrain, NSC-631570 (Nowicky Pharma), Tecogalan sodium (Daiichi Pharmaceutical), Tz-93 (Tsumura), TBC-1635 (Encysive Pharmaceuticals), TAN-1120 (Takeda), Semaxanib (Pfizer), BDI-7800 (Biopharmacopae), SD-186, SD-983 (Bristol-Meyers Squibb), SB-223245 (GlaxoSmithKline), SC-236 (Pfizer), RWJ-590973 (Johnson and Johnson), ILX-1850 (Genzyme), SC-68488, S-836 (Pfizer), CG-55069-11 (CuraGen), Ki-23057 (Kirin Brewery), CCX-700 (Chemoentryx), Pegaptanib octasodium (Giled Sciences), ANGIOCOL (available from Biostratum Inc.), or an analogue or derivative thereof.
22548. The method of item 22541 wherein the anti- fibrotic agent is an apoptosis antagonist.
22549. The method of item 22541 wherein the anti- fibrotic agent is an apoptosis activator. 22550. The method of item 22541 wherein the anti- fibrotic agent is a beta 1 integrin antagonist.
22551. The method of item 22541 wherein the anti- fibrotic agent is a beta tubulin inhibitor.
22552. The method of item 22541 wherein the anti- fibrotic agent is a blocker of enzyme production in Hepatitis C.
22553. The method of item 22541 wherein the anti- fibrotic agent is a Bruton's tyrosine kinase inhibitor.
22554. The method of item 22541 wherein the anti- fibrotic agent is a calcineurin inhibitor.
22555. The method of item 22541 wherein the anti- fibrotic agent is a caspase 3 inhibitor.
22556. The method of item 22541 wherein the anti- fibrotic agent is a CC chemokine receptor antagonist.
22557. The method of item 22541 wherein the anti- fibrotic agent is a cell cycle inhibitor selected from the group consisting of SNS-595 (Sunesis), synthadotin, KRX-0403, and an analogue or derivative thereof.
22558. The method of item 22541 wherein the anti- fibrotic agent is a cathepsin B inhibitor.
22559. The method of item 22541 wherein the anti- fibrotic agent is a cathepsin K inhibitor, wherein the cathepsin K inhibitor is 462795 (GlaxoSmithKline), 1NPL-022-D6 (Amura Therapeutics), or an analogue or derivative thereof.
22560. The method of item 22541 wherein the anti- fibrotic agent is a cathepsin L inhibitor.
22561. The method of item 22541 wherein the anti- fibrotic agent is a CD40 antagonist.
22562. The method of item 22541 wherein the anti- fibrotic agent is a chemokine receptor agonist.
22563. The method of item 22541 wherein the anti- fibrotic agent is a chymase inhibitor.
22564. The method of item 22541 wherein the anti- fibrotic agent is a collagenase antagonist.
22565. The method of item 22541 wherein the anti- fibrotic agent is a CXCR antagonist.
22566. The method of item 22541 wherein the anti- fibrotic agent is a cyclin dependent kinase inhibitor selected from the group consisting of a CDK-1 inhibitor, a CDK-2 inhibitor, a CDK- 4 inhibitor, a - CDK-6 inhibitor, a CAK1 inhibitor from GPC Biotech and Bristol-Myers Squibb, RGB-286199 (GPC Biotech), an anticancer agent from Astex Technology, a CAK1 inhibitor from GPC Biotech, a CDK inhibitor from Sanofi-Aventis, a CDK1/CDK2 inhibitor from Amgen, a CDK2 inhibitor from SUGEN-2 (Pfizer), a hearing loss therapy agent (Sound Pharmaceuticals), PD-0332991 (Pfizer), RGB-286199 (GPC Biotech), Ro-0505124 (Hoffmann- La Roche), a Ser/Thr kinase inhibitor from Lilly (EIi Lilly), CVT-2584 (CAS No. 199986-75-9) (CV Therapeutics), and an analogue or derivative thereof.
22567. The method of item 22541 wherein the anti- fibrotic agent is a cyclooxygenase 1 inhibitor.
22568. The method of item 22541 wherein the anti- fibrotic agent is a DHFR inhibitor.
22569. The method of item 22541 wherein the anti- fibrotic agent is a dual integrin inhibitor.
22570. The method of item 22541 wherein the anti- fibrotic agent is an elastase inhibitor.
22571. The method of item 22541 wherein the anti- fibrotic agent is an elongation factor-1 alpha inhibitor.
22572. The method of item 22541 wherein the anti- fibrotic agent is an endothelial growth factor antagonist.
22573. The method of item 22541 wherein the anti- fibrotic agent is an endothelial growth factor receptor kinase inhibitor selected from the group consisting of sorafenib tosylate (Bayer), AAL-993 (Novartis), ABP-309 (Novartis), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EXEL- 2880 (Exelixis), GW-654652 (G laxoSmith Kline), a KDR inhibitor from LG Life Sciences, CT-6685 and CT-6729 (UCB), KRN-633 and KRN-951 (Kirin Brewery), OSI-930 (OSI Pharmaceuticals), SP-5.2 (Supratek Pharma), SU-
11657 (Pfizer), a Tie-2 antagonist (Hybrigenics), a VEGFR-2 kinase inhibitor (Bristol-Myers Squibb), XL-647 (Exelixis), a KDR inhibitor from Abbott Laboratories, sorafenib tosylate, and an analogue or derivative thereof.
22574. The method of item 22541 wherein the anti- fibrotic agent is an endotoxin antagonist.
22575. The method of item 22541 wherein the anti- fibrotic agent is an epothilone and tubulin binder.
22576. The method of item 22541 wherein the anti- fibrotic agent is an estrogen receptor antagonist.
22577. The method of item 22541 wherein the anti- fibrotic agent is an FGF inhibitor.
22578. The method of item 22541 wherein the anti- fibrotic agent is a farnexyl transferase inhibitor.
22579. The method of item 22541 wherein the anti- fibrotic agent is famesyltransferase inhibitor selected from the group of A- 197574 (Abbott), a famesyltransferase inhibitor from Servier, FPTIII (Strathclyde Institute for Drug R), LB-42908 (LG Life Sciences), Pharmaprojects No. 5063 (Genzyme), Pharmaprojects No. 5597 (Ipsen), Yissum Project No. B-1055 (Yissum), and an analogue or derivative thereof.
22580. The method of item 22541 wherein the anti- fibrotic agent is an FLT-3 kinase inhibitor.
22581. The method of item 22541 wherein the anti- fibrotic agent is an FGF receptor kinase inhibitor. 22582. The method of item 22541 wherein the anti- fibrotic agent is a fibrinogen antagonist selected from the group consisting of AUV-201 (Auvation), MG-13926 (Sanofi-Aventis), plasminogen activator (CAS No. 105913-11-9) (from Sanofi-Aventis or UCB), plasminogen activator-2 (tPA-2) (Sanofi-Aventis), pro-urokinase (CAS No. 82657-92-9) (Sanofi-Aventis), and an analogue or derivative thereof.
22583. The method of item 22541 wherein the anti- fibrotic agent is a heat shock protein 90 antagonist selected from the group consisting of SRN-005 (Sirenade), geldanamycin, NSC-33050 (17- allylaminogeldanamycin; 17-AAG), 17-dimethylaminoethylamino-17- demethoxy-geldanamycin (17-DMAG), rifabutin (rifamycin XIV, 1',4- didehydro-1-deoxy-1 ,4-dihydro-5'-(2-methylpropyl)-1-oxo-), and an analogue or derivative thereof.
22584. The method of item 22541 wherein the anti- fibrotic agent is a histone deacetylase inhibitor.
22585. The method of item 22541 wherein the anti- fibrotic agent is an HMGCoA reductase inhibitor selected from the group consisting of an atherosclerosis therapeutic from Lipid Sciences, ATM 6000 (ARYx Therapeutics), KS-01-019 (Kos Pharmaceuticals), Pharmaprojects No. 2197 (Sanofi-Aventi), RP 61969 (Sanofi-Aventis), and an analogue or derivative thereof.
22586. The method of item 22541 wherein the anti- fibrotic agent is an ICAM inhibitor.
22587. The method of item 22541 wherein the anti- fibrotic agent is an IL, ICE and IRAK antagonist, wherein the antagonist is a CJ-14877, CP-424174 (Pfizer), NF-61 (Negma-Lerads), and an analogue or derivative thereof.
22588. The method of item 22541 wherein the anti- fibrotic agent is an IL-2 inhibitor.
22589. The method of item 22541 wherein the anti- fibrotic agent is an immunosuppressant selected from the group consisting of teriflunomide (Sanofi Aventis), chlorsulfaquinoxalone (NSC-339004), chlorsulfaquinoxalone sulfate, CS-712 (Sankyo), ismomultin alfa (CAS No. 457913-93-8) (Akzo Nobel), antiallergics from GenPat77, antiinflammatories or AT-005 (Androclus Therapeutics), autoimmune disease therapeutics from EpiVax, BN-007 (Bone), budesonide (CAS No. 51333-22- 3) (MAP Pharmaceuticals), CO-14 (Genzyme), edratide (CAS No. 433922- 67-9) (Teva), EP-314 (Enanta), eprovafen (CAS No. 101335-99-3) (Sanofi- Aventis), HWA-131 (CAS No. 118788-41-3) (Sanofi-Aventis), immunomodulators from MerLion Pharmaceuticals, immunosuppressives from Alchemia, IPL-12 (Inflazyme), MDL-9563 (CAS No. 27086-86-8) (Sanofi-Aventis), Pharmaprojects No. 2330 (Sanofi-Aventis), Pharmaprojects No. 6426 (Abgenix), PXS-25 (Pharmaxis), rosmarinic acid (CAS No. 20283-92-5) (Sanofi-Aventis), RP 42927 or RP 54745 (CAS No. 135330-08-4) (Sanofi-Aventis), SGN-35 (Seattle Genetics), ST-1959 (Sigma-Tau), type I diabetes therapy from SYNX Pharma, UNIL-88 (Debiopharm), VP-025 (Vasogen), VR-694 (Vectura), PRTX-001 (Protalex), and an analogue or derivative thereof.
22590. The method of item 22541 wherein the anti- fibrotic agent is an IMPDH (inosine monophosphate).
22591. The method of item 22541 wherein the anti- fibrotic agent is an integrin antagonist. 22592. The method of item 22541 wherein the anti- fibrotic agent is an interleukin antagonist.
22593. The method of item 22541 wherein the anti- fibrotic agent is an inhibitor of type III receptor tyrosine kinase.
22594. The method of item 22541 wherein the anti- fibrotic agent is an irreversible inhibitor of enzyme methionine aminopeptidase type 2.
22595. The method of item 22541 wherein the anti- fibrotic agent is an isozyme selective delta protein kinase C inhibitor.
22596. The method of item 22541 wherein the anti- fibrotic agent a JAK3 enzyme inhibitor.
22597. The method of item 22541 wherein the anti- fibrotic agent is a JNK inhibitor.
22598. The method of item 22541 wherein the anti- fibrotic agent is a kinase inhibitor.
22599. The method of item 22541 wherein the anti- fibrotic agent is kinesin antagonist.
22600. The method of item 22541 wherein the anti- fibrotic agent is a kinesin antagonist.
22601. The method of item 22541 wherein the anti- fibrotic agent is a leukotriene inhibitor and antagonist selected from the group consisting of ambicromil (CAS No. 58805-38-2) (Sanofi-Aventis), amelubant (CAS No. 346735-24-8) (Boehringer Ingelheim), DW-1141 (Dong Wha), ebselen (Daiichi Pharmaceutical), ibudilast (Kyorin), leucotriene inhibitors from Sanofi-Aventis, lymphotoxin-beta receptor (LT-D) from Biogen Idee, Pharmaprojects No. 1535 and 2728 (CAS No. 119340-33-9) (Sanofi-Aventis), R-112 (Rigel), Rev-5367 (CAS No. 92532-05-3) (Sanofi- Aventis), RG-14893 (CAS No. 141835-49-6) (Sanofi-Aventis), RG-5901-A (CAS No. 101910-24-1), 92532-23-5, RP 66153 (CAS No. 142422-79-5), RP 66364 (CAS No.186916-92-5), RP 69698 (CAS No. 141748-00-7) (Sanofi-Aventis), SC-411930 (Pfizer), SC-41930 (CAS No. 120072-59-5) (Pfizer), SC-50605 (CAS No. 138828-39-4) (Pfizer), SC-51146 (CAS No. 141059-52-1), SC-53228 (CAS No. 153633-01-3) (Pfizer), spaglumic acid (ZY-15106) (CAS No.3106-85-2), 80619-64-3 (Novartis), tipredane (CAS No. 85197-77-9) (Bristol-Myers Squibb), U-75302 (CAS No. 119477-85-9) (Pfizer), and analogue or derivative thereof.
22602. The method of item 22541 wherein the anti- fibrotic agent is an MAP kinase inhibitor.
22603. The method of item 22541 wherein the anti- fibrotic agent is a matrix metalloproteinase inhibitor.
22604. The method of item 22541 wherein the anti- fibrotic agent is an MCP-CCR2 inhibitor.
22605. The method of item 22541 wherein the anti- fibrotic agent is an mTOR inhibitor.
22606. The method of item 22541 wherein the anti- fibrotic agent is an mTOR kinase inhibitor. 22607. The method of item 22541 wherein the anti- fibrotic agent is a microtubule inhibitor selected from the group consisting of antibody-maytansinoid conjugates from Biogen Idee, colchicines (MantiCore Pharmaceuticals), anticancer immunoconjugates from Johnson & Johnson, DIME from Octamer, gni-1f (GNI), huC242-DM4, huMy9-6-DM1 (ImmunoGen), IDN-5404 (Indena), IMO-098, IMOderm (Imotep), mebendazole (Introgen Therapeutics), microtubule poisons from Cambridge Enterprise, paclitaxel such as LOTAX from Aphios (CAS No. 33069-62-4), Genexol-PM from Samyang, Pharmaprojects No. 6383 (Tapestry Pharmaceuticals), RPR-112378 (Sanofi-Aventis), SGN-75 (Seattle Genetics), SPL-7435 (Starpharma), SSR-250411 (Sanofi-Aventis), trastuzumab-DM1 (Genentech), vinorelbine, and an analogue or derivative thereof.
22608. The method of item 22541 wherein the anti- fibrotic agent is an MIF inhibitor.
22609. The method of item 22541 wherein the anti- fibrotic agent is an MMP inhibitor.
22610. The method of item 22541 wherein the anti- fibrotic agent is a neurokinin (NK) antagonist selected from the group consisting of anthrotainin (CAS No. 148084-40-6) (Sanofi-Aventis), an IBS thereapeutic from ArQuIe, MDL-105212A (CAS No. 167261-60-1) (Ssanofi- Aventis), Pharmaprojects No. 2744, 3258 (CAS No. 139167-47-8) 4006, 4201 , or 5986 (Sanofi-Aventis), RP 67580 (CAS No. 135911-02-3), SR- 144190 (CAS No. 201152-86-5), SSR-240600, SSR-241586 (Sanofi- Aventis), TKA-457 (Novartis), vestipitant mesylate (CAS No. 334476-64-1) (GlaxoSmithKline), Win-64821 (Sanofi-Aventis), PRX-96026 (Predix Pharmaceuticals), and an analogue or derivative thereof. 22611. The method of item 22541 wherein the anti- fibrotic agent is an NF kappa B inhibitor selected from the group consisting of AVE-0545 or AVE-0547 (Sanofi-Aventis), bortezomib (CAS No. 179324- 69-7) (Millennium Pharmaceuticals), dexanabinol (CAS No. 112924-45-5) (Pharmos), dexlipotam (Viatris), Pharmaprojects No. 6283 (INDRA) (OXiGENE), IPL-576092 (CAS No. 137571-30-3) (Inflazyme), NFKB decoy (Corgentech), NFKB decoy oligo (AnGes MG), NFKB's from Ariad, osteoporosis treatments or S5 (F005) from Fulcrum Pharmaceuticals, P61 (Phytopharm), R-flurbiprofen (CAS No. 5104-49-4) (Encore Pharmaceuticals), and an analogue or derivative thereof.
22612. The method of item 22541 wherein the anti- fibrotic agent is a nitric oxide agonist.
22613. The method of item 22541 wherein the anti- fibrotic agent is an ornithine decarboxylase inhibitor.
22614. The method of item 22541 wherein the anti- fibrotic agent is a p38 MAP kinase inhibitor selected from the group consisting of AZD-6703 (AstraZeneca), JX-401 (Jexys Pharmaceuticals), BMS-2 (Bristol-Myers Squibb), a p38 MAP kinase inhibitor from Novartis, a p38-alpha MAP kinase inhibitor from Amphora, Pharmaprojects No. 5704 (Pharmacopeia), RPR-200765A (Sanofi-Aventis), SD-282 (Johnson & Johnson), TAK-715 (Takeda), and an analogue or derivative thereof.
22615. The method of item 22541 wherein the anti- fibrotic agent is a palmitoyl-protein thioesterase inhibitor.
22616. The method of item 22541 wherein the anti- fibrotic agent is a PDGF receptor kinase inhibitor selected from the group consisting of AAL-993, AMN-107, or ABP-309 (Novartis), AMG-706 (Amgen), BAY-57-9352 (Bayer), CDP-860 (UCB), E-7080 (Eisai), imatinib (CAS No. 152459-95-5) (Novartis), OSI-930 (OSI Pharmaceuticals), RPR- 127963E (Sanofi-Aventis), RWJ-540973 (Johnson & Johnson), sorafenib tosylate (Bayer), SU-11657 (Pfizer), tandutinib (CAS No. 387867-13-2) (Millennium Pharmaceuticals), vatalanib (Novartis), ZK-CDK (Schering AG), and an analogue or derivative thereof.
22617. The method of item 22541 wherein the anti- fibrotic agent is (-)-halofenate (Metabolex), AMG-131 (Amgen), antidiabetics from Japan Tobacco, AZD-4619, AZD-8450, AZD-8677 (AstraZeneca), DRF-10945, balaglitazone (Dr Reddy's), CS-00088, CS-00098 (Chipscreen Biosciences), E-3030 (Eisai), etalocib (CAS No. 161172-51-6) (EIi Lilly), GSK-641597 (Ligand), GSK-677954 (GlaxoSmithKline), GW-409544 (Ligand), GW-590735 (GlaxoSmithKline), K-111 (Hoffmann-La Roche), LY- 518674 (EIi Lilly), LY-674 (Ligand), LY-929 (Ligand), MC-3001, MC-3002 (MaxoCore Pharmaceuticals), metformin HCI + pioglitazone (CAS No. 1115- 70-4 and 112529-15-4), ACTOPLUS MET from Andrx), muraglitazar (CAS No. 331741-94-7) (Bristol-Myers Squibb), naveglitazar (Ligand), oleoylethanolamide (Kadmus Pharmaceuticals), ONO-5129, pioglitazone hydrochloride (CAS No. 111025-46-8 and 112529-15-4) (Takeda), PLX-204 (Plexxikon), PPAR agonists from Genfit, PPAR delta agonists from EIi Lilly, PPAR-alpha agonists from CrystalGenomics, PPAR-gamma modulators and PPAR-β modulators from CareX, rosiglitazone maleate (CAS No. 122324- 73-4 or 155141-29-0) (GlaxoSmithKline), rosiglitazone maleate/glimepir (CAS No. 155141-29-0 and 93479-97-1), AVANDARYL, rosiglitazone maleate/metformin extend (CAS No. 155141-29-0 and 657-24-9), AVANDAMET, rosiglitazone maleate+metformin, AVANDAMET (GlaxoSmithKline), tesaglitazar (AstraZeneca), and an analogue or derivative thereof. 22618. The method of item 22541 wherein the anti- fibrotic agent is a phosphatase inhibitor.
22619. The method of item 22541 wherein the anti- fibrotic agent is a phosphodiesterase (PDE) inhibitor selected from the group consisting of avanafil (Tanabe Seiyaku), dasantafil (CAS No. 569351- 91-3) (Schering-Plough), A-906119 (CAS No. 134072-58-5), DL-850 (Sanofi-Aventis), GRC-3015, GRC-3566, GRC-3886 (Glenmark), HWA-153 (CAS No. 56395-66-5) (Sanofi-Aventis), hydroxypumafentrine (Altana), IBFB-130011, IBFB-14-016, IBFB-140301 , IBFB-150007, IBFB-211913 (IBFB Pharma), L-826141 (Merck & Co), medorinone (CAS No. 88296-61-1) (Sanofi-Aventis), MEM-1917 (Memory Pharmaceuticals), ND-1251 (Neuro3d), PDE inhibitors from ICOS, PDE IV inhibitors from Memory Pharmaceuticals and CrystalGenomics, Pharmaprojects No. 2742 and 6141 (Sanofi-Aventis), QAD-171 (Novartis), RHC-2963 (CAS No. 76993-12-9 and 76993-14-1), RPR-117658, RPR-122818 derivatives, SR-24870 , and RPR- 132294 (Sanofi-Aventis), SK-350 (ln2Gen), stroke therapy agents from deCODE Genetics, TAS-203 (Taiho), tofimilast (CAS No. 185954-27-2) (Pfizer), UK-371800 (Pfizer), WIN-65579 (CAS No. 158020-82-7) (Sanofi- Aventis), IBFB-130020 (IBFB Pharma), OPC-6535 (CAS No. 145739-56-6) (Otsuka), a phosphodiesterase III inhibitor, enoximone, a phosphodiesterase IV inhibitor, fosfosal, Atopik (Barrier Therapeutics), triflusal, a phosphodiesterase V inhibitor, and an analogue or derivative thereof.
22620. The method of item 22541 wherein the anti- fibrotic agent is a PKC inhibitor.
22621. The method of item 22541 wherein the anti- fibrotic agent is a platelet activating factor antagonist. 22622. The method of item 22541 wherein the anti- fibrotic agent is a platelet-derived growth factor receptor kinase inhibitor.
22623. The method of item 22541 wherein the anti- fibrotic agent is a prolyl hydroxylase inhibitor.
22624. The method of item 22541 wherein the anti- fibrotic agent is a polymorphonuclear neutrophil inhibitor.
22625. The method of item 22541 wherein the anti- fibrotic agent is a protein kinase B inhibitor.
22626. The method of item 22541 wherein the anti- fibrotic agent is a protein kinase C stimulant.
22627. The method of item 22541 wherein the anti- fibrotic agent is a purine nucleoside analogue.
22628. The method of item 22541 wherein the anti- fibrotic agent is a purinoreceptor P2X antagonist.
22629. The method of item 22541 wherein the anti- fibrotic agent is a Raf kinase inhibitor.
22630. The method of item 22541 wherein the anti- fibrotic agent is a reversible inhibitor of ErbB1 and ErbB2.
22631. The method of item 22541 wherein the anti- fibrotic agent is a ribonucleoside triphosphate reductase inhibitor. 22632. The method of item 22541 wherein the anti- fibrotic agent is an SDF-1 antagonist.
22633. The method of item 22541 wherein the anti- fibrotic agent is a sheddase inhibitor.
22634. The method of item 22541 wherein the anti- fibrotic agent is an SRC inhibitor.
22635. The method of item 22541 wherein the anti- fibrotic agent is a stromelysin inhibitor.
22636. The method of item 22541 wherein the anti- fibrotic agent is an Syk kinase inhibitor.
22637. The method of item 22541 wherein the anti- fibrotic agent is a telomerase inhibitor.
22638. The method of item 22541 wherein the anti- fibrotic agent is a TGF beta inhibitor selected from the group consisting of pirfenidone (CAS No. 53179-13-8) (MARNAC), tranilast (CAS No. 53902- 12-8) (Kissei), IN-1130 (ln2Gen), mannose-6-phosphate (BTG), TGF-β antagonists from Inflazyme (Pharmaprojects No. 6075), TGF-β antagonists from Sydney, non-industrial source), TGF-β I receptor kinase inhibitors from EIi Lilly, TGF-β receptor inhibitors from Johnson & Johnson, and an analogue or derivative thereof.
22639. The method of item 22541 wherein the anti- fibrotic agent is a TNFα antagonist or TACE inhibitor selected from the group consisting of adalimumab (CAS No. 331731-18-1) (Cambridge Antibody Technology), AGIX-4207 (AtheroGenics), AGT-1 (Advanced Biotherapy), an anti-inflammatory from Borean Pharma, Cellzome, or Paradigm Therapeutics, anti-inflammatory vaccine (TNF-alpha kinoid) from Neovacs, humanized anti-TNF antibody or an anti-TNF MAb (CB0006) Celltech (UCB), apratastat (CAS No. 287445-51-0) (Wyeth), BMS-561392 (Bristol-Myers Squibb), BN-006 (Bone), certolizumab pegol (CAS No. 428863-50-7 or CH-138 (UCB), cilomilast (CAS No. 153259-65-5) (GiaxoSmithKline), CR-1 (Nuada Pharmaceuticals), CRx-119 (CombinatoRx), D-5410 (UCB), dacopafant (CAS No. 125372-33-0) (Sanofi- Aventis), dersaiazine (CAS No. 188913-57-7/188913-58-8) (Uriach), etanercept (CAS No. 185243-69-0) (Amgen), ethyl pyruvate (Critical (Critical Therapeutics), golimumab (CAS No. 476181-74-5) (Johnson & Johnson), hormono-immunotherapy from Ipsen, CDP571 (e.g., Humicade from UCB), IC-485 (ICOS), infliximab (CAS No. 170277-31-3) (Johnson & Johnson), IP- 751 (Manhattan Pharmaceuticals), ISIS-104838 (CAS No. 250755-32-9) (ISIS Pharmaceuticals), lenalidomide (CAS No. 191732-72-6) (Celgene), lentinan (CAS No. 37339-90-5) (Ajinomoto), MDL-201112 (CAS No. 142130-73-2) (Sanofi-Aventis), medroxyprogesterone (CAS No. 520-85-4) (InKine Pharmaceutical), N-acetylcysteine (CAS No. 616-91-1) (Zambon), NBE-P2 (DIREVO Biotech), nerelimomab (CAS No. 162774-06-3) (Chiron), OM-294DP (OM PHARMA), onercept (CAS No. 199685-57-9) (Yeda), PASSTNF-alpha (Verigen), pentoxifylline or oxypentifylline (Sanofi-Aventis), Pharmaprojects No. 4091, 4241 , 4295, or 4488 (Sanofi-Aventis), Pharmaprojects No. 5480 (Amgen), Pharmaprojects No. 6749 (Cengent), pirfenidone (CAS No. 53179-13-8) (MARNAC), RPR-132294 (Sanofi- Aventis), S5 (F002) (Fulcrum Pharmaceuticals), simvastatin (CAS No. 79902-63-9) (Merck & Co), STA-6292 (Synta Pharmaceuticals), tacrolimus (CAS No. 104987-11-3) (Fujisawa LifeCycle Pharma), talactoferrin alfa (CAS No. 308240-58-6) (Agennix), thalidomide (CAS No. 50-35-1) (Celgene), TNF antagonists form ProStrakan, and Synergen, TNF inhibitors (Amgen), TNF-alpha antagonists from Dynavax Technologies and Jerina AG (Germany), TNF-alpha inhibitors from IBFB Pharma and Xencor (Xencor), torbafylline (CAS No. 105102-21-4) (Sanofi-Aventis), UR-1505 (Uriach), VT-346 (Viron Therapeutics), YSIL6 (Ys Therapeutics), YSTH2 (Y's Therapeutics), NPI-1302a-3 (Nereus Pharmaceuticals, a TNF antagonist from Jerina AG (Germany), dersalazine, and an analogue or derivative thereof.
22640. The method of item 22541 wherein the anti- fibrotic agent is a Toll receptor inhibitor.
22641. The method of item 22541 wherein the anti- fibrotic agent is a tubulin antagonist.
22642. The method of item 22541 wherein the anti- fibrotic agent is a tyrosine kinase inhibitor selected from the group consisting of SU-011248, SUTENT from Pfizer Inc. (New York, NY), BMS-354825, PN- 355 (Paracelsian Pharmaceuticals), AGN-199659 (Allergan), AAL-993 or ABP-309 (Novartis), adaphostin (NIH), AEE-788 (Novartis), AG-013736 (OSI Pharmaceuticals), AG-13736 (Pfizer), ALT-110 (Alteris Therapeutics), AMG-706 (Amgen), anticancer MAbs from Xencor, anti-EGFrvlll MAbs from Abgenix, anti-HER2 MAb from Abiogen, AZD-2171 or AZD-9935 (AstraZeneca), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), CEP-5214 (Cephalon), CEP-7055 (Cephalon), cetuximab (ImClone Systems), CHIR-200131 and CHIR-258 (Chiron), CP-547632 (OSI Pharmaceuticals), CP-724714 (Pfizer), CT-301 (Creabilis Therapeutics), D- 69491 (Baxter International), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EGFR/ErbB2 inhibitors from Array BioPharma, erlotinib (CAS No. 183319- 69-9) (OSI Pharmaceuticals), EXEL-2880 (Exelixis), FK-778 (Sanofi- Aventis) , gefitinib (CAS No. 184475-35-2) (AstraZeneca), GW-2286 or GW- 654652 (GlaxoSmithKline), her2/neu antigen from AlphaVax, HER-2/neu inhibitor from Generex, Herzyme (Medipad) (Sima Therapeutics), HKI-272 (Wyeth), HuMax-EGFr (Genmab), idronoxil (CAS No. 81267-65-4) (Novogen), IGF-1 inhibitors from Ontogen, IMC-11F8 (ImCIone Systems), kahalalide F (CAS No. 149204-42-2) (PharmaMar), KDR inhibitor from LG Life Sciences, KDR inhibitors from Abbott Laboratories, KDR kinase inhibitors (UCB), Kdr kinase inhibitors from Merck & Co, KRN-633 and KRN- 951 (Kirin Brewery), KSB-102 (Xenova), lapatinib ditosylate (CAS No. 388082-78-8) (GlaxoSmithKline), matuzumab (Merck KGaA), MDX-214 (Medarex), ME-103 (Pharmexa), MED-A300 (Gerolymatos), MNAC-13 (Lay Line Genomics), nimotuzumab (Center of Molecular Immunology), NSC- 330507 or NSC-707545 (NIH), NV-50 (Novogen), OSI-930 (OSI Pharmaceuticals), panitumumab (Abgenix), pelitinib (CAS No. 287933-82-7) (Wyeth), pertuzumab (CAS No. 380610-27-5) (Genentech), Pharmaprojects No. 3985 (Sanofi-Aventis), prostate cancer therapeutics from Sequenom (SQPC35, SQPC36, SQPC90), removab and remoxab (Trion Pharma), RG- 13022 (CAS No. 136831-48-6), RG-13291 (CAS No. 138989-50-1), or RG- 14620 (CAS No. 136831-49-7) (Sanofi-Aventis), RM-6427 (Romark), RNAi breast cancer therapy from Benitec, RP 53801 (CAS No. 125882-88-4) (Sanofi-Aventis), sorafenib tosylate (Bayer), SU-11657 (Pfizer), Tie-2 antagonists from Semaia (Hybrigenics), Tie-2 inhibitors from Ontogen, trastuzumab (CAS No. 180288-69-1) (Genentech), tyrosine kinase inhibitors from Sanofi-Aventis, U3-1287, U3-1565, U3-1784, U3-1800 (U3 Pharma), vatalanib (Novartis), VEGFR-2 kinase inhibitor from Bristol-Myers Squibb, XL-647 (Exelixis), ZD-6474 (AstraZeneca), ZK-CDK (Schering AG), an EGFR tyrosine kinase inhibitor, EKB-569 (Wyeth), and an analogue or derivative thereof.
22643. The method of item 22541 wherein the anti- fibrotic agent is a VEGF inhibitor.
22644. The method of item 22541 wherein the anti- fibrotic agent is a vitamin D receptor agonist. 22645. The method of item 22541 wherein the anti- fibrotic agent is ZD-6474 (an angiogenesis inhibitor).
22646. The method of item 22541 wherein the anti- fibrotic agent is AP-23573 (an mTOR inhibitor).
22647. The method of item 22541 wherein the anti- fibrotic agent is synthadotin (a tubulin antagonist).
22648. The method of item 22541 wherein the anti- fibrotic agent is S-0885 (a collagenase inhibitor).
22649. The method of item 22541 wherein the anti- fibrotic agent is apiidine (an elongation factor- 1 alpha inhibitor).
22650. The method of item 22541 wherein the anti- fibrotic agent is ixabepilone (an epithilone).
22651. The method of item 22541 wherein the anti- fibrotic agent is IDN-5390 (an angiogenesis inhibitor).
22652. The method of item 22541 wherein the anti- fibrotic agent is SB-2723005 (an angiogenesis inhibitor).
22653. The method of item 22541 wherein the anti- fibrotic agent is ABT-518 (an angiogenesis inhibitor).
22654. The method of item 22541 wherein the anti- fibrotic agent is combretastatin (an angiogenesis inhibitor). 22655. The method of item 22541 wherein the anti- fibrotic agent is anecortave acetate (an angiogenesis inhibitor).
22656. The method of item 22541 wherein the anti- fibrotic agent is SB-715992 (a kinesin antagonist).
22657. The method of item 22541 wherein the anti- fibrotic agent is temsirolimus (an mTOR inhibitor).
22658. The method of item 22541 wherein the anti- fibrotic agent is adalimumab (a TN Fa antagonist).
22659. The method of item 22541 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with an anti-fibrotic agent, and (b) implanting the medical device into the host.
22660. The method of item 22541 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with an anti-infective agent, and (b) implanting the medical device into the host.
22661. The method of item 22541 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with a polymer; and (b) implanting the medical device into the host.
22662. The method of item 22541 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with a composition comprising an anti-fibrotic agent and a polymer, and (b) implanting the medical device into the host. 22663. The method of item 22541 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with a composition comprising an anti-infective agent and a polymer, and (b) implanting the medical device into the host.
22664. The method of item 22541 comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with a composition comprising an anti-fibrotic agent, an anti- infective agent and a polymer, and (b) implanting the medical device into the host.
22665. The method of item 22541 wherein the anti- infective agent is an anthracycline.
22666. The method of item 22541 wherein the anti- infective agent is doxorubicin.
22667. The method of item 22541 wherein the anti- infective agent is is mitoxantrone.
22668. The method of item 22541 wherein the anti- infective agent is a fluoropyrimidine.
22669. The method of item 22541 wherein the anti- infective agent is 5-fluorouracil (5-FU).
22670. The method of item 22541 wherein the anti- infective agent is a folic acid antagonist.
22671. The method of item 22541 wherein the anti- infective agent is methotrexate. 22672. The method of item 22541 wherein the anti- infective agent is a podophylotoxin.
22673. The method of item 22541 wherein the anti- infective agent is etoposide.
22674. The method of item 22541 wherein the anti- infective agent is camptothecin.
22675. The method of item 22541 wherein the anti- infective agent is a hydroxyurea.
22676. The method of item 22541 wherein the anti- infective agent is a platinum complex.
22677. The method of item 22541 wherein the anti- infective agent is cisplatin.
22678. The method of item 22541 wherein the composition comprises an anti-thrombotic agent.
22679. The method of item 22541 wherein the polymer is formed from reactants comprising a naturally occurring polymer.
22680. The method of item 22541 wherein the polymer is formed from reactants comprising protein.
22681. The method of item 22541 wherein the polymer is formed from reactants comprising carbohydrate. 22682. The method of item 22541 wherein the polymer reactants comprising biodegradable polymer.
22683. The method of item 22541 wherein the polymer reactants comprising nonbiodegradable polymer.
22684. The method of item 22541 wherein the polymer reactants comprising collagen.
22685. The method of item 22541 wherein the polymer reactants comprising methylated collagen.
22686. The method of item 22541 wherein the polymer reactants comprising fibrinogen.
22687. The method of item 22541 wherein the polymer reactants comprising thrombin.
22688. The method of item 22541 wherein the polymer reactants comprising blood plasma.
22689. The method of item 22541 wherein the polymer reactants comprising calcium salt.
22690. The method of item 22541 wherein the polymer reactants comprising an antifibronolytic agent.
22691. The method of item 22541 wherein the polymer reactants comprising fibrinogen analog. 22692. The method of item 22541 wherein the polymer reactants comprising albumin.
22693. The method of item 22541 wherein the polymer reactants comprising plasminogen.
22694. The method of item 22541 wherein the polymer reactants comprising von Willebrands factor.
22695. The method of item 22541 wherein the polymer reactants comprising Factor VIII.
22696. The method of item 22541 wherein the polymer reactants comprising hypoallergenic collagen.
22697. The method of item 22541 wherein the polymer reactants comprising atelopeptidic collagen.
22698. The method of item 22541 wherein the polymer reactants comprising telopeptide collagen.
22699. The method of item 22541 wherein the polymer reactants comprising crosslinked collagen.
22700. The method of item 22541 wherein the polymer reactants comprising aprotinin.
22701. The method of item 22541 wherein the polymer reactants comprising epsilon-amino-n-caproic acid. 22702. The method of item 22541 wherein the polymer is formed from reactants comprising gelatin.
22703. The method of item 22541 wherein the polymer is formed from reactants comprising protein conjugates.
22704. The method of item 22541 wherein the polymer is formed from reactants comprising gelatin conjugates.
22705. The method of item 22541 wherein the polymer is formed from reactants comprising a synthetic polymer.
22706. The method of item 22541 wherein the polymer is formed from reactants comprising a synthetic isocyanate-containing compound.
22707. The method of item 22541 wherein the polymer is formed from reactants comprising a synthetic thiol-containing compound.
22708. The method of item 22541 wherein the polymer is formed from reactants comprising a synthetic compound containing at least two thiol groups.
22709. The method of item 22541 wherein the polymer is formed from reactants comprising a synthetic compound containing at least three thiol groups.
22710. The method of item 22541 wherein the polymer is formed from reactants comprising a synthetic compound containing at least four thiol groups. 22711. The method of item 22541 wherein the polymer is formed from reactants comprising a synthetic amino-containing compound.
22712. The method of item 22541 wherein the polymer is formed from reactants comprising a synthetic compound containing at least two amino groups.
22713. The method of item 22541 wherein the polymer is formed from reactants comprising a synthetic compound containing at least three amino groups.
22714. The method of item 22541 wherein the polymer is formed from reactants comprising a synthetic compound containing at least four amino groups.
22715. The method of item 22541 wherein the polymer is formed from reactants comprising a synthetic compound comprising a carbonyl-oxygen-succinimidyl group.
22716. The method of item 22541 wherein the polymer is formed from reactants comprising a synthetic compound comprising at least two carbonyl-oxygen-succinimidyl groups.
22717. The method of item 22541 wherein the polymer is formed from reactants comprising a synthetic compound comprising at least three carbonyl-oxygen-succinimidyl groups.
22718. The method of item 22541 wherein the polymer is formed from reactants comprising a synthetic compound comprising at least four carbonyl-oxygen-succinimidyl groups. 22719. The method of item 22541 wherein the polymer is formed from reactants comprising a synthetic polyalkylene oxide- containing compound.
22720. The method of item 22541 wherein the polymer is formed from reactants comprising a synthetic compound comprising both polyalkylene oxide and biodegradable polyester blocks.
22721. The method of item 22541 wherein the polymer is formed from reactants comprising a synthetic polyalkylene oxide- containing compound having reactive amino groups.
22722. The method of item 22541 wherein the polymer is formed from reactants comprising a synthetic polyalkylene oxide- containing compound having reactive thiol groups.
22723. The method of item 22541 wherein the polymer is formed from reactants comprising a synthetic polyalkylene oxide- containing compound having reactive carbonyl-oxygen-succinimidyl groups.
22724. The method of item 22541 wherein the polymer is formed from reactants comprising a synthetic compound comprising a biodegradable polyester block.
22725. The method of item 22541 wherein the polymer is formed from reactants comprising a synthetic polymer formed in whole or part from lactic acid or lactide.
22726. The method of item 22541 wherein the polymer is formed from reactants comprising a synthetic polymer formed in whole or part from glycolic acid or glycolide. 22727. The method of item 22541 wherein the polymer is formed from reactants comprising polylysine.
22728. The method of item 22541 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound comprising a polyalkylene oxide portion.
22729. The method of item 22541 wherein the polymer is formed from reactants comprising (a) protein and (b) polylysine.
22730. The method of item 22541 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound having at least four thiol groups.
22731. The method of item 22541 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound having at least four amino groups.
22732. The method of item 22541 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound having at least four carbonyl-oxygen-succinimide groups.
22733. The method of item 22541 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound having a biodegradable region formed from reactants selected from lactic acid, lactide, glycolic acid, glycolide, and epison-caprolactone.
22734. The method of item 22541 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound comprising a polyalkylene oxide portion. 22735. The method of item 22541 wherein the polymer is formed from reactants comprising (a) collagen and (b) polylysine.
22736. The method of item 22541 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound having at least four thiol groups.
22737. The method of item 22541 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound having at least four amino groups.
22738. The method of item 22541 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound having at least four carbonyl-oxygen-succinimide groups.
22739. The method of item 22541 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound having a biodegradable region formed from reactants selected from lactic acid, lactide, glycolic acid, glycolide, and epison-caprolactone.
22740. The method of item 22541 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound comprising a polyalkylene oxide portion.
22741. The method of item 22541 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) polylysine.
22742. The method of item 22541 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound having at least four thiol groups. 22743. The method of item 22541 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound having at least four amino groups.
22744. The method of item 22541 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound having at least four carbonyl-oxygen-succinimide groups.
22745. The method of item 22541 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound having a biodegradable region formed from reactants selected from lactic acid, lactide, glycolic acid, glycolide, and epison-caprolactone.
22746. The method of item 22541 wherein the polymer is formed from reactants comprising hyaluronic acid.
22747. The method of item 22541 wherein the polymer is formed from reactants comprising a hyaluronic acid derivative.
22748. The method of item 22541 wherein the polymer is formed from reactants comprising pentaerythritol poly(ethylene glycoQether tetra-sulfhydryl of number average molecular weight between 3,000 and 30,000.
22749. The method of item 22541 wherein the polymer is formed from reactants comprising pentaerythritol poly(ethylene glycol)ether tetra-amino of number average molecular weight between 3,000 and 30,000.
22750. The method of item 22541 wherein the polymer is formed from reactants comprising (a) a synthetic compound having a number average molecular weight between 3,000 and 30,000 and comprising a polyalkylene oxide region and multiple nucleophilic groups, and (b) a synthetic compound having a number average molecular weight between 3,000 and 30,000 and comprising a polyalkylene oxide region and multiple electrophilic groups.
22751. The method of item 22541 wherein the composition comprises a colorant.
22752. The method of item 22541 wherein the composition is sterile.
22753. A method for preventing surgical adhesions, comprising delivering (a) (i) a tissue-reactive polymeric composition to a site in need thereof to provide coated tissue, and (a) (ii) delivering a fibrosis- inhibiting agent to the coated tissue; (b) delivering to a site in need thereof an fibrosis-inhibiting agent; or (c) delivering to a site in need thereof a composition that comprises (i) a fibrosis-inhibiting agent and (ii) a polymer or a compound that forms a polymer in situ.
22754. A method of preventing surgical adhesions, comprising delivering a composition between a dural sleeve and paravertebral musculature in a patient post-laminectomy, where the composition prevents surgical adhesions.
22755. A method of preventing surgical adhesions, comprising coating a spinal nerve at a laminectomy site in a patient in need thereof with a composition, where the composition prevents surgical adhesions. 22756. A method of preventing surgical adhesions, comprising infiltrating a composition into tissue around a spinal nerve at a laminectomy site in a patient in need thereof, where the composition prevents surgical adhesions.
22757. A method of preventing surgical adhesions, comprising delivering a composition to a site of a surgical disc resection in a patient in need thereof, where the composition prevents surgical adhesions.
22758. A method of preventing surgical adhesions, comprising delivering a composition to a site of a microdiscectomy in a patient in need thereof, where the composition prevents surgical adhesions.
22759. A method of preventing surgical adhesions, comprising comprising delivering a composition to a site of a neurosurgical (brain) procedure in a patient in need thereof, where the composition prevents surgical adhesions.
22760. A method of preventing surgical adhesions, comprising infiltrating into a spinal surgical site of a patient in need thereof, a composition that prevents surgical adhesions.
22761. A method of preventing surgical adhesions, comprising delivering a composition to epidural tissue in a patient in need thereof, where the composition prevents surgical adhesions.
22762. A method of preventing surgical adhesions, comprising delivering a composition to dural tissue in a patient in need thereof, where the composition prevents surgical adhesions. 22763. A method of preventing surgical adhesions, comprising delivering a composition to a gynecological site in a patient in need thereof, where the composition prevents surgical adhesions.
22764. A method of preventing surgical adhesions, comprising delivering a composition to a tissue surface of the pelvic side wall in a patient in need thereof, where the composition prevents surgical adhesions.
22765. A method of preventing surgical adhesions, comprising delivering a composition to a peritoneal cavity in a patient in need thereof, where the composition prevents surgical adhesions.
22766. A method of preventing surgical adhesions, comprising delivering a composition to a pelvic cavity in a patient in need thereof, where the composition prevents surgical adhesions.
22767. A method of preventing surgical adhesions, comprising delivering a composition to a site of a laparotomy in a patient in need thereof, where the composition prevents surgical adhesions.
22768. A method of preventing surgical adhesions, comprising delivering a composition to a site of an endoscopic procedure in a patient in need thereof, where the composition prevents surgical adhesions.
22769. A method of preventing surgical adhesions, comprising delivering a composition to a site of a hernia repair in a patient in need thereof, where the composition prevents surgical adhesions. 22770. A method of preventing surgical adhesions, comprising delivering a composition to a site of cholecystectomy in a patient in need thereof, where the composition prevents surgical adhesions.
22771. A method of preventing surgical adhesions, comprising delivering a composition to a site of a cardiac procedure in a patient in need thereof, where the composition prevents surgical adhesions.
22772. A method of preventing surgical adhesions, comprising delivering a composition to a site of cardiac transplant surgery in a patient in need thereof, where the composition prevents surgical adhesions.
22773. A method of preventing surgical adhesions, comprising delivering a composition to a site of cardiac vascular repair in a patient in need thereof, where the composition prevents surgical adhesions.
2277 '4. A method of preventing surgical adhesions, comprising delivering a composition to a site of a heart valve replacement in a patient in need thereof, where the composition prevents surgical adhesions.
22775. A method of preventing surgical adhesions, comprising delivering a composition to a site of pericardial surgery in a patient in need thereof, where the composition prevents surgical adhesions.
22776. A method of preventing surgical adhesions, comprising delivering a composition to a site of an orthopedic surgical procedure in a patient in need thereof, where the composition prevents surgical adhesions. 22777. A method of preventing surgical adhesions, comprising delivering a composition to a site of a torn ligament in a patient in need thereof, where the composition prevents surgical adhesions.
22778. A method of preventing surgical adhesions, comprising delivering a composition to a site of joint injury in a patient in need thereof, where the composition prevents surgical adhesions.
22779. A method of preventing surgical adhesions, comprising delivering a composition to a site of a tendon injury in a patient in need thereof, where the composition prevents surgical adhesions.
22780. A method of preventing surgical adhesions, comprising delivering a composition to a site of a cartilage injury in a patient in need thereof, where the composition prevents surgical adhesions.
22781. A method of preventing surgical adhesions, comprising delivering a composition to a site of a muscle injury in a patient in need thereof, where the composition prevents surgical adhesions.
22782. A method of preventing surgical adhesions, comprising delivering a composition to a site of a nerve injury in a patient in need thereof, where the composition prevents surgical adhesions.
22783. A method of preventing surgical adhesions, comprising delivering a composition to a site of a cosmetic surgical procedure in a patient in need thereof, where the composition prevents surgical adhesions.
22784. A method of preventing surgical adhesions, comprising delivering a composition to a site of a reconstructive surgical procedure in a patient in need thereof, where the composition prevents surgical adhesions.
22785. A method of preventing surgical adhesions, comprising delivering a composition to a site of a breast implant in a patient in need thereof, where the composition prevents surgical adhesions.
22786. The method of any one of items 22753-22785 wherein the composition comprises an anti-fibrotic agent, and wherein the anti-fibrotic agent is an adensosine A2A receptor antagonist.
22787. The method of any one of items 22753-22785 wherein the composition comprises an anti-fibrotic agent, and wherein the anti-fibrotic agent is an AKT inhibitor.
22788. The method of any one of items 22753-22785 wherein the composition comprises an anti-fibrotic agent, and wherein the anti-fibrotic agent is Pharmaprojects No. 5754 (Merck KgaA).
22789. The method of any one of items 22753-22785 wherein the composition comprises an anti-fibrotic agent, and wherein the anti-fibrotic agent is an alpha 4 integrin antagonist.
22790. The method of any one of items 22753-22785 wherein the composition comprises an anti-fibrotic agent, and wherein the anti-fibrotic agent is an alpha 7 nicotinic receptor agonist.
22791. The method of any one of items 22753-22785 wherein the composition comprises an anti-fibrotic agent, and wherein the anti-fibrotic agent is an angiogenesis inhibitor selected from the group consisting of AG-12,958 (Pfizer), ATN-161 (Attenuon LLC), neovastat, an angiogenesis inhibitor from Jerina AG (Germany), NM-3 (Mercian), VGA- 1155 (Taisho), FCE-26644 (Pfizer), FCE-26950 (Pfizer), FPMA (Meiji Daries), FR-111142 (Fujisawa), GGTI-298, GM-1306 (Ligand), GPA-1734 (Novartis), NNC-47-0011 (Novo Nordisk), herbamycin (Nippon Kayaku), lenalidomide (Celegene), IP-10 (NIH)1 ABT-828 (Abbott), KIN-841 (Tokushima University, Japan), SF-1126 (Semafore Pharmaceuticals), laminin technology (NIH), CHIR-258 (Chiron), NVP-AEW541 (Novartis), NVP-AEW541 (Novartis), VM6907 (Alchemia), OXI-8007 (Oxigene), EG- 3306 (Ark Therapeutics), Maspin (Arriva), ABT-567 (Abbott), PPI-2458 (Praecis Pharmaceuticals), CC-5079, CC-4089 (Celgene), HIF-1 alpha inhibitors (Xenova), S-247 (Pfizer), AP-23573 (Ariad), AZD-9935 (Astra Zeneca), mebendazole (Introgen Therapeutics), MetAP-2 inhibitors (GlaxoSmithKline), AG-615 (Angiogene Pharmaceuticals), Tie-2 antagonists (Hybrigenics), NC-381 , CYC-381 , NC-169, NC-219, NC-383, NC-384, NC- 407 (Lorus Therapeutics), ATN-224 (Attenuon), ON-01370 (Onconova), Vitronectin antagonists (Amgen), SDX-103 (Salmedix), Vitronectin antagonists (Shire), CHP (Riemser), TEK (Amgen), Anecortave acetate (Alcon), T46.2 (Matrix Therapeutics), HG-2 (Heptagen), TEM antagonists (Genzyme), Oxi-4500 (Oxigene), ATN-161 (Attenuon), WX-293 (Wilex), M- 2025 (Metris Therapeutics), Alphastatin (BioActa), YH-16 (Yantai Rongchang), BIBF-1120 (Boehringer Ingelheim), BAY-57-9352 (Bayer), AS- 1404 (Cancer Research Technology), SC-77964 (Pfizer), glycomimetics (BioTie Therapies), TIE-2 Inhibitors (Ontogen), DIMI, Octamer (Octamer), ABR-215050 (Active Biotech), ABT-518 (Abbott), KDR inhibitors (Abbott), BSF-466895 (Abbott), SCH-221153 (Schering-Plough), DAC:antiangiogenic (ConjuChem), TFPI (EntreMed), AZD-2171 (Astra-Zenaca), CDC-394 (Celgene), LY290293 (EIi Lilly), IDN-5390 (Indena), Kdr Kinase Inhibitors (Merck), CT-113020, CT-116433, CT-116563, CT-31890, CT-32228) (Cell Therapeutics), A-299620 (Abbott), TWEAK Inhibitor (Amgen), VEGF modulators (Johnson and Johnson), Tum-N53, tumstatin (Genzyme), Thios- 1, Thios-2 (Thios Pharmaceuticals), MV-6401 (Miravant Medical Technologies), Spisulosine (PharmaMar), CEP-7055 (Cephalon), AUV-201 (Auvation), LM-609 (EIi Lilly), SKF-106615 (AnorMED), Oglufanide disodium (Cytran), BW-114 (Pharminox), Calreticulin (NIH), WX-678 (Wilex), SD-7784 (Pfizer), WX-UK1 (Wilex), SH-268 (Schering AG), 2-Me-PGA (Celgene), S- 137 (Pfizer), ZD-6126 (Angiogene Pharmaceuticals), SG-292 (SignalGen), Benefin (Lane Labs), A6, A36 (Angstrom), SB-2723005 (GlaxoSmithKline), SC-7 (Cell Therapeutics), ZEN-014 (AEterna Zentaris), 2-methoxyestradiol (EntreMed), NK-130119 (Nippon Kayaku), CC-10004 (Celgene), AVE- 8062A (Ajinomoto), Tacedinaline (Pfizer), Actinonin (Tokyo Metropolitan Institute of Medical Science), Lenalidomide (Celgene), VGA-1155, BTO-956 (SRI International), ER-68203-00 (Eisai), CT-6685 (UCB), JKC-362 (Phoenix Pharmaceuticals), DMI-3798 (DMI Biosciences, Angiomate (Ipsen), ZD-6474 (AstraZeneca), CEP-5214 (Cephalon), Canstatin (Genzyme), NM-3 (Mercian), Oridigm (MediQuest Therapeutics), Exherin (Adherex), BLS-0597 (Boston Life Sciences), PTC-299 (PTC Therapeutics), NPI-2358 (Nereus Pharmaceuticals), CGP-79787 (Novartis), AEE-788 (Novartis), CKD-732 (Chong Kun Dang), CP-564959 (OSI Pharmaceuticals), CM-101 (CarboMed), CT-2584, CT3501 (Cell Therapeutics), combretastatin and analogues and derivatives thereof (Oxigene), Rebimastat (Bristol-Meyers Squibb), Dextrin 2-sulfate (ML Laboratories), Cilengitide (Merk KGaA), NSC-706704 (Pharminox), KRN- 951 (Kirin Brewery), Ukrain, NSC-631570 (Nowicky Pharma), Tecogalan sodium (Daiichi Pharmaceutical), Tz-93 (Tsumura), TBC-1635 (Encysive Pharmaceuticals), TAN-1120 (Takeda), Semaxanib (Pfizer), BDI-7800 (Biopharmacopae), SD-186, SD-983 (Bristol-Meyers Squibb), SB-223245 (GlaxoSmithKline), SC-236 (Pfizer), RWJ-590973 (Johnson and Johnson), ILX-1850 (Genzyme), SC-68488, S-836 (Pfizer), CG-55069-11 (CuraGen), Ki-23057 (Kirin Brewery), CCX-700 (Chemoentryx), Pegaptanib octasodium (Giled Sciences), ANGIOCOL (available from Biostratum Inc.), or an analogue or derivative thereof. 22792. The method of any one of items 22753-22785 wherein the composition comprises an anti-fibrotic agent, and wherein the anti-fibrotic agent is an apoptosis antagonist.
22793. The method of any one of items 22753-22785 wherein the composition comprises an anti-fibrotic agent, and wherein the anti-fibrotic agent is an apoptosis activator.
22794. The method of any one of items 22753-22785 wherein the composition comprises an anti-fibrotic agent, and wherein the anti-fibrotic agent is a beta 1 integrin antagonist.
22795. The method of any one of items 22753-22785 wherein the composition comprises an anti-fibrotic agent, and wherein the anti-fibrotic agent is a beta tubulin inhibitor.
22796. The method of any one of items 22753-22785 wherein the composition comprises an anti-fibrotic agent, and wherein the anti-fibrotic agent is a blocker of enzyme production in Hepatitis C.
22797. The method of any one of items 22753-22785 wherein the composition comprises an anti-fibrotic agent, and wherein the anti-fibrotic agent is a Bruton's tyrosine kinase inhibitor.
22798. The method of any one of items 22753-22785 wherein the composition comprises an anti-fibrotic agent, and wherein the anti-fibrotic agent is a calcineurin inhibitor.
22799. The method of any one of items 22753-22785 wherein the composition comprises an anti-fibrotic agent, and wherein the anti-fibrotic agent is a caspase 3 inhibitor. 22800. The method of any one of items 22753-22785 wherein the composition comprises an anti-fibrotic agent, and wherein the anti-fibrotic agent is a CC chemokine receptor antagonist.
22801. The method of any one of items 22753-22785 wherein the composition comprises an anti-fibrotic agent, and wherein the anti-fibrotic agent is a cell cycle inhibitor selected from the group consisting of SNS-595 (Sunesis), synthadotin, KRX-0403, and an analogue or derivative thereof.
22802. The method of any one of items 22753-22785 wherein the composition comprises an anti-fibrotic agent, and wherein the anti-fibrotic agent is a cathepsin B inhibitor.
22803. The method of any one of items 22753-22785 wherein the composition comprises an anti-fibrotic agent, the anti-fibrotic agent is a cathepsin K inhibitor selected from the group consisting of 462795 (GlaxoSmithKline), INPL-022-D6 (Amura Therapeutics), and an analogue or derivative thereof.
22804. The method of any one of items 22753-22785 wherein the composition comprises an anti-fibrotic agent, and wherein the anti-fibrotic agent is a cathepsin L inhibitor.
22805. The method of any one of items 22753-22785 wherein the composition comprises an anti-fibrotic agent, and wherein the anti-fibrotic agent is a CD40 antagonist.
22806. The method of any one of items 22753-22785 wherein the composition comprises an anti-fibrotic agent, and wherein the anti-fibrotic agent is a chemokine receptor agonist. 22807. The method of any one of items 22753-22785 wherein the composition comprises an anti-fibrotic agent, and wherein the anti-fibrotic agent is a chymase inhibitor.
22808. The method of any one of items 22753-22785 wherein the composition comprises an anti-fibrotic agent, and wherein the anti-fibrotic agent is a collagenase antagonist.
22809. The method of any one of items 22753-22785 wherein the composition comprises an anti-fibrotic agent, and wherein the anti-fibrotic agent is a CXCR antagonist.
22810. The method of any one of items 22753-22785 wherein the composition comprises an anti-fibrotic agent, and wherein the anti-fibrotic agent is a cyclin dependent kinase inhibitor selected from the group consisting of a CDK-1 inhibitor, a CDK-2 inhibitor, a CDK- 4 inhibitor, a CDK-6 inhibitor, a CAK1 inhibitor from GPC Biotech and Bristol-Myers Squibb, RGB-286199 (GPC Biotech), an anticancer agent from Astex Technology, a CAK1 inhibitor from GPC Biotech, a CDK inhibitor from Sanofi-Aventis, a CDK1/CDK2 inhibitor from Amgen, a CDK2 inhibitor from SUGEN-2 (Pfizer), a hearing loss therapy agent (Sound Pharmaceuticals), PD-0332991 (Pfizer), RGB-286199 (GPC Biotech), Ro-0505124 (Hoffmann- La Roche), a Ser/Thr kinase inhibitor from Lilly (EIi Lilly), CVT-2584 (CAS No. 199986-75-9) (CV Therapeutics), and an analogue or derivative thereof.
22811. The method of any one of items 22753-22785 wherein the composition comprises an anti-fibrotic agent, and wherein the anti-fibrotic agent is a cyclooxygenase 1 inhibitor. 22812. The method of any one of items 22753-22785 wherein the composition comprises an anti-fibrotic agent, and wherein the anti-fibrotic agent is a DHFR inhibitor.
22813. The method of any one of items 22753-22785 wherein the composition comprises an anti-fibrotic agent, and wherein the anti-fibrotic agent is a dual integrin inhibitor.
22814. The method of any one of items 22753-22785 wherein the composition comprises an anti-fibrotic agent, and wherein the anti-fibrotic agent is an elastase inhibitor.
22815. The method of any one of items 22753-22785 wherein the composition comprises an anti-fibrotic agent, and wherein the anti-fibrotic agent is an elongation factor-1 alpha inhibitor.
22816. The method of any one of items 22753-22785 wherein the composition comprises an anti-fibrotic agent, and wherein the anti-fibrotic agent is an endothelial growth factor antagonist.
22817. The method of any one of items 22753-22785 wherein the composition comprises an anti-fibrotic agent, and wherein the anti-fibrotic agent is an endothelial growth factor receptor kinase inhibitor selected from the group consisting of sorafenib tosylate (Bayer), AAL-993 (Novartis), ABP-309 (Novartis), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EXEL- 2880 (Exelixis), GW-654652 (GlaxoSmithKline), a KDR inhibitor from LG Life Sciences, CT-6685 and CT-6729 (UCB), KRN-633 and KRN-951 (Kirin Brewery), OSI-930 (OSI Pharmaceuticals), SP-5.2 (Supratek Pharma), SU-
11657 (Pfizer), a Tie-2 antagonist (Hybrigenics), a VEGFR-2 kinase inhibitor (Bristol-Myers Squibb), XL-647 (Exeiixis), a KDR inhibitor from Abbott Laboratories, sorafenib tosylate, and an analogue or derivative thereof.
22818. The method of any one of items 22753-22785 wherein the composition comprises an anti-fibrotic agent, and wherein the anti-fibrotic agent is an endotoxin antagonist.
22819. The method of any one of items 22753-22785 wherein the composition comprises an anti-fibrotic agent, and wherein the anti-fibrotic agent is an epothilone and tubulin binder.
22820. The method of any one of items 22753-22785 wherein the composition comprises an anti-fibrotic agent, and wherein the anti-fibrotic agent is an estrogen receptor antagonist.
22821. The method of any one of items 22753-22785 wherein the composition comprises an anti-fibrotic agent, and wherein the anti-fibrotic agent is an FGF inhibitor.
22822. The method of any one of items 22753-22785 wherein the composition comprises an anti-fibrotic agent, and wherein the anti-fibrotic agent is a famexyl transferase inhibitor.
22823. The method of any one of items 22753-22785 wherein the composition comprises an anti-fibrotic agent, and wherein the anti-fibrotic agent is famesyltransferase inhibitor selected from the group of A-197574 (Abbott), a famesyltransferase inhibitor from Servier, FPTIII (Strathclyde Institute for Drug R), LB-42908 (LG Life Sciences), Pharmaprojects No. 5063 (Genzyme), Pharmaprojects No. 5597 (Ipsen), Yissum Project No. B-1055 (Yissum), and an analogue or derivative thereof. 22824. The method of any one of items 22753-22785 wherein the composition comprises an anti-fibrotic agent, and wherein the anti-fibrotic agent is an FLT-3 kinase inhibitor.
22825. The method of any one of items 22753-22785 wherein the composition comprises an anti-fibrotic agent, and wherein the anti-fibrotic agent is an FGF receptor kinase inhibitor.
22826. The method of any one of items 22753-22785 wherein the composition comprises an anti-fibrotic agent, and wherein the anti-fibrotic agent is a fibrinogen antagonist selected from the group consisting of AUV-201 (Auvation), MG-13926 (Sanofi-Aventis), plasminogen activator (CAS No. 105913-11-9) (from Sanofi-Aventis or UCB), plasminogen activator-2 (tPA-2) (Sanofi-Aventis), pro-urokinase (CAS No. 82657-92-9) (Sanofi-Aventis), and an analogue or derivative thereof.
22827. The method of any one of items 22753-22785 wherein the composition comprises an anti-fibrotic agent, and wherein the anti-fibrotic agent is a heat shock protein 90 antagonist selected from the group consisting of SRN-005 (Sirenade), geldanamycin, NSC-33050 (17- allylaminogeldanamycin; 17-AAG), 17-dimethylaminoethylamino-17- demethoxy-geldanamycin (17-DMAG), rifabutin (rifamycin XIV, 1',4- didehydro-1-deoxy-1 ,4-dihydro-5'-(2-methylpropyl)-1-oxo-), and an analogue or derivative thereof.
22828. The method of any one of items 22753-22785 wherein the composition comprises an anti-fibrotic agent, and wherein the anti-fibrotic agent is a histone deacetylase inhibitor.
22829. The method of any one of items 22753-22785 wherein the composition comprises an anti-fibrotic agent, and wherein the anti-fibrotic agent is an HMGCoA reductase inhibitor selected from the group consisting of an atherosclerosis therapeutic from Lipid Sciences, ATI- 16000 (ARYx Therapeutics), KS-01-019 (Kos Pharmaceuticals), Pharmaprojects No. 2197 (Sanofi-Aventi), RP 61969 (Sanofi-Aventis), and an analogue or derivative thereof.
22830. The method of any one of items 22753-22785 wherein the composition comprises an anti-fibrotic agent, and wherein the anti-fibrotic agent is an ICAM inhibitor.
22831. The method of any one of items 22753-22785 wherein the composition comprises an anti-fibrotic agent, and wherein the anti-fibrotic agent is an IL, ICE and IRAK antagonist, wherein the composition comprises an anti-fibrotic agent, and wherein the antagonist is a CJ-14877, CP-424174 (Pfizer), NF-61 (Negma-Lerads), and an analogue or derivative thereof.
22832. The method of any one of items 22753-22785 wherein the composition comprises an anti-fibrotic agent, and wherein the anti-fibrotic agent is an IL-2 inhibitor.
22833. The method of any one of items 22753-22785 wherein the composition comprises an anti-fibrotic agent, and wherein the anti-fibrotic agent is an immunosuppressant selected from the group consisting of teriflunomide (Sanofi Aventis), chlorsulfaquinoxalone (NSC- 339004), chlorsulfaquinoxalone sulfate, CS-712 (Sankyo), ismomultin alfa (CAS No. 457913-93-8) (Akzo Nobel), antiallergics from GenPat77, antiinflammatories or AT-005 (Androclus Therapeutics), autoimmune disease therapeutics from EpiVax, BN-007 (Bone), budesonide (CAS No. 51333-22- 3) (MAP Pharmaceuticals), CO-14 (Genzyme), edratide (CAS No. 433922- 67-9) (Teva), EP-314 (Enanta), eprovafen (CAS No. 101335-99-3) (Sanofi- Aventis), HWA-131 (CAS No. 118788-41-3) (Sanofi-Aventis), immunomodulators from Merϋon Pharmaceuticals, immunosuppressives from Alchemia, IPL-12 (Inflazyme), MDL-9563 (CAS No. 27086-86-8) (Sanofi-Aventis), Pharmaprojects No. 2330 (Sanofi-Aventis), Pharmaprojects No. 6426 (Abgenix), PXS-25 (Pharmaxis), rosmarinic acid (CAS No. 20283-92-5) (Sanofi-Aventis), RP 42927 or RP 54745 (CAS No. 135330-08-4) (Sanofi-Aventis), SGN-35 (Seattle Genetics), ST-1959 (Sigma-Tau), type I diabetes therapy from SYNX Pharma, UNIL-88 (Debiopharm), VP-025 (Vasogen), VR-694 (Vectura), PRTX-001 (Protalex), and an analogue or derivative thereof.
22834. The method of any one of items 22753-22785 wherein the composition comprises an anti-fibrotic agent, and wherein the anti-fibrotic agent is an IMPDH (inosine monophosphate).
22835. The method of any one of items 22753-22785 wherein the composition comprises an anti-fibrotic agent, and wherein the anti-fibrotic agent is an integrin antagonist.
22836. The method of any one of items 22753-22785 wherein the composition comprises an anti-fibrotic agent, and wherein the anti-fibrotic agent is an interleukin antagonist.
22837. The method of any one of items 22753-22785 wherein the composition comprises an anti-fibrotic agent, and wherein the anti-fibrotic agent is an inhibitor of type III receptor tyrosine kinase.
22838. The method of any one of items 22753-22785 wherein the composition comprises an anti-fibrotic agent, and wherein the anti-fibrotic agent is an irreversible inhibitor of enzyme methionine aminopeptidase type 2. 22839. The method of any one of items 22753-22785 wherein the composition comprises an anti-fibrotic agent, and wherein the anti-fibrotic agent is an isozyme selective delta protein kinase C inhibitor.
22840. The method of any one of items 22753-22785 wherein the composition comprises an anti-fibrotic agent, and wherein the anti-fibrotic agent a JAK3 enzyme inhibitor.
22841. The method of any one of items 22753-22785 wherein the composition comprises an anti-fibrotic agent, and wherein the anti-fibrotic agent is a JNK inhibitor.
22842. The method of any one of items 22753-22785 wherein the composition comprises an anti-fibrotic agent, and wherein the anti-fibrotic agent is a kinase inhibitor.
22843. The method of any one of items 22753-22785 wherein the composition comprises an anti-fibrotic agent, and wherein the anti-fibrotic agent is kinesin antagonist.
22844. The method of any one of items 22753-22785 wherein the composition comprises an anti-fibrotic agent, and wherein the anti-fibrotic agent is a kinesin antagonist.
22845. The method of any one of items 22753-22785 wherein the composition comprises an anti-fibrotic agent, and wherein the anti-fibrotic agent is a leukotriene inhibitor and antagonist selected from the group consisting of ambicromil (CAS No. 58805-38-2) (Sanofi-Aventis), amelubant (CAS No. 346735-24-8) (Boehringer Ingelheim), DW-1141 (Dong Wha), ebselen (Daiichi Pharmaceutical), ibudilast (Kyorin), leucotriene inhibitors from Sanofi-Aventis, lymphotoxin-beta receptor (LT-D) from Biogen Idee, Pharmaprojects No. 1535 and 2728 (CAS No. 119340-33-9) (Sanofi-Aventis), R-112 (Rigel), Rev-5367 (CAS No. 92532-05-3) (Sanofi- Aventis), RG-14893 (CAS No. 141835-49-6) (Sanofi-Aventis), RG-5901-A (CAS No. 101910-24-1), 92532-23-5, RP 66153 (CAS No. 142422-79-5), RP 66364 (CAS No.186916-92-5), RP 69698 (CAS No. 141748-00-7) (Sanofi-Aventis), SC-411930 (Pfizer), SC-41930 (CAS No. 120072-59-5) (Pfizer), SC-50605 (CAS No. 138828-39-4) (Pfizer), SC-51146 (CAS No. 141059-52-1), SC-53228 (CAS No. 153633-01-3) (Pfizer), spaglumic acid (ZY-15106) (CAS No.3106-85-2), 80619-64-3 (Novartis), tipredane (CAS No. 85197-77-9) (Bristol-Myers Squibb), U-75302 (CAS No. 119477-85-9) (Pfizer), and analogue or derivative thereof.
22846. The method of any one of items 22753-22785 wherein the composition comprises an anti-fibrotic agent, and wherein the anti-fibrotic agent is an MAP kinase inhibitor.
22847. The method of any one of items 22753-22785 wherein the composition comprises an anti-fibrotic agent, and wherein the anti-fibrotic agent is a matrix metalloproteinase inhibitor.
22848. The method of any one of items 22753-22785 wherein the composition comprises an anti-fibrotic agent, and wherein the anti-fibrotic agent is an MCP-CCR2 inhibitor.
22849. The method of any one of items 22753-22785 wherein the composition comprises an anti-fibrotic agent, and wherein the anti-fibrotic agent is an mTOR inhibitor.
22850. The method of any one of items 22753-22785 wherein the composition comprises an anti-fibrotic agent, and wherein the anti-fibrotic agent is an mTOR kinase inhibitor. 22851. The method of any one of items 22753-22785 wherein the composition comprises an anti-fibrotic agent, and wherein the anti-fibrotic agent is a microtubule inhibitor selected from the group consisting of antibody-maytansinoid conjugates from Biogen Idee, colchicines (MantiCore Pharmaceuticals), anticancer immunoconjugates from Johnson & Johnson, DIME from Octamer, gni-1f (GNI), huC242-DM4, huMy9-6-DM1 (ImmunoGen), IDN-5404 (Indena), IMO-098, IMOderm (Imotep), mebendazole (Introgen Therapeutics), microtubule poisons from Cambridge Enterprise, paclitaxel such as LOTAX from Aphios (CAS No. 33069-62-4), Genexol-PM from Samyang, Pharmaprojects No. 6383 (Tapestry Pharmaceuticals), RPR-112378 (Sanofi-Aventis), SGN-75 (Seattle Genetics), SPL-7435 (Starpharma), SSR-250411 (Sanofi-Aventis), trastuzumab-DM1 (Genentech), vinorelbine, and an analogue or derivative thereof.
22852. - The method of any one of items 2-2753-22785 wherein the composition comprises an anti-fibrotic agent, and wherein the anti-fibrotic agent is an MIF inhibitor.
22853. The method of any one of items 22753-22785 wherein the composition comprises an anti-fibrotic agent, and wherein the anti-fibrotic agent is an MMP inhibitor.
22854. The method of any one of items 22753-22785 wherein the composition comprises an anti-fibrotic agent, and wherein the anti-fibrotic agent is a neurokinin (NK) antagonist selected from the group consisting of anthrotainin (CAS No. 148084-40-6) (Sanofi-Aventis), an IBS thereapeutic from ArQuIe, MDL-105212A (CAS No. 167261-60-1) (Ssanofi- Aventis), Pharmaprojects No. 2744, 3258 (CAS No. 139167-47-8) 4006, 4201, or 5986 (Sanofi-Aventis), RP 67580 (CAS No. 135911-02-3), SR- 144190 (CAS No. 201152-86-5), SSR-240600, SSR-241586 (Sanofi- Aventis), TKA-457 (Novartis), vestipitant mesylate (CAS No. 334476-64-1) (GlaxoSmithKline), Win-64821 (Sanofi-Aventis), PRX-96026 (Predix Pharmaceuticals), and an analogue or derivative thereof.
22855. The method of any one of items 22753-22785 wherein the composition comprises an anti-fibrotic agent, and wherein the anti-fibrotic agent is an NF kappa B inhibitor selected from the group consisting of AVE-0545 or AVE-0547 (Sanofi-Aventis), bortezomib (CAS No. 179324-69-7) (Millennium Pharmaceuticals), dexanabinol (CAS No. 112924- 45-5) (Pharmos), dexlipotam (Viatris), Pharmaprojects No. 6283 (INDRA) (OXiGENE)1 IPL-576092 (CAS No. 137571-30-3) (Inflazyme), NFKB decoy (Corgentech), NFKB decoy oligo (AnGes MG), NFKB's from Ariad, osteoporosis treatments or S5 (F005) from Fulcrum Pharmaceuticals, P61 (Phytopharm), R-flurbiprofen (CAS No. 5104-49-4) (Encore Pharmaceuticals), and an analogue or derivative thereof.
22856. The method of any one of items 22753-22785 wherein the composition comprises an anti-fibrotic agent, and wherein the anti-fibrotic agent is a nitric oxide agonist.
22857. The method of any one of items 22753-22785 wherein the composition comprises an anti-fibrotic agent, and wherein the anti-fibrotic agent is an ornithine decarboxylase inhibitor.
22858. The method of any one of items 22753-22785 wherein the composition comprises an anti-fibrotic agent, and wherein the anti-fibrotic agent is a p38 MAP kinase inhibitor selected from the group consisting of AZD-6703 (AstraZeneca), JX-401 (Jexys Pharmaceuticals), BMS-2 (Bristol-Myers Squibb), a p38 MAP kinase inhibitor from Novartis, a p38-alpha MAP kinase inhibitor from Amphora, Pharmaprojects No. 5704 (Pharmacopeia), RPR-200765A (Sanofi-Aventis), SD-282 (Johnson & Johnson), TAK-715 (Takeda), and an analogue or derivative thereof.
22859. The method of any one of items 22753-22785 wherein the composition comprises an anti-fibrotic agent, and wherein the anti-fibrotic agent is a palmitoyl-protein thioesterase inhibitor.
22860. The method of any one of items 22753-22785 wherein the composition comprises an anti-fibrotic agent, and wherein the anti-fibrotic agent is a PDGF receptor kinase inhibitor selected from the group consisting of AAL-993, AMN-107, or ABP-309 (Novartis), AMG-706 (Amgen), BAY-57-9352 (Bayer), CDP-860 (UCB), E-7080 (Eisai), imatinib (CAS No. 152459-95-5) (Novartis), OSI-930 (OSI Pharmaceuticals), RPR- 127963E (Sanofi-Aventis), RWJ-540973 (Johnson & Johnson), sorafenib tosylate (Bayer), SU-11657 (Pfizer), tandutinib (CAS No. 387867-13-2) (Millennium Pharmaceuticals), vatalanib (Novartis), ZK-CDK (Schering AG)1 and an analogue or derivative thereof.
22861. The method of any one of items 22753-22785 wherein the composition comprises an anti-fibrotic agent, and wherein the anti-fibrotic agent is (-)-halofenate (Metabolex), AMG-131 (Amgen), antidiabetics from Japan Tobacco, AZD-4619, AZD-8450, AZD-8677 (AstraZeneca), DRF-10945, balaglitazone (Dr Reddy's), CS-00088, CS- 00098 (Chipscreen Biosciences), E-3030 (Eisai), etalocib (CAS No. 161172- 51-6) (EIi Lilly), GSK-641597 (Ligand), GSK-677954 (GlaxoSmithKline), GW-409544 (Ligand), GW-590735 (GlaxoSmithKline), K-111 (Hoffmann-La Roche), LY-518674 (EIi Lilly), LY-674 (Ligand), LY-929 (Ligand), MC-3001, MC-3002 (MaxoCore Pharmaceuticals), metformin HCI + pioglitazone (CAS No. 1115-70-4 and 112529-15-4), ACTOPLUS MET from Andrx), muraglitazar (CAS No. 331741-94-7) (Bristol-Myers Squibb), naveglitazar (Ligand), oleoylethanolamide (Kadmus Pharmaceuticals), ONO-5129, pioglitazone hydrochloride (CAS No. 111025-46-8 and 112529-15-4) (Takeda), PLX-204 (Plexxikon), PPAR agonists from Genfit, PPAR delta agonists from EIi Lilly, PPAR-alpha agonists from CrystalGenomics, PPAR- gamma modulators and PPAR-β modulators from CareX, rosiglitazone maleate (CAS No. 122324-73-4 or 155141-29-0) (GlaxoSmithKline), rosiglitazone maleate/glimepir (CAS No. 155141-29-0 and 93479-97-1), AVANDARYL, rosiglitazone maleate/metformin extend (CAS No. 155141- 29-0 and 657-24-9), AVANDAMET, rosiglitazone maleate+metformin, AVANDAMET (GlaxoSmithKline), tesaglitazar (AstraZeneca), and an analogue or derivative thereof.
22862. The method of any one of items 22753-22785 wherein the composition comprises an anti-fibrotic agent, and wherein the anti-fibrotic agent is a phosphatase inhibitor.
22863. The method of any one of items 22753-22785 wherein the composition comprises an anti-fibrotic agent, and wherein the anti-fibrotic agent is a phosphodiesterase (PDE) inhibitor selected from the group consisting of avanafil (Tanabe Seiyaku), dasantafil (CAS No. 569351- 91-3) (Schering-Plough), A-906119 (CAS No. 134072-58-5), DL-850 (Sanofi-Aventis), GRC-3015, GRC-3566, GRC-3886 (Glenmark), HWA-153 (CAS No. 56395-66-5) (Sanofi-Aventis), hydroxypumafentrine (Altana), IBFB-130011, IBFB-14-016, IBFB-140301, IBFB-150007, IBFB-211913 (IBFB Pharma), L-826141 (Merck & Co), medorinone (CAS No. 88296-61-1) (Sanofi-Aventis), MEM-1917 (Memory Pharmaceuticals), ND-1251 (Neuro3d), PDE inhibitors from ICOS, PDE IV inhibitors from Memory Pharmaceuticals and CrystalGenomics, Pharmaprojects No. 2742 and 6141 (Sanofi-Aventis), QAD-171 (Novartis), RHC-2963 (CAS No. 76993-12-9 and 76993-14-1), RPR-117658, RPR-122818 derivatives, SR-24870 , and RPR- 132294 (Sanofi-Aventis), SK-350 (ln2Gen), stroke therapy agents from deCODE Genetics, TAS-203 (Taiho), tofimilast (CAS No. 185954-27-2) (Pfizer), UK-371800 (Pfizer), WIN-65579 (CAS No. 158020-82-7) (Sanofi- Aventis), IBFB-130020 (IBFB Pharma), OPC-6535 (CAS No. 145739-56-6) (Otsuka), a phosphodiesterase III inhibitor, enoximone, a phosphodiesterase IV inhibitor, fosfosal, Atopik (Barrier Therapeutics), triflusal, a phosphodiesterase V inhibitor, and an analogue or derivative thereof.
22864. The method of any one of items 22753-22785 wherein the composition comprises an anti-fibrotic agent, and wherein the anti-fibrotic agent is a PKC inhibitor.
22865. The method of any one of items 22753-22785 wherein the composition comprises an anti-fibrotic agent, and wherein the anti-fibrotic agent is a platelet activating factor antagonist.
22866. The method of any one of items 22753-22785 wherein the composition comprises an anti-fibrotic agent, and wherein the anti-fibrotic agent is a platelet-derived growth factor receptor kinase inhibitor.
22867. The method of any one of items 22753-22785 wherein the composition comprises an anti-fibrotic agent, and wherein the anti-fibrotic agent is a prolyl hydroxylase inhibitor.
22868. The method of any one of items 22753-22785 wherein the composition comprises an anti-fibrotic agent, and wherein the anti-fibrotic agent is a polymorphonuclear neutrophil inhibitor.
22869. The method of any one of items 22753-22785 wherein the composition comprises an anti-fibrotic agent, and wherein the anti-fibrotic agent is a protein kinase B inhibitor. 22870. The method of any one of items 22753-22785 wherein the composition comprises an anti-fibrotic agent, and wherein the anti-fibrotic agent is a protein kinase C stimulant.
22871. The method of any one of items 22753-22785 wherein the composition comprises an anti-fibrotic agent, and wherein the anti-fibrotic agent is a purine nucleoside analogue.
22872. The method of any one of items 22753-22785 wherein the composition comprises an anti-fibrotic agent, and wherein the anti-fibrotic agent is a purinoreceptor P2X antagonist.
22873. The method of any one of items 22753-22785 wherein the composition comprises an anti-fibrotic agent, and wherein the anti-fibrotic agent is a Raf kinase inhibitor.
22874. The method of any one of items 22753-22785 wherein the composition comprises an anti-fibrotic agent, and wherein the anti-fibrotic agent is a reversible inhibitor of ErbB1 and ErbB2.
22875. The method of any one of items 22753-22785 wherein the composition comprises an anti-fibrotic agent, and wherein the anti-fibrotic agent is a ribonucleoside triphosphate reductase inhibitor.
22876. The method of any one of items 22753-22785 wherein the composition comprises an anti-fibrotic agent, and wherein the anti-fibrotic agent is an SDF-1 antagonist.
22877. The method of any one of items 22753-22785 wherein the composition comprises an anti-fibrotic agent, and wherein the anti-fibrotic agent is a sheddase inhibitor. 22878. The method of any one of items 22753-22785 wherein the composition comprises an anti-fibrotic agent, and wherein the anti-fibrotic agent is an SRC inhibitor.
22879. The method of any one of items 22753-22785 wherein the composition comprises an anti-fibrotic agent, and wherein the anti-fibrotic agent is a stromelysin inhibitor.
22880. The method of any one of items 22753-22785 wherein the composition comprises an anti-fibrotic agent, and wherein the anti-fibrotic agent is an Syk kinase inhibitor.
22881. The method of any one of items 22753-22785 wherein the composition comprises an anti-fibrotic agent, and wherein the anti-fibrotic agent is a telomerase inhibitor.
22882. The method of any one of items 22753-22785 wherein the composition comprises an anti-fibrotic agent, and wherein the anti-fibrotic agent is a TGF beta inhibitor selected from the group consisting of pirfenidone (CAS No. 53179-13-8) (MARNAC), tranilast (CAS No. 53902- 12-8) (Kissei), IN-1130 (ln2Gen), mannose-6-phosphate (BTG), TGF-β antagonists from Inflazyme (Pharmaprojects No. 6075), TGF-β antagonists from Sydney, non-industrial source), TGF-β I receptor kinase inhibitors from EIi Lilly, TGF-β receptor inhibitors from Johnson & Johnson, and an analogue or derivative thereof.
22883. The method of any one of items 22753-22785 wherein the composition comprises an anti-fibrotic agent, and wherein the anti-fibrotic agent is a TNFα antagonist or TACE inhibitor selected from the group consisting of adalimumab (CAS No. 331731-18-1) (Cambridge Antibody Technology), AGIX-4207 (AtheroGenics), AGT-1 (Advanced Biotherapy), an anti-inflammatory from Borean Pharma, Cellzome, or Paradigm Therapeutics, anti-inflammatory vaccine (TNF-alpha kinoid) from Neovacs, humanized anti-TNF antibody or an anti-TNF MAb (CB0006) Celltech (UCB), apratastat (CAS No. 287445-51-0) (Wyeth), BMS-561392 (Bristol-Myers Squibb), BN-006 (Bone), certolizumab pegol (CAS No. 428863-50-7 or CH-138 (UCB), cilomilast (CAS No. 153259-65-5) (GlaxoSmithKline), CR-1 (Nuada Pharmaceuticals), CRx-119 (CombinatoRx), D-5410 (UCB), dacopafant (CAS No. 125372-33-0) (Sanofi- Aventis), dersalazine (CAS No. 188913-57-7/188913-58-8) (Uriach), etanercept (CAS No. 185243-69^0) (Amgen), ethyl pyruvate (Critical (Critical Therapeutics), golimumab (CAS No. 476181-74-5) (Johnson & Johnson), hormono-immunotherapy from Ipsen, CDP571 {e.g., Humicade from UCB), IC-485 (ICOS), infliximab (CAS No. 170277-31-3) (Johnson & Johnson), IP- 751 (Manhattan Pharmaceuticals), ISIS-104838 (CAS No. 250755-32-9) (ISIS Pharmaceuticals), lenalidomide (CAS No. 191732-72-6) (Celgene), lentinan (CAS No. 37339-90-5) (Ajinomoto), MDL-201112 (CAS No. 142130-73-2) (Sanofi-Aventis), medroxyprogesterone (CAS No. 520-85-4) (InKine Pharmaceutical), N-acetylcysteine (CAS No. 616-91-1) (Zambon), NBE-P2 (DIREVO Biotech), nerelimomab (CAS No. 162774-06-3) (Chiron), OM-294DP (OM PHARMA), onercept (CAS No. 199685-57-9) (Yeda), PASSTNF-alpha (Verigen), pentoxifylline or oxypentifylline (Sanofi-Aventis), Pharmaprojects No. 4091 , 4241 , 4295, or 4488 (Sanofi-Aventis), Pharmaprojects No. 5480 (Amgen), Pharmaprojects No. 6749 (Cengent), pirfenidone (CAS No. 53179-13-8) (MARNAC), RPR-132294 (Sanofi- Aventis), S5 (F002) (Fulcrum Pharmaceuticals), simvastatin (CAS No. 79902-63-9) (Merck & Co), STA-6292 (Synta Pharmaceuticals), tacrolimus (CAS No. 104987-11-3) (Fujisawa LifeCycle Pharma), talactoferrin alfa (CAS No. 308240-58-6) (Agennix), thalidomide (CAS No. 50-35-1) (Celgene), TNF antagonists form ProStrakan, and Synergen, TNF inhibitors (Amgen), TNF-alpha antagonists from Dynavax Technologies and Jerina AG (Germany), TNF-alpha inhibitors from IBFB Pharma and Xencor (Xencor), torbafylline (CAS No. 105102-21-4) (Sanofi-Aventis), UFM 505 (Uriach), VT-346 (Viron Therapeutics), YSIL6 (Y1S Therapeutics), YSTH2 (Y's Therapeutics), NPI-1302a-3 (Nereus Pharmaceuticals, a TNF antagonist from Jerina AG (Germany), dersalazine, and an analogue or derivative thereof.
22884. The method of any one of items 22753-22785 wherein the composition comprises an anti-fibrotic agent, and wherein the anti-fibrotic agent is a Toll receptor inhibitor.
22885. The method of any one of items 22753-22785 wherein the composition comprises an anti-fibrotic agent, and wherein the anti-fibrotic agent is a tubulin antagonist.
22886. The method of any one of items 22753-22785 wherein the composition comprises an anti-fibrotic agent, and wherein the anti-fibrotic agent is a tyrosine kinase inhibitor selected from the group consisting of SU-011248, SUTENT from Pfizer Inc. (New York, NY), BMS- 354825, PN-355 (Paracelsian Pharmaceuticals), AGN-199659 (Allergan), AAL-993 or ABP-309 (Novartis), adaphostin (NIH), AEE-788 (Novartis), AG- 013736 (OSI Pharmaceuticals), AG-13736 (Pfizer), ALT-110 (Alteris Therapeutics), AMG-706 (Amgen), anticancer MAbs from Xencor, anti- EGFrvlll MAbs from Abgenix, anti-HER2 MAb from Abiogen, AZD-2171 or AZD-9935 (AstraZeneca), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), CEP-5214 (Cephalon), CEP-7055 (Cephalon), cetuximab (ImCIone Systems), CHIR-200131 and CHIR-258 (Chiron), CP-547632 (OSI Pharmaceuticals), CP-724714 (Pfizer), CT-301 (Creabilis Therapeutics), D- 69491 (Baxter International), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EGFR/ErbB2 inhibitors from Array BioPharma, erlotinib (CAS No. 183319- 69-9) (OSI Pharmaceuticals), EXEL-2880 (Exelixis), FK-778 (Sanofi- Aventis) , gefitinib (CAS No. 184475-35-2) (AstraZeneca), GW-2286 or GW- 654652 (GlaxoSmithKline), her2/neu antigen from AlphaVax, HER-2/neu inhibitor from Generex, Herzyme (Medipad) (Sirna Therapeutics), HKI-272 (Wyeth), HuMax-EGFr (Genmab), idronoxil (CAS No. 81267-65-4) (Novogen), IGF-1 inhibitors from Ontogen, IMC-11 F8 (ImCIone Systems), kahalalide F (CAS No. 149204-42-2) (PharmaMar), KDR inhibitor from LG Life Sciences, KDR inhibitors from Abbott Laboratories, KDR kinase inhibitors (UCB), Kdr kinase inhibitors from Merck & Co, KRN-633 and KRN- 951 (Kirin Brewery), KSB- 102 (Xenova), lapatinib ditosylate (CAS No. 388082-78-8) (GlaxoSmithKline), matuzumab (Merck KGaA), MDX-214 (Medarex), ME-103 (Pharmexa), MED-A300 (Gerolymatos), MNAC-13 (Lay Line Genomics), nimotuzumab (Center of Molecular Immunology), NSC- 330507 or NSC-707545 (NIH)1 NV-50 (Novogen), OSI-930 (OSI Pharmaceuticals), panitumumab (Abgenix), pelitinib (CAS No. 287933-82-7) (Wyeth), pertuzumab (CAS No. 380610-27-5) (Genentech), Pharmaprojects No. 3985 (Sanofi-Aventis), prostate cancer therapeutics from Sequenom (SQPC35, SQPC36, SQPC90), removab and remoxab (Trion Pharma), RG- 13022 (CAS No. 136831-48-6), RG-13291 (CAS No. 138989-50-1), or RG- 14620 (CAS No. 136831-49-7) (Sanofi-Aventis), RM-6427 (Romark), RNAi breast cancer therapy from Benitec, RP 53801 (CAS No. 125882-88-4) (Sanofi-Aventis), sorafenib tosylate (Bayer), SU-11657 (Pfizer), Tie-2 antagonists from Semaia (Hybrigenics), Tie-2 inhibitors from Ontogen, trastuzumab (CAS No. 180288-69-1) (Genentech), tyrosine kinase inhibitors from Sanofi-Aventis, U3-1287, U3-1565, U3-1784, U3-1800 (U3 Pharma), vatalanib (Novartis), VEGFR-2 kinase inhibitor from Bristol-Myers Squibb, XL-647 (Exelixis), ZD-6474 (AstraZeneca), ZK-CDK (Schering AG), an EGFR tyrosine kinase inhibitor, EKB-569 (Wyeth), and an analogue or derivative thereof.
22887. The method of any one of items 22753-22785 wherein the composition comprises an anti-fibrotic agent, and wherein the anti-fibrotic agent is a VEGF inhibitor. 22888. The method of any one of items 22753-22785 wherein the composition comprises an anti-fibrotic agent, and wherein the anti-fibrotic agent is a vitamin D receptor agonist.
22889. The method of any one of items 22753-22785 wherein the composition comprises an anti-fibrotic agent, and wherein the anti-fibrotic agent is ZD-6474 (an angiogenesis inhibitor).
22890. The method of any one of items 22753-22785 wherein the composition comprises an anti-fibrotic agent, and wherein the anti-fibrotic agent is AP-23573 (an mTOR inhibitor).
22891. The method of any one of items 22753-22785 wherein the composition comprises an anti-fibrotic agent, and wherein the anti-fibrotic agent is synthadotin (a tubulin antagonist).
22892. The method of any one of items 22753-22785 wherein the composition comprises an anti-fibrotic agent, and wherein the anti-fibrotic agent is S-0885 (a collagenase inhibitor).
22893. The method of any one of items 22753-22785 wherein the composition comprises an anti-fibrotic agent, and wherein the anti-fibrotic agent is aplidine (an elongation factor-1 alpha inhibitor).
22894. The method of any one of items 22753-22785 wherein the composition comprises an anti-fibrotic agent, and wherein the anti-fibrotic agent is ixabepilone (an epithilone).
22895. The method of any one of items 22753-22785 wherein the composition comprises an anti-fibrotic agent, and wherein the anti-fibrotic agent is IDN-5390 (an angiogenesis inhibitor). 22896. The method of any one of items 22753-22785 wherein the composition comprises an anti-fibrotic agent, and wherein the anti-fibrotic agent is SB-2723005 (an angiogenesis inhibitor).
22897. The method of any one of items 22753-22785 wherein the composition comprises an anti-fibrotic agent, and wherein the anti-fibrotic agent is ABT-518 (an angiogenesis inhibitor).
22898. The method of any one of items 22753-22785 wherein the composition comprises an anti-fibrotic agent, and wherein the anti-fibrotic agent is combretastatin (an angiogenesis inhibitor).
22899. The method of any one of items 22753-22785 wherein the composition comprises an anti-fibrotic agent, and wherein the anti-fibrotic agent is anecortave acetate (an angiogenesis inhibitor).
22900. The method of any one of items 22753-22785 wherein the composition comprises an anti-fibrotic agent, and wherein the anti-fibrotic agent is SB-715992 (a kinesin antagonist).
22901. The method of any one of items 22753-22785 wherein the composition comprises an anti-fibrotic agent, and wherein the anti-fibrotic agent is temsirolimus (an mTOR inhibitor).
22902. The method of any one of items 22753-22785 wherein the composition comprises an anti-fibrotic agent, and wherein the anti-fibrotic agent is adalimumab (a TNFα antagonist).
22903. A method of any of items 22753-22785 wherein the composition is delivered in conjunction with the placement of a medical implant. 22904. A method of any of items 22753-22785 wherein the composition is delivered in conjunction with the placement of a medical implant, and the composition is placed on tissue adjacent to the medical implant.
22905. A method of any of items 22753-22785 wherein the composition is delivered in conjunction with the placement of a medical implant, and the composition is placed on the medical implant.
22906. A method of any of items 22753-22785 wherein the composition is delivered via an endoscope.
22907. A method of any of items 22753-22785 wherein the composition is delivered through a needle.
22908. A method of any of items 22753-22785 wherein the composition is delivered through a catheter.
22909. A method of any of items 22753-22785 wherein the composition is delivered at the time of a surgery.
22910. A method of any of items 22753-22785 wherein the composition is delivered using fluoroscopic guidance.
22911. The method of any one of items 22753-22785 wherein the composition comprises an anti-thrombotic agent.
22912. The method of any one of items 22753-22785 wherein the composition comprises a naturally occurring polymer. 22913. The method of any one of items 22753-22785 wherein the composition comprises protein.
22914. The method of any one of items 22753-22785 wherein the composition comprises carbohydrate.
22915. The method of any one of items 22753-22785 wherein the composition comprises biodegradable polymer.
22916. The method of any one of items 22753-22785 wherein the composition comprises nonbiodegradable polymer.
22917. The method of any one of items 22753-22785 wherein the composition comprises collagen.
22918. The method of any one of items 22753-22785 wherein the composition comprises methylated collagen.
22919. The method of any one of items 22753-22785 wherein the composition comprises fibrinogen.
22920. The method of any one of items 22753-22785 wherein the composition comprises thrombin.
22921. The method of any one of items 22753-22785 wherein the composition comprises blood plasma.
22922. The method of any one of items 22753-22785 wherein the composition comprises calcium salt. 22923. The method of any one of items 22753-22785 wherein the composition comprises an antifibronolytic agent.
22924. The method of any one of items 22753-22785 wherein the composition comprises a fibrinogen analog.
22925. The method of any one of items 22753-22785 wherein the composition comprises albumin.
22926. The method of any one of items 22753-22785 wherein the composition comprises plasminogen.
22927. The method of any one of items 22753-22785 wherein the composition comprises von Willebrands factor.
22928. The method of any one of items 22753-22785 wherein the composition comprises Factor VIII.
22929. The method of any one of items 22753-22785 wherein the composition comprises hypoallergenic collagen.
22930. The method of any one of items 22753-22785 wherein the composition comprises atelopeptidic collagen.
22931. The method of any one of items 22753-22785 wherein the composition comprises telopeptide collagen.
22932. The method of any one of items 22753-22785 wherein the composition comprises crosslinked collagen. 22933. The method of any one of items 22753-22785 wherein the composition comprises aprotinin.
22934. The method of any one of items 22753-22785 wherein the composition comprises epsilon-amino-n-caproic acid.
22935. The method of any one of items 22753-22785 wherein the composition comprises gelatin.
22936. The method of any one of items 22753-22785 wherein the composition comprises protein conjugates.
22937. The method of any one of items 22753-22785 wherein the composition comprises gelatin conjugates.
22938. The method of any one of items 22753-22785 wherein the composition comprises a synthetic polymer.
22939. The method of any one of items 22753-22785 wherein the composition comprises a synthetic isocyanate-containing compound.
22940. The method of any one of items 22753-22785 wherein the composition comprises a synthetic thiol-containing compound.
22941. The method of any one of items 22753-22785 wherein the composition comprises a synthetic compound containing at least two thiol groups. 22942. The method of any one of items 22753-22785 wherein the composition comprises a synthetic compound containing at least three thiol groups.
22943. The method of any one of items 22753-22785 wherein the composition comprises a synthetic compound containing at least four thiol groups.
22944. The method of any one of items 22753-22785 wherein the composition comprises a synthetic amino-containing compound.
22945. The method of any one of items 22753-22785 wherein the composition comprises a synthetic compound containing at least two amino groups.
22946. The method of any one of items 22753-22785 wherein the composition comprises a synthetic compound containing at least three amino groups.
22947. The method of any one of items 22753-22785 wherein the composition comprises a synthetic compound containing at least four amino groups.
22948. The method of any one of items 22753-22785 wherein the composition comprises a synthetic compound comprising a carbonyl-oxygen-succinimidyl group.
22949. The method of any one of items 22753-22785 wherein the composition comprises a synthetic compound comprising at least two carbonyl-oxygen-succinimidyl groups. 22950. The method of any one of items 22753-22785 wherein the composition comprises a synthetic compound comprising at least three carbonyl-oxygen-succinimidyl groups.
22951. The method of any one of items 22753-22785 wherein the composition comprises a synthetic compound comprising at least four carbonyl-oxygen-succinimidyl groups.
22952. The method of any one of items 22753-22785 wherein the composition comprises a synthetic polyalkylene oxide- containing compound.
22953. The method of any one of items 22753-22785 wherein the composition comprises a synthetic compound comprising both polyalkylene oxide and biodegradable polyester blocks.
22954. The method of any one of items 22753-22785 wherein the composition comprises a synthetic polyalkylene oxide- containing compound having reactive amino groups.
22955. The method of any one of items 22753-22785 wherein the composition comprises a synthetic polyalkylene oxide- containing compound having reactive thiol groups.
22956. The method of any one of items 22753-22785 wherein the composition comprises a synthetic polyalkylene oxide- containing compound having reactive carbonyl-oxygen-succinimidyl groups.
22957. The method of any one of items 22753-22785 wherein the composition comprises a synthetic compound comprising a biodegradable polyester block. 22958. The method of any one of items 22753-22785 wherein the composition comprises a synthetic polymer formed in whole or part from lactic acid or lactide.
22959. The method of any one of items 22753-22785 wherein the composition comprises a synthetic polymer formed in whole or part from glycolic acid or glycolide.
22960. The method of any one of items 22753-22785 wherein the composition comprises polylysine.
22961. The method of any one of items 22753-22785 wherein the composition comprises (a) protein and (b) a compound comprising a polyalkylene oxide portion.
- 22962. . The method of any one of items 22753-22785 wherein the composition comprises (a) protein and (b) polylysine.
22963. The method of any one of items 22753-22785 wherein the composition comprises (a) protein and (b) a compound having at least four thiol groups.
22964. The method of any one of items 22753-22785 wherein the composition comprises (a) protein and (b) a compound having at least four amino groups.
22965. The method of any one of items 22753-22785 wherein the composition comprises (a) protein and (b) a compound having at least four carbonyl-oxygen-succinimide groups. 22966. The method of any one of items 22753-22785 wherein the composition comprises (a) protein and (b) a compound having a biodegradable region formed from reactants selected from lactic acid, lactide, glycolic acid, glycolide, and epison-caprolactone.
22967. The method of any one of items 22753-22785 wherein the composition comprises (a) collagen and (b) a compound comprising a polyalkylene oxide portion.
22968. The method of any one of items 22753-22785 wherein the composition comprises (a) collagen and (b) polylysine.
22969. The method of any one of items 22753-22785 wherein the composition comprises (a) collagen and (b) a compound having at least four thiol groups.
22970. The method of any one of items 22753-22785 wherein the composition comprises (a) collagen and (b) a compound having at least four amino groups.
22971. The method of any one of items 22753-22785 wherein the composition comprises (a) collagen and (b) a compound having at least four carbonyl-oxygen-succinimide groups.
22972. The method of any one of items 22753-22785 wherein the composition comprises (a) collagen and (b) a compound having a biodegradable region formed from reactants selected from lactic acid, lactide, glycolic acid, glycolide, and epison-caprolactone. 22973. The method of any one of items 22753-22785 wherein the composition comprises (a) methylated collagen and (b) a compound comprising a polyalkylene oxide portion.
22974. The method of any one of items 22753-22785 wherein the composition comprises (a) methylated collagen and (b) polylysine.
22975. The method of any one of items 22753-22785 wherein the composition comprises (a) methylated collagen and (b) a compound having at least four thiol groups.
22976. The method of any one of items 22753-22785 wherein the composition comprises (a) methylated collagen and (b) a compound having at least four amino groups.
22977. The method of any one of items 22753-22785 wherein the composition comprises (a) methylated collagen and (b) a compound having at least four carbonyl-oxygen-succinimide groups.
22978. The method of any one of items 22753-22785 wherein the composition comprises (a) methylated collagen and (b) a compound having a biodegradable region formed from reactants selected from lactic acid, lactide, glycolic acid, glycolide, and epison-caprolactone.
22979. The method of any one of items 22753-22785 wherein the composition comprises hyaluronic acid.
22980. The method of any one of items 22753-22785 wherein the composition comprises a hyaluronic acid derivative. 22981. The method of any one of items 22753-22785 wherein the composition comprises pentaerythritol poly(ethylene glycol)ether tetra-sulfhydryl of number average molecular weight between 3,000 and 30,000.
22982. The method of any one of items 22753-22785 wherein the composition comprises pentaerythritol poly(ethylene glycol)ether tetra-amino of number average molecular weight between 3,000 and 30,000.
22983. The method of any one of items 22753-22785 wherein the composition comprises (a) a synthetic compound having a number average molecular weight between 3,000 and 30,000 and comprising a polyalkylene oxide region and multiple nucleophilic groups, and (b) a synthetic compound having a number average molecular weight between 3,000 and 30,000 and comprising a polyalkylene oxide region and multiple electrophilic groups.
22984. The method of any one of items 22753-22785 wherein the composition comprises a colorant.
22985. The method of any one of items 22753-22785 wherein the composition is sterile.
22986. A method for treatment of inflammatory arthritis, comprising delivering to a patient in need thereof (i) an anti-fibrotic agent, or (ii) a therapeutic composition comprising a) a polymer and/or a compound that forms a polymer in situ and b) an anti-fibrotic agent.
22987. A method for prevention of inflammatory arthritis, comprising delivering to a patient in need thereof (i) an anti-fibrotic agent, or (ii) a therapeutic composition comprising a) a polymer and/or a compound that forms a polymer in situ and b) an anti-fibrotic agent.
22988. A method for treatment of osteoarthritis, comprising delivering to a patient in need thereof (i) an anti-fibrotic agent, or (ii) a therapeutic composition comprising a) a polymer and/or a compound that forms a polymer in situ and b) an anti-fibrotic agent.
22989. A method for prevention of osteoarthritis, comprising delivering to a patient in need thereof (i) an anti-fibrotic agent, or (ii) a therapeutic composition comprising a) a polymer and/or a compound that forms a polymer in situ and b) an anti-fibrotic agent.
22990. A method for treatment of primary osteoarthritis, comprising delivering to a patient in need thereof (i) an anti-fibrotic agent, or (ii) a therapeutic composition comprising a) a polymer and/or a compound that forms a polymer in situ and b) an anti-fibrotic agent.
22991. A method for prevention of primary osteoarthritis, comprising delivering to a patient in need thereof (i) an anti-fibrotic agent, or (ii) a therapeutic composition comprising a) a polymer and/or a compound that forms a polymer in situ and b) an anti-fibrotic agent.
22992. A method for treatment of secondary osteoarthritis, comprising delivering to a patient in need thereof (i) an anti- fibrotic agent, or (ii) a therapeutic composition comprising a) a polymer and/or a compound that forms a polymer in situ and b) an anti-fibrotic agent.
22993. A method for prevention of secondary osteoarthritis, comprising delivering to a patient in need thereof (i) an anti- fibrotic agent, or (ii) a therapeutic composition comprising a) a polymer and/or a compound that forms a polymer in situ and b) an anti-fibrotic agent.
22994. A method for treatment of rheumatoid arthritis, comprising delivering to a patient in need thereof (i) an anti-fibrotic agent, or (ii) a therapeutic composition comprising a) a polymer and/or a compound that forms a polymer in situ and b) an anti-fibrotic agent.
22995. A method for prevention of rheumatoid arthritis, comprising delivering to a patient in need thereof (i) an anti-fibrotic agent, or (ii) a therapeutic composition comprising a) a polymer and/or a compound that forms a polymer in situ and b) an anti-fibrotic agent.
22996. The method of any one of items 22986-22995 wherein the composition is delivered intravenously.
22997. The method of any one of items 22986-22995 wherein the composition is delivered orally.
22998. The method of any one of items 22986-22995 wherein the composition is delivered by subcutaneous injection.
22999. The method of any one of items 22986-22995 wherein the composition is delivered by intramuscular injection.
23000. The method of any one of items 22986-22995 wherein the composition is delivered intra-articularly.
23001. The method of any one of items 22986-22995 wherein the agent is an adensosine A2A receptor antagonist. 23002. The method of any one of items 22986-22995 wherein the agent is an AKT inhibitor.
23003. The method of any one of items 22986-22995 wherein the agent is an alpha 2 integrin antagonist, wherein the alpha 2 integrin antagonist is Pharmaprojects No. 5754 (Merck KgaA).
23004. The method of any one of items 22986-22995 wherein the agent is an alpha 4 integrin antagonist.
23005. The method of any one of items 22986-22995 wherein the agent is an alpha 7 nicotinic receptor agonist.
23006. The method of any one of items 22986-22995 wherein the agent is an angiogenesis inhibitor selected from the group consisting of AG-12,958 (Pfizer), ATN-161 (Attention LLC), neovastat, an angiogenesis inhibitor from Jerina AG (Germany), NM-3 (Mercian), VGA- 1155 (Taisho), FCE-26644 (Pfizer), FCE-26950 (Pfizer), FPMA (Meiji Daries), FR-111142 (Fujisawa), GGTI-298, GM-1306 (Ligand), GPA-1734 (Novartis), NNC-47-0011 (Novo Nordisk), herbamycin (Nippon Kayaku), lenalidomide (Celegene), IP-10 (NIH), ABT-828 (Abbott), KIN-841 (Tokushima University, Japan), SF-1126 (Semafore Pharmaceuticals), laminin technology (NIH), CHIR-258 (Chiron), NVP-AEW541 (Novartis), NVP-AEW541 (Novartis), Vt16907 (Alchemia), OXI-8007 (Oxigene), EG- 3306 (Ark Therapeutics), Maspin (Arriva), ABT-567 (Abbott), PPl-2458 (Praecis Pharmaceuticals), CC-5079, CC-4089 (Celgene), HIF-1 alpha inhibitors (Xenova), S-247 (Pfizer), AP-23573 (Ariad), AZD-9935 (Astra Zeneca), mebendazole (Introgen Therapeutics), MetAP-2 inhibitors (GlaxoSmithKline), AG-615 (Angiogene Pharmaceuticals), Tie-2 antagonists (Hybrigenics), NC-381, CYC-381, NC-169, NC-219, NC-383, NC-384, NC- 407 (Lorus Therapeutics), ATN-224 (Attenuon), ON-01370 (Onconova), Vitronectin antagonists (Amgen), SDX-103 (Salmedix), Vitronectin antagonists (Shire), CHP (Riemser), TEK (Amgen), Anecortave acetate (Alcon), T46.2 (Matrix Therapeutics), HG-2 (Heptagen), TEM antagonists (Genzyme), Oxi-4500 (Oxigene), ATN-161 (Attenuon), WX-293 (Wilex), M- 2025 (Metris Therapeutics), Alphastatin (BioActa), YH-16 (Yantai Rongchang), BIBF-1120 (Boehringer Ingelheim), BAY-57-9352 (Bayer), AS- 1404 (Cancer Research Technology), SC-77964 (Pfizer), glycomimetics (BioTie Therapies), TIE-2 Inhibitors (Ontogen), DIMI, Octamer (Octamer), ABR-215050 (Active Biotech), ABT-518 (Abbott), KDR inhibitors (Abbott), BSF-466895 (Abbott), SCH-221153 (Schering-Plough), DAC.antiangiogenic (ConjuChem), TFPl (EntreMed), AZD-2171 (Astra-Zenaca), CDC-394 (Celgene), LY290293 (EIi Lilly), IDN-5390 (Indena), Kdr Kinase Inhibitors (Merck), CT-113020, CT-116433, CT-116563, CT-31890, CT-32228) (Cell Therapeutics), A-299620 (Abbott), TWEAK Inhibitor (Amgen), VEGF modulators (Johnson and Johnson), Tum-N53, tumstatin (Genzyme), Thios- 1, Thios-2 (Thios Pharmaceuticals), MV-6401 (Miravant Medical Technologies), Spisulosine (PharmaMar), CEP-7055 (Cephalon), AUV-201 (Auvation), LM-609 (EIi Lilly), SKF-106615 (AnorMED), Oglufanide disodium (Cytran), BW-114 (Pharminox), Calreticulin (NIH), WX-678 (Wilex), SD-7784 (Pfizer), WX-UK1 (Wilex), SH-268 (Schering AG), 2-Me-PGA (Celgene), S- 137 (Pfizer), ZD-6126 (Angiogene Pharmaceuticals), SG-292 (SignalGen), Benefin (Lane Labs), A6, A36 (Angstrom), SB-2723005 (GlaxoSmithKline), SC-7 (Cell Therapeutics), ZEN-014 (AEterna Zentaris), 2-methoxyestradiol (EntreMed), NK-130119 (Nippon Kayaku), CC-10004 (Celgene), AVE- 8062A (Ajinomoto), Tacedinaline (Pfizer), Actinonin (Tokyo Metropolitan Institute of Medical Science), Lenalidomide (Celgene), VGA-1155, BTO-956 (SRI International), ER-68203-00 (Eisai), CT-6685 (UCB), JKC-362 (Phoenix Pharmaceuticals), DMI-3798 (DMI Biosciences, Angiomate (Ipsen), ZD-6474 (AstraZeneca), CEP-5214 (Cephalon), Canstatin (Genzyme), NM-3 (Mercian), Oridigm (MediQuest Therapeutics), Exherin (Adherex), BLS-0597 (Boston Life Sciences), PTC-299 (PTC Therapeutics), NPI-2358 (Nereus Pharmaceuticals), CGP-79787 (Novartis), AEE-788 (Novartis), CKD-732 (Chong Kun Dang), CP-564959 (OSI Pharmaceuticals), CM-101 (CarboMed), CT-2584, CT3501 (Cell Therapeutics), combretastatin and analogues and derivatives thereof (Oxigene), Rebimastat (Bristol-Meyers Squibb), Dextrin 2-sulfate (ML Laboratories), Cilengitide (Merk KGaA), NSC-706704 (Pharminox), KRN- 951 (Kirin Brewery), Ukrain, NSC-631570 (Nowicky Pharma), Tecogalan sodium (Daiichi Pharmaceutical), Tz-93 (Tsumura), TBC-1635 (Encysive Pharmaceuticals), TAN-1120 (Takeda), Semaxanib (Pfizer), BDI-7800 (Biopharmacopae), SD-186, SD-983 (Bristol-Meyers Squibb), SB-223245 (GlaxoSmithKline), SC-236 (Pfizer), RWJ-590973 (Johnson and Johnson), ILX-1850 (Genzyme), SC-68488, S-836 (Pfizer), CG-55069-11 (CuraGen), Ki-23057 (Kirin Brewery), CCX-700 (Chemoentryx), Pegaptanib octasodium (Giled Sciences), ANGIOCOL (available from Biostratum Inc.), or an analogue or derivative thereof.
23007. The method of any one of items 22986-22995 wherein the agent is an apoptosis antagonist.
23008. The method of any one of items 22986-22995 wherein the agent is an apoptosis activator.
23009. The method of any one of items 22986-22995 wherein the agent is a beta 1 integrin antagonist.
23010. The method of any one of items 22986-22995 wherein the agent is a beta tubulin inhibitor.
23011. The method of any one of items 22986-22995 wherein the agent is a blocker of enzyme production in Hepatitis C. 23012. The method of any one of items 22986-22995 wherein the agent is a Bruton's tyrosine kinase inhibitor.
23013. The method of any one of items 22986-22995 wherein the agent is a calcineurin inhibitor.
23014. The method of any one of items 22986-22995 wherein the agent is a caspase 3 inhibitor.
23015. The method of any one of items 22986-22995 wherein the agent is a CC chemokine receptor antagonist.
23016. The method of any one of items 22986-22995 wherein the agent is a cell cycle inhibitor selected from the group consisting of SNS-595 (Sunesis), synthadotin, KRX-0403, and an analogue or derivative thereof.
23017. The method of any one of items 22986-22995 wherein the agent is a cathepsin B inhibitor.
23018. The method of any one of items 22986-22995 wherein the agent is a cathepsin K inhibitor, wherein the cathepsin K inhibitor is 462795 (GlaxoSmithKline), INPL-022-D6 (Amura Therapeutics), or an analogue or derivative thereof.
23019. The method of any one of items 22986-22995 wherein the agent is a cathepsin L inhibitor.
23020. The method of any one of items 22986-22995 wherein the agent is a CD40 antagonist. 23021. The method of any one of items 22986-22995 wherein the agent is a chemokine receptor agonist.
23022. The method of any one of items 22986-22995 wherein the agent is a chymase inhibitor.
23023. The method of any one of items 22986-22995 wherein the agent is a collagenase antagonist.
23024. The method of any one of items 22986-22995 wherein the agent is a CXCR antagonist.
23025. The method of any one of items 22986-22995 wherein the agent is a cyclin dependent kinase inhibitor selected from the group consisting of a CDK-1 inhibitor, a CDK-2 inhibitor, a CDK- 4 inhibitor, a CDK-6 inhibitor, a CAK1 inhibitor from GPC Biotech and Bristol-Myers - Squibb, RGB-286199 (GPC Biotech), an anticancer agent from Astex Technology, a CAK1 inhibitor from GPC Biotech, a CDK inhibitor from Sanofi-Aventis, a CDK1/CDK2 inhibitor from Amgen, a CDK2 inhibitor from SUGEN-2 (Pfizer), a hearing loss therapy agent (Sound Pharmaceuticals), PD-0332991 (Pfizer), RGB-286199 (GPC Biotech), Ro-0505124 (Hoffmann- La Roche), a Ser/Thr kinase inhibitor from Lilly (EIi Lilly), CVT-2584 (CAS No. 199986-75-9) (CV Therapeutics), and an analogue or derivative thereof.
23026. The method of any one of items 22986-22995 wherein the agent is a cyclooxygenase 1 inhibitor.
23027. The method of any one of items 22986-22995 wherein the agent is a DHFR inhibitor. 23028. The method of any one of items 22986-22995 wherein the agent is a dual integrin inhibitor.
23029. The method of any one of items 22986-22995 wherein the agent is an elastase inhibitor.
23030. The method of any one of items 22986-22995 wherein the agent is an elongation factor-1 alpha inhibitor.
23031. The method of any one of items 22986-22995 wherein the agent is an endothelial growth factor antagonist.
23032. The method of any one of items 22986-22995 wherein the agent is an endothelial growth factor receptor kinase inhibitor selected from the group consisting of sorafenib tosylate (Bayer), AAL-993 (Novartis), ABP-309 (Novartis), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EXEL- 2880 (Exelixis), GW-654652 (GlaxoSmithKline), a KDR inhibitor from LG Life Sciences, CT-6685 and CT-6729 (UCB), KRN-633 and KRN-951 (Kirin Brewery), OSI-930 (OSI Pharmaceuticals), SP-5.2 (Supratek Pharma), SU- 11657 (Pfizer), a Tie-2 antagonist (Hybrigenics), a VEGFR-2 kinase inhibitor (Bristol-Myers Squibb), XL-647 (Exelixis), a KDR inhibitor from Abbott Laboratories, sorafenib tosylate, and an analogue or derivative thereof.
23033. The method of any one of items 22986-22995 wherein the agent is an endotoxin antagonist.
23034. The method of any one of items 22986-22995 wherein the agent is an epothilone and tubulin binder. 23035. The method of any one of items 22986-22995 wherein the agent is an estrogen receptor antagonist.
23036. The method of any one of items 22986-22995 wherein the agent is an FGF inhibitor.
23037. The method of any one of items 22986-22995 wherein the agent is a farnexyl transferase inhibitor.
23038. The method of any one of items 22986-22995 wherein the agent is farnesyltransferase inhibitor selected from the group of A-197574 (Abbott), a farnesyltransferase inhibitor from Servier, FPTIII (Strathclyde Institute for Drug R)1 LB-42908 (LG Life Sciences), Pharmaprojects No. 5063 (Genzyme), Pharmaprojects No. 5597 (Ipsen), Yissum Project No. B-1055 (Yissum), and an analogue or derivative thereof.
23039. The method of any one of items 22986-22995 wherein the agent is an FLT-3 kinase inhibitor.
23040. The method of any one of items 22986-22995 wherein the agent is an FGF receptor kinase inhibitor.
23041. The method of any one of items 22986-22995 wherein the agent is a fibrinogen antagonist selected from the group consisting of AUV-201 (Auvation), MG-13926 (Sanofi-Aventis), plasminogen activator (CAS No. 105913-11-9) (from Sanofi-Aventis or UCB), plasminogen activator-2 (tPA-2) (Sanofi-Aventis), pro-urokinase (CAS No. 82657-92-9) (Sanofi-Aventis), and an analogue or derivative thereof.
23042. The method of any one of items 22986-22995 wherein the agent is a heat shock protein 90 antagonist selected from the group consisting of SRN-005 (Sirenade), geldanamycin, NSC-33050 (17- allylaminogeldanamycin; 17-AAG), 17-dimethylaminoethylamino-17- demethoxy-geldanamycin (17-DMAG), rifabutin (rifamycin XIV, 1',4- didehydro-1-deoxy-1 ,4-dihydro-5'-(2-methylpropyl)-1-oxo-), and an analogue or derivative thereof.
23043. The method of any one of items 22986-22995 wherein the agent is a histone deacetylase inhibitor.
23044. The method of any one of items 22986-22995 wherein the agent is an HMGCoA reductase inhibitor selected from the group consisting of an atherosclerosis therapeutic from Lipid Sciences, ATI- 16000 (ARYx Therapeutics), KS-01-019 (Kos Pharmaceuticals), Pharmaprojects No. 2197 (Sanofi-Aventi), RP 61969 (Sanofi-Aventis), and an analogue or derivative thereof.
23045. The method of any one of items 22986-22995 wherein the agent is an ICAM inhibitor.
23046. The method of any one of items 22986-22995 wherein the agent is an IL, ICE and IRAK antagonist, wherein the antagonist is a CJ-14877, CP-424174 (Pfizer), NF-61 (Negma-Lerads), and an analogue or derivative thereof.
23047. The method of any one of items 22986-22995 wherein the agent is an IL-2 inhibitor.
23048. The method of any one of items 22986-22995 wherein the agent is an immunosuppressant selected from the group consisting of teriflunomide (Sanofi Aventis), chlorsulfaquinoxalone (NSC- 339004), chlorsulfaquinoxalone sulfate, CS-712 (Sankyo), ismomultin alfa (CAS No. 457913-93-8) (Akzo Nobel), antiallergics from GenPat77, antiinflammatories or AT-005 (Androclus Therapeutics), autoimmune disease therapeutics from EpiVax, BN-007 (Bone), budesonide (CAS No. 51333-22- 3) (MAP Pharmaceuticals), CO-14 (Genzyme), edratide (CAS No. 433922- 67-9) (Teva), EP-314 (Enanta), eprovafen (CAS No. 101335-99-3) (Sanofi- Aventis), HWA-131 (CAS No. 118788-41-3) (Sanofi-Aventis), immunomodulators from MerLion Pharmaceuticals, immunosuppressives from Alchemia, IPL-12 (Inflazyme), MDL-9563 (CAS No. 27086-86-8) (Sanofi-Aventis), Pharmaprojects No. 2330 (Sanofi-Aventis), Pharmaprojects No. 6426 (Abgenix), PXS-25 (Pharmaxis), rosmarinic acid (CAS No. 20283-92-5) (Sanofi-Aventis), RP 42927 or RP 54745 (CAS No. 135330-08-4) (Sanofi-Aventis), SGN-35 (Seattle Genetics), ST-1959 (Sigma-Tau), type I diabetes therapy from SYNX Pharma, UNIL-88 (Debiopharm), VP-025 (Vasogen), VR-694 (Vectura), PRTX-001 (Protalex), and an analogue or derivative thereof.
23049. The method of any one of items 22986-22995 wherein the agent is an IMPDH (inosine monophosphate).
23050. The method of any one of items 22986-22995 wherein the agent is an integrin antagonist.
23051. The method of any one of items 22986-22995 wherein the agent is an interleukin antagonist.
23052. The method of any one of items 22986-22995 wherein the agent is an inhibitor of type III receptor tyrosine kinase.
23053. The method of any one of items 22986-22995 wherein the agent is an irreversible inhibitor of enzyme methionine aminopeptidase type 2. 23054. The method of any one of items 22986-22995 wherein the agent is an isozyme selective delta protein kinase C inhibitor.
23055. The method of any one of items 22986-22995 wherein the agent a JAK3 enzyme inhibitor.
23056. The method of any one of items 22986-22995 wherein the agent is a JNK inhibitor.
23057. The method of any one of items 22986-22995 wherein the agent is a kinase inhibitor.
23058. The method of any one of items 22986-22995 wherein the agent is kinesin antagonist.
23059. The method of any one of items 22986-22995 wherein the agent is a kinesin antagonist.
23060. The method of any one of items 22986-22995 wherein the agent is a leukotriene inhibitor and antagonist selected from the group consisting of ambicromil (CAS No. 58805-38-2) (Sanofi-Aventis), amelubant (CAS No. 346735-24-8) (Boehringer Ingelheim), DW-1141 (Dong Wha), ebselen (Daiichi Pharmaceutical), ibudilast (Kyorin), leucotriene inhibitors from Sanofi-Aventis, lymphotoxin-beta receptor (LT-D) from Biogen Idee, Pharmaprojects No. 1535 and 2728 (CAS No. 119340-33-9) (Sanofi-Aventis), R-112 (Rigel), Rev-5367 (CAS No. 92532-05-3) (Sanofi- Aventis), RG-14893 (CAS No. 141835-49-6) (Sanofi-Aventis), RG-5901-A (CAS No. 101910-24-1), 92532-23-5, RP 66153 (CAS No. 142422-79-5), RP 66364 (CAS No.186916-92-5), RP 69698 (CAS No. 141748-00-7) (Sanofi-Aventis), SC-411930 (Pfizer), SC-41930 (CAS No. 120072-59-5) (Pfizer), SC-50605 (CAS No. 138828-39-4) (Pfizer), SC-51146 (CAS No. 141059-52-1), SC-53228 (CAS No. 153633-01-3) (Pfizer), spaglumic acid (ZY-15106) (CAS No.3106-85-2), 80619-64-3 (Novartis), tipredane (CAS No. 85197-77-9) (Bristol-Myers Squibb), U-75302 (CAS No. 119477-85-9) (Pfizer), and analogue or derivative thereof.
23061. The method of any one of items 22986-22995 wherein the agent is an MAP kinase inhibitor.
23062. The method of any one of items 22986-22995 wherein the agent is a matrix metalloproteinase inhibitor.
23063. The method of any one of items 22986-22995 wherein the agent is an MCP-CCR2 inhibitor.
23064. The method of any one of items 22986-22995 wherein the agent is an mTOR inhibitor.
23065. The method of any one of items 22986-22995 wherein the agent is an mTOR kinase inhibitor.
23066. The method of any one of items 22986-22995 wherein the agent is a microtubule inhibitor selected from the group consisting of antibody-maytansinoid conjugates from Biogen Idee, colchicines (MantiCore Pharmaceuticals), anticancer immunoconjugates from Johnson & Johnson, DIME from Octamer, gni-1f (GNI), huC242-DM4, huMy9-6-DM1 (ImmunoGen), IDN-5404 (Indena), IMO-098, IMOderm (Imotep), mebendazole (Introgen Therapeutics), microtubule poisons from Cambridge Enterprise, paclitaxel such as LOTAX from Aphios (CAS No. 33069-62-4), Genexol-PM from Samyang, Pharmaprojects No. 6383 (Tapestry Pharmaceuticals), RPR-112378 (Sanofi-Aventis), SGN-75 (Seattle Genetics), SPL-7435 (Starpharma), SSR-250411 (Sanofi-Aventis), trastuzumab-DM1 (Genentech), vinorelbine, and an analogue or derivative thereof.
23067. The method of any one of items 22986-22995 wherein the agent is an MIF inhibitor.
23068. The method of any one of items 22986-22995 wherein the agent is an MMP inhibitor.
23069. The method of any one of items 22986-22995 wherein the agent is a neurokinin (NK) antagonist selected from the group consisting of anthrotainin (CAS No. 148084-40-6) (Sanofi-Aventis), an IBS thereapeutic from ArQuIe, MDL-105212A (CAS No. 167261-60-1) (Ssanofi- Aventis), Pharmaprojects No. 2744, 3258 (CAS No. 139167-47-8) 4006, 4201, or 5986 (Sanofi-Aventis), RP 67580 (CAS No. 135911-02-3), SR- 144190 (CAS No. 201152-86-5), SSR-240600, SSR-241586 (Sanofi- Aventis), TKA-457 (Novartis), vestipitant mesylate (CAS No. 334476-64-1) (GlaxoSmithKline), Win-64821 (Sanofi-Aventis), PRX-96026 (Predix Pharmaceuticals), and an analogue or derivative thereof.
23070. The method of any one of items 22986-22995 wherein the agent is an NF kappa B inhibitor selected from the group consisting of AVE-0545 or AVE-0547 (Sanofi-Aventis), bortezomib (CAS No. 179324-69-7) (Millennium Pharmaceuticals), dexanabinol (CAS No. 112924- 45-5) (Pharmos), dexlipotam (Viatris), Pharmaprojects No. 6283 (INDRA) (OXiGENE), IPL-576092 (CAS No. 137571-30-3) (Inflazyme), NFKB decoy (Corgentech), NFKB decoy oligo (AnGes MG), NFKB's from Ariad, osteoporosis treatments or S5 (F005) from Fulcrum Pharmaceuticals, P61 (Phytopharm), R-flurbiprofen (CAS No. 5104-49-4) (Encore Pharmaceuticals), and an analogue or derivative thereof. 23071. The method of any one of items 22986-22995 wherein the agent is a nitric oxide agonist.
23072. The method of any one of items 22986-22995 wherein the agent is an ornithine decarboxylase inhibitor.
23073. The method of any one of items 22986-22995 wherein the agent is a p38 MAP kinase inhibitor selected from the group consisting of AZD-6703 (AstraZeneca), JX-401 (Jexys Pharmaceuticals), BMS-2 (Bristol-Myers Squibb), a p38 MAP kinase inhibitor from Novartis, a p38-alpha MAP kinase inhibitor from Amphora, Pharmaprojects No. 5704 (Pharmacopeia), RPR-200765A (Sanofi-Aventis), SD-282 (Johnson & Johnson), TAK-715 (Takeda), and an analogue or derivative thereof.
23074. The method of any one of items 22986-22995 wherein the agent is a palmitoyl-protein thioesterase inhibitor.
23075. The method of any one of items 22986-22995 wherein the agent is a PDGF receptor kinase inhibitor selected from the group consisting of AAL-993, AMN-107, or ABP-309 (Novartis), AMG-706 (Amgen), BAY-57-9352 (Bayer), CDP-860 (UCB), E-7080 (Eisai), imatinib (CAS No. 152459-95-5) (Novartis), OSI-930 (OSI Pharmaceuticals), RPR- 127963E (Sanofi-Aventis), RWJ-540973 (Johnson & Johnson), sorafenib tosylate (Bayer), SU-11657 (Pfizer), tandutinib (CAS No. 387867-13-2) (Millennium Pharmaceuticals), vatalanib (Novartis), ZK-CDK (Schering AG), and an analogue or derivative thereof.
23076. The method of any one of items 22986-22995 wherein the agent is (-)-halofenate (Metabolex), AMG-131 (Amgen), antidiabetics from Japan Tobacco, AZD-4619, AZD-8450, AZD-8677 (AstraZeneca), DRF-10945, balaglitazone (Dr Reddy's), CS-00088, CS- 00098 (Chipscreen Biosciences), E-3030 (Eisai), etalocib (CAS No. 161172- 51-6) (EIi Lilly), GSK-641597 (Ligand), GSK-677954 (GlaxoSmithKline), GW-409544 (Ligand), GW-590735 (GlaxoSmithKline), K-111 (Hoffmann-La Roche), LY-518674 (EIi Lilly), LY-674 (Ligand), LY-929 (Ligand), MC-3001 , MC-3002 (MaxoCore Pharmaceuticals), metformin HCI + pioglitazone (CAS No. 1115-70-4 and 112529-15-4), ACTOPLUS MET from Andrx), muraglitazar (CAS No. 331741-94-7) (Bristol-Myers Squibb), naveglitazar (Ligand), oleoylethanolamide (Kadmus Pharmaceuticals), ONO-5129, pioglitazone hydrochloride (CAS No. 111025-46-8 and 112529-15-4) (Takeda), PLX-204 (Plexxikon), PPAR agonists from Genfit, PPAR delta agonists from EIi Lilly, PPAR-alpha agonists from CrystalGenomics, PPAR- gamma modulators and PPAR-β modulators from CareX, rosiglitazone maleate (CAS No. 122324-73-4 or 155141-29-0) (GlaxoSmithKline), rosiglitazone maleate/glimepir (CAS No. 155141-29-0 and 93479-97-1), AVANDARYL, rosiglitazone maleate/metformin extend (CAS No. 155141- 29-0 and 657-24-9), AVANDAMET, rosiglitazone maleate+metformin, AVANDAMET (GlaxoSmithKline), tesaglitazar (AstraZeneca), and an analogue or derivative thereof.
23077. The method of any one of items 22986-22995 wherein the agent is a phosphatase inhibitor.
23078. The method of any one of items 22986-22995 wherein the agent is a phosphodiesterase (PDE) inhibitor selected from the group consisting of avanafil (Tanabe Seiyaku), dasantafil (CAS No. 569351- 91-3) (Schering-Plough), A-906119 (CAS No. 134072-58-5); DL-850 (Sanofi-Aventis), GRC-3015, GRC-3566, GRC-3886 (Glenmark), HWA-153 (CAS No. 56395-66-5) (Sanofi-Aventis), hydroxypumafentrine (Altana), IBFB-130011 , IBFB-14-016, IBFB-140301, IBFB-150007, IBFB-211913 (IBFB Pharma), L-826141 (Merck & Co), medorinone (CAS No. 88296-61-1) (Sanofi-Aventis), MEM-1917 (Memory Pharmaceuticals), ND-1251 (Neuro3d), PDE inhibitors from ICOS, PDE IV inhibitors from Memory Pharmaceuticals and CrystalGenomics, Pharmaprojects No. 2742 and 6141 (Sanofi-Aventis), QAD-171 (Novartis), RHC-2963 (CAS No. 76993-12-9 and 76993-14-1), RPR-117658, RPR-122818 derivatives, SR-24870 , and RPR- 132294 (Sanofi-Aventis), SK-350 (ln2Gen), stroke therapy agents from deCODE Genetics, TAS-203 (Taiho), tofimilast (CAS No. 185954-27-2) (Pfizer), UK-371800 (Pfizer), WlN-65579 (CAS No. 158020-82-7) (Sanofi- Aventis), IBFB-130020 (IBFB Pharma), OPC-6535 (CAS No. 145739-56-6) (Otsuka), a phosphodiesterase III inhibitor, enoximone, a phosphodiesterase IV inhibitor, fosfosal, Atopik (Barrier Therapeutics), triflusal, a phosphodiesterase V inhibitor, and an analogue or derivative thereof.
23079. The method of any one of items 22986-22995 wherein the agent is a PKC inhibitor.
23080. The method of any one of items 22986-22995 wherein the agent is a platelet activating factor antagonist.
23081. The method of any one of items 22986-22995 wherein the agent is a platelet-derived growth factor receptor kinase inhibitor.
23082. The method of any one of items 22986-22995 wherein the agent is a prolyl hydroxylase inhibitor.
23083. The method of any one of items 22986-22995 wherein the agent is a polymorphonuclear neutrophil inhibitor.
23084. The method of any one of items 22986-22995 wherein the agent is a protein kinase B inhibitor. 23085. The method of any one of items 22986-22995 wherein the agent is a protein kinase C stimulant.
23086. The method of any one of items 22986-22995 wherein the agent is a purine nucleoside analogue.
23087. The method of any one of items 22986-22995 wherein the agent is a purinoreceptor P2X antagonist.
23088. The method of any one of items 22986-22995 wherein the agent is a Raf kinase inhibitor.
23089. The method of any one of items 22986-22995 wherein the agent is a reversible inhibitor of ErbB1 and ErbB2.
23090. The method of any one of items 22986-22995 wherein the agent is a ribonucleoside triphosphate reductase inhibitor.
23091. The method of any one of items 22986-22995 wherein the agent is an SDF-1 antagonist.
23092. The method of any one of items 22986-22995 wherein the agent is a sheddase inhibitor.
23093. The method of any one of items 22986-22995 wherein the agent is an SRC inhibitor.
23094. The method of any one of items 22986-22995 wherein the agent is a stromelysin inhibitor. 23095. The method of any one of items 22986-22995 wherein the agent is an Syk kinase inhibitor.
23096. The method of any one of items 22986-22995 wherein the agent is a telomerase inhibitor.
23097. The method of any one of items 22986-22995 wherein the agent is a TGF beta inhibitor selected from the group consisting of pirfenidone (CAS No. 53179-13-8) (MARNAC), tranilast (CAS No. 53902- 12-8) (Kissei), IN-1130 (ln2Gen), mannose-6-phosphate (BTG), TGF-β antagonists from Inflazyme (Pharmaprojects No. 6075), TGF-β antagonists from Sydney, non-industrial source), TGF-β I receptor kinase inhibitors from EIi Lilly, TGF-β receptor inhibitors from Johnson & Johnson, and an analogue or derivative thereof.
23098. The method of any one of items 22986-22995 wherein the agent is a TNFα antagonist or TACE inhibitor selected from the group consisting of adalimumab (CAS No. 331731-18-1) (Cambridge Antibody Technology), AGIX-4207 (AtheroGenics), AGT-1 (Advanced Biotherapy), an anti-inflammatory from Borean Pharma, Cellzome, or Paradigm Therapeutics, anti-inflammatory vaccine (TNF-alpha kinoid) from Neovacs, humanized anti-TNF antibody or an anti-TNF MAb (CB0006) Celltech (UCB), apratastat (CAS No. 287445-51-0) (Wyeth), BMS-561392 (Bristol-Myers Squibb), BN-006 (Bone), certolizumab pegol (CAS No. 428863-50-7 or CH-138 (UCB), cilomilast (CAS No. 153259-65-5) (GlaxoSmithKline), CR-1 (Nuada Pharmaceuticals), CRx-119 (CombinatoRx), D-5410 (UCB), dacopafant (CAS No. 125372-33-0) (Sanofi- Aventis), dersalazine (CAS No. 188913-57-7/188913-58-8) (Uriach), etanercept (CAS No. 185243-69-0) (Amgen), ethyl pyruvate (Critical (Critical Therapeutics), golimumab (CAS No.476181-74-5) (Johnson & Johnson), hormono-immunotherapy from lpsen, CDP571 (e.g., Humicade from UCB), IC-485 (ICOS), infliximab (CAS No. 170277-31-3) (Johnson & Johnson), IP- 751 (Manhattan Pharmaceuticals), ISIS-104838 (CAS No. 250755-32-9) (ISIS Pharmaceuticals), lenalidomide (CAS No. 191732-72-6) (Celgene), lentinan (CAS No. 37339-90-5) (Ajinomoto), MDL-201112 (CAS No. 142130-73-2) (Sanofi-Aventis), medroxyprogesterone (CAS No. 520-85-4) (InKine Pharmaceutical), N-acetylcysteine (CAS No. 616-91-1) (Zambon), NBE-P2 (DIREVO Biotech), nerelimomab (CAS No. 162774-06-3) (Chiron), OM-294DP (OM PHARMA), onercept (CAS No. 199685-57-9) (Yeda), PASSTNF-alpha (Verigen), pentoxifylline or oxypentifylline (Sanofi-Aventis), Pharmaprojects No. 4091 , 4241, 4295, or 4488 (Sanofi-Aventis), Pharmaprojects No. 5480 (Amgen), Pharmaprojects No. 6749 (Cengent), pirfenidone (CAS No. 53179-13-8) (MARNAC), RPR-132294 (Sanofi- Aventis), S5 (F002) (Fulcrum Pharmaceuticals), simvastatin (CAS No. 79902-63-9) (Merck & Co), STA-6292 (Synta Pharmaceuticals), tacrolimus (CAS No. 104987-11-3) (Fujisawa LifeCycle Pharma), talactoferrin alfa (CAS No. 308240-58-6) (Agennix), thalidomide (CAS No. 50-35-1) (Celgene), TNF antagonists form ProStrakan, and Synergen, TNF inhibitors (Amgen), TNF-alpha antagonists from Dynavax Technologies and Jerina AG (Germany), TNF-alpha inhibitors from IBFB Pharma and Xencor (Xencor), torbafylline (CAS No. 105102-21-4) (Sanofi-Aventis), UR-1505 (Uriach), VT-346 (Viron Therapeutics), YSIL6 (Y1S Therapeutics), YSTH2 (Y's Therapeutics), NPI-1302a-3 (Nereus Pharmaceuticals, a TNF antagonist from Jerina AG (Germany), dersalazine, and an analogue or derivative thereof.
23099. The method of any one of items 22986-22995 wherein the agent is a Toll receptor inhibitor.
23100. The method of any one of items 22986-22995 wherein the agent is a tubulin antagonist. 23101. The method of any one of items 22986-22995 wherein the agent is a tyrosine kinase inhibitor selected from the group consisting of SU-011248, SUTENT from Pfizer Inc. (New York, NY), BMS- 354825, PN-355 (Paracelsian Pharmaceuticals), AGN-199659 (Allergan), AAL-993 or ABP-309 (Novartis), adaphostin (NIH), AEE-788 (Novartis), AG- 013736 (OSI Pharmaceuticals), AG-13736 (Pfizer), ALT-110 (Alteris Therapeutics), AMG-706 (Amgen), anticancer MAbs from Xencor, anti- EGFrvlll MAbs from Abgenix, anti-HER2 MAb from Abiogen, AZD-2171 or AZD-9935 (AstraZeneca), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), CEP-5214 (Cephalon), CEP-7055 (Cephalon), cetuximab (ImCIone Systems), CHIR-200131 and CHIR-258 (Chiron), CP-547632 (OSI Pharmaceuticals), CP-724714 (Pfizer), CT-301 (Creabilis Therapeutics), D- 69491 (Baxter International), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EGFR/ErbB2 inhibitors from Array BioPharma, erlotinib (CAS No. 183319- 69-9) (OSI Pharmaceuticals), EXEL-2880 (Exelixis), FK-778 (Sanofi- Aventis) , gefitinib (CAS No. 184475-35-2) (AstraZeneca), GW-2286 or GW- 654652 (GlaxoSmithKline), her2/neu antigen from AlphaVax, HER-2/neu inhibitor from Generex, Herzyme (Medipad) (Sirna Therapeutics), HKI-272 (Wyeth), HuMax-EGFr (Genmab), idronoxil (CAS No. 81267-65-4) (Novogen), IGF-1 inhibitors from Ontogen, IMC-11F8 (ImCIone Systems), kahalalide F (CAS No. 149204-42-2) (PharmaMar), KDR inhibitor from LG Life Sciences, KDR inhibitors from Abbott Laboratories, KDR kinase inhibitors (UCB), Kdr kinase inhibitors from Merck & Co, KRN-633 and KRN- 951 (Kirin Brewery), KSB-102 (Xenova), lapatinib ditosylate (CAS No. 388082-78-8) (GlaxoSmithKline), matuzumab (Merck KGaA), MDX-214 (Medarex), ME-103 (Pharmexa), MED-A300 (Gerolymatos), MNAC-13 (Lay Line Genomics), nimotuzumab (Center of Molecular Immunology), NSC- 330507 or NSC-707545 (NIH), NV-50 (Novogen), OSI-930 (OSI Pharmaceuticals), panitumumab (Abgenix), pelitinib (CAS No. 287933-82-7) (Wyeth), pertuzumab (CAS No. 380610-27-5) (Genentech), Pharmaprojects No. 3985 (Sanofi-Aventis), prostate cancer therapeutics from Sequenom (SQPC35, SQPC36, SQPC90), removab and remoxab (Trion Pharma), RG- 13022 (CAS No. 136831-48-6), RG-13291 (CAS No. 138989-50-1), or RG- 14620 (CAS No. 136831-49-7) (Sanofi-Aventis), RM-6427 (Romark), RNAi breast cancer therapy from Benitec, RP 53801 (CAS No. 125882-88-4) (Sanofi-Aventis), sorafenib tosylate (Bayer), SU-11657 (Pfizer), Tie-2 antagonists from Semaia (Hybrigenics), Tie-2 inhibitors from Ontogen, trastuzumab (CAS No. 180288-69-1) (Genentech), tyrosine kinase inhibitors from Sanofi-Aventis, U3-1287, U3-1565, U3-1784, U3-1800 (U3 Pharma), vatalanib (Novartis), VEGFR-2 kinase inhibitor from Bristol-Myers Squibb, XL-647 (Exelixis), ZD-6474 (AstraZeneca), ZK-CDK (Schering AG), an EGFR tyrosine kinase inhibitor, EKB-569 (Wyeth), and an analogue or derivative thereof.
23102. The method of any one of items 22986-22995 wherein the agent is a VEGF inhibitor.
23103. The method of any one of items 22986-22995 wherein the agent is a vitamin D receptor agonist.
23104. The method of any one of items 22986-22995 wherein the agent is ZD-6474 (an angiσgenesis inhibitor).
23105. The method of any one of items 22986-22995 wherein the agent is AP-23573 (an mTOR inhibitor).
23106. The method of any one of items 22986-22995 wherein the agent is synthadotin (a tubulin antagonist).
23107. The method of any one of items 22986-22995 wherein the agent is S-0885 (a collagenase inhibitor). 23108. The method of any one of items 22986-22995 wherein the agent is aplidine (an elongation factor-1 alpha inhibitor).
23109. The method of any one of items 22986-22995 wherein the agent is ixabepilone (an epithilone).
23110. The method of any one of items 22986-22995 wherein the agent is IDN-5390 (an angiogenesis inhibitor).
23111. The method of any one of items 22986-22995 wherein the agent is SB-2723005 (an angiogenesis inhibitor).
23112. The method of any one of items 22986-22995 wherein the agent is ABT-518 (an angiogenesis inhibitor).
23113. The method of any one of items 22986-22995 wherein the agent is combretastatin (an angiogenesis inhibitor).
23114. The method of any one of items 22986-22995 wherein the agent is anecortave acetate (an angiogenesis inhibitor).
23115. The method of any one of items 22986-22995 wherein the agent is SB-715992 (a kinesin antagonist).
23116. The method of any one of items 22986-22995 wherein the agent is temsirolimus (an mTOR inhibitor).
23117. The method of any one of items 22986-22995 wherein the agent is adalimumab (a TNFα antagonist). 23118. The method of any one of items 22986-22995 wherein the composition comprises an anti-thrombotic agent.
23119. The method of any one of items 22986-22995 wherein the composition comprises a naturally occurring polymer.
23120. The method of any one of items 22986-22995 wherein the composition comprises protein.
23121. The method of any one of items 22986-22995 wherein the composition comprises carbohydrate.
23122. The method of any one of items 22986-22995 wherein the composition comprises biodegradable polymer.
23123. The method of any one of items 22986-22995 wherein the composition comprises nonbiodegradable polymer.
23124. The method of any one of items 22986-22995 wherein the composition comprises collagen.
23125. The method of any one of items 22986-22995 wherein the composition comprises methylated collagen.
23126. The method of any one of items 22986-22995 wherein the composition comprises fibrinogen.
23127. The method of any one of items 22986-22995 wherein the composition comprises thrombin. 23128. The method of any one of items 22986-22995 wherein the composition comprises blood plasma.
23129. The method of any one of items 22986-22995 wherein the composition comprises calcium salt.
23130. The method of any one of items 22986-22995 wherein the composition comprises an antifibronolytic agent.
23131. The method of any one of items 22986-22995 wherein the composition comprises fibrinogen analog.
23132. The method of any one of items 22986-22995 wherein the composition comprises albumin.
23133. The method of any one of items 22986-22995 wherein the composition comprises plasminogen.
23134. The method of any one of items 22986-22995 wherein the composition comprises von Willebrands factor.
23135. The method of any one of items 22986-22995 wherein the composition comprises Factor VIII.
23136. The method of any one of items 22986-22995 wherein the composition comprises hypoallergenic collagen.
23137. The method of any one of items 22986-22995 wherein the composition comprises atelopeptidic collagen. 23138. The method of any one of items 22986-22995 wherein the composition comprises telopeptide collagen.
23139. The method of any one of items 22986-22995 wherein the composition comprises crosslinked collagen.
23140. The method of any one of items 22986-22995 wherein the composition comprises aprotinin.
23141. The method of any one of items 22986-22995 wherein the composition comprises epsilon-amino-n-caproic acid.
23142. The method of any one of items 22986-22995 wherein the composition comprises gelatin.
23143. The method of any one of items 22986-22995 wherein the composition comprises protein conjugates.
23144. The method of any one of items 22986-22995 wherein the composition comprises gelatin conjugates.
23145. The method of any one of items 22986-22995 wherein the composition comprises a synthetic polymer.
23146. The method of any one of items 22986-22995 wherein the composition comprises a synthetic isocyanate-containing compound.
23147. The method of any one of items 22986-22995 wherein the composition comprises a synthetic thiol-containing compound. 23148. The method of any one of items 22986-22995 wherein the composition comprises a synthetic compound containing at least two thiol groups.
23149. The method of any one of items 22986-22995 wherein the composition comprises a synthetic compound containing at least three thiol groups.
23150. The method of any one of items 22986-22995 wherein the composition comprises a synthetic compound containing at least four thiol groups.
23151. The method of any one of items 22986-22995 wherein the composition comprises a synthetic amino-containing compound.
23152. The method of any one of items 22986-22995 wherein the composition comprises a synthetic compound containing at least two amino groups.
23153. The method of any one of items 22986-22995 wherein the composition comprises a synthetic compound containing at least three amino groups.
23154. The method of any one of items 22986-22995 wherein the composition comprises a synthetic compound containing at least four amino groups.
23155. The method of any one of items 22986-22995 wherein the composition comprises a synthetic compound comprising a carbonyl-oxygen-succinimidyl group. 23156. The method of any one of items 22986-22995 wherein the composition comprises a synthetic compound comprising at least two carbonyl-oxygen-succinimidyl groups.
23157. The method of any one of items 22986-22995 wherein the composition comprises a synthetic compound comprising at least three carbonyl-oxygen-succinimidyl groups.
23158. The method of any one of items 22986-22995 wherein the composition comprises a synthetic compound comprising at least four carbonyl-oxygen-succinimidyl groups.
23159. The method of any one of items 22986-22995 wherein the composition comprises a synthetic polyalkylene oxide- containing compound.
23160. The method of any one of items 22986-22995 wherein the composition comprises a synthetic compound comprising both polyalkylene oxide and biodegradable polyester blocks.
23161. The method of any one of items 22986-22995 wherein the composition comprises a synthetic polyalkylene oxide- containing compound having reactive amino groups.
23162. The method of any one of items 22986-22995 wherein the composition comprises a synthetic polyalkylene oxide- containing compound having reactive thiol groups.
23163. The method of any one of items 22986-22995 wherein the composition comprises a synthetic polyalkylene oxide- containing compound having reactive carbonyl-oxygen-succinimidyl groups. 23164. The method of any one of items 22986-22995 wherein the composition comprises a synthetic compound comprising a biodegradable polyester block.
23165. The method of any one of items 22986-22995 wherein the composition comprises a synthetic polymer formed in whole or part from lactic acid or lactide.
23166. The method of any one of items 22986-22995 wherein the composition comprises a synthetic polymer formed in whole or part from glycolic acid or glycolide.
23167. The method of any one of items 22986-22995 wherein the composition comprises polylysine.
23168. The method of any one of items 22986-22995 wherein the composition comprises (a) protein and (b) a compound comprising a polyalkylene oxide portion.
23169. The method of any one of items 22986-22995 wherein the composition comprises (a) protein and (b) polylysine.
23170. The method of any one of items 22986-22995 wherein the composition comprises (a) protein and (b) a compound having at least four thiol groups.
23171. The method of any one of items 22986-22995 wherein the composition comprises (a) protein and (b) a compound having at least four amino groups. 23172. The method of any one of items 22986-22995 wherein the composition comprises (a) protein and (b) a compound having at least four carbonyl-oxygen-succinimide groups.
23173. The method of any one of items 22986-22995 wherein the composition comprises (a) protein and (b) a compound having a biodegradable region formed from reactants selected from lactic acid, lactide, glycolic acid, glycolide, and epison-caprolactone.
23174. The method of any one of items 22986-22995 wherein the composition comprises (a) collagen and (b) a compound comprising a polyalkylene oxide portion.
23175. The method of any one of items 22986-22995 wherein the composition comprises (a) collagen and (b) polylysine.
23176. The method of any one of items 22986-22995 wherein the composition comprises (a) collagen and (b) a compound having at least four thiol groups.
23177. The method of any one of items 22986-22995 wherein the composition comprises (a) collagen and (b) a compound having at least four amino groups.
23178. The method of any one of items 22986-22995 wherein the composition comprises (a) collagen and (b) a compound having at least four carbonyl-oxygen-succinimide groups.
23179. The method of any one of items 22986-22995 wherein the composition comprises (a) collagen and (b) a compound having a biodegradable region formed from reactants selected from lactic acid, lactide, glycolic acid, glycolide, and epison-caprolactone.
23180. The method of any one of items 22986-22995 wherein the composition comprises (a) methylated collagen and (b) a compound comprising a polyalkylene oxide portion.
23181. The method of any one of items 22986-22995 wherein the composition comprises (a) methylated collagen and (b) polylysine.
23182. The method of any one of items 22986-22995 wherein the composition comprises (a) methylated collagen and (b) a compound having at least four thiol groups.
23183. The method of any one of items 22986-22995 wherein the composition comprises (a) methylated collagen and (b) a compound having at least four amino groups.
23184. The method of any one of items 22986-22995 wherein the composition comprises (a) methylated collagen and (b) a compound having at least four carbonyl-oxygen-succinimide groups.
23185. The method of any one of items 22986-22995 wherein the composition comprises (a) methylated collagen and (b) a compound having a biodegradable region formed from reactants selected from lactic acid, lactide, glycolic acid, glycolide, and epison-caprolactone.
23186. The method of any one of items 22986-22995 wherein the composition comprises hyaluronic acid. 23187. The method of any one of items 22986-22995 wherein the composition comprises a hyaluronic acid derivative.
23188. The method of any one of items 22986-22995 wherein the composition comprises pentaerythritol poly(ethylene glycol)ether tetra-sulfhydryl of number average molecular weight between 3,000 and 30,000.
23189. The method of any one of items 22986-22995 wherein the composition comprises pentaerythritol poly(ethylene glycol)ether tetra-amino of number average molecular weight between 3,000 and 30,000.
23190. The method of any one of items 22986-22995 wherein the composition comprises (a) a synthetic compound having a number average molecular weight between 3,000 and 30,000 and comprising a polyalkylene oxide region and multiple nucleophilic groups, and (b) a synthetic compound having a number average molecular weight between 3,000 and 30,000 and comprising a polyalkylene oxide region and multiple electrophilic groups.
23191. The method of any one of items 22986-22995 wherein the composition comprises a colorant.
23192. The method of any one of items 22986-22995 wherein the composition is sterile.
23193. A method for treating a hypertrophic scar in a patient in need thereof, comprising delivering to the patient a) an anti-fibrotic agent or b) a composition comprising i) an anti-fibrotic agent and ii) a polymer and/or a compound that forms a polymer in situ. 23194. A method for treating a keloid in a patient in need thereof, comprising delivering to the patient a) an anti-fibrotic agent or b) a composition comprising i) an anti-fibrotic agent and ii) a polymer and/or a compound that forms a polymer in situ.
23195. The method of item 23193 or item 23194 wherein the agent or composition is directly injected into the scar or keloid.
23196. The method of item 23193 or item 23194 wherein the agent or composition is topically applied to the scar or keloid.
23197. The method of item 23193 or item 23194 wherein the agent is an adensosine A2A receptor antagonist.
23198. The method of item 23193 or item 23194 wherein the agent is an AKT inhibitor.
23199. The method of item 23193 or item 23194 wherein the agent is an alpha 2 integrin antagonist, wherein the alpha 2 integrin antagonist is Pharmaprojects No. 5754 (Merck KgaA).
23200. The method of item 23193 or item 23194 wherein the agent is an alpha 4 integrin antagonist.
23201. The method of item 23193 or item 23194 wherein the agent is an alpha 7 nicotinic receptor agonist.
23202. The method of item 23193 or item 23194 wherein the agent is an angiogenesis inhibitor selected from the group consisting of AG-12,958 (Pfizer), ATN-161 (Attenuon LLC), neovastat, an angiogenesis inhibitor from Jerina AG (Germany), NM-3 (Mercian), VGA- 1155 (Taisho), FCE-26644 (Pfizer), FCE-26950 (Pfizer), FPMA (Meiji Daries), FR-111142 (Fujisawa), GGTI-298, GM-1306 (Ligand), GPA-1734 (Novartis), NNC-47-0011 (Novo Nordisk), herbamycin (Nippon Kayaku), lenalidomide (Celegene), IP-10 (NIH)1 ABT-828 (Abbott), KIN-841 (Tokushima University, Japan), SF-1126 (Semafore Pharmaceuticals), laminin technology (NIH), CHIR-258 (Chiron), NVP-AEW541 (Novartis), NVP-AEW541 (Novartis), Vt16907 (Alchemia), OXI-8007 (Oxigene), EG- 3306 (Ark Therapeutics), Maspin (Arriva), ABT-567 (Abbott), PPI-2458 (Praecis Pharmaceuticals), CC-5079, CC-4089 (Celgene), HIF-1 alpha inhibitors (Xenova), S-247 (Pfizer), AP-23573 (Ariad), AZD-9935 (Astra Zeneca), mebendazole (Introgen Therapeutics), MetAP-2 inhibitors (GlaxoSmithKline), AG-615 (Angiogene Pharmaceuticals), Tie-2 antagonists (Hybrigenics), NC-381, CYC-381, NC-169, NC-219, NC-383, NC-384, NC- 407 (Lorus Therapeutics), ATN-224 (Attenuon), ON-01370 (Onconova), Vitronectin antagonists (Amgen), SDX-103 (Salmedix), Vitronectin antagonists (Shire), CHP (Riemser), TEK (Amgen), Anecortave acetate (Alcon), T46.2 (Matrix Therapeutics), HG-2 (Heptagen), TEM antagonists (Genzyme), Oxi-4500 (Oxigene), ATN-161 (Attenuon), WX-293 (Wilex), M- 2025 (Metris Therapeutics), Alphastatin (BioActa), YH-16 (Yantai Rongchang), BIBF-1120 (Boehringer Ingelheim), BAY-57-9352 (Bayer), AS- 1404 (Cancer Research Technology), SC-77964 (Pfizer), glycomimetics (BioTie Therapies), TIE-2 Inhibitors (Ontogen), DIMI, Octamer (Octamer), ABR-215050 (Active Biotech), ABT-518 (Abbott), KDR inhibitors (Abbott), BSF-466895 (Abbott), SCH-221153 (Schering-Plough), DAC:antiangiogenic (ConjuChem), TFPI (EntreMed), AZD-2171 (Astra-Zenaca), CDC-394 (Celgene), LY290293 (EIi Lilly), IDN-5390 (Indena), Kdr Kinase Inhibitors (Merck), CT-113020, CT-116433, CT-116563, CT-31890, CT-32228) (Cell Therapeutics), A-299620 (Abbott), TWEAK Inhibitor (Amgen), VEGF modulators (Johnson and Johnson), Tum-N53, tumstatin (Genzyme), Thios- 1 , Thios-2 (Thios Pharmaceuticals), MV-6401 (Miravant Medical Technologies), Spisulosine (PharmaMar), CEP-7055 (Cephalon), AUV-201 (Auvation), LM-609 (EIi Lilly), SKF-106615 (AnorMED), Oglufanide disodium (Cytran), BW-114 (Pharminox), Calreticulin (NIH), WX-678 (Wilex), SD-7784 (Pfizer), WX-UK1 (Wilex), SH-268 (Schering AG), 2-Me-PGA (Celgene), S- 137 (Pfizer), ZD-6126 (Angiogene Pharmaceuticals), SG-292 (SignalGen), Benefin (Lane Labs), A6, A36 (Angstrom), SB-2723005 (GlaxoSmithKline), SC-7 (Cell Therapeutics), ZEN-014 (AEterna Zentaris), 2-methoxyestradiol (EntreMed), NK-130119 (Nippon Kayaku), CC-10004 (Celgene), AVE- 8062A (Ajinomoto), Tacedinaline (Pfizer), Actinonin (Tokyo Metropolitan Institute of Medical Science), Lenalidomide (Celgene), VGA-1155, BTO-956 (SRI International), ER-68203-00 (Eisai), CT-6685 (UCB), JKC-362 (Phoenix Pharmaceuticals), DMI-3798 (DMI Biosciences, Angiomate (Ipsen), ZD-6474 (AstraZeneca), CEP-5214 (Cephalon), Canstatin (Genzyme), NM-3 (Mercian), Oridigm (MediQuest Therapeutics), Exherin (Adherex), BLS-0597 (Boston Life Sciences), PTC-299 (PTC Therapeutics), NPI-2358 (Nereus Pharmaceuticals), CGP-79787 (Novartis), AEE-788 (Novartis), CKD-732 (Chong Kun Dang), CP-564959 (OSI Pharmaceuticals), CM-101 (CarboMed), CT-2584, CT3501 (Cell Therapeutics), combretastatin and analogues and derivatives thereof (Oxigene), Rebimastat (Bristol-Meyers Squibb), Dextrin 2-sulfate (ML Laboratories), Cilengitide (Merk KGaA), NSC-706704 (Pharminox), KRN- 951 (Kirin Brewery), Ukrain, NSC-631570 (Nowicky Pharma), Tecogalan sodium (Daiichi Pharmaceutical), Tz-93 (Tsumura), TBC-1635 (Encysive Pharmaceuticals), TAN-1120 (Takeda), Semaxanib (Pfizer), BDI-7800 (Biopharmacopae), SD-186, SD-983 (Bristol-Meyers Squibb), SB-223245 (GlaxoSmithKline), SC-236 (Pfizer), RWJ-590973 (Johnson and Johnson), ILX-1850 (Genzyme), SC-68488, S-836 (Pfizer), CG-55069-11 (CuraGen), Ki-23057 (Kirϊn Brewery), CCX-700 (Chemoentryx), Pegaptanib octasodium (Giled Sciences), ANGIOCOL (available from Biostratum Inc.), or an analogue or derivative thereof. 23203. The method of item 23193 or item 23194 wherein the agent is an apoptosis antagonist.
23204. The method of item 23193 or item 23194 wherein the agent is an apoptosis activator.
23205. The method of item 23193 or item 23194 wherein the agent is a beta 1 integrin antagonist.
23206. The method of item 23193 or item 23194 wherein the agent is a beta tubulin inhibitor.
23207. The method of item 23193 or item 23194 wherein the agent is a blocker of enzyme production in Hepatitis C.
23208. The method of item 23193 or item 23194 wherein the agent is a Bruton's tyrosine kinase inhibitor.
23209. The method of item 23193 or item 23194 wherein the agent is a calcineurin inhibitor.
23210. The method of item 23193 or item 23194 wherein the agent is a caspase 3 inhibitor.
23211. The method of item 23193 or item 23194 wherein the agent is a CC chemokine receptor antagonist.
23212. The method of item 23193 or item 23194 wherein the agent is a cell cycle inhibitor selected from the group consisting of SNS-595 (Sunesis), synthadotin, KRX-0403, and an analogue or derivative thereof. 23213. The method of item 23193 or item 23194 wherein the agent is a cathepsin B inhibitor.
23214. The method of item 23193 or item 23194 wherein the agent is a cathepsin K inhibitor, wherein the cathepsin K inhibitor is 462795 (GlaxoSmithKline), INPL-022-D6 (Amura Therapeutics), or an analogue or derivative thereof.
23215. The method of item 23193 or item 23194 wherein the agent is a cathepsin L inhibitor.
23216. The method of item 23193 or item 23194 wherein the agent is a CD40 antagonist.
23217. The method of item 23193 or item 23194 wherein the agent is a chemokine receptor agonist.
23218. The method of item 23193 or item 23194 wherein the agent is a chymase inhibitor.
23219. The method of item 23193 or item 23194 wherein the agent is a collagenase antagonist.
23220. The method of item 23193 or item 23194 wherein the agent is a CXCR antagonist.
23221. The method of item 23193 or item 23194 wherein the agent is a cyclin dependent kinase inhibitor selected from the group consisting of a CDK-1 inhibitor, a CDK-2 inhibitor, a CDK- 4 inhibitor, a CDK-6 inhibitor, a CAK1 inhibitor from GPC Biotech and Bristol-Myers Squibb, RGB-286199 (GPC Biotech), an anticancer agent from Astex Technology, a CAK1 inhibitor from GPC Biotech, a CDK inhibitor from Sanofi-Aventis, a CDK1/CDK2 inhibitor from Amgen, a CDK2 inhibitor from SUGEN-2 (Pfizer), a hearing loss therapy agent (Sound Pharmaceuticals), PD-0332991 (Pfizer), RGB-286199 (GPC Biotech), Ro-0505124 (Hoffmann- La Roche), a Ser/Thr kinase inhibitor from Lilly (EIi Lilly), CVT-2584 (CAS No. 199986-75-9) (CV Therapeutics), and an analogue or derivative thereof.
23222. The method of item 23193 or item 23194 wherein the agent is a cyclooxygenase 1 inhibitor.
23223. The method of item 23193 or item 23194 wherein the agent is a DHFR inhibitor.
23224. The method of item 23193 or item 23194 wherein the agent is a dual integrin inhibitor.
23225. The method of item 23193 or item 23194 wherein the agent is an elastase inhibitor.
23226. The method of item 23193 or item 23194 wherein the agent is an elongation factor-1 alpha inhibitor.
23227. The method of item 23193 or item 23194 wherein the agent is an endothelial growth factor antagonist.
23228. The method of item 23193 or item 23194 wherein the agent is an endothelial growth factor receptor kinase inhibitor selected from the group consisting of sorafenib tosylate (Bayer), AAL-993 (Novartis), ABP-309 (Novartis), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EXEL- 2880 (Exelixis), GW-654652 (GlaxoSmithKline), a KDR inhibitor from LG Life Sciences, CT-6685 and CT-6729 (UCB), KRN-633 and KRN-951 (Kirin Brewery), OSI-930 (OSI Pharmaceuticals), SP-5.2 (Supratek Pharma), SU- 11657 (Pfizer), a Tie-2 antagonist (Hybrigenics), a VEGFR-2 kinase inhibitor (Bristol-Myers Squibb), XL-647 (Exelixis), a KDR inhibitor from Abbott Laboratories, sorafenib tosylate, and an analogue or derivative thereof.
23229. The method of item 23193 or item 23194 wherein the agent is an endotoxin antagonist.
23230. The method of item 23193 or item 23194 wherein the agent is an epothilone and tubulin binder.
23231. The method of item 23193 or item 23194 wherein the agent is an estrogen receptor antagonist.
23232. The method of item 23193 or item 23194 wherein the agent is an FGF inhibitor.
23233. The method of item 23193 or item 23194 wherein the agent is a farnexyl transferase inhibitor.
23234. The method of item 23193 or item 23194 wherein the agent is farnesyltransferase inhibitor selected from the group of A-197574 (Abbott), a farnesyltransferase inhibitor from Servier, FPTIII (Strathclyde Institute for Drug R), LB-42908 (LG Life Sciences), Pharmaprojects No. 5063 (Genzyme), Pharmaprojects No. 5597 (Ipsen), Yissum Project No. B-1055 (Yissum), and an analogue or derivative thereof.
23235. The method of item 23193 or item 23194 wherein the agent is an FLT-3 kinase inhibitor. 23236. The method of item 23193 or item 23194 wherein the agent is an FGF receptor kinase inhibitor.
23237. The method of item 23193 or item 23194 wherein the agent is a fibrinogen antagonist selected from the group consisting of AUV-201 (Auvation), MG-13926 (Sanofi-Aventis), plasminogen activator (CAS No. 105913-11-9) (from Sanofi-Aventis or UCB), plasminogen activator-2 (tPA-2) (Sanofi-Aventis), pro-urokinase (CAS No. 82657-92-9) (Sanofi-Aventis), and an analogue or derivative thereof.
23238. The method of item 23193 or item 23194 wherein the agent is a heat shock protein 90 antagonist selected from the group consisting of SRN-005 (Sirenade), geldanamycin, NSC-33050 (17- allylaminogeldanamycin; 17-AAG), 17-dimethylaminoethylamino-17- demethoxy-geldanamycin (17-DMAG), rifabutin (rifamycin XIV, 1',4- didehydro-1-deoxy-1 ,4-dihydro-5'-(2-methylpropyl)-1-oxo-), and an analogue _ .. or derivative thereof.
23239. The method of item 23193 or item 23194 wherein the agent is a histone deacetylase inhibitor.
23240. The method of item 23193 or item 23194 wherein the agent is an HMGCoA reductase inhibitor selected from the group consisting of an atherosclerosis therapeutic from Lipid Sciences, ATI- 16000 (ARYx Therapeutics), KS-01-019 (Kos Pharmaceuticals), Pharmaprojects No. 2197 (Sanofi-Aventi), RP 61969 (Sanofi-Aventis), and an analogue or derivative thereof.
23241. The method of item 23193 or item 23194 wherein the agent is an ICAM inhibitor. 23242. The method of item 23193 or item 23194 wherein the agent is an IL, ICE and IRAK antagonist, wherein the antagonist is a CJ-14877, CP-424174 (Pfizer), NF-61 (Negma-Lerads), and an analogue or derivative thereof.
23243. The method of item 23193 or item 23194 wherein the agent is an IL-2 inhibitor.
23244. The method of item 23193 or item 23194 wherein the agent is an immunosuppressant selected from the group consisting of teriflunomide (Sanofi Aventis), chlorsulfaquinoxalone (NSC- 339004), chlorsulfaquinoxalone sulfate, CS-712 (Sankyo), ismomultin alfa (CAS No. 457913-93-8) (Akzo Nobel), antiallergics from GenPat77, antiinflammatories or AT-005 (Androclus Therapeutics), autoimmune disease therapeutics from EpiVax, BN-007 (Bone), budesonide (CAS No. 51333-22- 3) (MAP Pharmaceuticals), CO-14 (Genzyme), edratide (CAS No. 433922- 67-9) (Teva), EP-314 (Enanta), eprovafen (CAS No. 101335-99-3) (Sanofi- Aventis), HWA-131 (CAS No. 118788-41-3) (Sanofi-Aventis), immunomodulators from MerLion Pharmaceuticals, immunosuppressives from Alchemia, IPL-12 (Inflazyme), MDL-9563 (CAS No. 27086-86-8) (Sanofi-Aventis), Pharmaprojects No. 2330 (Sanofi-Aventis), Pharmaprojects No. 6426 (Abgenix), PXS-25 (Pharmaxis), rosmarinic acid (CAS No. 20283-92-5) (Sanofi-Aventis), RP 42927 or RP 54745 (CAS No. 135330-08-4) (Sanofi-Aventis), SGN-35 (Seattle Genetics), ST-1959 (Sigma-Tau), type I diabetes therapy from SYNX Pharma, UNIL-88 (Debiopharm), VP-025 (Vasogen), VR-694 (Vectura), PRTX-001 (Protalex), and an analogue or derivative thereof.
23245. The method of item 23193 or item 23194 wherein the agent is an IMPDH (inosine monophosphate). 23246. The method of item 23193 or item 23194 wherein the agent is an integrin antagonist.
23247. The method of item 23193 or item 23194 wherein the agent is an interleukin antagonist.
23248. The method of item 23193 or item 23194 wherein the agent is an inhibitor of type III receptor tyrosine kinase.
23249. The method of item 23193 or item 23194 wherein the agent is an irreversible inhibitor of enzyme methionine aminopeptidase type 2.
23250. The method of item 23193 or item 23194 wherein the agent is an isozyme selective delta protein kinase C inhibitor.
23251. The method of item 23193 or item 23194 wherein the agent a JAK3 enzyme inhibitor.
23252. The method of item 23193 or item 23194 wherein the agent is a JNK inhibitor.
23253. The method of item 23193 or item 23194 wherein the agent is a kinase inhibitor.
23254. The method of item 23193 or item 23194 wherein the agent is kinesin antagonist.
23255. The method of item 23193 or item 23194 wherein the agent is a kinesin antagonist. 23256. The method of item 23193 or item 23194 wherein the agent is a leukotriene inhibitor and antagonist selected from the group consisting of ambicromil (CAS No. 58805-38-2) (Sanofi-Aventis), amelubant (CAS No. 346735-24-8) (Boehringer Ingelheim), DW-1141 (Dong Wha), ebselen (Daiichi Pharmaceutical), ibudilast (Kyorin), leucotriene inhibitors from Sanofi-Aventis, lymphotoxin-beta receptor (LT-D) from Biogen Idee, Pharmaprojects No. 1535 and 2728 (CAS No. 119340-33-9) (Sanofi-Aventis), R-112 (Rigel), Rev-5367 (CAS No. 92532-05-3) (Sanofi- Aventis), RG-14893 (CAS No. 141835-49-6) (Sanofi-Aventis), RG-5901-A (CAS No. 101910-24-1), 92532-23-5, RP 66153 (CAS No. 142422-79-5), RP 66364 (CAS No.186916-92-5), RP 69698 (CAS No. 141748-00-7) (Sanofi-Aventis), SC-411930 (Pfizer), SC-41930 (CAS No. 120072-59-5) (Pfizer), SC-50605 (CAS No. 138828-39-4) (Pfizer), SC-51146 (CAS No. 141059-52-1), SC-53228 (CAS No. 153633-01-3) (Pfizer), spaglumic acid (ZY-15106) (CAS No.3106-85-2), 80619-64-3 (Novartis), tipredane (CAS No. 85197-77-9) (Bristol-Myers Squibb), U-75302 (CAS No. 119477-85-9) (Pfizer), and analogue or derivative thereof.
23257. The method of item 23193 or item 23194 wherein the agent is an MAP kinase inhibitor.
23258. The method of item 23193 or item 23194 wherein the agent is a matrix metalloproteinase inhibitor.
23259. The method of item 23193 or item 23194 wherein the agent is an MCP-CCR2 inhibitor.
23260. The method of item 23193 or item 23194 wherein the agent is an mTOR inhibitor. 23261. The method of item 23193 or item 23194 wherein the agent is an mTOR kinase inhibitor.
23262. The method of item 23193 or item 23194 wherein the agent is a microtubule inhibitor selected from the group consisting of antibody-maytansinoid conjugates from Biogen Idee, colchicines (MantiCore Pharmaceuticals), anticancer immunoconjugates from Johnson & Johnson, DIME from Octamer, gni-1f (GNI), huC242-DM4, huMy9-6-DM1 (ImmunoGen), IDN-5404 (Indena), IMO-098, IMOderm (Imotep), mebendazole (Introgen Therapeutics), microtubule poisons from Cambridge Enterprise, paclitaxel such as LOTAX from Aphios (CAS No. 33069-62-4), Genexol-PM from Samyang, Pharmaprojects No. 6383 (Tapestry Pharmaceuticals), RPR-112378 (Sanofi-Aventis), SGN-75 (Seattle Genetics), SPL-7435 (Starpharma), SSR-250411 (Sanofi-Aventis), trastuzumab-DM1 (Genentech), vinorelbine, and an analogue or derivative thereof.
23263. The method of item 23193 or item 23194 wherein the agent is an MIF inhibitor.
23264. The method of item 23193 or item 23194 wherein the agent is an MMP inhibitor.
23265. The method of item 23193 or item 23194 wherein the agent is a neurokinin (NK) antagonist selected from the group consisting of anthrotainin (CAS No. 148084-40-6) (Sanofi-Aventis), an IBS thereapeutic from ArQuIe, MDL-105212A (CAS No. 167261-60-1) (Ssanofi- Aventis), Pharmaprojects No. 2744, 3258 (CAS No. 139167-47-8) 4006, 4201 , or 5986 (Sanofi-Aventis), RP 67580 (CAS No. 135911-02-3), SR- 144190 (CAS No. 201152-86-5), SSR-240600, SSR-241586 (Sanofi- Aventis), TKA-457 (Novartis), vestipitant mesylate (CAS No. 334476-64-1) (GlaxoSmithKline), Win-64821 (Sanofi-Aventis), PRX-96026 (Predix Pharmaceuticals), and an analogue or derivative thereof.
23266. The method of item 23193 or item 23194 wherein the agent is an NF kappa B inhibitor selected from the group consisting of AVE-0545 or AVE-0547 (Sanofi-Aventis), bortezomib (CAS No. 179324-69-7) (Millennium Pharmaceuticals), dexanabinol (CAS No. 112924- 45-5) (Pharmos), dexlipotam (Viatris), Pharmaprojects No. 6283 (INDRA) (OXiGENE), IPL-576092 (CAS No. 137571-30-3) (Inflazyme), NFKB decoy (Corgentech), NFKB decoy oligo (AnGes MG), NFKB's from Ariad, osteoporosis treatments or S5 (F005) from Fulcrum Pharmaceuticals, P61 (Phytopharm), R-flurbiprofen (CAS No. 5104-49-4) (Encore Pharmaceuticals), and an analogue or derivative thereof.
23267. The method of item 23193 or item 23194 wherein the agent is a nitric oxide agonist.
23268. The method of item 23193 or item 23194 wherein the agent is an ornithine decarboxylase inhibitor.
23269. The method of item 23193 or item 23194 wherein the agent is a p38 MAP kinase inhibitor selected from the group consisting of AZD-6703 (AstraZeneca), JX-401 (Jexys Pharmaceuticals), BMS-2 (Bristol-Myers Squibb), a p38 MAP kinase inhibitor from Novartis, a p38-alpha MAP kinase inhibitor from Amphora, Pharmaprojects No. 5704 (Pharmacopeia), RPR-200765A (Sanofi-Aventis), SD-282 (Johnson & Johnson), TAK-715 (Takeda), and an analogue or derivative thereof.
23270. The method of item 23193 or item 23194 wherein the agent is a palmitoyl-protein thioesterase inhibitor. 23271. The method of item 23193 or item 23194 wherein the agent is a PDGF receptor kinase inhibitor selected from the group consisting of AAL-993, AMN-107, or ABP-309 (Novartis), AMG-706 (Amgen), BAY-57-9352 (Bayer), CDP-860 (UCB), E-7080 (Eisai), imatinib (CAS No. 152459-95-5) (Novartis), OSI-930 (OSI Pharmaceuticals), RPR- 127963E (Sanofi-Aventis), RWJ-540973 (Johnson & Johnson), sorafenib tosylate (Bayer), SU-11657 (Pfizer), tandutinib (CAS No. 387867-13-2) (Millennium Pharmaceuticals), vatalanib (Novartis), ZK-CDK (Schering AG)1 and an analogue or derivative thereof.
23272. The method of item 23193 or item 23194 wherein the agent is (-)-halofenate (Metabolex), AMG-131 (Amgen), antidiabetics from Japan Tobacco, AZD-4619, AZD-8450, AZD-8677 (AstraZeneca), DRF-10945, balaglitazone (Dr Reddy's), CS-00088, CS- 00098 (Chipscreen Biosciences), E-3030 (Eisai), etalocib (CAS No. 161172- 51-6) (EIi Lilly), GSK-641597 (Ligand), GSK-677954 (GlaxoSmithKline), GW-409544 (Ligand), GW-590735 (GlaxoSmithKline), K-111 (Hoffmann-La Roche), LY-518674 (EIi Lilly), LY-674 (Ligand), LY-929 (Ligand), MC-3001, MC-3002 (MaxoCore Pharmaceuticals), metformin HCI + pioglitazone (CAS No. 1115-70-4 and 112529-15-4), ACTOPLUS MET from Andrx), muraglitazar (CAS No. 331741-94-7) (Bristol-Myers Squibb), naveglitazar (Ligand), oleoylethanolamide (Kadmus Pharmaceuticals), ONO-5129, pioglitazone hydrochloride (CAS No. 111025-46-8 and 112529-15-4) (Takeda), PLX-204 (Plexxikon), PPAR agonists from Genfit, PPAR delta agonists from EIi Lilly, PPAR-alpha agonists from CrystalGenomics, PPAR- gamma modulators and PPAR-β modulators from CareX, rosiglitazone maleate (CAS No. 122324-73-4 or 155141-29-0) (GlaxoSmithKline), rosiglitazone maleate/glimepir (CAS No. 155141-29-0 and 93479-97-1), AVANDARYL, rosiglitazone maleate/metformin extend (CAS No. 155141- 29-0 and 657-24-9), AVANDAMET, rosiglitazone maleate+metformin, AVANDAMET (GlaxoSmithKline), tesaglitazar (AstraZeneca), and an analogue or derivative thereof.
23273. The method of item 23193 or item 23194 wherein the agent is a phosphatase inhibitor.
23274. The method of item 23193 or item 23194 wherein the agent is a phosphodiesterase (PDE) inhibitor selected from the group consisting of avanafil (Tanabe Seiyaku), dasantafil (CAS No. 569351- 91-3) (Schering-Plough), A-906119 (CAS No. 134072-58-5), DL-850 (Sanofi-Aventis), GRC-3015, GRC-3566, GRC-3886 (Glenmark), HWA-153 (CAS No. 56395-66-5) (Sanofi-Aventis), hydroxypumafentrine (Altana), IBFB-130011 , IBFB-14-016, IBFB-140301 , IBFB-150007, IBFB-211913 (IBFB Pharma), L-826141 (Merck & Co), medorinone (CAS No. 88296-61-1) (Sanofi-Aventis), MEM-1917 (Memory Pharmaceuticals), ND-1251 (Neuro3d), PDE inhibitors from ICOS, PDE IV inhibitors from Memory Pharmaceuticals and CrystalGenomics, Pharmaprojects No. 2742 and 6141 (Sanofi-Aventis), QAD-171 (Novartis), RHC-2963 (CAS No. 76993-12-9 and 76993-14-1), RPR-117658, RPR-122818 derivatives, SR-24870 , and RPR- 132294 (Sanofi-Aventis), SK-350 (ln2Gen), stroke therapy agents from deCODE Genetics, TAS-203 (Taiho), tofimilast (CAS No. 185954-27-2) (Pfizer), UK-371800 (Pfizer), WIN-65579 (CAS No. 158020-82-7) (Sanofi- Aventis), IBFB-130020 (IBFB Pharma), OPC-6535 (CAS No. 145739-56-6) (Otsuka), a phosphodiesterase III inhibitor, enoximone, a phosphodiesterase IV inhibitor, fosfosal, Atopik (Barrier Therapeutics), triflusal, a phosphodiesterase V inhibitor, and an analogue or derivative thereof.
23275. The method of item 23193 or item 23194 wherein the agent is a PKC inhibitor. 23276. The method of item 23193 or item 23194 wherein the agent is a platelet activating factor antagonist.
23277. The method of item 23193 or item 23194 wherein the agent is a platelet-derived growth factor receptor kinase inhibitor.
23278. The method of item 23193 or item 23194 wherein the agent is a prolyl hydroxylase inhibitor.
23279. The method of item 23193 or item 23194 wherein the agent is a polymorphonuclear neutrophil inhibitor.
23280. The method of item 23193 or item 23194 wherein the agent is a protein kinase B inhibitor.
23281. The method of item 23193 or item 23194 wherein the agent is a protein kinase C stimulant.
23282. The method of item 23193 or item 23194 wherein the agent is a purine nucleoside analogue.
23283. The method of item 23193 or item 23194 wherein the agent is a purinoreceptor P2X antagonist.
23284. The method of item 23193 or item 23194 wherein the agent is a Raf kinase inhibitor.
23285. The method of item 23193 or item 23194 wherein the agent is a reversible inhibitor of ErbB1 and ErbB2. 23286. The method of item 23193 or item 23194 wherein the agent is a ribonucleoside triphosphate reductase inhibitor.
23287. The method of item 23193 or item 23194 wherein the agent is an SDF-1 antagonist.
23288. The method of item 23193 or item 23194 wherein the agent is a sheddase inhibitor.
23289. The method of item 23193 or item 23194 wherein the agent is an SRC inhibitor.
23290. The method of item 23193 or item 23194 wherein the agent is a stromelysin inhibitor.
23291. The method of item 23193 or item 23194 wherein the agent is an Syk kinase inhibitor.
23292. The method of item 23193 or item 23194 wherein the agent is a telomerase inhibitor.
23293. The method of item 23193 or item 23194 wherein the agent is a TGF beta inhibitor selected from the group consisting of pirfenidone (CAS No. 53179-13-8) (MARNAC), tranilast (CAS No. 53902- 12-8) (Kissei), IN-1130 (ln2Gen), mannose-6-phosphate (BTG), TGF-β antagonists from Inflazyme (Pharmaprojects No. 6075), TGF-β antagonists from Sydney, non-industrial source), TGF-β I receptor kinase inhibitors from EIi Lilly, TGF-β receptor inhibitors from Johnson & Johnson, and an analogue or derivative thereof. 23294. The method of item 23193 or item 23194 wherein the agent is a TNFα antagonist or TACE inhibitor selected from the group consisting of adaiimumab (CAS No. 331731-18-1) (Cambridge Antibody Technology), AGIX-4207 (AtheroGenics), AGT-1 (Advanced Biotherapy), an anti-inflammatory from Borean Pharma, Cellzome, or Paradigm Therapeutics, anti-inflammatory vaccine (TNF-alpha kinoid) from Neovacs, humanized anti-TNF antibody or an anti-TNF MAb (CB0006) Celltech (UCB), apratastat (CAS No. 287445-51-0) (Wyeth), BMS-561392 (Bristol-Myers Squibb), BN-006 (Bone), certolizumab pegol (CAS No. 428863-50-7 or CH-138 (UCB), cilomilast (CAS No. 153259-65-5) (GlaxoSmithKline), CR-1 (Nuada Pharmaceuticals), CRx-119 (CombinatoRx), D-5410 (UCB), dacopafant (CAS No. 125372-33-0) (Sanofi- Aventis), dersalazine (CAS No. 188913-57-7/188913-58-8) (Uriach), etanercept (CAS No. 185243-69-0) (Amgen), ethyl pyruvate (Critical (Critical Therapeutics), golimumab (CAS No. 476181-74-5) (Johnson & Johnson), hormono-immunotherapy from Ipsen, CDP571 (e.g., Humicade from UCB), IC-485 (ICOS), infliximab (CAS No. 170277-31-3) (Johnson & Johnson), IP- 751 (Manhattan Pharmaceuticals), ISIS-104838 (CAS No. 250755-32-9) (ISIS Pharmaceuticals), lenalidomide (CAS No. 191732-72-6) (Celgene), lentinan (CAS No. 37339-90-5) (Ajinomoto), MDL-201112 (CAS No. 142130-73-2) (Sanofi-Aventis), medroxyprogesterone (CAS No. 520-85-4) (InKine Pharmaceutical), N-acetylcysteine (CAS No. 616-91-1) (Zambon), NBE-P2 (DIREVO Biotech), nerelimomab (CAS No. 162774-06-3) (Chiron), OM-294DP (OM PHARMA), onercept (CAS No. 199685-57-9) (Yeda), PASSTNF-alpha (Verigen), pentoxifylline or oxypentifylline (Sanofi-Aventis), Pharmaprojects No. 4091 , 4241 , 4295, or 4488 (Sanofi-Aventis), Pharmaprojects No. 5480 (Amgen), Pharmaprojects No. 6749 (Cengent), pirfenidone (CAS No. 53179-13-8) (MARNAC), RPR-132294 (Sanofi- Aventis), S5 (F002) (Fulcrum Pharmaceuticals), simvastatin (CAS No. 79902-63-9) (Merck & Co), STA-6292 (Synta Pharmaceuticals), tacrolimus (CAS No. 104987-11-3) (Fujisawa LifeCycle Pharma), talactoferrin alfa (CAS No. 308240-58-6) (Agennix), thalidomide (CAS No. 50-35-1) (Celgene), TNF antagonists form ProStrakan, and Synergen, TNF inhibitors (Amgen), TNF-alpha antagonists from Dynavax Technologies and Jerina AG (Germany), TNF-alpha inhibitors from IBFB Pharma and Xencor (Xencor), torbafylline (CAS No. 105102-21-4) (Sanofi-Aventis), UR-1505 (Uriach), VT-346 (Viron Therapeutics), YSIL6 (Y's Therapeutics), YSTH2 (Y's Therapeutics), NPI-1302a-3 (Nereus Pharmaceuticals, a TNF antagonist from Jerina AG (Germany), dersalazine, and an analogue or derivative thereof.
23295. The method of item 23193 or item 23194 wherein the agent is a Toll receptor inhibitor.
23296. The method of item 23193 or item 23194 wherein the agent is a tubulin antagonist.
23297. The method of item 23193 or item 23194 wherein the agent is a tyrosine kinase inhibitor selected from the group consisting of SU-011248, SUTENT from Pfizer Inc. (New York, NY), BMS- 354825, PN-355 (Paracelsian Pharmaceuticals), AGN-199659 (Allergan), AAL-993 or ABP-309 (Novartis), adaphostin (NIH), AEE-788 (Novartis), AG- 013736 (OSI Pharmaceuticals), AG-13736 (Pfizer), ALT-110 (Alteris Therapeutics), AMG-706 (Amgen), anticancer MAbs from Xencor, anti- EGFrvlll MAbs from Abgenix, anti-HER2 MAb from Abiogen, AZD-2171 or AZD-9935 (AstraZeneca), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), CEP-5214 (Cephalon), CEP-7055 (Cephalon), cetuximab (ImClone Systems), CHIR-200131 and CHIR-258 (Chiron), CP-547632 (OSI Pharmaceuticals), CP-724714 (Pfizer), CT-301 (Creabilis Therapeutics), D- 69491 (Baxter International), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EGFR/ErbB2 inhibitors from Array BioPharma, erlotinib (CAS No. 183319- 69-9) (OSI Pharmaceuticals), EXEL-2880 (Exelixis), FK-778 (Sanofi- Aventis) , gefitinib (CAS No. 184475-35-2) (AstraZeneca), GW-2286 or GW- 654652 (GlaxoSmithKline), her2/neu antigen from AlphaVax, HER-2/neu inhibitor from Generex, Herzyme (Medipad) (Sirna Therapeutics), HKI-272 (Wyeth), HuMax-EGFr (Genmab), idronoxil (CAS No. 81267-65-4) (Novogen), IGF-1 inhibitors from Ontogen, IMC-11F8 (ImCIone Systems), kahalalide F (CAS No. 149204-42-2) (PharmaMar), KDR inhibitor from LG Life Sciences, KDR inhibitors from Abbott Laboratories, KDR kinase inhibitors (UCB), Kdr kinase inhibitors from Merck & Co, KRN-633 and KRN- 951 (Kirin Brewery), KSB-102 (Xenova), lapatinib ditosylate (CAS No. 388082-78-8) (GlaxoSmithKline), matuzumab (Merck KGaA), MDX-214 (Medarex), ME-103 (Pharmexa), MED-A300 (Gerolymatos), MNAC-13 (Lay Line Genomics), nimotuzumab (Center of Molecular Immunology), NSC- 330507 or NSC-707545 (NIH), NV-50 (Novogen), OSI-930 (OSI Pharmaceuticals), panitumumab (Abgenix), pelitinib (CAS No. 287933-82-7) (Wyeth), pertuzumab (CAS No. 380610-27-5) (Genentech), Pharmaprojects No. 3985 (Sanofi-Aventis), prostate cancer therapeutics from Sequenom (SQPC35, SQPC36, SQPC90), removab and remoxab (Trion Pharma), RG- 13022 (CAS No. 136831-48-6), RG-13291 (CAS No. 138989-50-1), or RG- 14620 (CAS No. 136831-49-7) (Sanofi-Aventis), RM-6427 (Romark), RNAi breast cancer therapy from Benitec, RP 53801 (CAS No. 125882-88-4) (Sanofi-Aventis), sorafenib tosylate (Bayer), SU-11657 (Pfizer), Tie-2 antagonists from Semaia (Hybrigenics), Tie-2 inhibitors from Ontogen, trastuzumab (CAS No. 180288-69-1) (Genentech), tyrosine kinase inhibitors from Sanofi-Aventis, U3-1287, U3-1565, U3-1784, U3-1800 (U3 Pharma), vatalanib (Novartis), VEGFR-2 kinase inhibitor from Bristol-Myers Squibb, XL-647 (Exelixis), ZD-6474 (AstraZeneca), ZK-CDK (Schering AG), an EGFR tyrosine kinase inhibitor, EKB-569 (Wyeth), and an analogue or derivative thereof.
23298. The method of item 23193 or item 23194 wherein the agent is a VEGF inhibitor. 23299. The method of item 23193 or item 23194 wherein the agent is a vitamin D receptor agonist.
23300. The method of item 23193 or item 23194 wherein the agent is ZD-6474 (an angiogenesis inhibitor).
23301. The method of item 23193 or item 23194 wherein the agent is AP-23573 (an mTOR inhibitor).
23302. The method of item 23193 or item 23194 wherein the agent is synthadotin (a tubulin antagonist).
23303. The method of item 23193 or item 23194 wherein the agent is S-0885 (a collagenase inhibitor).
23304. The method of item 23193 or item 23194 wherein the agent is aplidine (an elongation factor-1 alpha inhibitor).
23305. The method of item 23193 or item 23194 wherein the agent is ixabepilone (an epithilone).
23306. The method of item 23193 or item 23194 wherein the agent is IDN-5390 (an angiogenesis inhibitor).
23307. The method of item 23193 or item 23194 wherein the agent is SB-2723005 (an angiogenesis inhibitor).
23308. The method of item 23193 or item 23194 wherein the agent is ABT-518 (an angiogenesis inhibitor). 23309. The method of item 23193 or item 23194 wherein the agent is combretastatin (an angiogenesis inhibitor).
23310. The method of item 23193 or item 23194 wherein the agent is anecortave acetate (an angiogenesis inhibitor).
23311. The method of item 23193 or item 23194 wherein the agent is SB-715992 (a kinesin antagonist).
23312. The method of item 23193 or item 23194 wherein the agent is temsirolimus (an mTOR inhibitor).
23313. The method of item 23193 or item 23194 wherein the agent is adalimumab (a TNFα antagonist).
23314. The method of item 23193 or item 23194 wherein the composition comprises an anti-thrombotic agent.
23315. The method of item 23193 or item 23194 wherein the composition comprises a naturally occurring polymer.
23316. The method of item 23193 or item 23194 wherein the composition comprises protein.
23317. The method of item 23193 or item 23194 wherein the composition comprises carbohydrate.
23318. The method of item 23193 or item 23194 wherein the composition comprises biodegradable polymer. 23319. The method of item 23193 or item 23194 wherein the composition comprises nonbiodegradable polymer.
23320. The method of item 23193 or item 23194 wherein the composition comprises collagen.
23321. The method of item 23193 or item 23194 wherein the composition comprises methylated collagen.
23322. The method of item 23193 or item 23194 wherein the composition comprises fibrinogen.
23323. The method of item 23193 or item 23194 wherein the composition comprises thrombin.
23324. The method of item 23193 or item 23194 wherein the composition comprises blood plasma.
23325. The method of item 23193 or item 23194 wherein the composition comprises calcium salt.
23326. The method of item 23193 or item 23194 wherein the composition comprises an antifibronolytic agent.
23327. The method of item 23193 or item 23194 wherein the composition comprises fibrinogen analog.
23328. The method of item 23193 or item 23194 wherein the composition comprises albumin. 23329. The method of item 23193 or item 23194 wherein the composition comprises plasminogen.
23330. The method of item 23193 or item 23194 wherein the composition comprises von Willebrands factor.
23331. The method of item 23193 or item 23194 wherein the composition comprises Factor VIII.
23332. The method of item 23193. or item 23194 wherein the composition comprises hypoallergenic collagen.
23333. The method of item 23193 or item 23194 wherein the composition comprises atelopeptidic collagen.
23334. The method of item 23193 or item 23194 wherein the composition comprises telopeptide collagen.
23335. The method of item 23193 or item 23194 wherein the composition comprises crosslinked collagen.
23336. The method of item 23193 or item 23194 wherein the composition comprises aprotinin.
23337. The method of item 23193 or item 23194 wherein the composition comprises epsilon-amino-n-caproic acid.
23338. The method of item 23193 or item 23194 wherein the composition comprises gelatin. 23339. The method of item 23193 or item 23194 wherein the composition comprises protein conjugates.
23340. The method of item 23193 or item 23194 wherein the composition comprises gelatin conjugates.
23341. The method of item 23193 or item 23194 wherein the composition comprises a synthetic polymer.
23342. The method of item 23193 or item 23194 wherein the composition comprises a synthetic isocyanate-containing compound.
23343. The method of item 23193 or item 23194 wherein the composition comprises a synthetic thiol-containing compound.
23344. The method of item 23193 or item 23194 wherein the composition comprises a synthetic compound containing at least two thiol groups.
23345. The method of item 23193 or item 23194 wherein the composition comprises a synthetic compound containing at least three thiol groups.
23346. The method of item 23193 or item 23194 wherein the composition comprises a synthetic compound containing at least four thiol groups.
23347. The method of item 23193 or item 23194 wherein the composition comprises a synthetic amino-containing compound. 23348. The method of item 23193 or item 23194 wherein the composition comprises a synthetic compound containing at least two amino groups.
23349. The method of item 23193 or item 23194 wherein the composition comprises a synthetic compound containing at least three amino groups.
23350. The method of item 23193 or item 23194 wherein the composition comprises a synthetic compound containing at least four amino groups.
23351. The method of item 23193 or item 23194 wherein the composition comprises a synthetic compound comprising a carbonyl-oxygen-succinimidyl group.
23352. The method of item 23193 or item 23194 wherein the composition comprises a synthetic compound comprising at least two carbonyl-oxygen-succinimidyl groups.
23353. The method of item 23193 or item 23194 wherein the composition comprises a synthetic compound comprising at least three carbonyl-oxygen-succinimidyl groups.
23354. The method of item 23193 or item 23194 wherein the composition comprises a synthetic compound comprising at least four carbonyl-oxygen-succinimidyl groups.
23355. The method of item 23193 or item 23194 wherein the composition comprises a synthetic polyalkylene oxide- containing compound. 23356. The method of item 23193 or item 23194 wherein the composition comprises a synthetic compound comprising both polyalkylene oxide and biodegradable polyester blocks.
23357. The method of item 23193 or item 23194 wherein the composition comprises a synthetic polyalkylene oxide- containing compound having reactive amino groups.
23358. The method of item 23193 or item 23194 wherein the composition comprises a synthetic polyalkylene oxide- containing compound having reactive thiol groups.
23359. The method of item 23193 or item 23194 wherein the composition comprises a synthetic polyalkylene oxide- containing compound having reactive carbonyl-oxygen-succinimidyl groups.
23360. The method of item 23193 or item 23194 wherein the composition comprises a synthetic compound comprising a biodegradable polyester block.
23361. The method of item 23193 or item 23194 wherein the composition comprises a synthetic polymer formed in whole or part from lactic acid or lactide.
23362. The method of item 23193 or item 23194 wherein the composition comprises a synthetic polymer formed in whole or part from glycolic acid or glycolide.
23363. The method of item 23193 or item 23194 wherein the composition comprises polylysine. 23364. The method of item 23193 or item 23194 wherein the composition comprises (a) protein and (b) a compound comprising a polyalkylene oxide portion.
23365. The method of item 23193 or item 23194 wherein the composition comprises (a) protein and (b) polylysine.
23366. The method of item 23193 or item 23194 wherein the composition comprises (a) protein and (b) a compound having at least four thiol groups.
23367. The method of item 23193 or item 23194 wherein the composition comprises (a) protein and (b) a compound having at least four amino groups.
23368. The method of item 23193 or item 23194 wherein the composition comprises (a) protein and (b) a compound having at least four carbonyl-oxygen-succinimide groups.
23369. The method of item 23193 or item 23194 wherein the composition comprises (a) protein and (b) a compound having a biodegradable region formed from reactants selected from lactic acid, lactide, glycolic acid, glycolide, and epison-caprolactone.
23370. The method of item 23193 or item 23194 wherein the composition comprises (a) collagen and (b) a compound comprising a polyalkylene oxide portion.
23371. The method of item 23193 or item 23194 wherein the composition comprises (a) collagen and (b) polylysine. 23372. The method of item 23193 or item 23194 wherein the composition comprises (a) collagen and (b) a compound having at least four thiol groups.
23373. The method of item 23193 or item 23194 wherein the composition comprises (a) collagen and (b) a compound having at least four amino groups.
23374. The method of item 23193 or item 23194 wherein the composition comprises (a) collagen and (b) a compound having at least four carbonyl-oxygen-succinimide groups.
23375. The method of item 23193 or item 23194 wherein the composition comprises (a) collagen and (b) a compound having a biodegradable region formed from reactants selected from lactic acid, lactide, glycolic acid, glycolide, and epison-caprolactone.
23376. The method of item 23193 or item 23194 wherein the composition comprises (a) methylated collagen and (b) a compound comprising a polyalkylene oxide portion.
23377. The method of item 23193 or item 23194 wherein the composition comprises (a) methylated collagen and (b) polylysine.
23378. The method of item 23193 or item 23194 wherein the composition comprises (a) methylated collagen and (b) a compound having at least four thiol groups. 23379. The method of item 23193 or item 23194 wherein the composition comprises (a) methylated collagen and (b) a compound having at least four amino groups.
23380. The method of item 23193 or item 23194 wherein the composition comprises (a) methylated collagen and (b) a compound having at least four carbonyl-oxygen-succinimide groups.
23381. The method of item 23193 or item 23194 wherein the composition comprises (a) methylated collagen and (b) a compound having a biodegradable region formed from reactants selected from lactic acid, lactide, glycolic acid, glycolide, and epison-caprolactone.
23382. The method of item 23193 or item 23194 wherein the composition comprises hyaluronic acid.
23383. The method of item 23193 or item 23194 wherein the composition comprises a hyaluronic acid derivative.
23384. The method of item 23193 or item 23194 wherein the composition comprises pentaerythritol poly(ethylene glycol)ether tetra-sulfhydryl of number average molecular weight between 3,000 and 30,000.
23385. The method of item 23193 or item 23194 wherein the composition comprises pentaerythritol poly(ethylene glycol)ether tetra-amino of number average molecular weight between 3,000 and 30,000.
23386. The method of item 23193 or item 23194 wherein the composition comprises (a) a synthetic compound having a number average molecular weight between 3,000 and 30,000 and comprising a polyalkylene oxide region and multiple nucleophilic groups, and (b) a synthetic compound having a number average molecular weight between 3,000 and 30,000 and comprising a polyalkylene oxide region and multiple electrophilic groups.
23387. The method of item 23193 or item 23194 wherein the composition comprises a colorant.
23388. The method of item 23193 or item 23194 wherein the composition is sterile.
23389. A method for reducing cartilage loss following an injury to a joint in a patient in need thereof, comprising delivering to the patient a) an anti-fibrotic agent or b) a composition comprising i) an anti- fibrotic agent and ii) a polymer and/or a compound that forms a polymer in situ.
23390. A method for preventing cartilage loss following an injury to a joint in a patient in need thereof, comprising delivering to the patient a) an anti-fibrotic agent or b) a composition comprising i) an anti- fibrotic agent and ii) a polymer and/or a compound that forms a polymer in situ.
23391. A method for reducing cartilage loss following a cruciate ligament tear in a patient in need thereof, comprising delivering to the patient a) an anti-fibrotic agent or b) a composition comprising i) an anti- fibrotic agent and ii) a polymer and/or a compound that forms a polymer in situ. 23392. A method for preventing cartilage loss following a cruciate ligament tear in a patient in need thereof, comprising delivering to the patient a) an anti-fibrotic agent or b) a composition comprising i) an anti- fibrotic agent and ii) a polymer and/or a compound that forms a polymer in situ.
23393. A method for reducing cartilage loss following a meniscal tear in a patient in need thereof, comprising delivering to the patient a) an anti-fibrotic agent or b) a composition comprising i) an anti- fibrotic agent and ii) a polymer and/or a compound that forms a polymer in situ.
23394. A method for preventing cartilage loss following a meniscal ligament tear in a patient in need thereof, comprising delivering to the patient a) an anti-fibrotic agent or b) a composition comprising i) an anti- fibrotic agent and ii) a polymer and/or a compound that forms a polymer in situ.
23395. The method of any one of items 23389-23394 wherein the agent or composition is delivered intra-articularly.
23396. The method of any one of items 23389-23394 wherein the agent is an adensosine A2A receptor antagonist.
23397. The method of any one of items 23389-23394 wherein the agent is an AKT inhibitor.
23398. The method of any one of items 23389-23394 wherein the agent is an alpha 2 integrin antagonist, wherein the alpha 2 integrin antagonist is Pharmaprojects No. 5754 (Merck KgaA). 23399. The method of any one of items 23389-23394 wherein the agent is an alpha 4 integrin antagonist.
23400. The method of any one of items 23389-23394 wherein the agent is an alpha 7 nicotinic receptor agonist.
23401. The method of any one of items 23389-23394 wherein the agent is an angiogenesis inhibitor selected from the group consisting of AG-12,958 (Pfizer), ATN-161 (Attention LLC)1 neovastat, an angiogenesis inhibitor from Jerina AG (Germany), NM-3 (Mercian), VGA- 1155 (Taisho), FCE-26644 (Pfizer), FCE-26950 (Pfizer), FPMA (Meiji Daries), FR-111142 (Fujisawa), GGTI-298, GM-1306 (Ligand), GPA-1734 (Novartis), NNC-47-0011 (Novo Nordisk), herbamycin (Nippon Kayaku), lenalidomide (Celegene), IP-10 (NIH), ABT-828 (Abbott), KIN-841 (Tokushima University, Japan), SF-1126 (Semafore Pharmaceuticals), laminin technology (N[H), CHIR-258 (Chiron), NVP-AEW541 (Novartis), NVP-AEW541 (Novartis), Vt16907 (Alchemia), OXI-8007 (Oxigene), EG- 3306 (Ark Therapeutics), Maspin (Arriva), ABT-567 (Abbott), PPI-2458 (Praecis Pharmaceuticals), CC-5079, CC-4089 (Celgene), HIF-1 alpha inhibitors (Xenova), S-247 (Pfizer), AP-23573 (Ariad), AZD-9935 (Astra Zeneca), mebendazole (Introgen Therapeutics), MetAP-2 inhibitors (GlaxoSmithKline), AG-615 (Angiogene Pharmaceuticals), Tie-2 antagonists (Hybrigenics), NC-381, CYC-381 , NC-169, NC-219, NC-383, NC-384, NC- 407 (Lorus Therapeutics), ATN-224 (Attenuon), ON-01370 (Onconova), Vitronectin antagonists (Amgen), SDX-103 (Salmedix), Vitronectin antagonists (Shire), CHP (Riemser), TEK (Amgen), Anecortave acetate (Alcon), T46.2 (Matrix Therapeutics), HG-2 (Heptagen), TEM antagonists (Genzyme), Oxi-4500 (Oxigene), ATN-161 (Attenuon), WX-293 (Wilex), M- 2025 (Metris Therapeutics), Alphastatin (BioActa), YH-16 (Yantai Rongchang), BIBF-1120 (Boehringer Ingelheim), BAY-57-9352 (Bayer), AS- 1404 (Cancer Research Technology), SC-77964 (Pfizer), glycomimetics (BioTie Therapies), TIE-2 Inhibitors (Ontogen), DIMI, Octamer (Octamer), ABR-215050 (Active Biotech), ABT-518 (Abbott), KDR inhibitors (Abbott), BSF-466895 (Abbott), SCH-221153 (Schering-Plough), DAC:antiangiogenic (ConjuChem), TFPI (EntreMed), AZD-2171 (Astra-Zenaca), CDC-394 (Celgene), LY290293 (EIi Lilly), IDN-5390 (Indena), Kdr Kinase Inhibitors (Merck), CT-113020, CT-116433, CT-116563, CT-31890, CT-32228) (Cell Therapeutics), A-299620 (Abbott), TWEAK Inhibitor (Amgen), VEGF modulators (Johnson and Johnson), Tum-N53, tumstatin (Genzyme), Thios- 1 , Thios-2 (Thios Pharmaceuticals), MV-6401 (Miravant Medical Technologies), Spisulosine (PharmaMar), CEP-7055 (Cephalon), AUV-201 (Auvation), LM-609 (EIi Lilly), SKF-106615 (AnorMED), Oglufanide disodium (Cytran), BW-114 (Pharminox), Calreticulin (NIH), WX-678 (Wilex), SD-7784 (Pfizer), WX-UK1 (Wilex), SH-268 (Schering AG), 2-Me-PGA (Celgene), S- 137 (Pfizer), ZD-6126 (Angiogene Pharmaceuticals), SG-292 (SignalGen), Benefin (Lane Labs), A6, A36 (Angstrom), SB-2723005 (GlaxoSmithKline), SC-7 (Cell Therapeutics), ZEN-014 (AEterna Zentaris), 2-methoxyestradiol (EntreMed), NK-130119 (Nippon Kayaku), CC-10004 (Celgene), AVE- 8062A (Ajinomoto), Tacedinaline (Pfizer), Actinonin (Tokyo Metropolitan Institute of Medical Science), Lenalidomide (Celgene), VGA-1155, BTO-956 (SRI International), ER-68203-00 (Eisai), CT-6685 (UCB), JKC-362 (Phoenix Pharmaceuticals), DMI-3798 (DMI Biosciences, Angiomate (Ipsen), ZD-6474 (AstraZeneca), CEP-5214 (Cephalon), Canstatin (Genzyme), NM-3 (Mercian), Oridigm (MediQuest Therapeutics), Exherin (Adherex), BLS-0597 (Boston Life Sciences), PTC-299 (PTC Therapeutics), NPI-2358 (Nereus Pharmaceuticals), CGP-79787 (Novartis), AEE-788 (Novartis), CKD-732 (Chong Kun Dang), CP-564959 (OSI Pharmaceuticals), CM-101 (CarboMed), CT-2584, CT3501 (Cell Therapeutics), combretastatin and analogues and derivatives thereof (Oxigene), Rebimastat (Bristol-Meyers Squibb), Dextrin 2-sulfate (ML Laboratories), Cilengitide (Merk KGaA), NSC-706704 (Pharminox), KRN- 951 (Kirin Brewery), Ukrain, NSC-631570 (Nowicky Pharma), Tecogalan sodium (Daiichi Pharmaceutical), Tz-93 (Tsumura), TBC-1635 (Encysive Pharmaceuticals), TAN-1120 (Takeda), Semaxanib (Pfizer), BDI-7800 (Biopharmacopae), SD-186, SD-983 (Bristol-Meyers Squibb), SB-223245 (GlaxoSmithKline), SC-236 (Pfizer), RWJ-590973 (Johnson and Johnson), ILX-1850 (Genzyme), SC-68488, S-836 (Pfizer), CG-55069-11 (CuraGen), Ki-23057 (Kirin Brewery), CCX-700 (Chemoentryx), Pegaptanib octasodium (Giled Sciences), ANGIOCOL (available from Biostratum Inc.), or an analogue or derivative thereof.
23402. The method of any one of items 23389-23394 wherein the agent is an apoptosis antagonist.
23403. The method of any one of items 23389-23394 wherein the agent is an apoptosis activator.
23404. The method of any one of items 23389-23394 wherein the agent is a beta 1 integrin antagonist.
23405. The method of any one of items 23389-23394 wherein the agent is a beta tubulin inhibitor.
23406. The method of any one of items 23389-23394 wherein the agent is a blocker of enzyme production in Hepatitis C.
23407. The method of any one of items 23389-23394 wherein the agent is a Bruton's tyrosine kinase inhibitor.
23408. The method of any one of items 23389-23394 wherein the agent is a calcineurin inhibitor. 23409. The method of any one of items 23389-23394 wherein the agent is a caspase 3 inhibitor.
23410. The method of any one of items 23389-23394 wherein the agent is a CC chemokine receptor antagonist.
23411. The method of any one of items 23389-23394 wherein the agent is a cell cycle inhibitor selected from the group consisting of SNS-595 (Sunesis), synthadotin, KRX-0403, and an analogue or derivative thereof.
23412. The method of any one of items 23389-23394 wherein the agent is a cathepsin B inhibitor.
23413. The method of any one of items 23389-23394 wherein the agent is a cathepsin K inhibitor, wherein the cathepsin K inhibitor is 462795 (GlaxoSmithKline), INPL-022-D6 (Amura Therapeutics), or an analogue or derivative thereof.
23414. The method of any one of items 23389-23394 wherein the agent is a cathepsin L inhibitor.
23415. The method of any one of items 23389-23394 wherein the agent is a CD40 antagonist.
23416. The method of any one of items 23389-23394 wherein the agent is a chemokine receptor agonist.
23417. The method of any one of items 23389-23394 wherein the agent is a chymase inhibitor. 23418. The method of any one of items 23389-23394 wherein the agent is a collagenase antagonist.
23419. The method of any one of items 23389-23394 wherein the agent is a CXCR antagonist.
23420. The method of any one of items 23389-23394 wherein the agent is a cyclin dependent kinase inhibitor selected from the group consisting of a CDK- 1 inhibitor, a CDK-2 inhibitor, a CDK- 4 inhibitor, a CDK-6 inhibitor, a CAK1 inhibitor from GPC Biotech and Bristol-Myers Squibb, RGB-286199 (GPC Biotech), an anticancer agent from Astex Technology, a CAK1 inhibitor from GPC Biotech, a CDK inhibitor from Sanofi-Aventis, a CDK1/CDK2 inhibitor from Amgen, a CDK2 inhibitor from SUGEN-2 (Pfizer), a hearing loss therapy agent (Sound Pharmaceuticals), PD-0332991 (Pfizer), RGB-286199 (GPC Biotech), Ro-0505124 (Hoffmann- La Roche), a Ser/Thr kinase inhibitor from Lilly (EIi Lilly), CVT-2584 (CAS No. 199986-75-9) (CV Therapeutics), and an analogue or derivative thereof.
23421. The method of any one of items 23389-23394 wherein the agent is a cyclooxygenase 1 inhibitor.
23422. The method of any one of items 23389-23394 wherein the agent is a DHFR inhibitor.
23423. The method of any one of items 23389-23394 wherein the agent is a dual integrin inhibitor.
23424. The method of any one of items 23389-23394 wherein the agent is an elastase inhibitor. 23425. The method of any one of items 23389-23394 wherein the agent is an elongation factor- 1 alpha inhibitor.
23426. The method of any one of items 23389-23394 wherein the agent is an endothelial growth factor antagonist.
23427. The method of any one of items 23389-23394 wherein the agent is an endothelial growth factor receptor kinase inhibitor selected from the group consisting of sorafenib tosylate (Bayer), AAL-993 (Novartis), ABP-309 (Novartis), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EXEL- 2880 (Exelixis), GW-654652 (GlaxoSmith Kline), a KDR inhibitor from LG Life Sciences, CT-6685 and CT-6729 (UCB), KRN-633 and KRN-951 (Kirin Brewery), OSI-930 (OSI Pharmaceuticals), SP-5.2 (Supratek Pharma), SU- 11657 (Pfizer), a Tie-2 antagonist (Hybrigenϊcs), a VEGFR-2 kinase inhibitor (Bristol-Myers Squibb), XL-647 (Exelixis), a KDR inhibitor from Abbott Laboratories, sorafenib tosylate, and an analogue or derivative thereof.
23428. The method of any one of items 23389-23394 wherein the agent is an endotoxin antagonist.
23429. The method of any one of items 23389-23394 wherein the agent is an epothilone and tubulin binder.
23430. The method of any one of items 23389-23394 wherein the agent is an estrogen receptor antagonist.
23431. The method of any one of items 23389-23394 wherein the agent is an FGF inhibitor. 23432. The method of any one of items 23389-23394 wherein the agent is a farnexyl transferase inhibitor.
23433. The method of any one of items 23389-23394 wherein the agent is famesyltransferase inhibitor selected from the group of A-197574 (Abbott), a famesyltransferase inhibitor from Servier, FPTIII (Strathclyde Institute for Drug R), LB-42908 (LG Life Sciences), Pharmaprojects No. 5063 (Genzyme), Pharmaprojects No. 5597 (Ipsen), Yissum Project No. B-1055 (Yissum), and an analogue or derivative thereof.
23434. The method of any one of items 23389-23394 wherein the agent is an FLT-3 kinase inhibitor.
23435. The method of any one of items 23389-23394 wherein the agent is an FGF receptor kinase inhibitor.
23436. The method of any one of items 23389-23394 wherein the agent is a fibrinogen antagonist selected from the group consisting of AUV-201 (Auvation), MG-13926 (Sanofi-Aventis), plasminogen activator (CAS No. 105913-11-9) (from Sanofi-Aventis or UCB), plasminogen activator-2 (tPA-2) (Sanofi-Aventis), pro-urokinase (CAS No. 82657-92-9) (Sanofi-Aventis), and an analogue or derivative thereof.
23437. The method of any one of items 23389-23394 wherein the agent is a heat shock protein 90 antagonist selected from the group consisting of SRN-005 (Sirenade), geldanamycin, NSC-33050 (17- allylaminogeldanamycin; 17-AAG), 17-dimethylaminoethylamino-17- demethoxy-geldanamycin (17-DMAG), rifabutin (rifamycin XIV, 1',4- didehydro-1-deoxy-1,4-dihydro-5'-(2-methylpropyl)-1-oxo-), and an analogue or derivative thereof. 23438. The method of any one of items 23389-23394 wherein the agent is a histone deacetylase inhibitor.
23439. The method of any one of items 23389-23394 wherein the agent is an HMGCoA reductase inhibitor selected from the group consisting of an atherosclerosis therapeutic from Lipid Sciences, ATI- 16000 (ARYx Therapeutics), KS-01-019 (Kos Pharmaceuticals), Pharmaprojects No. 2197 (Sanofi-Aventi), RP 61969 (Sanofi-Aventis), and an analogue or derivative thereof.
23440. The method of any one of items 23389-23394 wherein the agent is an ICAM inhibitor.
23441. The method of any one of items 23389-23394 wherein the agent is an IL, ICE and IRAK antagonist, wherein the antagonist is a CJ-14877, CP-424174 (Pfizer), NF-61 (Negma-Lerads), and an analogue or derivative thereof.
23442. The method of any one of items 23389-23394 wherein the agent is an IL-2 inhibitor.
23443. The method of any one of items 23389-23394 wherein the agent is an immunosuppressant selected from the group consisting of teriflunomide (Sanofi Aventis), chlorsulfaquinoxalone (NSC- 339004), chlorsulfaquinoxalone sulfate, CS-712 (Sankyo), ismomultin alfa (CAS No. 457913-93-8) (Akzo Nobel), antiallergics from GenPat77, antiinflammatories or AT-005 (Androclus Therapeutics), autoimmune disease therapeutics from EpiVax, BN-007 (Bone), budesonide (CAS No. 51333-22- 3) (MAP Pharmaceuticals), CO-14 (Genzyme), edratide (CAS No. 433922- 67-9) (Teva), EP-314 (Enanta), eprovafen (CAS No. 101335-99-3) (Sanofi- Aventis), HWA-131 (CAS No. 118788-41-3) (Sanofi-Aventis), immunomodufators from MerLion Pharmaceuticals, immunosuppressives from Alchemia, IPL-12 (Inflazyme), MDL-9563 (CAS No. 27086-86-8) (Sanofi-Aventis), Pharmaprojects No. 2330 (Sanofi-Aventis), Pharmaprojects No. 6426 (Abgenix), PXS-25 (Pharmaxis), rosmarinic acid (CAS No. 20283-92-5) (Sanofi-Aventis), RP 42927 or RP 54745 (CAS No. 135330-08-4) (Sanofi-Aventis), SGN-35 (Seattle Genetics), ST-1959 (Sigma-Tau), type I diabetes therapy from SYNX Pharma, UNIL-88 (Debiopharm), VP-025 (Vasogen), VR-694 (Vectura), PRTX-001 (Protalex), and an analogue or derivative thereof.
23444. The method of any one of items 23389-23394 wherein the agent is an IMPDH (inosine monophosphate).
23445. The method of any one of items 23389-23394 wherein the agent is an integrin antagonist.
23446. The method of any one of items 23389-23394 wherein the agent is an interleukin antagonist.
23447. The method of any one of items 23389-23394 wherein the agent is an inhibitor of type III receptor tyrosine kinase.
23448. The method of any one of items 23389-23394 wherein the agent is an irreversible inhibitor of enzyme methionine aminopeptidase type 2.
23449. The method of any one of items 23389-23394 wherein the agent is an isozyme selective delta protein kinase C inhibitor.
23450. The method of any one of items 23389-23394 wherein the agent a JAK3 enzyme inhibitor. 23451. The method of any one of items 23389-23394 wherein the agent is a JNK inhibitor.
23452. The method of any one of items 23389-23394 wherein the agent is a kinase inhibitor.
23453. The method of any one of items 23389-23394 wherein the agent is kinesin antagonist.
23454. The method of any one of items 23389-23394 wherein the agent is a kinesin antagonist.
23455. The method of any one of items 23389-23394 wherein the agent is a leukotriene inhibitor and antagonist selected from the group consisting of ambicromil (CAS No. 58805-38-2) (Sanofi-Aventis), amelubant (CAS No. 346735-24-8) (Boehringer Ingelheim), DW-1141 (Dong Wha), ebselen (Daiichi Pharmaceutical), ibudilast (Kyorin), leucotriene inhibitors from Sanofi-Aventis, lymphotoxin-beta receptor (LT-D) from Biogen Idee, Pharmaprojects No. 1535 and 2728 (CAS No. 119340-33-9) (Sanofi-Aventis), R-112 (Rigel), Rev-5367 (CAS No. 92532-05-3) (Sanofi- Aventis), RG-14893 (CAS No. 141835-49-6) (Sanofi-Aventis), RG-5901-A (CAS No. 101910-24-1), 92532-23-5, RP 66153 (CAS No. 142422-79-5), RP 66364 (CAS No.186916-92-5), RP 69698 (CAS No. 141748-00-7) (Sanofi-Aventis), SC-411930 (Pfizer), SC-41930 (CAS No. 120072-59-5) (Pfizer), SC-50605 (CAS No. 138828-39-4) (Pfizer), SC-51146 (CAS No. 141059-52-1), SC-53228 (CAS No. 153633-01-3) (Pfizer), spaglumic acid (ZY-15106) (CAS No.3106-85-2), 80619-64-3 (Novartis), tipredane (CAS No. 85197-77-9) (Bristol-Myers Squibb), U-75302 (CAS No. 119477-85-9) (Pfizer), and analogue or derivative thereof. 23456. The method of any one of items 23389-23394 wherein the agent is an MAP kinase inhibitor.
23457. The method of any one of items 23389-23394 wherein the agent is a matrix metalloproteinase inhibitor.
23458. The method of any one of items 23389-23394 wherein the agent is an MCP-CCR2 inhibitor.
23459. The method of any one of items 23389-23394 wherein the agent is an mTOR inhibitor.
23460. The method of any one of items 23389-23394 wherein the agent is an mTOR kinase inhibitor.
23461. The method of any one of items 23389-23394 - wherein the agent is a microtubule inhibitor selected from the group consisting of antibody-maytansinoid conjugates from Biogen Idee, colchicines {MantiCore Pharmaceuticals), anticancer immunoconjugates from Johnson & Johnson, DIME from Octamer, gni-1f (GNI), huC242-DM4, huMy9-6-DM1 (ImmunoGen), IDN-5404 (Indena), IMO-098, IMOderm (Imotep), mebendazole (Introgen Therapeutics), microtubule poisons from Cambridge Enterprise, paclitaxel such as LOTAX from Aphios (CAS No. 33069-62-4), Genexol-PM from Samyang, Pharmaprojects No. 6383 (Tapestry Pharmaceuticals), RPR-112378 (Sanofi-Aventis), SGN-75 (Seattle Genetics), SPL-7435 (Starpharma), SSR-250411 (Sanofi-Aventis), trastuzumab-DM1 (Genentech), vinorelbine, and an analogue or derivative thereof.
23462. The method of any one of items 23389-23394 wherein the agent is an MIF inhibitor. 23463. The method of any one of items 23389-23394 wherein the agent is an MMP inhibitor.
23464. The method of any one of items 23389-23394 wherein the agent is a neurokinin (NK) antagonist selected from the group consisting of anthrotainin (CAS No. 148084-40-6) (Sanofi-Aventis), an IBS thereapeutic from ArQuIe, MDL-105212A (CAS No. 167261-60-1) (Ssanofi- Aventis), Pharmaprojects No. 2744, 3258 (CAS No. 139167-47-8) 4006, 4201 , or 5986 (Sanofi-Aventis), RP 67580 (CAS No. 135911-02-3), SR- 144190 (CAS No. 201152-86-5), SSR-240600, SSR-241586 (Sanofi- Aventis), TKA-457 (Novartis), vestipitant mesylate (CAS No. 334476-64-1) (GlaxoSmithKline), Win-64821 (Sanofi-Aventis), PRX-96026 (Predix Pharmaceuticals), and an analogue or derivative thereof.
23465. The method of any one of items 23389-23394 wherein the agent is an NF kappa B inhibitor selected from the group consisting of AVE-0545 or AVE-0547 (Sanofi-Aventis), bortezomib (CAS No. 179324-69-7) (Millennium Pharmaceuticals), dexanabinol (CAS No. 112924- 45-5) (Pharmos), dexlipotam (Viatris), Pharmaprojects No. 6283 (INDRA) (OXiGENE), IPL-576092 (CAS No. 137571-30-3) (Inflazyme), NFKB decoy (Corgentech), NFKB decoy oligo (AnGes MG)1 NFKB's from Ariad, osteoporosis treatments or S5 (F005) from Fulcrum Pharmaceuticals, P61 (Phytopharm), R-flurbiprofen (CAS No. 5104-49-4) (Encore Pharmaceuticals), and an analogue or derivative thereof.
23466. The method of any one of items 23389-23394 wherein the agent is a nitric oxide agonist.
23467. The method of any one of items 23389-23394 wherein the agent is an ornithine decarboxylase inhibitor. 23468. The method of any one of items 23389-23394 wherein the agent is a p38 MAP kinase inhibitor selected from the group consisting of AZD-6703 (AstraZeneca), JX-401 (Jexys Pharmaceuticals), BMS-2 (Bristol-Myers Squibb), a p38 MAP kinase inhibitor from Novartis, a p38-alpha MAP kinase inhibitor from Amphora, Pharmaprojects No. 5704 (Pharmacopeia), RPR-200765A (Sanofi-Aventis), SD-282 (Johnson & Johnson), TAK-715 (Takeda), and an analogue or derivative thereof.
23469. The method of any one of items 23389-23394 wherein the agent is a palmitoyl-protein thioesterase inhibitor.
23470. The method of any one of items 23389-23394 wherein the agent is a PDGF receptor kinase inhibitor selected from the group consisting of AAL-993, AMN-107, or ABP-309 (Novartis), AMG-706 (Amgen), BAY-57-9352 (Bayer), CDP-860 (UCB), E-7080 (Eisai), imatinib (CAS No. 152459-95-5) (Novartis), OSI-930 (OSI Pharmaceuticals), RPR- 127963E (Sanofi-Aventis), RWJ-540973 (Johnson & Johnson), sorafenib tosylate (Bayer), SU-11657 (Pfizer), tandutinib (CAS No. 387867-13-2) (Millennium Pharmaceuticals), vatalanib (Novartis), ZK-CDK (Schering AG), and an analogue or derivative thereof.
23471. The method of any one of items 23389-23394 wherein the agent is (-)-halofenate (Metabolex), AMG-131 (Amgen), antidiabetics from Japan Tobacco, AZD-4619, AZD-8450, AZD-8677 (AstraZeneca), DRF-10945, balaglitazone (Dr Reddy's), CS-00088, CS- 00098 (Chipscreen Biosciences), E-3030 (Eisai), etalocib (CAS No. 161172- 51-6) (EIi Lilly), GSK-641597 (Ligand), GSK-677954 (GlaxoSmithKline), GW-409544 (Ligand), GW-590735 (GlaxoSmithKline), K-111 (Hoffmann-La Roche), LY-518674 (EIi Lilly), LY-674 (Ligand), LY-929 (Ligand), MC-3001, MC-3002 (MaxoCore Pharmaceuticals), metformin HCl + pioglitazone (CAS No. 1115-70-4 and 112529-15-4), ACTOPLUS MET from Andrx), muraglitazar (CAS No. 331741-94-7) (Bristol-Myers Squibb), naveglitazar (Ligand), oleoylethanolamide (Kadmus Pharmaceuticals), ONO-5129, pioglitazone hydrochloride (CAS No. 111025-46-8 and 112529-15-4) (Takeda), PLX-204 (Plexxikon), PPAR agonists from Genfit, PPAR delta agonists from EIi Lilly, PPAR-alpha agonists from CrystalGenomics, PPAR- gamma modulators and PPAR-β modulators from CareX, rosiglitazone maleate (CAS No. 122324-73-4 or 155141-29-0) (GlaxoSmithKline), rosiglitazone maleate/glimepir (CAS No. 155141-29-0 and 93479-97-1), AVANDARYL, rosiglitazone maleate/metformin extend (CAS No. 155141- 29-0 and 657-24-9), AVANDAMET, rosiglitazone maleate+metformin, AVANDAMET (GlaxoSmithKline), tesaglitazar (AstraZeneca), and an analogue or derivative thereof.
23472. The method of any one of items 23389-23394 wherein the agent is a phosphatase inhibitor.
23473. The method of any one of items 23389-23394 wherein the agent is a phosphodiesterase (PDE) inhibitor selected from the group consisting of avanafil (Tanabe Seiyaku), dasantafil (CAS No. 569351- 91-3) (Schering-Plough), A-906119 (CAS No. 134072-58-5), DL-850 (Sanofi-Aventis), GRC-3015, GRC-3566, GRC-3886 (Glenmark), HWA-153 (CAS No. 56395-66-5) (Sanofi-Aventis), hydroxypumafentrine (Altana), IBFB-130011 , IBFB-14-016, IBFB-140301, IBFB-150007, IBFB-211913 (IBFB Pharma), L-826141 (Merck & Co), medorinone (CAS No. 88296-61-1) (Sanofi-Aventis), MEM-1917 (Memory Pharmaceuticals), ND-1251 (Neuro3d), PDE inhibitors from ICOS, PDE IV inhibitors from Memory Pharmaceuticals and CrystalGenomics, Pharmaprojects No. 2742 and 6141 (Sanofi-Aventis), QAD-171 (Novartis), RHC-2963 (CAS No. 76993-12-9 and 76993-14-1), RPR-117658, RPR-122818 derivatives, SR-24870 , and RPR- 132294 (Sanofi-Aventis), SK-350 (ln2Gen), stroke therapy agents from deCODE Genetics, TAS-203 (Taiho), tofimilast (CAS No. 185954-27-2) (Pfizer), UK-371800 (Pfizer), WIN-65579 (CAS No. 158020-82-7) (Sanofi- Aventis), IBFB-130020 (IBFB Pharma), OPC-6535 (CAS No. 145739-56-6) (Otsuka), a phosphodiesterase III inhibitor, enoximone, a phosphodiesterase IV inhibitor, fosfosal, Atopik (Barrier Therapeutics), triflusal, a phosphodiesterase V inhibitor, and an analogue or derivative thereof.
23474. The method of any one of items 23389-23394 wherein the agent is a PKC inhibitor.
23475. The method of any one of items 23389-23394 wherein the agent is a platelet activating factor antagonist.
23476. The method of any one of items 23389-23394 wherein the agent is a platelet-derived growth factor receptor kinase inhibitor.
23477. The method of any one of items 23389-23394 wherein the agent is a prolyl hydroxylase inhibitor.
23478. The method of any one of items 23389-23394 wherein the agent is a polymorphonuclear neutrophil inhibitor.
23479. The method of any one of items 23389-23394 wherein the agent is a protein kinase B inhibitor.
23480. The method of any one of items 23389-23394 wherein the agent is a protein kinase C stimulant.
23481. The method of any one of items 23389-23394 wherein the agent is a purine nucleoside analogue. 23482. The method of any one of items 23389-23394 wherein the agent is a purinoreceptor P2X antagonist.
23483. The method of any one of items 23389-23394 wherein the agent is a Raf kinase inhibitor.
23484. The method of any one of items 23389-23394 wherein the agent is a reversible inhibitor of ErbB1 and ErbB2.
23485. The method of any one of items 23389-23394 wherein the agent is a ribonucleoside triphosphate reductase inhibitor.
23486. The method of any one of items 23389-23394 wherein the agent is an SDF-1 antagonist.
23487. The method of any one of items 23389-23394 wherein the agent is a sheddase inhibitor.
23488. The method of any one of items 23389-23394 wherein the agent is an SRC inhibitor.
23489. The method of any one of items 23389-23394 wherein the agent is a stromelysin inhibitor.
23490. The method of any one of items 23389-23394 wherein the agent is an Syk kinase inhibitor.
23491. The method of any one of items 23389-23394 wherein the agent is a telomerase inhibitor. 23492. The method of any one of items 23389-23394 wherein the agent is a TGF beta inhibitor selected from the group consisting of pirfenidone (CAS No. 53179-13-8) (MARNAC), tranilast (CAS No. 53902- 12-8) (Kissei), IN-1130 (ln2Gen), mannose-6-phosphate (BTG), TGF-β antagonists from Inflazyme (Pharmaprojects No. 6075), TGF-β antagonists from Sydney, non-industrial source), TGF-β I receptor kinase inhibitors from EIi Lilly, TGF-β receptor inhibitors from Johnson & Johnson, and an analogue or derivative thereof.
23493. The method of any one of items 23389-23394 wherein the agent is a TNFα antagonist or TACE inhibitor selected from the group consisting of adalimumab (CAS No. 331731-18-1) (Cambridge Antibody Technology), AGIX-4207 (AtheroGenics), AGT-1 (Advanced Biotherapy), an anti-inflammatory from Borean Pharma, Cellzome, or Paradigm Therapeutics, anti-inflammatory vaccine (TNF-alpha kinoid) from Neovacs, humanized anti-TNF antibody or an anti-TNF MAb (CB0006) Celltech (UCB), apratastat (CAS No. 287445-51-0) (Wyeth), BMS-561392 (Bristol-Myers Squibb), BN-006 (Bone), certolizumab pegol (CAS No. 428863-50-7 or CH-138 (UCB), cilomilast (CAS No. 153259-65-5) (GlaxoSmithKline), CR-1 (Nuada Pharmaceuticals), CRx-119 (CombinatoRx), D-5410 (UCB), dacopafant (CAS No. 125372-33-0) (Sanofi- Aventis), dersalazine (CAS No. 188913-57-7/188913-58-8) (Uriach), etanercept (CAS No. 185243-69-0) (Amgen), ethyl pyruvate (Critical (Critical Therapeutics), golimumab (CAS No. 476181-74-5) (Johnson & Johnson), hormono-immunotherapy from Ipsen, CDP571 (e.g., Humicade from UCB), IC-485 (ICOS), infliximab (CAS No. 170277-31-3) (Johnson & Johnson), IP- 751 (Manhattan Pharmaceuticals), ISIS-104838 (CAS No. 250755-32-9) (ISIS Pharmaceuticals), lenalidomide (CAS No. 191732-72-6) (Celgene), lentinan (CAS No. 37339-90-5) (Ajinomoto), MDL-201112 (CAS No. 142130-73-2) (Sanofi-Aventis), medroxyprogesterone (CAS No. 520-85-4) (InKine Pharmaceutical), N-acetylcysteine (CAS No. 616-91-1) (Zambon), NBE-P2 (DIREVO Biotech), nerelimomab (CAS No. 162774-06-3) (Chiron), OM-294DP (OM PHARMA), onercept (CAS No. 199685-57-9) (Yeda), PASSTNF-alpha (Verigen), pentoxifylline or oxypentifylline (Sanofi-Aventis), Pharmaprojects No. 4091, 4241 , 4295, or 4488 (Sanofi-Aventis), Pharmaprojects No. 5480 (Amgen), Pharmaprojects No. 6749 (Cengent), pirfenidone (CAS No. 53179-13-8) (MARNAC), RPR-132294 (Sanofi- Aventis), S5 (F002) (Fulcrum Pharmaceuticals), simvastatin (CAS No. 79902-63-9) (Merck & Co), STA-6292 (Synta Pharmaceuticals), tacrolimus (CAS No. 104987-11-3) (Fujisawa LifeCycle Pharma), talactoferrin alfa (CAS No. 308240-58-6) (Agennix), thalidomide (CAS No. 50-35-1) (Celgene), TNF antagonists form ProStrakan, and Synergen, TNF inhibitors (Amgen), TNF-alpha antagonists from Dynavax Technologies and Jerina AG (Germany), TNF-alpha inhibitors from IBFB Pharma and Xencor (Xencor), torbafylline (CAS No. 105102-21-4) (Sanofi-Aventis), UR-1505 (Uriach), VT-346 (Viron Therapeutics), YSIL6 (Y1S Therapeutics), YSTH2 (Y's Therapeutics), NPI-1302a-3 (Nereus Pharmaceuticals, a TNF antagonist from Jerina AG (Germany), dersalazine, and an analogue or derivative thereof.
23494. The method of any one of items 23389-23394 wherein the agent is a Toll receptor inhibitor.
23495. The method of any one of items 23389-23394 wherein the agent is a tubulin antagonist.
23496. The method of any one of items 23389-23394 wherein the agent is a tyrosine kinase inhibitor selected from the group consisting of SU-011248, SUTENT from Pfizer Inc. (New York, NY), BMS- 354825, PN-355 (Paracelsian Pharmaceuticals), AGN-199659 (Allergan), AAL-993 or ABP-309 (Novartis), adaphostin (NIH), AEE-788 (Novartis), AG- 013736 (OSI Pharmaceuticals), AG-13736 (Pfizer), ALT-110 (Alteris Therapeutics), AMG-706 (Amgen), anticancer MAbs from Xencor, anti- EGFrvlll MAbs from Abgenix, anti-HER2 MAb from Abiogen, AZD-2171 or AZD-9935 (AstraZeneca), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), CEP-5214 (Cephalon), CEP-7055 (Cephalon), cetuximab (ImClone Systems), CHIR-200131 and CHIR-258 (Chiron), CP-547632 (OSI Pharmaceuticals), CP-724714 (Pfizer), CT-301 (Creabilis Therapeutics), D- 69491 (Baxter International), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EGFR/ErbB2 inhibitors from Array BioPharma, erlotinib (CAS No. 183319- 69-9) (OSI Pharmaceuticals), EXEL-2880 (Exelixis), FK-778 (Sanofi- Aventis) , gefitinib (CAS No. 184475-35-2) (AstraZeneca), GW-2286 or GW- 654652 (GlaxoSmithKline), her2/neu antigen from AlphaVax, HER-2/neu inhibitor from Generex, Herzyme (Medipad) (Sirna Therapeutics), HKI-272 (Wyeth), HuMax-EGFr (Genmab), idronoxil (CAS No. 81267-65-4) (Novogen), IGF-1 inhibitors from Ontogen, IMC-11F8 (ImClone Systems), kahalalide F (CAS No. 149204-42-2) (PharmaMar), KDR inhibitor from LG Life Sciences, KDR inhibitors from Abbott Laboratories, KDR kinase inhibitors (UCB), Kdr kinase inhibitors from Merck & Co, KRN-633 and KRN- 951 (Kirin Brewery), KSB-102 (Xenova), lapatinib ditosylate (CAS No. 388082-78-8) (GlaxoSmithKline), matuzumab (Merck KGaA), MDX-214 (Medarex), ME-103 (Pharmexa), MED-A300 (Gerolymatos), MNAC-13 (Lay Line Genomics), nimotuzumab (Center of Molecular Immunology), NSC- 330507 or NSC-707545 (NIH), NV-50 (Novogen), OSI-930 (OSI Pharmaceuticals), panitumumab (Abgenix), pelitinib (CAS No. 287933-82-7) (Wyeth), pertuzumab (CAS No. 380610-27-5) (Genentech), Pharmaprojects No. 3985 (Sanofi-Aventis), prostate cancer therapeutics from Sequenom (SQPC35, SQPC36, SQPC90), removab and remoxab (Trion Pharma), RG- 13022 (CAS No. 136831-48-6), RG-13291 (CAS No. 138989-50-1), or RG- 14620 (CAS No. 136831-49-7) (Sanofi-Aventis), RM-6427 (Romark), RNAi breast cancer therapy from Benitec, RP 53801 (CAS No. 125882-88-4) (Sanofi-Aventis), sorafenib tosylate (Bayer), SU-11657 (Pfizer), Tie-2 antagonists from Semaia (Hybrigenics), Tie-2 inhibitors from Ontogen, trastuzumab (CAS No. 180288-69-1) (Genentech), tyrosine kinase inhibitors from Sanofi-Aventis, U3-1287, U3-1565, U3-1784, U3-1800 (U3 Pharma), vatalanib (Novartis), VEGFR-2 kinase inhibitor from Bristol-Myers Squibb, XL-647 (Exelixis), ZD-6474 (AstraZeneca), ZK-CDK (Schering AG), an EGFR tyrosine kinase inhibitor, EKB-569 (Wyeth), and an analogue or derivative thereof.
23497. The method of any one of items 23389-23394 wherein the agent is a VEGF inhibitor.
23498. The method of any one of items 23389-23394 wherein the agent is a vitamin D receptor agonist.
23499. The method of any one of items 23389-23394 wherein the agent is ZD-6474 (an angiogenesis inhibitor).
23500. The method of any one of items 23389-23394 wherein the agent is AP-23573 (an mTOR inhibitor).
23501. The method of any one of items 23389-23394 wherein the agent is synthadotin (a tubulin antagonist).
23502. The method of any one of items 23389-23394 wherein the agent is S-0885 (a collagenase inhibitor).
23503. The method of any one of items 23389-23394 wherein the agent is aplidine (an elongation factor-1 alpha inhibitor).
23504. The method of any one of items 23389-23394 wherein the agent is ixabepilone (an epithilone). 23505. The method of any one of items 23389-23394 wherein the agent is 1DN-5390 (an angiogenesis inhibitor).
23506. The method of any one of items 23389-23394 wherein the agent is SB-2723005 (an angiogenesis inhibitor).
23507. The method of any one of items 23389-23394 wherein the agent is ABT-518 (an angiogenesis inhibitor).
23508. The method of any one of items 23389-23394 wherein the agent is combretastatin (an angiogenesis inhibitor).
23509. The method of any one of items 23389-23394 wherein the agent is anecortave acetate (an angiogenesis inhibitor).
23510. The method of any one of items 23389-23394 wherein the agent is SB-715992 (a kinesin antagonist).
23511. The method of any one of items 23389-23394 wherein the agent is temsirolimus (an mTOR inhibitor).
23512. The method of any one of items 23389-23394 wherein the agent is adalimumab (a TNFα antagonist).
23513. The method of any one of items 23389-23394 wherein the composition comprises an antithrombotic agent.
23514. The method of any one of items 23389-23394 wherein the composition comprises a naturally occurring polymer. 23515. The method of any one of items 23389-23394 wherein the composition comprises protein.
23516. The method of any one of items 23389-23394 wherein the composition comprises carbohydrate.
23517. The method of any one of items 23389-23394 wherein the composition comprises biodegradable polymer.
23518. The method of any one of items 23389-23394 wherein the composition comprises nonbiodegradable polymer.
23519. The method of any one of items 23389-23394 wherein the composition comprises collagen.
23520. The method of any one of items 23389-23394 wherein the composition comprises methylated collagen.
23521. The method of any one of items 23389-23394 wherein the composition comprises fibrinogen.
23522. The method of any one of items 23389-23394 wherein the composition comprises thrombin.
23523. The method of any one of items 23389-23394 wherein the composition comprises blood plasma.
23524. The method of any one of items 23389-23394 wherein the composition comprises calcium salt. 23525. The method of any one of items 23389-23394 wherein the composition comprises an antifibronolytic agent.
23526. The method of any one of items 23389-23394 wherein the composition comprises fibrinogen analog.
23527. The method of any one of items 23389-23394 wherein the composition comprises albumin.
23528. The method of any one of items 23389-23394 wherein the composition comprises plasminogen.
23529. The method of any one of items 23389-23394 wherein the composition comprises von Willebrands factor.
23530. The method of any one of items 23389-23394 wherein the composition comprises Factor VIII.
23531. The method of any one of items 23389-23394 wherein the composition comprises hypoallergenic collagen.
23532. The method of any one of items 23389-23394 wherein the composition comprises atelopeptidic collagen.
23533. The method of any one of items 23389-23394 wherein the composition comprises telopeptide collagen.
23534. The method of any one of items 23389-23394 wherein the composition comprises crosslinked collagen. 23535. The method of any one of items 23389-23394 wherein the composition comprises aprotinin.
23536. The method of any one of items 23389-23394 wherein the composition comprises epsilon-amino-n-caproic acid.
23537. The method of any one of items 23389-23394 wherein the composition comprises gelatin.
23538. The method of any one of items 23389-23394 wherein the composition comprises protein conjugates.
23539. The method of any one of items 23389-23394 wherein the composition comprises gelatin conjugates.
23540. The method of any one of items 23389-23394 wherein the composition comprises a synthetic polymer.
23541. The method of any one of items 23389-23394 wherein the composition comprises a synthetic isocyanate-containing compound.
23542. The method of any one of items 23389-23394 wherein the composition comprises a synthetic thiol-containing compound.
23543. The method of any one of items 23389-23394 wherein the composition comprises a synthetic compound containing at least two thiol groups. 23544. The method of any one of items 23389-23394 wherein the composition comprises a synthetic compound containing at least three thiol groups.
23545. The method of any one of items 23389-23394 wherein the composition comprises a synthetic compound containing at least four thiol groups.
23546. The method of any one of items 23389-23394 wherein the composition comprises a synthetic amino-containing compound.
23547. The method of any one of items 23389-23394 wherein the composition comprises a synthetic compound containing at least two amino groups.
23548. The method of any one of items 23389-23394 wherein the composition comprises a synthetic compound containing at least three amino groups.
23549. The method of any one of items 23389-23394 wherein the composition comprises a synthetic compound containing at least four amino groups.
23550. The method of any one of items 23389-23394 wherein the composition comprises a synthetic compound comprising a carbonyl-oxygen-succinimidyl group.
23551. The method of any one of items 23389-23394 wherein the composition comprises a synthetic compound comprising at least two carbonyl-oxygen-succinimidyl groups. 23552. The method of any one of items 23389-23394 wherein the composition comprises a synthetic compound comprising at least three carbonyl-oxygen-succinimidyl groups.
23553. The method of any one of items 23389-23394 wherein the composition comprises a synthetic compound comprising at least four carbonyl-oxygen-succinimidyl groups.
23554. The method of any one of items 23389-23394 wherein the composition comprises a synthetic polyalkylene oxide- containing compound.
23555. The method of any one of items 23389-23394 wherein the composition comprises a synthetic compound comprising both polyalkylene oxide and biodegradable polyester blocks.
23556. The method of any one of items 23389-23394 wherein the composition comprises a synthetic polyalkylene oxide- containing compound having reactive amino groups.
23557. The method of any one of items 23389-23394 wherein the composition comprises a synthetic polyalkylene oxide- containing compound having reactive thiol groups.
23558. The method of any one of items 23389-23394 wherein the composition comprises a synthetic polyalkylene oxide- containing compound having reactive carbonyl-oxygen-succinimidyl groups.
23559. The method of any one of items 23389-23394 wherein the composition comprises a synthetic compound comprising a biodegradable polyester block. 23560. The method of any one of items 23389-23394 wherein the composition comprises a synthetic polymer formed in whole or part from lactic acid or lactide.
23561. The method of any one of items 23389-23394 wherein the composition comprises a synthetic polymer formed in whole or part from glycolic acid or glycolide.
23562. The method of any one of items 23389-23394 wherein the composition comprises polylysine.
23563. The method of any one of items 23389-23394 wherein the composition comprises (a) protein and (b) a compound comprising a polyalkylene oxide portion.
23564. The method of any one of items 23389-23394 wherein the composition comprises (a) protein and (b) polylysine.
23565. The method of any one of items 23389-23394 wherein the composition comprises (a) protein and (b) a compound having at least four thiol groups.
23566. The method of any one of items 23389-23394 wherein the composition comprises (a) protein and (b) a compound having at least four amino groups.
23567. The method of any one of items 23389-23394 wherein the composition comprises (a) protein and (b) a compound having at least four carbonyl-oxygen-succinirnide groups. 23568. The method of any one of items 23389-23394 wherein the composition comprises (a) protein and (b) a compound having a biodegradable region formed from reactants selected from lactic acid, lactide, glycolic acid, glycolide, and epison-caprolactone.
23569. The method of any one of items 23389-23394 wherein the composition comprises (a) collagen and (b) a compound comprising a polyalkylene oxide portion.
23570. The method of any one of items 23389-23394 wherein the composition comprises (a) collagen and (b) polylysine.
23571. The method of any one of items 23389-23394 wherein the composition comprises (a) collagen and (b) a compound having at least four thiol groups.
23572. The method of any one of items 23389-23394 wherein the composition comprises (a) collagen and (b) a compound having at least four amino groups.
23573. The method of any one of items 23389-23394 wherein the composition comprises (a) collagen and (b) a compound having at least four carbonyl-oxygen-succinimide groups.
23574. The method of any one of items 23389-23394 wherein the composition comprises (a) collagen and (b) a compound having a biodegradable region formed from reactants selected from lactic acid, lactide, glycolic acid, glycolide, and epison-caprolactone. 23575. The method of any one of items 23389-23394 wherein the composition comprises (a) methylated collagen and (b) a compound comprising a polyalkylene oxide portion.
23576. The method of any one of items 23389-23394 wherein the composition comprises (a) methylated collagen and (b) polylysine.
23577. The method of any one of items 23389-23394 wherein the composition comprises (a) methylated collagen and (b) a compound having at least four thiol groups.
23578. The method of any one of items 23389-23394 wherein the composition comprises (a) methylated collagen and (b) a compound having at least four amino groups.
23579. The method of any one of items 23389-23394 wherein the composition comprises (a) methylated collagen and (b) a compound having at least four carbonyl-oxygen-succinimide groups.
23580. The method of any one of items 23389-23394 wherein the composition comprises (a) methylated collagen and (b) a compound having a biodegradable region formed from reactants selected from lactic acid, lactide, glycolic acid, glycolide, and epison-caprolactone.
23581. The method of any one of items 23389-23394 wherein the composition comprises hyaluronic acid.
23582. The method of any one of items 23389-23394 wherein the composition comprises a hyaluronic acid derivative. 23583. The method of any one of items 23389-23394 wherein the composition comprises pentaerythritol poly(ethylene glycol)ether tetra-sulfhydryl of number average molecular weight between 3,000 and 30,000.
23584. The method of any one of items 23389-23394 wherein the composition comprises pentaerythritol poly(ethylene glycol)ether tetra-amino of number average molecular weight between 3,000 and 30,000.
23585. The method of any one of items 23389-23394 wherein the composition comprises (a) a synthetic compound having a number average molecular weight between 3,000 and 30,000 and comprising a polyalkylene oxide region and multiple nucleophilic groups, and (b) a synthetic compound having a number average molecular weight between 3,000 and 30,000 and comprising a polyalkylene oxide region and multiple electrophilic groups.
23586. The method of any one of items 23389-23394 wherein the composition comprises a colorant.
23587. The method of any one of items 23389-23394 wherein the composition is sterile.
23588. A method for treating vascular disease in a patient in need thereof, comprising delivering to the patient a) an anti-fibrotic agent or b) a composition comprising i) an anti-fibrotic agent and ii) a polymer and/or a compound that forms a polymer in situ.
23589. A method for treating stenosis in a patient in need thereof, comprising delivering to the patient a) an anti-fibrotic agent or b) a composition comprising i) an anti-fibrotic agent and ii) a polymer and/or a compound that forms a polymer in situ.
23590. A method for treating restenosis in a patient in need thereof, comprising delivering to the patient a) an anti-fibrotic agent or b) a composition comprising i) an anti-fibrotic agent and ii) a polymer and/or a compound that forms a polymer in situ.
23591. A method for treating atherosclerosis in a patient in need thereof, comprising delivering to the patient a) an anti-fibrotic agent or b) a composition comprising i) an anti-fibrotic agent and ii) a polymer and/or a compound that forms a polymer in situ.
23592. The method of any one of items 23588-23591 wherein the agent or composition is delivered perivascularly.
23593. The method of any one of items 23588-23591 wherein the agent is an adensosine A2A receptor antagonist.
23594. The method of any one of items 23588-23591 wherein the agent is an AKT inhibitor.
23595. The method of any one of items 23588-23591 wherein the agent is an alpha 2 integrin antagonist, wherein the alpha 2 integrin antagonist is Pharmaprojects No. 5754 (Merck KgaA).
23596. The method of any one of items 23588-23591 wherein the agent is an alpha 4 integrin antagonist.
23597. The method of any one of items 23588-23591 wherein the agent is an alpha 7 nicotinic receptor agonist. 23598. The method of any one of items 23588-23591 wherein the agent is an angiogenesis inhibitor selected from the group consisting of AG-12,958 (Pfizer), ATN-161 (Attention LLC), neovastat, an angiogenesis inhibitor from Jerina AG (Germany), NM-3 (Mercian), VGA- 1155 (Taisho), FCE-26644 (Pfizer), FCE-26950 (Pfizer), FPMA (Meiji Daries), FR-111142 (Fujisawa), GGTI-298, GM-1306 (Ligand), GPA-1734 (Novartis), NNC-47-0011 (Novo Nordisk), herbamycin (Nippon Kayaku), lenalidomide (Celegene), IP-10 (NIH), ABT-828 (Abbott), KIN-841 (Tokushima University, Japan), SF-1126 (Semafore Pharmaceuticals), laminin technology (NIH), CHIR-258 (Chiron), NVP-AEW541 (Novartis), NVP-AEW541 (Novartis), Vt16907 (Alchemia), OXI-8007 (Oxigene), EG- 3306 (Ark Therapeutics), Maspin (Arriva), ABT-567 (Abbott), PPI-2458 (Praecis Pharmaceuticals), CC-5079, CC-4089 (Celgene), HIF-1 alpha inhibitors (Xenova), S-247 (Pfizer), AP-23573 (Ariad), AZD-9935 (Astra Zeneca), mebendazole (Introgen Therapeutics), MetAP-2 inhibitors (GlaxoSmithKline), AG-615 (Angiogene Pharmaceuticals), Tie-2 antagonists (Hybrigenics), NC-381, CYC-381 , NC-169, NC-219, NC-383, NC-384, NC- 407 (Lorus Therapeutics), ATN-224 (Attenuon), ON-01370 (Onconova), Vitronectin antagonists (Amgen), SDX-103 (Salmedix), Vitronectin antagonists (Shire), CHP (Riemser), TEK (Amgen), Anecortave acetate (Alcon), T46.2 (Matrix Therapeutics), HG-2 (Heptagen), TEM antagonists (Genzyme), Oxi-4500 (Oxigene), ATN-161 (Attenuon), WX-293 (Wilex), M- 2025 (Metris Therapeutics), Alphastatin (BioActa), YH-16 (Yantai Rongchang), BIBF-1120 (Boehringer Ingelheim), BAY-57-9352 (Bayer), AS- 1404 (Cancer Research Technology), SC-77964 (Pfizer), glycomimetics (BioTie Therapies), TIE-2 Inhibitors (Ontogen), DIMI, Octamer (Octamer), ABR-215050 (Active Biotech), ABT-518 (Abbott), KDR inhibitors (Abbott), BSF-466895 (Abbott), SCH-221153 (Schering-Plough), DAC.antiangiogenic (ConjuChem), TFPI (EntreMed), AZD-2171 (Astra-Zenaca), CDC-394 (Celgene), LY290293 (EIi Lilly), IDN-5390 (Indena), Kdr Kinase Inhibitors (Merck), CT-113020, CT-116433, CT-116563, CT-31890, CT-32228) (Cell Therapeutics), A-299620 (Abbott), TWEAK Inhibitor (Amgen), VEGF modulators (Johnson and Johnson), Tum-N53, tumstatin (Genzyme), Thios- 1, Thios-2 (Thios Pharmaceuticals), MV-6401 (Miravant Medical Technologies), Spisulosine (PharmaMar), CEP-7055 (Cephalon), AUV-201 (Auvation), LM-609 (EIi Lilly), SKF-106615 (AnorMED), Oglufanide disodium (Cytran), BW-114 (Pharminox), Calreticulin (NIH), WX-678 (Wilex), SD-7784 (Pfizer), WX-UK1 (Wilex), SH-268 (Schering AG), 2-Me-PGA (Celgene), S- 137 (Pfizer), ZD-6126 (Angiogene Pharmaceuticals), SG-292 (SignalGen), Benefin (Lane Labs), A6, A36 (Angstrom), SB-2723005 (GlaxoSmithKline), SC-7 (Cell Therapeutics), ZEN-014 (AEterna Zentaris), 2-methoxyestradiol (EntreMed), NK-130119 (Nippon Kayaku), CC-10004 (Celgene), AVE- 8062A (Ajinomoto), Tacedinaline (Pfizer), Actinonin (Tokyo Metropolitan Institute of Medical Science), Lenalidomide (Celgene), VGA-1155, BTO-956 (SRI International), ER-68203-00 (Eisai), CT-6685 (UCB), JKC-362 (Phoenix Pharmaceuticals), DMI-3798 (DMI Biosciences, Angiomate (Ipsen), ZD-6474 (AstraZeneca), CEP-5214 (Cephalon), Canstatin - (Genzyme), NM-3 (Mercian), Oridigm (MediQuest Therapeutics), Exherin (Adherex), BLS-0597 (Boston Life Sciences), PTC-299 (PTC Therapeutics), NPI-2358 (Nereus Pharmaceuticals), CGP-79787 (Novartis), AEE-788 (Novartis), CKD-732 (Chong Kun Dang), CP-564959 (OSI Pharmaceuticals), CM-101 (CarboMed), CT-2584, CT3501 (Cell Therapeutics), combretastatin and analogues and derivatives thereof (Oxigene), Rebimastat (Bristol-Meyers Squibb), Dextrin 2-sulfate (ML Laboratories), Cilengitide (Merk KGaA), NSC-706704 (Pharminox), KRN- 951 (Kirin Brewery), Ukrain, NSC-631570 (Nowicky Pharma), Tecogalan sodium (Daiichi Pharmaceutical), Tz-93 (Tsumura), TBC-1635 (Encysive Pharmaceuticals), TAN-1120 (Takeda), Semaxanib (Pfizer), BDI-7800 (Biopharmacopae), SD-186, SD-983 (Bristol-Meyers Squibb), SB-223245 (GlaxoSmithKline), SC-236 (Pfizer), RWJ-590973 (Johnson and Johnson), ILX-1850 (Genzyme), SC-68488, S-836 (Pfizer), CG-55069-11 (CuraGen), Ki-23057 (Kirin Brewery), CCX-700 (Chemoentryx), Pegaptanib octasodium (Giled Sciences), ANGIOCOL (available from Biostratum Inc.), or an analogue or derivative thereof.
23599. The method of any one of items 23588-23591 wherein the agent is an apoptosis antagonist.
23600. The method of any one of items 23588-23591 wherein the agent is an apoptosis activator.
23601. The method of any one of items 23588-23591 wherein the agent is a beta 1 integrin antagonist.
23602. The method of any one of items 23588-23591 wherein the agent is a beta tubulin inhibitor.
23603. The method of any one of items 23588-23591 wherein the agent is a blocker of enzyme production in Hepatitis C.
23604. The method of any one of items 23588-23591 wherein the agent is a Bruton's tyrosine kinase inhibitor.
23605. The method of any one of items 23588-23591 wherein the agent is a calcineurin inhibitor.
23606. The method of any one of items 23588-23591 wherein the agent is a caspase 3 inhibitor.
23607. The method of any one of items 23588-23591 wherein the agent is a CC chemokine receptor antagonist. 23608. The method of any one of items 23588-23591 wherein the agent is a cell cycle inhibitor selected from the group consisting of SNS-595 (Sunesis), synthadotin, KRX-0403, and an analogue or derivative thereof.
23609. The method of any one of items 23588-23591 wherein the agent is a cathepsin B inhibitor.
23610. The method of any one of items 23588-23591 wherein the agent is a cathepsin K inhibitor, wherein the cathepsin K inhibitor is 462795 (GlaxoSmithKline), INPL-022-D6 (Amura Therapeutics), or an analogue or derivative thereof.
23611. The method of any one of items 23588-23591 wherein the agent is a cathepsin L inhibitor.
23612. The method of any one of items 23588-23591 wherein the agent is a CD40 antagonist.
23613. The method of any one of items 23588-23591 wherein the agent is a chemokine receptor agonist.
23614. The method of any one of items 23588-23591 wherein the agent is a chymase inhibitor.
23615. The method of any one of items 23588-23591 wherein the agent is a collagenase antagonist.
23616. The method of any one of items 23588-23591 wherein the agent is a CXCR antagonist. 23617. The method of any one of items 23588-23591 wherein the agent is a cyclin dependent kinase inhibitor selected from the group consisting of a CDK-1 inhibitor, a CDK-2 inhibitor, a CDK- 4 inhibitor, a CDK-6 inhibitor, a CAK1 inhibitor from GPC Biotech and Bristol-Myers Squibb, RGB-286199 (GPC Biotech), an anticancer agent from Astex Technology, a CAK1 inhibitor from GPC Biotech, a CDK inhibitor from Sanofi-Aventis, a CDK1/CDK2 inhibitor from Amgen, a CDK2 inhibitor from SUGEN-2 (Pfizer), a hearing loss therapy agent (Sound Pharmaceuticals), PD-0332991 (Pfizer), RGB-286199 (GPC Biotech), Ro-0505124 (Hoffmann- La Roche), a Ser/Thr kinase inhibitor from Lilly (EIi Lilly), CVT-2584 (CAS No. 199986-75-9) (CV Therapeutics), and an analogue or derivative thereof.
23618. The method of any one of items 23588-23591 wherein the agent is a cyclooxygenase 1 inhibitor.
23619. The method of any one of items 23588-23591 wherein the agent is a DHFR inhibitor.
23620. The method of any one of items 23588-23591 wherein the agent is a dual integrin inhibitor.
23621. The method of any one of items 23588-23591 wherein the agent is an elastase inhibitor.
23622. The method of any one of items 23588-23591 wherein the agent is an elongation factor-1 alpha inhibitor.
23623. The method of any one of items 23588-23591 wherein the agent is an endothelial growth factor antagonist. 23624. The method of any one of items 23588-23591 wherein the agent is an endothelial growth factor receptor kinase inhibitor selected from the group consisting of sorafenib tosylate (Bayer), AAL-993 (Novartis), ABP-309 (Novartis), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EXEL- 2880 (Exelixis), GW-654652 (GlaxoSmithKline), a KDR inhibitor from LG Life Sciences, CT-6685 and CT-6729 (UCB)1 KRN-633 and KRN-951 (Kirin Brewery), OSI-930 (OSI Pharmaceuticals), SP-5.2 (Supratek Pharma), SU-
11657 (Pfizer), a Tie-2 antagonist (Hybrigenics), a VEGFR-2 kinase inhibitor (Bristol-Myers Squibb), XL-647 (Exelixis), a KDR inhibitor from Abbott Laboratories, sorafenib tosylate, and an analogue or derivative thereof.
23625. The method of any one of items 23588-23591 wherein the agent is an endotoxin antagonist.
23626. The method of any one of items 23588-23591 wherein the agent is an epothilone and tubulin binder.
23627. The method of any one of items 23588-23591 wherein the agent is an estrogen receptor antagonist.
23628. The method of any one of items 23588-23591 wherein the agent is an FGF inhibitor.
23629. The method of any one of items 23588-23591 wherein the agent is a farnexyl transferase inhibitor.
23630. The method of any one of items 23588-23591 wherein the agent is famesyltransferase inhibitor selected from the group of A-197574 (Abbott), a famesyltransferase inhibitor from Servier, FPTIII (Strathclyde Institute for Drug R), LB-42908 (LG Life Sciences), Pharmaprojects No. 5063 (Genzyme), Pharmaprojects No. 5597 (Ipsen), Yissum Project No. B-1055 (Yissum), and an analogue or derivative thereof.
23631. The method of any one of items 23588-23591 wherein the agent is an FLT-3 kinase inhibitor.
23632. The method of any one of items 23588-23591 wherein the agent is an FGF receptor kinase inhibitor.
23633. The method of any one of items 23588-23591 wherein the agent is a fibrinogen antagonist selected from the group consisting of AUV-201 (Auvation), MG-13926 (Sanofi-Aventis), plasminogen activator (CAS No. 105913-11-9) (from Sanofi-Aventis or UCB), plasminogen activator-2 (tPA-2) (Sanofi-Aventis), pro-urokinase (CAS No. 82657-92-9) (Sanofi-Aventis), and an analogue or derivative thereof.
23634. The method of any one of items 23588-23591 wherein the agent is a heat shock protein 90 antagonist selected from the group consisting of SRN-005 (Sirenade), geldanamycin, NSC-33050 (17- allylaminogeldanamycin; 17-AAG), 17-dimethylaminoethylamino-17- demethoxy-geldanamycin (17-DMAG), rifabutin (rifamycin XIV, 1',4- didehydro-1-deoxy-1 ,4-dihydro-5'-(2-methylpropyl)-1-oxo-), and an analogue or derivative thereof.
23635. The method of any one of items 23588-23591 wherein the agent is a histone deacetylase inhibitor.
23636. The method of any one of items 23588-23591 wherein the agent is an HMGCoA reductase inhibitor selected from the group consisting of an atherosclerosis therapeutic from Lipid Sciences, ATI- 16000 (ARYx Therapeutics), KS-01-019 (Kos Pharmaceuticals), Pharmaprojects No. 2197 (Sanofi-Aventi), RP 61969 (Sanofi-Aventis), and an analogue or derivative thereof.
23637. The method of any one of items 23588-23591 wherein the agent is an ICAM inhibitor.
23638. The method of any one of items 23588-23591 wherein the agent is an IL, ICE and IRAK antagonist, wherein the antagonist is a CJ-14877, CP-424174 (Pfizer), NF-61 (Negma-Lerads), and an analogue or derivative thereof.
23639. The method of any one of items 23588-23591 wherein the agent is an IL-2 inhibitor.
23640. The method of any one of items 23588-23591 wherein the agent is an immunosuppressant selected from the group consisting of teriflunomide (Sanofi Aventis), chlorsulfaquinoxalone (NSC- 339004), chlorsulfaquinoxalone sulfate, CS-712 (Sankyo), ismomultin alfa (CAS No. 457913-93-8) (Akzo Nobel), antiallergics from GenPat77, antiinflammatories or AT-005 (Androclus Therapeutics), autoimmune disease therapeutics from EpiVax, BN-007 (Bone), budesonide (CAS No. 51333-22- 3) (MAP Pharmaceuticals), CO-14 (Genzyme), edratide (CAS No. 433922- 67-9) (Teva), EP-314 (Enanta), eprovafen (CAS No. 101335-99-3) (Sanofi- Aventis), HWA-131 (CAS No. 118788-41-3) (Sanofi-Aventis), immunomodulators from MerLion Pharmaceuticals, immunosuppressives from Alchemia, IPL-12 (Inflazyme), MDL-9563 (CAS No. 27086-86-8) (Sanofi-Aventis), Pharmaprojects No. 2330 (Sanofi-Aventis), Pharmaprojects No. 6426 (Abgenix), PXS-25 (Pharmaxis), rosmarinic acid (CAS No. 20283-92-5) (Sanofi-Aventis), RP 42927 or RP 54745 (CAS No. 135330-08-4) (Sanofi-Aventis), SGN-35 (Seattle Genetics), ST-1959 (Sigma-Tau), type I diabetes therapy from SYNX Pharma, UNIL-88 (Debiopharm), VP-025 (Vasogen), VR-694 (Vectura), PRTX-001 (Protalex), and an analogue or derivative thereof.
23641. The method of any one of items 23588-23591 wherein the agent is an IMPDH (inosine monophosphate).
23642. The method of any one of items 23588-23591 wherein the agent is an integrin antagonist.
23643. The method of any one of items 23588-23591 wherein the agent is an interleukin antagonist.
23644. The method of any one of items 23588-23591 wherein the agent is an inhibitor of type III receptor tyrosine kinase.
23645. The method of any one of items 23588-23591 - wherein the agent is an irreversible inhibitor of enzyme methionine aminopeptidase type 2.
23646. The method of any one of items 23588-23591 wherein the agent is an isozyme selective delta protein kinase C inhibitor.
23647. The method of any one of items 23588-23591 wherein the agent a JAK3 enzyme inhibitor.
23648. The method of any one of items 23588-23591 wherein the agent is a JNK inhibitor.
23649. The method of any one of items 23588-23591 wherein the agent is a kinase inhibitor. 23650. The method of any one of items 23588-23591 wherein the agent is kinesin antagonist.
23651. The method of any one of items 23588-23591 wherein the agent is a kinesin antagonist.
23652. The method of any one of items 23588-23591 wherein the agent is a leukotriene inhibitor and antagonist selected from the group consisting of ambicromil (CAS No. 58805-38-2) (Sanofi-Aventis), amelubant (CAS No. 346735-24-8) (Boehringer Ingelheim), DW-1141 (Dong Wha), ebselen (Daiichi Pharmaceutical), ibudilast (Kyorin), leucotriene inhibitors from Sanofi-Aventis, lymphotoxin-beta receptor (LT-D) from Biogen Idee, Pharmaprojects No. 1535 and 2728 (CAS No. 119340-33-9) (Sanofi-Aventis), R-112 (Rigel), Rev-5367 (CAS No. 92532-05-3) (Sanofi- Aventis), RG-14893 (CAS No. 141835-49-6) (Sanofi-Aventis), RG-5901-A (CAS No. 101910-24-1), 92532-23-5, RP 66153 (CAS No. 142422-79-5), RP 66364 (CAS No.186916-92-5), RP 69698 (CAS No. 141748-00-7) (Sanofi-Aventis), SC-411930 (Pfizer), SC-41930 (CAS No. 120072-59-5) (Pfizer), SC-50605 (CAS No. 138828-39-4) (Pfizer), SC-51146 (CAS No. 141059-52-1), SC-53228 (CAS No. 153633-01-3) (Pfizer), spaglumic acid (ZY-15106) (CAS No.3106-85-2), 80619-64-3 (Novartis), tipredane (CAS No. 85197-77-9) (Bristol-Myers Squibb), U-75302 (CAS No. 119477-85-9) (Pfizer), and analogue or derivative thereof.
23653. The method of any one of items 23588-23591 wherein the agent is an MAP kinase inhibitor.
23654. The method of any one of items 23588-23591 wherein the agent is a matrix metalloproteinase inhibitor. 23655. The method of any one of items 23588-23591 wherein the agent is an MCP-CCR2 inhibitor.
23656. The method of any one of items 23588-23591 wherein the agent is an mTOR inhibitor.
23657. The method of any one of items 23588-23591 wherein the agent is an mTOR kinase inhibitor.
23658. The method of any one of items 23588-23591 wherein the agent is a microtubule inhibitor selected from the group consisting of antibody-maytansinoid conjugates from Biogen Idee, colchicines (MantiCore Pharmaceuticals), anticancer immunoconjugates from Johnson & Johnson, DIME from Octamer, gni-1f (GNI)1 huC242-DM4, huMy9-6-DM1 (ImmunoGen), IDN-5404 (Indena), IMO-098, IMOderm (Imotep), mebendazole (Introgen Therapeutics), microtubule poisons from Cambridge Enterprise, paclitaxel such as LOTAX from Aphios (CAS No. 33069-62-4), Genexol-PM from Samyang, Pharmaprojects No. 6383 (Tapestry Pharmaceuticals), RPR-112378 (Sanofi-Aventis), SGN-75 (Seattle Genetics), SPL-7435 (Starpharma), SSR-250411 (Sanofi-Aventis), trastuzumab-DM1 (Genentech), vinorelbine, and an analogue or derivative thereof.
23659. The method of any one of items 23588-23591 wherein the agent is an MIF inhibitor.
23660. The method of any one of items 23588-23591 wherein the agent is an MMP inhibitor.
23661. The method of any one of items 23588-23591 wherein the agent is a neurokinin (NK) antagonist selected from the group consisting of anthrotainin (CAS No. 148084-40-6) (Sanofi-Aventis), an IBS thereapeutic from ArQuIe, MDL-105212A (CAS No. 167261-60-1) (Ssanofi- Aventis), Pharmaprojects No. 2744, 3258 (CAS No. 139167-47-8) 4006, 4201, or 5986 (Sanofi-Aventis), RP 67580 (CAS No. 135911-02-3), SR- 144190 (CAS No. 201152-86-5), SSR-240600, SSR-241586 (Sanofi- Aventis), TKA-457 (Novartis), vestipitant mesylate (CAS No. 334476-64-1) (GlaxoSmithKline), Win-64821 (Sanofi-Aventis), PRX-96026 (Predix Pharmaceuticals), and an analogue or derivative thereof.
23662. The method of any one of items 23588-23591 wherein the agent is an NF kappa B inhibitor selected from the group consisting of AVE-0545 or AVE-0547 (Sanofi-Aventis), bortezomib (CAS No. 179324-69-7) (Millennium Pharmaceuticals), dexanabinol (CAS No. 112924- 45-5) (Pharmos), dexlipotam (Viatris), Pharmaprojects No. 6283 (INDRA) (OXiGENE), IPL-576092 (CAS No. 137571-30-3) (Inflazyme), NFKB decoy (Corgentech), NFKB decoy oligo (AnGes MG), NFKB's from Ariad, osteoporosis treatments or S5 (F005) from Fulcrum Pharmaceuticals, P61 (Phytopharm), R-flurbiprofen (CAS No. 5104-49-4) (Encore Pharmaceuticals), and an analogue or derivative thereof.
23663. The method of any one of items 23588-23591 wherein the agent is a nitric oxide agonist.
23664. The method of any one of items 23588-23591 wherein the agent is an ornithine decarboxylase inhibitor.
23665. The method of any one of items 23588-23591 wherein the agent is a p38 MAP kinase inhibitor selected from the group consisting of AZD-6703 (AstraZeneca), JX-401 (Jexys Pharmaceuticals), BMS-2 (Bristol-Myers Squibb), a p38 MAP kinase inhibitor from Novartis, a p38-alpha MAP kinase inhibitor from Amphora, Pharmaprojects No. 5704 (Pharmacopeia), RPR-200765A (Sanofi-Aventis), SD-282 (Johnson & Johnson), TAK-715 (Takeda), and an analogue or derivative thereof.
23666. The method of any one of items 23588-23591 wherein the agent is a palmitoyl-protein thioesterase inhibitor.
23667. The method of any one of items 23588-23591 wherein the agent is a PDGF receptor kinase inhibitor selected from the group consisting of AAL-993, AMN-107, or ABP-309 (Novartis), AMG-706 (Amgen), BAY-57-9352 (Bayer), CDP-860 (UCB), E-7080 (Eisai), imatinib (CAS No. 152459-95-5) (Novartis), OSI-930 (OSI Pharmaceuticals), RPR- 127963E (Sanofi-Aventis), RWJ-540973 (Johnson & Johnson), sorafenib tosylate (Bayer), SU-11657 (Pfizer), tandutinib (CAS No. 387867-13-2) (Millennium Pharmaceuticals), vatalanib (Novartis), ZK-CDK (Schering AG), and an analogue or derivative thereof.
23668. The method of any one of items 23588-23591 wherein the agent is (-)-halofenate (Metabolex), AMG-131 (Amgen), antidiabetics from Japan Tobacco, AZD-4619, AZD-8450, AZD-8677 (AstraZeneca), DRF-10945, balaglitazone (Dr Reddy's), CS-00088, CS- 00098 (Chipscreen Biosciences), E-3030 (Eisai), etalocib (CAS No. 161172- 51-6) (EIi Lilly), GSK-641597 (Ligand), GSK-677954 (GlaxoSmithKline), GW-409544 (Ligand), GW-590735 (GlaxoSmithKline), K-111 (Hoffmann-La Roche), LY-518674 (EIi Lilly), LY-674 (Ligand), LY-929 (Ligand), MC-3001 , MC-3002 (MaxoCore Pharmaceuticals), metformin HCI + pioglitazone (CAS No. 1115-70-4 and 112529-15-4), ACTOPLUS MET from Andrx), muraglitazar (CAS No. 331741-94-7) (Bristol-Myers Squibb), naveglitazar (Ligand), oleoylethanolamide (Kadmus Pharmaceuticals), ONO-5129, pioglitazone hydrochloride (CAS No. 111025-46-8 and 112529-15-4) (takeda), PLX-204 (Plexxikon), PPAR agonists from Genfit, PPAR delta agonists from EIi Lilly, PPAR-alpha agonists from CrystalGenomics, PPAR- gamma modulators and PPAR-β modulators from CareX, rosiglitazone maleate (CAS No. 122324-73-4 or 155141-29-0) (GlaxoSmithKline), rosiglitazone maleate/glimepir (CAS No. 155141-29-0 and 93479-97-1), AVANDARYL, rosiglitazone maleate/metformin extend (CAS No. 155141- 29-0 and 657-24-9), AVANDAMET, rosiglitazone maleate+metformin, AVANDAMET (GlaxoSmithKline), tesaglitazar (AstraZeneca), and an analogue or derivative thereof.
23669. The method of any one of items 23588-23591 wherein the agent is a phosphatase inhibitor.
23670. The method of any one of items 23588-23591 wherein the agent is a phosphodiesterase (PDE) inhibitor selected from the group consisting of avanafil (Tanabe Seiyaku), dasantafil (CAS No. 569351- 91-3) (Schering-Plough), A-906119 (CAS No. 134072-58-5), DL-850 (Sanofi-Aventis), GRC-3015, GRC-3566, GRC-3886 (Glenmark), HWA-153 (CAS No. 56395-66-5) (Sanofi-Aventis), hydroxypumafentrine (Altana), IBFB-130011 , IBFB-14-016, IBFB-140301 , IBFB-150007, IBFB-211913 (IBFB Pharma), L-826141 (Merck & Co), medorinone (CAS No. 88296-61-1) (Sanofi-Aventis), MEM-1917 (Memory Pharmaceuticals), ND-1251 (Neuro3d), PDE inhibitors from ICOS, PDE IV inhibitors from Memory Pharmaceuticals and CrystalGenomics, Pharmaprojects No. 2742 and 6141 (Sanofi-Aventis), QAD-171 (Novartis), RHC-2963 (CAS No. 76993-12-9 and 76993-14-1), RPR-117658, RPR-122818 derivatives, SR-24870 , and RPR- 132294 (Sanofi-Aventis), SK-350 (ln2Gen), stroke therapy agents from deCODE Genetics, TAS-203 (Taiho), tofimilast (CAS No. 185954-27-2) (Pfizer), UK-371800 (Pfizer), WIN-65579 (CAS No. 158020-82-7) (Sanofi- Aventis), IBFB-130020 (IBFB Pharma), OPC-6535 (CAS No. 145739-56-6) (Otsuka), a phosphodiesterase III inhibitor, enoximone, a phosphodiesterase IV inhibitor, fosfosal, Atopik (Barrier Therapeutics), triflusal, a phosphodiesterase V inhibitor, and an analogue or derivative thereof.
23671. The method of any one of items 23588-23591 wherein the agent is a PKC inhibitor.
23672. The method of any one of items 23588-23591 wherein the agent is a platelet activating factor antagonist.
23673. The method of any one of items 23588-23591 wherein the agent is a platelet-derived growth factor receptor kinase inhibitor.
23674. The method of any one of items 23588-23591 wherein the agent is a prolyl hydroxylase inhibitor.
23675. The method of any one of items 23588-23591 wherein the agent is a polymorphonuclear neutrophil inhibitor.
23676. The method of any one of items 23588-23591 wherein the agent is a protein kinase B inhibitor.
23677. The method of any one of items 23588-23591 wherein the agent is a protein kinase C stimulant.
23678. The method of any one of items 23588-23591 wherein the agent is a purine nucleoside analogue.
23679. The method of any one of items 23588-23591 wherein the agent is a purinoreceptor P2X antagonist. 23680. The method of any one of items 23588-23591 wherein the agent is a Raf kinase inhibitor.
23681. The method of any one of items 23588-23591 wherein the agent is a reversible inhibitor of ErbB1 and ErbB2.
23682. The method of any one of items 23588-23591 wherein the agent is a ribonucleoside triphosphate reductase inhibitor.
23683. The method of any one of items 23588-23591 wherein the agent is an SDF-1 antagonist.
23684. The method of any one of items 23588-23591 wherein the agent is a sheddase inhibitor.
23685. The method of any one of items 23588-23591 wherein the agent is an SRC inhibitor.
23686. The method of any one of items 23588-23591 wherein the agent is a stromelysin inhibitor.
23687. The method of any one of items 23588-23591 wherein the agent is an Syk kinase inhibitor.
23688. The method of any one of items 23588-23591 wherein the agent is a telomerase inhibitor.
23689. The method of any one of items 23588-23591 wherein the agent is a TGF beta inhibitor selected from the group consisting of pirfenidone (CAS No. 53179-13-8) (MARNAC), tranilast (CAS No. 53902- 12-8) (Kissei), IN-1130 (ln2Gen), mannose-6-phosphate (BTG), TGF-β antagonists from Inflazyme (Pharmaprojects No. 6075), TGF-β antagonists from Sydney, non-industrial source), TGF-β I receptor kinase inhibitors from EIi Lilly, TGF-β receptor inhibitors from Johnson & Johnson, and an analogue or derivative thereof.
23690. The method of any one of items 23588-23591 wherein the agent is a TNFα antagonist or TACE inhibitor selected from the group consisting of adalimumab (CAS No. 331731-18-1) (Cambridge Antibody Technology), AGIX-4207 (AtheroGenics), AGT-1 (Advanced Biotherapy), an anti-inflammatory from Borean Pharma, Cellzome, or Paradigm Therapeutics, anti-inflammatory vaccine (TNF-alpha kinoid) from Neovacs, humanized anti-TNF antibody or an anti-TNF MAb (CB0006) Celltech (UCB), apratastat (CAS No. 287445-51-0) (Wyeth), BMS-561392 (Bristol-Myers Squibb), BN-006 (Bone), certolizumab pegol (CAS No. 428863-50-7 or CH-138 (UCB), cilomilast (CAS No. 153259-65-5) (GlaxoSmithKline), CR-1 (Nuada Pharmaceuticals), CRx^119 (CombinatoRx), D-5410 (UCB), dacopafant (CAS No. 125372-33-0) (Sanofi- Aventis), dersalazine (CAS No. 188913-57-7/188913-58-8) (Uriach), etanercept (CAS No. 185243-69-0) (Amgen), ethyl pyruvate (Critical (Critical Therapeutics), golimumab (CAS No. 476181-74-5) (Johnson & Johnson), hormono-immunotherapy from Ipsen, CDP571 {e.g., Humicade from UCB), IC-485 (ICOS), infliximab (CAS No. 170277-31-3) (Johnson & Johnson), IP- 751 (Manhattan Pharmaceuticals), ISIS-104838 (CAS No. 250755-32-9) (ISIS Pharmaceuticals), lenalidomide (CAS No. 191732-72-6) (Celgene), lentinan (CAS No. 37339-90-5) (Ajinomoto), MDL-201112 (CAS No. 142130-73-2) (Sanofi-Aventis), medroxyprogesterone (CAS No. 520-85-4) (InKine Pharmaceutical), N-acetylcysteine (CAS No. 616-91-1) (Zambon), NBE-P2 (DIREVO Biotech), nerelimomab (CAS No. 162774-06-3) (Chiron), OM-294DP (OM PHARMA), onercept (CAS No. 199685-57-9) (Yeda), PASSTNF-alpha (Verigen), pentoxifylline or oxypentifylline (Sanofi-Aventis), Pharmaprojects No. 4091, 4241 , 4295, or 4488 (Sanofi-Aventis), Pharmaprojects No. 5480 (Amgen), Pharmaprojects No. 6749 (Cengent), pirfenidone (CAS No. 53179-13-8) (MARNAC)1 RPR-132294 (Sanofi- Aventis), S5 (F002) (Fulcrum Pharmaceuticals), simvastatin (CAS No. 79902-63-9) (Merck & Co), STA-6292 (Synta Pharmaceuticals), tacrolimus (CAS No. 104987-11-3) (Fujisawa LifeCycle Pharma), talactoferrin alfa (CAS No. 308240-58-6) (Agennix), thalidomide (CAS No. 50-35-1) (Celgene), TNF antagonists form ProStrakan, and Synergen, TNF inhibitors (Amgen), TNF-alpha antagonists from Dynavax Technologies and Jerina AG (Germany), TNF-alpha inhibitors from IBFB Pharma and Xencor (Xencor), torbafylline (CAS No. 105102-21-4) (Sanofi-Aventis), UR-1505 (Uriach), VT-346 (Viron Therapeutics), YSIL6 (Y1S Therapeutics), YSTH2 (Y's Therapeutics), NPI-1302a-3 (Nereus Pharmaceuticals, a TNF antagonist from Jerina AG (Germany), dersalazine, and an analogue or derivative thereof.
23691. The method of any one of items 23588-23591 wherein the agent is a Toll receptor inhibitor.
23692. The method of any one of items 23588-23591 wherein the agent is a tubulin antagonist.
23693. The method of any one of items 23588-23591 wherein the agent is a tyrosine kinase inhibitor selected from the group consisting of SU-011248, SUTENT from Pfizer Inc. (New York, NY)1 BMS- 354825, PN-355 (Paracelsian Pharmaceuticals), AGN-199659 (Allergan), AAL-993 or ABP-309 (Novartis), adaphostin (NIH), AEE-788 (Novartis), AG- 013736 (OSI Pharmaceuticals), AG-13736 (Pfizer), ALT-110 (Alteris Therapeutics), AMG-706 (Amgen), anticancer MAbs from Xencor, anti- EGFrvlll MAbs from Abgenix, anti-HER2 MAb from Abiogen, AZD-2171 or AZD-9935 (AstraZeneca), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), CEP-5214 (Cephalon), CEP-7055 (Cephalon), cetuximab (ImCIone Systems), CHIR-200131 and CHIR-258 (Chiron), CP-547632 (OSI Pharmaceuticals), CP-724714 (Pfizer), CT-301 (Creabilis Therapeutics), D- 69491 (Baxter International), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EGFR/ErbB2 inhibitors from Array BioPharma, erlotinib (CAS No. 183319- 69-9) (OSI Pharmaceuticals), EXEL-2880 (Exelixis), FK-778 (Sanofi- Aventis) , gefitinib (CAS No. 184475-35-2) (AstraZeneca), GW-2286 or GW- 654652 (GlaxoSmithKline), her2/neu antigen from AlphaVax, HER-2/neu inhibitor from Generex, Herzyme (Medipad) (Sirna Therapeutics), HKI-272 (Wyeth), HuMax-EGFr (Genmab), idronoxil (CAS No. 81267-65-4) (Novogen), IGF-1 inhibitors from Ontogen, IMC-11F8 (ImCIone Systems), kahalalide F (CAS No. 149204-42-2) (PharmaMar), KDR inhibitor from LG Life Sciences, KDR inhibitors from Abbott Laboratories, KDR kinase inhibitors (UCB), Kdr kinase inhibitors from Merck & Co, KRN-633 and KRN- 951 (Kirin Brewery), KSB-102 (Xenova), lapatinib ditosylate (CAS No. 388082-78-8) (GlaxoSmithKline), matuzumab (Merck KGaA), MDX-214 (Medarex), ME-103 (Pharmexa), MED-A300 (Gerolymatos), MNAC-13 (Lay Line Genomics), nimotuzumab (Center of Molecular Immunology), NSC- 330507 or NSC-707545 (NIH), NV-50 (Novogen), OSI-930 (OSI Pharmaceuticals), panitumumab (Abgenix), pelitinib (CAS No. 287933-82-7) (Wyeth), pertuzumab (CAS No. 380610-27-5) (Genentech), Pharmaprojects No. 3985 (Sanofi-Aventis), prostate cancer therapeutics from Sequenom (SQPC35, SQPC36, SQPC90), removab and remoxab (Trion Pharma), RG- 13022 (CAS No. 136831-48-6), RG-13291 (CAS No. 138989-50-1), or RG- 14620 (CAS No. 136831-49-7) (Sanofi-Aventis), RM-6427 (Romark), RNAi breast cancer therapy from Benitec, RP 53801 (CAS No. 125882-88-4) (Sanofi-Aventis), sorafenib tosylate (Bayer), SU-11657 (Pfizer), Tie-2 antagonists from Semaia (Hybrigenics), Tie-2 inhibitors from Ontogen, trastuzumab (CAS No. 180288-69-1) (Genentech), tyrosine kinase inhibitors from Sanofi-Aventis, U3-1287, U3-1565, U3-1784, U3-1800 (U3 Pharma), vatalanib (Novartis), VEGFR-2 kinase inhibitor from Bristol-Myers Squibb, XL-647 (Exelixis), ZD-6474 (AstraZeneca), ZK-CDK (Schering AG), an EGFR tyrosine kinase inhibitor, EKB-569 (Wyeth), and an analogue or derivative thereof.
23694. The method of any one of items 23588-23591 wherein the agent is a VEGF inhibitor.
23695. The method of any one of items 23588-23591 wherein the agent is a vitamin D receptor agonist.
23696. The method of any one of items 23588-23591 wherein the agent is ZD-6474 (an angiogenesis inhibitor).
23697. The method of any one of items 23588-23591 wherein the agent is AP-23573 (an mTOR inhibitor).
23698. The method of any one of items 23588-23591 wherein the agent is synthadotin (a tubulin antagonist).
23699. The method of any one of items 23588-23591 wherein the agent is S-0885 (a collagenase inhibitor).
23700. The method of any one of items 23588-23591 wherein the agent is aplidine (an elongation factor-1 alpha inhibitor).
23701. The method of any one of items 23588-23591 wherein the agent is ixabepilone (an epithilone).
23702. The method of any one of items 23588-23591 wherein the agent is IDN-5390 (an angiogenesis inhibitor). 23703. The method of any one of items 23588-23591 wherein the agent is SB-2723005 (an angiogenesis inhibitor).
23704. The method of any one of items 23588-23591 wherein the agent is ABT-518 (an angiogenesis inhibitor).
23705. The method of any one of items 23588-23591 wherein the agent is combretastatin (an angiogenesis inhibitor).
23706. The method of any one of items 23588-23591 wherein the agent is anecortave acetate (an angiogenesis inhibitor).
23707. The method of any one of items 23588-23591 wherein the agent is SB-715992 (a kinesin antagonist).
23708. The method of any one of items 23588-23591 wherein the agent is temsirolimus (an mTOR inhibitor).
23709. The method of any one of items 23588-23591 wherein the agent is adalimumab (a TNFα antagonist).
23710. The method of any one of items 23588-23591 wherein the composition comprises an anti-thrombotic agent.
23711. The method of any one of items 23588-23591 wherein the composition comprises a naturally occurring polymer.
23712. The method of any one of items 23588-23591 wherein the composition comprises protein. 23713. The method of any one of items 23588-23591 wherein the composition comprises carbohydrate.
23714. The method of any one of items 23588-23591 wherein the composition comprises biodegradable polymer.
23715. The method of any one of items 23588-23591 wherein the composition comprises nonbiodegradable polymer.
23716. The method of any one of items 23588-23591 wherein the composition comprises collagen.
23717. The method of any one of items 23588-23591 wherein the composition comprises methylated collagen.
23718. The method of any one of items 23588-23591 wherein the composition comprises fibrinogen.
23719. The method of any one of items 23588-23591 wherein the composition comprises thrombin.
23720. The method of any one of items 23588-23591 wherein the composition comprises blood plasma.
23721. The method of any one of items 23588-23591 wherein the composition comprises calcium salt.
23722. The method of any one of items 23588-23591 wherein the composition comprises an antifibronolytic agent. 23723. The method of any one of items 23588-23591 wherein the composition comprises fibrinogen analog.
23724. The method of any one of items 23588-23591 wherein the composition comprises albumin.
23725. The method of any one of items 23588-23591 wherein the composition comprises plasminogen.
23726. The method of any one of items 23588-23591 wherein the composition comprises von Willebrands factor.
23727. The method of any one of items 23588-23591 wherein the composition comprises Factor VIII.
23728. The method of any one of items 23588-23591 wherein the composition comprises hypoallergenic collagen.
23729. The method of any one of items 23588-23591 wherein the composition comprises atelopeptidic collagen.
23730. The method of any one of items 23588-23591 wherein the composition comprises telopeptide collagen.
23731. The method of any one of items 23588-23591 wherein the composition comprises crosslinked collagen.
23732. The method of any one of items 23588-23591 wherein the composition comprises aprotinin. 23733. The method of any one of items 23588-23591 wherein the composition comprises epsilon-amino-n-caproic acid.
23734. The method of any one of items 23588-23591 wherein the composition comprises gelatin.
23735. The method of any one of items 23588-23591 wherein the composition comprises protein conjugates.
23736. The method of any one of items 23588-23591 wherein the composition comprises gelatin conjugates.
23737. The method of any one of items 23588-23591 wherein the composition comprises a synthetic polymer.
23738. The method of any one of items 23588-23591 wherein the composition comprises a synthetic isocyanate-containing compound.
23739. The method of any one of items 23588-23591 wherein the composition comprises a synthetic thiol-containing compound.
23740. The method of any one of items 23588-23591 wherein the composition comprises a synthetic compound containing at least two thiol groups.
23741. The method of any one of items 23588-23591 wherein the composition comprises a synthetic compound containing at least three thiol groups. 23742. The method of any one of items 23588-23591 wherein the composition comprises a synthetic compound containing at least four thiol groups.
23743. The method of any one of items 23588-23591 wherein the composition comprises a synthetic amino-containing compound.
23744. The method of any one of items 23588-23591 wherein the composition comprises a synthetic compound containing at least two amino groups.
23745. The method of any one of items 23588-23591 wherein the composition comprises a synthetic compound containing at least three amino groups.
23746. The method of any one of items 23588-23591 wherein the composition comprises a synthetic compound containing at least four amino groups.
23747. The method of any one of items 23588-23591 wherein the composition comprises a synthetic compound comprising a carbonyl-oxygen-succinimidyl group.
23748. The method of any one of items 23588-23591 wherein the composition comprises a synthetic compound comprising at least two carbonyl-oxygen-succinimidyl groups.
23749. The method of any one of items 23588-23591 wherein the composition comprises a synthetic compound comprising at least three carbonyl-oxygen-succinimidyl groups. 23750. The method of any one of items 23588-23591 wherein the composition comprises a synthetic compound comprising at least four carbonyl-oxygen-succinimidyl groups.
23751. The method of any one of items 23588-23591 wherein the composition comprises a synthetic polyalkylene oxide- containing compound.
23752. The method of any one of items 23588-23591 wherein the composition comprises a synthetic compound comprising both polyalkylene oxide and biodegradable polyester blocks.
23753. The method of any one of items 23588-23591 wherein the composition comprises a synthetic polyalkylene oxide- containing compound having reactive amino groups.
23754. The method of any one of items 23588-23591 wherein the composition comprises a synthetic polyalkylene oxide- containing compound having reactive thiol groups.
23755. The method of any one of items 23588-23591 wherein the composition comprises a synthetic polyalkylene oxide- containing compound having reactive carbonyl-oxygen-succinimidyl groups.
23756. The method of any one of items 23588-23591 wherein the composition comprises a synthetic compound comprising a biodegradable polyester block.
23757. The method of any one of items 23588-23591 wherein the composition comprises a synthetic polymer formed in whole or part from lactic acid or lactide. 23758. The method of any one of items 23588-23591 wherein the composition comprises a synthetic polymer formed in whole or part from glycolic acid or glycolide.
23759. The method of any one of items 23588-23591 wherein the composition comprises polylysine.
23760. The method of any one of items 23588-23591 wherein the composition comprises (a) protein and (b) a compound comprising a polyalkylene oxide portion.
23761. The method of any one of items 23588-23591 wherein the composition comprises (a) protein and (b) polylysine.
23762. The method of any one of items 23588-23591 wherein the composition comprises (a) protein and (b) a compound having at least four thiol groups.
23763. The method of any one of items 23588-23591 wherein the composition comprises (a) protein and (b) a compound having at least four amino groups.
23764. The method of any one of items 23588-23591 wherein the composition comprises (a) protein and (b) a compound having at least four carbonyl-oxygen-succinimide groups.
23765. The method of any one of items 23588-23591 wherein the composition comprises (a) protein and (b) a compound having a biodegradable region formed from reactants selected from lactic acid, lactide, glycolic acid, glycolide, and epison-caprolactone. 23766. The method of any one of items 23588-23591 wherein the composition comprises (a) collagen and (b) a compound comprising a polyalkylene oxide portion.
23767. The method of any one of items 23588-23591 wherein the composition comprises (a) collagen and (b) polylysine.
23768. The method of any one of items 23588-23591 wherein the composition comprises (a) collagen and (b) a compound having at least four thiol groups.
23769. The method of any one of items 23588-23591 wherein the composition comprises (a) collagen and (b) a compound having at least four amino groups.
23770. The method of any one of items 23588-23591 wherein the composition comprises (a) collagen and (b) a compound having at least four carbonyl-oxygen-succinimide groups.
23771. The method of any one of items 23588-23591 wherein the composition comprises (a) collagen and (b) a compound having a biodegradable region formed from reactants selected from lactic acid, lactide, glycolic acid, glycolide, and epison-caprolactone.
23772. The method of any one of items 23588-23591 wherein the composition comprises (a) methylated collagen and (b) a compound comprising a polyalkylene oxide portion.
23773. The method of any one of items 23588-23591 wherein the composition comprises (a) methylated collagen and (b) polylysine. 23774. The method of any one of items 23588-23591 wherein the composition comprises (a) methylated collagen and (b) a compound having at least four thiol groups.
23775. The method of any one of items 23588-23591 wherein the composition comprises (a) methylated collagen and (b) a compound having at least four amino groups.
23776. The method of any one of items 23588-23591 wherein the composition comprises (a) methylated collagen and (b) a compound having at least four carbonyl-oxygen-succinimide groups.
23777. The method of any one of items 23588-23591 wherein the composition comprises (a) methylated collagen and (b) a compound having a biodegradable region formed from reactants selected from lactic acid, lactide, glycolic acid, glycolide, and epison-caprolactone.
23778. The method of any one of items 23588-23591 wherein the composition comprises hyaluronic acid.
23779. The method of any one of items 23588-23591 wherein the composition comprises a hyaluronic acid derivative.
23780. The method of any one of items 23588-23591 wherein the composition comprises pentaerythritol poly(ethylene glycol)ether tetra-sulfhydryl of number average molecular weight between 3,000 and 30,000.
23781. The method of any one of items 23588-23591 wherein the composition comprises pentaerythritol poly(ethylene glycol)ether tetra-amino of number average molecular weight between 3,000 and 30,000.
23782. The method of any one of items 23588-23591 wherein the composition comprises (a) a synthetic compound having a number average molecular weight between 3,000 and 30,000 and comprising a polyalkylene oxide region and multiple nucleophilic groups, and (b) a synthetic compound having a number average molecular weight between 3,000 and 30,000 and comprising a polyalkylene oxide region and multiple electrophilic groups.
23783. The method of any one of items 23588-23591 wherein the composition comprises a colorant.
23784. The method of any one of items 23588-23591 wherein the composition is sterile.
23785. A composition comprising i) an anti-fibrotic agent and ii) a polymer or a compound that forms a polymer in situ.
23786. The composition of item 23785 wherein the agent is an adensosine A2A receptor antagonist.
23787. The composition of item 23785 wherein the agent is an AKT inhibitor.
23788. The composition of item 23785 wherein the agent is an alpha 2 integrin antagonist, wherein the alpha 2 integrin antagonist is Pharmaprojects No. 5754 (Merck KgaA). 23789. The composition of item 23785 wherein the agent is an alpha 4 integrin antagonist.
23790. The composition of item 23785 wherein the agent is an alpha 7 nicotinic receptor agonist.
23791. The composition of item 23785 wherein the agent is an angiogenesis inhibitor selected from the group consisting of AG- 12,958 (Pfizer), ATN-161 (Attenuon LLC), neovastat, an angiogenesis inhibitor from Jerina AG (Germany), NM-3 (Mercian), VGA-1155 (Taisho), FCE-26644 (Pfizer), FCE-26950 (Pfizer), FPMA (Meiji Daries), FR-111142 (Fujisawa), GGTI-298, GM-1306 (Ligand), GPA-1734 (Novartis), NNC-47- 0011 (Novo Nordisk), herbamycin (Nippon Kayaku), lenalidomide (Celegene), IP-10 (NIH), ABT-828 (Abbott), KIN-841 (Tokushima University, Japan), SF-1126 (Semafore Pharmaceuticals), laminin technology (NIH), CHIR-258 (Chiron), NVP-AEW541 (Novartis), NVP-AEW541 (Novartis), Vt16907 (Alchemia), OXI-8007 (Oxigene), EG-3306 (Ark Therapeutics), Maspin (Arriva), ABT-567 (Abbott), PPI-2458 (Praecis Pharmaceuticals), CC-5079, CC-4089 (Celgene), HIF-1 alpha inhibitors (Xenova), S-247 (Pfizer), AP-23573 (Ariad), AZD-9935 (Astra Zeneca), mebendazole (Introgen Therapeutics), MetAP-2 inhibitors (GlaxoSmithKline), AG-615 (Angiogene Pharmaceuticals), Tie-2 antagonists (Hybrigenics), NC-381, CYC-381, NC-169, NC-219, NC-383, NC-384, NC-407 (Lorus Therapeutics), ATN-224 (Attenuon), ON-01370 (Onconova), Vitronectin antagonists (Amgen), SDX-103 (Salmedix), Vitronectin antagonists (Shire), CHP (Riemser), TEK (Amgen), Anecortave acetate (Alcon), T46.2 (Matrix Therapeutics), HG-2 (Heptagen), TEM antagonists (Genzyme), Oxi-4500 (Oxigene), ATN-161 (Attenuon), WX-293 (Wilex), M-2025 (Metris Therapeutics), Alphastatin (BioActa), YH-16 (Yantai Rongchang), BIBF- 1120 (Boehringer Ingelheim), BAY-57-9352 (Bayer), AS-1404 (Cancer Research Technology), SC-77964 (Pfizer), glycomimetics (BioTie Therapies), TIE-2 Inhibitors (Ontogen), DIMI, Octamer (Octamer), ABR- 215050 (Active Biotech), ABT-518 (Abbott), KDR inhibitors (Abbott), BSF- 466895 (Abbott), SCH-221153 (Schering-Plough), DAC.antiangiogenic (ConjuChem), TFPI (EntreMed), AZD-2171 (Astra-Zenaca), CDC-394 (Celgene), LY290293 (EIi Lilly), IDN-5390 (Indena), Kdr Kinase Inhibitors (Merck), CT-113020, CT-116433, CT-116563, CT-31890, CT-32228) (Cell Therapeutics), A-299620 (Abbott), TWEAK Inhibitor (Amgen), VEGF modulators (Johnson and Johnson), Tum-N53, tumstatin (Genzyme), Thios- 1, Thios-2 (Thios Pharmaceuticals), MV-6401 (Miravant Medical Technologies), Spisulosine (PharmaMar), CEP-7055 (Cephalon), AUV-201 (Auvation), LM-609 (EIi Lilly), SKF-106615 (AnorMED), Oglufanide disodium (Cytran), BW-114 (Pharminox), Calreticulin (NIH), WX-678 (Wilex), SD-7784 (Pfizer), WX-UK1 (Wilex), SH-268 (Schering AG), 2-Me-PGA (Celgene), S- 137 (Pfizer), ZD-6126 (Angiogene Pharmaceuticals), SG-292 (SignalGen), Benefin (Lane Labs), A6, A36 (Angstrom), SB-2723005 (GlaxoSmithKline), SC-7 (Cell Therapeutics), ZEN-014 (AEterna Zentaris), 2-methoxyestradiol (EntreMed), NK-130119 (Nippon Kayaku), CC-10004 (Celgene), AVE- 8062A (Ajinomoto), Tacedinaline (Pfizer), Actinonin (Tokyo Metropolitan Institute of Medical Science), Lenalidomide (Celgene), VGA-1155, BTO-956 (SRI International), ER-68203-00 (Eisai), CT-6685 (UCB), JKC-362 (Phoenix Pharmaceuticals), DMI-3798 (DMI Biosciences, Angiomate (Ipsen), ZD-6474 (AstraZeneca), CEP-5214 (Cephalon), Canstatin (Genzyme), NM-3 (Mercian), Oridigm (MediQuest Therapeutics), Exherin (Adherex), BLS-0597 (Boston Life Sciences), PTC-299 (PTC Therapeutics), NPI-2358 (Nereus Pharmaceuticals), CGP-79787 (Novartis), AEE-788 (Novartis), CKD-732 (Chong Kun Dang), CP-564959 (OSI Pharmaceuticals), CM-101 (CarboMed), CT-2584, CT3501 (Cell Therapeutics), combretastatin and analogues and derivatives thereof (Oxigene), Rebimastat (Bristol-Meyers Squibb), Dextrin 2-sulfate (ML Laboratories), Cilengitide (Merk KGaA), NSC-706704 (Pharminox), KRN- 951 (Kirin Brewery), Ukrain, NSC-631570 (Nowicky Pharma), Tecogalan sodium (Daiichi Pharmaceutical), Tz-93 (Tsumura), TBC-1635 (Encysive Pharmaceuticals), TAN-1120 (Takeda), Semaxanib (Pfizer), BDI-7800 (Biopharmacopae), SD-186, SD-983 (Bristol-Meyers Squibb), SB-223245 (GlaxoSmithKline), SC-236 (Pfizer), RWJ-590973 (Johnson and Johnson), ILX-1850 (Genzyme), SC-68488, S-836 (Pfizer), CG-55069-11 (CuraGen), Ki-23057 (Kirin Brewery), CCX-700 (Chemoentryx), Pegaptanib octasodium (Giled Sciences), ANGIOCOL (available from Biostratum Inc.), or an analogue or derivative thereof.
23792. The composition of item 23785 wherein the agent is an apoptosis antagonist.
23793. The composition of item 23785 wherein the agent is an apoptosis activator.
23794. The composition of item 23785 wherein the agent is a beta 1 integrin antagonist.
23795. The composition of item 23785 wherein the agent is a beta tubulin inhibitor.
23796. The composition of item 23785 wherein the agent is a blocker of enzyme production in Hepatitis C.
23797. The composition of item 23785 wherein the agent is a Bruton's tyrosine kinase inhibitor.
23798. The composition of item 23785 wherein the agent is a calcineurin inhibitor. 23799. The composition of item 23785 wherein the agent is a caspase 3 inhibitor.
23800. The composition of item 23785 wherein the agent is a CC chemokine receptor antagonist.
23801. The composition of item 23785 wherein the agent is a cell cycle inhibitor selected from the group consisting of SNS-595 (Sunesis), synthadotin, KRX-0403, and an analogue or derivative thereof.
23802. The composition of item 23785 wherein the agent is a cathepsin B inhibitor.
23803. The composition of item 23785 wherein the agent is a cathepsin K inhibitor, wherein the cathepsin K inhibitor is 462795 (GlaxoSmithKline), INPL-022-D6 (Amura Therapeutics), or an analogue or derivative thereof.
23804. The composition of item 23785 wherein the agent is a cathepsin L inhibitor.
23805. The composition of item 23785 wherein the agent is a CD40 antagonist.
23806. The composition of item 23785 wherein the agent is a chemokine receptor agonist.
23807. The composition of item 23785 wherein the agent is a chymase inhibitor. 23808. The composition of item 23785 wherein the agent is a collagenase antagonist.
23809. The composition of item 23785 wherein the agent is a CXCR antagonist.
23810. The composition of item 23785 wherein the agent is a cyclin dependent kinase inhibitor selected from the group consisting of a CDK-1 inhibitor, a CDK-2 inhibitor, a CDK- 4 inhibitor, a CDK-6 inhibitor, a CAK1 inhibitor from GPC Biotech and Bristol-Myers Squibb, RGB-286199 (GPC Biotech), an anticancer agent from Astex Technology, a CAK1 inhibitor from GPC Biotech, a CDK inhibitor from Sanofi-Aventis, a CDK1/CDK2 inhibitor from Amgen, a CDK2 inhibitor from SUGEN-2 (Pfizer), a hearing loss therapy agent (Sound Pharmaceuticals), PD-0332991 (Pfizer), RGB-286199 (GPC Biotech), Ro-0505124 (Hoffmann- La Roche), a Ser/Thr kinase inhibitor from Lilly (EIi Lilly), CVT-2584 (CAS No. 199986-75-9) (CV Therapeutics), and an analogue or derivative thereof.
23811. The composition of item 23785 wherein the agent is a cyclooxygenase 1 inhibitor.
23812. The composition of item 23785 wherein the agent is a DHFR inhibitor.
23813. The composition of item 23785 wherein the agent is a dual integrin inhibitor.
23814. The composition of item 23785 wherein the agent is an elastase inhibitor. 23815. The composition of item 23785 wherein the agent is an elongation factor-1 alpha inhibitor.
23816. The composition of item 23785 wherein the agent is an endothelial growth factor antagonist.
23817. The composition of item 23785 wherein the agent is an endothelial growth factor receptor kinase inhibitor selected from the group consisting of sorafenib tosylate (Bayer), AAL-993 (Novartis), ABP- 309 (Novartis), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), E- 7080 (Eisai), EG-3306 (Ark Therapeutics), EXEL-2880 (Exelixis), GW- 654652 (GlaxoSmithKline), a KDR inhibitor from LG Life Sciences, CT-6685 and CT-6729 (UCB), KRN-633 and KRN-951 (Kirin Brewery), OSI-930 (OSI Pharmaceuticals), SP-5.2 (Supratek Pharma), SU-11657 (Pfizer), a Tie-2 antagonist (Hybrigenics), a VEGFR-2 kinase inhibitor (Bristol-Myers Squibb), XL-647 (Exelixis), a KDR inhibitor from Abbott Laboratories, sorafenib tosylate, and an analogue or derivative thereof.
23818. The composition of item 23785 wherein the agent is an endotoxin antagonist.
23819. The composition of item 23785 wherein the agent is an epothilone and tubulin binder.
23820. The composition of item 23785 wherein the agent is an estrogen receptor antagonist.
23821. The composition of item 23785 wherein the agent is an FGF inhibitor. 23822. The composition of item 23785 wherein the agent is a famexyl transferase inhibitor.
23823. The composition of item 23785 wherein the agent is famesyltransferase inhibitor selected from the group of A-197574 (Abbott), a famesyltransferase inhibitor from Servier, FPTIIl (Strathclyde Institute for Drug R), LB-42908 (LG Life Sciences), Pharmaprojects No. 5063 (Genzyme), Pharmaprojects No. 5597 (Ipsen), Yissum Project No. B- 1055 (Yissum), and an analogue or derivative thereof.
23824. The composition of item 23785 wherein the agent is an FLT-3 kinase inhibitor.
23825. The composition of item 23785 wherein the agent is an FGF receptor kinase inhibitor.
23826. The composition of item 23785 wherein the agent is a fibrinogen antagonist selected from the group consisting of AUV- 201 (Auvation), MG-13926 (Sanofi-Aventis), plasminogen activator (CAS No. 105913-11-9) (from Sanofi-Aventis or UCB), plasminogen activator-2 (tPA-2) (Sanofi-Aventis), pro-urokinase (CAS No. 82657-92-9) (Sanofi- Aventis), and an analogue or derivative thereof.
23827. The composition of item 23785 wherein the agent is a heat shock protein 90 antagonist selected from the group consisting of SRN-005 (Sirenade), geldanamycin, NSC-33050 (17- allylaminogeldanamycin; 17-AAG), 17-dimethylaminoethylamino-17- demethoxy-geldanamycin (17-DMAG), rifabutin (rifamycin XIV, 1',4- didehydro-1-deoxy-1 ,4-dihydro-5'-(2-methylpropyl)-1-oxo-), and an analogue or derivative thereof. 23828. The composition of item 23785 wherein the agent is a histone deacetylase inhibitor.
23829. The composition of item 23785 wherein the agent is an HMGCoA reductase inhibitor selected from the group consisting of an atherosclerosis therapeutic from Lipid Sciences, ATI-16000 (ARYx Therapeutics), KS-01-019 (Kos Pharmaceuticals), Pharmaprojects No. 2197 (Sanofi-Aventi), RP 61969 (Sanofi-Aventis), and an analogue or derivative thereof.
23830. The composition of item 23785 wherein the agent is an ICAM inhibitor.
23831. The composition of item 23785 wherein the agent is an IL, ICE and IRAK antagonist, wherein the antagonist is a CJ- 14877, CP-424174 (Pfizer), NF-61 (Negma-Lerads), and an analogue or derivative thereof.
23832. The composition of item 23785 wherein the agent is an IL-2 inhibitor.
23833. The composition of item 23785 wherein the agent is an immunosuppressant selected from the group consisting of teriflunomide (Sanofi Aventis), chlorsulfaquinoxalone (NSC-339004), chlorsulfaquinoxalone sulfate, CS-712 (Sankyo), ismomultin alfa (CAS No. 457913-93-8) (Akzo Nobel), antiallergics from GenPat77, antiinflammatories or AT-005 (Androclus Therapeutics), autoimmune disease therapeutics from EpiVax, BN-007 (Bone), budesonide (CAS No. 51333-22- 3) (MAP Pharmaceuticals), CO-14 (Genzyme), edratide (CAS No. 433922- 67-9) (Teva), EP-314 (Enanta), eprovafen (CAS No. 101335-99-3) (Sanofi- Aventis), HWA-131 (CAS No. 118788-41-3) (Sanofi-Aventis), immunomodulators from MerLion Pharmaceuticals, immunosuppressives from Alchemia, IPL-12 (Inflazyme), MDL-9563 (CAS No. 27086-86-8) (Sanofi-Aventis), Pharmaprojects No. 2330 (Sanofi-Aventis), Pharmaprojects No. 6426 (Abgenix), PXS-25 (Pharmaxis), rosmarinic acid (CAS No. 20283-92-5) (Sanofi-Aventis), RP 42927 or RP 54745 (CAS No. 135330-08-4) (Sanofi-Aventis), SGN-35 (Seattle Genetics), ST-1959 (Sigma-Tau), type I diabetes therapy from SYNX Pharma, UNIL-88 (Debiopharm), VP-025 (Vasogen), VR-694 (Vectura), PRTX-001 (Protalex), and an analogue or derivative thereof.
23834. The composition of item 23785 wherein the agent is an IMPDH (inosine monophosphate).
23835. The composition of item 23785 wherein the agent is an integrin antagonist.
23836. The composition of item 23785 wherein the agent is an interleukin antagonist.
23837. The composition of item 23785 wherein the agent is an inhibitor of type III receptor tyrosine kinase.
23838. The composition of item 23785 wherein the agent is an irreversible inhibitor of enzyme methionine aminopeptidase type 2.
23839. The composition of item 23785 wherein the agent is an isozyme selective delta protein kinase C inhibitor.
23840. The composition of item 23785 wherein the agent a JAK3 enzyme inhibitor. 23841. The composition of item 23785 wherein the agent is a JNK inhibitor.
23842. The composition of item 23785 wherein the agent is a kinase inhibitor.
23843. The composition of item 23785 wherein the agent is kinesin antagonist.
23844. The composition of item 23785 wherein the agent is a kinesin antagonist.
23845. The composition of item 23785 wherein the agent is a leukotriene inhibitor and antagonist selected from the group consisting of ambicromil (CAS No. 58805-38-2) (Sanofi-Aventis), amelubant (CAS No. 346735-24-8) (Boehringer Ingelheim), DW-1141 (Dong Wha), ebselen (Daiichi Pharmaceutical), ibudilast (Kyorin), leucotriene inhibitors from Sanofi-Aventis, lymphotoxin-beta receptor (LT-D) from Biogen Idee, Pharmaprojects No. 1535 and 2728 (CAS No. 119340-33-9) (Sanofi- Aventis), R-112 (Rigel), Rev-5367 (CAS No. 92532-05-3) (Sanofi-Aventis), RG-14893 (CAS No. 141835-49-6) (Sanofi-Aventis), RG-5901-A (CAS No. 101910-24-1), 92532-23-5, RP 66153 (CAS No. 142422-79-5), RP 66364 (CAS No.186916-92-5), RP 69698 (CAS No. 141748-00-7) (Sanofi-Aventis), SC-411930 (Pfizer), SC-41930 (CAS No. 120072-59-5) (Pfizer), SC-50605 (CAS No. 138828-39-4) (Pfizer), SC-51146 (CAS No. 141059-52-1), SC- 53228 (CAS No. 153633-01-3) (Pfizer), spaglumic acid (ZY-15106) (CAS No.3106-85-2), 80619-64-3 (Novartis), tipredane (CAS No. 85197-77-9) (Bristol-Myers Squibb), U-75302 (CAS No. 119477-85-9) (Pfizer), and analogue or derivative thereof. 23846. The composition of item 23785 wherein the agent is an MAP kinase inhibitor.
23847. The composition of item 23785 wherein the agent is a matrix metalloproteinase inhibitor.
23848. The composition of item 23785 wherein the agent is an MCP-CCR2 inhibitor.
23849. The composition of item 23785 wherein the agent is an mTOR inhibitor.
23850. The composition of item 23785 wherein the agent is an mTOR kinase inhibitor.
23851. The composition of item 23785 wherein the agent is a microtubule inhibitor selected from the group consisting of antibody-maytansinoid conjugates from Biogen Idee, colchicines (MantiCore Pharmaceuticals), anticancer immunoconjugates from Johnson & Johnson, DIME from Octamer, gni-1f (GNI), huC242-DM4, huMy9-6-DM1 (ImmunoGen), IDN-5404 (Indena), IMO-098, IMOderm (Imotep), mebendazole (Introgen Therapeutics), microtubule poisons from Cambridge Enterprise, paclitaxel such as LOTAX from Aphios (CAS No. 33069-62-4), Genexol-PM from Samyang, Pharmaprojects No. 6383 (Tapestry Pharmaceuticals), RPR-112378 (Sanofi-Aventis), SGN-75 (Seattle Genetics), SPL-7435 (Starpharma), SSR-250411 (Sanofi-Aventis), trastuzumab-DM1 (Genentech), vinorelbine, and an analogue or derivative thereof.
23852. The composition of item 23785 wherein the agent is an MIF inhibitor. 23853. The composition of item 23785 wherein the agent is an MMP inhibitor.
23854. The composition of item 23785 wherein the agent is a neurokinin (NK) antagonist selected from the group consisting of anthrotainin (CAS No. 148084-40-6) (Sanofi-Aventis), an IBS thereapeutic from ArQuIe, MDL-105212A (CAS No. 167261-60-1) (Ssanofi-Aventis), Pharmaprojects No. 2744, 3258 (CAS No. 139167-47-8) 4006, 4201, or 5986 (Sanofi-Aventis), RP 67580 (CAS No. 135911-02-3), SR-144190 (CAS No. 201152-86-5), SSR-240600, SSR-241586 (Sanofi-Aventis), TKA-457 (Novartis), vestipitant mesylate (CAS No. 334476-64-1) (GlaxoSmithKline), Win-64821 (Sanofi-Aventis), PRX-96026 (Predix Pharmaceuticals), and an analogue or derivative thereof.
23855. The composition of item 23785 wherein the agent is an NF kappa B inhibitor selected from the group consisting of AVE- 0545 or AVE-0547 (Sanofi-Aventis), bortezomib (CAS No. 179324-69-7) (Millennium Pharmaceuticals), dexanabinol (CAS No. 112924-45-5) (Pharmos), dexlipotam (Viatris), Pharmaprojects No. 6283 (INDRA) (OXiGENE), IPL-576092 (CAS No. 137571-30-3) (Inflazyme), NFKB decoy (Corgentech), NFKB decoy oligo (AnGes MG), NFKB's from Ariad, osteoporosis treatments or S5 (F005) from Fulcrum Pharmaceuticals, P61 (Phytopharm), R-flurbiprofen (CAS No. 5104-49-4) (Encore Pharmaceuticals), and an analogue or derivative thereof.
23856. The composition of item 23785 wherein the agent is a nitric oxide agonist.
23857. The composition of item 23785 wherein the agent is an ornithine decarboxylase inhibitor. 23858. The composition of item 23785 wherein the agent is a p38 MAP kinase inhibitor selected from the group consisting of AZD-6703 (AstraZeneca), JX-401 (Jexys Pharmaceuticals), BMS-2 (Bristol- Myers Squibb), a p38 MAP kinase inhibitor from Novartis, a p38-alpha MAP kinase inhibitor from Amphora, Pharmaprojects No. 5704 (Pharmacopeia), RPR-200765A (Sanofi-Aventis), SD-282 (Johnson & Johnson), TAK-715 (Takeda), and an analogue or derivative thereof.
23859. The composition of item 23785 wherein the agent is a palmitoyl-protein thioesterase inhibitor.
23860. The composition of item 23785 wherein the agent is a PDGF receptor kinase inhibitor selected from the group consisting of AAL-993, AMN-107, or ABP-309 (Novartis), AMG-706 (Amgen), BAY-57- 9352 (Bayer), CDP-860 (UCB), E-7080 (Eisai), imatinib (CAS No. 152459- 95-5) (Novartis), OSI-930 (OSI Pharmaceuticals), RPR-127963E (Sanofi- . Aventis), RWJ-540973 (Johnson & Johnson), sorafenib tosylate (Bayer), SU-11657 (Pfizer), tandutinib (CAS No. 387867-13-2) (Millennium Pharmaceuticals), vatalanib (Novartis), ZK-CDK (Schering AG), and an analogue or derivative thereof.
23861. The composition of item 23785 wherein the agent is (-)-halofenate (Metabolex), AMG-131 (Amgen), antidiabetics from Japan Tobacco, AZD-4619, AZD-8450, AZD-8677 (AstraZeneca), DRF- 10945, balaglitazone (Dr Reddy's), CS-00088, CS-00098 (Chipscreen Biosciences), E-3030 (Eisai), etalocib (CAS No. 161172-51-6) (EIi Lilly), GSK-641597 (Ligand), GSK-677954 (GlaxoSmithKline), GW-409544 (Ligand), GW-590735 (GlaxoSmithKline), K-111 (Hoffmann-La Roche), LY- 518674 (EIi Lilly), LY-674 (Ligand), LY-929 (Ligand), MC-3001, MC-3002 (MaxoCore Pharmaceuticals), metformin HCI + pioglitazone (CAS No. 1115- 70-4 and 112529-15-4), ACTOPLUS MET from Andrx), muraglitazar (CAS No. 331741-94-7) (Bristol-Myers Squibb), naveglitazar (Ligand), oleoylethanolamide (Kadmus Pharmaceuticals), ONO-5129, pioglitazone hydrochloride (CAS No. 111025-46-8 and 112529-15-4) (Takeda), PLX-204 (Plexxikon), PPAR agonists from Genfit, PPAR delta agonists from EIi Lilly, PPAR-alpha agonists from CrystalGenomics, PPAR-gamma modulators and PPAR-β modulators from CareX, rosiglitazone maleate (CAS No. 122324- 73-4 or 155141-29-0) (GlaxoSmithKline), rosiglitazone maleate/glimepir (CAS No. 155141-29-0 and 93479-97-1), AVANDARYL1 rosiglitazone maleate/metformin extend (CAS No. 155141-29-0 and 657-24-9), AVANDAMET1 rosiglitazone maleate+metformin, AVANDAMET (GlaxoSmithKline), tesaglitazar (AstraZeneca), and an analogue or derivative thereof.
23862. The composition of item 23785 wherein the agent is a phosphatase inhibitor.
23863. The composition of item 23785 wherein the agent is a phosphodiesterase (PDE) inhibitor selected from the group consisting of avanafil (Tanabe Seiyaku), dasantafil (CAS No. 569351-91-3) (Schering-Plough), A-906119 (CAS No. 134072-58-5), DL-850 (Sanofi- Aventis), GRC-3015, GRC-3566, GRC-3886 (Glenmark), HWA-153 (CAS No. 56395-66-5) (Sanofi-Aventis), hydroxypumafentrine (Altana), IBFB- 130011, IBFB-14-016, IBFB-140301, IBFB-150007, IBFB-211913 (IBFB Pharma), L-826141 (Merck & Co), medorinone (CAS No. 88296-61-1) (Sanofi-Aventis), MEM-1917 (Memory Pharmaceuticals), ND-1251 (Neuro3d), PDE inhibitors from ICOS, PDE IV inhibitors from Memory Pharmaceuticals and CrystalGenomics, Pharmaprojects No. 2742 and 6141 (Sanofi-Aventis), QAD-171 (Novartis), RHC-2963 (CAS No. 76993-12-9 and 76993-14-1), RPR-117658, RPR-122818 derivatives, SR-24870 , and RPR- 132294 (Sanofi-Aventis), SK-350 (ln2Gen), stroke therapy agents from deCODE Genetics, TAS-203 (Taiho), tofimilast (CAS No. 185954-27-2) (Pfizer), UK-371800 (Pfizer), WIN-65579 (CAS No. 158020-82-7) (Sanofi- Aventis), IBFB-130020 (IBFB Pharma), OPC-6535 (CAS No. 145739-56-6) (Otsuka), a phosphodiesterase III inhibitor, enoximone, a phosphodiesterase IV inhibitor, fosfosal, Atopik (Barrier Therapeutics), triflusal, a phosphodiesterase V inhibitor, and an analogue or derivative thereof.
23864. The composition of item 23785 wherein the agent is a PKC inhibitor.
23865. The composition of item 23785 wherein the agent is a platelet activating factor antagonist.
23866. The composition of item 23785 wherein the agent is a platelet-derived growth factor receptor kinase inhibitor.
23867. The composition of item 23785 wherein the agent is a prolyl hydroxylase inhibitor.
23868. The composition of item 23785 wherein the agent is a polymorphonuclear neutrophil inhibitor.
23869. The composition of item 23785 wherein the agent is a protein kinase B inhibitor.
23870. The composition of item 23785 wherein the agent is a protein kinase C stimulant.
23871. The composition of item 23785 wherein the agent is a purine nucleoside analogue. 23872. The composition of item 23785 wherein the agent is a purinoreceptor P2X antagonist.
23873. The composition of item 23785 wherein the agent is a Raf kinase inhibitor.
23874. The composition of item 23785 wherein the agent is a reversible inhibitor of ErbB1 and ErbB2.
23875. The composition of item 23785 wherein the agent is a ribonucleoside triphosphate reductase inhibitor.
23876. The composition of item 23785 wherein the agent is an SDF-1 antagonist.
23877. The composition of item 23785 wherein the agent is a sheddase inhibitor.
23878. The composition of item 23785 wherein the agent is an SRC inhibitor.
23879. The composition of item 23785 wherein the agent is a stromelysin inhibitor.
23880. The composition of item 23785 wherein the agent is an Syk kinase inhibitor.
23881. The composition of item 23785 wherein the agent is a telomerase inhibitor. 23882. The composition of item 23785 wherein the agent is a TGF beta inhibitor selected from the group consisting of pirfenidone (CAS No. 53179-13-8) (MARNAC), tranilast (CAS No. 53902- 12-8) (Kissei), IN-1130 (ln2Gen), mannose-6-phosphate (BTG), TGF-β antagonists from Inflazyme (Pharmaprojects No. 6075), TGF-β antagonists from Sydney, non-industrial source), TGF-β I receptor kinase inhibitors from EIi Lilly, TGF-β receptor inhibitors from Johnson & Johnson, and an analogue or derivative thereof.
23883. The composition of item 23785 wherein the agent is a TNFα antagonist or TACE inhibitor selected from the group consisting of adalimumab (CAS No. 331731-18-1) (Cambridge Antibody Technology), AGIX-4207 (AtheroGenics), AGT-1 (Advanced Biotherapy), an anti-inflammatory from Borean Pharma, Cellzome, or Paradigm Therapeutics, anti-inflammatory vaccine (TNF-alpha kinoid) from Neovacs, humanized anti-TNF antibody or an anti-TNF MAb (CB0006) Celltech (UCB), apratastat (CAS No. 287445-51-0) (Wyeth), BMS-561392 (Bristol- Myers Squibb), BN-006 (Bone), certolizumab pegol (CAS No. 428863-50-7 or CH-138 (UCB), cilomilast (CAS No. 153259-65-5) (GlaxoSmithKline), CR- 1 (Nuada Pharmaceuticals), CRx-119 (CombinatoRx), D-5410 (UCB), dacopafant (CAS No. 125372-33-0) (Sanofi-Aventis), dersalazine (CAS No. 188913-57-7/188913-58-8) (Uriach), etanercept (CAS No. 185243-69-0) (Amgen), ethyl pyruvate (Critical (Critical Therapeutics), golimumab (CAS No. 476181-74-5) (Johnson & Johnson), hormono-immunotherapy from Ipsen, CDP571 (e.g., Humicade from UCB), IC-485 (ICOS), infliximab (CAS No. 170277-31-3) (Johnson & Johnson), IP-751 (Manhattan Pharmaceuticals), ISIS-104838 (CAS No. 250755-32-9) (ISIS Pharmaceuticals), lenalidomide (CAS No. 191732-72-6) (Celgene), lentinan (CAS No. 37339-90-5) (Ajinomoto), MDL-201112 (CAS No. 142130-73-2) (Sanofi-Aventis), medroxyprogesterone (CAS No. 520-85-4) (InKine Pharmaceutical), N-acetylcysteine (CAS No. 616-91-1) (Zambon), NBE-P2 (DIREVO Biotech), nerelimomab (CAS No. 162774-06-3) (Chiron), OM- 294DP (OM PHARMA), onercept (CAS No. 199685-57-9) (Yeda), PASSTNF-alpha (Verigen), pentoxifylline or oxypentifylline (Sanofi-Aventis), Pharmaprojects No.4091, 4241, 4295, or 4488 (Sanofi-Aventis), Pharmaprojects No. 5480 (Amgen), Pharmaprojects No. 6749 (Cengent), pirfenidone (CAS No. 53179-13-8) (MARNAC), RPR-132294 (Sanofi- Aventis), S5 (F002) (Fulcrum Pharmaceuticals), simvastatin (CAS No. 79902-63-9) (Merck & Co), STA-6292 (Synta Pharmaceuticals), tacrolimus (CAS No. 104987-11-3) (Fujisawa LifeCycle Pharma), talactoferrin alfa (CAS No. 308240-58-6) (Agennix), thalidomide (CAS No. 50-35-1) (Celgene), TNF antagonists form ProStrakan, and Synergen, TNF inhibitors (Amgen), TNF-alpha antagonists from Dynavax Technologies and Jerina AG (Germany), TNF-alpha inhibitors from IBFB Pharma and Xencor (Xencor), torbafylline (CAS No. 105102-21-4) (Sanofi-Aventis), UR-1505 (Uriach), VT-346 (Won Therapeutics), YSIL6 (Y's Therapeutics), YSTH2 (Y's Therapeutics), NPI-1302a-3 (Nereus Pharmaceuticals, a TNF antagonist from Jerina AG (Germany), dersalazine, and an analogue or derivative thereof.
23884. The composition of item 23785 wherein the agent is a Toll receptor inhibitor.
23885. The composition of item 23785 wherein the agent is a tubulin antagonist.
23886. The composition of item 23785 wherein the agent is a tyrosine kinase inhibitor selected from the group consisting of SU- 011248, SUTENT from Pfizer Inc. (New York, NY), BMS-354825, PN-355 (Paracelsian Pharmaceuticals), AGN-199659 (Allergan), AAL-993 or ABP- 309 (Novartis), adaphostin (NIH), AEE-788 (Novartis), AG-013736 (OSI Pharmaceuticals), AG-13736 (Pfizer), ALT-110 (Alteris Therapeutics), AMG- 706 (Amgen), anticancer MAbs from Xencor, anti-EGFrvlll MAbs from Abgenix, anti-HER2 MAb from Abiogen, AZD-2171 or AZD-9935 (AstraZeneca), BAY-57-9352 (Bayer), BIBF-1120 (Boehringer Ingelheim), CEP-5214 (Cephalon), CEP-7055 (Cephalon), cetuximab (ImCIone Systems), CHIR-200131 and CHIR-258 (Chiron), CP-547632 (OSI Pharmaceuticals), CP-724714 (Pfizer), CT-301 (Creabilis Therapeutics), D- 69491 (Baxter International), E-7080 (Eisai), EG-3306 (Ark Therapeutics), EGFR/ErbB2 inhibitors from Array BioPharma, erlotinib (CAS No. 183319- 69-9) (OSI Pharmaceuticals), EXEL-2880 (Exelixis), FK-778 (Sanofi- Aventis) , gefitinib (CAS No. 184475-35-2) (AstraZeneca), GW-2286 or GW- 654652 (GlaxoSmithKline), her2/neu antigen from AlphaVax, HER-2/neu inhibitor from Generex, Herzyme (Medipad) (Sirna Therapeutics), HKI-272 (Wyeth), HuMax-EGFr (Genmab), idronoxil (CAS No. 81267-65-4) (Novogen), IGF-1 inhibitors from Ontogen, IMC-11F8 (ImCIone Systems), kahalalide F (CAS No. 149204-42-2) (PharmaMar), KDR inhibitor from LG Life Sciences, KDR inhibitors from Abbott Laboratories, KDR kinase inhibitors (UCB), Kdr kinase inhibitors from Merck & Co, KRN-633 and KRN- 951 (Kirin Brewery), KSB-102 (Xenova), lapatinib ditosylate (CAS No. 388082-78-8) (GlaxoSmithKline), matuzumab (Merck KGaA), MDX-214 (Medarex), ME-103 (Pharmexa), MED-A300 (Gerolymatos), MNAC-13 (Lay Line Genomics), nimotuzumab (Center of Molecular Immunology), NSC- 330507 or NSC-707545 (NIH), NV-50 (Novogen), OSI-930 (OSI Pharmaceuticals), panitumumab (Abgenix), pelitinib (CAS No. 287933-82-7) (Wyeth), pertuzumab (CAS No. 380610-27-5) (Genentech), Pharmaprojects No. 3985 (Sanofi-Aventis), prostate cancer therapeutics from Sequenom (SQPC35, SQPC36, SQPC90), removab and remoxab (Trion Pharma), RG- 13022 (CAS No. 136831-48-6), RG-13291 (CAS No. 138989-50-1), or RG- 14620 (CAS No. 136831-49-7) (Sanofi-Aventis), RM-6427 (Romark), RNAi breast cancer therapy from Benitec, RP 53801 (CAS No. 125882-88-4) (Sanofi-Aventis), sorafenib tosylate (Bayer), SU-11657 (Pfizer), Tie-2 antagonists from Semaia (Hybrigenics), Tie-2 inhibitors from Ontogen, trastuzumab (CAS No. 180288-69-1) (Genentech), tyrosine kinase inhibitors from Sanofi-Aventis, U3-1287, U3-1565, U3-1784, U3-1800 (U3 Pharma), vatalanib (Novartis), VEGFR-2 kinase inhibitor from Bristol-Myers Squibb, XL-647 (Exelixis), ZD-6474 (AstraZeneca), ZK-CDK (Schering AG)1 an EGFR tyrosine kinase inhibitor, EKB-569 (Wyeth), and an analogue or derivative thereof.
23887. The composition of item 23785 wherein the agent is a VEGF inhibitor.
23888. The composition of item 23785 wherein the agent is a vitamin D receptor agonist.
23889. The composition of item 23785 wherein the agent is ZD-6474 (an angiogenesis inhibitor).
23890. The composition of item 23785 wherein the agent is AP-23573 (an mTOR inhibitor).
23891. The composition of item 23785 wherein the agent is synthadotin (a tubulin antagonist).
23892. The composition of item 23785 wherein the agent is S-0885 (a collagenase inhibitor).
23893. The composition of item 23785 wherein the agent is aplidine (an elongation factor-1 alpha inhibitor).
23894. The composition of item 23785 wherein the agent is ixabepilone (an epithilone). 23895. The composition of item 23785 wherein the agent is IDN-5390 (an angiogenesis inhibitor).
23896. The composition of item 23785 wherein the agent is SB-2723005 (an angiogenesis inhibitor).
23897. The composition of item 23785 wherein the agent is ABT-518 (an angiogenesis inhibitor).
23898. The composition of item 23785 wherein the agent is combretastatin (an angiogenesis inhibitor).
23899. The composition of item 23785 wherein the agent is anecortave acetate (an angiogenesis inhibitor).
23900. The composition of item 23785 wherein the agent is SB-715992 (a kinesin antagonist).
23901. The composition of item 23785 wherein the agent is temsirolimus (an mTOR inhibitor).
23902. The composition of item 23785 wherein the agent is adalimumab (a TNFα antagonist).
23903. The composition of any one of items 23785- 23902 wherein the polymer is crosslinked.
23904. The composition of any one of items 23785- 23902 wherein the polymer reacts with mammalian tissue. 23905. The composition of any one of items 23785- 23902 wherein the polymer is a naturally occurring polymer.
23906. The composition of any one of items 23785- 23902 wherein the polymer is protein.
23907. The composition of any one of items 23785- 23902 wherein the polymer is carbohydrate.
23908. The composition of any one of items 23785- 23902 wherein the polymer is biodegradable polymer.
23909. The composition of any one of items 23785- 23902 wherein the polymer is crosslinked and biodegradable.
23910. The composition of any one of items 23785- 23902 wherein the polymer is nonbiodegradable polymer.
23911. The composition of any one of items 23785- 23902 wherein the polymer is collagen.
23912. The composition of any one of items 23785- 23902 wherein the polymer is methylated collagen.
23913. The composition of any one of items 23785- 23902 comprising fibrinogen.
23914. The composition of any one of items 23785- 23902 comprising thrombin. 23915. The composition of any one of items 23785- 23902 comprising calcium salt.
23916. The composition of any one of items 23785- 23902 comprising an antifibronolytic agent.
23917. The composition of any one of items 23785- 23902 comprising a fibrinogen analog.
23918. The composition of any one of items 23785- 23902 comprising albumin.
23919. The composition of any one of items 23785- 23902 comprising plasminogen.
23920. The composition of any one of items 23785- 23902 comprising von Willebrands factor.
23921. The composition of any one of items 23785- 23902 comprising Factor VIII.
23922. The composition of any one of items 23785- 23902 comprising hypoallergenic collagen.
23923. The composition of any one of items 23785- 23902, comprising atelopeptidic collagen.
23924. The composition of any one of items 23785- 23902, comprising telopeptide collagen. 23925. The composition of any one of items 23785- 23902, comprising crosslinked collagen.
23926. The composition of any one of items 23785- 23902, comprising aprotinin.
23927. The composition of any one of items 23785- 23902 comprising epsilon-amino-n-caproic acid.
23928. The composition of any one of items 23785- 23902 comprising gelatin.
23929. The composition of any one of items 23785- 23902 comprising protein conjugates.
23930. The composition _of any one of items 23785- 23902 comprising gelatin conjugates.
23931. The composition of any one of items 23785- 23902 comprising hyaluronic acid.
23932. The composition of any one of items 23785- 23902 comprising a hyaluronic acid derivative.
23933. The composition of any one of items 23785- 23902 comprising a synthetic polymer.
23934. The composition of any one of items 23785- 23902 wherein the polymer is formed from reactants comprising a synthetic isocyanate-containing compound. 23935. The composition of any one of items 23785- 23902 comprising a synthetic isocyanate-containing compound.
23936. The composition of any one of items 23785- 23902 wherein the polymer is formed from reactants comprising a synthetic thioi-containing compound.
23937. The composition of any one of items 23785- 23902 comprising a synthetic thiol-containing compound.
23938. The composition of any one of items 23785- 23902 wherein the polymer is formed from reactants comprising a synthetic compound containing at least two thiol groups.
23939. The composition of any one of items 23785- 23902 comprising a synthetic compound containing at least two thiol groups..
23940. The composition of any one of items 23785- 23902 wherein the polymer is formed from reactants comprising a synthetic compound containing at least three thiol groups.
23941. The composition of any one of items 23785- 23902 comprising a synthetic compound containing at least three thiol groups.
23942. The composition of any one of items 23785- 23902 wherein the polymer is formed from reactants comprising a synthetic compound containing at least four thiol groups. 23943. The composition of any one of items 23785- 23902 comprising a synthetic compound containing at least four thiol groups.
23944. The composition of any one of items 23785- 23902 wherein the polymer is formed from reactants comprising a synthetic amino-containing compound.
23945. The composition of any one of items 23785- 23902 comprising a synthetic amino-containing compound.
23946. The composition of any one of items 23785- 23902 wherein the polymer is formed from reactants comprising a synthetic compound containing at least two amino groups.
23947. The composition of any one of items 23785- 23902 comprising a synthetic compound containing at least two amino groups.
23948. The composition of any one of items 23785- 23902 wherein the polymer is formed from reactants comprising a synthetic compound containing at least three amino groups.
23949. The composition of any one of items 23785- 23902 comprising a synthetic compound containing at least three amino groups.
23950. The composition of any one of items 23785- 23902 wherein the polymer is formed from reactants comprising a synthetic compound containing at least four amino groups. 23951. The composition of any one of items 23785- 23902 comprising a synthetic compound containing at least four amino groups.
23952. The composition of any one of items 23785- 23902 wherein the polymer is formed from reactants comprising a synthetic compound comprising a carbonyl-oxygen-succinimidyl group.
23953. The composition of any one of items 23785- 23902 comprising a synthetic compound comprising a carbonyl-oxygen- succinimidyl group.
23954. The composition of any one of items 23785- 23902 wherein the polymer is formed from reactants comprising a synthetic compound comprising at least two carbonyl-oxygen-succinimidyl groups.
23955. The composition of any one of items 23785- 23902 comprising a synthetic compound comprising at least two carbonyl- oxygen-succinimidyl groups.
23956. The composition of any one of items 23785- 23902 wherein the polymer is formed from reactants comprising a synthetic compound comprising at least three carbonyl-oxygen-succinimidyl groups.
23957. The composition of any one of items 23785- 23902 comprising a synthetic compound comprising at least three carbonyl- oxygen-succinimidyl groups.
23958. The composition of any one of items 23785- 23902 wherein the polymer is formed from reactants comprising a synthetic compound comprising at least four carbonyl-oxygen-succinimidyl groups. 23959. The composition of any one of items 23785- 23902 comprising a synthetic compound comprising at least four carbonyl- oxygen-succinimidyl groups.
23960. The composition of any one of items 23785- 23902 wherein the polymer is formed from reactants comprising a synthetic polyalkylene oxide-containing compound.
23961. The composition of any one of items 23785- 23902 comprising a synthetic polyalkylene oxide-containing compound.
23962. The composition of any one of items 23785- 23902 wherein the polymer is formed from reactants comprising a synthetic compound comprising both polyalkylene oxide and biodegradable polyester blocks.
23963. The composition of any one of items 23785- 23902 comprising a synthetic compound comprising both polyalkylene oxide and biodegradable polyester blocks.
23964. The composition of any one of items 23785- 23902 wherein the polymer is formed from reactants comprising a synthetic polyalkylene oxide-containing compound having reactive amino groups.
23965. The composition of any one of items 23785- 23902 comprising a synthetic polyalkylene oxide-containing compound having reactive amino groups.
23966. The composition of any one of items 23785- 23902 wherein the polymer is formed from reactants comprising a synthetic polyalkylene oxide-containing compound having reactive thiol groups. 23967. The composition of any one of items 23785- 23902 comprising a synthetic polyalkylene oxide-containing compound having reactive thiol groups.
23968. The composition of any one of items 23785- 23902 wherein the polymer is formed from reactants comprising a synthetic polyalkylene oxide-containing compound having reactive carbonyl-oxygen- succinimidyl groups.
23969. The composition of any one of items 23785- 23902 comprising a synthetic polyalkylene oxide-containing compound having reactive carbonyl-oxygen-succinimidyl groups.
23970. The composition of any one of items 23785- 23902 wherein the polymer is formed from reactants comprising a synthetic compound comprising a biodegradable polyester block.
23971. The composition of any one of items 23785- 23902 comprising a synthetic compound comprising a biodegradable polyester block.
23972. The composition of any one of items 23785- 23902 wherein the polymer is formed from reactants comprising a synthetic polymer formed in whole or part from lactic acid or lactide.
23973. The composition of any one of items 23785- 23902 comprising a synthetic polymer formed in whole or part from lactic acid or lactide. 23974. The composition of any one of items 23785- 23902 wherein the polymer is formed from reactants comprising a synthetic polymer formed in whole or part from glycolic acid or glycolide.
23975. The composition of any one of items 23785- 23902 comprising a synthetic polymer formed in whole or part from glycolic acid or glycolide.
23976. The composition of any one of items 23785- 23902 wherein the polymer is formed from reactants comprising polylysine.
23977. The composition of any one of items 23785- 23902 comprising polylysine.
23978. The composition of any one of items 23785- 23902 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound comprising a polyalkylene oxide portion.
23979. The composition of any one of items 23785- 23902 comprising (a) protein and (b) a compound comprising a polyalkylene oxide portion.
23980. The composition of any one of items 23785- 23902 wherein the polymer is formed from reactants comprising (a) protein and (b) polylysine.
23981. The composition of any one of items 23785- 23902 comprising (a) protein and (b) polylysine. 23982. The composition of any one of items 23785- 23902 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound having at least four thiol groups.
23983. The composition of any one of items 23785- 23902 comprising (a) protein and (b) a compound having at least four thiol groups.
23984. The composition of any one of items 23785- 23902 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound having at least four amino groups.
23985. The composition of any one of items 23785- 23902 comprising (a) protein and (b) a compound having at least four amino groups.
23986. The composition of any one of items 23785- 23902 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound having at least four carbonyl-oxygen-succinimide groups.
23987. The composition of any one of items 23785- 23902 comprising (a) protein and (b) a compound having at least four carbonyl-oxygen-succinimide groups.
23988. The composition of any one of items 23785- 23902 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound having a biodegradable region formed from reactants selected from lactic acid, lactide, glycolic acid, glycolide, and epsilon- caprolactone. 23989. The composition of any one of items 23785- 23902 comprising (a) protein and (b) a compound having a biodegradable region formed from reactants selected from lactic acid, lactide, glycolic acid, glycolide, and epsilon-caprolactone.
23990. The composition of any one of items 23785- 23902 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound comprising a polyalkylene oxide portion.
23991. The composition of any one of items 23785- 23902 comprising (a) collagen and (b) a compound comprising a polyalkylene oxide portion.
23992. The composition of any one of items 23785- 23902 wherein the polymer is formed from reactants comprising (a) collagen and (b) polylysine.
23993. The composition of any one of items 23785- 23902 comprising (a) collagen and (b) polylysine.
23994. The composition of any one of items 23785- 23902 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound having at least four thiol groups.
23995. The composition of any one of items 23785- 23902 comprising (a) collagen and (b) a compound having at least four thiol groups.
23996. The composition of any one of items 23785- 23902 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound having at least four amino groups. 23997. The composition of any one of items 23785- 23902 comprising (a) collagen and (b) a compound having at least four amino groups.
23998. The composition of any one of items 23785- 23902 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound having at least four carbonyl-oxygen-succinimide groups.
23999. The composition of any one of items 23785- 23902 comprising (a) collagen and (b) a compound having at least four carbonyl-oxygen-succinimide groups.
24000. The composition of any one of items 23785- 23902 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound having a biodegradable region formed from-reactants selected from lactic acid, lactide, glycolic acid, glycolide, and epsilon- caprolactone.
24001. The composition of any one of items 23785- 23902 comprising (a) collagen and (b) a compound having a biodegradable region formed from reactants selected from lactic acid, lactide, glycolic acid, glycolide, and epsilon-caprolactone.
24002. The composition of any one of items 23785- 23902 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound comprising a polyalkylene oxide portion. 24003. The composition of any one of items 23785- 23902 comprising (a) methylated collagen and (b) a compound comprising a polyalkylene oxide portion.
24004. The composition of any one of items 23785- 23902 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) polylysine.
24005. The composition of any one of items 23785- 23902 comprising (a) methylated collagen and (b) polylysine.
24006. The composition of any one of items item 23785- 23902 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound having at least four thiol groups.
24007. The composition of any one of items 23785- 23902 comprising (a) methylated collagen and (b) a compound having at least four thiol groups.
24008. The composition of any one of items 23785- 23902 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound having at least four amino groups.
24009. The composition of any one of items 23785- 23902 comprising (a) methylated collagen and (b) a compound having at least four amino groups.
24010. The composition of any one of items 23785- 23902 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound having at least four carbonyl- oxygen-succinimide groups. 24011. The composition of any one of items 23785- 23902 comprising (a) methylated collagen and (b) a compound having at least four carbonyl-oxygen-succinimide groups.
24012. The composition of any one of items 23785- 23902 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound having a biodegradable region formed from reactants selected from lactic acid, lactide, glycolic acid, glycolide, and epsilon-caprolactone.
24013. The composition of any one of items 23785- 23902 comprising (a) methylated collagen and (b) a compound having a biodegradable region formed from reactants selected from lactic acid, lactide, glycolic acid, glycolide, and epsilon-caprolactone.
24014. The composition of any one of items 23785- 23902 wherein the polymer is formed from reactants comprising hyaluronic acid.
24015. The composition of any one of items 23785- 23902 comprising hyaluronic acid.
24016. The composition of any one of items 23785- 23902 wherein the polymer is formed from reactants comprising a hyaluronic acid derivative.
24017. The composition of any one of items 23785- 23902 comprising a hyaluronic acid derivative.
24018. The composition of any one of items 23785- 23902 wherein the polymer is formed from reactants comprising pentaerythritol poly(ethylene glycol)ether tetra-sulfhyclryl of number average molecular weight between 3,000 and 30,000.
24019. The composition of any one of items 23785- 23902 comprising pentaerythritol poly(ethylene glyco!)ether tetra-sulfhydryl of number average molecular weight between 3,000 and 30,000.
24020. The composition of any one of items 23785- 23902 wherein the polymer is formed from reactants comprising pentaerythritol poly(ethylene glycol)ether tetra-amino of number average molecular weight between 3,000 and 30,000.
24021. The composition of any one of items 23785- 23902 comprising pentaerythritol poly(ethylene glycol)ether tetra-amino of number average molecular weight between 3,000 and 30,000.
24022. The composition of any one of items 23785- 23902 wherein the polymer is formed from reactants comprising (a) a synthetic compound having a number average molecular weight between 3,000 and 30,000 and comprising a polyalkylene oxide region and multiple nucleophilic groups, and (b) a synthetic compound having a number average molecular weight between 3,000 and 30,000 and comprising a polyalkylene oxide region and multiple electrophilic groups.
24023. The composition of any one of items 23785- 23902 comprising (a) a synthetic compound having a number average molecular weight between 3,000 and 30,000 and comprising a polyalkylene oxide region and multiple nucleophilic groups, and (b) a synthetic compound having a number average molecular weight between 3,000 and 30,000 and comprising a polyalkylene oxide region and multiple electrophilic groups. 24024. The composition of any one of items 23785- 23902 wherein the composition comprises a colorant.
24025. The composition of any one of items 23785- 23902 wherein the composition is sterile.
24026. The device of item 1 wherein the agent is a cyclin dependent kinase inhibitor selected from the group consisting of CGP 74514A, bohemine, olomoucine (CAS No. 101622-51-9), indole-3-carbinol (CAS No. 700-06-1), and an analogue or derivative thereof.
24027. The device of item 1 wherein the agent is an endothelial growth factor receptor (EGF-R) kinase inhibitor selected from the group consisting of lavendustin A (CAS No. 125697-92-9), SU 1498 (a VEGF-R inhibitor), and an analogue or derivative thereof.
24028. The device of item 1 wherein the agent is mevastatin (a fibrinogen antagonist).
24029. The device of item 1 wherein the agent is vincamine (a microtubule inhibitor).
24030. The device of item 1 wherein the agent is emodin (CAS No. 518-82-1) (an NF kappa B inhibitor) or Bay 11-7085 (an NF kappa B inhibitor).
24031. The device of item 1 wherein the agent is SKF86002 (CAS No. 72873-74-6) (a p38 MAP kinase inhibitor).
24032. The device of item 1 wherein the agent is a peroxisome proliferators-activated receptor (PPAR) agonist selected from the group consisting of LBM642, WY-14,643 (CAS No. 50892-23-4), GW7647, fenofibric acid (CAS No. 42017-89-0), MCC-555 (CAS No. 161600-01-7), GW9662, GW1929, GW501516, L-165,041 (CAS No. 79558- 09-1), and an analogue or derivative thereof.
24033. The device of item 1 wherein the agent is a phosphodiesterase (PDE) inhibitor selected from the group consisting of theobromine (CAS No. 83-67-0), papverine hydrochloride (CAS No. 61-25- 6), quercetin dehydrate (CAS No. 6151-25-3), YM 976 (CAS No. 191219- 80-4), irsogladine (CAS No. 57381-26-7), and an analogue or derivative thereof.
24034. The device of item 1 wherein the agent is a tumor necrosis factor antagonist.
24035. The device of item 1 wherein the agent is herbimycin A (a tyrosine kinase inhibitor).
24036. The device of item 1 wherein the agent is erucylphosphocholine.
24037. The device of item 1 wherein the agent is alphastatin.
24038. The device of item 1 wherein the agent is etanercept.
24039. The device of item 1 wherein the agent is humicade. 24040. The device of item 1 wherein the agent is gefitinib.
24041. The device of item 1 wherein the agent is a histamine receptor antagonist selected from the group consisting of phenothiazines (e.g., promethazine), alkylamines (e.g., chlorpheniramine (CAS No. 7054-11-7), brompheniramine (CAS No. 980-71-2), fexofenadine hydrochloride, promethazine hydrochloride, loratadine, ketotifen fumarate salt, and acrivastine), methylxanthines (e.g., theophylline, theobromine, and caffeine), cimetidine (available under the tradename TAGAMET from SmithKline Beecham Phamaceutical Co., Wilmington, DE), ranitidine (available under the tradename ZANTAC from Warner Lambert Company, Morris Plains, NJ), famotidine (available under the tradename PEPCID from Merck & Co., Whitehouse Station, NJ), nizatidine (available under the tradename AXID from Reliant Pharmaceuticals, Inc., Liberty Corner, NJ), nizatidine, and roxatidine acetate (CAS No. 78628-28-1), H3 receptor antagonists (e.g., thioperamide and thioperamide maleate salt), and antihistamines (e.g., tricyclic dibenozoxepins, ethanolamines, ethylenediamines, piperizines, piperidines, and pthalazinones).
24042. The device of item 1 wherein the agent is an alpha adrenergic receptor antagonist.
24043. The device of item 1 wherein the agent is an anti-psychotic compound.
24044. The device of item 1 wherein the agent is a CaM kinase Il inhibitor.
24045. The device of item 1 wherein the agent is a G protein agonist. 24046. The device of item 1 wherein the agent is an antibiotic selected from the group consisting of apigenin (Cas No. 520-36-5), ampicillin sodium salt (CAS No. 69-52-3), puromycin, and an analogue or derivative thereof.
24047. The device of item 1 wherein the agent is an anti-microbial agent.
24048. The device of item 1 wherein the agent is a DNA topoisomerase inhibitor selected from the group consisting of D-lapachone (CAS No. 4707-32-8)~-arctigenin (CAS No. 7770-78-7), aurintricarboxylic acid, and an analogue or derivative thereof.
24049. The device of item 1 wherein the agent is a thromboxane A2 receptor inhibitor selected from the group consisting of BM- 531 (CAS No. 284464-46-6), ozagrel hydrochloride (CAS No. 78712-43-3), and an analogue or derivative thereof.
24050. The device of item 1 wherein the agent is a D2 dopamine receptor antagonist.
24051. The device of item 1 wherein the agent is a Peptidyl-Prolyl Cis/Trans lsomerase Inhibitor.
24052. The device of item 1 wherein the agent is a dopamine antagonist.
24053. The device of item 1 wherein the agent is an anesthetic compound. 24054. The device of item 1 wherein the agent is a clotting factor.
24055. The device of item 1 wherein the agent is a lysyl hydrolase inhibitor.
24056. The device of item 1 wherein the agent is a muscarinic receptor inhibitor.
24057. The device of item 1 wherein the agent is a superoxide anion generator.
24058. The device of item 1 wherein the agent is a steroid.
24059. The device of item 1 wherein the agent is an antiproliferative agent selected from the group consisting of silibinin (CAS No. 22888-70-6), silymarin (CAS No. 65666-07-1), 1 ,2-hexanediol, dioctyl phthalate (CAS No. 117-81-7), zirconium (IV) oxide, glycyrrhizic acid, spermidine trihydrochloride, tetrahydrochloride, CGP 74514A, spermine tetrahydrochloride, NG-methyl-L-arginine acetate salt, galardin, and an analogue or derivative thereof.
24060. The device of item 1 wherein the agent is a diuretic.
24061. The device of item 1 wherein the agent is an anti-coagulant.
24062. The device of item 1 wherein the agent is a cyclic GMP agonist. 24063. The device of item 1 wherein the agent is an adenylate cyclase agonist.
24064. The device of item 1 wherein the agent is an antioxidant.
24065. The device of item 1 wherein the agent is a nitric oxide synthase inhibitor.
24066. The device of item 1 wherein the agent is an anti-neoplastic agent selected from tirapazamine (CAS No. 27314-97-2), fludarabine (CAS No. 21679-14-1), cladribine, imatinib mesilate, and an analogue or derivative thereof.
24067. The device of item 1 wherein the agent is a DNA synthesis inhibitor.
24068. The device of item 1 wherein the agent is a DNA alkylating agent selected from dacarbazine (CAS No. 4342-03-4), temozolomide, procarbazine HCI, and an analogue or derivative thereof.
24069. The device of item 1 wherein the agent is a DNA methylation inhibitor.
24070. The device of item 1 wherein the agent is a NSAID agent.
24071. The device of item 1 wherein the agent is a peptidylglycine alpha-hydroxylating monooxygenase inhibitor. 24072. The device of item 1 wherein the agent is an MEK1/MEK 2 inhibitor.
24073. The device of item 1 wherein the agent is a NO synthase inhibitor.
24074. The device of item 1 wherein the agent is a retinoic acid receptor antagonist selected from isotretinoin (CAS No. 4759- 48-2) and an analogue or derivative thereof.
24075. The device of item 1 wherein the agent is an ACE inhibitor.
24076. The device of item 1 wherein the agent is a glycosylation inhibitor.
24077. The device of item 1 wherein the agent is an intracellular calcium influx inhibitor.
24078. The device of item 1 wherein the agent is an anti-emetic agent.
24079. The device of item 1 wherein the agent is an acetylcholinesterase inhibitor.
24080. The device of item 1 wherein the agent is an ALK-5 receptor antagonist.
24081. The device of item 1 wherein the agent is a RAR/RXT antagonist. 24082. The device of item 1 wherein the agent is an elF- 2a inhibitor.
24083. The device of item 1 wherein the agent is an S- adenosyl-L-homocysteine hydrolase inhibitor.
24084. The device of item 1 wherein the agent is an estrogen agonist.
24085. The device of item 1 wherein the agent is a serotonin receptor inhibitor.
24086. The device of item 1 wherein the agent is an anti-thrombotic agent.
24087. The device of item 1 wherein the agent is a tryptase inhibitor.
24088. The device of item 1 wherein the agent is a pesticide.
24089. The device of item 1 wherein the agent is a bone mineralization promotor.
24090. The device of item 1 wherein the agent is a bisphosphonate compound selected from risedronate and an analogue or derivative thereof.
24091. The device of item 1 wherein the agent is an anti-inflammatory compound. 24092. The device of item 1 wherein the agent is a DNA methylation promotor.
24093. The device of item 1 wherein the agent is an anti-spasmodic agent.
24094. The device of item 1 wherein the agent is a protein synthesis inhibitor.
24095. The device of item 1 wherein the agent is an D- glucosidase inhibitor.
24096. The device of item 1 wherein the agent is a calcium channel blocker.
24097. The device of item 1 wherein the agent is a pyruvate dehydrogenase activator.
24098. The device of item 1 wherein the agent is a prostaglandin inhibitor. (
24099. The device of item 1 wherein the agent is a sodium channel inhibitor.
24100. The device of item 1 wherein the agent is a serine protease inhibitor.
24101. The device of item 1 wherein the agent is an intracellular calcium flux inhibitor. 24102. The device of item 1 wherein the agent is a JAK2 inhibitor.
24103. The device of item 1 wherein the agent is an androgen inhibitor.
24104. The device of item 1 wherein the agent is an aromatase inhibitor.
24105. The device of item 1 wherein the agent is an anti-viral agent.
24106. The device of item 1 wherein the agent is a 5-HT inhibitor.
24107. The device of item 1 wherein the agent is an FXR antagonist.
24108. The device of item 1 wherein the agent is an actin polymerization and stabilization promotor.
24109. The device of item 1 wherein the agent is an AXOR12 agonist.
24110. The device of item 1 wherein the agent is an angiotensin Il receptor agonist.
24111. The device of item 1 wherein the agent is a platelet aggregation inhibitor. 24112. The device of item 1 wherein the agent is a CB1/CB2 receptor agonist.
24113. The device of item 1 wherein the agent is a norepinephrine reuptake inhibitor.
24114. The device of item 1 wherein the agent is a selective serotonin reuptake inhibitor.
24115. The device of item 1 wherein the agent is a reducing agent.
24116. The device of item 1 wherein the agent is a immuno-modulator selected from Bay 11-7085, (-)-arctigenin, idazoxan hydrochloride, and an analogue or derivative thereof.
24117. The device of item 1 wherein the agent is isotretinoin.
24118. The device of item 1 wherein the agent is radicicol.
24119. The device of item 1 wherein the agent is clobetasol propionate.
24120. The device of item 1 wherein the agent is homoharringtonine.
24121. The device of item 1 wherein the agent is trichostatin A. 24122. The device of item 1 wherein the agent is brefeldin A.
24123. The device of item 1 wherein the agent is thapsigargin.
24124. The device of item 1 wherein the agent is dolastatin 15.
24125. The device of item 1 wherein the agent is cerivastatin.
24126. The device of item 1 wherein the agent is jasplakinolide.
24127. The device of item 1 wherein the agent is herbimycin A.
24128. The device of item 1 wherein the agent is pirfenidone.
24129. The device of item 1 wherein the agent is vinorelbine.
24130. The device of item 1 wherein the agent is 17- DMAG.
24131. The device of item 1 wherein the agent is tacrolimus. 24132. The device of item 1 wherein the agent is loteprednol etabonate.
24133. The device of item 1 wherein the agent is juglone.
24134. The device of item 1 wherein the agent is prednisolone.
24135. The device of item 1 wherein the agent is puromycin.
24136. The device of item 1 wherein the agent is 3- BAABE.
24137. The device of item 1 wherein the agent is cladribine.
24138. The device of item 1 wherein the agent is mannose-6-phosphate.
24139. The device of item 1 wherein the agent is 5- azacytidine.
24140. The device of item 1 wherein the agent is Ly333531 (ruboxistaurin).
24141. The device of item 1 wherein the agent is simvastatin. 24142. The device of any one of items 24026-24141 , wherein the medical device is an intravascular device.
24143. The device of any one of items 24026-24141 , wherein the medical device is a gastrointestinal stent.
24144. The device of any one of items 24026-24141 , wherein the medical device is a tracheal and bronchial stent.
24145. The device of any one of items 24026-24141 , wherein the medical device is a genital urinary stent.
24146. The device of any one of items 24026-24141 , wherein the medical device is an ear and nose stent.
24147. The device of any one of items 24026-24141 , wherein the medical device is an ear ventilation device.
24148. The device of any one of items 24026-24141 , wherein the medical device is an intraocular implant.
24149. The device of any one of items 24026-24141 , wherein the medical device is a vascular graft.
24150. The device of any one of items 24026-24141 , wherein the medical device comprises a film or a mesh.
24151. The device of any one of items 24026-24141 , wherein the medical device is a glaucoma drainage device. 24152. The device of any one of items 24026-24141 , wherein the medical device is a prosthetic heart valve or a component thereof.
24153. The device of any one of items 24026-24141 , wherein the medical device is a penile implant.
24154. The device of any one of items 24026-24141 , wherein the medical device is an endotracheal or tracheostomy tube.
24155. The device of any one of items 24026-24141 , wherein the medical device is a peritoneal dialysis catheter.
24156. The device of any one of items 24026-24141 , wherein the medical device is a central nervous system shunt or a pressure monitoring device.
24157. The device of any one of items 24026-24141 , wherein the medical device is an inferior vena cava filter.
24158. The device of any one of items 24026-24141 , wherein the medical device is a gastrointestinal device.
24159. The device of any one of items 24026-24141 , wherein the medical device is a central venous catheter.
24160. The device of any one of items 24026-24141 , wherein the medical device is a ventricular assist device.
24161. The device of any one of items 24026-24141 , wherein the medical device is a spinal implant. 24162. The device of any one of items 24026-24141 , wherein the medical device is for treating hypertropic scar or keloid.
24163. The device of any one of items 24026-24141 , wherein the medical device is a hemodialysis access device.
24164. The device of any one of items 24026-24141 , wherein the medical device is a surgical adhesion barrier.
24165. The method of item 5493 wherein the agent is a cyclin dependent kinase inhibitor selected from the group consisting of CGP 74514A, bohemine, olomoucine (CAS No. 101622-51-9), indole-3-carbinol (CAS No. 700-06-1), and an analogue or derivative thereof.
24166. The method of item 5493 wherein the agent is an endothelial growth factor receptor (EGF-R) kinase inhibitor selected from the group consisting of lavendustin A (CAS No. 125697-92-9), SU 1498 (a VEGF-R inhibitor), and an analogue or derivative thereof.
24167. The method of item 5493 wherein the agent is mevastatin (a fibrinogen antagonist).
24168. The method of item 5493 wherein the agent is vincamine (a microtubule inhibitor).
24169. The method of item 5493 wherein the agent is emodin (CAS No. 518-82-1) (an NF kappa B inhibitor) or Bay 11-7085 (an NF kappa B inhibitor).
24170. The method of item 5493 wherein the agent is SKF86002 (CAS No. 72873-74-6) (a p38 MAP kinase inhibitor). 24171. The method of item 5493 wherein the agent is a peroxisome proliferators-activated receptor (PPAR) agonist selected from the group consisting of LBM642, WY-14,643 (CAS No. 50892-23-4), GW7647, fenofibric acid (CAS No. 42017-89-0), MCC-555 (CAS No. 161600-01-7), GW9662, GW1929, GW501516, L-165,041 (CAS No. 79558- 09-1), and an analogue or derivative thereof.
24172. The method of item 5493 wherein the agent is a phosphodiesterase (PDE) inhibitor selected from the group consisting of theobromine (CAS No. 83-67-0), papverine hydrochloride (CAS No. 61-25- 6), quercetin dehydrate (CAS No. 6151-25-3), YM 976 (CAS No. 191219- 80-4), irsogladine (CAS No. 57381-26-7), and an analogue or derivative thereof.
24173. The method of item 5493 wherein the agent is a tumor necrosis factor antagonist.
24174. The method of item 5493 wherein the agent is herbimycin A (a tyrosine kinase inhibitor).
24175. The method of item 5493 wherein the agent is erucylphosphocholine.
24176. The method of item 5493 wherein the agent is alphastatin.
24177. The method of item 5493 wherein the agent is etanercept.
24178. The method of item 5493 wherein the agent is humicade. 24179. The method of item 5493 wherein the agent is gefitinib.
24180. The method of item 5493 wherein the agent is a histamine receptor antagonist selected from the group consisting of phenothiazines (e.g., promethazine), alkylamines {e.g., chlorpheniramine (CAS No. 7054-11-7), brompheniramine (CAS No. 980-71-2), fexofenadine hydrochloride, promethazine hydrochloride, loratadine, ketotifen fumarate salt, and acrivastine), methylxanthines (e.g., theophylline, theobromine, and caffeine), cimetidine (available under the tradename TAGAMET from SmithKline Beecham Phamaceutical Co., Wilmington, DE), ranitidine (available under the tradename ZANTAC from Warner Lambert Company, Morris Plains, NJ), famotidine (available under the tradename PEPCID from Merck & Co., Whitehouse Station, NJ), nizatidine (available under the tradename AXID from Reliant Pharmaceuticals, Inc., Liberty Corner, NJ), nizatidine, and roxatidine acetate (CAS No. 78628-28-1), H3 receptor antagonists (e.g., thioperamide and thioperamide maleate salt), and antihistamines (e.g., tricyclic dibenozoxepins, ethanolamines, ethylenediamines, piperizines, piperidines, and pthalazinones).
24181. The method of item 5493 wherein the agent is an alpha adrenergic receptor antagonist.
24182. The method of item 5493 wherein the agent is an anti-psychotic compound.
24183. The method of item 5493 wherein the agent is a CaM kinase Il inhibitor.
24184. The method of item 5493 wherein the agent is a G protein agonist. 24185. The method of item 5493 wherein the agent is an antibiotic selected from the group consisting of apigenin (Cas No. 520-36-5), ampicillin sodium salt (CAS No. 69-52-3), puromycin, and an analogue or derivative thereof.
24186. The method of item 5493 wherein the agent is an anti-microbial agent.
24187. The method of item 5493 wherein the agent is a DNA topoisomerase inhibitor selected from the group consisting of D- lapachone (CAS No. 4707-32-8)~-arctigenin (CAS No. 7770-78-7), aurintricarboxylic acid, and an analogue or derivative thereof.
24188. The method of item 5493 wherein the agent is a thromboxane A2 receptor inhibitor selected from the group consisting of BM- 531 (CAS No. 284464-46-6), ozagrel hydrochloride (CAS No. 78712-43-3), and an analogue or derivative thereof.
24189. The method of item 5493 wherein the agent is a D2 dopamine receptor antagonist.
24190. The method of item 5493 wherein the agent is a Peptidyl-Prolyl Cis/Trans lsomerase Inhibitor.
24191. The method of item 5493 wherein the agent is a dopamine antagonist.
24192. The method of item 5493 wherein the agent is an anesthetic compound. 24193. The method of item 5493 wherein the agent is a clotting factor.
24194. The method of item 5493 wherein the agent is a lysyl hydrolase inhibitor.
24195. The method of item 5493 wherein the agent is a muscarinic receptor inhibitor.
24196. The method of item 5493 wherein the agent is a superoxide anion generator.
24197. The method of item 5493 wherein the agent is a steroid.
24198. The method of item 5493 wherein the agent is an anti-proliferative agent selected from the group consisting of silibinin (CAS No. 22888-70-6), silymarin (CAS No. 65666-07-1), 1 ,2-hexanediol, dioctyl phthalate (CAS No. 117-81-7), zirconium (IV) oxide, glycyrrhizic acid, spermidine trihydrochloride, tetrahydrochloride, CGP 74514A, spermine tetrahydrochloride, NG-methyl-L-arginine acetate salt, galardin, and an analogue or derivative thereof.
24199. The method of item 5493 wherein the agent is a diuretic.
24200. The method of item 5493 wherein the agent is an anti-coagulant.
24201. The method of item 5493 wherein the agent is a cyclic GMP agonist. 24202. The method of item 5493 wherein the agent is an adenylate cyclase agonist.
24203. The method of item 5493 wherein the agent is an antioxidant.
24204. The method of item 5493 wherein the agent is a nitric oxide synthase inhibitor.
24205. The method of item 5493 wherein the agent is an anti-neoplastic agent selected from tirapazamine (CAS No. 27314-97-2), fludarabine (CAS No. 21679-14-1), cladribine, imatinib mesilate, and an analogue or derivative thereof.
24206. The method of item 5493 wherein the agent is a DNA synthesis inhibitor.
24207. The method of item 5493 wherein the agent is a DNA alkylating agent selected from dacarbazine (CAS No. 4342-03-4), temozolomide, procarbazine HCI, and an analogue or derivative thereof.
24208. The method of item 5493 wherein the agent is a DNA methylation inhibitor.
24209. The method of item 5493 wherein the agent is a NSAID agent.
24210. The method of item 5493 wherein the agent is a peptidylglycine alpha-hydroxylating monooxygenase inhibitor. 24211. The method of item 5493 wherein the agent is an MEK1/MEK 2 inhibitor.
24212. The method of item 5493 wherein the agent is a NO synthase inhibitor.
24213. The method of item 5493 wherein the agent is a retinoic acid receptor antagonist selected from isotretinoin (CAS No. 4759- 48-2) and an analogue or derivative thereof.
24214. The method of item 5493 wherein the agent is an ACE inhibitor.
24215. The method of item 5493 wherein the agent is a glycosylation inhibitor.
24216. The method of item 5493 wherein the agent is an intracellular calcium influx inhibitor.
24217. The method of item 5493 wherein the agent is an anti-emetic agent.
24218. The method of item 5493 wherein the agent is an acetylcholinesterase inhibitor.
24219. The method of item 5493 wherein the agent is an ALK-5 receptor antagonist.
24220. The method of item 5493 wherein the agent is a RAR/RXT antagonist. 24221. The method of item 5493 wherein the agent is an elF-2a inhibitor.
24222. The method of item 5493 wherein the agent is an S-adenosyl-L-homocysteine hydrolase inhibitor.
24223. The method of item 5493 wherein the agent is an estrogen agonist.
24224. The method of item 5493 wherein the agent is a serotonin receptor inhibitor.
24225. The method of item 5493 wherein the agent is an anti-thrombotic agent.
24226. The method of item 5493 wherein the agent is a tryptase inhibitor.
24227. The method of item 5493 wherein the agent is a pesticide.
24228. The method of item 5493 wherein the agent is a bone mineralization promotor.
24229. The method of item 5493 wherein the agent is a bisphosphonate compound selected from risedronate and an analogue or derivative thereof.
24230. The method of item 5493 wherein the agent is an anti-inflammatory compound. 24231. The method of item 5493 wherein the agent is a DNA methylation promotor.
24232. The method of item 5493 wherein the agent is an anti-spasmodic agent.
24233. The method of item 5493 wherein the agent is a protein synthesis inhibitor.
24234. The method of item 5493 wherein the agent is an D-glucosidase inhibitor.
24235. The method of item 5493 wherein the agent is a calcium channel blocker.
24236. The method of item 5493 wherein the agent is a pyruvate dehydrogenase activator.
24237. The method of item 5493 wherein the agent is a prostaglandin inhibitor.
24238. The method of item 5493 wherein the agent is a sodium channel inhibitor.
24239. The method of item 5493 wherein the agent is a serine protease inhibitor.
24240. The method of item 5493 wherein the agent is an intracellular calcium flux inhibitor. 24241. The method of item 5493 wherein the agent is a JAK2 inhibitor.
24242. The method of item 5493 wherein the agent is an androgen inhibitor.
24243. The method of item 5493 wherein the agent is an aromatase inhibitor.
24244. The method of item 5493 wherein the agent is an anti-viral agent.
24245. The method of item 5493 wherein the agent is a 5-HT inhibitor.
24246. The method of item 5493 wherein the agent is an FXR antagonist.
24247. The method of item 5493 wherein the agent is an actin polymerization and stabilization promotor.
24248. The method of item 5493 wherein the agent is an AXOR12 agonist.
24249. The method of item 5493 wherein the agent is an angiotensin Il receptor agonist.
24250. The method of item 5493 wherein the agent is a platelet aggregation inhibitor. 24251. The method of item 5493 wherein the agent is a CB1/CB2 receptor agonist.
24252. The method of item 5493 wherein the agent is a norepinephrine reuptake inhibitor.
24253. The method of item 5493 wherein the agent is a selective serotonin reuptake inhibitor.
24254. The method of item 5493 wherein the agent is a reducing agent.
24255. The method of item 5493 wherein the agent is a immuno-modulator selected from Bay 11-7085, (-)-arctigenin, idazoxan hydrochloride, and an analogue or derivative thereof.
24256. The method of item 5493 wherein the agent is isotretinoin.
24257. The method of item 5493 wherein the agent is radicicol.
24258. The method of item 5493 wherein the agent is clobetasol propionate.
24259. The method of item 5493 wherein the agent is homoharringtonine.
24260. The method of item 5493 wherein the agent is trichostatin A. 24261. The method of item 5493 wherein the agent is brefeldin A.
24262. The method of item 5493 wherein the agent is thapsigargin.
24263. The method of item 5493 wherein the agent is dolastatin 15.
24264. The method of item 5493 wherein the agent is cerivastatin.
24265. The method of item 5493 wherein the agent is jasplakinolide.
24266. The method of item 5493 wherein the agent is herbimycin A.
24267. The method of item 5493 wherein the agent is pirfenidone.
24268. The method of item 5493 wherein the agent is vinorelbine.
24269. The method of item 5493 wherein the agent is 17-DMAG.
24270. The method of item 5493 wherein the agent is tacrolimus. 24271. The method of item 5493 wherein the agent is loteprednol etabonate.
24272. The method of item 5493 wherein the agent is juglone.
24273. The method of item 5493 wherein the agent is prednisolone.
24274. The method of item 5493 wherein the agent is puromycin.
24275. The method of item 5493 wherein the agent is 3- BAABE.
24276. The method of item 5493 wherein the agent is cladribine.
24277. The method of item 5493 wherein the agent is mannose-6-phosphate.
24278. The method of item 5493 wherein the agent is 5- azacytidine.
24279. The method of item 5493 wherein the agent is Ly333531 (ruboxistaurin).
24280. The method of item 5493 wherein the agent is simvastatin. 24281. The method of any one of items 24165-24280, wherein the medical device is an intravascular device.
24282. The method of any one of items 24165-24280, wherein the medical device is a gastrointestinal stent.
24283. The method of any one of items 24165-24280, wherein the medical device is a tracheal and bronchial stent.
24284. The method of any one of items 24165-24280, wherein the medical device is a genital urinary stent.
24285. The method of any one of items 24165-24280, wherein the medical device is an ear and nose stent.
24286. The method of any one of items 24165-24280, wherein the medical device is an ear ventilation device.
24287. The method of any one of items 24165-24280, wherein the medical device is an intraocular implant.
24288. The method of any one of items 24165-24280, wherein the medical device is a vascular graft.
24289. The method of any one of items 24165-24280, wherein the medical device comprises a film or a mesh.
24290. The method of any one of items 24165-24280, wherein the medical device is a glaucoma drainage device. 24291. The method of any one of items 24165-24280, wherein the medical device is a prosthetic heart valve or a component thereof.
24292. The method of any one of items 24165-24280, wherein the medical device is a penile implant.
24293. The method of any one of items 24165-24280, wherein the medical device is an endotracheal or tracheostomy tube.
24294. The method of any one of items 24165-24280, wherein the medical device is a peritoneal dialysis catheter.
24295. The method of any one of items 24165-24280, wherein the medical device is a central nervous system shunt or a pressure monitoring device.
24296. The method of any one of items 24165-24280, wherein the medical device is an inferior vena cava filter.
24297. The method of any one of items 24165-24280, wherein the medical device is a gastrointestinal device.
24298. The method of any one of items 24165-24280, wherein the medical device is a central venous catheter.
24299. The method of any one of items 24165-24280, wherein the medical device is a ventricular assist device.
24300. The method of any one of items 24165-24280, wherein the medical device is a spinal implant. 24301. The method of any one of items 24165-24280, wherein the medical device is for treating hypertropic scar or keloid.
24302. The method of any one of items 24165-24280, wherein the medical device is a hemodialysis access device.
24303. The method of any one of items 24165-24280, wherein the medical device is a surgical adhesion barrier.
24304. The method of item 11030 wherein the agent is a cyclin dependent kinase inhibitor selected from the group consisting of CGP 74514A, bohemine, olomoucine (CAS No. 101622-51-9), indole-3-carbinol (CAS No. 700-06-1), and an analogue or derivative thereof.
24305. The method of item 11030 wherein the agent is an endothelial growth factor receptor (EGF-R) kinase inhibitor selected from the group consisting of lavendustin A (CAS No. 125697-92-9), SU 1498 (a VEGF-R inhibitor), and an analogue or derivative thereof.
24306. The method of item 11030 wherein the agent is mevastatin (a fibrinogen antagonist).
24307. The method of item 11030 wherein the agent is vincamine (a microtubule inhibitor).
24308. The method of item 11030 wherein the agent is emodin (CAS No. 518-82-1) (an NF kappa B inhibitor) or Bay 11-7085 (an NF kappa B inhibitor).
24309. The method of item 11030 wherein the agent is SKF86002 (CAS No. 72873-74-6) (a p38 MAP kinase inhibitor). 24310. The method of item 11030 wherein the agent is a peroxisome proliferators-activated receptor (PPAR) agonist selected from the group consisting of LBM642, WY-14,643 (CAS No. 50892-23-4), GW7647, fenofibric acid (CAS No. 42017-89-0), MCC-555 (CAS No. 161600-01-7), GW9662, GW1929, GW501516, L-165,041 (CAS No. 79558- 09-1), and an analogue or derivative thereof.
24311. The method of item 11030 wherein the agent is a phosphodiesterase (PDE) inhibitor selected from the group consisting of theobromine (CAS No. 83-67-0), papverine hydrochloride (CAS No. 61-25- 6), quercetin dehydrate (CAS No. 6151-25-3), YM 976 (CAS No. 191219- 80-4), irsogladine (CAS No. 57381-26-7), and an analogue or derivative thereof.
24312. The method of item 11030 wherein the agent is a tumor necrosis factor antagonist.
24313. The method of item 11030 wherein the agent is herbimycin A (a tyrosine kinase inhibitor).
24314. The method of item 11030 wherein the agent is erucylphosphocholine.
24315. The method of item 11030 wherein the agent is alphastatin.
24316. The method of item 11030 wherein the agent is etanercept.
24317. The method of item 11030 wherein the agent is humicade. 24318. The method of item 11030 wherein the agent is gefitinib.
24319. The method of item 11030 wherein the agent is a histamine receptor antagonist selected from the group consisting of phenothiazines (e.g., promethazine), alkylamines (e.g., chlorpheniramine (CAS No. 7054-11-7), brompheniramine (CAS No. 980-71-2), fexofenadine hydrochloride, promethazine hydrochloride, loratadine, ketotifen fumarate salt, and acrivastine), methylxanthines (e.g., theophylline, theobromine, and caffeine), cimetidine (available under the tradename TAGAMET from SmithKline Beecham Phamaceutical Co., Wilmington, DE), ranitidine (available under the tradename ZANTAC from Warner Lambert Company, Morris Plains, NJ), famotidine (available under the tradename PEPCID from Merck & Co., Whitehouse Station, NJ), nizatidine (available under the tradename AXID from Reliant Pharmaceuticals, Inc., Liberty Comer, NJ), nizatidine, and roxatidine acetate (CAS No. 78628-28-1), H3 receptor antagonists (e.g., thioperamide and thioperamide maleate salt), and antihistamines (e.g., tricyclic dibenozoxepins, ethanolamines, ethylenediamines, piperizines, piperidines, and pthalazinones).
24320. The method of item 11030 wherein the agent is an alpha adrenergic receptor antagonist.
24321. The method of item 11030 wherein the agent is an anti-psychotic compound.
24322. The method of item 11030 wherein the agent is a CaM kinase Il inhibitor.
24323. The method of item 11030 wherein the agent is a G protein agonist. 24324. The method of item 11030 wherein the agent is an antibiotic selected from the group consisting of apigenin (Cas No. 520- 36-5), ampicillin sodium salt (CAS No. 69-52-3), puromycin, and an analogue or derivative thereof.
24325. The method of item 11030 wherein the agent is an anti-microbial agent.
24326. The method of item 11030 wherein the agent is a DNA topoisomerase inhibitor selected from the group consisting of D- lapachone (CAS No. 4707-32-8)~-arctigenin (CAS No. 7770-78-7), aurintricarboxylic acid, and an analogue or derivative thereof.
24327. The method of item 11030 wherein the agent is a thromboxane A2 receptor inhibitor selected from the group consisting of BM- 531 (CAS No. 284464-46-6), ozagrel hydrochloride (CAS No. 78712-43-3), and an analogue or derivative thereof.
24328. The method of item 11030 wherein the agent is a D2 dopamine receptor antagonist.
24329. The method of item 11030 wherein the agent is a Peptidyl-Prolyl Cis/Trans lsomerase Inhibitor.
24330. The method of item 11030 wherein the agent is a dopamine antagonist.
24331. The method of item 11030 wherein the agent is an anesthetic compound. 24332. The method of item 11030 wherein the agent is a clotting factor.
24333. The method of item 11030 wherein the agent is a lysyl hydrolase inhibitor.
24334. The method of item 11030 wherein the agent is a muscarinic receptor inhibitor.
24335. The method of item 11030 wherein the agent is a superoxide anion generator.
24336. The method of item 11030 wherein the agent is a steroid.
24337. The method of item 11030 wherein the agent is an anti-proliferative agent selected from the group consisting of silibinin (CAS No. 22888-70-6), silymarin (CAS No. 65666-07-1), 1 ,2-hexanediol, dioctyl phthalate (CAS No. 117-81-7), zirconium (IV) oxide, glycyrrhizic acid, spermidine trihydrochloride, tetrahydrochloride, CGP 74514A, spermine tetrahydrochloride, NG-methyl-L-arginine acetate salt, galardin, and an analogue or derivative thereof.
24338. The method of item 11030 wherein the agent is a diuretic.
24339. The method of item 11030 wherein the agent is an anti-coagulant.
24340. The method of item 11030 wherein the agent is a cyclic GMP agonist. 24341. The method of item 11030 wherein the agent is an adenylate cyclase agonist.
24342. The method of item 11030 wherein the agent is an antioxidant.
24343. The method of item 11030 wherein the agent is a nitric oxide synthase inhibitor.
24344. The method of item 11030 wherein the agent is an anti-neoplastic agent selected from tirapazamine (CAS No. 27314-97-2), fludarabine (CAS No. 21679-14-1), cladribine, imatinib mesilate, and an analogue or derivative thereof.
24345. The method of item 11030 wherein the agent is a DNA synthesis inhibitor.
24346. The method of item 11030 wherein the agent is a DNA alkylating agent selected from dacarbazine (CAS No. 4342-03-4), temozolomide, procarbazine HCI, and an analogue or derivative thereof.
24347. The method of item 11030 wherein the agent is a DNA methylation inhibitor.
24348. The method of item 11030 wherein the agent is a NSAID agent.
24349. The method of item 11030 wherein the agent is a peptidylglycine alpha-hydroxylating monooxygenase inhibitor. 24350. The method of item 11030 wherein the agent is an MEK1/MEK 2 inhibitor.
24351. The method of item 11030 wherein the agent is a NO synthase inhibitor.
24352. The method of item 11030 wherein the agent is a retinoic acid receptor antagonist selected from isotretinoin (CAS No. 4759- 48-2) and an analogue or derivative thereof.
24353. The method of item 11030 wherein the agent is an ACE inhibitor.
24354. The method of item 11030 wherein the agent is a glycosylation inhibitor.
24355. The method of item 11030 wherein the agent is an intracellular calcium influx inhibitor.
24356. The method of item 11030 wherein the agent is an anti-emetic agent.
24357. The method of item 11030 wherein the agent is an acetylcholinesterase inhibitor.
24358. The method of item 11030 wherein the agent is an ALK-5 receptor antagonist.
24359. The method of item 11030 wherein the agent is a RAR/RXT antagonist. 24360. The method of item 11030 wherein the agent is an elF-2a inhibitor.
24361. The method of item 11030 wherein the agent is an S-adenosyl-L-homocysteine hydrolase inhibitor.
24362. The method of item 11030 wherein the agent is an estrogen agonist.
24363. The method of item 11030 wherein the agent is a serotonin receptor inhibitor.
24364. The method of item 11030 wherein the agent is an anti-thrombotic agent.
24365. The method of item 11030 wherein the agent is a tryptase inhibitor.
24366. The method of item 11030 wherein the agent is a pesticide.
24367. The method of item 11030 wherein the agent is a bone mineralization promotor.
24368. The method of item 11030 wherein the agent is a bisphosphonate compound selected from risedronate and an analogue or derivative thereof.
24369. The method of item 11030 wherein the agent is an anti-inflammatory compound. 24370. The method of item 11030 wherein the agent is a DNA methylation promotor.
24371. The method of item 11030 wherein the agent is an anti-spasmodic agent.
24372. The method of item 11030 wherein the agent is a protein synthesis inhibitor.
24373. The method of item 11030 wherein the agent is an D-glucosidase inhibitor.
24374. The method of item 11030 wherein the agent is a calcium channel blocker.
24375. The method of item 11030 wherein the agent is a pyruvate dehydrogenase activator.
24376. The method of item 11030 wherein the agent is a prostaglandin inhibitor.
24377. The method of item 11030 wherein the agent is a sodium channel inhibitor.
24378. The method of item 11030 wherein the agent is a serine protease inhibitor.
24379. The method of item 11030 wherein the agent is an intracellular calcium flux inhibitor. 24380. The method of item 11030 wherein the agent is a JAK2 inhibitor.
24381. The method of item 11030 wherein the agent is an androgen inhibitor.
24382. The method of item 11030 wherein the agent is an aromatase inhibitor.
24383. The method of item 11030 wherein the agent is an anti-viral agent.
24384. The method of item 11030 wherein the agent is a 5-HT inhibitor.
24385. The method of item 11030 wherein the agent is - an FXR antagonist.
24386. The method of item 11030 wherein the agent is an actin polymerization and stabilization promotor.
24387. The method of item 11030 wherein the agent is an AXOR 12 agonist.
24388. The method of item 11030 wherein the agent is an angiotensin Il receptor agonist.
24389. The method of item 11030 wherein the agent is a platelet aggregation inhibitor. 24390. The method of item 11030 wherein the agent is a CB1/CB2 receptor agonist.
24391. The method of item 11030 wherein the agent is a norepinephrine reuptake inhibitor.
24392. The method of item 11030 wherein the agent is a selective serotonin reuptake inhibitor.
24393. The method of item 11030 wherein the agent is a reducing agent.
24394. The method of item 11030 wherein the agent is a immuno-modulator selected from Bay 11-7085, (-)-arctigenin, idazoxan hydrochloride, and an analogue or derivative thereof.
24395. The method of item 11030 wherein the agent is isotretinoin.
24396. The method of item 11030 wherein the agent is radicicol.
24397. The method of item 11030 wherein the agent is clobetasol propionate.
24398. The method of item 11030 wherein the agent is homoharringtonine.
24399. The method of item 11030 wherein the agent is trichostatin A. 24400. The method of item 11030 wherein the agent is brefeldin A.
24401. The method of item 11030 wherein the agent is thapsigargin.
24402. The method of item 11030 wherein the agent is dolastatin 15.
24403. The method of item 11030 wherein the agent is cerivastatin.
24404. The method of item 11030 wherein the agent is jasplakinolide.
24405. The method of item 11030 wherein thejagent is herbimycin A.
24406. The method of item 11030 wherein the agent is pirfenidone.
24407. The method of item 11030 wherein the agent is vinorelbine.
24408. The method of item 11030 wherein the agent is 17-DMAG.
24409. The method of item 11030 wherein the agent is tacrolimus. 24410. The method of item 11030 wherein the agent is loteprednol etabonate.
24411. The method of item 11030 wherein the agent is juglone.
24412. The method of item 11030 wherein the agent is prednisolone.
24413. The method of item 11030 wherein the agent is puromycin.
24414. The method of item 11030 wherein the agent is 3-BAABE.
24415. The method of item 11030 wherein the agent is cladribine.
24416. The method of item 11030 wherein the agent is mannose-6-phosphate.
24417. The method of item 11030 wherein the agent is 5-azacytidine.
24418. The method of item 11030 wherein the agent is Ly333531 (ruboxistaurin).
24419. The method of item 11030 wherein the agent is simvastatin. 24420. The method of any one of items 24304-24419, wherein the medical device is an intravascular device.
24421. The method of any one of items 24304-24419, wherein the medical device is a gastrointestinal stent.
24422. The method of any one of items 24304-24419, wherein the medical device is a tracheal and bronchial stent.
24423. The method of any one of items 24304-24419, wherein the medical device is a genital urinary stent.
24424. The method of any one of items 24304-24419, wherein the medical device is an ear and nose stent.
24425. The method of any one of items 24304-24419, wherein the medical device is an ear ventilation device.
24426. The method of any one of items 24304-24419, wherein the medical device is an intraocular implant.
24427. The method of any one of items 24304-24419, wherein the medical device is a vascular graft.
24428. The method of any one of items 24304-24419, wherein the medical device comprises a film or a mesh.
24429. The method of any one of items 24304-24419, wherein the medical device is a glaucoma drainage device. 24430. The method of any one of items 24304-24419, wherein the medical device is a prosthetic heart valve or a component thereof.
24431. The method of any one of items 24304-24419, wherein the medical device is a penile implant.
24432. The method of any one of items 24304-24419, wherein the medical device is an endotracheal or tracheostomy tube.
24433. The method of any one of items 24304-24419, wherein the medical device is a peritoneal dialysis catheter.
24434. The method of any one of items 24304-24419, wherein the medical device is a central nervous system shunt or a pressure monitoring device.
24435. The method of any one of items 24304-24419, wherein the medical device is an inferior vena cava filter.
24436. The method of any one of items 24304-24419, wherein the medical device is a gastrointestinal device.
24437. The method of any one of items 24304-24419, wherein the medical device is a central venous catheter.
24438. The method of any one of items 24304-24419, wherein the medical device is a ventricular assist device.
24439. The method of any one of items 24304-24419, wherein the medical device is a spinal implant. 24440. The method of any one of items 24304-24419, wherein the medical device is for treating hypertropic scar or keloid.
24441. The method of any one of items 24304-24419, wherein the medical device is a hemodialysis access device.
24442. The method of any one of items 24304-24419, wherein the medical device is a surgical adhesion barrier.
24443. The method of item 17360 wherein the agent is a cyclin dependent kinase inhibitor selected from the group consisting of CGP 74514A, bohemine, olomoucine (CAS No. 101622-51-9), indole-3-carbinol (CAS No. 700-06-1), and an analogue or derivative thereof.
24444. The method of item 17360 wherein the agent is an endothelial growth factor receptor (EGF-R) kinase inhibitor selected from the group consisting of lavendustin A (CAS No. 125697-92-9), SU 1498 (a VEGF-R inhibitor), and an analogue or derivative thereof.
24445. The method of item 17360 wherein the agent is mevastatin (a fibrinogen antagonist).
24446. The method of item 17360 wherein the agent is vincamine (a microtubule inhibitor).
24447. The method of item 17360 wherein the agent is emodin (CAS No. 518-82-1) (an NF kappa B inhibitor) or Bay 11-7085 (an NF kappa B inhibitor).
24448. The method of item 17360 wherein the agent is SKF86002 (CAS No. 72873-74-6) (a p38 MAP kinase inhibitor). 24449. The method of item 17360 wherein the agent is a peroxisome proliferators-activated receptor (PPAR) agonist selected from the group consisting of LBM642, WY-14,643 (CAS No. 50892-23-4), GW7647, fenofibric acid (CAS No. 42017-89-0), MCC-555 (CAS No. 161600-01-7), GW9662, GW1929, GW501516, L-165,041 (CAS No. 79558- 09-1), and an analogue or derivative thereof.
24450. The method of item 17360 wherein the agent is a phosphodiesterase (PDE) inhibitor selected from the group consisting of theobromine (CAS No. 83-67-0), papverine hydrochloride (CAS No. 61-25- 6), quercetin dehydrate (CAS No. 6151-25-3), YM 976 (CAS No. 191219- 80-4), irsogladine (CAS No. 57381-26-7), and an analogue or derivative thereof.
24451. The method of item 17360 wherein the agent is a tumor necrosis factor antagonist.
24452. The method of item 17360 wherein the agent is herbimycin A (a tyrosine kinase inhibitor).
24453. The method of item 17360 wherein the agent is erucylphosphocholine.
24454. The method of item 17360 wherein the agent is alphastatin.
24455. The method of item 17360 wherein the agent is etanercept.
24456. The method of item 17360 wherein the agent is humicade. 24457. The method of item 17360 wherein the agent is gefitinib.
24458. The method of item 17360 wherein the agent is a histamine receptor antagonist selected from the group consisting of phenothiazines (e.g., promethazine), alkylamines (e.g., chlorpheniramine (CAS No. 7054-11-7), brompheniramine (CAS No. 980-71-2), fexofenadine hydrochloride, promethazine hydrochloride, loratadine, ketotifen fumarate salt, and acrivastine), methylxanthines (e.g., theophylline, theobromine, and caffeine), cimetidine (available under the tradename TAGAMET from SmithKline Beecham Phamaceutical Co., Wilmington, DE), ranitidine (available under the tradename ZANTAC from Warner Lambert Company, Morris Plains, NJ), famotidine (available under the tradename PEPCID from Merck & Co., Whitehouse Station, NJ), nizatidine (available under the tradename AXID from Reliant Pharmaceuticals, Inc., Liberty Corner, NJ), nizatidine, and roxatidine acetate (CAS No. 78628-28-1), H3 receptor antagonists (e.g., thioperamide and thioperamide maleate salt), and antihistamines (e.g., tricyclic dibenozoxepins, ethanolamines, ethylenediamines, piperizines, piperidines, and pthalazinones).
24459. The method of item 17360 wherein the agent is an alpha adrenergic receptor antagonist.
24460. The method of item 17360 wherein the agent is an anti-psychotic compound.
24461. The method of item 17360 wherein the agent is a CaM kinase Il inhibitor.
24462. The method of item 17360 wherein the agent is a G protein agonist. 24463. The method of item 17360 wherein the agent is an antibiotic selected from the group consisting of apigenin (Cas No. 520- 36-5), ampicillin sodium salt (CAS No. 69-52-3), puromycin, and an analogue or derivative thereof.
24464. The method of item 17360 wherein the agent is an anti-microbial agent.
24465. The method of item 17360 wherein the agent is a DNA topoisomerase inhibitor selected from the group consisting of D- lapachone (CAS No. 4707-32-8)~-arctigenin (CAS No. 7770-78-7), aurintricarboxylic acid, and an analogue or derivative thereof.
24466. The method of item 17360 wherein the agent is a thromboxane A2 receptor inhibitor selected from the group consisting of BM- 531 (CAS No. 284464-46-6), ozagrel hydrochloride (CAS No. 78712-43-3), and an analogue or derivative thereof.
24467. The method of item 17360 wherein the agent is a D2 dopamine receptor antagonist.
24468. The method of item 17360 wherein the agent is a Peptidyl-Prolyl Cis/Trans lsomerase Inhibitor.
24469. The method of item 17360 wherein the agent is a dopamine antagonist.
24470. The method of item 17360 wherein the agent is an anesthetic compound. 24471. The method of item 17360 wherein the agent is a clotting factor.
24472. The method of item 17360 wherein the agent is a lysyl hydrolase inhibitor.
24473. The method of item 17360 wherein the agent is a muscarinic receptor inhibitor.
24474. The method of item 17360 wherein the agent is a superoxide anion generator.
24475. The method of item 17360 wherein the agent is a steroid.
24476. The method of item 17360 wherein the agent is an antiproliferative agent selected from the group consisting of silibinin (CAS No. 22888-70-6), silymarin (CAS No. 65666-07-1), 1 ,2-hexanediol, dioctyl phthalate (CAS No. 117-81-7), zirconium (IV) oxide, glycyrrhizic acid, spermidine trihydrochloride, tetrahydrochloride, CGP 74514A, spermine tetrahydrochloride, NG-methyl-L-arginine acetate salt, galardin, and an analogue or derivative thereof.
24477. The method of item 17360 wherein the agent is a diuretic.
24478. The method of item 17360 wherein the agent is an anti-coagulant.
24479. The method of item 17360 wherein the agent is a cyclic GMP agonist. 24480. The method of item 17360 wherein the agent is an adenylate cyclase agonist.
24481. The method of item 17360 wherein the agent is an antioxidant.
24482. The method of item 17360 wherein the agent is a nitric oxide synthase inhibitor.
24483. The method of item 17360 wherein the agent is an anti-neoplastic agent selected from tirapazamine (CAS No. 27314-97-2), fludarabine (CAS No. 21679-14-1), cladribine, imatinib mesilate, and an analogue or derivative thereof.
24484. The method of item 17360 wherein the agent is a DNA synthesis inhibitor.
24485. The method of item 17360 wherein the agent is a DNA alkylating agent selected from dacarbazine (CAS No. 4342-03-4), temozolomide, procarbazine HCI, and an analogue or derivative thereof.
24486. The method of item 17360 wherein the agent is a DNA methylation inhibitor.
24487. The method of item 17360 wherein the agent is a NSAID agent.
24488. The method of item 17360 wherein the agent is a peptidylglycine alpha-hydroxylating monooxygenase inhibitor. 24489. The method of item 17360 wherein the agent is an MEK1/MEK 2 inhibitor.
24490. The method of item 17360 wherein the agent is a NO synthase inhibitor.
24491. The method of item 17360 wherein the agent is a retinoic acid receptor antagonist selected from isotretinoin (CAS No. 4759- 48-2) and an analogue or derivative thereof.
24492. The method of item 17360 wherein the agent is an ACE inhibitor.
24493. The method of item 17360 wherein the agent is a glycosylation inhibitor.
24494. The method of item 17360 wherein the agent is an intracellular calcium influx inhibitor.
24495. The method of item 17360 wherein the agent is an anti-emetic agent.
24496. The method of item 17360 wherein the agent is an acetylcholinesterase inhibitor.
24497. The method of item 17360 wherein the agent is an ALK-5 receptor antagonist.
24498. The method of item 17360 wherein the agent is a RAR/RXT antagonist. 24499. The method of item 17360 wherein the agent is an elF-2a inhibitor.
24500. The method of item 17360 wherein the agent is an S-adenosyl-L-homocysteine hydrolase inhibitor.
24501. The method of item 17360 wherein the agent is an estrogen agonist.
24502. The method of item 17360 wherein the agent is a serotonin receptor inhibitor.
24503. The method of item 17360 wherein the agent is an anti-thrombotic agent.
24504. The method of item 17360 wherein the agent is a tryptase inhibitor.
24505. The method of item 17360 wherein the agent is a pesticide.
24506. The method of item 17360 wherein the agent is a bone mineralization promotor.
24507. The method of item 17360 wherein the agent is a bisphosphonate compound selected from risedronate and an analogue or derivative thereof.
24508. The method of item 17360 wherein the agent is an anti-inflammatory compound. 24509. The method of item 17360 wherein the agent is a DNA methylation promoter.
24510. The method of item 17360 wherein the agent is an anti-spasmodic agent.
24511. The method of item 17360 wherein the agent is a protein synthesis inhibitor.
24512. The method of item 17360 wherein the agent is an D-glucosidase inhibitor.
24513. The method of item 17360 wherein the agent is a calcium channel blocker.
24514. The method of item 17360 wherein the agent is a pyruvate dehydrogenase activator.
24515. The method of item 17360 wherein the agent is a prostaglandin inhibitor.
24516. The method of item 17360 wherein the agent is a sodium channel inhibitor.
24517. The method of item 17360 wherein the agent is a serine protease inhibitor.
24518. The method of item 17360 wherein the agent is an intracellular calcium flux inhibitor. 24519. The method of item 17360 wherein the agent is a JAK2 inhibitor.
24520. The method of item 17360 wherein the agent is an androgen inhibitor.
24521. The method of item 17360 wherein the agent is an aromatase inhibitor.
24522. The method of item 17360 wherein the agent is an anti-viral agent.
24523. The method of item 17360 wherein the agent is a 5-HT inhibitor.
24524. The method of item 17360 wherein the agent is an FXR antagonist.
24525. The method of item 17360 wherein the agent is an actin polymerization and stabilization promotor.
24526. The method of item 17360 wherein the agent is an AXOR12 agonist.
24527. The method of item 17360 wherein the agent is an angiotensin Il receptor agonist.
24528. The method of item 17360 wherein the agent is a platelet aggregation inhibitor. 24529. The method of item 17360 wherein the agent is a CB1/CB2 receptor agonist.
24530. The method of item 17360 wherein the agent is a norepinephrine reuptake inhibitor.
24531. The method of item 17360 wherein the agent is a selective serotonin reuptake inhibitor.
24532. The method of item 17360 wherein the agent is a reducing agent.
24533. The method of item 17360 wherein the agent is a immuno-modulator selected from Bay 11-7085, (-)-arctigenin, idazoxan hydrochloride, and an analogue or derivative thereof.
24534. The method of item 17360 wherein the agent is isotretinoin.
24535. The method of item 17360 wherein the agent is radicicol.
24536. The method of item 17360 wherein the agent is clobetasol propionate.
24537. The method of item 17360 wherein the agent is homoharringtonine.
24538. The method of item 17360 wherein the agent is trichostatin A. 24539. The method of item 17360 wherein the agent is brefeldin A.
24540. The method of item 17360 wherein the agent is thapsigargin.
24541. The method of item 17360 wherein the agent is dolastatin 15.
24542. The method of item 17360 wherein the agent is cerivastatin.
24543. The method of item 17360 wherein the agent is jasplakinolide.
24544. The method of item 17360 wherein the agent is herbimycin A.
24545. The method of item 17360 wherein the agent is pirfenidone.
24546. The method of item 17360 wherein the agent is vinorelbine.
24547. The method of item 17360 wherein the agent is 17-DMAG.
24548. The method of item 17360 wherein the agent is tacrolimus. 24549. The method of item 17360 wherein the agent is loteprednol etabonate.
24550. The method of item 17360 wherein the agent is juglone.
24551. The method of item 17360 wherein the agent is prednisolone.
24552. The method of item 17360 wherein the agent is puromycin.
24553. The method of item 17360 wherein the agent is 3-BAABE.
24554. The method of item 17360 wherein the agent is cladribine.
24555. The method of item 17360 wherein the agent is mannose-6-phosphate.
24556. The method of item 17360 wherein the agent is 5-azacytidine.
24557. The method of item 17360 wherein the agent is Ly333531 (ruboxistaurin).
24558. The method of item 17360 wherein the agent is simvastatin. 24559. The method of any one of items 24443-24558, wherein the medical device is an intravascular device.
24560. The method of any one of items 24443-24558, wherein the medical device is a gastrointestinal stent.
24561. The method of any one of items 24443-24558, wherein the medical device is a tracheal and bronchial stent.
24562. The method of any one of items 24443-24558, wherein the medical device is a genital urinary stent.
24563. The method of any one of items 24443-24558, wherein the medical device is an ear and nose stent.
24564. The method of any one of items 24443-24558, wherein the medical device is an ear ventilation device.
24565. The method of any one of items 24443-24558, wherein the medical device is an intraocular implant.
24566. The method of any one of items 24443-24558, wherein the medical device is a vascular graft.
24567. The method of any one of items 24443-24558, wherein the medical device comprises a film or a mesh.
24568. The method of any one of items 24443-24558, wherein the medical device is a glaucoma drainage device. 24569. The method of any one of items 24443-24558, wherein the medical device is a prosthetic heart valve or a component thereof.
24570. The method of any one of items 24443-24558, wherein the medical device is a penile implant.
24571. The method of any one of items 24443-24558, wherein the medical device is an endotracheal or tracheostomy tube.
24572. The method of any one of items 24443-24558, wherein the medical device is a peritoneal dialysis catheter.
24573. The method of any one of items 24443-24558, wherein the medical device is a central nervous system shunt or a pressure monitoring device.
24574. The method of any one of items 24443-24558, wherein the medical device is an inferior vena cava filter.
24575. The method of any one of items 24443-24558, wherein the medical device is a gastrointestinal device.
24576. The method of any one of items 24443-24558, wherein the medical device is a central venous catheter.
24577. The method of any one of items 24443-24558, wherein the medical device is a ventricular assist device.
24578. The method of any one of items 24443-24558, wherein the medical device is a spinal implant. 24579. The method of any one of items 24443-24558, wherein the medical device is for treating hypertropic scar or keloid.
24580. The method of any one of items 24443-24558, wherein the medical device is a hemodialysis access device.
24581. The method of any one of items 24443-24558, wherein the medical device is a surgical adhesion barrier.
24582. The method of item 22753 wherein the agent is a cyclin dependent kinase inhibitor selected from the group consisting of CGP 74514A, bohemine, olomoucine (CAS No. 101622-51-9), indole-3-carbinol (CAS No. 700-06-1), and an analogue or derivative thereof.
24583. The method of item 22753 wherein the agent is an endothelial growth factor receptor (EGF-R) kinase inhibitor selected from - the group consisting of lavendustin A (CAS No. 125697-92-9), SU 1498 (a VEGF-R inhibitor), and an analogue or derivative thereof.
24584. The method of item 22753 wherein the agent is mevastatin (a fibrinogen antagonist).
24585. The method of item 22753 wherein the agent is vincamine (a microtubule inhibitor).
24586. The method of item 22753 wherein the agent is emodin (CAS No. 518-82-1) (an NF kappa B inhibitor) or Bay 11-7085 (an NF kappa B inhibitor).
24587. The method of item 22753 wherein the agent is SKF86002 (CAS No. 72873-74-6) (a p38 MAP kinase inhibitor). 24588. The method of item 22753 wherein the agent is a peroxisome proliferators-activated receptor (PPAR) agonist selected from the group consisting of LBM642, WY-14,643 (CAS No. 50892-23-4), GW7647, fenofibric acid (CAS No. 42017-89-0), MCC-555 (CAS No. 161600-01-7), GW9662, GW1929, GW501516, L-165,041 (CAS No. 79558- 09-1), and an analogue or derivative thereof.
24589. The method of item 22753 wherein the agent is a phosphodiesterase (PDE) inhibitor selected from the group consisting of theobromine (CAS No. 83-67-0), papverine hydrochloride (CAS No. 61-25- 6), quercetin dehydrate (CAS No. 6151-25-3), YM 976 (CAS No. 191219- 80-4), irsogladine (CAS No. 57381-26-7), and an analogue or derivative thereof.
24590. The method of item 22753 wherein the agent is a tumor necrosis factor antagonist.
24591. The method of item 22753 wherein the agent is herbimycin A (a tyrosine kinase inhibitor).
24592. The method of item 22753 wherein the agent is erucylphosphocholine.
24593. The method of item 22753 wherein the agent is alphastatin.
24594. The method of item 22753 wherein the agent is etanercept.
24595. The method of item 22753 wherein the agent is humicade. 24596. The method of item 22753 wherein the agent is gefitinib.
24597. The method of item 22753 wherein the agent is a histamine receptor antagonist selected from the group consisting of phenothiazines (e.g., promethazine), alkylamines (e.g., chlorpheniramine (CAS No. 7054-11-7), brompheniramine (CAS No. 980-71-2), fexofenadine hydrochloride, promethazine hydrochloride, loratadine, ketotifen fumarate salt, and acrivastine), methylxanthines (e.g., theophylline, theobromine, and caffeine), cimetidine (available under the tradename TAGAMET from SmithKline Beecham Phamaceutical Co., Wilmington, DE), ranitidine (available under the tradename ZANTAC from Warner Lambert Company, Morris Plains, NJ), famotidine (available under the tradename PEPCID from Merck & Co., Whitehouse Station, NJ), nizatidine (available under the tradename AXID from Reliant Pharmaceuticals, Inc., Liberty Corner, NJ)1 nizatidine, and roxatidine acetate (CAS No. 78628-28-1), H3 receptor antagonists (e.g., thioperamide and thioperamide maleate salt), and antihistamines (e.g., tricyclic dibenozoxepins, ethanolamines, ethylenediamines, piperizines, piperidines, and pthalazinones).
24598. The method of item 22753 wherein the agent is an alpha adrenergic receptor antagonist.
24599. The method of item 22753 wherein the agent is an anti-psychotic compound.
24600. The method of item 22753 wherein the agent is a CaM kinase Il inhibitor.
24601. The method of item 22753 wherein the agent is a G protein agonist. 24602. The method of item 22753 wherein the agent is an antibiotic selected from the group consisting of apigenin (Cas No. 520- 36-5), ampicillin sodium salt (CAS No. 69-52-3), puromycin, and an analogue or derivative thereof.
24603. The method of item 22753 wherein the agent is an anti-microbial agent.
24604. The method of item 22753 wherein the agent is a DNA topoisomerase inhibitor selected from the group consisting of D- lapachone (CAS No. 4707-32-8)" -arctigenin (CAS No. 7770-78-7), aurintricarboxylic acid, and an analogue or derivative thereof.
24605. The method of item 22753 wherein the agent is a thromboxane A2 receptor inhibitor selected from the group consisting of BM- 531 (CAS No. 284464-46-6), ozagrel hydrochloride (CAS No. 78712-43-3), and an analogue or derivative thereof.
24606. The method of item 22753 wherein the agent is a D2 dopamine receptor antagonist.
24607. The method of item 22753 wherein the agent is a Peptidyl-Prolyl Cis/Trans lsomerase Inhibitor.
24608. The method of item 22753 wherein the agent is a dopamine antagonist.
24609. The method of item 22753 wherein the agent is an anesthetic compound. 24610. The method of item 22753 wherein the agent is a clotting factor.
24611. The method of item 22753 wherein the agent is a lysyl hydrolase inhibitor.
24612. The method of item 22753 wherein the agent is a muscarinic receptor inhibitor.
24613. The method of item 22753 wherein the agent is a superoxide anion generator.
24614. The method of item 22753 wherein the agent is a steroid.
24615. The method of item 22753 wherein the agent is - an anti-proliferative agent selected from the group consisting of silibinin (CAS No. 22888-70-6), silymarin (CAS No. 65666-07-1), 1,2-hexanediol, dioctyl phthalate (CAS No. 117-81-7), zirconium (IV) oxide, glycyrrhizic acid, spermidine trihydrochloride, tetrahydrochloride, CGP 74514A, spermine tetrahydrochloride, NG-methyl-L-arginine acetate salt, galardin, and an analogue or derivative thereof.
24616. The method of item 22753 wherein the agent is a diuretic.
24617. The method of item 22753 wherein the agent is an anti-coagulant.
24618. The method of item 22753 wherein the agent is a cyclic GMP agonist. 24619. The method of item 22753 wherein the agent is an adenylate cyclase agonist.
24620. The method of item 22753 wherein the agent is an antioxidant.
24621. The method of item 22753 wherein the agent is a nitric oxide synthase inhibitor.
24622. The method of item 22753 wherein the agent is an anti-neoplastic agent selected from tirapazamine (CAS No. 27314-97-2), fludarabine (CAS No. 21679-14-1), cladribine, imatinib mesilate, and an analogue or derivative thereof.
24623. The method of item 22753 wherein the agent is a DNA synthesis inhibitor.
24624. The method of item 22753 wherein the agent is a DNA alkylating agent selected from dacarbazine (CAS No. 4342-03-4), temozolomide, procarbazine HCI, and an analogue or derivative thereof.
24625. The method of item 22753 wherein the agent is a DNA methylation inhibitor.
24626. The method of item 22753 wherein the agent is a NSAID agent.
24627. The method of item 22753 wherein the agent is a peptidylglycine alpha-hydroxylating monooxygenase inhibitor. 24628. The method of item 22753 wherein the agent is an MEK1/MEK 2 inhibitor.
24629. The method of item 22753 wherein the agent is a NO synthase inhibitor.
24630. The method of item 22753 wherein the agent is a retinoic acid receptor antagonist selected from isotretinoin (CAS No. 4759- 48-2) and an analogue or derivative thereof.
24631. The method of item 22753 wherein the agent is an ACE inhibitor.
24632. The method of item 22753 wherein the agent is a glycosylation inhibitor.
24633. The method of item 22753 wherein the agent is an intracellular calcium influx inhibitor.
24634. The method of item 22753 wherein the agent is an anti-emetic agent.
24635. The method of item 22753 wherein the agent is an acetylcholinesterase inhibitor.
24636. The method of item 22753 wherein the agent is an ALK-5 receptor antagonist.
24637. The method of item 22753 wherein the agent is a RAR/RXT antagonist. 24638. The method of item 22753 wherein the agent is an elF-2a inhibitor.
24639. The method of item 22753 wherein the agent is an S-adenosyl-L-homocysteine hydrolase inhibitor.
24640. The method of item 22753 wherein the agent is an estrogen agonist.
24641. The method of item 22753 wherein the agent is a serotonin receptor inhibitor.
24642. The method of item 22753 wherein the agent is an anti-thrombotic agent.
24643. The method of item 22753 wherein the agent is a tryptase inhibitor.
24644. The method of item 22753 wherein the agent is a pesticide.
24645. The method of item 22753 wherein the agent is a bone mineralization promotor.
24646. The method of item 22753 wherein the agent is a bisphosphonate compound selected from risedronate and an analogue or derivative thereof.
24647. The method of item 22753 wherein the agent is an anti-inflammatory compound. 24648. The method of item 22753 wherein the agent is a DNA methylation promotor.
24649. The method of item 22753 wherein the agent is an anti-spasmodic agent.
24650. The method of item 22753 wherein the agent is a protein synthesis inhibitor.
24651. The method of item 22753 wherein the agent is an D-glucosidase inhibitor.
24652. The method of item 22753 wherein the agent is a calcium channel blocker.
24653. The method of item 22753 wherein the agent is a pyruvate dehydrogenase activator.
24654. The method of item 22753 wherein the agent is a prostaglandin inhibitor.
24655. The method of item 22753 wherein the agent is a sodium channel inhibitor.
24656. The method of item 22753 wherein the agent is a serine protease inhibitor.
24657. The method of item 22753 wherein the agent is an intracellular calcium flux inhibitor. 24658. The method of item 22753 wherein the agent is a JAK2 inhibitor.
24659. The method of item 22753 wherein the agent is an androgen inhibitor.
24660. The method of item 22753 wherein the agent is an aromatase inhibitor.
24661. The method of item 22753 wherein the agent is an anti-viral agent.
24662. The method of item 22753 wherein the agent is a 5-HT inhibitor.
24663. The method of item 22753 wherein the agent is an FXR antagonist.
24664. The method of item 22753 wherein the agent is an actin polymerization and stabilization promotor.
24665. The method of item 22753 wherein the agent is an AXOR12 agonist.
24666. The method of item 22753 wherein the agent is an angiotensin Il receptor agonist.
24667. The method of item 22753 wherein the agent is a platelet aggregation inhibitor. 24668. The method of item 22753 wherein the agent is a CB1/CB2 receptor agonist.
24669. The method of item 22753 wherein the agent is a norepinephrine reuptake inhibitor.
24670. The method of item 22753 wherein the agent is a selective serotonin reuptake inhibitor.
24671. The method of item 22753 wherein the agent is a reducing agent.
24672. The method of item 22753 wherein the agent is a immuno-modulator selected from Bay 11-7085, (-)-arctigenin, idazoxan hydrochloride, and an analogue or derivative thereof.
24673. The method of item 22753 wherein the agent is isotretinoin.
24674. The method of item 22753 wherein the agent is radicicol.
24675. The method of item 22753 wherein the agent is clobetasol propionate.
24676. The method of item 22753 wherein the agent is homoharringtonine.
24677. The method of item 22753 wherein the agent is trichostatin A. 24678. The method of item 22753 wherein the agent is brefeldin A.
24679. The method of item 22753 wherein the agent is thapsigargin.
24680. The method of item 22753 wherein the agent is dolastatin 15.
24681. The method of item 22753 wherein the agent is cerivastatin.
24682. The method of item 22753 wherein the agent is jasplakinolide.
24683. The method of item 22753 wherein the agent is herbimycin A.
24684. The method of item 22753 wherein the agent is pirfenidone.
24685. The method of item 22753 wherein the agent is vinorelbine.
24686. The method of item 22753 wherein the agent is 17-DMAG.
24687. The method of item 22753 wherein the agent is tacrolimus. 24688. The method of item 22753 wherein the agent is loteprednol etabonate.
24689. The method of item 22753 wherein the agent is juglone.
24690. The method of item 22753 wherein the agent is prednisolone.
24691. The method of item 22753 wherein the agent is puromycin.
24692. The method of item 22753 wherein the agent is 3-BAABE.
24693. The method of item 22753 wherein the agent is cladribine.
24694. The method of item 22753 wherein the agent is mannose-6-phosphate.
24695. The method of item 22753 wherein the agent is 5-azacytidine.
24696. The method of item 22753 wherein the agent is Ly333531 (ruboxistaurin).
24697. The method of item 22753 wherein the agent is simvastatin. 24698. The method of any one of items 24582-24697 wherein the site in need thereof is between a dural sleeve and paravertebral musculature in a patient post-laminectomy.
24699. The method of any one of items 24582-24697 wherein the site in need thereof is a spinal nerve at a laminectomy site in a patient.
24700. The method of any one of items 24582-24697 wherein the site in need thereof is a tissue around a spinal nerve at a laminectomy site.
24701. The method of any one of items 24582-24697 wherein the site in need thereof is a site of a surgical disc resection.
24702. The method of any one of items 24582-24697 wherein the site in need thereof is a site of a microdiscectomy.
24703. The method of any one of items 24582-24697 wherein the site in need thereof is a site of a neurosurgical (brain) procedure.
24704. The method of any one of items 24582-24697 wherein the site in need thereof is a spinal surgical site.
24705. The method of any one of items 24582-24697 wherein the site in need thereof is an epidural tissue.
24706. The method of any one of items 24582-24697 wherein the site in need thereof is a dural tissue. 24707. The method of any one of items 24582-24697 wherein the site in need thereof is a gynecological site.
24708. The method of any one of items 24582-24697 wherein the site in need thereof is a tissue surface of the pelvic side wall.
24709. The method of any one of items 24582-24697 wherein the site in need thereof is a peritoneal cavity.
24710. The method of any one of items 24582-24697 wherein the site in need thereof is a pelvic cavity.
24711. The method of any one of items 24582-24697 wherein the site in need thereof is a site of a laparotomy.
24712. The method of any one of items 24582-24697 wherein the site in need thereof is an endoscopic procedure.
24713. The method of any one of items 24582-24697 wherein the site in need thereof is a site of a hernia repair.
24714. The method of any one of items 24582-24697 wherein the site in need thereof is a site of cholecystectomy.
24715. The method of any one of items 24582-24697 wherein the site in need thereof is a site of a cardiac procedure.
24716. The method of any one of items 24582-24697 wherein the site in need thereof is a site of cardiac transplant surgery. 24717. The method of any one of items 24582-24697 wherein the site in need thereof is a site of cardiac vascular repair.
24718. The method of any one of items 24582-24697 wherein the site in need thereof is a site of a heart valve replacement.
24719. The method of any one of items 24582-24697 wherein the site in need thereof is a site of pericardial surgery.
24720. The method of any one of items 24582-24697 wherein the site in need thereof is a site of an orthopedic surgical procedure.
24721. The method of any one of items 24582-24697 wherein the site in need thereof is a site of a torn ligament.
24722. The method of any one of items 24582-24697 wherein the site in need thereof is a site of a joint injury.
24723. The method of any one of items 24582-24697 wherein the site in need thereof is a site of a tendon injury.
24724. The method of any one of items 24582-24697 wherein the site in need thereof is a site of a cartilage injury.
24725. The method of any one of items 24582-24697 wherein the site in need thereof is a site of a muscle injury.
24726. The method of any one of items 24582-24697 wherein the site in need thereof is a site of a nerve injury. 24727. The method of any one of items 24582-24697 wherein the site in need thereof is a site of a cosmetic surgical procedure.
24728. The method of any one of items 24582-24697 wherein the site in need thereof is a site of a reconstructive surgical procedure.
24729. The method of any one of items 24582-24697 wherein the site in need thereof is a site of a breast implant.
24730. The method of any one of items 24582-24697 wherein the composition is delivered in conjunction with the placement of a medical implant.
24731. The method of any one of items 24582-24697 wherein the composition is delivered in conjunction with the placement of a medical implant, and the composition is placed on tissue adjacent to the medical implant.
24732. The method of any one of items 24582-24697 wherein the composition is delivered in conjunction with the placement of a medical implant, and the composition is placed on the medical implant.
24733. The method of any one of items 24582-24697 wherein the composition is delivered via an endoscope.
24734. The method of any one of items 24582-24697 wherein the composition is delivered through a needle.
24735. The method of any one of items 24582-24697 wherein the composition is delivered through a catheter. 24736. The method of any one of items 24582-24697 wherein the composition is delivered at the time of a surgery.
24737. The method of any one of items 24582-24697 wherein the composition is delivered using fluoroscopic guidance.
24738. The method of any one of items 24582-24697 wherein the composition comprises an antithrombotic agent.
24739. The method of any one of items 24582-24697 wherein the composition comprises a naturally occurring polymer.
24740. The method of any one of items 24582-24697 wherein the composition comprises protein.
24741. The method of any one of items 24582-24697 wherein the composition comprises carbohydrate.
24742. The method of any one of items 24582-24697 wherein the composition comprises biodegradable polymer.
24743. The method of any one of items 24582-24697 wherein the composition comprises nonbiodegradable polymer.
24744. The method of any one of items 24582-24697 wherein the composition comprises collagen.
24745. The method of any one of items 24582-24697 wherein the composition comprises methylated collagen. 24746. The method of any one of items 24582-24697 wherein the composition comprises fibrinogen.
24747. The method of any one of items 24582-24697 wherein the composition comprises thrombin.
24748. The method of any one of items 24582-24697 wherein the composition comprises blood plasma.
24749. The method of any one of items 24582-24697 wherein the composition comprises calcium salt.
24750. The method of any one of items 24582-24697 wherein the composition comprises an antifibronolytic agent.
24751. The method of any one of items 24582-24697 wherein the composition comprises a fibrinogen analog.
24752. The method of any one of items 24582-24697 wherein the composition comprises albumin.
24753. The method of any one of items 24582-24697 wherein the composition comprises plasminogen.
24754. The method of any one of items 24582-24697 wherein the composition comprises von Willebrands factor.
24755. The method of any one of items 24582-24697 wherein the composition comprises Factor VIII. 24756. The method of any one of items 24582-24697 wherein the composition comprises hypoallergenic collagen.
24757. The method of any one of items 24582-24697 wherein the composition comprises atelopeptidic collagen.
24758. The method of any one of items 24582-24697 wherein the composition comprises telopeptide collagen.
24759. The method of any one of items 24582-24697 wherein the composition comprises crosslinked collagen.
24760. The method of any one of items 24582-24697 wherein the composition comprises aprotinin.
24761. The method of any one of items 24582-24697 . wherein the composition comprises epsilon-amino-n-caproic acid.
24762. The method of any one of items 24582-24697 wherein the composition comprises gelatin.
24763. The method of any one of items 24582-24697 wherein the composition comprises protein conjugates.
24764. The method of any one of items 24582-24697 wherein the composition comprises gelatin conjugates.
24765. The method of any one of items 24582-24697 wherein the composition comprises a synthetic polymer. 24766. The method of any one of items 24582-24697 wherein the composition comprises a synthetic isocyanate-containing compound.
24767. The method of any one of items 24582-24697 wherein the composition comprises a synthetic thiol-containing compound.
24768. The method of any one of items 24582-24697 wherein the composition comprises a synthetic compound containing at least two thiol groups.
24769. The method of any one of items 24582-24697 wherein the composition comprises a synthetic compound containing at least three thiol groups.
24770. The method of any one of items 24582-24697 wherein the composition comprises a synthetic compound containing at least four thiol groups.
24771. The method of any one of items 24582-24697 wherein the composition comprises a synthetic amino-containing compound.
24772. The method of any one of items 24582-24697 wherein the composition comprises a synthetic compound containing at least two amino groups.
24773. The method of any one of items 24582-24697 wherein the composition comprises a synthetic compound containing at least three amino groups. 24774. The method of any one of items 24582-24697 wherein the composition comprises a synthetic compound containing at least four amino groups.
24775. The method of any one of items 24582-24697 wherein the composition comprises a synthetic compound comprising a carbonyl-oxygen-succinimidyl group.
24776. The method of any one of items 24582-24697 wherein the composition comprises a synthetic compound comprising at least two carbonyl-oxygen-succinimidyl groups.
24777. The method of any one of items 24582-24697 wherein the composition comprises a synthetic compound comprising at least three carbonyl-oxygen-succinimidyl groups.
24778. The method of any one of items 24582-24697 wherein the composition comprises a synthetic compound comprising at least four carbonyl-oxygen-succinimidyl groups.
24779. The method of any one of items 24582-24697 wherein the composition comprises a synthetic polyalkylene oxide- containing compound.
24780. The method of any one of items 24582-24697 wherein the composition comprises a synthetic compound comprising both polyalkylene oxide and biodegradable polyester blocks.
24781. The method of any one of items 24582-24697 wherein the composition comprises a synthetic polyalkylene oxide- containing compound having reactive amino groups. 24782. The method of any one of items 24582-24697 wherein the composition comprises a synthetic polyalkylene oxide- containing compound having reactive thiol groups.
24783. The method of any one of items 24582-24697 wherein the composition comprises a synthetic polyalkylene oxide- containing compound having reactive carbonyl-oxygen-succinimidyl groups.
24784. The method of any one of items 24582-24697 wherein the composition comprises a synthetic compound comprising a biodegradable polyester block.
24785. The method of any one of items 24582-24697 wherein the composition comprises a synthetic polymer formed in whole or part from lactic acid or lactide.
24786. The method of any one of items 24582-24697 wherein the composition comprises a synthetic polymer formed in whole or part from glycolic acid or glycolide.
24787. The method of any one of items 24582-24697 wherein the composition comprises polylysine.
24788. The method of any one of items 24582-24697 wherein the composition comprises (a) protein and (b) a compound comprising a polyalkylene oxide portion.
24789. The method of any one of items 24582-24697 wherein the composition comprises (a) protein and (b) polylysine. 24790. The method of any one of items 24582-24697 wherein the composition comprises (a) protein and (b) a compound having at least four thiol groups.
24791. The method of any one of items 24582-24697 wherein the composition comprises (a) protein and (b) a compound having at least four amino groups.
24792. The method of any one of items 24582-24697 wherein the composition comprises (a) protein and (b) a compound having at least four carbonyl-oxygen-succinimide groups.
24793. The method of any one of items 24582-24697 wherein the composition comprises (a) protein and (b) a compound having a biodegradable region formed from reactants selected from lactic acid, lactide, glycolic acid, glycolide, and epison-caprolactone.
24794. The method of any one of items 24582-24697 wherein the composition comprises (a) collagen and (b) a compound comprising a polyalkylene oxide portion.
24795. The method of any one of items 24582-24697 wherein the composition comprises (a) collagen and (b) polylysine.
24796. The method of any one of items 24582-24697 wherein the composition comprises (a) collagen and (b) a compound having at least four thiol groups.
24797. The method of any one of items 24582-24697 wherein the composition comprises (a) collagen and (b) a compound having at least four amino groups. 24798. The method of any one of items 24582-24697 wherein the composition comprises (a) collagen and (b) a compound having at least four carbonyl-oxygen-succinimide groups.
24799. The method of any one of items 24582-24697 wherein the composition comprises (a) collagen and (b) a compound having a biodegradable region formed from reactants selected from lactic acid, lactide, glycolic acid, glycolide, and epison-caprolactone.
24800. The method of any one of items 24582-24697 wherein the composition comprises (a) methylated collagen and (b) a compound comprising a polyalkylene oxide portion.
24801. The method of any one of items 24582-24697 wherein the composition comprises (a) methylated collagen and (b) polylysine.
24802. The method of any one of items 24582-24697 wherein the composition comprises (a) methylated collagen and (b) a compound having at least four thiol groups.
24803. The method of any one of items 24582-24697 wherein the composition comprises (a) methylated collagen and (b) a compound having at least four amino groups.
24804. The method of any one of items 24582-24697 wherein the composition comprises (a) methylated collagen and (b) a compound having at least four carbonyl-oxygen-succinimide groups.
24805. The method of any one of items 24582-24697 wherein the composition comprises (a) methylated collagen and (b) a compound having a biodegradable region formed from reactants selected from lactic acid, lactide, giycolic acid, glycolide, and epison-caprolactone.
24806. The method of any one of items 24582-24697 wherein the composition comprises hyaluronic acid.
24807. The method of any one of items 24582-24697 wherein the composition comprises a hyaluronic acid derivative.
24808. The method of any one of items 24582-24697 wherein the composition comprises pentaerythritol poly(ethylene glycol)ether tetra-sulfhydryl of number average molecular weight between 3,000 and 30,000.
24809. The method of any one of items 24582-24697 wherein the composition comprises pentaerythritol poly(ethylene _ glycol)ether tetra-amino of number average molecular weight between 3,000 and 30,000.
24810. The method of any one of items 24582-24697 wherein the composition comprises (a) a synthetic compound having a number average molecular weight between 3,000 and 30,000 and comprising a polyalkylene oxide region and multiple nucleophilic groups, and (b) a synthetic compound having a number average molecular weight between 3,000 and 30,000 and comprising a polyalkylene oxide region and multiple electrophilic groups.
24811. The method of any one of items 24582-24697 wherein the composition comprises a colorant. 24812. The method of any one of items 24582-24697 wherein the composition is sterile.
24813. The method of item 22986 or 22987 wherein the agent is a cyclin dependent kinase inhibitor selected from the group consisting of CGP 74514A, bohemine, olomoucine (CAS No. 101622-51-9), indole-3-carbinol (CAS No. 700-06-1), and an analogue or derivative thereof.
24814. The method of item 22986 or 22987 wherein the agent is an endothelial growth factor receptor (EGF-R) kinase inhibitor selected from the group consisting of lavendustin A (CAS No. 125697-92-9), SU 1498 (a VEGF-R inhibitor), and an analogue or derivative thereof.
24815. The method of item 22986 or 22987 wherein the agent is mevastatin (a fibrinogen antagonist).
24816. The method of item 22986 or 22987 wherein the agent is vincamine (a microtubule inhibitor).
24817. The method of item 22986 or 22987 wherein the agent is emodin (CAS No. 518-82-1) (an NF kappa B inhibitor) or Bay 11- 7085 (an NF kappa B inhibitor).
24818. The method of item 22986 or 22987 wherein the agent is SKF86002 (CAS No. 72873-74-6) (a p38 MAP kinase inhibitor).
24819. The method of item 22986 or 22987 wherein the agent is a peroxisome proliferators-activated receptor (PPAR) agonist selected from the group consisting of LBM642, WY-14,643 (CAS No. 50892- 23-4), GW7647, fenofibric acid (CAS No. 42017-89-0), MCC-555 (CAS No. 161600-01-7), GW9662, GW1929, GW501516, L-165,041 (CAS No. 79558- 09-1), and an analogue or derivative thereof.
24820. The method of item 22986 or 22987 wherein the agent is a phosphodiesterase (PDE) inhibitor selected from the group consisting of theobromine (CAS No. 83-67-0), papverine hydrochloride (CAS No. 61-25-6), quercetin dehydrate (CAS No. 6151-25-3), YM 976 (CAS No. 191219-80-4), irsogladine (CAS No. 57381-26-7), and an analogue or derivative thereof.
24821. The method of item 22986 or 22987 wherein the agent is a tumor necrosis factor antagonist.
24822. The method of item 22986 or 22987 wherein the agent is herbimycin A (a tyrosine kinase inhibitor).
24823. The method of item 22986 or 22987 wherein the agent is erucylphosphocholine.
24824. The method of item 22986 or 22987 wherein the agent is alphastatin.
24825. The method of item 22986 or 22987 wherein the agent is etanercept.
24826. The method of item 22986 or 22987 wherein the agent is humicade.
24827. The method of item 22986 or 22987 wherein the agent is gefitinib. 24828. The method of item 22986 or 22987 wherein the agent is a histamine receptor antagonist selected from the group consisting of phenothiazines (e.g., promethazine), alkylamines (e.g., chlorpheniramine (CAS No. 7054-11-7), brompheniramine (CAS No. 980-71-2), fexofenadine hydrochloride, promethazine hydrochloride, loratadine, ketotifen fumarate salt, and acrivastine), methylxanthines (e.g., theophylline, theobromine, and caffeine), cimetidine (available under the tradename TAGAMET from SmithKline Beecham Phamaceutical Co., Wilmington, DE), ranitidine (available under the tradename ZANTAC from Warner Lambert Company, Morris Plains, NJ), famotidine (available under the tradename PEPCID from Merck & Co., Whitehouse Station, NJ), nizatidine (available under the tradename AXID from Reliant Pharmaceuticals, Inc., Liberty Corner, NJ), nizatidine, and roxatidine acetate (CAS No. 78628-28-1), H3 receptor antagonists (e.g., thioperamide and thioperamide maleate salt), and antihistamines (e.g., tricyclic dibenozoxepins, ethanolamines, ethylenediamines, piperizines, piperidines, and pthalazinones).
24829. The method of item 22986 or 22987 wherein the agent is an alpha adrenergic receptor antagonist.
24830. The method of item 22986 or 22987 wherein the agent is an anti-psychotic compound.
24831. The method of item 22986 or 22987 wherein the agent is a CaM kinase Il inhibitor.
24832. The method of item 22986 or 22987 wherein the agent is a G protein agonist.
24833. The method of item 22986 or 22987 wherein the agent is an antibiotic selected from the group consisting of apigenin (Cas No. 520-36-5), ampicillin sodium salt (CAS No. 69-52-3), puromycin, and an analogue or derivative thereof.
24834. The method of item 22986 or 22987 wherein the agent is an anti-microbial agent.
24835. The method of item 22986 or 22987 wherein the agent is a DNA topoisomerase inhibitor selected from the group consisting of D-lapachone (CAS No. 4707-32-8)~-arctigenin (CAS No. 7770-78-7), aurintricarboxylic acid, and an analogue or derivative thereof.
24836. The method of item 22986 or 22987 wherein the agent is a thromboxane A2 receptor inhibitor selected from the group consisting of BM-531 (CAS No. 284464-46-6), ozagrel hydrochloride (CAS No. 78712-43-3), and an analogue or derivative thereof.
24837. The method of item 22986 or 22987 wherein the agent is a D2 dopamine receptor antagonist.
24838. The method of item 22986 or 22987 wherein the agent is a Peptidyl-Prolyl Cis/Trans lsomerase Inhibitor.
24839. The method of item 22986 or 22987 wherein the agent is a dopamine antagonist.
24840. The method of item 22986 or 22987 wherein the agent is an anesthetic compound.
24841. The method of item 22986 or 22987 wherein the agent is a clotting factor. 24842. The method of item 22986 or 22987 wherein the agent is a lysyl hydrolase inhibitor.
24843. The method of item 22986 or 22987 wherein the agent is a muscarinic receptor inhibitor.
24844. The method of item 22986 or 22987 wherein the agent is a superoxide anion generator.
24845. The method of item 22986 or 22987 wherein the agent is a steroid.
24846. The method of item 22986 or 22987 wherein the agent is an anti-proliferative agent selected from the group consisting of silibinin (CAS No. 22888-70-6), silymarin (CAS No. 65666-07-1), 1 ,2- hexanediol, dioctyl phthalate (CAS No. 117-81-7), zirconium (IV) oxide, glycyrrhizic acid, spermidine trihydrochloride, tetrahydrochloride, CGP 74514A, spermine tetrahydrochloride, NG-methyl-L-arginine acetate salt, galardin, and an analogue or derivative thereof.
24847. The method of item 22986 or 22987 wherein the agent is a diuretic.
24848. The method of item 22986 or 22987 wherein the agent is an anti-coagulant.
24849. The method of item 22986 or 22987 wherein the agent is a cyclic GMP agonist.
24850. The method of item 22986 or 22987 wherein the agent is an adenylate cyclase agonist. 24851. The method of item 22986 or 22987 wherein the agent is an antioxidant.
24852. The method of item 22986 or 22987 wherein the agent is a nitric oxide synthase inhibitor.
24853. The method of item 22986 or 22987 wherein the agent is an anti-neoplastic agent selected from tirapazamine (CAS No. 27314-97-2), fludarabine (CAS No. 21679-14-1), cladribine, imatinib mesilate, and an analogue or derivative thereof.
24854. The method of item 22986 or 22987 wherein the agent is a DNA synthesis inhibitor.
24855. The method of item 22986 or 22987 wherein the agent is a DNA alkylating agent selected from dacarbazine (CAS No. 4342- 03-4), temozolomide, procarbazine HCI, and an analogue or derivative thereof.
24856. The method of item 22986 or 22987 wherein the agent is a DNA methylation inhibitor.
24857. The method of item 22986 or 22987 wherein the agent is a NSAID agent.
24858. The method of item 22986 or 22987 wherein the agent is a peptidylglycine alpha-hydroxylating monooxygenase inhibitor.
24859. The method of item 22986 or 22987 wherein the agent is an MEK1/MEK 2 inhibitor. 24860. The method of item 22986 or 22987 wherein the agent is a NO synthase inhibitor.
24861. The method of item 22986 or 22987 wherein the agent is a retinoic acid receptor antagonist selected from isotretinoin (CAS No. 4759-48-2) and an analogue or derivative thereof.
24862. The method of item 22986 or 22987 wherein the agent is an ACE inhibitor.
24863. The method of item 22986 or 22987 wherein the agent is a glycosylation inhibitor.
24864. The method of item 22986 or 22987 wherein the agent is an intracellular calcium influx inhibitor.
24865. The method of item 22986 or 22987 wherein the agent is an anti-emetic agent.
24866. The method of item 22986 or 22987 wherein the agent is an acetylcholinesterase inhibitor.
24867. The method of item 22986 or 22987 wherein the agent is an ALK-5 receptor antagonist.
24868. The method of item 22986 or 22987 wherein the agent is a RAR/RXT antagonist.
24869. The method of item 22986 or 22987 wherein the agent is an elF-2a inhibitor. 24870. The method of item 22986 or 22987 wherein the agent is an S-adenosyl-L-homocysteine hydrolase inhibitor.
24871. The method of item 22986 or 22987 wherein the agent is an estrogen agonist.
24872. The method of item 22986 or 22987 wherein the agent is a serotonin receptor inhibitor.
24873. The method of item 22986 or 22987 wherein the agent is an anti-thrombotic agent.
24874. The method of item 22986 or 22987 wherein the agent is a tryptase inhibitor.
24875. The method of item 22986 or 22987 wherein the agent is a pesticide.
24876. The method of item 22986 or 22987 wherein the agent is a bone mineralization promotor.
24877. The method of item 22986 or 22987 wherein the agent is a bisphosphonate compound selected from risedronate and an analogue or derivative thereof.
24878. The method of item 22986 or 22987 wherein the agent is an anti-inflammatory compound.
24879. The method of item 22986 or 22987 wherein the agent is a DNA methylation promotor. 24880. The method of item 22986 or 22987 wherein the agent is an anti-spasmodic agent.
24881. The method of item 22986 or 22987 wherein the agent is a protein synthesis inhibitor.
24882. The method of item 22986 or 22987 wherein the agent is an D-glucosidase inhibitor.
24883. The method of item 22986 or 22987 wherein the agent is a calcium channel blocker.
24884. The method of item 22986 or 22987 wherein the agent is a pyruvate dehydrogenase activator.
24885. The method of item 22986 or 22987 wherein the agent is a prostaglandin inhibitor.
24886. The method of item 22986 or 22987 wherein the agent is a sodium channel inhibitor.
24887. The method of item 22986 or 22987 wherein the agent is a serine protease inhibitor.
24888. The method of item 22986 or 22987 wherein the agent is an intracellular calcium flux inhibitor.
24889. The method of item 22986 or 22987 wherein the agent is a JAK2 inhibitor. 24890. The method of item 22986 or 22987 wherein the agent is an androgen inhibitor.
24891. The method of item 22986 or 22987 wherein the agent is an aromatase inhibitor.
24892. The method of item 22986 or 22987 wherein the agent is an anti-viral agent.
24893. The method of item 22986 or 22987 wherein the agent is a 5-HT inhibitor.
24894. The method of item 22986 or 22987 wherein the agent is an FXR antagonist.
24895. The method of item 22986 or 22987 wherein the agent is an actin polymerization and stabilization promotor.
24896. The method of item 22986 or 22987 wherein the agent is an AXOR12 agonist.
24897. The method of item 22986 or 22987 wherein the agent is an angiotensin Il receptor agonist.
24898. The method of item 22986 or 22987 wherein the agent is a platelet aggregation inhibitor.
24899. The method of item 22986 or 22987 wherein the agent is a CB1/CB2 receptor agonist. 24900. The method of item 22986 or 22987 wherein the agent is a norepinephrine reuptake inhibitor.
24901. The method of item 22986 or 22987 wherein the agent is a selective serotonin reuptake inhibitor.
24902. The method of item 22986 or 22987 wherein the agent is a reducing agent.
24903. The method of item 22986 or 22987 wherein the agent is a immuno-modulator selected from Bay 11-7085, (-)-arctigenin, idazoxan hydrochloride, and an analogue or derivative thereof.
24904. The method of item 22986 or 22987 wherein the agent is isotretinoin.
' 24905. The method of item 22986 or 22987 wherein the agent is radicicol.
24906. The method of item 22986 or 22987 wherein the agent is clobetasol propionate.
24907. The method of item 22986 or 22987 wherein the agent is homoharringtonine.
24908. The method of item 22986 or 22987 wherein the agent is trichostatin A.
24909. The method of item 22986 or 22987 wherein the agent is brefeldin A. 24910. The method of item 22986 or 22987 wherein the agent is thapsigargin.
24911. The method of item 22986 or 22987 wherein the agent is dolastatin 15.
24912. The method of item 22986 or 22987 wherein the agent is cerivastatin.
24913. The method of item 22986 or 22987 wherein the agent is jasplakinolide.
24914. The method of item 22986 or 22987 wherein the agent is herbimycin A.
24915. The method of item 22986 or 22987 wherein the agent is pirfenidone.
24916. The method of item 22986 or 22987 wherein the agent is vinorelbine.
24917. The method of item 22986 or 22987 wherein the agent is 17-DMAG.
24918. The method of item 22986 or 22987 wherein the agent is tacrolimus.
24919. The method of item 22986 or 22987 wherein the agent is loteprednol etabonate. 24920. The method of item 22986 or 22987 wherein the agent is juglone.
24921. The method of item 22986 or 22987 wherein the agent is prednisolone.
24922. The method of item 22986 or 22987 wherein the agent is puromycin.
24923. The method of item 22986 or 22987 wherein the agent is 3-BAABE.
24924. The method of item 22986 or 22987 wherein the agent is cladribine.
24925. The method of item 22986 or 22987 wherein the agent is mannose-6-phosphate.
24926. The method of item 22986 or 22987 wherein the agent is 5-azacytidine.
24927. The method of item 22986 or 22987 wherein the agent is Ly333531 (ruboxistaurin).
24928. The method of item 22986 or 22987 wherein the agent is simvastatin. '
24929. The method of any one of items 24813-24928 wherein the inflammatory arthritis is osteoarthritis. 24930. The method of any one of items 24813-24928 wherein the inflammatory arthritis is primary osteoarthritis.
24931. The method of any one of items 24813-24928 wherein the inflammatory arthritis is secondary osteoarthritis.
24932. The method of any one of items 24813-24928 wherein the inflammatory arthritis is rheumatoid arthritis.
24933. The method of any one of items 24813-24928 wherein the composition is delivered intravenously.
24934. The method of any one of items 24813-24928 wherein the composition is delivered orally.
24935. The method of any one of items 24813-24928 wherein the composition is delivered by subcutaneous injection.
24936. The method of any one of items 24813-24928 wherein the composition is delivered by intramuscular injection.
24937. The method of any one of items 24813-24928 wherein the composition is delivered intra-articularly.
24938. The method of any one of items 24813-24928 wherein the composition comprises an anti-thrombotic agent.
24939. The method of any one of items 24813-24928 wherein the composition comprises a naturally occurring polymer. 24940. The method of any one of items 24813-24928 wherein the composition comprises protein.
24941. The method of any one of items 24813-24928 wherein the composition comprises carbohydrate.
24942. The method of any one of items 24813-24928 wherein the composition comprises biodegradable polymer.
24943. The method of any one of items 24813-24928 wherein the composition comprises nonbiodegradable polymer.
24944. The method of any one of items 24813-24928 wherein the composition comprises collagen.
- 24945. The method of any one of items 24813-24928 wherein the composition comprises methylated collagen.
24946. The method of any one of items 24813-24928 wherein the composition comprises fibrinogen.
24947. The method of any one of items 24813-24928 wherein the composition comprises thrombin.
24948. The method of any one of items 24813-24928 wherein the composition comprises blood plasma.
24949. The method of any one of items 24813-24928 wherein the composition comprises calcium salt. 24950. The method of any one of items 24813-24928 wherein the composition comprises an antifibronolytic agent.
24951. The method of any one of items 24813-24928 wherein the composition comprises fibrinogen analog.
24952. The method of any one of items 24813-24928 wherein the composition comprises albumin.
24953. The method of any one of items 24813-24928 wherein the composition comprises plasminogen.
24954. The method of any one of items 24813-24928 wherein the composition comprises von Willebrands factor.
24955. The method of any one of items 24813-24928 wherein the composition comprises Factor VIII.
24956. The method of any one of items 24813-24928 wherein the composition comprises hypoallergenic collagen.
24957. The method of any one of items 24813-24928 wherein the composition comprises atelopeptidic collagen.
24958. The method of any one of items 24813-24928 wherein the composition comprises telopeptide collagen.
24959. The method of any one of items 24813-24928 wherein the composition comprises crosslinked collagen. 24960. The method of any one of items 24813-24928 wherein the composition comprises aprotinin.
24961. The method of any one of items 24813-24928 wherein the composition comprises epsilon-amino-n-caproic acid.
24962. The method of any one of items 24813-24928 wherein the composition comprises gelatin.
24963. The method of any one of items 24813-24928 wherein the composition comprises protein conjugates.
24964. The method of any one of items 24813-24928 wherein the composition comprises gelatin conjugates.
24965. The method of any one of items 24813-24928 wherein the composition comprises a synthetic polymer.
24966. The method of any one of items 24813-24928 wherein the composition comprises a synthetic isocyanate-containing compound.
24967. The method of any one of items 24813-24928 wherein the composition comprises a synthetic thiol-containing compound.
24968. The method of any one of items 24813-24928 wherein the composition comprises a synthetic compound containing at least two thiol groups. 24969. The method of any one of items 24813-24928 wherein the composition comprises a synthetic compound containing at least three thiol groups.
24970. The method of any one of items 24813-24928 wherein the composition comprises a synthetic compound containing at least four thiol groups.
24971. The method of any one of items 24813-24928 wherein the composition comprises a synthetic amino-containing compound.
24972. The method of any one of items 24813-24928 wherein the composition comprises a synthetic compound containing at least two amino groups.
24973. The method of any one of items 24813-24928 wherein the composition comprises a synthetic compound containing at least three amino groups.
24974. The method of any one of items 24813-24928 wherein the composition comprises a synthetic compound containing at least four amino groups.
24975. The method of any one of items 24813-24928 wherein the composition comprises a synthetic compound comprising a carbonyl-oxygen-succinimidyl group.
24976. The method of any one of items 24813-24928 wherein the composition comprises a synthetic compound comprising at least two carbonyl-oxygen-succinimidyl groups. 24977. The method of any one of items 24813-24928 wherein the composition comprises a synthetic compound comprising at least three carbonyl-oxygen-succinimidyl groups.
24978. The method of any one of items 24813-24928 wherein the composition comprises a synthetic compound comprising at least four carbonyl-oxygen-succinimidyl groups.
24979. The method of any one of items 24813-24928 wherein the composition comprises a synthetic polyalkylene oxide- containing compound.
24980. The method of any one of items 24813-24928 wherein the composition comprises a synthetic compound comprising both polyalkylene oxide and biodegradable polyester blocks.
24981. The method of any one of items 24813-24928 wherein the composition comprises a synthetic polyalkylene oxide- containing compound having reactive amino groups.
24982. The method of any one of items 24813-24928 wherein the composition comprises a synthetic polyalkylene oxide- containing compound having reactive thiol groups.
24983. The method of any one of items 24813-24928 wherein the composition comprises a synthetic polyalkylene oxide- containing compound having reactive carbonyl-oxygen-succinimidyl groups.
24984. The method of any one of items 24813-24928 wherein the composition comprises a synthetic compound comprising a biodegradable polyester block. 24985. The method of any one of items 24813-24928 wherein the composition comprises a synthetic polymer formed in whole or part from lactic acid or lactide.
24986. The method of any one of items 24813-24928 wherein the composition comprises a synthetic polymer formed in whole or part from glycolic acid or glycolide.
24987. The method of any one of items 24813-24928 wherein the composition comprises polylysine.
24988. The method of any one of items 24813-24928 wherein the composition comprises (a) protein and (b) a compound comprising a polyalkylene oxide portion.
24989. The method of any one of items 24813-24928 _ wherein the composition comprises (a) protein and (b) polylysine.
24990. The method of any one of items 24813-24928 wherein the composition comprises (a) protein and (b) a compound having at least four thiol groups.
24991. The method of any one of items 24813-24928 wherein the composition comprises (a) protein and (b) a compound having at least four amino groups.
24992. The method of any one of items 24813-24928 wherein the composition comprises (a) protein and (b) a compound having at least four carbonyl-oxygen-succinimide groups. 24993. The method of any one of items 24813-24928 wherein the composition comprises (a) protein and (b) a compound having a biodegradable region formed from reactants selected from lactic acid, lactide, glycolic acid, glycolide, and epison-caprolactone.
24994. The method of any one of items 24813-24928 wherein the composition comprises (a) collagen and (b) a compound comprising a polyalkylene oxide portion.
24995. The method of any one of items 24813-24928 wherein the composition comprises (a) collagen and (b) polylysine.
24996. The method of any one of items 24813-24928 wherein the composition comprises (a) collagen and (b) a compound having at least four thiol groups.
24997. The method of any one of items 24813-24928 wherein the composition comprises (a) collagen and (b) a compound having at least four amino groups.
24998. The method of any one of items 24813-24928 wherein the composition comprises (a) collagen and (b) a compound having at least four carbonyl-oxygen-succinimide groups.
24999. The method of any one of items 24813-24928 wherein the composition comprises (a) collagen and (b) a compound having a biodegradable region formed from reactants selected from lactic acid, lactide, glycolic acid, glycolide, and epison-caprolactone. 25000. The method of any one of items 24813-24928 wherein the composition comprises (a) methylated collagen and (b) a compound comprising a polyalkylene oxide portion.
25001. The method of any one of items 24813-24928 wherein the composition comprises (a) methylated collagen and (b) polylysine.
25002. The method of any one of items 24813-24928 wherein the composition comprises (a) methylated collagen and (b) a compound having at least four thiol groups.
25003. The method of any one of items 24813-24928 wherein the composition comprises (a) methylated collagen and (b) a compound having at least four amino groups.
25004. The method of any one of items 24813-24928 wherein the composition comprises (a) methylated collagen and (b) a compound having at least four carbonyl-oxygen-succinimide groups.
25005. The method of any one of items 24813-24928 wherein the composition comprises (a) methylated collagen and (b) a compound having a biodegradable region formed from reactants selected from lactic acid, lactide, glycolic acid, glycolide, and epison-caprolactone.
25006. The method of any one of items 24813-24928 wherein the composition comprises hyaluronic acid.
25007. The method of any one of items 24813-24928 wherein the composition comprises a hyaluronic acid derivative. 25008. The method of any one of items 24813-24928 wherein the composition comprises pentaerythritol poly(ethylene glycol)ether tetra-sulfhydryl of number average molecular weight between 3,000 and 30,000.
25009. The method of any one of items 24813-24928 wherein the composition comprises pentaerythritol poly(ethylene glycol)ether tetra-amino of number average molecular weight between 3,000 and 30,000.
25010. The method of any one of items 24813-24928 wherein the composition comprises (a) a synthetic compound having a number average molecular weight between 3,000 and 30,000 and comprising a polyalkylene oxide region and multiple nucleophilic groups, and (b) a synthetic compound having a number average molecular weight between 3,000 and 30,000 and comprising a polyalkylene oxide region and multiple electrophilic groups.
25011. The method of any one of items 24813-24928 wherein the composition comprises a colorant.
25012. The method of any one of items 24813-24928 wherein the composition is sterile.
25013. The method of item 23193 or 23194 wherein the agent is a cyclin dependent kinase inhibitor selected from the group consisting of CGP 74514A, bohemine, olomoucine (CAS No. 101622-51-9), indole-3-carbinol (CAS No. 700-06-1), and an analogue or derivative thereof. 25014. The method of item 23193 or 23194 wherein the agent is an endothelial growth factor receptor (EGF-R) kinase inhibitor selected from the group consisting of lavendustin A (CAS No. 125697-92-9), SU 1498 (a VEGF-R inhibitor), and an analogue or derivative thereof.
25015. The method of item 23193 or 23194 wherein the agent is mevastatin (a fibrinogen antagonist).
25016. The method of item 23193 or 23194 wherein the agent is vincamine (a microtubule inhibitor).
25017. The method of item 23193 or 23194 wherein the agent is emodin (CAS No. 518-82-1) (an NF kappa B inhibitor) or Bay 11- 7085 (an NF kappa B inhibitor).
25018. The method of item 23193 or 23194 wherein the agent is SKF86002 (CAS No. 72873-74-6) (a p38 MAP kinase inhibitor).
25019. The method of item 23193 or 23194 wherein the agent is a peroxisome proliferators-activated receptor (PPAR) agonist selected from the group consisting of LBM642, WY-14,643 (CAS No. 50892- 23-4), GW7647, fenofibric acid (CAS No. 42017-89-0), MCC-555 (CAS No. 161600-01-7), GW9662, GW1929, GW501516, L-165,041 (CAS No. 79558- 09-1), and an analogue or derivative thereof.
25020. The method of item 23193 or 23194 wherein the agent is a phosphodiesterase (PDE) inhibitor selected from the group consisting of theobromine (CAS No. 83-67-0), papverine hydrochloride (CAS No. 61-25-6), quercetin dehydrate (CAS No. 6151-25-3), YM 976 (CAS No. 191219-80-4), irsogladine (CAS No. 57381-26-7), and an analogue or derivative thereof. 25021. The method of item 23193 or 23194 wherein the agent is a tumor necrosis factor antagonist.
25022. The method of item 23193 or 23194 wherein the agent is herbimycin A (a tyrosine kinase inhibitor).
25023. The method of item 23193 or 23194 wherein the agent is erucylphosphocholine.
25024. The method of item 23193 or 23194 wherein the agent is alphastatin.
25025. The method of item 23193 or 23194 wherein the agent is etanercept.
25026. The method of item 23193 or 23194 wherein ihe agent is humicade.
25027. The method of item 23193 or 23194 wherein the agent is gefitinib.
25028. The method of item 23193 or 23194 wherein the agent is a histamine receptor antagonist selected from the group consisting of phenothiazines (e.g., promethazine), alkylamines (e.g., chlorpheniramine (CAS No. 7054-11-7), brompheniramine (CAS No. 980-71-2), fexofenadine hydrochloride, promethazine hydrochloride, loratadine, ketotifen fumarate salt, and acrivastine), methylxanthines (e.g., theophylline, theobromine, and caffeine), cimetidine (available under the tradename TAGAMET from SmithKline Beecham Phamaceutical Co., Wilmington, DE), ranitidine (available under the tradename ZANTAC from Warner Lambert Company, Morris Plains, NJ), famotidine (available under the tradename PEPCID from Merck & Co., Whitehouse Station, NJ), nizatidine (available under the tradename AXID from Reliant Pharmaceuticals, Inc., Liberty Corner, NJ), nizatidine, and roxatidine acetate (CAS No. 78628-28-1), H3 receptor antagonists (e.g., thioperamide and thioperamide maleate salt), and antihistamines (e.g., tricyclic dibenozoxepins, ethanolamines, ethylenediamines, piperizines, piperidines, and pthalazinones).
25029. The method of item 23193 or 23194 wherein the agent is an alpha adrenergic receptor antagonist.
25030. The method of item 23193 or 23194 wherein the agent is an anti-psychotic compound.
25031. The method of item 23193 or 23194 wherein the agent is a CaM kinase Il inhibitor.
25032. The method of item 23193 or 23194 wherein the agent is a G protein agonist.
25033. The method of item 23193 or 23194 wherein the agent is an antibiotic selected from the group consisting of apigenin (Cas No. 520-36-5), ampicillin sodium salt (CAS No. 69-52-3), puromycin, and an analogue or derivative thereof.
25034. The method of item 23193 or 23194 wherein the agent is an anti-microbial agent.
25035. The method of item 23193 or 23194 wherein the agent is a DNA topoisomerase inhibitor selected from the group consisting of D-lapachone (CAS No. 4707-32-8)~-arctigenin (CAS No. 7770-78-7), aurintricarboxylic acid, and an analogue or derivative thereof. 25036. The method of item 23193 or 23194 wherein the agent is a thromboxane A2 receptor inhibitor selected from the group consisting of BM-531 (CAS No. 284464-46-6), ozagrel hydrochloride (CAS No. 78712-43-3), and an analogue or derivative thereof.
25037. The method of item 23193 or 23194 wherein the agent is a D2 dopamine receptor antagonist.
25038. The method of item 23193 or 23194 wherein the agent is a Peptidyl-Prolyl Cis/Trans lsomerase Inhibitor.
25039. The method of item 23193 or 23194 wherein the agent is a dopamine antagonist.
25040. The method of item 23193 or 23194 wherein the agent is an anesthetic compound.
25041. The method of item 23193 or 23194 wherein the agent is a clotting factor.
25042. The method of item 23193 or 23194 wherein the agent is a lysyl hydrolase inhibitor.
25043. The method of item 23193 or 23194 wherein the agent is a muscarinic receptor inhibitor.
25044. The method of item 23193 or 23194 wherein the agent is a superoxide anion generator.
25045. The method of item 23193 or 23194 wherein the agent is a steroid. 25046. The method of item 23193 or 23194 wherein the agent is an anti-proliferative agent selected from the group consisting of silibinin (CAS No. 22888-70-6), silymarin (CAS No. 65666-07-1), 1,2- hexanediol, dioctyl phthalate (CAS No. 117-81-7), zirconium (IV) oxide, glycyrrhizic acid, spermidine trihydrochloride, tetrahydrochloride, CGP 74514A, spermine tetrahydrochloride, NG-methyl-L-arginine acetate salt, galardin, and an analogue or derivative thereof.
25047. The method of item 23193 or 23194 wherein the agent is a diuretic.
25048. The method of item 23193 or 23194 wherein the agent is an anti-coagulant.
25049. The method of item 23193 or 23194 wherein the agent is a cyclic GMP agonist.
25050. The method of item 23193 or 23194 wherein the agent is an adenylate cyclase agonist.
25051. The method of item 23193 or 23194 wherein the agent is an antioxidant.
25052. The method of item 23193 or 23194 wherein the agent is a nitric oxide synthase inhibitor.
25053. The method of item 23193 or 23194 wherein the agent is an anti-neoplastic agent selected from tirapazamine (CAS No. 27314-97-2), fludarabine (CAS No. 21679-14-1), cladribine, imatinib mesilate, and an analogue or derivative thereof. 25054. The method of item 23193 or 23194 wherein the agent is a DNA synthesis inhibitor.
25055. The method of item 23193 or 23194 wherein the agent is a DNA alkylating agent selected from dacarbazine (CAS No. 4342- 03-4), temozolomide, procarbazine HCI, and an analogue or derivative thereof.
25056. The method of item 23193 or 23194 wherein the agent is a DNA methylation inhibitor.
25057. The method of item 23193 or 23194 wherein the agent is a NSAID agent.
25058. The method of item 23193 or 23194 wherein the agent is a peptidylglycine alpha-hydroxylating monooxygenase inhibitor.
25059. The method of item 23193 or 23194 wherein the agent is an MEK1/MEK 2 inhibitor.
25060. The method of item 23193 or 23194 wherein the agent is a NO synthase inhibitor.
25061. The method of item 23193 or 23194 wherein the agent is a retinoic acid receptor antagonist selected from isotretinoin (CAS No. 4759-48-2) and an analogue or derivative thereof.
25062. The method of item 23193 or 23194 wherein the agent is an ACE inhibitor. 25063. The method of item 23193 or 23194 wherein the agent is a glycosylation inhibitor.
25064. The method of item 23193 or 23194 wherein the agent is an intracellular calcium influx inhibitor.
25065. The method of item 23193 or 23194 wherein the agent is an anti-emetic agent.
25066. The method of item 23193 or 23194 wherein the agent is an acetylcholinesterase inhibitor.
25067. The method of item 23193 or 23194 wherein the agent is an ALK-5 receptor antagonist.
25068. The method of item 23193 or 23194 wherein the agent is a RAR/RXT antagonist.
25069. The method of item 23193 or 23194 wherein the agent is an elF-2a inhibitor.
25070. The method of item 23193 or 23194 wherein the agent is an S-adenosyl-L-homocysteine hydrolase inhibitor.
25071. The method of item 23193 or 23194 wherein the agent is an estrogen agonist.
25072. The method of item 23193 or 23194 wherein the agent is a serotonin receptor inhibitor. 25073. The method of item 23193 or 23194 wherein the agent is an anti-thrombotic agent.
25074. The method of item 23193 or 23194 wherein the agent is a tryptase inhibitor.
25075. The method of item 23193 or 23194 wherein the agent is a pesticide.
25076. The method of item 23193 or 23194 wherein the agent is a bone mineralization promotor.
25077. The method of item 23193 or 23194 wherein the agent is a bisphosphonate compound selected from risedronate and an analogue or derivative thereof.
25078. The method of item 23193 or 23194 wherein the agent is an anti-inflammatory compound.
25079. The method of item 23193 or 23194 wherein the agent is a DNA methylation promotor.
25080. The method of item 23193 or 23194 wherein the agent is an anti-spasmodic agent.
25081. The method of item 23193 or 23194 wherein the agent is a protein synthesis inhibitor.
25082. The method of item 23193 or 23194 wherein the agent is an D-glucosidase inhibitor. 25083. The method of item 23193 or 23194 wherein the agent is a calcium channel blocker.
25084. The method of item 23193 or 23194 wherein the agent is a pyruvate dehydrogenase activator.
25085. The method of item 23193 or 23194 wherein the agent is a prostaglandin inhibitor.
25086. The method of item 23193 or 23194 wherein the agent is a sodium channel inhibitor.
25087. The method of item 23193 or 23194 wherein the agent is a serine protease inhibitor.
25088. The method of item 23193 or 23194 wherein the agent is an intracellular calcium flux inhibitor.
25089. The method of item 23193 or 23194 wherein the agent is a JAK2 inhibitor.
25090. The method of item 23193 or 23194 wherein the agent is an androgen inhibitor.
25091. The method of item 23193 or 23194 wherein the agent is an aromatase inhibitor.
25092. The method of item 23193 or 23194 wherein the agent is an anti-viral agent. 25093. The method of item 23193 or 23194 wherein the agent is a 5-HT inhibitor.
25094. The method of item 23193 or 23194 wherein the agent is an FXR antagonist.
25095. The method of item 23193 or 23194 wherein the agent is an actin polymerization and stabilization promotor.
25096. The method of item 23193 or 23194 wherein the agent is an AXOR12 agonist.
25097. The method of item 23193 or 23194 wherein the agent is an angiotensin Il receptor agonist.
25098. The method of item 23193 or 23194 wherein the agent is a platelet aggregation inhibitor.
25099. The method of item 23193 or 23194 wherein the agent is a CB1/CB2 receptor agonist.
25100. The method of item 23193 or 23194 wherein the agent is a norepinephrine reuptake inhibitor.
25101. The method of item 23193 or 23194 wherein the agent is a selective serotonin reuptake inhibitor.
25102. The method of item 23193 or 23194 wherein the agent is a reducing agent. 25103. The method of item 23193 or 23194 wherein the agent is a immuno-modulator selected from Bay 11-7085, (-)-arctigenin, idazoxan hydrochloride, and an analogue or derivative thereof.
25104. The method of item 23193 or 23194 wherein the agent is isotretinoin.
25105. The method of item 23193 or 23194 wherein the agent is radicicol.
25106. The method of item 23193 or 23194 wherein the agent is clobetasol propionate.
25107. The method of item 23193 or 23194 wherein the agent is homoharringtonine.
25108. The method of item 23193 or 23194 wherein the agent is trichostatin A.
25109. The method of item 23193 or 23194 wherein the agent is brefeldin A.
25110. The method of item 23193 or 23194 wherein the agent is thapsigargin.
25111. The method of item 23193 or 23194 wherein the agent is dolastatin 15.
25112. The method of item 23193 or 23194 wherein the agent is cerivastatin. 25113. The method of item 23193 or 23194 wherein the agent is jasplakinolide.
25114. The method of item 23193 or 23194 wherein the agent is herbimycin A.
25115. The method of item 23193 or 23194 wherein the agent is pirfenidone.
25116. The method of item 23193 or 23194 wherein the agent is vinorelbine.
25117. The method of item 23193 or 23194 wherein the agent is 17-DMAG.
25118. The method of item 23193 or 23194 wherein the agent is tacrolimus.
25119. The method of item 23193 or 23194 wherein the agent is loteprednol etabonate.
25120. The method of item 23193 or 23194 wherein the agent is juglone.
25121. The method of item 23193 or 23194 wherein the agent is prednisolone.
25122. The method of item 23193 or 23194 wherein the agent is puromycin. 25123. The method of item 23193 or 23194 wherein the agent is 3-BAABE.
25124. The method of item 23193 or 23194 wherein the agent is cladribine.
25125. The method of item 23193 or 23194 wherein the agent is mannose-6-phosphate.
25126. The method of item 23193 or 23194 wherein the agent is 5-azacytidine.
25127. The method of item 23193 or 23194 wherein the agent is Ly333531 (ruboxistaurin).
25128. The method of item 23193 or 23194 wherein the agent is simvastatin.
25129. The method of any one of items 25013-25128 wherein the agent or composition is directly injected into the scar or keloid.
25130. The method of any one of items 25013-25128 wherein the agent or composition is topically applied to the scar or keloid.
25131. The method of any one of items 25013-25128 wherein the composition comprises an anti-thrombotic agent.
25132. The method of any one of items 25013-25128 wherein the composition comprises a naturally occurring polymer. 25133. The method of any one of items 25013-25128 wherein the composition comprises protein.
25134. The method of any one of items 25013-25128 wherein the composition comprises carbohydrate.
25135. The method of any one of items 25013-25128 wherein the composition comprises biodegradable polymer.
25136. The method of any one of items 25013-25128 wherein the composition comprises nonbiodegradable polymer.
25137. The method of any one of items 25013-25128 wherein the composition comprises collagen.
25138. The method of any one of items 25013-25128 wherein the composition comprises methylated collagen.
25139. The method of any one of items 25013-25128 wherein the composition comprises fibrinogen.
25140. The method of any one of items 25013-25128 wherein the composition comprises thrombin.
25141. The method of any one of items 25013-25128 wherein the composition comprises blood plasma.
25142. The method of any one of items 25013-25128 wherein the composition comprises calcium salt. 25143. The method of any one of items 25013-25128 wherein the composition comprises an antifibronolytic agent.
25144. The method of any one of items 25013-25128 wherein the composition comprises fibrinogen analog.
25145. The method of any one of items 25013-25128 wherein the composition comprises albumin.
25146. The method of any one of items 25013-25128 wherein the composition comprises plasminogen.
25147. The method of any one of items 25013-25128 wherein the composition comprises von Willebrands factor.
25148. The method of any one of items 25013-25128 wherein the composition comprises Factor VIII.
25149. The method of any one of items 25013-25128 wherein the composition comprises hypoallergenic collagen.
25150. The method of any one of items 25013-25128 wherein the composition comprises atelopeptidic collagen.
25151. The method of any one of items 25013-25128 wherein the composition comprises telopeptide collagen.
25152. The method of any one of items 25013-25128 wherein the composition comprises crosslinked collagen. 25153. The method of any one of items 25013-25128 wherein the composition comprises aprotinin.
25154. The method of any one of items 25013-25128 wherein the composition comprises epsilon-amino-n-caproic acid.
25155. The method of any one of items 25013-25128 wherein the composition comprises gelatin.
25156. The method of any one of items 25013-25128 wherein the composition comprises protein conjugates.
25157. The method of any one of items 25013-25128 wherein the composition comprises gelatin conjugates.
25158. The method of any one of items 25013-25128 wherein the composition comprises a synthetic polymer.
25159. The method of any one of items 25013-25128 wherein the composition comprises a synthetic isocyanate-containing compound.
25160. The method of any one of items 25013-25128 wherein the composition comprises a synthetic thiol-containing compound.
25161. The method of any one of items 25013-25128 wherein the composition comprises a synthetic compound containing at least two thiol groups. 25162. The method of any one of items 25013-25128 wherein the composition comprises a synthetic compound containing at least three thiol groups.
25163. The method of any one of items 25013-25128 wherein the composition comprises a synthetic compound containing at least four thiol groups.
25164. The method of any one of items 25013-25128 wherein the composition comprises a synthetic amino-containing compound.
25165. The method of any one of items 25013-25128 wherein the composition comprises a synthetic compound containing at least two amino groups.
25166. The method of any one of items 25013-25128 wherein the composition comprises a synthetic compound containing at least three amino groups.
25167. The method of any one of items 25013-25128 wherein the composition comprises a synthetic compound containing at least four amino groups.
25168. The method of any one of items 25013-25128 wherein the composition comprises a synthetic compound comprising a carbonyl-oxygen-succinimidyl group.
25169. The method of any one of items 25013-25128 wherein the composition comprises a synthetic compound comprising at least two carbonyl-oxygen-succinimidyl groups. 25170. The method of any one of items 25013-25128 wherein the composition comprises a synthetic compound comprising at least three carbonyl-oxygen-succinimidyl groups.
25171. The method of any one of items 25013-25128 wherein the composition comprises a synthetic compound comprising at least four carbonyl-oxygen-succinimidyl groups.
25172. The method of any one of items 25013-25128 wherein the composition comprises a synthetic polyalkylene oxide- containing compound.
25173. The method of any one of items 25013-25128 wherein the composition comprises a synthetic compound comprising both polyalkylene oxide and biodegradable polyester blocks.
25174. The method of any one of items 25013-25128 wherein the composition comprises a synthetic polyalkylene oxide- containing compound having reactive amino groups.
25175. The method of any one of items 25013-25128 wherein the composition comprises a synthetic polyalkylene oxide- containing compound having reactive thiol groups.
25176. The method of any one of items 25013-25128 wherein the composition comprises a synthetic polyalkylene oxide- containing compound having reactive carbonyl-oxygen-succinimidyl groups.
25177. The method of any one of items 25013-25128 wherein the composition comprises a synthetic compound comprising a biodegradable polyester block. 25178. The method of any one of items 25013-25128 wherein the composition comprises a synthetic polymer formed in whole or part from lactic acid or lactide.
25179. The method of any one of items 25013-25128 wherein the composition comprises a synthetic polymer formed in whole or part from glycolic acid or glycolide.
25180. The method of any one of items 25013-25128 wherein the composition comprises polylysine.
25181. The method of any one of items 25013-25128 wherein the composition comprises (a) protein and (b) a compound comprising a polyalkylene oxide portion.
25182. The method of any one of items 25013-25128 wherein the composition comprises (a) protein and (b) polylysine.
25183. The method of any one of items 25013-25128 wherein the composition comprises (a) protein and (b) a compound having at least four thiol groups.
25184. The method of any one of items 25013-25128 wherein the composition comprises (a) protein and (b) a compound having at least four amino groups.
25185. The method of any one of items 25013-25128 wherein the composition comprises (a) protein and (b) a compound having at least four carbonyl-oxygen-succinimide groups. 25186. The method of any one of items 25013-25128 wherein the composition comprises (a) protein and (b) a compound having a biodegradable region formed from reactants selected from lactic acid, lactide, glycolic acid, glycolide, and epison-caprolactone.
25187. The method of any one of items 25013-25128 wherein the composition comprises (a) collagen and (b) a compound comprising a polyalkylene oxide portion.
25188. The method of any one of items 25013-25128 wherein the composition comprises (a) collagen and (b) polylysine.
25189. The method of any one of items 25013-25128 wherein the composition comprises (a) collagen and (b) a compound having at least four thiol groups.
25190. The method of any one of items 25013-25128 wherein the composition comprises (a) collagen and (b) a compound having at least four amino groups.
25191. The method of any one of items 25013-25128 wherein the composition comprises (a) collagen and (b) a compound having at least four carbonyl-oxygen-succinimide groups.
25192. The method of any one of items 25013-25128 wherein the composition comprises (a) collagen and (b) a compound having a biodegradable region formed from reactants selected from lactic acid, lactide, glycolic acid, glycolide, and epison-caprolactone. 25193. The method of any one of items 25013-25128 wherein the composition comprises (a) methylated collagen and (b) a compound comprising a polyalkylene oxide portion.
25194. The method of any one of items 25013-25128 wherein the composition comprises (a) methylated collagen and (b) polylysine.
25195. The method of any one of items 25013-25128 wherein the composition comprises (a) methylated collagen and (b) a compound having at least four thiol groups.
25196. The method of any one of items 25013-25128 wherein the composition comprises (a) methylated collagen and (b) a compound having at least four amino groups.
25197. The method of any one of items 25013-25128 wherein the composition comprises (a) methylated collagen and (b) a compound having at least four carbonyl-oxygen-succinimide groups.
25198. The method of any one of items 25013-25128 wherein the composition comprises (a) methylated collagen and (b) a compound having a biodegradable region formed from reactants selected from lactic acid, lactide, glycolic acid, glycolide, and epison-caprolactone.
25199. The method of any one of items 25013-25128 wherein the composition comprises hyaluronic acid.
25200. The method of any one of items 25013-25128 wherein the composition comprises a hyaluronic acid derivative. 25201. The method of any one of items 25013-25128 wherein the composition comprises pentaerythritol poly(ethylene glycol)ether tetra-sulfhydryl of number average molecular weight between 3,000 and 30,000.
25202. The method of any one of items 25013-25128 wherein the composition comprises pentaerythritol poly(ethylene glycol)ether tetra-amino of number average molecular weight between 3,000 and 30,000.
25203. The method of any one of items 25013-25128 wherein the composition comprises (a) a synthetic compound having a number average molecular weight between 3,000 and 30,000 and comprising a polyalkylene oxide region and multiple nucleophilic groups, and (b) a synthetic compound having a number average molecular weight between 3,000 and 30,000 and comprising a polyalkylene oxide region and multiple electrophilic groups.
25204. The method of any one of items 25013-25128 wherein the composition comprises a colorant.
25205. The method of any one of items 25013-25128 wherein the composition is sterile.
25206. The method of item 23389 or 23390 wherein the agent is a cyclin dependent kinase inhibitor selected from the group consisting of CGP 74514A, bohemine, olomoucine (CAS No. 101622-51-9), indole-3-carbinol (CAS No. 700-06-1), and an analogue or derivative thereof. 25207. The method of item 23389 or 23390 wherein the agent is an endothelial growth factor receptor (EGF-R) kinase inhibitor selected from the group consisting of lavendustin A (CAS No. 125697-92-9), SU 1498 (a VEGF-R inhibitor), and an analogue or derivative thereof.
25208. The method of item 23389 or 23390 wherein the agent is mevastatin (a fibrinogen antagonist).
25209. The method of item 23389 or 23390 wherein the agent is vincamine (a microtubule inhibitor).
25210. The method of item 23389 or 23390 wherein the agent is emodin (CAS No. 518-82-1) (an NF kappa B inhibitor) or Bay 11- 7085 (an NF kappa B inhibitor).
25211. The method of item 23389 or 23390 wherein the agent is SKF86002 (CAS No. 72873-74-6) (a p38 MAP kinase inhibitor).
25212. The method of item 23389 or 23390 wherein the agent is a peroxisome proliferators-activated receptor (PPAR) agonist selected from the group consisting of LBM642, WY-14,643 (CAS No. 50892- 23-4), GW7647, fenofibric acid (CAS No. 42017-89-0), MCC-555 (CAS No. 161600-01-7), GW9662, GW1929, GW501516, L-165,041 (CAS No. 79558- 09-1), and an analogue or derivative thereof.
25213. The method of item 23389 or 23390 wherein the agent is a phosphodiesterase (PDE) inhibitor selected from the group consisting of theobromine (CAS No. 83-67-0), papverine hydrochloride (CAS No. 61-25-6), quercetin dehydrate (CAS No. 6151-25-3), YM 976 (CAS No. 191219-80-4), irsogladine (CAS No. 57381-26-7), and an analogue or derivative thereof. 25214. The method of item 23389 or 23390 wherein the agent is a tumor necrosis factor antagonist.
25215. The method of item 23389 or 23390 wherein the agent is herbimycin A (a tyrosine kinase inhibitor).
25216. The method of item 23389 or 23390 wherein the agent is erucylphosphocholine.
25217. The method of item 23389 or 23390 wherein the agent is alphastatin.
25218. The method of item 23389 or 23390 wherein the agent is etanercept.
25219. The method of item 23389 or 23390 wherein the agent is humicade.
25220. The method of item 23389 or 23390 wherein the agent is gefitinib.
25221. The method of item 23389 or 23390 wherein the agent is a histamine receptor antagonist selected from the group consisting of phenothiazines (e.g., promethazine), alkylamines (e.g., chlorpheniramine (CAS No. 7054-11-7), brompheniramine (CAS No. 980-71-2), fexofenadine hydrochloride, promethazine hydrochloride, loratadine, ketotifen fumarate salt, and acrivastine), methylxanthines (e.g., theophylline, theobromine, and caffeine), cimetidine (available under the tradename TAGAMET from SmithKline Beecham Phamaceutical Co., Wilmington, DE), ranitidine (available under the tradename ZANTAC from Warner Lambert Company, Morris Plains, NJ), famotidine (available under the tradename PEPCID from Merck & Co., Whitehouse Station, NJ), nizatidine (available under the tradename AXID from Reliant Pharmaceuticals, Inc., Liberty Corner, NJ), nizatidine, and roxatidine acetate (CAS No. 78628-28-1), H3 receptor antagonists (e.g., thioperamide and thioperamide maleate salt), and antihistamines (e.g., tricyclic dibenozoxepins, ethanolamines, ethylenediamines, piperizines, piperidines, and pthalazinones).
25222. The method of item 23389 or 23390 wherein the agent is an alpha adrenergic receptor antagonist.
25223. The method of item 23389 or 23390 wherein the agent is an anti-psychotic compound.
25224. The method of item 23389 or 23390 wherein the agent is a CaM kinase Il inhibitor.
25225. The method of item 23389 or 23390 wherein the agent is a G protein agonist.
25226. The method of item 23389 or 23390 wherein the agent is an antibiotic selected from the group consisting of apigenin (Cas No. 520-36-5), ampicillin sodium salt (CAS No. 69-52-3), puromycin, and an analogue or derivative thereof.
25227. The method of item 23389 or 23390 wherein the agent is an anti-microbial agent.
25228. The method of item 23389 or 23390 wherein the agent is a DNA topoisomerase inhibitor selected from the group consisting of D-lapachone (CAS No. 4707-32-8)~-arctigenin (CAS No. 7770-78-7), aurintricarboxylic acid, and an analogue or derivative thereof. 25229. The method of item 23389 or 23390 wherein the agent is a thromboxane A2 receptor inhibitor selected from the group consisting of BM-531 (CAS No. 284464-46-6), ozagrel hydrochloride (CAS No. 78712-43-3), and an analogue or derivative thereof.
25230. The method of item 23389 or 23390 wherein the agent is a D2 dopamine receptor antagonist.
25231. The method of item 23389 or 23390 wherein the agent is a Peptidyl-Prolyl Cis/Trans lsomerase Inhibitor.
25232. The method of item 23389 or 23390 wherein the agent is a dopamine antagonist.
25233. The method of item 23389 or 23390 wherein the agent is an anesthetic compound.
25234. The method of item 23389 or 23390 wherein the agent is a clotting factor.
25235. The method of item 23389 or 23390 wherein the agent is a lysyl hydrolase inhibitor.
25236. The method of item 23389 or 23390 wherein the agent is a muscarinic receptor inhibitor.
25237. The method of item 23389 or 23390 wherein the agent is a superoxide anion generator.
25238. The method of item 23389 or 23390 wherein the agent is a steroid. 25239. The method of item 23389 or 23390 wherein the agent is an anti-proliferative agent selected from the group consisting of silibinin (CAS No. 22888-70-6), silymarin (CAS No. 65666-07-1), 1,2- hexanediol, dioctyl phthalate (CAS No. 117-81-7), zirconium (IV) oxide, glycyrrhizic acid, spermidine trihydrochloride, tetrahydrochloride, CGP 74514A, spermine tetrahydrochloride, NG-methyl-L-arginine acetate salt, galardin, and an analogue or derivative thereof.
25240. The method of item 23389 or 23390 wherein the agent is a diuretic.
25241. The method of item 23389 or 23390 wherein the agent is an anti-coagulant.
25242. The method of item 23389 or 23390 wherein the agent is a cyclic GMP agonist.
25243. The method of item 23389 or 23390 wherein the agent is an adenylate cyclase agonist.
25244. The method of item 23389 or 23390 wherein the agent is an antioxidant.
25245. The method of item 23389 or 23390 wherein the agent is a nitric oxide synthase inhibitor.
25246. The method of item 23389 or 23390 wherein the agent is an anti-neoplastic agent selected from tirapazamine (CAS No. 27314-97-2), fludarabine (CAS No. 21679-14-1), cladribine, imatinib mesilate, and an analogue or derivative thereof. 25247. The method of item 23389 or 23390 wherein the agent is a DNA synthesis inhibitor.
25248. The method of item 23389 or 23390 wherein the agent is a DNA alkylating agent selected from dacarbazine (CAS No. 4342- 03-4), temozolomide, procarbazine HCI, and an analogue or derivative thereof.
25249. The method of item 23389 or 23390 wherein the agent is a DNA methylation inhibitor.
25250. The method of item 23389 or 23390 wherein the agent is a NSAID agent.
25251. The method of item 23389 or 23390 wherein the agent is a peptidylglycine alpha-hydroxylating monooxygenase inhibitor.
25252. The method of item 23389 or 23390 wherein the agent is an MEK1/MEK 2 inhibitor.
25253. The method of item 23389 or 23390 wherein the agent is a NO synthase inhibitor.
25254. The method of item 23389 or 23390 wherein the agent is a retinoic acid receptor antagonist selected from isotretinoin (CAS No. 4759-48-2) and an analogue or derivative thereof.
25255. The method of item 23389 or 23390 wherein the agent is an ACE inhibitor. 25256. The method of item 23389 or 23390 wherein the agent is a glycosylation inhibitor.
25257. The method of item 23389 or 23390 wherein the agent is an intracellular calcium influx inhibitor.
25258. The method of item 23389 or 23390 wherein the agent is an anti-emetic agent.
25259. The method of item 23389 or 23390 wherein the agent is an acetylcholinesterase inhibitor.
25260. The method of item 23389 or 23390 wherein the agent is an ALK-5 receptor antagonist.
25261. The method of item 23389 or 23390 wherein the agent is a RAR/RXT antagonist.
25262. The method of item 23389 or 23390 wherein the agent is an elF-2a inhibitor.
25263. The method of item 23389 or 23390 wherein the agent is an S-adenosyl-L-homocysteine hydrolase inhibitor.
25264. The method of item 23389 or 23390 wherein the agent is an estrogen agonist.
25265. The method of item 23389 or 23390 wherein the agent is a serotonin receptor inhibitor. 25266. The method of item 23389 or 23390 wherein the agent is an anti-thrombotic agent.
25267. The method of item 23389 or 23390 wherein the agent is a tryptase inhibitor.
25268. The method of item 23389 or 23390 wherein the agent is a pesticide.
25269. The method of item 23389 or 23390 wherein the agent is a bone mineralization promotor.
25270. The method of item 23389 or 23390 wherein the agent is a bisphosphonate compound selected from risedronate and an analogue or derivative thereof.
25271. The method of item 23389 or 23390 wherein the agent is an anti-inflammatory compound.
25272. The method of item 23389 or 23390 wherein the agent is a DNA methylation promotor.
25273. The method of item 23389 or 23390 wherein the agent is an anti-spasmodic agent.
25274. The method of item 23389 or 23390 wherein the agent is a protein synthesis inhibitor.
25275. . The method of item 23389 or 23390 wherein the agent is an D-glucosidase inhibitor. 25276. The method of item 23389 or 23390 wherein the agent is a calcium channel blocker.
25277. The method of item 23389 or 23390 wherein the agent is a pyruvate dehydrogenase activator.
25278. The method of item 23389 or 23390 wherein the agent is a prostaglandin inhibitor.
25279. The method of item 23389 or 23390 wherein the agent is a sodium channel inhibitor.
25280. The method of item 23389 or 23390 wherein the agent is a serine protease inhibitor.
25281. The method of item 23389 or 23390 wherein the agent is an intracellular calcium flux inhibitor.
25282. The method of item 23389 or 23390 wherein the agent is a JAK2 inhibitor.
25283. The method of item 23389 or 23390 wherein the agent is an androgen inhibitor.
25284. The method of item 23389 or 23390 wherein the agent is an aromatase inhibitor.
25285. The method of item 23389 or 23390 wherein the agent is an anti-viral agent. 25286. The method of item 23389 or 23390 wherein the agent is a 5-HT inhibitor.
25287. The method of item 23389 or 23390 wherein the agent is an FXR antagonist.
25288. The method of item 23389 or 23390 wherein the agent is an actin polymerization and stabilization promotor.
25289. The method of item 23389 or 23390 wherein the agent is an AXOR12 agonist.
25290. The method of item 23389 or 23390 wherein the agent is an angiotensin Il receptor agonist.
25291. The method of item 23389 or 23390 wherein the agent is a platelet aggregation inhibitor.
25292. The method of item 23389 or 23390 wherein the agent is a CB1/CB2 receptor agonist.
25293. The method of item 23389 or 23390 wherein the agent is a norepinephrine reuptake inhibitor.
25294. The method of item 23389 or 23390 wherein the agent is a selective serotonin reuptake inhibitor.
25295. The method of item 23389 or 23390 wherein the agent is a reducing agent. 25296. The method of item 23389 or 23390 wherein the agent is a immuno-modulator selected from Bay 11-7085, (-)-arctigenin, idazoxan hydrochloride, and an analogue or derivative thereof.
25297. The method of item 23389 or 23390 wherein the agent is isotretinoin.
25298. The method of item 23389 or 23390 wherein the agent is radicicol.
25299. The method of item 23389 or 23390 wherein the agent is clobetasol propionate.
25300. The method of item 23389 or 23390 wherein the agent is homoharringtonine.
25301. The method of item 23389 or 23390 wherein the agent is trichostatin A.
25302. The method of item 23389 or 23390 wherein the agent is brefeldin A.
25303. The method of item 23389 or 23390 wherein the agent is thapsigargin.
25304. The method of item 23389 or 23390 wherein the agent is dolastatin 15.
25305. The method of item 23389 or 23390 wherein the agent is cerivastatin. 25306. The method of item 23389 or 23390 wherein the agent is jasplakinolide.
25307. The method of item 23389 or 23390 wherein the agent is herbimycin A.
25308. The method of item 23389 or 23390 wherein the agent is pirfenidone.
25309. The method of item 23389 or 23390 wherein the agent is vinorelbine.
25310. The method of item 23389 or 23390 wherein the agent is 17-DMAG.
25311. The method of item 23389 or 23390 wherein the agent is tacrolimus.
25312. The method of item 23389 or 23390 wherein the agent is loteprednol etabonate.
25313. The method of item 23389 or 23390 wherein the agent is juglone.
25314. The method of item 23389 or 23390 wherein the agent is prednisolone.
25315. The method of item 23389 or 23390 wherein the agent is puromycin. 25316. The method of item 23389 or 23390 wherein the agent is 3-BAABE.
25317. The method of item 23389 or 23390 wherein the agent is cladribine.
25318. The method of item 23389 or 23390 wherein the agent is mannose-6-phosphate.
25319. The method of item 23389 or 23390 wherein the agent is 5-azacytidine.
25320. The method of item 23389 or 23390 wherein the agent is Ly333531 (ruboxistaurin).
25321. The method, of item 23389 or 23390 wherein the agent is simvastatin.
25322. The method of any one of items 25206-25321 wherein the agent or composition is delivered intra-articularly.
25323. The method of any one of items 25206-25321 wherein the composition comprises an antithrombotic agent.
25324. The method of any one of items 25206-25321 wherein the composition comprises a naturally occurring polymer.
25325. The method of any one of items 25206-25321 wherein the composition comprises protein. 25326. The method of any one of items 25206-25321 wherein the composition comprises carbohydrate.
25327. The method of any one of items 25206-25321 wherein the composition comprises biodegradable polymer.
25328. The method of any one of items 25206-25321 wherein the composition comprises nonbiodegradable polymer.
25329. The method of any one of items 25206-25321 wherein the composition comprises collagen.
25330. The method of any one of items 25206-25321 wherein the composition comprises methylated collagen.
25331. The method of any one of items 25206-25321 wherein the composition comprises fibrinogen.
25332. The method of any one of items 25206-25321 wherein the composition comprises thrombin.
25333. The method of any one of items 25206-25321 wherein the composition comprises blood plasma.
25334. The method of any one of items 25206-25321 wherein the composition comprises calcium salt.
25335. The method of any one of items 25206-25321 wherein the composition comprises an antifibronolytic agent. 25336. The method of any one of items 25206-25321 wherein the composition comprises fibrinogen analog.
25337. The method of any one of items 25206-25321 wherein the composition comprises albumin.
25338. The method of any one of items 25206-25321 wherein the composition comprises plasminogen.
25339. The method of any one of items 25206-25321 wherein the composition comprises von Willebrands factor.
25340. The method of any one of items 25206-25321 wherein the composition comprises Factor VIII.
25341. The method of any one of items 25206-25321 wherein the composition comprises hypoallergenic collagen.
25342. The method of any one of items 25206-25321 wherein the composition comprises atelopeptidic collagen.
25343. The method of any one of items 25206-25321 wherein the composition comprises telopeptide collagen.
25344. The method of any one of items 25206-25321 wherein the composition comprises crosslinked collagen.
25345. The method of any one of items 25206-25321 wherein the composition comprises aprotinin. 25346. The method of any one of items 25206-25321 wherein the composition comprises epsilon-amino-n-caproic acid.
25347. The method of any one of items 25206-25321 wherein the composition comprises gelatin.
25348. The method of any one of items 25206-25321 wherein the composition comprises protein conjugates.
25349. The method of any one of items 25206-25321 wherein the composition comprises gelatin conjugates.
25350. The method of any one of items 25206-25321 wherein the composition comprises a synthetic polymer.
25351. The method of any one of items 25206-25321 wherein the composition comprises a synthetic isocyanate-containing compound.
25352. The method of any one of items 25206-25321 wherein the composition comprises a synthetic thiol-containing compound.
25353. The method of any one of items 25206-25321 wherein the composition comprises a synthetic compound containing at least two thiol groups.
25354. The method of any one of items 25206-25321 wherein the composition comprises a synthetic compound containing at least three thiol groups. 25355. The method of any one of items 25206-25321 wherein the composition comprises a synthetic compound containing at least four thiol groups.
25356. The method of any one of items 25206-25321 wherein the composition comprises a synthetic amino-containing compound.
25357. The method of any one of items 25206-25321 wherein the composition comprises a synthetic compound containing at least two amino groups.
25358. The method of any one of items 25206-25321 wherein the composition comprises a synthetic compound containing at least three amino groups.
25359. _ The method of any one of items 25206-25321 wherein the composition comprises a synthetic compound containing at least four amino groups.
25360. The method of any one of items 25206-25321 wherein the composition comprises a synthetic compound comprising a carbonyl-oxygen-succinimidyl group.
25361. The method of any one of items 25206-25321 wherein the composition comprises a synthetic compound comprising at least two carbonyl-oxygen-succinimidyl groups.
25362. The method of any one of items 25206-25321 wherein the composition comprises a synthetic compound comprising at least three carbonyl-oxygen-succinimidyl groups. 25363. The method of any one of items 25206-25321 wherein the composition comprises a synthetic compound comprising at least four carbonyl-oxygen-succinimidyl groups.
25364. The method of any one of items 25206-25321 wherein the composition comprises a synthetic polyalkylene oxide- containing compound.
25365. The method of any one of items 25206-25321 wherein the composition comprises a synthetic compound comprising both polyalkylene oxide and biodegradable polyester blocks.
25366. The method of any one of items 25206-25321 wherein the composition comprises a synthetic polyalkylene oxide- containing compound having reactive amino groups.
25367. The method of any one of items 25206-25321 wherein the composition comprises a synthetic polyalkylene oxide- containing compound having reactive thiol groups.
25368. The method of any one of items 25206-25321 wherein the composition comprises a synthetic polyalkylene oxide- containing compound having reactive carbonyl-oxygen-succinimidyl groups.
25369. The method of any one of items 25206-25321 wherein the composition comprises a synthetic compound comprising a biodegradable polyester block.
25370. The method of any one of items 25206-25321 wherein the composition comprises a synthetic polymer formed in whole or part from lactic acid or lactide. 25371. The method of any one of items 25206-25321 wherein the composition comprises a synthetic polymer formed in whole or part from glycolic acid or glycolide.
25372. The method of any one of items 25206-25321 wherein the composition comprises polylysine.
25373. The method of any one of items 25206-25321 wherein the composition comprises (a) protein and (b) a compound comprising a polyalkylene oxide portion.
25374. The method of any one of items 25206-25321 wherein the composition comprises (a) protein and (b) polylysine.
25375. The method of any one of items 25206-25321 wherein the composition comprises (a) protein and (b) a compound having at least four thiol groups.
25376. The method of any one of items 25206-25321 wherein the composition comprises (a) protein and (b) a compound having at least four amino groups.
25377. The method of any one of items 25206-25321 wherein the composition comprises (a) protein and (b) a compound having at least four carbonyl-oxygen-succinimide groups.
25378. The method of any one of items 25206-25321 wherein the composition comprises (a) protein and (b) a compound having a biodegradable region formed from reactants selected from lactic acid, lactide, glycolic acid, glycolide, and epison-caprolactone. 25379. The method of any one of items 25206-25321 wherein the composition comprises (a) collagen and (b) a compound comprising a polyalkylene oxide portion.
25380. The method of any one of items 25206-25321 wherein the composition comprises (a) collagen and (b) polylysine.
25381. The method of any one of items 25206-25321 wherein the composition comprises (a) collagen and (b) a compound having at least four thiol groups.
25382. The method of any one of items 25206-25321 wherein the composition comprises (a) collagen and (b) a compound having at least four amino groups.
25383. The method of any one of items 25206-25321 wherein the composition comprises (a) collagen and (b) a compound having at least four carbonyl-oxygen-succinimide groups.
25384. The method of any one of items 25206-25321 wherein the composition comprises (a) collagen and (b) a compound having a biodegradable region formed from reactants selected from lactic acid, lactide, glycolic acid, glycolide, and epison-caprolactone.
25385. The method of any one of items 25206-25321 wherein the composition comprises (a) methylated collagen and (b) a compound comprising a polyalkylene oxide portion.
25386. The method of any one of items 25206-25321 wherein the composition comprises (a) methylated collagen and (b) polylysine. 25387. The method of any one of items 25206-25321 wherein the composition comprises (a) methylated collagen and (b) a compound having at least four thiol groups.
25388. The method of any one of items 25206-25321 wherein the composition comprises (a) methylated collagen and (b) a compound having at least four amino groups.
25389. The method of any one of items 25206-25321 wherein the composition comprises (a) methylated collagen and (b) a compound having at least four carbonyl-oxygen-succinimide groups.
25390. The method of any one of items 25206-25321 wherein the composition comprises (a) methylated collagen and (b) a compound having a biodegradable region formed from reactants selected from lactic acid, lactide, glycolic acid, glycolide, and epison-caprolactone.
25391. The method of any one of items 25206-25321 wherein the composition comprises hyaluronic acid.
25392. The method of any one of items 25206-25321 wherein the composition comprises a hyaluronic acid derivative.
25393. The method of any one of items 25206-25321 wherein the composition comprises pentaerythritol poly(ethylene glycol)ether tetra-sulfhydryl of number average molecular weight between 3,000 and 30,000.
25394. The method of any one of items 25206-25321 wherein the composition comprises pentaerythritol poly(ethylene glycol)ether tetra-amino of number average molecular weight between 3,000 and 30,000.
25395. The method of any one of items 25206-25321 wherein the composition comprises (a) a synthetic compound having a number average molecular weight between 3,000 and 30,000 and comprising a polyalkylene oxide region and multiple nucleophilic groups, and (b) a synthetic compound having a number average molecular weight between 3,000 and 30,000 and comprising a polyalkylene oxide region and multiple electrophilic groups.
25396. The method of any one of items 25206-25321 wherein the composition comprises a colorant.
25397. The method of any one of items 25206-25321 wherein the composition is sterile.
25398. The method of item 23588 wherein the agent is a cyclin dependent kinase inhibitor selected from the group consisting of CGP 74514A, bohemine, olomoucine (CAS No. 101622-51-9), indole-3-carbinol (CAS No. 700-06-1), and an analogue or derivative thereof.
25399. The method of item 23588 wherein the agent is an endothelial growth factor receptor (EGF-R) kinase inhibitor selected from the group consisting of lavendustin A (CAS No. 125697-92-9), SU 1498 (a VEGF-R inhibitor), and an analogue or derivative thereof.
25400. The method of item 23588 wherein the agent is mevastatin (a fibrinogen antagonist). 25401. The method of item 23588 wherein the agent is vincamine (a microtubule inhibitor).
25402. The method of item 23588 wherein the agent is emodin (CAS No. 518-82-1) (an NF kappa B inhibitor) or Bay 11-7085 (an NF kappa B inhibitor).
25403. The method of item 23588 wherein the agent is SKF86002 (CAS No. 72873-74-6) (a p38 MAP kinase inhibitor).
25404. The method of item 23588 wherein the agent is a peroxisome proliferators-activated receptor (PPAR) agonist selected from the group consisting of LBM642, WY-14,643 (CAS No. 50892-23-4), GW7647, fenofibric acid (CAS No. 42017-89-0), MCC-555 (CAS No. 161600-01-7), GW9662, GW1929, GW501516, L-165,041 (CAS No. 79558- 09-1), and an analogue or derivative thereof .
25405. The method of item 23588 wherein the agent is a phosphodiesterase (PDE) inhibitor selected from the group consisting of theobromine (CAS No. 83-67-0), papverine hydrochloride (CAS No. 61-25- 6), quercetin dehydrate (CAS No. 6151-25-3), YM 976 (CAS No. 191219- 80-4), irsogladine (CAS No. 57381-26-7), and an analogue or derivative thereof.
25406. The method of item 23588 wherein the agent is a tumor necrosis factor antagonist.
25407. The method of item 23588 wherein the agent is herbimycin A (a tyrosine kinase inhibitor). 25408. The method of item 23588 wherein the agent is erucylphosphocholine.
25409. The method of item 23588 wherein the agent is alphastatin.
25410. The method of item 23588 wherein the agent is etanercept.
25411. The method of item 23588 wherein the agent is humicade.
25412. The method of item 23588 wherein the agent is gefitinib.
25413. The method of item 23588 wherein the agent is a histamine receptor antagonist selected from the group consisting of phenothiazines (e.g., promethazine), alkylamines (e.g., chlorpheniramine (CAS No. 7054-11-7), brompheniramine (CAS No. 980-71-2), fexofenadine hydrochloride, promethazine hydrochloride, loratadine, ketotifen fumarate salt, and acrivastine), methylxanthines (e.g., theophylline, theobromine, and caffeine), cimetidine (available under the tradename TAGAMET from SmithKline Beecham Phamaceutical Co., Wilmington, DE), ranitidine (available under the tradename ZANTAC from Warner Lambert Company, Morris Plains, NJ), famotidine (available under the tradename PEPCID from Merck & Co., Whitehouse Station, NJ), nizatidine (available under the tradename AXID from Reliant Pharmaceuticals, Inc., Liberty Corner, NJ), nizatidine, and roxatidine acetate (CAS No. 78628-28-1), H3 receptor antagonists (e.g., thioperamide and thioperamide maleate salt), and antihistamines (e.g., tricyclic dibenozoxepins, ethanolamines, ethylenediamines, piperizines, piperidines, and pthalazinones). 25414. The method of item 23588 wherein the agent is an alpha adrenergic receptor antagonist.
25415. The method of item 23588 wherein the agent is an anti-psychotic compound.
25416. The method of item 23588 wherein the agent is a CaM kinase Il inhibitor.
25417. The method of item 23588 wherein the agent is a G protein agonist.
25418. The method of item 23588 wherein the agent is an antibiotic selected from the group consisting of apigenin (Cas No. 520- 36-5), ampicillin sodium salt (CAS No. 69-52-3), puromycin, and an analogue or derivative thereof. .
25419. The method of item 23588 wherein the agent is an anti-microbial agent.
25420. The method of item 23588 wherein the agent is a DNA topoisomerase inhibitor selected from the group consisting of D- lapachone (CAS No. 4707-32-8)" -arctigenin (CAS No. 7770-78-7), aurintricarboxylic acid, and an analogue or derivative thereof.
25421. The method of item 23588 wherein the agent is a thromboxane A2 receptor inhibitor selected from the group consisting of BM- 531 (CAS No. 284464-46-6), ozagrel hydrochloride (CAS No. 78712-43-3), and an analogue or derivative thereof. 25422. The method of item 23588 wherein the agent is a D2 dopamine receptor antagonist.
25423. The method of item 23588 wherein the agent is a Peptidyl-Prolyl Cis/Trans lsomerase Inhibitor.
25424. The method of item 23588 wherein the agent is a dopamine antagonist.
25425. The method of item 23588 wherein the agent is an anesthetic compound.
25426. The method of item 23588 wherein the agent is a clotting factor.
25427. The method of item 23588 wherein the agent is a lysyl hydrolase inhibitor.
25428. The method of item 23588 wherein the agent is a muscarinic receptor inhibitor.
25429. The method of item 23588 wherein the agent is a superoxide anion generator.
25430. The method of item 23588 wherein the agent is a steroid.
25431. The method of item 23588 wherein the agent is an anti-proliferative agent selected from the group consisting of silibinin (CAS No. 22888-70-6), silymarin (CAS No. 65666-07-1), 1 ,2-hexanediol, dioctyl phthalate (CAS No. 117-81-7), zirconium (IV) oxide, glycyrrhizic acid, spermidine trihydrochloride, tetrahydrochloride, CGP 74514A, spermine tetrahydrochloride, NG-methyl-L-arginine acetate salt, galardin, and an analogue or derivative thereof.
25432. The method of item 23588 wherein the agent is a diuretic.
25433. The method of item 23588 wherein the agent is an anti-coagulant.
25434. The method of item 23588 wherein the agent is a cyclic GMP agonist.
25435. The method of item 23588 wherein the agent is an adenylate cyclase agonist.
25436. The method of item 23588 wherein the agent is an antioxidant.
25437. The method of item 23588 wherein the agent is a nitric oxide synthase inhibitor.
25438. The method of item 23588 wherein the agent is an anti-neoplastic agent selected from tirapazamine (CAS No. 27314-97-2), fludarabine (CAS No. 21679-14-1), cladribine, imatinib mesilate, and an analogue or derivative thereof.
25439. The method of item 23588 wherein the agent is a DNA synthesis inhibitor. 25440. The method of item 23588 wherein the agent is a DNA alkylating agent selected from dacarbazine (CAS No. 4342-03-4), temozolomide, procarbazine HCI, and an analogue or derivative thereof.
25441. The method of item 23588 wherein the agent is a DNA methylation inhibitor.
25442. The method of item 23588 wherein the agent is a NSAID agent.
25443. The method of item 23588 wherein the agent is a peptidylglycine alpha-hydroxylating monooxygenase inhibitor.
25444. The method of item 23588 wherein the agent is an MEK1/MEK 2 inhibitor.
25445. The method of item 23588 wherein the agent is a NO synthase inhibitor.
25446. The method of item 23588 wherein the agent is a retinoic acid receptor antagonist selected from isotretinoin (CAS No. 4759- 48-2) and an analogue or derivative thereof.
25447. The method of item 23588 wherein the agent is an ACE inhibitor.
25448. The method of item 23588 wherein the agent is a glycosylation inhibitor.
25449. The method of item 23588 wherein the agent is an intracellular calcium influx inhibitor. 25450. The method of item 23588 wherein the agent is an anti-emetic agent.
25451. The method of item 23588 wherein the agent is an acetylcholinesterase inhibitor.
25452. The method of item 23588 wherein the agent is an ALK-5 receptor antagonist.
25453. The method of item 23588 wherein the agent is a RAR/RXT antagonist.
25454. The method of item 23588 wherein the agent is an elF-2a inhibitor.
25455. The method of item 23588 wherein the agent is an S-adenosyl-L-homocysteine hydrolase inhibitor.
25456. The method of item 23588 wherein the agent is an estrogen agonist.
25457. The method of item 23588 wherein the agent is a serotonin receptor inhibitor.
25458. The method of item 23588 wherein the agent is an anti-thrombotic agent.
25459. The method of item 23588 wherein the agent is a tryptase inhibitor. 25460. The method of item 23588 wherein the agent is a pesticide.
25461. The method of item 23588 wherein the agent is a bone mineralization promotor.
25462. The method of item 23588 wherein the agent is a bisphosphonate compound selected from risedronate and an analogue or derivative thereof.
25463. The method of item 23588 wherein the agent is an anti-inflammatory compound.
25464. The method of item 23588 wherein the agent is a DNA methylation promotor.
25465. The method of item 23588 wherein the agent is an anti-spasmodic agent.
25466. The method of item 23588 wherein the agent is a protein synthesis inhibitor.
25467. The method of item 23588 wherein the agent is an D-glucosidase inhibitor.
25468. The method of item 23588 wherein the agent is a calcium channel blocker.
25469. The method of item 23588 wherein the agent is a pyruvate dehydrogenase activator. 25470. The method of item 23588 wherein the agent is a prostaglandin inhibitor.
25471. The method of item 23588 wherein the agent is a sodium channel inhibitor.
25472. The method of item 23588 wherein the agent is a serine protease inhibitor.
25473. The method of item 23588 wherein the agent is an intracellular calcium flux inhibitor.
25474. The method of item 23588 wherein the agent is a JAK2 inhibitor.
25475. The method of item 23588 wherein the agent is _ an androgen inhibitor.
25476. The method of item 23588 wherein the agent is an aromatase inhibitor.
25477. The method of item 23588 wherein the agent is an anti-viral agent.
25478. The method of item 23588 wherein the agent is a 5-HT inhibitor.
25479. The method of item 23588 wherein the agent is an FXR antagonist. 25480. The method of item 23588 wherein the agent is an actin polymerization and stabilization promotor.
25481. The method of item 23588 wherein the agent is an AXOR12 agonist.
25482. The method of item 23588 wherein the agent is an angiotensin Il receptor agonist.
25483. The method of item 23588 wherein the agent is a platelet aggregation inhibitor.
25484. The method of item 23588 wherein the agent is a CB1/CB2 receptor agonist.
25485. The method of item 23588 wherein the agent is a norepinephrine reuptake inhibitor.
25486. The method of item 23588 wherein the agent is a selective serotonin reuptake inhibitor.
25487. The method of item 23588 wherein the agent is a reducing agent.
25488. The method of item 23588 wherein the agent is a immuno-modulator selected from Bay 11-7085, (-)-arctigenin, idazoxan hydrochloride, and an analogue or derivative thereof.
25489. The method of item 23588 wherein the agent is isotretinoin. 25490. The method of item 23588 wherein the agent is radicicol.
25491. The method of item 23588 wherein the agent is clobetasol propionate.
25492. The method of item 23588 wherein the agent is homoharringtonine.
25493. The method of item 23588 wherein the agent is trichostatin A.
25494. The method of item 23588 wherein the agent is brefeldin A.
25495. The method of item 23588 wherein the agent is thapsigargin.
25496. The method of item 23588 wherein the agent is dolastatin 15.
25497. The method of item 23588 wherein the agent is cerivastatin.
25498. The method of item 23588 wherein the agent is jasplakinolide.
25499. The method of item 23588 wherein the agent is herbimycin A. 25500. The method of item 23588 wherein the agent is pirfenidone.
25501. The method of item 23588 wherein the agent is vinorelbine.
25502. The method of item 23588 wherein the agent is 17-DMAG.
25503. The method of item 23588 wherein the agent is tacrolimus.
25504. The method of item 23588 wherein the agent is loteprednol etabonate.
25505. The method of item 23588 wherein the agent is juglone.
25506. The method of item 23588 wherein the agent is prednisolone.
25507. The method of item 23588 wherein the agent is puromycin.
25508. The method of item 23588 wherein the agent is 3-BAABE.
25509. The method of item 23588 wherein the agent is cladribine. 25510. The method of item 23588 wherein the agent is mannose-6-phosphate.
25511. The method of item 23588 wherein the agent is 5-azacytidine.
25512. The method of item 23588 wherein the agent is Ly333531 (ruboxistaurin).
25513. The method of item 23588 wherein the agent is simvastatin.
25514. The method of any one of items 25398-25513 wherein the vascular disease is stenosis.
25515. The method of any one of items 25398-25513 wherein the vascular disease is restenosis.
25516. The method of any one of items 25398-25513 wherein the vascular disease is atherosclerosis.
25517. The method of any one of items 25398-25513 wherein the agent or composition is delivered perivascularly.
25518. The method of any one of items 25398-25513 wherein the composition comprises an anti-thrombotic agent.
25519. The method of any one of items 25398-25513 wherein the composition comprises a naturally occurring polymer. 25520. The method of any one of items 25398-25513 wherein the composition comprises protein.
25521. The method of any one of items 25398-25513 wherein the composition comprises carbohydrate.
25522. The method of any one of items 25398-25513 wherein the composition comprises biodegradable polymer.
25523. The method of any one of items 25398-25513 wherein the composition comprises nonbiodegradable polymer.
25524. The method of any one of items 25398-25513 wherein the composition comprises collagen.
25525. The method of any one of items 25398-25513 wherein the composition comprises methylated collagen.
25526. The method of any one of items 25398-25513 wherein the composition comprises fibrinogen.
25527. The method of any one of items 25398-25513 wherein the composition comprises thrombin.
25528. The method of any one of items 25398-25513 wherein the composition comprises blood plasma.
25529. The method of any one of items 25398-25513 wherein the composition comprises calcium salt. 25530. The method of any one of items 25398-25513 wherein the composition comprises an antifibronolytic agent.
25531. The method of any one of items 25398-25513 wherein the composition comprises fibrinogen analog.
25532. The method of any one of items 25398-25513 wherein the composition comprises albumin.
25533. The method of any one of items 25398-25513 wherein the composition comprises plasminogen.
25534. The method of any one of items 25398-25513 wherein the composition comprises von Willebrands factor.
25535. The method of any one of items 25398-25513 wherein the composition comprises Factor VIII.
25536. The method of any one of items 25398-25513 wherein the composition comprises hypoallergenic collagen.
25537. The method of any one of items 25398-25513 wherein the composition comprises atelopeptidic collagen.
25538. The method of any one of items 25398-25513 wherein the composition comprises telopeptide collagen.
25539. The method of any one of items 25398-25513 wherein the composition comprises crosslinked collagen. 25540. The method of any one of items 25398-25513 wherein the composition comprises aprotinin.
25541. The method of any one of items 25398-25513 wherein the composition comprises epsilon-amino-n-caproic acid.
25542. The method of any one of items 25398-25513 wherein the composition comprises gelatin.
25543. The method of any one of items 25398-25513 wherein the composition comprises protein conjugates.
25544. The method of any one of items 25398-25513 wherein the composition comprises gelatin conjugates.
25545. The method of any one of items 25398-25513 wherein the composition comprises a synthetic polymer.
25546. The method of any one of items 25398-25513 wherein the composition comprises a synthetic isocyanate-containing compound.
25547. The method of any one of items 25398-25513 wherein the composition comprises a synthetic thiol-containing compound.
25548. The method of any one of items 25398-25513 wherein the composition comprises a synthetic compound containing at least two thiol groups. 25549. The method of any one of items 25398-25513 wherein the composition comprises a synthetic compound containing at least three thiol groups.
25550. The method of any one of items 25398-25513 wherein the composition comprises a synthetic compound containing at least four thiol groups.
25551. The method of any one of items 25398-25513 wherein the composition comprises a synthetic amino-containing compound.
25552. The method of any one of items 25398-25513 wherein the composition comprises a synthetic compound containing at least two amino groups.
25553. The method of any one of items 25398-25513. wherein the composition comprises a synthetic compound containing at least three amino groups.
25554. The method of any one of items 25398-25513 wherein the composition comprises a synthetic compound containing at least four amino groups.
25555. The method of any one of items 25398-25513 wherein the composition comprises a synthetic compound comprising a carbonyl-oxygen-succinimidyl group.
25556. The method of any one of items 25398-25513 wherein the composition comprises a synthetic compound comprising at least two carbonyl-oxygen-succinimidyl groups. 25557. The method of any one of items 25398-25513 wherein the composition comprises a synthetic compound comprising at least three carbonyl-oxygen-succinimidyl groups.
25558. The method of any one of items 25398-25513 wherein the composition comprises a synthetic compound comprising at least four carbonyl-oxygen-succinimidyl groups.
25559. The method of any one of items 25398-25513 wherein the composition comprises a synthetic polyalkylene oxide- containing compound.
25560. The method of any one of items 25398-25513 wherein the composition comprises a synthetic compound comprising both polyalkylene oxide and biodegradable polyester blocks.
25561. The method of any one of items 25398-25513 wherein the composition comprises a synthetic polyalkylene oxide- containing compound having reactive amino groups.
25562. The method of any one of items 25398-25513 wherein the composition comprises a synthetic polyalkylene oxide- containing compound having reactive thiol groups.
25563. The method of any one of items 25398-25513 wherein the composition comprises a synthetic polyalkylene oxide- containing compound having reactive carbonyl-oxygen-succinimidyl groups.
25564. The method of any one of items 25398-25513 wherein the composition comprises a synthetic compound comprising a biodegradable polyester block. 25565. The method of any one of items 25398-25513 wherein the composition comprises a synthetic polymer formed in whole or part from lactic acid or lactide.
25566. The method of any one of items 25398-25513 wherein the composition comprises a synthetic polymer formed in whole or part from glycolic acid or glycolide.
25567. The method of any one of items 25398-25513 wherein the composition comprises polylysine.
25568. The method of any one of items 25398-25513 wherein the composition comprises (a) protein and (b) a compound comprising a polyalkylene oxide portion.
25569. The method of any one of items 25398-25513 . . wherein the composition comprises (a) protein and (b) polylysine.
25570. The method of any one of items 25398-25513 wherein the composition comprises (a) protein and (b) a compound having at least four thiol groups.
25571. The method of any one of items 25398-25513 wherein the composition comprises (a) protein and (b) a compound having at least four amino groups.
25572. The method of any one of items 25398-25513 wherein the composition comprises (a) protein and (b) a compound having at least four carbonyl-oxygen-succinimide groups. 25573. The method of any one of items 25398-25513 wherein the composition comprises (a) protein and (b) a compound having a biodegradable region formed from reactants selected from lactic acid, lactide, glycolic acid, glycolide, and epison-caprolactone.
25574. The method of any one of items 25398-25513 wherein the composition comprises (a) collagen and (b) a compound comprising a polyalkylene oxide portion.
25575. The method of any one of items 25398-25513 wherein the composition comprises (a) collagen and (b) polylysine.
25576. The method of any one of items 25398-25513 wherein the composition comprises (a) collagen and (b) a compound having at least four thiol groups.
25577. The method of any one of items 25398-25513 wherein the composition comprises (a) collagen and (b) a compound having at least four amino groups.
25578. The method of any one of items 25398-25513 wherein the composition comprises (a) collagen and (b) a compound having at least four carbonyl-oxygen-succinimide groups.
25579. The method of any one of items 25398-25513 wherein the composition comprises (a) collagen and (b) a compound having a biodegradable region formed from reactants selected from lactic acid, lactide, glycolic acid, glycolide, and epison-caprolactone. 25580. The method of any one of items 25398-25513 wherein the composition comprises (a) methylated collagen and (b) a compound comprising a polyalkylene oxide portion.
25581. The method of any one of items 25398-25513 wherein the composition comprises (a) methylated collagen and (b) polylysine.
25582. The method of any one of items 25398-25513 wherein the composition comprises (a) methylated collagen and (b) a compound having at least four thiol groups.
25583. The method of any one of items 25398-25513 wherein the composition comprises (a) methylated collagen and (b) a compound having at least four amino groups.
25584. The method of any one of items 25398-25513 wherein the composition comprises (a) methylated collagen and (b) a compound having at least four carbonyl-oxygen-succinimide groups.
25585. The method of any one of items 25398-25513 wherein the composition comprises (a) methylated collagen and (b) a compound having a biodegradable region formed from reactants selected from lactic acid, lactide, glycolic acid, glycolide, and epison-caprolactone.
25586. The method of any one of items 25398-25513 wherein the composition comprises hyaluronic acid.
25587. The method of any one of items 25398-25513 wherein the composition comprises a hyaluronic acid derivative. 25588. The method of any one of items 25398-25513 wherein the composition comprises pentaerythritol poly(ethylene glycol)ether tetra-sulfhydryl of number average molecular weight between 3,000 and 30,000.
25589. The method of any one of items 25398-25513 wherein the composition comprises pentaerythritol poly(ethylene glycol)ether tetra-amino of number average molecular weight between 3,000 and 30,000.
25590. The method of any one of items 25398-25513 wherein the composition comprises (a) a synthetic compound having a number average molecular weight between 3,000 and 30,000 and comprising a polyalkylene oxide region and multiple nucleophilic groups, and (b) a synthetic compound having a number average molecular weight between 3,000 and 30,000 and comprising a polyalkylene oxide region and multiple electrophilic groups.
25591. The method of any one of items 25398-25513 wherein the composition comprises a colorant.
25592. The method of any one of items 25398-25513 wherein the composition is sterile.
25593. The composition of item 23785 wherein the agent is a cyclin dependent kinase inhibitor selected from the group consisting of CGP 74514A, bohemine, olomoucine (CAS No. 101622-51-9), indole-3-carbinol (CAS No. 700-06-1), and an analogue or derivative thereof. 25594. The composition of item 23785 wherein the agent is an endothelial growth factor receptor (EGF-R) kinase inhibitor selected from the group consisting of lavendustin A (CAS No. 125697-92-9), SU 1498 (a VEGF-R inhibitor), and an analogue or derivative thereof.
25595. The composition of item 23785 wherein the agent is mevastatin (a fibrinogen antagonist).
25596. The composition of item 23785 wherein the agent is vincamine (a microtubule inhibitor).
25597. The composition of item 23785 wherein the agent is emodin (CAS No. 518-82-1) (an NF kappa B inhibitor) or Bay 11- 7085 (an NF kappa B inhibitor).
25598. The composition of item 23785 wherein the agent is SKF86002 (CAS No. 72873-74-6) (a p38 MAP kinase inhibitor).
25599. The composition of item 23785 wherein the agent is a peroxisome proliferators-activated receptor (PPAR) agonist selected from the group consisting of LBM642, WY-14,643 (CAS No. 50892- 23-4), GW7647, fenofibric acid (CAS No. 42017-89-0), MCC-555 (CAS No. 161600-01-7), GW9662, GW1929, GW501516, L-165,041 (CAS No. 79558- 09-1), and an analogue or derivative thereof.
25600. The composition of item 23785 wherein the agent is a phosphodiesterase (PDE) inhibitor selected from the group consisting of theobromine (CAS No. 83-67-0), papverine hydrochloride (CAS No. 61-25-6), quercetin dehydrate (CAS No. 6151-25-3), YM 976 (CAS No. 191219-80-4), irsogladine (CAS No. 57381-26-7), and an analogue or derivative thereof. 25601. The composition of item 23785 wherein the agent is a tumor necrosis factor antagonist.
25602. The composition of item 23785 wherein the agent is herbimycin A (a tyrosine kinase inhibitor).
25603. The composition of item 23785 wherein the agent is erucylphosphocholine.
25604. The composition of item 23785 wherein the agent is alphastatin.
25605. The composition of item 23785 wherein the agent is etanercept.
25606. The composition of item 23785 wherein the agent is humicade.
25607. The composition of item 23785 wherein the agent is gefitinib.
25608. The composition of item 23785 wherein the agent is a histamine receptor antagonist selected from the group consisting of phenothiazines (e.g., promethazine), alkylamines {e.g., chlorpheniramine (CAS No. 7054-11-7), brompheniramine (CAS No. 980-71-2), fexofenadine hydrochloride, promethazine hydrochloride, loratadine, ketotifen fumarate salt, and acrivastine), methylxanthines (e.g., theophylline, theobromine, and caffeine), cimetidine (available under the tradename TAGAMET from SmithKline Beecham Phamaceutical Co., Wilmington, DE), ranitidine (available under the tradename ZANTAC from Warner Lambert Company, Morris Plains, NJ), famotidine (available under the tradename PEPCID from Merck & Co., Whitehouse Station, NJ), nizatidine (available under the tradename AXID from Reliant Pharmaceuticals, Inc., Liberty Corner, NJ)1 nizatidine, and roxatidine acetate (CAS No. 78628-28-1), H3 receptor antagonists (e.g., thioperamide and thioperamide maleate salt), and antihistamines (e.g., tricyclic dibenozoxepins, ethanolamines, ethylenediamines, piperizines, piperidines, and pthalazinones).
25609. The composition of item 23785 wherein the agent is an alpha adrenergic receptor antagonist.
25610. The composition of item 23785 wherein the agent is an anti-psychotic compound.
25611. The composition of item 23785 wherein the agent is a CaM kinase Il inhibitor.
25612. The composition of item 23785 wherein the agent is a G protein agonist.
25613. The composition of item 23785 wherein the agent is an antibiotic selected from the group consisting of apigenin (Cas No. 520-36-5), ampicillin sodium salt (CAS No. 69-52-3), puromycin, and an analogue or derivative thereof.
25614. The composition of item 23785 wherein the agent is an anti-microbial agent.
25615. The composition of item 23785 wherein the agent is a DNA topoisomerase inhibitor selected from the group consisting of D-lapachone (CAS No. 4707-32-8)~-arctigenin (CAS No. 7770-78-7), aurintricarboxylic acid, and an analogue or derivative thereof. 25616. The composition of item 23785 wherein the agent is a thromboxane A2 receptor inhibitor selected from the group consisting of BM-531 (CAS No. 284464-46-6), ozagrel hydrochloride (CAS No. 78712-43-3), and an analogue or derivative thereof.
25617. The composition of item 23785 wherein the agent is a D2 dopamine receptor antagonist.
25618. The composition of item 23785 wherein the agent is a Peptidyl-Prolyl Cis/Trans lsomerase Inhibitor.
25619. The composition of item 23785 wherein the agent is a dopamine antagonist.
25620. The composition of item 23785 wherein the agent is an anesthetic compound.
25621. The composition of item 23785 wherein the agent is a clotting factor.
25622. The composition of item 23785 wherein the agent is a lysyl hydrolase inhibitor.
25623. The composition of item 23785 wherein the agent is a muscarinic receptor inhibitor.
25624. The composition of item 23785 wherein the agent is a superoxide anion generator.
25625. The composition of item 23785 wherein the agent is a steroid. 25626. The composition of item 23785 wherein the agent is an anti-proliferative agent selected from the group consisting of silibinin (CAS No. 22888-70-6), silymarin (CAS No. 65666-07-1), 1,2- hexanediol, dioctyl phthalate (CAS No. 117-81-7), zirconium (IV) oxide, glycyrrhizic acid, spermidine trihydrochloride, tetrahydrochloride, CGP 74514A, spermine tetrahydrochloride, NG-methyl-L-arginine acetate salt, galardin, and an analogue or derivative thereof.
25627. The composition of item 23785 wherein the agent is a diuretic.
25628. The composition of item 23785 wherein the agent is an anti-coagulant.
25629. The composition of item 23785 wherein the agent is a cyclic GMP agonist.
25630. The composition of item 23785 wherein the agent is an adenylate cyclase agonist.
25631. The composition of item 23785 wherein the agent is an antioxidant.
25632. The composition of item 23785 wherein the agent is a nitric oxide synthase inhibitor.
25633. The composition of item 23785 wherein the agent is an anti-neoplastic agent selected from tirapazamine (CAS No. 27314-97-2), fludarabine (CAS No. 21679-14-1), cladribine, imatinib mesilate, and an analogue or derivative thereof. 25634. The composition of item 23785 wherein the agent is a DNA synthesis inhibitor.
25635. The composition of item 23785 wherein the agent is a DNA alkylating agent selected from dacarbazine (CAS No. 4342- 03-4), temozolomide, procarbazine HCI, and an analogue or derivative thereof.
25636. The composition of item 23785 wherein the agent is a DNA methylation inhibitor.
25637. The composition of item 23785 wherein the agent is a NSAID agent.
25638. The composition of item 23785 wherein the agent is a peptidylglycine alpha-hydroxylating monooxygenase inhibitor.
25639. The composition of item 23785 wherein the agent is an MEK1/MEK 2 inhibitor.
25640. The composition of item 23785 wherein the agent is a NO synthase inhibitor.
25641. The composition of item 23785 wherein the agent is a retinoic acid receptor antagonist selected from isotretinoin (CAS No. 4759-48-2) and an analogue or derivative thereof.
25642. The composition of item 23785 wherein the agent is an ACE inhibitor. 25643. The composition of item 23785 wherein the agent is a glycosylation inhibitor.
25644. The composition of item 23785 wherein the agent is an intracellular calcium influx inhibitor.
25645. The composition of item 23785 wherein the agent is an anti-emetic agent.
25646. The composition of item 23785 wherein the agent is an acetylcholinesterase inhibitor.
25647. The composition of item 23785 wherein the agent is an ALK-5 receptor antagonist.
25648. The composition of item 23785 wherein the agent is a RAR/RXT antagonist.
25649. The composition of item 23785 wherein the agent is an elF-2a inhibitor.
25650. The composition of item 23785 wherein the agent is an S-adenosyl-L-homocysteine hydrolase inhibitor.
25651. The composition of item 23785 wherein the agent is an estrogen agonist.
25652. The composition of item 23785 wherein the agent is a serotonin receptor inhibitor. 25653. The composition of item 23785 wherein the agent is an antithrombotic agent.
25654. The composition of item 23785 wherein the agent is a tryptase inhibitor.
25655. The composition of item 23785 wherein the agent is a pesticide.
25656. The composition of item 23785 wherein the agent is a bone mineralization promotor.
25657. The composition of item 23785 wherein the agent is a bisphosphonate compound selected from risedronate and an analogue or derivative thereof.
25658. The composition of item 23785 wherein the agent is an anti-inflammatory compound.
25659. The composition of item 23785 wherein the agent is a DNA methylation promotor.
25660. The composition of item 23785 wherein the agent is an anti-spasmodic agent.
25661. The composition of item 23785 wherein the agent is a protein synthesis inhibitor.
25662. The composition of item 23785 wherein the agent is an D-glucosidase inhibitor. 25663. The composition of item 23785 wherein the agent is a calcium channel blocker.
25664. The composition of item 23785 wherein the agent is a pyruvate dehydrogenase activator.
25665. The composition of item 23785 wherein the agent is a prostaglandin inhibitor.
25666. The composition of item 23785 wherein the agent is a sodium channel inhibitor.
25667. The composition of item 23785 wherein the agent is a serine protease inhibitor.
25668. The composition of item 23785 wherein the agent is an intracellular calcium flux inhibitor.
25669. The composition of item 23785 wherein the agent is a JAK2 inhibitor.
25670. The composition of item 23785 wherein the agent is an androgen inhibitor.
25671. The composition of item 23785 wherein the agent is an aromatase inhibitor.
25672. The composition of item 23785 wherein the agent is an anti-viral agent. 25673. The composition of item 23785 wherein the agent is a 5-HT inhibitor.
25674. The composition of item 23785 wherein the agent is an FXR antagonist.
25675. The composition of item 23785 wherein the agent is an actin polymerization and stabilization promotor.
25676. The composition of item 23785 wherein the agent is an AXOR12 agonist.
25677. The composition of item 23785 wherein the agent is an angiotensin Il receptor agonist.
25678. The composition of item 23785 wherein the agent is a platelet aggregation inhibitor.
25679. The composition of item 23785 wherein the agent is a CB1/CB2 receptor agonist.
25680. The composition of item 23785 wherein the agent is a norepinephrine reuptake inhibitor.
25681. The composition of item 23785 wherein the agent is a selective serotonin reuptake inhibitor.
25682. The composition of item 23785 wherein the agent is a reducing agent. 25683. The composition of item 23785 wherein the agent is a immuno-modulator selected from Bay 11-7085, (-)-arctigenin, idazoxan hydrochloride, and an analogue or derivative thereof.
25684. The composition of item 23785 wherein the agent is isotretinoin.
25685. The composition of item 23785 wherein the agent is radicicol.
25686. The composition of item 23785 wherein the agent is clobetasol propionate.
25687. The composition of item 23785 wherein the agent is homoharringtonine.
25688. The composition of item 23785 wherein the agent is trichostatin A.
25689. The composition of item 23785 wherein the agent is brefeldin A.
25690. The composition of item 23785 wherein the agent is thapsigargin.
25691. The composition of item 23785 wherein the agent is dolastatin 15.
25692. The composition of item 23785 wherein the agent is cerivastatin. 25693. The composition of item 23785 wherein the agent is jasplakinolide.
25694. The composition of item 23785 wherein the agent is herbimycin A.
25695. The composition of item 23785 wherein the agent is pirfenidone.
25696. The composition of item 23785 wherein the agent is vinorelbine.
25697. The composition of item 23785 wherein the agent is 17-DMAG.
25698. The composition of item 23785 wherein the agent is tacrolimus.
25699. The composition of item 23785 wherein the agent is loteprednol etabonate.
25700. The composition of item 23785 wherein the agent is juglone.
25701. The composition of item 23785 wherein the agent is prednisolone.
25702. The composition of item 23785 wherein the agent is puromycin. 25703. The composition of item 23785 wherein the agent is 3-BAABE.
25704. The composition of item 23785 wherein the agent is cladribine.
25705. The composition of item 23785 wherein the agent is mannose-6-phosphate.
25706. The composition of item 23785 wherein the agent is 5-azacytidine.
25707. The composition of item 23785 wherein the agent is Ly333531 (ruboxistaurin).
25708. The composition of item 23785 wherein the agent is simvastatin.
25709. The composition of any one of items 25593- 25708 wherein the polymer is crosslinked.
25710. The composition of any one of items 25593- 25708 wherein the polymer reacts with mammalian tissue.
25711. The composition of any one of items 25593- 25708 wherein the polymer is a naturally occurring polymer.
25712. The composition of any one of items 25593- 25708 wherein the polymer is protein. 25713. The composition of any one of items 25593- 25708 wherein the polymer is carbohydrate.
25714. The composition of any one of items 25593- 25708 wherein the polymer is biodegradable polymer.
25715. The composition of any one of items 25593- 25708 wherein the polymer is crosslinked and biodegradable.
25716. The composition of any one of items 25593- 25708 wherein the polymer is nonbiodegradable polymer.
25717. The composition of any one of items 25593- 25708 wherein the polymer is collagen.
25718. The composition of any one of items 25593- 25708 wherein the polymer is methylated collagen.
25719. The composition of any one of items 25593- 25708 comprising fibrinogen.
25720. The composition of any one of items 25593- 25708 comprising thrombin.
25721. The composition of any one of items 25593- 25708 comprising calcium salt.
25722. The composition of any one of items 25593- 25708 comprising an antifibronolytic agent. 25723. The composition of any one of items 25593- 25708 comprising a fibrinogen analog.
25724. The composition of any one of items 25593- 25708 comprising albumin.
25725. The composition of any one of items 25593- 25708 comprising plasminogen.
25726. The composition of any one of items 25593- 25708 comprising von Willebrands factor.
25727. The composition of any one of items 25593- 25708 comprising Factor VIII.
25728. The composition of any one of items 25593- 25708 comprising hypoallergenic collagen.
25729. The composition of any one of items 25593- 25708, comprising atelopeptidic collagen.
25730. The composition of any one of items 25593- 25708, comprising telopeptide collagen.
25731. The composition of any one of items 25593- 25708, comprising crosslinked collagen.
25732. The composition of any one of items 25593- 25708, comprising aprotinin. 25733. The composition of any one of items 25593- 25708 comprising epsilon-amino-n-caproic acid.
25734. The composition of any one of items 25593- 25708 comprising gelatin.
25735. The composition of any one of items 25593- 25708 comprising protein conjugates.
25736. The composition of any one of items 25593- 25708 comprising gelatin conjugates.
25737. The composition of any one of items 25593- 25708 comprising hyaluronic acid.
25738. The composition of any one of items 25593- 25708 comprising a hyaluronic acid derivative.
25739. The composition of any one of items 25593- 25708 comprising a synthetic polymer.
25740. The composition of any one of items 25593- 25708 wherein the polymer is formed from reactants comprising a synthetic isocyanate-containing compound.
25741. The composition of any one of items 25593- 25708 comprising a synthetic isocyanate-containing compound.
25742. The composition of any one of items 25593- 25708 wherein the polymer is formed from reactants comprising a synthetic thiol-containing compound. 25743. The composition of any one of items 25593- 25708 comprising a synthetic thiol-containing compound.
25744. The composition of any one of items 25593- 25708 wherein the polymer is formed from reactants comprising a synthetic compound containing at least two thiol groups.
25745. The composition of any one of items 25593- 25708 comprising a synthetic compound containing at least two thiol groups.
25746. The composition of any one of items 25593- 25708 wherein the polymer is formed from reactants comprising a synthetic compound containing at least three thiol groups.
25747. The composition of any one of items 25593- 25708 comprising a synthetic compound containing at least three thiol groups.
25748. The composition of any one of items 25593- 25708 wherein the polymer is formed from reactants comprising a synthetic compound containing at least four thiol groups.
25749. The composition of any one of items 25593- 25708 comprising a synthetic compound containing at least four thiol groups.
25750. The composition of any one of items 25593- 25708 wherein the polymer is formed from reactants comprising a synthetic amino-containing compound. 25751. The composition of any one of items 25593- 25708 comprising a synthetic amino-containing compound.
25752. The composition of any one of items 25593- 25708 wherein the polymer is formed from reactants comprising a synthetic compound containing at least two amino groups.
25753. The composition of any one of items 25593- 25708 comprising a synthetic compound containing at least two amino groups.
25754. The composition of any one of items 25593- 25708 wherein the polymer is formed from reactants comprising a synthetic compound containing at least three amino groups.
25755. The composition of any one of items 25593- 25708 comprising a synthetic compound containing at least three amino groups.
25756. The composition of any one of items 25593- 25708 wherein the polymer is formed from reactants comprising a synthetic compound containing at least four amino groups.
25757. The composition of any one of items 25593- 25708 comprising a synthetic compound containing at least four amino groups.
25758. The composition of any one of items 25593- 25708 wherein the polymer is formed from reactants comprising a synthetic compound comprising a carbonyl-oxygen-succinimidyl group. 25759. The composition of any one of items 25593- 25708 comprising a synthetic compound comprising a carbonyl-oxygen- succinimidyl group.
25760. The composition of any one of items 25593- 25708 wherein the polymer is formed from reactants comprising a synthetic compound comprising at least two carbonyl-oxygen-succinimidyl groups.
25761. The composition of any one of items 25593- 25708 comprising a synthetic compound comprising at least two carbonyl- oxygen-succinimidyl groups.
25762. The composition of any one of items 25593- 25708 wherein the polymer is formed from reactants comprising a synthetic compound comprising at least three carbonyl-oxygen-succinimidyl groups.
25763. The composition of any one of items 25593- 25708 comprising a synthetic compound comprising at least three carbonyl- oxygen-succinimidyl groups.
25764. The composition of any one of items 25593- 25708 wherein the polymer is formed from reactants comprising a synthetic compound comprising at least four carbonyl-oxygen-succinimidyl groups.
25765. The composition of any one of items 25593- 25708 comprising a synthetic compound comprising at least four carbonyl- oxygen-succinimidyl groups.
25766. The composition of any one of items 25593- 25708 wherein the polymer is formed from reactants comprising a synthetic polyalkylene oxide-containing compound. 25767. The composition of any one of items 25593- 25708 comprising a synthetic polyalkylene oxide-containing compound.
25768. The composition of any one of items 25593- 25708 wherein the polymer is formed from reactants comprising a synthetic compound comprising both polyalkylene oxide and biodegradable polyester blocks.
25769. The composition of any one of items 25593- 25708 comprising a synthetic compound comprising both polyalkylene oxide and biodegradable polyester blocks.
25770. The composition of any one of items 25593- 25708 wherein the polymer is formed from reactants comprising a synthetic polyalkylene oxide-containing compound having reactive amino groups.
25771. The composition of any one of items 25593- 25708 comprising a synthetic polyalkylene oxide-containing compound having reactive amino groups.
25772. The composition of any one of items 25593- 25708 wherein the polymer is formed from reactants comprising a synthetic polyalkylene oxide-containing compound having reactive thiol groups.
25773. The composition of any one of items 25593- 25708 comprising a synthetic polyalkylene oxide-containing compound having reactive thiol groups.
25774. The composition of any one of items 25593- 25708 wherein the polymer is formed from reactants comprising a synthetic polyalkylene oxide-containing compound having reactive carbonyl-oxygen- succinimidyl groups.
25775. The composition of any one of items 25593- 25708 comprising a synthetic polyalkylene oxide-containing compound having reactive carbonyl-oxygen-succinimidyl groups.
25776. The composition of any one of items 25593- 25708 wherein the polymer is formed from reactants comprising a synthetic compound comprising a biodegradable polyester block.
25777. The composition of any one of items 25593- 25708 comprising a synthetic compound comprising a biodegradable polyester block.
25778. The composition of any one of items 25593- 25708 wherein the polymer is formed from reactants comprising a synthetic polymer formed in whole or part from lactic acid or lactide.
25779. The composition of any one of items 25593- 25708 comprising a synthetic polymer formed in whole or part from lactic acid or lactide.
25780. The composition of any one of items 25593- 25708 wherein the polymer is formed from reactants comprising a synthetic polymer formed in whole or part from glycolic acid or glycolide.
25781. The composition of any one of items 25593- 25708 comprising a synthetic polymer formed in whole or part from glycolic acid or glycolide. 25782. The composition of any one of items 25593- 25708 wherein the polymer is formed from reactants comprising polylysine.
25783. The composition of any one of items 25593- 25708 comprising polylysine.
25784. The composition of any one of items 25593- 25708 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound comprising a polyalkylene oxide portion.
25785. The composition of any one of items 25593- 25708 comprising (a) protein and (b) a compound comprising a polyalkylene oxide portion.
25786. The composition of any one of items 25593- 25708 wherein the polymer is formed from reactants comprising (a) protein and (b) polylysine.
25787. The composition of any one of items 25593- 25708 comprising (a) protein and (b) polylysine.
25788. The composition of any one of items 25593- 25708 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound having at least four thiol groups.
25789. The composition of any one of items 25593- 25708 comprising (a) protein and (b) a compound having at least four thiol groups. 25790. The composition of any one of items 25593- 25708 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound having at least four amino groups.
25791. The composition of any one of items 25593- 25708 comprising (a) protein and (b) a compound having at least four amino groups.
25792. The composition of any one of items 25593- 25708 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound having at least four carbonyl-oxygen-succinimide groups.
25793. The composition of any one of items 25593- 25708 comprising (a) protein and (b) a compound having at least four carbonyl-oxygen-succinimide groups.
25794. The composition of any one of items 25593- 25708 wherein the polymer is formed from reactants comprising (a) protein and (b) a compound having a biodegradable region formed from reactants selected from lactic acid, lactide, glycolic acid, glycolide, and epsilon- caprolactone.
25795. The composition of any one of items 25593- 25708 comprising (a) protein and (b) a compound having a biodegradable region formed from reactants selected from lactic acid, lactide, glycolic acid, glycolide, and epsilon-caprolactone.
25796. The composition of any one of items 25593- 25708 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound comprising a polyalkylene oxide portion. 25797. The composition of any one of items 25593- 25708 comprising (a) collagen and (b) a compound comprising a polyalkylene oxide portion.
25798. The composition of any one of items 25593- 25708 wherein the polymer is formed from reactants comprising (a) collagen and (b) polylysine.
25799. The composition of any one of items 25593- 25708 comprising (a) collagen and (b) polylysine.
25800. The composition of any one of items 25593- 25708 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound having at least four thiol groups.
25801. The composition of any one of items 25593- 25708 comprising (a) collagen and (b) a compound having at least four thiol groups.
25802. The composition of any one of items 25593- 25708 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound having at least four amino groups.
25803. The composition of any one of items 25593- 25708 comprising (a) collagen and (b) a compound having at least four amino groups.
25804. The composition of any one of items 25593- 25708 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound having at least four carbonyl-oxygen-succinimide groups. 25805. The composition of any one of items 25593- 25708 comprising (a) collagen and (b) a compound having at least four carbonyl-oxygen-succinimide groups.
25806. The composition of any one of items 25593- 25708 wherein the polymer is formed from reactants comprising (a) collagen and (b) a compound having a biodegradable region formed from reactants selected from lactic acid, lactide, glycolic acid, glycolide, and epsilon- caprolactone.
25807. The composition of any one of items 25593- 25708 comprising (a) collagen and (b) a compound having a biodegradable region formed from reactants selected from lactic acid, lactide, glycolic acid, glycolide, and epsilon-caprolactone.
25808. The composition of any one of items 25593- 25708 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound comprising a polyalkylene oxide portion.
25809. The composition of any one of items 25593- 25708 comprising (a) methylated collagen and (b) a compound comprising a polyalkylene oxide portion.
25810. The composition of any one of items 25593- 25708 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) polylysine.
25811. The composition of any one of items 25593- 25708 comprising (a) methylated collagen and (b) polylysine. 25812. The composition of any one of items item 23785- 23902 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound having at least four thiol groups.
25813. The composition of any one of items 25593- 25708 comprising (a) methylated collagen and (b) a compound having at least four thiol groups.
25814. The composition of any one of items 25593- 25708 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound having at least four amino groups.
25815. The composition of any one of items 25593- 25708 comprising (a) methylated collagen and (b) a compound having at least four amino groups.
25816. The composition of any one of items 25593- 25708 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound having at least four carbonyl- oxygen-succinimide groups.
25817. The composition of any one of items 25593- 25708 comprising (a) methylated collagen and (b) a compound having at least four carbonyl-oxygen-succinimide groups.
25818. The composition of any one of items 25593- 25708 wherein the polymer is formed from reactants comprising (a) methylated collagen and (b) a compound having a biodegradable region formed from reactants selected from lactic acid, lactide, glycolic acid, glycolide, and epsilon-caprolactone. 25819. The composition of any one of items 25593- 25708 comprising (a) methylated collagen and (b) a compound having a biodegradable region formed from reactants selected from lactic acid, lactide, glycolic acid, glycolide, and epsilon-caprolactone.
25820. The composition of any one of items 25593- 25708 wherein the polymer is formed from reactants comprising hyaluronic acid.
25821. The composition of any one of items 25593- 25708 comprising hyaluronic acid.
25822. The composition of any one of items 25593- 25708 wherein the polymer is formed from reactants comprising a hyaluronic acid derivative.
25823. The composition of any one of items 25593- 25708 comprising a hyaluronic acid derivative.
25824. The composition of any one of items 25593- 25708 wherein the polymer is formed from reactants comprising pentaerythritol poly(ethylene glycol)ether tetra-sulfhydryl of number average molecular weight between 3,000 and 30,000.
25825. The composition of any one of items 25593- 25708 comprising pentaerythritol poly(ethylene glycol)ether tetra-sulfhydryl of number average molecular weight between 3,000 and 30,000.
25826. The composition of any one of items 25593- 25708 wherein the polymer is formed from reactants comprising pentaerythritol poly(ethylene glycol)ether tetra-amino of number average molecular weight between 3,000 and 30,000.
25827. The composition of any one of items 25593- 25708 comprising pentaerythritol poly(ethylene glycol)ether tetra-amino of number average molecular weight between 3,000 and 30,000.
25828. The composition of any one of items 25593- 25708 wherein the polymer is formed from reactants comprising (a) a synthetic compound having a number average molecular weight between 3,000 and 30,000 and comprising a polyalkylene oxide region and multiple nucleophilic groups, and (b) a synthetic compound having a number average molecular weight between 3,000 and 30,000 and comprising a polyalkylene oxide region and multiple electrophilic groups.
25829. The composition of any one of items 25593- 25708 comprising (a) a synthetic compound having a number average molecular weight between 3,000 and 30,000 and comprising a polyalkylene oxide region and multiple nucleophilic groups, and (b) a synthetic compound having a number average molecular weight between 3,000 and 30,000 and comprising a polyalkylene oxide region and multiple electrophilic groups.
25830. The composition of any one of items 25593- 25708 wherein the composition comprises a colorant.
25831. The composition of any one of items 25593- 25708 wherein the composition is sterile. The following examples are offered by way of illustration, and limitation.
EXAMPLES
EXAMPLE 1 PREPARATION OF DRUG-LOADED MICROSPHERES BY SPRAY DRYING
3.6 grams of methoxy poly(ethylene glycol 5000))-block-(poly (DL-lactide) (65:35 MePEGPDLLA weight ratio) is dissolved in 200 ml methylene chloride. 100 mg of a drug, juglone, that is previously sieved through a 100 μm stainless steel sieve, is added and the resulting solution is spray dried. Inlet temperature 50cC, outlet temperature < 390C, aspirator 100%, flow rate 700 l/hr. The collected microspheres are dried under vacuum at room temperature overnight to produce uniform, spherical particles having size ranges of less than about 10 microns (typically about 0.5 to about 2 microns). This process for preparing drug-containing microspheres may be used with ZD-6474, AP-23573, synthadotin, S-0885, aplidine, ixabepilone, IDN-5390, SB-2723005, ABT-518, combretastatin, anecortave acetate, SB-715992, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17- DMAG, tacrolimus, temsirolimus, loteprednol etabonate, prednisolone, puromycin, 3-BAABE, cladribine, mannosemannose-6-phosphate, 5- azacytidine, Ly333531 (ruboxistaurin), dolastatin 15, and simvastatin, respectively.
EXAMPLE 2 DRUG-LOADED MICROSPHERES BY AN EMULSION PROCESS
100 ml of freshly prepared 10% polyvinyl alcohol (PVA) solution is added into a 600 ml beaker. The PVA solution is stirred at 2000 rpm for 30 minutes. Meanwhile, 40 mg jugalone is added to a solution of 800 mg MePEG5000-PDLLA (65:35) in 20 ml dichloromethane. This mixture is stirred for 20 min. The resultant mixture is added dropwise to the PVA solution while stirring at 2000 rpm with a Fisher DYNA-MIX stirrer. After addition is complete, the solution is allowed to stir for an additional 45 minutes. The microsphere solution is transferred to several disposable graduated polypropylene conical centrifuge tubes, washed with deionized water, and centrifuged at 2600 rpm for 10 minutes. The aqueous layer is decanted and the washing, centrifuging and decanting is repeated 3 times. The combined, washed microspheres are freeze-dried and vacuum dried to remove any excess water. This process for preparing drug-containing microspheres may be used with ZD-6474, AP-23573, synthadotin, S-0885, aplidine, ixabepilone, IDN-5390, SB-2723005, ABT-518, combretastatin, anecortave acetate, SB-715992, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17- DMAG, tacrolimus, temsirolimus, loteprednol etabonate, prednisolone, puromycin, 3-BAABE, cladribine, mannosemannose-6-phosphate, 5- azacytidine, Ly333531 (ruboxistaurin), dolastatin 15, and simvastatin, respectively.
EXAMPLE 3
DRUG-CONTAINING MICROSPHERES (50-100 MICRON) BY THE W/O/W EMULSION
PROCESS
Microspheres having an average size of about 50-100 microns are prepared using a 1 % PVA solution and 500 rpm stirring rate using the same procedure described in Example 2. EXAMPLE 4 JUGLONE CONTAINING MICELLES
MePEG2000 (4.1 g) and MePEG2000-PDLLA (60:40) (41 g) are combined in a vessel and heated to 75°C with stirring. After the polymers are completely melted and mixed, the temperature was decreased to 55°C. A juglone solution in tetrahydrofuran (4.6 g / 20 ml) is prepared and is poured into the polymer solution under constant stirring. Stirring is continued for an additional hour. The juglone containing micelles are dried at 50°C under vacuum to remove solvent. The resultant solid material is ground on a 2 mm mesh screen after cooling.
EXAMPLE 5
TETRA FUNCTIONAL POLY (ETHYLENE GLYCOL) SUCCINIMIDYL GLUTARATE, (PEG-SG4), NON-GELLING FORMULATION
. . . - A-1 ml syringe (syringe 1) equipped with a luer-lock mixing connector is filled with PEG-SG4 (100mg) (Sunbio, Inc., Orinda, CA). A 1 ml capped syringe (syringe 2) is filled with 0.25 ml of 6.3 mM HCI solution (pH 2.1). A 1 ml capped syringe (syringe 3) is filled with 0.25 ml 0.12 M monobasic sodium phosphate and 0.2 M sodium carbonate (pH 9.7) buffer. The solid contents of syringe 1 and the acidic solution of syringe 2 are mixed through a mixing connector by repeatedly transferring the contents from one syringe to the other. After complete mixing, the entire mixture is pushed into one of the syringes. The syringe containing the mixture then is attached to one inlet of an applicator (MICROMEDICS air assisted spray- applicator (Model SA-6105)). Syringe 3 containing the pH 9.7 solution is attached onto another inlet of the applicator. The formulation is applied to a tissue surface as specified by the applicator manufacturer. EXAMPLE 6 GELLING FORMULATION (PREMIX) I
A 1 ml syringe (syringe 1) equipped with a luer-lock mixing connector is filled with a mixture of PEG-SG4 (50mg) and PEG-SH4 (tetra functional poly (ethylene glycol) thiol) (50 mg) (Sunbio, Inc.) (referred to as "premix"). A 1 ml capped syringe (syringe 2) is filled with 0.25 ml of 6.3 mM HCI solution (pH 2.1). A 1 ml capped syringe (syringe 3) is filled with 0.25 ml 0.12 M monobasic sodium phosphate and 0.2 M sodium carbonate (pH 9.7) buffer. The components are mixed and applied to a tissue surface using the procedure described in Example 5.
EXAMPLE 7
TETRA FUNCTIONAL POLY (ETHYLENE GLYCOL) AMINE, (PEG-N4) GELLING
FORMULATION
A 1 ml syringe (syringe 1) equipped with a luer-lock mixing connector is filled with PEG-SG4 (50mg) and PEG-SH4 (tetra functional poly (ethylene glycol) thiol) (10, 25 or 40 mg). A 1 ml capped syringe (syringe 2) is filled with 0.25 ml of 6.3 mM HCI solution (pH 2.1). A 1 ml capped syringe (syringe 3) is filled with 0.25 ml 0.12 M monobasic sodium phosphate and 0.2 M sodium carbonate (pH 9.7) buffer. A 1 ml syringe (syringe 4) equipped with luer-lock mixing connector is filled with PEG-N4 (Sunbio, Inc.) (40, 25 or 10 mg) to make a mixture (50 mg total) of PEG-SH4 (in syringe 1) and PEG-N4 (in syringe 4). The contents of syringe 1 and syringe 2 are mixed through the mixing connector by repeatedly transferring the contents from one syringe to the other. After complete mixing, all of the formulation is pushed into one of the syringes which is then attached to one inlet of an applicator (MICROMEDICS air assisted spray-applicator (Model SA-6105)). Syringe 4 is attached to syringe 3 containing the pH 9.7 solution with a mixing connector. After complete mixing of the contents of syringe 3 and 4, the mixture is pushed into one of the syringes, which is then attached onto a second inlet of the applicator. The formulation is applied to a tissue surface as specified by the applicator manufacturer.
EXAMPLE 8 JUGLONE IN PEG-SG4
A 1 ml syringe (syringe 1) equipped with a luer-lock mixing connector is filled with PEG-SG4 (100 mg). A 1 ml capped syringe (syringe 2) is filled with 0.25 ml of 6.3 mM HCI solution (pH 2.1). A 1 ml capped syringe (syringe 3) is filled with 0.25 ml 0.12 M monobasic sodium phosphate and 0.2 M sodium carbonate (pH 9.7) buffer. A 1 ml syringe (syringe 4) equipped with luer-lock mixing connector is filled with 5 mg juglone, which is previously sieved through a 100um stainless steel sieve. The contents of syringe 4 and syringe 2 are mixed through a mixing connector by repeatedly transferring the contents from one syringe to the other. This solution is then used to reconstitute the solids in syringe 1. After complete mixing, all of the formulation is pushed into one of the syringes that is then attached to one inlet of an applicator (MICROMEDICS air assisted spray-applicator (Model SA-6105)). Syringe 3 containing the pH 9.7 solution is attached onto the other inlet of the applicator. The formulation is applied to a tissue surface as specified by the applicator manufacturer. This process for preparing drug-containing in-situ forming hydrogels may be repeated using ZD-6474, AP-23573, synthadotin, S-0885, aplidine, ixabepilone, IDN-5390, SB-2723005, ABT-518, combretastatin, anecortave acetate, SB-715992, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17-DMAG, tacrolimus, temsirolimus, loteprednol etabonate, prednisolone, puromycin, 3-BAABE, cladribine, mannosemannose-6-phosphate, 5-azacytidine, Ly333531 (ruboxistaurin), dolastatin 15, and simvastatin, respectively. EXAMPLE 9 JUGLONE IN PREMIX
A 1 ml syringe (syringe 1) equipped with a luer-lock mixing connector is filled with a mixture of PEG-SH4 (50 mg), PEG-SG4 (50 mg), and juglone (5 mg, sifted to less than 100 micron using a 100 micron stainless steel sieve). A 1 ml capped syringe (syringe 2) is filled with 0.25 ml of 6.3mM HCI solution (pH 2.1). A 1 ml capped syringe (syringe 3) is filled with 0.35 ml 0.24 M monobasic sodium phosphate and 0.4 M sodium carbonate (pH 10.0) buffer. The components are mixed and applied to a tissue surface using the procedure described in Example 7. This process for preparing drug-containing in-situ forming hydrogels may be repeated using ZD-6474, AP-23573, synthadotin, S-0885, aplidine, ixabepilone, IDN-5390, SB-2723005, ABT-518, combretastatin, anecortave acetate, SB-715992, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17-DMAG, tacrolimus, temsirolimus, loteprednol etabonate, prednisolone, puromycin, 3-BAABE, cladribine, mannosemannose-6-phosphate, 5-azacytidine, Ly333531 (ruboxistaurin), dolastatin 15, and simvastatin, respectively.
EXAMPLE 10
Two DRUGS COMBINED FOR INCORPORATION INTO AN IN SITU FORMING
HYDROGEL
A 1 ml syringe (syringe 1) equipped with a luer-lock mixing connector is filled with a mixture of PEG-SG4 (50 mg) and PEG-SH4 (50 mg). A 1 ml capped syringe (syringe 2) is filled with 0.25 ml of 6.3 rnM HCI solution (pH 2.1). A 1 ml capped syringe (syringe 3) is filled with 0.25 ml 0.12 M monobasic sodium phosphate and 0.2 M sodium carbonate (pH 9.7) buffer. Juglone (1 mg) and ABT-518 (1 mg), both sifted to less than 100 micron through a 100 micron stainless steel sieve, are then added to syringe 1. The components are mixed and applied to a tissue surface using the procedure described in Example 7.
EXAMPLE 11 JUGLONE LOADED MICROSPHERES IN PREMIX
A 1 ml syringe (syringe 1) equipped with a luer-lock mixing connector is filled with a mixture of PEG-SG4 (50 mg), PEG-SH4 (50 mg), and 2.7% juglone loaded MePEG5000-PDLLA (65:35) microspheres prepared by spray drying (0.5 or 2 mg) (prepared using the procedure described in Example 1). A 1 ml capped syringe (syringe 2) is filled with 0.25 ml of 6.3 mM HCI solution (pH 2.1). A 1 ml capped syringe (syringe 3) is filled 0.25 ml 0.12 M monobasic sodium phosphate and 0.2 M sodium carbonate (pH 9.7) buffer. The components are mixed and applied to a tissue surface using the procedure described in Example 7. This process for preparing drug-containing in situ forming hydrogel may be repeated using ZD-6474, AP-23573, synthadotin, S-0885, aplidine, ixabepilone, IDN-5390, SB-2723005, ABT-518, combretastatin, anecortave acetate, SB-715992, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17-DMAG, tacrolimus, temsirolimus, loteprednol etabonate, prednisolone, puromycin, 3-BAABE, cladribine, mannosemannose-6-phosphate, 5-azacytidine, Ly333531 (ruboxistaurin), dolastatin 15, and simvastatin, respectively.
EXAMPLE 12 INCORPORATION OF JUGLONE LOADED MICELLES INTO PREMIX
A 1 ml syringe (syringe 1) equipped with a luer-lock mixing connector is filled with a mixture of PEG-SG4 (50 mg) and PEG-SH4 (50 mg). A 2 ml serum vial is filled with 1.5 ml of 6.3 mM HCI solution (pH 2.1). A 1 ml capped syringe (syringe 2) is filled with 0.25 ml 0.12 M monobasic sodium phosphate and 0.2 M sodium carbonate (pH 9.7) buffer. A 2 ml serum vial is filled with juglone loaded micelles (2 mg or 8 mg) (prepared as in Example 4) and reconstituted with 1 ml of the pH 2.1 solution. 0.25 ml of the micelle solution is removed with a 1 ml syringe; the syringe is attached to syringe 1 containing the solids PEG-SG4 and PEG-SH4; and the components are mixed through the mixing connector by repeatedly transferring the contents from one syringe to the other. After complete mixing, the entire mixture is pushed into one of the syringes, which is then attached to one inlet of an applicator (MICROMEDICS air assisted spray- applicator (Model SA-6105)). Syringe 3 containing the pH 9.7 solution is attached onto the other inlet of the applicator. The formulation is applied to a tissue surface as specified by the applicator manufacturer.
EXAMPLE 13
TETRA FUNCTIONAL POLY (ETHYLENE GLYCOL) SUCCINIMIDYL GLUTARATE (PEG-
SG4), NON GELLING FORMULATION
A 3 ml syringe (syringe 1) equipped with a luer-lock mixing connector is filled with containing PEG-SG4 (400mg). A 3 ml capped syringe (syringe 2) is filled with 1.0 ml of 6.3 mM HCI solution (pH 2.1). A 3 ml capped syringe (syringe 3) is filled 1 ml 0.12 M monobasic sodium phosphate and 0.2 M sodium carbonate (pH 9.7) buffer. The components are mixed and applied to a tissue surface using the procedure described in Example 7.
EXAMPLE 14 GELLING FORMULATION (PREMIX) Il
A 3 ml syringe (syringe 1) equipped with a luer-lock mixing connector is filled with a mixture of PEG-SG4 (200mg) and PEG-SH4 (200 mg). A 3 ml capped syringe (syringe 2) is filled with 1.0 ml of 6.3 mM HCI solution (pH 2.1). A 3 ml capped syringe (syringe 3) is filled 1 ml 0.12 M monobasic sodium phosphate and 0.2 M sodium carbonate (pH 9.7) buffer. The components are mixed and applied to a tissue surface using the procedure described in Example 7.
EXAMPLE 15 JUGLONE LOADED PREMIX
A 3 ml syringe (syringe 1) equipped with a luer-lock mixing connector is filled with a mixture of PEG-SG4 (200mg), PEG-SH4 (200 mg), and juglone (7 mg or 14 mg). A 3 ml capped syringe (syringe 2) is filled with 1 ml of 6.3 mM HCI solution (pH 2.1). A 3 ml capped syringe (syringe 3) is filled 1.5 ml 0.24 M monobasic sodium phosphate and 0.4 M sodium carbonate (pH 10) buffer. The components are mixed and applied to a tissue surface using the procedure described in Example 7.
EXAMPLE 16
SCREENING ASSAY FOR ASSESSING THE EFFECT OF VARIOUS THERAPEUTIC AGENTS ON NITRIC OXIDE PRODUCTION BY MACROPHAGES
This example describes an assay for determining the effect of an agent on nitric oxide production by murine macrophages ((Chem. Ber. (1879) 12: 426; J. AOAC (1977) 60-594; Ann. Rev. Biochem. (1994) 63: 175). The murine macrophage cell line RAW 264.7 was trypsinized to remove cells from flasks and plated in individual wells of a 6-well plate. Approximately 2 x 106 cells were plated in 2 ml of media containing 5% heat-inactivated fetal bovine serum (FBS). RAW 264.7 cells were incubated at 37°C for 1.5 hours to allow adherence to plastic. The agent was prepared in DMSO at a concentration of 10~2 M and serially diluted 10-fold to give a range of stock concentrations (10"8 M to 10"2 M). Media was then removed and cells are incubated in 1 ng/ml of recombinant murine IFNγ and 5 ng/ml of LPS with or without mitoxantrone in fresh media containing 5% FBS. The agent was added to cells by directly adding agent DMSO stock solutions, prepared earlier, at a 1/1000 dilution, to each well. Plates containing IFNγ, LPS plus or minus the agent were incubated at 37°C for 24 hours.
At the end of the 24 hour period, supematants were collected from the cells and assayed for the production of nitrites. Each sample was tested in triplicate by aliquoting 50 μL of supernatant in a 96-well plate and adding 50 μl of Greiss Reagent A (0.5 g sulfanilamide, 1.5 ml H3PO4, 48.5 ml ddH2O) and 50 μl of Greiss Reagent B (0.05 g N-(i-naphthyl)- ethylenediamine, 1.5 ml H3PO4, 48.5 ml ddH2O). Optical density was read immediately on microplate spectrophotometer at 562 nm absorbance. Absorbance over triplicate wells was averaged after subtracting background and concentration values obtained from the nitrite standard curve (1μM to 2 mM). Inhibitory concentration of 50% (IC50) was determined by comparing average nitrite concentration to the positive control (cell stimulated with I FNy and LPS). An average of n=4 replicate experiments was used to determine . IC50 values for the agent. IC50 values were obtained for the following compounds: herbimycin A (112 nM); geldanamycin (51 nM); 17-AAG (743 nM); 17-DMAG (50 nM); ruboxistaurin (2299 nM); actinomycin D (28 nM); CGP 74514A (700 nM); Radicicol from Humicola fuscoatra, Humicola fuscoatra (21 nM); loteprednol etabonate (221 nM); clobetasol propionate (99 nM); homoharringtonine (75 nM)); trichostatin A from Streptomyces sp. (310 nM); thapsigargin (33 nM); 3-BAABE (637 nM); jasplakinolide (153 nM). The following compounds showed an IC50 value greater than 10,00OnM or no effect in this assay: simvastatin, tacrolimus, ribavarin, vinorelbine, pioglitazone, cladribine, pirfenidone, and mannose-6- phosphate.
Other compounds that may be tested for IC50 values in this assay include: e.g., ZD-6474, AP-23573, synthadotin, S-0885, aplidine, ixabepilone, IDN-5390, SB-2723005, ABT-518, combretastatin, anecortave acetate, SB-715992, temsirolimus, juglone, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, isotretinoin, brefeldin A, cerivastatin, dolastatin 15, prednisolone, puromycin, and 5-azacytidine.
EXAMPLE 17
SCREENING ASSAY FOR ASSESSING THE EFFECT OF VARIOUS THERAPEUTIC AGENTS ON TNF-ALPHA PRODUCTION BY MACROPHAGES
This example describes an assay for determining the effect of an agent on TNF-α production by human macrophages (J. Immunol. (2000) 165: 411-418; J. Immunol. (2000) 164: 4804-4811 ; J. Immunol Meth. (2000) 235 (1-2): 33-40). The human macrophage cell line, THP-1, was plated in a 12 well plate such that each well contains 1 X 106 cells in 2 ml of media containing 10% FCS. Opsonized zymosan was prepared by resuspending 20 mg of zymosan A in 2 ml of ddH2O and homogenizing until a uniform suspension was obtained. Homogenized zymosan was pelleted at 250 g and resuspended in 4 ml of human serum for a final concentration of 5 mg/ml and incubate in a 37°C water bath for 20 minutes to enable opsonization. An agent was prepared in DMSO at a concentration of 10"2 M and serially diluted 10-fold to give a range of stock concentrations (10"8 M to 10-2 M).
THP-1 cells were stimulated to produce TNFα by the addition of 1 mg/ml opsonized zymosan. The agent was added to THP-1 cells by directly adding DMSO stock solutions, prepared earlier, at a 1/1000 dilution, to each well. Each drug concentration was tested in triplicate wells. Plates were incubated at 37°C for 24 hours.
After 24 hour stimulation, supernatants were collected to quantify TNFα production. TNFα concentrations in the supernatants were determined by ELISA using recombinant human TNFα to obtain a standard curve. A 96-well MaxiSorb plate was coated with 100 μL of anti-human TNFα Capture Antibody diluted in Coating Buffer (0.1 M sodium carbonate pH 9.5) overnight at 4°C. The dilution of Capture Antibody used was lot- specific and was determined empirically. Capture antibody was then aspirated and the plate washed 3 times with Wash Buffer (PBS, 0.05% TWEEN-20). Plates were blocked for 1 hour at room temperature with 200 μL/well of Assay Diluent (PBS, 10% FCS pH 7.0). After blocking, plates were washed 3 times with Wash Buffer. Standards and sample dilutions were prepared as follows: (a) sample supernatants were diluted V8 and V16; (b) recombinant human TN Fa was prepared at 500 pg/ml and serially diluted to yield as standard curve of 7.8 pg/ml to 500 pg/ml. Sample supernatants and standards were assayed in triplicate and were incubated at room temperature for 2 hours after addition to the plate coated with Capture Antibody. The plates were washed 5 times and incubated with 100 μl_ of Working Detector (biotinylated anti-human TNFα detection antibody + avidin-HRP) for 1 hour at room temperature. Following this incubation, the plates were washed 7 times and 100 μl_ of Substrate Solution (tetramethylbenzidine, H2O2) was added to plates and incubated for 30 minutes at room temperature. Stop Solution (2 N HaSO4) was then added to the wells and a yellow color reaction was read at 450 nm with λ correction at 570 nm. Mean absorbance was determined from triplicate data readings and the mean background was subtracted. TNFα concentration values were obtained from the standard curve. Inhibitory concentration of 50% (IC50) was determined by comparing average TNFα concentration to the positive control (THP-1 cells stimulated with opsonized zymosan). An average of n=4 replicate experiments were used to determine IC50 values. IC50 values were obtained for the following compounds: herbimycin A (207 nM); geldanamycin (16 nM); 17-AAG (877 nM); 17-DMAG (112 nM); tacrolimus (0.5 nM); vinorelbine (244 nM); simvastatin (8876 nM); SB 203580 (171 nM); puromycin dihydrochloride from Streptomyces alboniger (751 nM); radicicol from Humicola fuscoatra, ( 212 nM); homoharringtonine (32 nM); brefeldin A (53 nM); dolastatin 15 (28 nM). The following compounds showed an IC50 value greater than 10,00OnM or no effect in this assay: ribavarin, pioglitazone, and mannose-6-phosphate. Other compounds that may be tested for IC50 values in this assay include: e.g., ZD-6474, AP-23573, synthadotin, S-0885, aplidine, ixabepilone, IDN-5390, SB-2723005, ABT-518, combretastatin, anecortave acetate, SB-715992, temsirolimus, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, isotretinoin, clobetasol propionatetrichostatin A, thapsigargin, cerivastatin, jasplakinolide, pirfenidone, loteprednol etabonate, juglone, prednisolone, 3-BAABE, cladribine, 5-azacytidine, and Ly333531(ruboxistaurin).
EXAMPLE 18 SURGICAL ADHESIONS MODEL TO ASSESS FIBROSIS INHIBITING AGENTS IN RATS
The rat caecal sidewall model is used to as to assess the anti- fibrotic capacity of formulations in vivo. Sprague Dawley rats are anesthetized with halothane. Using aseptic precautions, the abdomen is opened via a midline incision. The caecum is exposed and lifted out of the abdominal cavity. Dorsal and ventral aspects of the caecum are successively scraped a total of 45 times over the terminal 1.5 cm using a #10 scalpel blade. Blade angle and pressure are controlled to produce punctate bleeding while avoiding severe tissue damage. The left side of the abdomen is retracted and everted to expose a section of the peritoneal wall that lies proximal to the caecum. The superficial layer of muscle (transverses abdominis) is excised over an area of 1 X 2 cm2, leaving behind torn fibers from the second layer of muscle (internal oblique muscle). Abraded surfaces are tamponaded until bleeding stops. The abraded caecum is then positioned over the sidewall wound and attached by two sutures. The formulation is applied over both sides of the abraded caecum and over the abraded peritoneal sidewall. A further two sutures are placed to attach the caecum to the injured sidewall by a total of 4 sutures and the abdominal incision is closed in two layers. After 7 days, animals are evaluated post mortem with the extent and severity of adhesions being scored both quantitatively and qualitatively. Exemplary compounds that may be tested in this model include: e.g., ZD-6474, AP-23573, synthadotin, S-0885, aplidine, ixabepilone, IDN-5390, SB-2723005, ABT-518, combretastatin, anecortave acetate, SB-715992, temsirolimus, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, herbimycin A, geldanamycin, 17-AAG, 17-DMAG, cladribine, simvastatin, pirfenidone, mannose-6-phosphate, tacrolimus, vinorelbine, isotretinoin, radicicol, brefeldin A, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, temsirolimus, loteprednol etabonate, prednisolone, puromycin, mannose-6- phosphate, 5-azacytidine, dolastatin 15, and Ly333531(ruboxistaurin).
EXAMPLE 19
SURGICAL ADHESIONS MODEL TO ASSESS FIBROSIS INHIBITING AGENTS IN
RABBITS
The rabbit uterine horn model is used to assess the anti- fibrotic capacity of formulations in vivo. Mature New Zealand White (NZW) female rabbits are placed under general anesthetic. Using aseptic precautions, the abdomen is opened in two layers at the midline to expose the uterus. Both uterine horns are lifted out of the abdominal cavity and assessed for size on the French Scale of catheters. Horns between #8 and #14 on the French Scale (2.5-4.5 mm diameter) are deemed suitable for this model. Both uterine horns and the opposing peritoneal wall are abraded with a #10 scalpel blade at a 45° angle over an area 2.5 cm in length and 0.4 cm in width until punctuate bleeding is observed. Abraded surfaces are tamponaded until bleeding stops. The individual horns are then opposed to the peritoneal wall and secured by two sutures placed 2 mm beyond the edges of the abraded area. The formulation is applied and the abdomen is closed in three layers. After 14 days, animals are evaluated postmortem with the extent and severity of adhesions being scored both quantitatively and qualitatively. Exemplary compounds that may be tested in this model include: e.g., ZD-6474, AP-23573, synthadotin, S-0885, aplidine, ixabepilone, IDN-5390, SB-2723005, ABT-518, combretastatin, anecortave acetate, SB-715992, temsirolimus, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, herbimycin A, geldanamycin, 17-AAG, 17-DMAG, cladribine, simvastatin, pirfenidone, mannose-6- phosphate, tacrolimus, vinorelbine, isotretinoin, radicicol, brefeldin A, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, temsirolimus, loteprednol etabonate, prednisolone, puromycin, mannose-6-phosphate, 5- azacytidine, dolastatin 15, and Ly333531(ruboxistaurin).
EXAMPLE 20
SCREENING ASSAY FOR ASSESSING THE EFFECT OF VARIOUS THERAPEUTIC AGENTS ON FIBROBLAST CELL PROLIFERATION
This example describes an assay for determining the effect of an agent on fibroblast cell proliferation ((In vitro toxicol. (1990) 3: 219; Biotech. Histochem. (1993) 68: 29; Anal. Biochem. (1993) 213: 426). Fibroblasts at 70-90% confluency were trypsinized, replated at 600 cells/well in media in 96-well plates and allowed to" attach overnight. The agent was prepared in DMSO at a concentration of 10"2 M and diluted 10-fold to give a range of stock concentrations (10"8 M to 10'2 M). Drug dilutions were diluted 1/1000 in media and added to cells to give a total volume of 200 μL/well. Each drug concentration was tested in triplicate wells. Plates containing fibroblasts and the agent were incubated at 37°C for 72 hours.
To terminate the assay, the media was removed by gentle aspiration. A 1/400 dilution of CYQUANT 400X GR dye indicator (Molecular Probes; Eugene, OR) was added to 1X Cell Lysis buffer, and 200 μL of the mixture was added to the wells of the plate. Plates were incubated at room temperature, protected from light for 3-5 minutes. Fluorescence was read in a fluorescence microplate reader at ~480 nm excitation wavelength and -520 nm emission maxima. Inhibitory concentration of 50% (IC5o) was determined by taking the average of triplicate wells and comparing average relative fluorescence units to the DMSO control. An average of n=4 replicate experiments was used to determine IC50 values. IC50 values were obtained for the following compounds: herbimycin A (4.5 nM); geldanamycin (2.6 nM); 17-AAG (18 nM); 17-DMAG (6 nM); vinorelbine (4 nM); ruboxostaurin (4626 nM); cladribine (184 nM); simvastatin (896 nM); actinomycin D (0.5 nM); juglone (314 nM); Bay 11-7085 (173 nM); puromycin dihydrochloride from Streptomyces alboniger (254 nM); decitabine (486 nM); penfluridol (878 nM); radicicol from Humicola fuscoatra (26 nM); 5-azacytidine (806 nM); homoharringtonine (17 nM); trichostatin A from Streptomyces sp. (17 nM); brefeldin A (56 nM); thapsigargin (3 nM); AG-490 (623 nM); ammonium pyrrolidinedithiocarbamate (318 nM); dolastatin 15 (0.2 nM); cervistatin Na (33 nM); 3-BAABE (369 nM); jasplakinolide (3 nM); spermine tetrahydrochloride (283 nM). The following compounds showed an IC50 value greater than 10,00OnM or no effect in this assay: tacrolimus, ribavarin, pioglitazone, pirfenidone, and mannose-6- phosphate.
Other compounds that may be tested for IC50 values in this assay include: e.g., ZD-6474, AP-23573, synthadotin, S-0885, aplidine, ixabepilone, IDN-5390, SB-2723005, ABT-518, combretastatin, anecortave acetate, SB-715992, temsirolimus, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, isotretinoin, clobetasol propionate, loteprednol etabonate, and prednisolone.
EXAMPLE 2OA JUGLONE-LOADED DEGRADABLE MESH
A PLGA mesh (10/90 -VICRYL Mesh [VKMM], Ethicon (Somerville, NJ) of dimensions 5 cm x 5 cm is dipped into a solution of isopropanol (HPLC grade). The mesh is then dried under vacuum. 3 g PLGA (50/50, IV=0.15, Durect Corporation, Pelham, AL) is dossolved in 15 ml ethyl acetate (HPLC Grade, Fisher) in a 20 mL glass scintillation vial. 10.3 mg juglone is added to the polymer solution and the resultant mixture was vortexed for 10 minutes. The mesh is held with a pair of tweezers and is dipped into this polymer drug solution. The excess solution is removed by touching the mesh to the neck of the vial. The mesh is air dried by holding the coated mesh in front of a small electrical fan for 2-3 minutes. The mesh is placed in a PTFE Petri dish and is further dried in a fume hood for 2 hours. The coated mesh is further dried under vacuum in a vacuum oven for 24 hours. The coated mesh is then placed between 2 pieces of release liner (REXAM A10, 6 cm x 6 cm) after which it is sealed in a heat sealable plastic pouch. The mesh is then sterilized by gamma radiation. The mesh can then be used to wrap around all or a portion of the electrical device immediately prior to use. This process for preparing drug-containing mesh may be repeated using ZD-6474, AP-23573, synthadotin, S-0885, aplidine, ixabepilone, IDN-5390, SB-2723005, ABT-518, combretastatin, anecortave acetate, SB-715992, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, jasplakinolide, pirfenidone, loteprednol etabonate, juglone, prednisolone, 3-BAABE, cladribine, mannose-6-phosphate, 5-azacytidine, puromycin, tacrolimus, paclitaxel, simvastatin, isotretinoin, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, temsirolimus, 5-azacytidine, dolastatin 15, and Ly333531 (ruboxistaurin),
EXAMPLE 21
EVALUATION OF JUGLONE CONTAINING MESH ON INTIMAL HYPERPLASIA
DEVELOPMENT IN A RAT BALLOON INJURY CAROTID ARTERY MODEL AS AN
EXAMPLE TO EVALUATE FIBROSIS INHIBITING AGENTS
A rat balloon injury carotid artery model is used to demonstrate the efficacy of a fibrosis-inhibiting agent (e.g., juglone) containing mesh system on the development of intimal hyperplasia fourteen days following placement. Control Group
Wistar rats weighing 400 - 500 g are anesthetized with 1.5% halothane in oxygen, and the left external carotid artery is exposed. An A 2 French FOGARTY balloon embolectomy catheter (Baxter, Irvine, CA) is advanced through an arteriotomy in the external carotid artery down the left common carotid artery to the aorta. The balloon is inflated with enough saline to generate slight resistance (approximately 0.02 ml) and it is withdrawn with a twisting motion to the carotid bifurcation. The balloon is then deflated and the procedure repeated twice more. This technique produces distension of the arterial wall and denudation of the endothelium. The external carotid artery is ligated after removal of the catheter. The right common carotid artery is not injured and is used as a control.
Local Perivascular Juglone Treatment
Immediately after injury of the left common carotid artery, a 1 cm long distal segment of the artery is exposed and treated with a 1x1 cm juglone-containing mesh (345 ug juglone in a 50:50 PLG coating on a 10:90 PLG mesh - Example 61). The wound is then closed, and the animals are kept for 14 days.
Histology and immunohistochemistry
At the time of sacrifice, the animals are euthanized with carbon dioxide and pressure perfused at 100 mmHg with 10% phosphate buffered formaldehyde for 15 minutes. Both carotid arteries are harvested and left overnight in fixative. The fixed arteries are processed and embedded in paraffin wax. Serial cross-sections are cut at 3 μm thickness every 2 mm within and outside the implant region of the injured left carotid artery and at corresponding levels in the control right carotid artery. Cross-sections are stained with Mayer's hematoxylin-and-eosin for cell count and with Movat's pentachrome stains for morphometry analysis and for extracellular matrix composition assessment. Other compounds that may be tested in this model include, e.g., ZD-6474, AP-23573, synthadotin, S-0885, aplidine, ixabepilone, IDN- 5390, SB-2723005, ABT-518, combretastatin, anecortave acetate, SB- 715992, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, dolastatin 15, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17- DMAG, tacrolimus, temsirolimus, loteprednol etabonate, prednisolone, puromycin, 3-BAABE, cladribine, mannose-6-phosphate, 5-azacytidine, Ly333531 (ruboxistaurin), simvastatin geldanamycin, and 17-AAG,
EXAMPLE 22
EFFECT OF PACLITAXEL AND OTHER ANTI-MICROTUBULE AGENTS ON MATRIX METALLOPROTEINASE PRODUCTION
A. Materials and Methods
1) 1L-1 stimulated AP-1 transcriptional activity is inhibited by paclitaxel
Chondrocytes were transfected with constructs containing an AP-1 driven CAT reporter gene, and stimulated with IL-I , IL-1 (50 ng/ml) was added and incubated for 24 hours in the absence and presence of paclitaxel at various concentrations. Paclitaxel treatment decreased CAT activity in a concentration dependent manner (mean ± SD). The data noted with an asterisk (*) have significance compared with IL-1 -induced CAT activity according to a t-test, P<0.05. The results shown are representative of three independent experiments. 2) Effect of paclitaxel on 1L-1 induced AP-1 DNA binding activity, AP-1 DNA
Binding activity was assayed with a radiolabeled human AP-1 sequence probe and gel mobility shift assay. Extracts from chondrocytes untreated or treated with various amounts of paclitaxel (10"7 to 10'5 M) followed by IL-1β (20 ng/ml) were incubated with excess probe on ice for 30 minutes, followed by non-denaturing gel electrophoresis. The "com" lane contains excess unlabeled AP-1 oligonucleotide. The results shown are representative of three independent experiments.
3) Effect of paclitaxel on IL-1 induced MMP-1 and MMP-3 mRNA expression
Cells were treated with paclitaxel at various concentrations (10~7 to 10"5 M) for 24 hours, then treated with IL-1β (20 ng/ml) for additional 18 hours in the presence of paclitaxel. Total RNA was isolated, and the MMP-1 mRNA levels were determined by Northern blot analysis. The blots . were subsequently stripped and reprobed with 32P-radiolabeled rat GAPDH cDNA, which was used as a housekeeping gene. The results shown are representative of four independent experiments. Quantitation of collagenase-1 and stromelysin-expression mRNA levels were conducted. The MMP-1 and MMP-3 expression levels were normalized with GAPDH.
4) Effect of other anti-microtυbυles on collagenase expression Primary chondrocyte cultures were freshly isolated from calf cartilage. The cells were plated at 2.5 x 106 per ml in 100 x 20 mm culture dishes and incubated in Ham's F12 medium containing 5% FBS overnight at 370C. The cells were starved in serum-free medium overnight and then treated with anti-microtubule agents at various concentrations for 6 hours. IL-1 (20 ng/ml) was then added to each plate and the plates incubated for an additional 18 hours. Total RNA was isolated by the acidified guanidine isothiocyanate method and subjected to electrophoresis on a denatured gel. Denatured RNA samples (15 μg) were analyzed by gel electrophoresis in a 1% denatured gel, transferred to a nylon membrane and hydridized with the 32P-labeled collagenase cDNA probe. 32P-labeled glyceraldehyde phosphate dehydrase (GAPDH) cDNA as an internal standard to ensure roughly equal loading. The exposed films were scanned and quantitatively analyzed with IMAGEQUANT.
B. Results
1) Promoters on the family of matrix metalloproteinases Figure 1 shows that all matrix metalloproteinases contained the transcriptional elements AP-1 and PEA-3 with the exception of gelatinase B. It has been well established that expression of matrix metalloproteinases such as collagenases and stromelysins are dependent on the activation of the transcription factors AP-1. Thus inhibitors of AP-1 may inhibit the expression of matrix metalloproteinases.
2) Effect of paclitaxel on AP-1 transcriptional activity As demonstrated in Figure 2, IL-1 stimulated AP-1 transcriptional activity 5-fold. Pretreatment of transiently transfected chondrocytes with paclitaxel reduced IL-1 induced AP-1 reporter gene CAT activity. Thus, IL-1 induced AP-1 activity was reduced in chondrocytes by paclitaxel in a concentration dependent manner (10"7 to 10"5 M). These data demonstrated that paclitaxel was a potent inhibitor of AP-1 activity in chondrocytes.
3) Effect of paclitaxel on AP-1 DNA binding activity
To confirm that paclitaxel inhibition of AP-1 activity was not due to nonspecific effects, the effect of paclitaxel on IL-1 induced AP-1 binding to oligonucleotides using chondrocyte nuclear lysates was examined. As shown in Figure 3, IL-1 induced binding activity decreased in lysates from chondrocyte which had been pretreated with paclitaxel at concentration 10"7 to 10"5 M for 24 hours. Paclitaxel inhibition of AP- 1 transcriptional activity closely correlated with the decrease in AP-1 binding to DNA.
4) Effect of paclitaxel on collagenase and stromelysin expression Since paclitaxel was a potent inhibitor of AP-1 activity, the effect of paclitaxel or IL-1 induced collagenase and stromelysin expression, two important matrix metalloproteinases involved in inflammatory diseases was examined. Briefly, as shown in Figure 4, IL-1 induction increases collagenase and stromelysin mRNA levels in chondrocytes. Pretreatment of chondrocytes with paclitaxel for 24 hours significantly reduced the levels of collagenase and stromelysin mRNA. At 10"5 M paclitaxel, there was complete inhibition. The results show that paclitaxel completely inhibited the expression of two matrix metalloproteinases at concentrations similar to which it inhibits AP-1 activity.
5) Effect of other anti-microtubules on collagenase expression Figures 5A-H demonstrate that anti-microtubule agents inhibited collagenase expression. Expression of collagenase was stimulated by the addition of IL-1 which is a proinflammatory cytokine. Preincubation of chondrocytes with various anti-microtubule agents, specifically LY290181, hexylene glycol, deuterium oxide, glycine ethyl ester, ethylene glycol bis-(succinimidylsuccinate), tubercidin, AIF3, and epothilone, all prevented IL-1-induced collagenase expression at concentrations as low as 1 x 10"7 M.
C. Discussion
Paclitaxel was capable of inhibiting collagenase and stromelysin expression in vitro at concentrations of 10 M. Since this inhibition may be explained by the inhibition of AP-1 activity, a required step in the induction of all matrix metalloproteinases with the exception of gelatinase B, it is expected that paclitaxel may inhibit other matrix metalloproteinases which are AP-1 dependent. The levels of these matrix metalloproteinases are elevated in all inflammatory diseases and play a principle role in matrix degradation, cellular migration and proliferation, and angiogenesis. Thus, paclitaxel inhibition of expression of matrix metalloproteinases such as collagenase and stromelysin can have a beneficial effect in inflammatory diseases.
In addition to paclitaxel's inhibitory effect on collagenase expression, LY290181 , hexylene glycol, deuterium oxide, glycine ethyl ester, AIF3, tubercidin epothilone, and ethylene glycol bis- (succinimidylsuccinate), all prevented IL-1 -induced collagenase expression at concentrations as low as 1 x 10~7 M. Thus, anti-microtubule agents are capable of inhibiting the AP-1 pathway at varying concentrations. The following compounds are additional examples that may also be tested using these methods: ZD-6474, AP-23573, synthadotin, S-0885, aplidine, ixabepilone, IDN-5390, juglone, SB-2723005, ABT-518, combretastatin, anecortave acetate, SB-715992, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17- DMAG, tacrolimus, temsirolimus, loteprednol etabonate, prednisolone, puromycin, 3-BAABE, cladribine, mannose-6-phosphate, 5-azacytidine, Ly333531(ruboxistaurin), dolastatin 15, and simvastatin.
EXAMPLE 23 INHIBITION OF ANGIOGENESIS BY PACLITAXEL
Chick Chorioallantoic Membrane ("CAM") Assays
Fertilized, domestic chick embryos were incubated for 3 days prior to shell-less culturing. In this procedure, the egg contents were emptied by removing the shell located around the air space. The interior shell membrane was then severed and the opposite end of the shell was perforated to allow the contents of the egg to gently slide out from the blunted end. The egg contents were emptied into round-bottom sterilized glass bowls and covered with petri dish covers. These were then placed into an incubator at 90% relative humidity and 3% CO2 and incubated for 3 days.
Paclitaxel (Sigma, St. Louis, Ml) was mixed at concentrations of 0.25, 0.5, 1 , 5, 10, 30 μg per 10 ul aliquot of 0.5% aqueous methylcellulose. Since paclitaxel is insoluble in water, glass beads were used to produce fine particles. Ten microliter aliquots of this solution were dried on parafilm for 1 hour forming disks 2 mm in diameter. The dried disks containing paclitaxel were then carefully placed at the growing edge of each CAM at day 6 of incubation. Controls were obtained by placing paclitaxel- free methylcellulose disks on the CAMs over the same time course. After a 2 day exposure (day 8 of incubation) the vasculature was examined with the aid of a stereomicroscope. Liposyn II, a white opaque solution, was injected into the CAM to increase the visibility of the vascular details. The vasculature of unstained, living embryos were imaged using a Zeiss stereomicroscope which was interfaced with a video camera (Dage-MTI Inc., Michigan City, IN). These video signals were then displayed at 16Ox magnification and captured using an image analysis system (Vidas, Kontron; Etching, Germany). Image negatives were then made on a graphics recorder (Model 3000; Matrix Instruments, Orangeburg, NY).
The membranes of the 8 day-old shell-less embryo were flooded with 2% glutaraldehyde in 0.1 M sodium cacodylate buffer; additional fixative was injected under the CAM. After 10 minutes in situ, the CAM was removed and placed into fresh fixative for 2 hours at room temperature. The tissue was then washed overnight in cacodylate buffer containing 6% sucrose. The areas of interest were postfixed in 1 % osmium tetroxide for 1.5 hours at 4°C. The tissues were then dehydrated in a graded series of ethanols, solvent exchanged with propylene oxide, and embedded in Spurr resin. Thin sections were cut with a diamond knife, placed on copper grids, stained, and examined in a Joel 1200EX electron microscope. Similarly, 0.5 mm sections were cut and stained with toluene blue for light microscopy.
At day 11 of development, chick embryos were used for the corrosion casting technique. Mercox resin (Ted PeIIa, Inc., Redding, CA) was injected into the CAM vasculature using a 30-gauge hypodermic needle. The casting material consisted of 2.5 grams of Mercox CL-2B polymer and 0.05 grams of catalyst (55% benzoyl peroxide) having a 5 minute polymerization time. After injection, the plastic was allowed to sit in situ for an hour at room temperature and then overnight in an oven at 650C. The CAM was then placed in 50% aqueous solution of sodium hydroxide to digest all organic components. The plastic casts were washed extensively in distilled water, air-dried, coated with gold/palladium, and viewed with the Philips 501 B scanning electron microscope.
Results of the assay were as follows. At day 6 of incubation, the embryo was centrally positioned to a radially expanding network of blood vessels; the CAM developed adjacent to the embryo. These growing vessels lie close to the surface and are readily visible making this system an idealized model for the study of angiogenesis. Living, unstained capillary networks of the CAM may be imaged noninvasively with a stereomicroscope.
Transverse sections through the CAM show an outer ectoderm consisting of a double cell layer, a broader mesodermal layer containing capillaries which lie subjacent to the ectoderm, adventitial cells, and an inner, single endodermal cell layer. At the electron microscopic level, the typical structural details of the CAM capillaries are demonstrated. Typically, these vessels lie in close association with the inner cell layer of ectoderm.
After 48 hours exposure to paclitaxel at concentrations of 0.25, 0.5, 1 , 5, 10, or 30 μg, each CAM was examined under living conditions with a stereomicroscope equipped with a video/computer interface in order to evaluate the effects on angiogenesis. This imaging setup was used at a magnification of 16Ox which permitted the direct visualization of blood cells within the capillaries; thereby blood flow in areas of interest may be easily assessed and recorded. For this study, the inhibition of angiogenesis was defined as an area of the CAM (measuring 2-6 mm in diameter) lacking a capillary network and vascular blood flow. Throughout the experiments, avascular zones were assessed on a 4 point avascular gradient (Table 1). This scale represents the degree of overall inhibition with maximal inhibition represented as a 3 on the avascular gradient scale. Paclitaxel was very consistent and induced a maximal avascular zone (6 mm in diameter or a 3 on the avascular gradient scale) within 48 hours depending on its concentration.
Table 1 Avascular Gradient
0 — normal vascularity
1 - lacking some microvascular movement
2*- small avascular zone approximately 2 mm in diameter 3*- avascularity extending beyond the disk (6 mm in diameter)
* - indicates a positive antiangiogenesis response
The dose-dependent, experimental data of the effects of paclitaxel at different concentrations are shown in Table 2.
Table 2
Agent Delivery Vehicle Concentration Inhibition/n paclitaxel methylcellulose (10 ul) 0.25 ug 2/11 methylcellulose (10 ul) 0.5 ug 6/11 methylcellulose (10 ul) 1 ug 6/15 methylcellulose (10 ul) 5 ug 20/27 methylcellulose (10 ul) 10 ug 16/21 methylcellulose (10 ul) 30 ug 31/31
Typical paclitaxel-treated CAMs are also shown with the transparent methylcellulose disk centrally positioned over the avascular zone measuring 6 mm in diameter. At a slightly higher magnification, the periphery of such avascular zones was clearly evident; the surrounding functional vessels were often redirected away from the source of paclitaxel. Such angular redirecting of blood flow was never observed under normal conditions. Another feature of the effects of paclitaxel was the formation of blood islands within the avascular zone representing the aggregation of blood cells.
In summary, this study demonstrated that 48 hours after paclitaxel application to the CAM, angiogenesis was inhibited. The blood vessel inhibition formed an avascular zone which was represented by three transitional phases of paclitaxePs effect. The central, most affected area of the avascular zone contained disrupted capillaries with extravasated red blood cells; this indicated that intercellular junctions between endothelial cells were absent. The cells of the endoderm and ectoderm maintained their intercellular junctions and therefore these germ layers remained intact; however, they were slightly thickened. As the normal vascular area was approached, the blood vessels retained their junctional complexes and therefore also remained intact. At the periphery of the paclitaxel-treated zone, further blood vessel growth was inhibited which was evident by the typical redirecting or "elbowing" effect of the blood vessels. Exemplary compounds that may be tested in this model include: e.g., ZD-6474, AP- 23573, synthadotin, S-0885, aplidine, ixabepilone, IDN-5390, SB-2723005, ABT-518, combretastatin, anecortave acetate, SB-715992, temsirolimus, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, herbimycin A, geldanamycin, 17-AAG, 17-DMAG, cladribine, simvastatin, pirfenidone, mannose-6-phosphate, tacrolimus, vinorelbine, dolastatin 15, and ruboxistaurin.
EXAMPLE 24
SCREENING ASSAY FOR ASSESSING THE EFFECT OF PACLITAXEL ON SMOOTH
MUSCLE CELL MIGRATION
This example describes an assay for determining the effect of an agent on human smooth muscle cell migration (Biotechniques (2000) 29: 81 ; J. Immunol Methods (2001) 254: 85). Primary human smooth muscle cells were starved of serum in smooth muscle cell basal media containing insulin and human basic fibroblast growth factor (bFGF) for 16 hours prior to the assay. For the migration assay, cells were trypsinized to remove cells from flasks, washed with migration media and diluted to a concentration of 2-2.5 X 105 cells/ml in migration media. Migration media consisted of phenol red free Dulbecco's Modified Eagle Medium (DMEM) containing 0.35% human serum albumin. A 100 μL volume of smooth muscle cells (approximately 20,000-25,000 cells) was added to the top of a Boyden chamber assembly (Chemicon QCM CHEMOTAXIS 96-well migration plate). To the bottom wells, the chemotactic agent, recombinant human platelet derived growth factor (rhPDGF-BB) was added at a concentration of 10 ng/ml in a total volume of 150 μL. An agent ( e.g., paclitaxel) was prepared in DMSO at a concentration of 10~2 M and serially diluted 10-fold to give a range of stock concentrations (10"8 M to 10'2 M). The agent was added to cells by directly adding the agent-DMSO stock solutions, prepared earlier, at a 1/1000 dilution, to the cells in the top chamber. Plates were incubated for 4 hours to allow cell migration. At the end of the 4 hour period, cells in the top chamber were discarded and the smooth muscle cells attached to the underside of the filter were detached for 30 minutes at 37°C in Cell Detachment Solution (Chemicon). Dislodged cells were lysed in lysis buffer containing the DNA binding CYQUANT GR dye and incubated at room temperature for 15 minutes. Fluorescence was read in a fluorescence microplate reader at ~480 nm excitation wavelength and ~520 nm emission maxima. Relative fluorescence units from triplicate wells were averaged after subtracting background fluorescence (control chamber without chemoattractant) and average number of cells migrating was obtained from a standard curve of smooth muscle cells serially diluted from 25,000 cells/well down to 98 cells/well. Inhibitory concentration of 50% (IC50) was determined by comparing the average number of cells migrating in the presence of the agent to the positive control (smooth muscle cell chemotaxis in response to rhPDGF-BB). IC5O values were obtained for the following compounds: paclitaxel (0.76 nM; see Figure 6); herbimycin A (738 nM); geldanamycin (86 nM); 17-AAG (429 nM); puromycin dihydrochloride from Streptomyces alboniger (324 nM); homoharringtonine (83 nM); cervistatin Na (90 nM). Simvastatin had an IC50 value greater than 10,00OnM or no effect in this assay.
Other compounds that may be tested in this assay include: e.g., juglone, ZD-6474, AP-23573, synthadotin, S-0885, aplidine, ixabepilone, IDN-5390, SB-2723005, ABT-518, combretastatin, anecortave acetate, SB-715992, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, isotretinoin, radicicol, clobetasol propionate, trichostatin A, brefeldin A, thapsigargin, dolastatin 15, jasplakinolide, pirfenidone, vinorelbine, 17-DMAG, tacrolimus, temsirolimus, loteprednol etabonate, prednisolone, 3-BAABE, cladribine, mannose-6-phosphate, 5- azacytidine, and Ly333531(ruboxistaurin). EXAMPLE 25
SCREENING ASSAY FOR ASSESSING THE EFFECT OF VARIOUS COMPOUNDS ON IL-
1β PRODUCTION BY MACROPHAGES
This example describes a screening assay for assessing the effect of an agent on IL-1β production by human macrophages. The assay protocol is based on the following references: J. Immunol. (2000) 165: 411- 418; J. Immunol. (2000) 164: 4804-4811 ; J. Immunol Meth. (2000) 235 (1- 2): 33-40). The human macrophage cell line, THP-1 , was plated in a 12 well plate such that each well contains 1 X 106 cells in 2 ml of media containing 10% FCS. Opsonized zymosan was prepared by resuspending 20 mg of zymosan A in 2 ml of ddhfeO and homogenizing until a uniform suspension was obtained. Homogenized zymosan was pelleted at 250 g and resuspended in 4 ml of human serum for a final concentration of 5 mg/ml and incubated in a 37°C water bath for 20 minutes to enable opsonization. An agent was prepared in DMSO at a concentration of 10"2 M and serially diluted 10-fold to give a range of stock concentrations (10'8 M to 10~2 M).
THP-1 cells were stimulated to produce IL-1β by the addition of 1 mg/ml opsonized zymosan. Geldanamycin was added to THP-1 cells by directly adding DMSO stock solutions, prepared earlier, at a 1/1000 dilution, to each well. Each drug concentration was tested in triplicate wells. Plates were incubated at 37°C for 24 hours.
After a 24 hour stimulation, supematants were collected to quantify IL-1β production. IL-1β concentrations in the supematants were determined by ELISA using recombinant human IL-1β to obtain a standard curve. A 96-well MaxiSorb plate was coated with 100 μL of anti-human IL- 1β Capture Antibody diluted in Coating Buffer (0.1 M Sodium carbonate pH 9.5) overnight at 4°C. The dilution of Capture Antibody used was lot-specific and was determined empirically. Capture antibody was then aspirated and the plate washed 3 times with Wash Buffer (PBS, 0.05% TWEEN-20). Plates were blocked for 1 hour at room temperature with 200 μL/well of Assay Diluent (PBS, 10% FCS pH 7.0). After blocking, plates were washed 3 times with Wash Buffer. Standards and sample dilutions were prepared as follows: (a) sample supernatants were diluted % and %; (b) recombinant human IL-1β was prepared at 1000 pg/ml and serially diluted to yield as standard curve of 15.6 pg/ml to 1000 pg/ml. Sample supernatants and standards were assayed in triplicate and were incubated at room temperature for 2 hours after addition to the plate coated with Capture Antibody. The plates were washed 5 times and incubated with 100 μl_ of Working Detector (biotinylated anti-human IL-1β detection antibody + avidin- HRP) for 1 hour at room temperature. Following this incubation, the plates were washed 7 times and 100 μl_ of Substrate Solution (Tetramethylbenzidine, H2O2) was added to plates and incubated for 30 minutes at room temperature. Stop Solution (2 N H2SO4) was then added to the wells and a yellow color reaction was read at 450 nm with λ correction at 570 nm. Mean absorbance was determined from triplicate data readings and the mean background is subtracted. IL-1β concentration values were obtained from the standard curve. Inhibitory concentration of 50% (IC50) was determined by comparing average IL-1β concentration to the positive control (THP-1 cells stimulated with opsonized zymosan). IC50 values were measured for the following compounds: herbimycin A (1027 nM); geldanamycin (267 nM); 17-AAG (253 nM); 17-DMAG (78 nM); vinorelbine (2359 nM); simvastatin (9572 nM), pirfenidone (80 nM); prednisolone 21- acetate (6.5 nM); NG-methyl-L-arginine acetate salt (<0.02 nM); tinidazole (96 nM); radicicol from Humicola fuscoatra (369 nM); loteprednol etabonate (185 nM); homoharringtonine (7 nM); brefeldin A (42 nM). The following compounds showed an IC50 value greater than 10,00OnM or no effect in this assay: mannose-6-phosphate, ribavarin, tacrolimus, pioglitazone, ruboxostaurin, and cladribine.
Other compounds that may be tested for IC50 values in this assay include: e.g., ZD-6474, AP-23573, synthadotin, S-0885, aplidine, ixabepilone, IDN-5390, SB-2723005, ABT-518, combretastatin, anecortave acetate, SB-715992, temsirolimus, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, juglone, isotretinoin, clobetasol propionate, trichostatin A1 thapsigargin, dolastatin 15, cerivastatin, jasplakinolide, hpuromycin, 3-BAABE, and 5-azacytidine.
EXAMPLE 26
SCREENING ASSAY FOR ASSESSING THE EFFECT OF VARIOUS COMPOUNDS ON IL-
8 PRODUCTION BY MACROPHAGES
This example describes a screening assay for assessing the effect of an agent on IL-8 production by human macrophages. The assay protocol is based on the following references: J. Immunol. (2000) 165: 411- 418; J. Immunol. (2000) 164: 4804-4811; J. Immunol Meth. (2000) 235 (1- 2): 33-40. The human macrophage cell line, THP- 1 was plated in a 12 well plate such that each well contains 1 X 106 cells in 2 ml of media containing 10% FCS. Opsonized zymosan was prepared by resuspending 20 mg of zymosan A in 2 ml of ddHaO and homogenizing until a uniform suspension was obtained. Homogenized zymosan was pelleted at 250 g, resuspended in 4 ml of human serum for a final concentration of 5 mg/ml, and incubated in a 37°C water bath for 20 minutes to enable opsonization. The agent was prepared in DMSO at a concentration of 10'2 M and serially diluted 10-fold to give a range of stock concentrations (10~8 M to 10~2 M).
THP-1 cells were stimulated to produce IL-8 by the addition of 1 mg/ml opsonized zymosan. An agent was added to THP-1 cells by directly adding DMSO stock solutions, prepared earlier, at a 1/1000 dilution, to each well. Each drug concentration was tested in triplicate wells. Plates were incubated at 37°C for 24 hours.
After a 24 hour stimulation, supematants were collected to quantify IL-8 production. IL-8 concentrations in the supematants were determined by ELISA using recombinant human IL-8 to obtain a standard curve. A 96-well MAXISORB plate was coated with 100 μL of anti-human IL-8 Capture Antibody diluted in Coating Buffer (0.1 M sodium carbonate pH 9.5) overnight at 4°C. The dilution of Capture Antibody used was lot-specific and was determined empirically. Capture Antibody was then aspirated and the plate washed 3 times with Wash Buffer (PBS, 0.05% TWEEN-20). Plates were blocked for 1 hour at room temperature with 200 μL/well of Assay Diluent (PBS, 10% FCS pH 7.0). After blocking, plates were washed 3 times with Wash Buffer. Standards and sample dilutions were prepared as follows: (a) sample supematants were diluted Vioo and Vioooi (b) recombinant human IL-8 was prepared at 200 pg/ml and serially diluted to yield as standard curve of 3.1 pg/ml to 200 pg/ml. Sample supematants and standards were assayed in triplicate and were incubated at room temperature for 2 hours after addition to the plate coated with Capture Antibody. The plates were washed 5 times and incubated with 100 μl_ of Working Detector (biotinylated anti-human IL-8 detection antibody + avidin- HRP) for 1 hour at room temperature. Following this incubation, the plates were washed 7 times and 100 μl_ of Substrate Solution (tetramethylbenzidine, H2O2) was added to plates and incubated for 30 minutes at room temperature. Stop Solution (2 N H2SO4) was then added to the wells and a yellow color reaction was read at 450 nm with λ correction at 570 nm. Mean absorbance was determined from triplicate data readings and the mean background was subtracted. IL-8 concentration values were obtained from the standard curve. Inhibitory concentration of 50% (IC5o) was determined by comparing average IL-8 concentration to the positive control (THP-1 cells stimulated with opsonized zymosan). IC50 values were measured for the following compounds: herbimycin A (1431 nM); tacrolimus (188 nM); geldanamycin (19 nM); 17-AAG (209 nM); 17-DMAG (62 nM); mannose-6-phosphate (220 nM); simvastatin (9587 nM); isotretinoin (126 nM); puromycin dihydrochloride from Streptomyces alboniger (936 nM); NPC-15437 dihydrochloride (514 nM); radicicol from Humicola fuscoatra (67 nM). The following compounds showed an IC50 value greater than 10,00OnM or no effect in this assay: ribavarin, pirfenidone, vinorelbine, pioglitazone, ruboxostaurin, and cladribine. Other compounds that may be tested for IC50 values in this assay include: e.g., ZD-6474, AP-23573, synthadotin, S-0885, aplidine, ixabepilone, IDN-5390, SB-2723005, ABT-518, combretastatin, anecortave acetate, SB-715992, temsirolimus, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, juglone, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, dolastatin 15, cerivastatin, jasplakinolide, loteprednol etabonate, prednisolone, 3-BAABE, and 5-azacytidine.
EXAMPLE 27
SCREENING ASSAY FOR ASSESSING THE EFFECT OF VARIOUS COMPOUNDS ON MCP-1 PRODUCTION BY MACROPHAGES
This example describes a screening assay for assessing the effect of an agent on MCP-1 production by human macrophages. The assay protocol is based on the following references: J. Immunol. (2000) 165: 411-418; J, Immunol. (2000) 164: 4804-4811; J. Immunol Meth. (2000) 235 (1-2): 33-40. The human macrophage cell line, THP-1, was plated in a 12 well plate such that each well contains 1 X 106 cells in 2 ml of media containing 10% FCS. Opsonized zymosan was prepared by resuspending 20 mg of zymosan A in 2 ml of ddhbO and homogenizing until a uniform suspension was obtained. Homogenized zymosan was pelleted at 250 g and resuspended in 4 ml of human serum for a final concentration of 5 mg/ml and incubated in a 370C water bath for 20 minutes to enable opsonization. The agent was prepared in DMSO at a concentration of 10~2 M and serially diluted 10-fold to give a range of stock concentrations (10~8 M to 10"2 M).
THP-1 cells were stimulated to produce MCP-1 by the addition of 1 mg/ml opsonized zymosan. An agent was added to THP-1 cells by directly adding DMSO stock solutions, prepared earlier, at a 1/1000 dilution, to each well. Each drug concentration was tested in triplicate wells. Plates were incubated at 37°C for 24 hours. After 24 hour stimulation, supernatants were collected to quantify MCP-1 production. MCP-1 concentrations in the supernatants were determined by ELISA using recombinant human MCP-1 to obtain a standard curve. A 96-well MaxiSorb plate was coated with 100 μl_ of anti-human MCP-1 Capture Antibody diluted in Coating Buffer (0.1M sodium carbonate pH 9.5) overnight at 40C. The dilution of Capture Antibody used was lot- specific and was determined empirically. Capture antibody was then aspirated and the plate washed 3 times with Wash Buffer (PBS1 0.05% TWEEN-20). Plates were blocked for 1 hour at room temperature with 200 μL/well of Assay Diluent (PBS, 10% FCS pH 7.0). After blocking, plates were washed 3 times with Wash Buffer. Standards and sample dilutions were prepared as follows: (a) sample supernatants were diluted V-ioo and 1/iooo; (b) recombinant human MCP-1 was prepared at 500 pg/ml and serially diluted to yield as standard curve of 7.8 pg/ml to 500 pg/ml. Sample supernatants and standards were assayed in triplicate and were incubated at room temperature for 2 hours after addition to the plate coated with Capture Antibody. The plates were washed 5 times and incubated with 100 μl_ of Working Detector (biotinylated anti-human MCP-1 detection antibody + avidin-HRP) for 1 hour at room temperature. Following this incubation, the plates were washed 7 times and 100 μl_ of Substrate Solution (tetramethylbenzidine, H2O2) was added to plates and incubated for 30 minutes at room temperature. Stop Solution (2 N H2SO4)was then added to the wells and a yellow color reaction was read at 450 nm with λ correction at 570 nm. Mean absorbance was determined from triplicate data readings and the mean background was subtracted. MCP-1 concentration values were obtained from the standard curve. Inhibitory concentration of 50% (IC5o) was determined by comparing average MCP-1 concentration to the positive control (THP-1 cells stimulated with opsonized zymosan). IC50 values were measured for the following compounds: herbimycin A (77 nM); geldanamycin (15 nM); 17-AAG (306 nM); 17-DMAG (59 nM); vinorelbine (38 nM); simvastatin (3031 nM); galardin (232 nM); idazoxan hydrochloride (15 nM); thioperamide maleate salt (63 nM); tirapazamine (554 nM); puromycin dihydrochloride from Streptomyces alboniger (161 nM); NPC- 15437 dihydrochloride (402 nM); YM 976 (24 nM); radicicol from Humicola fuscoatra (72 nM); risedronate (17 nM); aucubin (30 nM); homoharringtonine (8 nM); geniposidic acid (0.01 nM); geniposide (<0.01 nM); brefeldin A (33 nM); thapsigargin (4 nM); (-)-arctigenin (24 nM); dolastatin 15 (2 nM). The following compounds showed an IC50 value greater than 10,00OnM or no effect in this assay: tacrolimus, mannose-6-phosphate, pioglitazone, ruboxistaurin, cladribine, ribavarin, and pirfenidone.
Other compounds that may be tested for IC50 values in this assay include: e.g., ZD-6474, AP-23573, synthadotin, S-0885, aplidine, ixabepilone, IDN-5390, SB-2723005, ABT-518, combretastatin, anecortave acetate, SB-715992, temsirolimus, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, juglone, isotretinoin, clobetasol propionate, trichostatin A, cerivastatin, jasplakinolide, loteprednol etabonate, prednisolone, 3-BAABE, 5-azacytidine, and gefitinib.
EXAMPLE 28
SCREENING ASSAY FOR ASSESSING THE EFFECT OF COMPOUNDS ON SMOOTH
CELL PROLIFERATION
This example describes a screening assay for assessing the effect of an agent on smooth muscle cell proliferation (In vitro toxicol. (1990) 3: 219; Biotech. Histochem. (1993) 68: 29; Anal. Biochem. (1993) 213: 426). This assay also may be used assess the effect of compounds on proliferation of fibroblasts and murine macrophage cell line RAW 264.7.
Smooth muscle cells at 70-90% confluency were trypsinized, replated at 600 cells/well in media in 96-well plates and allowed to attach overnight. An agent (e.g., paclitaxel, geldanamycin, or herbimycin A) was prepared in DMSO at a concentration of 10'2 M and diluted 10-fold to give a range of stock concentrations (10"8 M to 10"2 M). Drug dilutions were diluted 1/1000 in media and added to cells to give a total volume of 200 μL/well. Each drug concentration was tested in triplicate wells. Plates containing cells and paclitaxel were incubated at 37°C for 72 hours.
To terminate the assay, the media was removed by gentle aspiration. A 1/400 dilution of CYQUANT 400X GR dye indicator (Molecular Probes; Eugene, OR) was added to 1X Cell Lysis buffer, and 200 μl of the mixture was added to the wells of the plate. Plates were incubated at room temperature, protected from light for 3-5 minutes. Fluorescence was read in a fluorescence microplate reader at ~480 nm excitation wavelength and ~520 nm emission maxima. Inhibitory concentration of 50% (IC5o) was determined by taking the average of triplicate wells and comparing average relative fluorescence units to the DMSO control. An average of n=3 replicate experiments was used to determine IC5O values. IC50 values were measured for the following compounds: herbimycin A (237 nM); geldanamycin (7.5 nM); 17-AAG (69 nM); 17-DMAG (17 nM); vinorelbine (15 nM); simvastatin (996 nM), ruboxistaurin (1825 nM), cladribine (137 nM); PD 98059 (<0.01 nM); actinomycin D (0.3 nM); juglone (902 nM); prednisolone 21 -acetate (320 nM); puromycin dihydrochloride from Streptomyces alboniger (441 nM); dacarbazine (<0.01 nM); temozolomide (<0.01 nM); radicicol from Humicola fuscoatra (34 nM); (-)-epicatechin gallate from green tea (428 nM); 5-azacytidine (542 nM); homoharringtonine (9 nM); trichostatin A from Streptomyces sp. (16 nM); NS-398 (51 nM); betulinic acid (<0.01 nM); thapsigargin (5 nM); ammonium pyrrolidinedithiocarbamate (294 nM); dolastatin 15 (0.9 nM); cervistatin Na (12 nM); roxatidine acetate (<0.01 nM); benzoyl peroxide (<0.01 nM); 3- BAABE (403 nM); jasplakinolide (3 nM). The following compounds showed an IC50 value greater than 10,00OnM or no effect in this assay: pioglitazone, mannose-6-phosphate, tacrolimus, ribavarin, and pirfenidone.
Other compounds that may be tested for IC50 values in this assay include: e.g., ZD-6474, AP-23573, synthadotin, S-0885, aplidine, ixabepilone, IDN-5390, SB-2723005, ABT-518, combretastatin, anecortave acetate, SB-715992, temsirolimus, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, isotretinoin, clobetasol propionate, brefeldin A, and loteprednol etabonate.
EXAMPLE 28A PREPARATION OF DRUG-LOADED POLYURETHANE (PU) FILMS
2 g CHRONOFLEX - AL 85A (CT Biomaterials, Woburn, Mass.) is added to a 20 ml glass scintillation vial. 10 ml THF is added together with a magnetic stirrer bar and the vial is capped with the screw top lid. The mixture is stirred using a magnetic stirrer at a temperature of 500C until the polymer is dissolved. The solution is allowed to cool and the desired mass of the drug (10mg, 25 mg, 50 mg, 100 mg, 200 mg, 300 mg, 400 mg, or 500 mg ) is added to the solution. The solution is stirred to ensure proper mixing of the drug. Depending on the drug solubility in the THF, the drug may be dissolved in a suitable organic solvent prior to addition to the polymer solution. Solvents that can be used include DCM, acetone, dimethylacetamide and ethanol. Depending on the drug, these mixtures may be solutions or may have the some or all of the drug in suspension. The mixture is then poured onto a sheet of release liner (rexam a10, grade 10393, silicone coated PET) and a casting knife ( 40 mil) is used to draw the mixture into a film. The film is covered with a sheet of glass (setup in such a manner that the glass is about 10mm from the wet film, i.e., not touching the wet film) and is allowed to air dry for 10 min. The glass sheet is removed and the film is dair dried in a hood for a further 60 min. The film is then placed in a force air oven (500C) for 2 hours afer which the film is placed in a vacuum oven for 24-48 hours. Once dired, the film is covered with another sheet of the release liner. This release liner laminated film is then cut into squared of approx. 8 mm x 8 mm. The films are then placed in a plastic pouch which is heat sealed closed. The films are then sterilized using either gamma sterilization or e-beam sterilization. Films containing the following drugs may be made using the described process: ZD-6474, AP-23573, synthadotin, S-0885, aplidine, ixabepilone, IDN-5390, tacrolimus, juglone, SB-2723005, ABT-518, combretastatin, anecortave acetate, SB-715992, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, cerivastatin, jasplakinolide, herbimycin A1 pirfenidone, vinorelbine, 17-DMAG, temsirolimus, loteprednol etabonate, prednisolone, puromycin, 3-BAABE, cladribine, mannose-6-phosphate, 5-azacytidine, Ly333531 (ruboxistaurin), dolastatin 15, and simvastatin.
EXAMPLE 29
PERIVASCULAR ADMINISTRATION OF THERAPEUTIC COMPOUNDS TO ASSESS
INHIBITION OF FIBROSIS
WISTAR rats weighing 250 - 300 g are anesthetized by the intramuscular injection of Innovar (0.33 ml/kg). Once sedated, they are then placed under Halothane anesthesia. After general anesthesia is established, fur over the neck region is shaved, the skin clamped and swabbed with betadine. A vertical incision is made over the left carotid artery and the external carotid artery exposed. Two ligatures are placed around the external carotid artery and a transverse arteriotomy is made. A number 2 French Fogarty balloon catheter is then introduced into the carotid artery and passed into the left common carotid artery and the balloon is inflated with saline. The catheter is passed up and down the carotid artery three times. The catheter is then removed and the ligature is tied off on the left external carotid artery.
A drug-loaded PU film (Example 28A) is then placed in a circumferential fashion around the common carotid artery). Five rats from each group are sacrificed at 14 days and the final five at 28 days. The rats are observed for weight loss or other signs of systemic illness. After 14 or 28 days the animals are anesthetized and the left carotid artery is exposed in the manner of the initial experiment. The carotid artery is isolated, fixed at 10% buffered formaldehyde and examined for histology. A statistically significant reduction in the degree of initimal hyperplasia, as measured by standard morphometric analysis, indicates a drug induced reduction in fibrotic response. Other compounds that may be tested in this model include: e.g., ZD-6474, AP-23573, synthadotin, S-0885, aplidine, ixabepilone, IDN-5390, SB-2723005, ABT-518, combretastatin, anecortave acetate, SB-715992, temsirolimus, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, herbimycin A, geldanamycin, 17-AAG, 17-DMAG, cladribine, simvastatin, pirfenidone, mannose-6- phosphate, tacrolimus, vinorelbine, dolastatin 15, and ruboxistaurin.
EXAMPLE 30 MIC DETERMINATION BY MICROTITRE BROTH DILUTION METHOD
A. MIC assay of various gram negative and positive bacteria
MIC assays were conducted essentially as described by Amsterdam, Dr 1996, "Susceptibility testing of antimicrobials in liquid media", p.52-111, in Loman, V., ed. Antibiotics in laboratory medicine, 4th ed. Williams and Wilkins, Baltimore, MD. Briefly, a variety of compounds were tested for antibacterial activity against isolates of P. aeruginosa, K, pneumoniae, E. coli, S. epidermidus and S. aureus in the MIC (minimum inhibitory concentration assay under aerobic conditions using 96 well polystyrene microtitre plates (Falcon 1177), and Mueller Hinton broth at 37°C incubated for 24h. (MHB was used for most testing except C721 (S. pyogenes), which used Todd Hewitt broth, and Haemophilus influenzae, which used Haemophilus test medium (HTM)) Tests were conducted in triplicate. The results are provided below in Table 3. TABLE 3: MINIMUM INHIBITORY CONCENTRATIONS OF THERAPEUTIC AGENTS AGAINST VARIOUS GRAM NEGATIVE AND POSITIVE BACTERIA
Figure imgf004594_0001
Activities are in Molar concentrations Wt = wild type N = No activity
D. MIC of antibiotic-resistant bacteria
Various concentrations of the following compounds, mitoxantrone, cisplatin, tubercidin, methotrexate, 5-fluorouracil, etoposide, 2-mercaptopurine, doxorubicin, 6-mercaptopurine, camptothecin, hydroxyurea and cytarabine were tested for antibacterial activity against clinical isolates of a methicillin resistant S. aureus and a vancomycin resistant pediocoocus clinical isolate in an MIC assay as described above. Compounds which showed inhibition of growth (MIC value of <1.0x 10-3) included: mitoxantrone (both strains), methotrexate (vancomycin resistant pediococcus), 5-fluorouracil (both strains), etoposide (both strains), and 2- mercaptopurine (vancomycin resistant pediococcus). EXAMPLE 31 PREPARATION OF RELEASE BUFFER
The release buffer is prepared by adding 8.22 g sodium chloride, 0.32 g sodium phosphate monobasic (monohydrate) and 2.60 g sodium phosphate dibasic (anhydrous) to a beaker. 1L HPLC grade water is added and the solution is stirred until all the salts are dissolved. If required, the pH of the solution is adjusted to pH 7.4 ± 0.2 using either 0.1 N NaOH or 0.1 N phosphoric acid.
EXAMPLE 32
RELEASE STUDY TO DETERMINE RELEASE PROFILE OF A THERAPEUTIC AGENT FROM A POLYMERIC COMPOSITION
The release profile of a therapeutic agent from a polymeric composition can be determined according to the following procedure.
Release and Extraction
A sample is placed in a 16 x 125 mm screw capped culture tube. 16 ml release buffer (Example 31) is added to the tube. The samples are placed on a rotating wheel (30 rpm) in a 370C oven. At the various time intervals (2h, 5h, 8h, 24h and then daily), the sample tubes are taken from the oven, placed in a rack and the caps are removed in a fume hood. As much of the release buffer as possible is removed from the tube and placed in a second culture tube. 16 ml of release media is then added to the sample containing tube using an Oxford pipettor bottle. The samples are capped with a new PTFE lined cap. All samples are returned to the rotating wheel device in the oven.
Using a p1000 pipettor (PIPETMAN) and a clean pipette tip, remove and discard 1 ml of release media from each sample. Add 1 ml of dichloromethane to each sample using an oxford pipettor bottle. Cap each sample tube with the respective PTFE lined screw cap. Hand shake each sample vigorously for 5 seconds. Place samples on the labquake rotator and rotate for 15 min. Centrifuge samples at 1500 rpm for 10 minutes. Transfer the sample tubes to a fume hood and uncap. Remove most of the supernatant (aqueous phase) using a Pasteur pipette and vacuum system. Remove the final portion of the supernatant with a glass syringe. Transfer sample tubes to the pierce drying system, set the heating block to 1.5 (45°c) and turn on the system. Dry all samples on the pierce drying system under a stream of nitrogen gas (approximately 45 min.). Re-cap the sample tubes, place in a plastic bag, label bag with date and time of sample, and store at - 20°c (freezer) until analysis.
External Standard Preparation
100 mg of the drug to be analysed is accurately weighed, quantitatively transferred and made up to volume with ACN in a 100 ml volumetric flask (1 mg/ml). 5 ml of this standard solution is transferred, using a volumetric pipette, to a 100 ml volumetric flask and is made up to volume with ACN (50 μg/ml). Serial dilutions (5 ml qs ad 10 ml with ACN) are used to prepare 25, 12.5, 6.25, 3.13, 1.56, 0.781 and 0.391 μg/ml solutions respectively. On the day of HPLC analysis of samples, an aliquot (~100 μl) of each standard is placed into separate autosampler vials using small volume inserts and is transferred to the HPLC.
Sample Reconstitution
Remove samples to be analyzed from the freezer, place in a fume hood, and allow tubes to come to room temperature. Uncap and add 1 ml of water/acetonitrile (50/50) to each tube with an Oxford pipettor. Recap sample tubes and vortex for 60 s. Centrifuge sample tubes at 1500 rpm for 15 min. In a fume hood, transfer approximately 500 μl of each sample to a separate HPLC autosampler vial with a clean Pasteur pipette. Cap each autosampler vial and transfer to the HPLC. Dispose of the sample tube and Pasteur pipette. The samples are then analysed for drug content using HPLC.
EXAMPLE 33 FORMULATION OF A DRUG IN A VEHICLE COMPRISING A TRIBLOCK COPOLYMER
Juglone is incorporated into a formulation comprising a triblock copolymer and a diluent (described below) by dissolving the juglone in the diluent with stirring at ambient temperature for at least two hours, then adding the triblock copolymer, again with stirring for at least 2 hours. Longer periods of time are used to add triblock copolymer at higher concentrations. For example, the addition of 33% triblock copolymer is accomplished by stirring for at least 15 hours (overnight). The diluent is PEG 300 NF or PEG 400 derivatized by end addition of trimethylene carbonate 90%/glycolide 10% in a ratio of 400:100. The triblock copolymer is an ABA copolymer with blocks A containing polymerized trimethylene carbonate (90%) and glycolide (10%), having a total molecular weight of about 900 g/mol and the B block containing PEG 400. Juglone is effectively incorporated into this formulation at a concentration of 0.015 to 0.45 mg/ml. The amount of triblock copolymer in the formulation is varied from 2.3 to 50%w/w using PEG 400 as the diluent.
EXAMPLE 34 FORMULATION OF A DRUG IN A CO-SOLVENT VEHICLE
Juglone is incorporated into a formulation comprising water and PEG 300 NF. The juglone is first dissolved in a 90:10 mixture of PEG 300 NF.water by stirring at ambient temperature for at least two hours. Once the drug was dissolved, the composition is combined with equal parts of a 50:50 mixture of PEG 300 NF:water. The final composition is juglone dissolved in a mixture of 70:30 PEG 300 NF.water. Juglone is incorporated at concentrations of 0.45 to 4.5 mg/ml. The composition is passed through a 0.22 μm filter to render it sterile.
EXAMPLE 35
DETERMINATION THE MAXIMUM TOLERATED DOSE (MTD) OF A DRUG AFTER
INTRA-ARTICULAR INJECTION
Male Hartley guinea pigs, at least 6 weeks old, were anaesthetized using 5% isoflurane in an enclosed chamber. The animals were weighed and then transferred to the surgical table where anesthesia was maintained by nose cone with 2% isoflurane. The knee area on both legs was shaved and knee width at the head of the femur was measured on both knees. The skin on the right knee was sterilized. A 25G needle was introduced into the synovial cavity using a medial approach and 0.1 mL of the test formulation was injected. Three or seven days after the injection, the animals were sacrificed by cardiac injection of 0.7 mL Euthanyl under deep anesthesia (5% isoflurane). Sample size was N=3 for each formulation.
Knee function was assessed before sacrifice by recording changes in walking behavior and signs of tenderness. The animal was weighed immediately after sacrifice. The width of both knees at the head of the femur was then measured with calipers. The knee joint was dissected open by transecting the quadriceps tendon, cutting through the lateral and medial articular capsule and flipping the patella over the tibia. Knee inflammation was assessed by recording signs of swelling, vascularization, fluid accumulation and change in color in subcutaneous tissue as well as inner joint structures. Photographs were taken to document findings. All data was recorded by observers blinded to the treatment groups.
The MTD of the drug in the test formulation was determined to be that for which knee inflammation was not observed.
The MTD of paclitaxel in the triblock gel formulation prepared as in Example 34 was 0.075 mg/ml, based upon evaluation at 7 days. Evaluation of this formulation after three days showed that doses up to 0.15 mg/ml were tolerated. The 0.015 mg/ml dose showed signs of inflammation only after seven days. The MTD of paclitaxel in the co-solvent formulation was found to be 1.5 mg/ml, based upon a 3 day evaluation.
Other compounds that may be tested in this model include: ZD-6474, AP-23573, synthadotin, S-0885, aplidine, ixabepilone, IDN-5390, SB-2723005, ABT-518, combretastatin, anecortave acetate, SB-715992, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17-DMAG, tacrolimus, temsirolimus, loteprednol etabonate, prednisolone, puromycin, 3-BAABE, cladribine, mannosemannose-6-phosphate, 5-azacytidine, Ly333531 (ruboxistaurin), dolastatin 15, and simvastatin.
EXAMPLE 36
EVALUATION OF LOCAL TISSUE DISTRIBUTION OF A DRUG AFTER INTRAARTICULAR INJECTION
Animals are injected in the knee joint as described above in Example 35 with the drug MTD dose identified for each formulation. Three or seven days after injection the animals are euthanized with an intracardiac injection of Euthanyl. The knee joint is dissected open and the synovial membrane, the anterior cruciate ligament, the fat pad, the menisci and the cartilage are harvested. Each tissue is briefly rinsed in saline solution, blotted dry and stored individually in a scintillation vial at -20°C until drug analysis.
The drug is extracted from a weighed pooled sample from three animals by homogenization using a Polytron PT2000 homogenizer. The instrument setting is 3 to 9 and the extraction time is 1 minute. The extraction solution is 1 mL of 50/50 acetonitrile (ACN)/water containing 0.2 μg/mL 10-deacetyl taxol (10-DAT) and 0.1% formic acid. The extract is centrifuged using a Beckman J6-HC centrifuge for 10 minutes at 3000 rpm. The supernatant is filtered through an Acrodisc CR (13 mm, 0.45 μ) syringe filter into an HPLC vial for LC/MS/MS analysis. Some fat pad samples that did not produce a clear supernatant arecentrifuged again prior to filtration using an IEC Micromax centrifuge for 10 minutes at 10000 rpm.
The drug content in the extract is determined by an LC/MS/MS method using an internal calibration. For example, a calibration curve may range from 0.01 to 1 μg/mL for drug with 0.2 μg/mL 10-DAT. The LC/MS/MS system consists of a Waters 2695 separation module and a Waters Micromass QuattoMicro triple-Quad mass spectrometer
EXAMPLE 37
EVALUATION OF LOCAL TISSUE DISTRIBUTION OF A DRUG AFTER INTRAARTICULAR INJECTION
Animals are treated in the manner described in Example 35. Rabbits are evaluated by intra-articular injection of 0.5 ml of formulation. Drug is extracted from individual tissue sample from three animals by homogenization using a Freezer/Mill, SPEX CertiPrep 6850. The ground sample is extracted with 12 mL solution containing acetic acid (3.4 mM) and LiCI (4 to 8 μM) in 50/50 ACN/water. Extraction is performed on a tube rotator (Labquake Shaker) for 30 minutes at room temperature. The extract is filtered through an Acrodisc CR (13 mm, 0.45 μ) syringe filter into an HPLC vial for LC/MS/MS analysis.
The drug content in the extract is determined by an LC/MS/MS method using an external calibration. The calibration curve ranges from 0.01 to 1 μg/mL for paclitaxel. The LC/MS/MS system consists of a Waters 2695 separation module and a Waters Micromass QUATTOMICRO triple- Quad mass spectrometer. EXAMPLE 38
ASSESSMENT OF PACLITAXEL IN THE INHIBITION OF CARTILAGE DAMAGE IN THE ACL INJURED HARTLEY GUINEA PIG MODEL OF OSTEOARTHRITIS
The purpose of this study was to determine whether a fibrosis- inhibiting agent (e.g., paclitaxel) administered in a hyaluronic acid formulation can delay or prevent the development of osteoarthritis in guinea pig knees.
Surgical procedures.
Male Hartley guinea pigs, at least 6 weeks old, were anaesthetised using 5% isoflurane in an enclosed chamber. The animals were weighed and then transferred to the surgical table where anaesthesia was maintained by nose cone with 2% isoflurane. The knee area on the both legs was shaved and knee width at the head of the femur was measured on both knees. The skin on the right knee was sterilized. A 2OG needle was introduced in the knee joint using a medial approach and the anterior cruciate ligament was cut with the sharp end of the needle. This procedure was practiced in a preliminary experiment that showed that the anterior cruciate ligament could be sectioned reliably using this technique.
Two weeks after the initial procedure, the animals were anesthetized with isoflurane (5% induction - 2% maintenance) and weighed. The knee area on both legs was shaved and knee width at the head of the femur was measured on both knees. The skin of the injured knee was sterilised. A 25G needle was introduced into the synovial cavity using a medial approach and 0.1ml of the test formulation was injected. Injections were repeated weekly for a total of 5 injections. Sample size was N=12 for each formulation. Two doses of paclitaxel and control formulation were tested.
Ten weeks after injury, the animals were sacrificed by cardiac injection of 0.7 ml Euthanyl under deep anaesthesia (5% isoflurane) and weighed. A final knee measurement was taken. The skin over the knee area was removed without damaging subcutaneous tissues. The knee joints were then harvested en bloc and placed into a formaldehyde (37%)/ acetic acid solution (5:1 ratio) for fixation. Samples were sent to an independent laboratory for the conduct of histological preparation of joints and assessment by a pathologist for signs of cartilage damage.
Briefly knee sections were made to examine cartilage and slides were stained with H&E stain. A pathologist scored slides in a blinded fashion from each animal using corresponding knee sections according to the following scale: no damage to cartilage, loss of proteoglycans, fraying of cartilage, loss of cartilage to the tidemark, and loss of cartilage to the bone. Bar graphs were constructed from each group and compared. Paclitaxel treatment at a low dose (dose 1) and medium dose (dose 2) showed a statistical reduction in cartilage damage relative to control. See Figures 7 and 8A-C.
Exemplary compounds that may be tested in this model include: juglone, ZD-6474, AP-23573, synthadotin, S-0885, aplidine, ixabepilone, IDN-5390, SB-2723005, ABT-518, combretastatin, anecortave acetate, SB-715992, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17-DMAG, tacrolimus, temsirolimus, loteprednol etabonate, prednisolone, puromycin, 3-BAABE, cladribine, mannosemannose-6-phosphate, 5-azacytidine, Ly333531 (ruboxistaurin), dolastatin 15, and simvastatin.
EXAMPLE 39
PROTEOGLYCAN LOSS INDEX IN THE CARRAGEENIN-INDUCED AND ANTIGEN- INDUCED RABBIT MODELS OF ARTHRITIS FOLLOWING TREATMENT WITH
PACLITAXEL
Microspheres were made using the oil in water solvent evaporation method described by Liggins and Burt (2001). The external phase was 100 ml of 1-5% PVA in water. The internal phase was 10 ml of a dichloromethane solution containing 5% w/v total solids (polymer and drug). The dispersion was stirred for 2 hours at room temperature to form microspheres. By varying the stirring speed between 900 and 2100 rpm and the PVA concentration, various size ranges were produced. The microspheres were separated from the external phase and rinsed with distilled water. Some microspheres were further divided into discrete size ranges by sieving the microspheres suspension through sieves having mesh sizes of 38, 53, 75 and 106 μm. Microsphere size distributions were determined using a Coulter LS130 particle size analyzer. Microspheres were suspended in water with a small amount of Tween 80 to prevent aggregation prior to particles size analysis. Chitosan microparticle size ranges were determined by optical microscopy using a microscope slide marked with 5 μm gradations. Optical microscopy was performed on both dry and wetted samples.
Thermal properties of the microspheres were determined using a Dupont Thermal Analysis DSC. Approximately 5 mg of microspheres were placed in unsealed aluminum pans and thermograms were obtained at a heating rate of 10°C/min. Evidence of crystallinity was obtained by X-ray powder diffraction measurements using a Rigaku X-ray diffractometer. Samples were scanned with a CuKa X-ray source through 5- 35°2Θ at a rate of 1°2θ/min with a step increment of 0.02°2θ.
The surface morphology of microspheres was determined using a Hitachi scanning electron microscope. Microspheres were coated with a 100 A gold-palladium coat and visualized at a magnification of 1000x.
The drug content and in vitro release from microspheres were determined using the methods similar to that of Liggins & Burt (2001). For total content analysis, approximately 5 mg (accurately weighed) of microspheres were dissolved in 1 ml of dichloromethane followed by vigorous mixing with 15 ml of 60:40 acetonitrile:water. The solvent mixture was allowed to separate into two approximately equal volumes with a precipitated mass of polymer between the two. The amount of paclitaxel in each of the two fractions was then determined by HPLC using a Waters HPLC system.
Antigen induced arthritis was reproduced in rabbits using a previously described method (Kim et al., J. Rheumatol 1995:22:1714-21.). Briefly, female New Zealand white rabbits weighing 2.5 - 2.8 kg were used in biocompatibility and efficacy studies. Animals were housed in suspended caging with free access to food and water. Animals were acclimated for seven days prior to all experiments. Arthritis was induced in some animals for use as positive controls in biocompatibility testing and for use in efficacy studies. All knee joint injections were carried out under anaethesia induced by intramuscular injection of ketamine HCI (40 mg/kg) and xylazine (5 mg/kg). At the end of the in-life portion of the study, animals were sacrificed using intravenous T-61. The knee joints were dissected immediately after sacrifice and fixed in 10% formalin prior to histological analysis.
Antigen induced arthritis was established by three injections of bovine serum albumin (BSA) in Freund's complete adjuvant (FCA). The first injection consisted of 5 mg BSA emulsified in 1 ml FCA and diluted in 1 ml PBS. Three weeks later, each rabbit received a subcutaneous booster injection of 2.5 mg of BSA emulsified in 1 ml FCA diluted with 1ml PBS. After four weeks, each rabbit received a second booster of 0.5 mg BSA in 0.3 mL pyrogen-free PBS injected into the knee joint. Five days after the final booster, the rabbits were treated by intra-articular injection with test articles.
Carrageenan induced arthritis was established in rabbits and the rabbits were treated in the same manner as for the antigen induced arthritis model. All rabbits in the carrageenan groups were injected with 0.3 ml of 1% carrageenan in pyrogen free PBS on days 1, 3, 8, 16 and 21. Half the animals were also injected with 35 mg of 20% drug-loaded microspheres on day 6. All animals were sacrificed on day 29 and the joints were dissected for histological analysis. Synovial inflammation was assessed after sacrificing the rabbits. The joints were fixed in formalin and decalcified in 10% formic acid with repeated changes. The decalcified joints were embedded in paraffin and sections containing synovium, cartilage and bone were prepared. Sections were stained for cellularity with hematoxylin and eosin (H&E) and for proteoglycan content with safranin O. Synovial inflammation and cartilage degradation were evaluated by blinded histological evaluation of parapatellar synovium and femoral condylar articular cartilage, respectively. Villus hyperplasia, fibroblast proliferation, fibrosis, angiogenesis, mononuclear cell and polymorphonuclear cell infiltrations were graded as indicators of synovial inflammation. For cartilage degradation, surface erosion, proteoglycan content and chondrocyte necrosis were graded. Grading of cellular infiltration and swelling was scored with an integer from 0 to 4 based on increasing erythema, swelling and cellular infiltration (0, normal; 4, maximum). For slight effects, a score of 0.5 was assigned; this was the only non-integer score used. Proteoglycan loss was also scored from 0 (normal) to 4 (almost total loss of stained proteoglycans).
The efficacy of drug-loaded polyester microspheres given by intra-articular injection in treating antigen-induced arthritis was assessed using control and 20% loaded 10-35 and 35-105 μm PLA microspheres. Groups of five rabbits were treated with 40 mg of microspheres or PBS alone in the right joint. The left joint received PBS alone. The animals were sacrificed fourteen days after treatment and examined histologically for synovial inflammation and cartilage degradation as described above.
PLA microspheres containing 20% paclitaxel were selected for the efficacy study. Table 4 shows the results of the injection of 40 mg of control and paclitaxel-loaded PLA microspheres in rabbits with antigen induced arthritis. Untreated arthritic rabbits had a joint swelling score of 3 and 4.9 x 107 cells in the joint fluid. Paclitaxel-loaded microspheres in the 10-35 um size range did not reduce antigen induced arthritis. In fact, the amount of cellular infiltration was elevated in this group relative to untreated arthritic rabbits (Table 4). However, the injection of 35-105 μm paclitaxel- loaded microspheres significantly reduced both the joint swelling and the number of cells in the joint fluid (about a 50% decrease) relative to control (Table 4). Cartilage degradation expressed as proteoglycan loss and chondrocyte necrosis was also assessed in the control groups and the paclitaxel-loaded 35-105 μm microspheres group. There was no effect on either proteoglycan loss or chondrocyte necrosis by the injection of control PLA microspheres in diseased animals. However, animals treated with paclitaxel-loaded microspheres had significantly less proteoglycan loss than the untreated animals (Table 4 and Figures 9A-9C). Figure 9A illustrates a knee having a normal histological appearance, with a continuous top layer of cartilage and no loss of stain color indicating normal proteoglycan content (score 0). Figure 9B shows a control microspheres arthritic knee with proteoglycan loss down to the bottom third layer of the section, which is termed heavy loss (score 3). In Figure 9C, a paclitaxel microspheres treated arthritic knee shows only slight loss of proteoglycan at the surface layer of cartilage, with an intact surface (score 1).
The effect of paclitaxel-loaded microspheres in preventing proteoglycan loss in carrageenan-induced arthritis was not as prominent as in antigen induced arthritis (Figures 9D-F). Figure 9E shows severe loss of proteoglycan throughout all layers of cartilage, but the surface layer remained intact (score 4). Treatment of carrageenan-induced knees with paclitaxel microspheres resulted in less reduction of stain color (Figure 9F, score 2), but the protective effect was not as pronounced as observed in the antigen induced model (Figure 9C).
Antigen induced arthritis was used to determine efficacy in these studies. Although this animal model takes some time to develop, it mirrors many aspects of human rheumatoid arthritis such as the production of inflammatory cytokines (such as TNF-α), the loss of proteoglycans and the infiltration of white blood cells into the joint with chronic inflammation. Results from this model are compared to those from carrageenan-induced arthritis which is quick to establish in the rabbits and offers a method of inducing intense and reproducible levels of acute (rather than chronic) forms of arthritis. Because carrageenan-induced arthritis is characterized by severe proteoglycan loss, this model was also used in this study to measure the effect of intraarticular paclitaxel on proteoglycan loss. Efficacy studies that included measurements of joint swelling, cell infiltration, proteoglycan loss and chondrocyte necrosis demonstrated that the single injection of 40 mg of 20% paclitaxel-loaded, 35-105 μm microspheres significantly reduced all aspects of the chronic arthritic condition in rabbits (Table 4 and Figures 9A-C). The effect of paclitaxel-loaded microspheres in preventing proteoglycan loss in the carrageenan induced arthritis model was not as pronounced as for the antigen induced arthritis model. Other compounds that may be tested in this model include: e.g., ZD-6474, AP-23573, synthadotin, S-0885, aplidine, ixabepilone, IDN-5390, SB-2723005, ABT- 518, combretastatin, anecortave acetate, SB-715992, temsirolimus, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, herbimycin A, geldanamycin, 17-AAG, 17-DMAG, cladribine, simvastatin, pirfenidone, mannose-6-phosphate, tacrolimus, vinorelbine, dolastatin 15, and ruboxistaurin.
TABLE 4. EFFICACY OF 40 MG OF CONTROL AND 20% PACLITAXEL-LOADED PLA MICROSPHERES IN THE SIZE RANGES OF 10-35 AND 35-105 MM, ASSESSED IN TERMS OF MEAN SCORES (N=5) FOR SWELLING, CELLULAR INFILTRATION, LOSS OF PROTEOGLYCAN AND CHONDROCYTE NECROSIS
Treatment Swelling Number of Proteoglyca Chondrocyte score (0-4) cell in joint n loss (0-4) necrosis (0- fluid 3) healthy, untreated
0 7.0 x 105 Not tested Not tested control
35-105 μm, control 3 4.9 x 107 2 ± 0.6 1 ± 0.3
10-35 μm, 20% paclitaxel 3 8.4 x 107 Not tested Not tested
35-105 μm, 20% paclitaxel 1 2.4 x 107 1 ± 0.3 0 ± 0.1
EXAMPLE 40 DRUG-LOADED MICROSPHERES BY AN EMULSION PROCESS
100 ml of freshly prepared 10% polyvinyl alcohol (PVA) solution is added into a 600 ml beaker. The PVA solution is stirred at 2000 rpm for 30 minutes. Meanwhile, 80 mg cerivastatin is added to a solution of 800 mg plga (50/50, iv= 0.35, Durect Corporation Pelham, AL) in 20 ml dichloromethane. This mixture is stirred for 20 min. The resultant mixture is added dropwise to the PVA solution while stirring at 2000 rpm with a fisher dyna-mix stirrer. After addition is complete, the solution is allowed to stir for an additional 45 minutes. The microsphere solution is transferred to several disposable graduated polypropylene conical centrifuge tubes, washed with deionized water, and centrifuged at 2600 rpm for 10 minutes. The aqueous layer is decanted and the washing, centrifuging and decanting is repeated 3 times. The combined, washed microspheres are freeze-dried and vacuum dried to remove any excess water. This process for preparing drug- containing microspheres may be used with ZD-6474, AP-23573, synthadotin, S-0885, aplidine, ixabepilone, IDN-5390, SB-2723005, ABT- 518, combretastatin, anecortave acetate, SB-715992, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin a, brefeldin a, thapsigargin, jasplakinolide, pirfenidone, loteprednol etabonate, juglone, prednisolone, 3-BAABE, puromycin, tacrolimus, cladribine, mannose-6-phosphate, 5-azacytidine, dolastatin 15, and simvastatin
Example 41 CERIVASTATIN-LOADED GEL
A mass of microspheres (as prepared in Example 40) equivalent to 10 mg cerivastatin is added to a 3 mL plastic syringe which has been capped. 2 mL hyaluronic acid gel (SYNVISC, Genzyme, Cambridge, MA) is added to the 3 mL syringe. The contents of the drug/hyaluronic acid syringe is mixed through a mixing connector connected to a second syringe by repeatedly transferring the contents from one syringe to the other. After complete mixing, the entire mixture is pushed into one of the syringes. The syringe is removed from the mixing connector. The drug- loaded gel can be applied to the electrical device (e.g. neurostimulator) immediately prior to implantation or following implantation of the electrical device (e.g. neurostimulator) the drug-loaded gel can be injected around the device. This process for preparing drug-containing gel may be repeated using ZD-6474, AP-23573, synthadotin, S-0885, aplidine, ixabepilone, IDN- 5390, SB-2723005, ABT-518, combretastatin, anecortave acetate, SB- 715992, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, jasplakinolide, pirfenidone, loteprednol etabonate, juglone, prednisolone, 3-BAABE, cladribine, tacrolimus, puromycin, mannose-6-phosphate, 5-azacytidine, dolastatin 15, and simvastatin.
EXAMPLE 42
SPINAL SURGICAL ADHESIONS MODEL TO ASSESS FIBROSIS INHIBITING AGENTS IN
RABBITS
Extensive scar formation and adhesions often occur after lumbar spine surgery involving the vertebrae. The dense and thick fibrous tissue adherent to the spine and adjacent muscles must be removed by surgery. Unfortunately, fibrous adhesions usually reform after the secondary surgery. Adhesions are formed by proliferation and migration of fibroblasts from the surrounding tissue at the site of surgery. These cells are responsible for the healing response after tissue injury. Once they have migrated to the wound they lay down proteins such as collagen to repair the injured tissue. Overproliferation and secretion by these cells induce local obstruction, compression and contraction of the surrounding tissues with accompanying side effects.
The rabbit laminectomy spinal adhesion model described herein is used to investigate spinal adhesion prevention by local slow release of antifibrotic drugs.
Five to six animals are included in each experimental group to allow for meaningful statistical analysis. Formulations with various concentrations of antifibrotic drugs (as prepared in Example 41) are tested against control animals to assess inhibition of adhesion formation.
Rabbits are anesthetized with an IM injection of ketamine/zylazine. An endotracheal tube is inserted for maintenance of anesthesia with halothane. The animal is placed prone on the operating table on top of a heating pad and the skin over the lower half of the back is shaved and prepared for sterile surgery. A longitudinal midline skin incision is made from L-1 to L-5 and down the lumbosacral fascia. The fascia is incised to expose the tips of the spinous processes. The paraspinous muscles are dissected and retracted from the spinous process and lamina of L-4. A laminectomy is performed at L-4 by removal of the spinal process with careful bilateral excision of the laminae, thus creating a small 5x1 Omm laminectomy defect. Hemostasis is obtained with Gelfoam. The test formulations are applied to the injury site and the wound is closed in layers with Vicryl sutures. The animals are placed in an incubator until recovery from anesthesia and then returned to their cage.
Two weeks after surgery, the animals are anesthetized using procedures similar to those described above. The animals are euthanized with Euthanyl. After a skin incision, the laminectomy site is analyzed by dissection and the amount of adhesion is scored using scoring systems published in the scientific literature for this type of injury.
Exemplary compounds that may be tested in this model include: e.g., juglone, ZD-6474, AP-23573, synthadotin, S-0885, aplidine, ixabepilone, IDN-5390, SB-2723005, ABT-518, combretastatin, anecortave acetate, SB-715992, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17-DMAG, tacrolimus, temsirolimus, loteprednol etabonate, prednisolone, puromycin, 3-BAABE, cladribine, mannosemannose-6-phosphate, 5-azacytidine, Ly333531 (ruboxistaurin), dolastatin 15, and simvastatin.
EXAMPLE 43
TENDON SURGICAL ADHESIONS MODEL TO ASSESS FIBROSIS INHIBITING AGENTS
IN RABBITS
This model is used to investigate whether adhesion of the tendons can be prevented by local slow release of drugs known to inhibit fibrosis. Formulations are loaded with drugs (as described in 41B) and implanted around injured tendons in rabbits. In animals without fibrosis- inhibiting formulations, adhesions develop within 3 weeks of flexor tendon injury if immobilization of the tendon is maintained during that period. An advantage of rabbits is that their tendon anatomy and cellular behaviour during tendon healing are similar to those in man except for the rate of healing that is much faster in rabbits.
Rabbits are anesthetized and the skin over the right hindlimb is shaved and prepared for sterile surgery. Sterile surgery is performed aided by an operating microscope. A longitudinal midline skin incision is made on the volvar aspect of the proximal phalange in digits 2 and 4. The synovial sheath of the tendons is carefully exposed and incised transversally to access the flexor digitorum profundus distal to the flexor digitorum superficialis bifurcation. Tendon injury is performed by gently lifting the flexor digitorum profundus with curved forceps and incising transversally through half of its substance. The formulation containing the test drug formulation is applied around the tendons in the sheath of one of the two digits randomly selected. The other digit is left untreated and is used as a control. The sheath is then repaired with 6-0 nylon suture. An immobilizing 6-0 nylon suture is inserted through the transverse metacarpal ligament into the tendon / sheath complex to immobilize the tendon and the sheath as a single unit to encourage adhesion formation. The wound is closed with 4-0 interrupted sutures. A bandage is applied around the hindpaw to further augment immobilization of the digits and ensure comfort and ambulation of the animals. The animals are recovered and returned to their cage.
Three weeks after surgery, the animals are anesthetized. After a skin incision, the tissue plane around the synovial sheath is dissected and the tendon - sheath complex harvested en block and transferred in 10% phosphate buffered formaldehyde for histopathology analysis. The animals are then euthanized. After paraffin embedding, serial 5-um thin cross- sections are cut every 2 mm through the sheath and tendon complex. Sections are stained with H&E and Movat's stains to evaluate adhesion growth. Each slide is digitized using a computer connected to a digital microscope camera (Nikon Micropublisher cooled camera). Morphometry analysis is then performed using image analysis software (ImagePro). Thickness and area of adhesion defined as the substance obliterating the synovial space are measured and compared between formulation-treated and control animals. Exemplary compounds that may be tested in this model include: e.g., juglone, ZD-6474, AP-23573, synthadotin, S-0885, aplidine, ixabepilone, IDN-5390, SB-2723005, ABT-518, combretastatin, anecortave acetate, SB-715992, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17-DMAG, tacrolimus, temsirolimus, loteprednol etabonate, prednisolone, puromycin, 3-BAABE, cladribine, mannosemannose-6-phosphate, 5-azacytidine, Ly333531(ruboxistaurin), dolastatin 15, and simvastatin..
EXAMPLE 44 PARYLENE COATING
The metallic portion of a coronary stent is washed by dipping it into HPLC grade isopropanol. The cleaned device is then coated with a parylene coating using a parylene coater and either di-p-xylylene or dichloro-di-p-xylylene as the coating feed material. This procedure may be used to coat other types of medical devices that include a metallic portion (e.g., peripheral stents, covered stents, guidewires, shunts, Gl drainage tubes, and anastomotic connectors).
EXAMPLE 45 LOTEPREDNOL ETABONATE COATING - END COATING
loteprednol etabonate solutions are prepared by dissolving paclitaxel in 5 mL HPLC grade THF. The ends of a parylene coated coronary stent (prepared as in Example 44) are then dipped into the paclitaxel/THF solution. After various incubation times, the devices are removed and dried in a forced air oven (500C). The device is then further dried in a vacuum oven overnight. The amount of paclitaxel used in each solution is varied such that the amount of paclitaxel coated onto the ends of the device is in the range of 0.06 mg/mm2 to 10 mg/mm2. In addition to paclitaxel, the following are exemplary compounds that may be used to coat the device: ZD-6474, AP-23573, synthadotin, S-0885, aplidine, ixabepilone, IDN-5390, SB-2723005, ABT-518, combretastatin, anecortave acetate, SB- 715992, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17-DMAG, tacrolimus, temsirolimus, jugalone, prednisolone, puromycin, 3-BAABE, cladribine, mannosemannose-6-phosphate, 5-azacytidine, Ly333531 (ruboxistaurin), dolastatin 15, and simvastatin.
This procedure may be used to coat other types of devices that include a metallic portion (e.g., peripheral stents, covered stents, guidewires, Gl drainage tubes, shunts, and anastomotic connectors).
EXAMPLE 46 JUGLONE COATING - COMPLETE COATING
Juglone solutions are prepared by dissolving paclitaxel in 5 mL HPLC grade THF. A parylene coated coronary stent (as prepared in Example 44) is then dipped entirely into the Juglone/THF solution. After various incubation times, the device is removed and dried in a forced air oven (500C). The device is then further dried in a vacuum oven overnight. The amount of juglone used in each solution is varied such that the amount of paclitaxel coated onto the ends of the device is in the range of 0.06 mg/mm2 to 10 mg/mm2. In addition to Juglone, the following are exemplary compounds that may be also used to coat the device: ZD-6474, AP-23573, synthadotin, S-0885, aplidine, ixabepilone, IDN-5390, SB-2723005, ABT- 518, combretastatin, anecortave acetate, SB-715992, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17-DMAG, tacrolimus, temsirolimus, loteprednol etabonate, prednisolone, puromycin, 3-BAABE, cladribine, mannosemannose-6-phosphate, 5-azacytidine, Ly333531 (ruboxistaurin), dolastatin 15, and simvastatin.
This procedure may be used to coat other types of parylene coated devices that include a metallic portion (e.g., peripheral stents, covered stents, guidewires, Gl drainage tubes, shunts, and anastomotic connectors).
EXAMPLE 47 APPLICATION OF A PARYLENE OVERCOAT
A jugalone coated device is placed in a parylene coater and an additional thin layer of parylene is deposited on the jugalone coated device (see, Example 44The coating duration is altered such that the parylene topcoat thickness is varied such that different elution profiles of the jugalone may be obtained.
EXAMPLE 48 APPLICATION OF AN ECHOGENIC COATING LAYER
DESMODUR (Bayer AG, Germany), an isocyanate pre- polymer, is dissolved in a 50:50 mixture of dimethylsulfoxide and tetrahydrofuran. A juglone /parylene overcoated coronary stent (prepared as in Example 47) is then dipped into the pre-polymer solution. The device is then removed and the coating is then partially dried at room temperature for 3 to 5 minutes. The device is then immersed in a beaker of water (room temperature) for 3-5 minutes to cause the polymerization reaction to occur rapidly. An echogenic coating is formed. This procedure may be used to coat other types of devices (e.g., peripheral stents, covered stents, guidewires, Gl drainage tubes, shunts, and anastomotic connectors).
EXAMPLE 49 LOTEPREDNOL ETABONATE /POLYMER COATING - END COATING
5% solutions of poly(ethylene-co-vinyl acetate) (EVA) (60% vinyl acetate) are prepared using THF as the solvent. Various amounts of loteprednol etabonate are added to each of the EVA solutions. The ends of a corornary stent are dipped into the loteprednol etabonate /EVA solution. After removing the end-coated device from the solution, the coating is dried by placing the device in a forced air oven (400C) for 3 hours. The coated device is then further dried under vacuum for 24 hours. The dip coating process may be repeated to increase the amount of polymer/ loteprednol etabonatecoated onto the device. In addition to loteprednol etabonate, the following are exemplary compounds that may also be used to coat the device: ZD-6474, AP-23573, synthadotin, S-0885, aplidine, ixabepilone, IDN-5390, SB-2723005, ABT-518, combretastatin, anecortave acetate, SB- 715992, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, cerivastatin, jasplakinolide, loteprednol etabonate, herbimycin A, pirfenidone, vinorelbine, 17-DMAG, tacrolimus, temsirolimus, prednisolone, puromycin, 3-BAABE, cladribine, mannosemannose-6-phosphate, 5-azacytidine, Ly333531 (ruboxistaurin), dolastatin 15, and simvastatin.
This procedure may be used to coat other types of devices (e.g., central venous catheters, ventricular assist devices, peripheral stents, and nasal stents). EXAMPLE 50 JUGLONE-HEPARIN COATING - END COATING
5% solutions of poly(ethylene-co-vinyl acetate) (EVA) (60% vinyl acetate) are prepared using THF as the solvent. Various amounts of Juglone and a solution of tridodecyl methyl ammonium chloride-heparin complex (PolySciences) are added to each of the EVA solutions. The ends of an anastomotic connector device are dipped into the Juglone/EVA solution. After removing the end-coated device from the solution, the coating is dried by placing the anastomotic device in a forced air oven (40°C) for 3 hours. The coated anastomotic device is then further dried under vacuum for 24 hours. In addition to juglone, the following are exemplary compounds that may be used to coat the device: ZD-6474, AP- 23573, synthadotin, S-0885, aplidine, ixabepilone, IDN-5390, SB-2723005, ABT-518, combretastatin, anecortave acetate, SB-715992, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17-DMAG, tacrolimus, temsirolimus, loteprednol etabonate, prednisolone, puromycin, 3-BAABE, cladribine, mannosemannose-6-phosphate, 5-azacytidine, Ly333531 (ruboxistaurin), dolastatin 15, and simvastatin.
This procedure may be used to coat other types of devices including peritoneal dialysis catheters, coronary stents, peripheral stents, hemodialysis access devices, guidewires, shunts, and VAD's.
EXAMPLE 51 JUGLONE - HEPARIN/HEPARIN COATING
The uncoated portions of Juglone-heparin coated devices (Example 50) are dipped into a 5% EVA solution containing different amounts of a tridodecyl methyl ammonium chloride-heparin complex solution (PolySciences). After removing the end-coated device from the solution, the coating is dried by placing the anastomotic device in a forced air oven (400C) for 3 hours. The coated device is then further dried under vacuum for 24 hours. This provides a device with a Juglone/heparin coating on the ends of the device and a heparin coating on the remaining parts of the device. In addition to juglone, the following are exemplary compounds that may be used to coat the device: ZD-6474, AP-23573, synthadotin, S- 0885, aplidine, ixabepilone, IDN-5390, SB-2723005, ABT-518, combretastatin, anecortave acetate, SB-715992, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17-DMAG, tacrolimus, temsirolimus, loteprednol etabonate, prednisolone, puromycin, 3-BAABE, cladribine, mannosemannose-6-phosphate, 5-azacytidine, Ly333531 (ruboxistaurin), dolastatin 15, and simvastatin.
This procedure may be used to coated other types of devices including peritoneal dialysis catheters, coronary stents, peripheral stents, hemodialysis access devices, guidewires, shunts, and VAD's
EXAMPLE 52 JUGLONE/POLYMER COATING - END COATING
5% solutions of poly(styrene-co-isobutylene-styrene) (SIBS) are prepared using THF as the solvent. Various amounts of juglone are added to each of the SIBS solutions. The ends of a central venous catheter device are dipped into the Juglone/SIBS solution. After removing the end- coated device from the solution, the coating is dried by placing the device in a forced air oven (400C) for 3 hours. The coated device is then further dried under vacuum for 24 hours. The dip coating process may be repeated to increase the amount of polymer/Juglone coated onto the device. In addition to Juglone, the following exemplary compounds that may be used to coat the device: ZD-6474, AP-23573, synthadotin, S-0885, aplidine, ixabepilone, IDN-5390, SB-2723005, ABT-518, combretastatin, anecortave acetate, SB- 715992, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17-DMAG, tacrolimus, temsirolimus, loteprednol etabonate, prednisolone, puromycin, 3-BAABE, cladribine, mannosemannose-6-phosphate, 5-azacytidine, Ly333531 (ruboxistaurin), dolastatin 15, and simvastatin.
This procedure may be used to coat other types of devices including peritoneal dialysis catheters, coronary stents, non-vascular stents, peripheral stents, hemodialysis access devices, guidewires, shunts, and anastomotic connectors, LVAD's.
EXAMPLE 53 JUGLONE/POLYMER COATING - ECHOGENIC OVERCOAT
A coated CVC device from Example 52 is dipped into a DESMODUR solution (50:50 mixture of dimethylsulfoxide and tetrahydrofuran). The anastomotic device is then removed and the coating is then partially dried at room temperature for 3 to 5 minutes. The device is then immersed in a beaker of water (room temperature) for 3-5 minutes to cause the polymerization reaction to occur rapidly. An echogenic coating is formed. In addition to juglone, the following are exemplary compounds that may be used to coat the device: ZD-6474, AP-23573, synthadotin, S-0885, aplidine, ixabepilone, IDN-5390, SB-2723005, ABT-518, combretastatin, anecortave acetate, SB-715992, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, isotretinoin, radicicot, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17- DMAG, tacrolimus, temsirolimus, loteprednol etabonate, prednisolone, puromycin, 3-BAABE, cladribine, mannosemannose-6-phosphate, 5- azacytidine, Ly333531 (ruboxistaurin), and simvastatin.
EXAMPLE 54 POLYMER/ECHOGENIC COATING
5% solutions of poly(styrene-co-isobutylene-styrene) (SIBS) are prepared using THF as the solvent. A LVAD device is dipped into the SIBS solution. After removing the device from the solution, the coating is dried by placing the device in a forced air oven (400C) for 3 hours. The coated device is then further dried under vacuum for 24 hours.
The coated device is dipped into a DESMODUR solution (50:50 mixture of dimethylsulfoxide and tetrahydrofuran). The device is then removed and the coating is then partially dried at room temperature for 3 to 5 minutes. The device is then immersed in a beaker of water (room temperature) for 3-5 minutes to cause the polymerization reaction to occur rapidly. The device is dried under vacuum for 24 hours at room temperature. The ends of the coated device are immersed into a solution of Juglone. The device is removed and dried at 400C for 1 hour and then under vacuum for 24 hours.
The amount of Juglone absorbed by the polymeric coating may be altered by changing the Juglone concentration, the immersion time as well as the solvent composition of the Juglone solution. In addition to juglone, the following are exemplary compounds that may be used to coat the device: ZD-6474, AP-23573, synthadotin, S-0885, aplidine, ixabepilone, IDN-5390, SB-2723005, ABT-518, combretastatin, anecortave acetate, SB- 715992, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, isotretinoin, radicicot, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17-DMAG, tacrolimus, temsirolimus, loteprednol etabonate, prednisolone, puromycin, 3-BAABE, cladribine, mannosemannose-6-phosphate, 5-azacytidine, Ly333531 (ruboxistaurin), dolastatin 15, and simvastatin.
This procedure may be used to coat other types of devices including peritoneal dialysis catheters, coronary stents, non-vascular stents, peripheral stents, hemodialysis access devices, guidewires, shunts, anastomotic connectors, CVCs.
EXAMPLE 55 COMBRETASTATIN / SlLOXANE COATING - END COATING
A central venous catheter is coated with a silioxane layer by exposing the device to gaseous tetramethylcyclotetrasiloxane that is then polymerized by low energy plasma polymerization onto the device surface. The thickness of the siloxane layer may be increased by increasing the polymerization time. The ends of the device are then immersed into a combretastatin / THF solution. The combretastatin is absorbed into the siloxane coating: The device is then removed from the solution and is dried for 2 hours at 400C in a forced air oven. The device is then further dried under vacuum at room temperature for 24 hours. The amount of combretastatin coated onto the device ends may be varied by altering the concentration of the combretastatin / THF solution as well as altering the immersion time of the device ends in the combretastatin THF solution. In addition to combretastatin, the following are exemplary compounds that may be used to coat the device: ZD-6474, AP-23573, synthadotin, S-0885, aplidine, ixabepilone, IDN-5390, SB-2723005, ABT-518, anecortave acetate, SB-715992, juglone, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17- DMAG, tacrolimus, temsirolimus, loteprednol etabonate, prednisolone, puromycin, 3-BAABE, cladribine, mannosemannose-6-phosphate, 5- azacytidine, Ly333531 (ruboxistaurin), dolastatin 15, and simvastatin. This procedure may be used to coat other types of devices including peritoneal dialysis catheters, coronary stents, non-vascular stents, peripheral stents, hemodialysis access devices, guidewires, Gl drainage tubes, shunts, and anastomotic connectors.
EXAMPLE 56 HEPARIN COATING
A CNS shunt device is dipped into a solution containing different amounts of a tridodecyl methyl ammonium chloride-heparin complex solution (PolySciences). After various incubation times, the device is removed and dried in a forced air oven (500C). The device is then further dried in a vacuum oven overnight. Other types of devices that may be coated with this procedure include coronary stents, peripheral stents, nasal and sinus stents, tracheal stents, peritoneal dialysis catheters, vascular grafts, hemodialysis access devices, guidewires, shunts, and anastomotic connectors.
EXAMPLE 57 SPRAY-COATED DEVICES
2% solutions poly(styrene-co-isobutylene-styrene) (SIBS) are prepared using THF as the solvent. Various amounts of Juglone are added to each solution. A device (e.g., a stent, central venous catheter, LVAD, anastomotic connector, or shunt) is held with a pair of tweezers and is then spray coated with one of the Juglone/polymer solutions using an airbrush. The device is then air-dried. The device is then held in a new location using the tweezers and a second coat of juglone/polymer is applied. The device is air-dried and is then dried under vacuum overnight. The total amount of juglone coated onto the device may be altered by changing the juglone content in the solution as well as by increasing the number of coatings applied. In addition to Juglone, the following are exemplary compounds that may be used to coat the device: ZD-6474, AP-23573, synthadotin, S-0885, aplidine, ixabepilone, IDN-5390, SB-2723005, ABT-518, combretastatin, anecortave acetate, SB-715992, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17- DMAG, tacrolimus, temsirolimus, loteprednol etabonate, prednisolone, puromycin, 3-BAABE, cladribine, mannosemannose-6-phosphate, 5- azacytidine, Ly333531 (ruboxistaurin), dolastatin 15, and simvastatin. .
EXAMPLE 58 DRUG COATED COVERED STENT-NON-DEGRADABLE
A covered stent (WALLGRAFT, Boston Scientific Corporation) is attached to a rotating mandrel. A solution of juglone (5% w/w O'uglone:polyurethane)) in a polyurethane (CHRONOFLEX 85A) / THF solution (2.5% w/v) is then sprayed onto the outer surface of the covered stent. The solution is sprayed on at a rate that ensures that the graft material is not damaged or saturated with the sprayed solution. The covered stent is allowed to air dry after which it is dried under vacuum for 24 hours. In addition to juglone, the following are exemplary compounds that may be used to coat the device: ZD-6474, AP-23573, synthadotin, S-0885, aplidine, ixabepilone, IDN-5390, SB-2723005, ABT-518, combretastatin, anecortave acetate, SB-715992, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17- DMAG, tacrolimus, temsirolimus, loteprednol etabonate, prednisolone, puromycin, 3-BAABE, cladribine, mannosemannose-6-phosphate, 5- azacytidine, Ly333531 (ruboxistaurin), dolastatin 15, and simvastatin. EXAMPLE 59 DRUG COATED COVERED STENT- DEGRADABLE
A WALLGRAFT stent is attached to a rotating mandrel. Juglone (5% w/w (juglone:PLGA)) in a PLGA/ethyl acetate solution (2.5% w/v) is then sprayed onto the outer surface of the covered stent. The solution is sprayed on at a rate that ensures that the graft material is not damaged or saturated with the sprayed solution. The covered stent is allowed to air dry after which it is dried under vacuum for 24 hours. In addition to Juglone, the following are exemplary compounds that may also be used to coat the device: ZD-6474, AP-23573, synthadotin, S-0885, aplidine, ixabepilone, IDN-5390, SB-2723005, ABT-518, combretastatin, anecortave acetate, SB-715992, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17- DMAG, tacrolimus, temsirolimus, lotepredήol etabonate, prednisolone, puromycin, 3-BAABE, cladribine, mannosemannose-6-phosphate, 5- azacytidine, Ly333531 (ruboxistaurin), dolastatin 15, and simvastatin.
EXAMPLE 60 DRUG COATED COVERED STENT - DEGRADABLE OVERCOAT
A drug-coated WALLGRAFT stent from either Example 58 or Example 59 is attached to a rotating mandrel. A PLGA/ethyl acetate solution (2.5% w/v) is then sprayed onto the outer surface of the covered stent such that a coating is formed over the initial drug containing coating. The solution is sprayed on at a rate that ensures that the graft material is not damaged or saturated with the sprayed solution. The covered stent is allowed to air dry after which it is dried under vacuum for 24 hours. EXAMPLE 61 DRUG-LOADED MICROSPHERE FORMULATION
Juglone (10% w/w (juglone.PLGA)) is added to a solution of PLGA (50/50, Mw « 54,000) in DCM (5% w/v (PLGA:DCM)). The solution is vortexed and then poured into a stirred (overhead stirrer with a 3 bladed TEFLON coated stirrer) aqueous PVA (approximately 89% hydrolyzed, Mw « 13,000, 2% w/v). The solution is stirred for 6 hours after which the solution is centrifuged to sediment the microspheres. The microspheres were resuspended in water. The centrifugation - washing process is repeated 4 times. The final microsphere solution is flash frozen in an acetone/dry-ice bath. The frozen solution is then freeze-dried to produce a fine powder. The size of the microspheres formed may be altered by changing the stirring speed and/or the PVA solution concentration. The freeze dried powder may be resuspended in PBS or saline and may be used for direct injection, as an incubation fluid or as an irrigation fluid. In addition to juglone, the following are exemplary compounds that may be used to coat the device: ZD-6474, AP-23573, synthadotin, S-0885, aplidine, ixabepilone, IDN-5390, SB-2723005, ABT-518, combretastatin, anecortave acetate, SB- 715992, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17-DMAG, tacrolimus, temsirolimus, loteprednol etabonate, prednisolone, puromycin, 3-BAABE, cladribine, mannosemannose-6-phosphate, 5-azacytidine, Ly333531 (ruboxistaurin), dolastatin 15, and simvastatin.
EXAMPLE 62 DRUG COATED STENT (EXTERIOR COATING)
A stent is dipped into a polyurethane (CHRONOFLEX 85A)ZTHF solution (2.5% w/v). The coated stent is allowed to air dry for 10 seconds. The stent is then rolled in powdered, loteprednol etabonate that is spread thinly on a piece of release liner. The rolling process is done in such a manner that the , loteprednol etabonate powder predominantly adheres to the exterior side of the coated stent. The stents are air-dried for 1 hour followed by vacuum drying for 24 hours. In addition to , loteprednol etabonate the following are exemplary compounds that may be used to coat the device: ZD-6474, AP-23573, synthadotin, S-0885, aplidine, ixabepilone, IDN-5390, SB-2723005, ABT-518, combretastatin, anecortave acetate, SB- 715992, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, isotretinoin, jugalone, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17-DMAG, tacrolimus, temsirolimus, prednisolone, puromycin, 3-BAABE, cladribine, mannosemannose-6-phosphate, 5-azacytidine, Ly333531 (ruboxistaurin), dolastatin 15, and simvastatin.
EXAMPLE 63 DRUG COATED STENT (EXTERIOR COATING) WITH A HEPARIN COATING
The drug-coated stent from Example 62 is further coated with a heparin coating. The stents that are prepared in Example 62 are dipped into a solution of heparin-benzalkonium chloride complex (1.5% (w/v) in isopropanol, STS Biopolymers). The stents are removed from the solution and are air-dried for 1 hour followed by vacuum drying for 24 hours. This process results in both the interior and exterior surfaces of the covered stent being coated with heparin.
EXAMPLE 64 PARTIAL DRUG COATING OF A COVERED STENT
A WALLGRAFT covered stent is attached to a rotating mandrel. A mask system is set up so that only the middle of the outer surface of the covered stent may be sprayed. A solution of juglone (5% w/w (juglone.polyurethane)) in a polyurethane (CHRONOFLEX 85A) / THF solution (2.5% w/v) is then sprayed onto the outer surface of the covered stent. The solution is sprayed on at a rate that ensures that the graft material is not damaged or saturated with the sprayed solution. The covered stent is allowed to air dry after which it is dried under vacuum for 24 hours. In addition to Juglone, the following are exemplary compounds that also may be used to coat the device: ZD-6474, AP-23573, synthadotin, S- 0885, aplidine, ixabepilone, IDN-5390, SB-2723005, ABT-518, combretastatin, anecortave acetate, SB-715992, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17-DMAG, tacrolimus, temsirolimus, loteprednol etabonate, prednisolone, puromycin, 3-BAABE, cladribine, mannosemannose-6-phosphate, 5-azacytidine, Ly333531 (ruboxistaurin), dolastatin 15, and simvastatin.
EXAMPLE 65 DRUG - DEXAMETHASONE COATED COVERED STENT
A WALLGRAFT covered stent is attached to a rotating mandrel. A mask system is set up so that only the middle of the outer surface of the covered stent may be sprayed. A solution of juglone (5% w/w (juglone:polyurethane)) in a polyurethane (CHRONOFLEX 85A) / THF solution (2.5% w/v) is then sprayed onto the outer surface of the covered stent. The solution is sprayed on at a rate that ensures that the graft material is not damaged or saturated with the sprayed solution. The covered stent is allowed to air dry. The mask is then rearranged so that only the ends of the outer surface of the covered stent may be sprayed. The ends of the outer surface of the covered stent are then sprayed with a dexamethasone (10% w/w)/ polyurethane (CHRONOFLEX 85A) / THF solution (2.5% w/v). The sample is air dried after which it is dried under vacuum for 24 hours. In addition to Juglone, the following are exemplary compounds that also may be used to coat the device: ZD-6474, AP-23573, synthadotin, S-0885, aplidine, ixabepilone, IDN-5390, SB-2723005, ABT- 518, combretastatin, anecortave acetate, SB-715992, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17-DMAG, tacrolimus, temsirolimus, loteprednol etabonate, prednisolone, puromycin, 3-BAABE, cladribine, mannosemannose-6-phosphate, 5-azacytidine, Ly333531 (ruboxistaurin), dolastatin 15, and simvastatin.
EXAMPLE 66 DRUG - HEPARIN COATED COVERED STENT
A WALLGRAFT covered stent is attached to a rotating mandrel. A mask system is set up so that only the middle of the outer surface of the covered stent may be sprayed. A solution of juglone (5% w/w Ouglone:polyurethane)) in a polyurethane (CHRONOFLEX 85A) / THF solution (2.5% w/v) is then sprayed onto the outer surface of the covered stent. The solution is sprayed on at a rate that ensures that the graft material is not damaged or saturated with the sprayed solution. The covered stent is allowed to air dry. The mask is then rearranged so that only the ends of the outer surface of the covered stent may be sprayed. The ends of the outer surface of the covered stent are then sprayed with a heparin-benzalkonium chloride complex (1.5% (w/v) in isopropanol, STS Biopolymers). The sample is air dried after which it is dried under vacuum for 24 hours. In addition to Juglone, the following are exemplary compounds that may be used to coat the device: ZD-6474, AP-23573, synthadotin, S-0885, aplidine, ixabepilone, IDN-5390, SB-2723005, ABT- 518, combretastatin, anecortave acetate, SB-715992, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17-DMAG, tacrolimus, temsirolimus, loteprednol etabonate, prednisolone, puromycin, 3-BAABE, cladribine, mannosemannose-6-phosphate, 5-azacytidine, Ly333531 (ruboxistaurin), dolastatin 15, and simvastatin.
EXAMPLE 67 DRUG-DEXAMETHAXONE COATED COVERED STENT
A WALLGRAFT stent is attached to a rotating mandrel. A solution of juglone (5% w/w (juglone:PLGA)) and dexamethazone (5%w/w (dexamethazone:PLGA)) in a PLGA (50/50, Mw « 54,000) / ethyl acetate solution (2.5% w/v) is sprayed onto the outer surface of the covered stent. The solution is sprayed on at a rate that ensures that the graft material is not damaged or saturated with the sprayed solution. The covered stent is allowed to air dry after which it is dried under vacuum for 24 hours. In addition to Juglone, the following are exemplary compounds that also may be used to coat the device: ZD-6474, AP-23573, synthadotin, S-0885, aplidine, ixabepilone, IDN-5390, SB-2723005, ABT-518, combretastatin, anecortave acetate, SB-715992, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17- DMAG, tacrolimus, temsirolimus, loteprednol etabonate, prednisolone, puromycin, 3-BAABE, cladribine, mannosemannose-6-phosphate, 5- azacytidine, Ly333531 (ruboxistaurin), dolastatin 15, and simvastatin. EXAMPLE 68
DRUG-DEXAMETHASONE COATED COVERED STENT (SEQUENTIAL COATING)
A WALLGRAFT stent is attached to a rotating mandrel. A solution of juglone (5% w/w (juglone: PLGA)) in a PLGA (50/50, Mw » 54,000) / ethyl acetate solution (2.5% w/v) is sprayed onto the outer surface of the covered stent. The solution is sprayed on at a rate that ensures that the graft material is not damaged or saturated with the sprayed solution. The covered stent is allowed to air dry. A methanol solution of dexamethasone is then sprayed onto the outer surface of the covered stent (at a rate that ensures that the graft material is not damaged or saturated with the sprayed solution). The covered stent is allowed to air dry after which it is dried under vacuum for 24 hours. In addition to Juglone, the following are exemplary compounds that may be used to coat the device: ZD-6474, AP-23573, synthadotin, S-0885, aplidine, ixabepilone, IDN-5390, SB-2723005, ABT-518, combretastatin, anecortave acetate, SB-715992, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17-DMAG, tacrolimus, temsirolimus, loteprednol etabonate, prednisolone, puromycin, 3-BAABE, cladribine, mannosemannose-6-phosphate, 5-azacytidine, Ly333531 (ruboxistaurin), dolastatin 15, and simvastatin.
EXAMPLE 69
RELEASE STUDY TO DETERMINE RELEASE PROFILE OF THE THERAPEUTIC AGENT
FROM A COATED DEVICE
A sample of the therapeutic agent-loaded catheter is placed in a 15 ml culture tube. 15 ml release buffer (Example 32) is added to the culture tube. The tube is sealed with a TEFLON lined screw cap and is placed on a rotating wheel in a 370C oven. At various time points, the buffer is withdrawn from the culture tube and is replaced with fresh buffer. The withdrawn buffer is then analyzed for the amount of therapeutic agent contained in this buffer solution using HPLC.
EXAMPLE 70 COMPLETE COATING - DIP COATING A VENA CAVA FILTER
Poly(ethylene-co-vinyl acetate) {28% vinyl acetate} [p(EVA)] is dissolved in 10 ml THF to produce a solution that has a polymer concentration of approximately 40 mg/mL. Juglone is added to the pEVA solution to produce a final Juglone concentration of 3 mg/mL. A vena cava filter is cleaned by immersing the filter into isopropanol for 30 minutes and then rinsing 3 times with isopropanol. The filter is air dried. The filter is dip coated by completely immersing the cleaned filter into the pEVA - juglone solution. The filter is the removed from the solution and is air dried. This process may be repeated until the desired Juglone dose is achieved. The filter is then dried under vacuum. Other fibrosis-inhbiting agents that may be coated onto a vena cava filter device using this procedure include: ZD- 6474, AP-23573, synthadotin, S-0885, aplidine, ixabepilone, IDN-5390, SB- 2723005, ABT-518, combretastatin, anecortave acetate, SB-715992, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17-DMAG, tacrolimus, temsirolimus, loteprednol etabonate, prednisolone, puromycin, 3-BAABE, cladribine, mannosemannose-6-phosphate, 5-azacytidine, Ly333531 (ruboxistaurin), dolastatin 15, and simvastatin.
EXAMPLE 71 PARTIAL COATING - DIP COATING A VENA CAVA FILTER
Polyurethane (CHRONOFLEX AL 85A) is dissolved in 10 ml THF to produce a solution that has a polymer concentration of approximately 400 mg/mL Juglone is added to the polyurethane solution to produce a final everolimus concentration of 3 mg/mL. A vena cava filter is cleaned by immersing the filter into isopropanol for 30 minutes and then rinsing 3 times with isopropanol. The filter is air dried. The filter is dip coated by immersing only the portions of the cleaned filter that will come into contact with the body tissue into the polyurethane -juglone solution. The filter is the removed from the solution and is air dried. This process may be repeated until the desired everolimus dose is achieved. The filter is then dried under vacuum. Other fibrosis-inhbiting agents that may be coated onto a vena cava filter device using this procedure include: ZD-6474, AP- 23573, synthadotin, S-0885, aplidine, ixabepilone, IDN-5390, SB-2723005, ABT-518, combretastatin, anecortave acetate, SB-715992, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17-DMAG, tacrolimus, temsirolimus, loteprednol etabonate, prednisolone, puromycin, 3-BAABE, cladribine, mannosemannose-6-phosphate, 5-azacytidine, Ly333531 (ruboxistaurin), dolastatin 15, and simvastatin.
EXAMPLE 72 COMPLETE COATING - SPRAY COATING
A 2% solution poly(styrene-co-isobutylene-styrene) (SIBS) is prepared using THF as the solvent. Juglone is added to the SIBS solution to produce a final Juglone concentration of 3 mg/mL. The SIBS -juglone solution is then transferred to the reservoir of an artist's air brush tool. A vena cava filter is cleaned by immersing the filter into isopropanol for 30 minutes and then rinsing 3 times with isopropanol. The filter is air dried. Using a crocodile clip, the filter is suspended in the air and is spray coated from several angles to ensure complete coating of the filter. Once the coating is dry to the touch, the filter is removed from the clip and the uncoated portion is spray coated. The filter is then air dried and/or vacuum dried to remove the solvent. This process may be repeated until the desired Juglone dose is achieved. The filter is then dried under vacuum. Other fibrosis-inhbiting agents that may be coated onto a vena cava filter device using this procedure include: ZD-6474, AP-23573, synthadotin, S-0885, aplidine, ixabepilone, IDN-5390, SB-2723005, ABT-518, combretastatin, anecortave acetate, SB-715992, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17- DMAG, tacrolimus, temsirolimus, loteprednol etabonate, prednisolone, puromycin, 3-BAABE, cladribine, mannosemannose-6-phosphate, 5- azacytidine, Ly333531 (ruboxistaurin), dolastatin 15, and simvastatin.
EXAMPLE 73 PARTIAL COATING - SPRAY COATING A VENA CAVA FILTER
A 2% solution poly(styrene-co-isobutylene-styrene) (SIBS) is prepared using THF as the solvent. Juglone is added to the SIBS solution to produce a final concentration of 3 mg/mL. The SIBS -juglone solution is then transferred to the reservoir of an artist's air brush tool. A vena cava filter is cleaned by immersing the filter into isopropanol for 30 minutes and then rinsing 3 times with isopropanol. The filter is air dried. Using a crocodile clip that is attached to a portion of the filter that is not to be coated, the filter is suspended in the air and is spray coated through a mask to ensure that only the desired portions of the filter are coated. The filter is then air dried and/or vacuum dried to remove the solvent. This process may be repeated until the desired Juglone dose is achieved. The filter is then dried under vacuum. Other fibrosis-inhbiting agents that may be coated onto a vena cava filter device using this procedure include: ZD-6474, AP-23573, synthadotin, S-0885, aplidine, ixabepilone, IDN-5390, SB-2723005, ABT- 518, combretastatin, anecortave acetate, SB-715992, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, isotretinoin, radicicol, ciobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17-DMAG1 tacrolimus, temsirolimus, loteprednol etabonate, prednisolone, puromycin, 3-BAABE, cladribine, mannosemannose-6-phosphate, 5-azacytidine, Ly333531 (ruboxistaurin), dolastatin 15, and simvastatin.
EXAMPLE 74 APPLICATION OF A SECOND COATING TO A VENA CAVA FILTER
Poly(ethylene-co-vinyl acetate) {28% vinyl acetate) [p(EVA)] is dissolved in 10 ml THF to produce a solution that has a polymer concentration of approximately 40 mg/mL. Juglone is added to the pEVA solution to produce a final Juglone concentration of 3 mg/mL. A vena cava filter is cleaned by immersing the filter into isopropanol for 30 minutes and then rinsing 3 times with isopropanol. The filter is air dried. The filter is dip coated by completely immersing the cleaned filter into the pEVA - juglone solution. The filter is the removed from the solution and is air dried. This process may be repeated until the desired Juglone dose is achieved. The filter is then dried under vacuum to remove the residual solvent. The filter is then dipped into an aqueous solution of sodium hyaluronate [HA] (mw approximately 1- 1.5 x 106 kDa, 10 mg/mL). The water is removed by air drying at 37 0C. The process is repeated until the desired amount of HA is coated onto the filter. The filter is then dried under vacuum. Other fibrosis- inhbiting agents that may be coated onto a vena cava filter device using this procedure include: ZD-6474, AP-23573, synthadotin, S-0885, aplidine, ixabepilone, IDN-5390, SB-2723005, ABT-518, combretastatin, anecortave acetate, SB-715992, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17-DMAG, tacrolimus, temsirolimus, loteprednol etabonate, prednisolone, puromycin, 3-BAABE, cladribine, mannosemannose-6-phosphate, 5-azacytidine, Ly333531 (rυboxistaurin), dolastatin 15, and simvastatin.
EXAMPLE 75 COATING CONTAINING TWO BIOACTIVE AGENTS FOR A VENA CAVA FILTER
Poly(ethylene-co-vinyl acetate) {28% vinyl acetate) [p(EVA)] is dissolved in 10 ml THF to produce a solution that has a polymer concentration of approximately 40 mg/mL Juglone is added to the pEVA solution to produce a final Juglone concentration of 3 mg/mL. Heparin- benzalkonium chloride is then added to the pEVA solution to achieve a final concentration of 1 mg/ml. A vena cava filter is cleaned by immersing the filter into isopropanol for 30 minutes and then rinsing 3 times with isopropanol. The filter is air dried. The filter is dip coated by completely immersing the cleaned filter into the pEVA - juglone solution. The filter is the removed from the solution and is air dried. This process may be repeated until the desired Juglone dose is achieved. The filter is then dried under vacuum. Other fibrosis-inhbiting agents that may be coated onto a vena cava filter device using this procedure include: ZD-6474, AP-23573, synthadotin, S-0885, aplidine, ixabepilone, IDN-5390, SB-2723005, ABT- 518, combretastatin, anecortave acetate, SB-715992, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17-DMAG, tacrolimus, temsirolimus, loteprednol etabonate, prednisolone, puromycin, 3-BAABE, cladribine, mannosemannose-6-phosphate, 5-azacytidine, Ly333531 (ruboxistaurin), dolastatin 15, and simvastatin.
EXAMPLE 76
Two COATING LAYERS CONTAINING TWO DIFFERENT BIOACTIVE AGENTS FOR A
VENA CAVA FILTER
Poly(ethylene-co-vinyl acetate) {28% vinyl acetate) [p(EVA)] is dissolved in 10 ml THF to produce a solution that has a polymer concentration of approximately 40 mg/mL Juglone is added to the pEVA solution to produce a final Juglone concentration of 3 mg/mL. A vena cava filter is cleaned by immersing the filter into isopropanol for 30 minutes and then rinsing 3 times with isopropanol. The filter is air dried. The filter is dip coated by completely immersing the cleaned filter into the pEVA -juglone solution. The filter is the removed from the solution and is air dried. This process may be repeated until the desired Juglone dose is achieved. The filter is then dried under vacuum to remove the residual solvent. The filter is then dipped into an aqueous solution of sodium hyaluronate [HA] (mw approximately 1- 1.5 x 106 kDa, 10 mg/mL) that contains 1 mg/ml heparin. The water is removed by air drying at 37 0C. The process is repeated until the desired amount of HA is coated onto the filter. The filter is then dried under vacuum. Other fibrosis-inhbiting agents that may be coated onto a vena cava filter device using this procedure include: ZD-6474, AP-23573, synthadotin, S-0885, aplidine, ixabepilone, IDN-5390, SB-2723005, ABT- 518, combretastatin, anecortave acetate, SB-715992, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17-DMAG, tacrolimus, temsirolimus, loteprednol etabonate, prednisolone, puromycin, 3-BAABE, cladribine, mannosemannose-6-phosphate, 5-azacytidine, Ly333531 (ruboxistaurin), dolastatin 15, and simvastatin. EXAMPLE 77 DRUG INCORPORATION INTO A VASCULAR GRAFT
A solution of Juglone is prepared by dissolving 70 mg Juglone in 10 mL water/ethanol (1:1) in a 20 mL glass scintillation vial. A 5 cm piece of an ePTFE vascular graft (IMPRA, 6 mm) is immersed in the solution. The solution is placed in an ultrasonic bath (Fisher) for 1 min. The graft is removed using a pair of tweezers. The graft is air dried for 3 hours after which it is dried under vacuum for 24 hours. Other fibrosis-inhbiting agents that may be coated onto a vascular graft device using this procedure include ZD-6474, AP-23573, synthadotin, S-0885, aplidine, ixabepilone, IDN-5390, SB-2723005, ABT-518, combretastatin, anecortave acetate, SB-715992, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17-DMAG, tacrolimus, temsirolimus, loteprednol etabonate, prednisolone, puromycin, 3-BAABE, cladribine, mannosemannose-6-phosphate, 5-azacytidine, Ly333531 (ruboxistaurin), dolastatin 15, and simvastatin.
EXAMPLE 78 DRUG INCORPORATION INTO A TYMPANOSTOMY TUBE
Five 15 mL solutions of Juglone at 5 mg/ml are prepared in methanol in a 20 mL scintillation vial. A soft silicone T-tube ((Medco Catalogue Number T5030) is then immersed in each of the Juglone solutions. The tubes are removed from the juglone solutions at 30 min, 1 hour, 2 hours, 6 hours and 24 hours. The tubes are air dried and then dried under vacuum for 24 hours. Other fibrosis-inhbiting agents that may be coated onto a tympanostomy tube device using this procedure include: ZD- 6474, AP-23573, synthadotin, S-0885, aplidine, ixabepilone, IDN-5390, SB- 2723005, ABT-518, combretastatin, anecortave acetate, SB-715992, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17-DMAG, tacrolimus, temsirolimus, loteprednol etabonate, prednisolone, puromycin, 3-BAABE, cladribine, mannosemannose-6-phosphate, 5-azacytidine, Ly333531 (ruboxistaurin), dolastatin 15, and simvastatin.
EXAMPLE 79 DRUG INCORPORATION INTO A TYMPANOSTOMY TUBE
Five 15 ml_ solution of juglone (5 mg/mL) and 5-fluorouracil (4 mg/mL) are prepared in methanol in a 20 ml_ scintillation vial. A soft silicone T-tube ((Medco Catalogue Number T5030) is then immersed in each of the Juglone solutions. The tubes are removed from the juglone solutions at 30 ηnin, 1 hour, 2 hours, 6 hours and 24 hours. The tubes are air dried and then dried under vacuum for 24 hours. Other fibrosis-inhbiting agents that may be coated onto a tympanostomy tube device using this procedure include: ZD-6474, AP-23573, synthadotin, S-0885, aplidine, ixabepilone, IDN-5390, SB-2723005, ABT-518, combretastatin, anecortave acetate, SB-715992, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17-DMAG, tacrolimus, temsirolimus, loteprednol etabonate, prednisolone, puromycin, 3-BAABE, cladribine, mannosemannose-6-phosphate, 5-azacytidine, Ly333531 (ruboxistaurin), dolastatin 15, and simvastatin.
EXAMPLE 80 DRUG INCORPORATION INTO AN INTRAOCULAR LENS
Five 15 mL solution of juglone (1mg/mL) are prepared in methanol in a 20 mL scintillation vial. An intra-ocular lens (STAAR) then immersed in each of the juglone solutions. The lenses are removed from the Juglone solutions at 30 min, 1 hour, 2 hours, 6 hours and 24 hours. The lenses are air dried and then dried under vacuum for 24 hours. Other fibrosis-inhbiting agents that may be coated onto an intraocular lens device using this procedure include: ZD-6474, AP-23573, synthadotin, S-0885, aplidine, ixabepilone, IDN-5390, SB-2723005, ABT-518, combretastatin, anecortave acetate, SB-715992, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17- DMAG, tacrolimus, temsirolimus, loteprednol etabonate, prednisolone, puromycin, 3-BAABE, cladribine, mannosemannose-6-phosphate, 5- azacytidine, Ly333531 (ruboxistaurin), dolastatin 15, and simvastatin.
EXAMPLE 81
SCREENING ASSAY TO ASSESS THE EFFECT OF COMPOUNDS ON MATRIX METALLOPROTEASE 1 ACTIVITY
Cultures of primary human dermal fibroblasts were grown to confluence, re-seeded in 12-well plates (50,000 cells/well) and grown to 80- 90% confluency. Prior to growth factor stimulation, cells were starved under low serum conditions (0.5% fetal bovine serum) for 24 hours. Cells were then stimulated with 10 ng/mL of platelet-derived growth factor-BB (PDGF- BB) and 1 ng/mL of IL-1 β for 72 hours. Compounds to be tested for Matrix Metalloprotease 1 (MMP-1) inhibitory activity were added concurrently with growth factors at concentrations between 10"11 M and 10'5 M. After 72 hours, tissue culture media containing MMP-1 was collected and stored at - 20°C. To isolate MMP-1, an MMP-1 specific monoclonal antibody that binds both the pro- and mature forms of MMP-1 was coated onto black 96-well microplates and tissue culture media is added. After washing away unbound material, the pro- form of MMP-1 was converted to the mature, active form by addition of the activation reagent, p-aminophenylmercuric acetate (APMA). To assay for MMP-1 activity, a MMP-1 peptide substrate (200 μl_ of a 1mM solution) was added to the wells following a wash in wash buffer and the plates were incubated at 37°C for 20 hours in the dark. In the presence of active MMP-1 , the peptide was cleaved and the product was detected by a fluorescent microplate reader. Upon exitation at ~320 nm, the cleaved product would emit a fluorescent signal at ~405nm, which was proportional to the amount of MMP-1 specific enzymatic activity in the sample. Relative fluorescence units from triplicate wells were averaged after subtracting background fluorescence. Fluoresence units were converted to activity units utilizing a standard curve generated with recombinant pro-MMP-1 serially diluted from 12.5 ng/mL to 0.39 ng/mL. Inhibitory concentration of 50% (IC50) was determined for test compounds by comparing the average MMP-1 activity obtained in presence of compound to the positive control (supernatants from cells stimulated in the absence of compound). From one to, optimally, four replicate experiments were conducted to determine average IC5O values. The IC5O of selected drugs on fibroblast MMP-1 activity were determined as follows: geldanamycin (1.3 nM); 17-AAG (30 nM); 17-DMAG (8 nM); simvastatin (no inhibitory activity); mycophenolic acid (518 nM); 1a,25-dihydroxyvitamin D3, vitamin D3 (2249 nM); SP600125 (1869 nM); halofuginone hydrobromide, HBr (15 nM); fascaplysin (1073 nM); puromycin dihydrochloride from Streptomyces alboniger (626 nM); celastrol, celastrus scandens, tripterin (405 nM); imatinib mesilate, STI571, CGP 57148B (109 nM); CPKC 412A, midostaurin, N-benzoylstaurosporine, PKC412A (80 nM); LBM642 (1032 nM); radicicol from Humicola fuscoatra (700 nM); farglitazar, GI262570X (2197 nM); SC12267 (902 nM); homoharringtonine (26 nM); prednisolone 21-acetate (86 nM). Other exemplary compounds that may be tested in this assay include, ZD-6474, AP-23573, synthadotin, S-0885, aplidine, ixabepilone, IDN-5390, SB-2723005, ABT-518, combretastatin, anecortave acetate, SB- 715992, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, juglone, isotretinoin, clobetasol propionate, trichostatin A, brefeldin A, thapsigargin, doiastatin 15, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, tacrolimus, temsirolimus, loteprednol etabonate, 3-BAABE, cladribine, mannose-6-phosphate, 5-azacytidine, and Ly333531 (ruboxistaurin).
All of the above U.S. patents, U.S. patent application publications, U.S. patent applications, foreign patents, foreign patent applications and non-patent publications referred to in this specification and/or listed in the Application Data Sheet, are incorporated herein by reference, in their entirety.
From the foregoing it will be appreciated that, although specific embodiments of the invention have been described herein for purposes of illustration, various modifications may be made without deviating from the spirit and scope of the invention. Accordingly, the invention is not limited except as by the appended claims.

Claims

1. A device, comprising a medical device (i.e., an implant) and an anti-scarring agent or a composition comprising an anti-scarring agent, wherein the agent inhibits scarring between the device and a host into which the device is implanted.
2. The device of claim 1 wherein the medical device is an intravascular device, a gastrointestinal stent, a tracheal and bronchial stent, a genital urinary stent, an ear and nose stent, an ear ventilation tube, an intraocular implant, a medical device for treating hypertropic scar or keloid, a vascular graft, a hemodialysis access device, a medical device comprising a film or a mesh, a glaucoma drainage device, a prosthetic heart valve or component thereof, a penile implant, an endotracheal or tracheostomy tube, a peritoneal dialysis catheter, a central nervous system shunt or pressure monitor device, an inferior vena cava filter, a gastrointestinal device, a central venous catheter, a ventricular assist device, a spinal implant, a surgical adhesion barrier.
3. The device of claim 1 or claim 2 wherein the anti- scarring agent is an antimicrobial compound.
4. The device of claim 3 wherein the antimicrobial compound is brefeldin A.
5. The device of claim 1 or claim 2 wherein the anti- scarring agent is selected from a histamine receptor antagonist, an alpha adrenergic receptor antagonist, an anti-psychotic compound, a CaM kinase Il inhibitor, a G protein agonist, an antibiotic selected from the group consisting of apigenin, ampicillin sodium salt, puromycin, an anti-microbial agent, a DNA topoisomerase inhibitor, a thromboxane A2 receptor inhibitor, a D2 dopamine receptor antagonist, a Peptidyl-Prolyl Cis/Trans lsomerase Inhibitor, a dopamine antagonist, an anesthetic compound, a clotting factor, a lysyl hydrolase inhibitor, a muscarinic receptor inhibitor, a superoxide anion generator, a steroid, an antiproliferative agent, a diuretic, an anticoagulant, a cyclic GMP agonist, an adenylate cyclase agonist, an antioxidant, a nitric oxide synthase inhibitor, an anti-neoplastic agent, a DNA synthesis inhibitor, a DNA alkylating agent, a DNA methylation inhibitor, a NSAID agent, a peptidylglycine alpha-hydroxylating monooxygenase inhibitor, an MEK1/MEK 2 inhibitor, a NO synthase inhibitor, a retinoic acid receptor antagonist, an ACE inhibitor, a glycosylation inhibitor, an intracellular calcium influx inhibitor, an anti-emetic agent, an acetylcholinesterase inhibitor, an ALK-5 receptor antagonist, a RAR/RXT antagonist, an elF-2a inhibitor, an S-adenosyl-L-homocysteine hydrolase inhibitor, an estrogen agonist, a serotonin receptor inhibitor, an antithrombotic agent, a tryptase inhibitor, a pesticide, a bone mineralization promoter, a bisphosphonate compound selected from risedronate and an analogue or derivative thereof, an anti-inflammatory compound, a DNA methylation promoter, an anti-spasmodic agent, a protein synthesis inhibitor, an D-glucosidase inhibitor, a calcium channel blocker, a pyruvate dehydrogenase activator, a prostaglandin inhibitor, a sodium channel inhibitor, a serine protease inhibitor, an intracellular calcium flux inhibitor, a JAK2 inhibitor, an androgen inhibitor, an aromatase inhibitor, an anti-viral agent, a 5-HT inhibitor, an FXR antagonist, an actin polymerization and stabilization promoter, an AXOR12 agonist, an angiotensin Il receptor agonist, a platelet aggregation inhibitor, a CB1/CB2 receptor agonist, a norepinephrine reuptake inhibitor, a selective serotonin reuptake inhibitor, a reducing agent, and a immuno-modulator selected from Bay 11-7085, (-)- arctigenin, idazoxan hydrochloride.
6. The device of claim 1 or claim 2 wherein the anti- scarring agent is selected from an angiogenesis inhibitor, an apoptosis antagonist, a Raf kinase inhibitor, reversible inhibitor of ErbB1 and ErbB2, ribonucleoside triphosphate reductase inhibitor, an SDF-1 antagonist, a sheddase inhibitor, an SRC inhibitor, a stromelysin inhibitor, an Syk kinase inhibitor, a telomerase inhibitor, a TGF beta inhibitor, a TNF-alpha antagonist or TACE inhibitor, a tumor necrosis factor antagonist, a Toll receptor inhibitor, a tubulin antagonist, a protein tyrosine kinase inhibitor, a VEGF inhibitor, and a vitamin D receptor agonist.
7. The device of claim 1 or claim 2 wherein the anti- scarring agent is selected from a retinoic acid receptor antagonist, a heat shock protein 90 antagonist, a steroid, a cell cycle inhibitor, a histone deacetylase inhibitor, an anti-microbial agent, an intracellular calcium flux inhibitor, an microtubule inhibitor, an HMGCoA reductase inhibitor, an actin polymerization and stabilization promoter, a tyrosine kinase inhibitor, a TGF beta inhibitor, a TNF-alpha antagonist, a TACE inhibitor, a calcineurin inhibitor, a peptidyl-prolyl cis/trans isomerase inhibitor, an apoptosis activator, an antimetabolite and anti-neoplastic agent, a TGF beta inhibitor, a DNA methylation promoter, and a PKC inhibitor.
8. The device of claim 1 or claim 2 wherein the anti- scarring agent is selected from ZD-6474, AP-23573, synthadotin, S-0885, aplidine, ixabepilone, IDN-5390, SB-2723005, ABT-518, combretastatin, anecortave acetate, SB-715992, temsirolimus, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, dolastatin 15, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17-DMAG, tacrolimus, loteprednol etabonate, juglone, prednisolone, puromycin, 3-BAABE, cladribine, mannose-6-phosphate, 5-azacytidine, Ly333531 (ruboxistaurin), and simvastatin.
4643 antagonist, an apoptosis activator, a beta 1 integrin antagonist, a beta tubulin inhibitor, a blocker of enzyme production in Hepatitis C, a Bruton's tyrosine kinase inhibitor, a calcineurin inhibitor, a caspase 3 inhibitor, a CC chemokine receptor antagonist, a cell cycle inhibitor, a cathepsin B inhibitor, a cathepsin K inhibitor, a cathepsin L inhibitor, a CD40 antagonist, a chemokine receptor antagonist, a chymase inhibitor, a collagenase antagonist, a CXCR antagonist, a cyclin dependent kinase inhibitor, a cyclooxygenase 1 inhibitor, a DHFR inhibitor, a cual integrin inhibitor, an elastase inhibitor, an elongation factor-1 alpha inhibitor, an endothelial growth factor antagonist, an endothelial growth factor receptor kinase inhibitor, an endotoxin antagonist, an epothilone and tubulin binder, an estrogen receptor antagonist, an FGF inhibitor, a famexyl transferase inhibitor, a farnesyltransferase inhibitor, an FLT-3 kinase inhibitor, an FGF receptor kinase inhibitor, a fibrinogen antagonist, a histone deacetylase inhibitor, an HMGCoA reductase inhibitor, an ICAM inhibitor, an IL, ICE, and IRAK antagonist, an IL-2 inhibitor, an immunosuppressant, an inosine monophosphate inhibitor, an integrin antagonist, an interleukin antagonist, an inhibitor of type III receptor tyrosine kinase, an irreversible inhibitor of enzyme methionine aminopeptidase type 2, an isozyme selective delta protein kinase C inhibitor, a JAK3 enzyme inhibitor, a JNK inhibitor, a kinase inhibitor, a kinesin antagonist, a leukotriene inhibitor and antagonist, a MAP kinase inhibitor, a matrix metalloproteinase inhibitor, an MCP-CCR2 inhibitor, an mTOR inhibitor, an mTOR kinase inhibitor, a microtubule inhibitor, an MIF inhibitor, a neurokinin antagonist, an NF kappa B inhibitor, a nitric oxide agonist, an ornithine decarboxylase inhibitor, a p38 MAP kinase inhibitor, a palmitoyl-protein thioesterase inhibitor, a PDGF receptor kinase inhibitor, a peroxisome proliferator-activated receptor (PPAR) agonist, a phosphatase inhibitor, a phosphodiesterase inhibitor, a PKC inhibitor, a platelet activating factor antagonist, a prolyl hydroxylase inhibitor, a polymorphonuclear neutrophil inhibitor, protein kinase B inhibitor, protein kinase C stimulant, purine nucleoside analogue, a purineoreceptor P2X
4642
9. The device of any one of claims 1-8 further comprising a coating wherein the coating comprises (a) the anti-scarring agent or (b) the anti-scarring agent and a polymer.
10. The device of any one of claims 1-8 further comprising a polymer or further comprising a polymeric carrier.
11. The device of any one of claims 1-8 further comprising a second pharmaceutically active agent.
12. The device of any one of claims 1-8 further comprising at least one of an anti-inflammatory agent, an agent that inhibits infection, anthracycline, doxorubicin, mitoxantrone, fluoropyrimidine, 5-fluorouracil, a folic acid antagonist, methotrexate, podophylotoxin, etoposide, camptothecin, hydroxyurea, a platinum complex, cisplatin, an antithrombotic agent, a visualization agent, and an echogenic material.
13. The device of any one of claims 1 -8 wherein the device is adapted for delivering the anti-scarring agent locally to tissue proximate to the device.
14. The device of any one of claims 1-8 wherein the anti- scarring agent is released into tissue in the vicinity of the device after deployment of the device.
15. The device of any one of claims 1-8 wherein the device comprises (a) about 0.01 μg to about 10 μg of the anti-scarring agent; (b) about 10 μg to about 10 mg of the anti-scarring agent; (c) about 10 mg to about 250 mg of the anti-scarring agent; (d) about 250 mg to about 1000 mg of the anti-scarring agent; or (e) about 1000 mg to about 2500 mg of the anti-scarring agent.
16. The device of any one of claims 1-8 wherein the device comprises (a) about 0.01 μg to about 1 μg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied; (b) about 1 μg to about 10 μg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied; (c) about 10 μg to about 250 μg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied; (d) about 250 μg to about 1000 μg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied; or (e) about 1000 μg to about 2500 μg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
17. The device of any one of claims 1-8 wherein the device comprises (a) about 0.01 μg to about 100 μg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied; (b) about 0.01 μg to about 200 μg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied; or (c) about 0.01 μg to about 500 μg of the anti-scarring agent per mm2 of device surface to which the anti- scarring agent is applied.
18. A method for inhibiting scarring comprising placing a device that comprises a medical device and an anti-scarring agent or a composition comprising an anti-scarring agent into an animal host, wherein the agent inhibits scarring.
19. The method of claim 18 wherein the medical device is an intravascular device, a gastrointestinal stent, a tracheal and bronchial stent, a genital urinary stent, an ear and nose stent, an ear ventilation tube, an intraocular implant, a medical device for treating hypertropic scar or keloid, a vascular graft, a hemodialysis access device, a medical device comprising a film or a mesh, a glaucoma drainage device, a prosthetic heart valve or component thereof, a penile implant, an endotracheal or tracheostomy tube, a peritoneal dialysis catheter, a central nervous system shunt or pressure monitor device, an inferior vena cava filter, a gastrointestinal device, a central venous catheter, a ventricular assist device, a spinal implant, a surgical adhesion barrier.
20. The method of claim 18 or claim 19 wherein the anti- scarring agent is an antimicrobial compound.
21. The method of claim 20 wherein the antimicrobial compound is brefeldin A.
22. The method of claim 18 or claim 19 wherein the anti- scarring agent is selected from a histamine receptor antagonist, an alpha adrenergic receptor antagonist, an anti-psychotic compound, a CaM kinase Il inhibitor, a G protein agonist, an antibiotic selected from the group consisting of apigenin, ampicillin sodium salt, puromycin, an anti-microbial agent, a DNA topoisomerase inhibitor, a thromboxane A2 receptor inhibitor, a D2 dopamine receptor antagonist, a Peptidyl-Prolyl Cis/Trans lsomerase Inhibitor, a dopamine antagonist, an anesthetic compound, a clotting factor, a lysyl hydrolase inhibitor, a muscarinic receptor inhibitor, a superoxide anion generator, a steroid, an antiproliferative agent, a diuretic, an anticoagulant, a cyclic GMP agonist, an adenylate cyclase agonist, an antioxidant, a nitric oxide synthase inhibitor, an anti-neoplastic agent, a DNA synthesis inhibitor, a DNA alkylating agent, a DNA methylation inhibitor, a NSAID agent, a peptidylglycine alpha-hydroxylating monooxygenase inhibitor, an MEK1/MEK 2 inhibitor, a NO synthase inhibitor, a retinoic acid receptor antagonist, an ACE inhibitor, a glycosylation inhibitor, an intracellular calcium influx inhibitor, an anti-emetic agent, an acetylcholinesterase inhibitor, an ALK-5 receptor antagonist, a RAR/RXT antagonist, an elF-2a inhibitor, an S-adenosyl-L-homocysteine hydrolase inhibitor, an estrogen agonist, a serotonin receptor inhibitor, an anti- thrombotic agent, a tryptase inhibitor, a pesticide, a bone mineralization promoter, a bisphosphonate compound selected from risedronate and an analogue or derivative thereof, an anti-inflammatory compound, a DNA methylation promoter, an anti-spasmodic agent, a protein synthesis inhibitor, an α-glucosidase inhibitor, a calcium channel blocker, a pyruvate dehydrogenase activator, a prostaglandin inhibitor, a sodium channel inhibitor, a serine protease inhibitor, an intracellular calcium flux inhibitor, a JAK2 inhibitor, an androgen inhibitor, an aromatase inhibitor, an anti-viral agent, a 5-HT inhibitor, an FXR antagonist, an actin polymerization and stabilization promoter, an AXOR12 agonist, an angiotensin Il receptor agonist, a platelet aggregation inhibitor, a CB1/CB2 receptor agonist, a norepinephrine reuptake inhibitor, a selective serotonin reuptake inhibitor, a reducing agent, and a immuno-modulator selected from Bay 11-7085, (-)- arctigenin, idazoxan hydrochloride.
23. The method of claim 18 or claim 19 wherein the anti- scarring agent is selected from an angiogenesis inhibitor, an apoptosis antagonist, an apoptosis activator, a beta 1 integrin antagonist, a beta tubulin inhibitor, a blocker of enzyme production in Hepatitis C, a Bruton's tyrosine kinase inhibitor, a calcineurin inhibitor, a caspase 3 inhibitor, a CC chemokine receptor antagonist, a cell cycle inhibitor, a cathepsin B inhibitor, a cathepsin K inhibitor, a cathepsin L inhibitor, a CD40 antagonist, a chemokine receptor antagonist, a chymase inhibitor, a collagenase antagonist, a CXCR antagonist, a cyclin dependent kinase inhibitor, a cyclooxygenase 1 inhibitor, a DHFR inhibitor, a cual integrin inhibitor, an elastase inhibitor, an elongation factor-1 alpha inhibitor, an endothelial growth factor antagonist, an endothelial growth factor receptor kinase inhibitor, an endotoxin antagonist, an epothilone and tubulin binder, an estrogen receptor antagonist, an FGF inhibitor, a farnexyl transferase inhibitor, a farnesyltransferase inhibitor, an FLT-3 kinase inhibitor, an FGF receptor kinase inhibitor, a fibrinogen antagonist, a histone deacetylase inhibitor, an HMGCoA reductase inhibitor, an ICAM inhibitor, an IL, ICE, and IRAK antagonist, an IL-2 inhibitor, an immunosuppressant, an inosine monophosphate inhibitor, an integrin antagonist, an interleukin antagonist, an inhibitor of type III receptor tyrosine kinase, an irreversible inhibitor of enzyme methionine aminopeptidase type 2, an isozyme selective delta protein kinase C inhibitor, a JAK3 enzyme inhibitor, a JNK inhibitor, a kinase inhibitor, a kinesin antagonist, a leukotriene inhibitor and antagonist, a MAP kinase inhibitor, a matrix metalloproteinase inhibitor, an MCP-CCR2 inhibitor, an mTOR inhibitor, an mTOR kinase inhibitor, a microtubule inhibitor, an MIF inhibitor, a neurokinin antagonist, an NF kappa B inhibitor, a nitric oxide agonist, an ornithine decarboxylase inhibitor, a p38 MAP kinase inhibitor, a palmitoyl-protein thioesterase inhibitor, a PDGF receptor kinase inhibitor, a peroxisome proliferator-activated receptor (PPAR) agonist, a phosphatase inhibitor, a phosphodiesterase inhibitor, a PKC inhibitor, a platelet activating factor antagonist, a prolyl hydroxylase inhibitor, a polymorphonuclear neutrophil inhibitor, protein kinase B inhibitor, protein kinase C stimulant, purine nucleoside analogue, a purineoreceptor P2X antagonist, a Raf kinase inhibitor, reversible inhibitor of ErbB1 and ErbB2, ribonucleoside triphosphate reductase inhibitor, an SDF-1 antagonist, a sheddase inhibitor, an SRC inhibitor, a stromelysin inhibitor, an Syk kinase inhibitor, a telomerase inhibitor, a TGF beta inhibitor, a TNF-alpha antagonist or TACE inhibitor, a tumor necrosis factor antagonist, a Toll receptor inhibitor, a tubulin antagonist, a protein tyrosine kinase inhibitor, a VEGF inhibitor, and a vitamin D receptor agonist.
24. The method of claim 18 or claim 19 wherein the anti- scarring agent is selected from a retinoic acid receptor antagonist, a heat shock protein 90 antagonist, a steroid, a cell cycle inhibitor, a histone deacetylase inhibitor, an anti-microbial agent, an intracellular calcium flux inhibitor, an microtubule inhibitor, an HMGCoA reductase inhibitor, an actin polymerization and stabilization promoter, a tyrosine kinase inhibitor, a TGF beta inhibitor, a TNF-alpha antagonist, a TACE inhibitor, a calcineurin inhibitor, a peptidyl-prolyl cis/trans isomerase inhibitor, an apoptosis activator, an antimetabolite and anti-neoplastic agent, a TGF beta inhibitor, a DNA methylation promoter, and a PKC inhibitor.
25. The method of claim 18 or claim 19 wherein the anti- scarring agent is selected from ZD-6474, AP-23573, synthadotin, S-0885, aplidine, ixabepilone, IDN-5390, SB-2723005, ABT-518, combretastatin, anecortave acetate, SB-715992, temsirolimus, adaiimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, dolastatin 15, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17-DMAG, tacrolimus, loteprednol etabonate, juglone, prednisolone, pυromycin, 3-BAABE, cladribine, mannose-6-phosphate, 5-azacytidine, Ly333531 (ruboxistaurin), and simvastatin.
26. The method of any one of claims 18-25 wherein the device comprises a coating wherein the coating comprises (a) the anti- scarring agent or (b) the anti-scarring agent and a polymer.
27. The method of any one of claims 18-25 wherein the device further comprises a polymer or a polymeric carrier.
28. The method of any one of claims 18-25 wherein the device further comprises a second pharmaceutically active agent.
29. The method of any one of claims 18-25 wherein the device further comprises at least one of an anti-inflammatory agent, an agent that inhibits infection, anthracycline, doxorubicin, mitoxantrone, fluoropyrimidine, 5-fluorouracil, a folic acid antagonist, methotrexate, podophylotoxin, etoposide, camptothecin, hydroxyurea, a platinum complex, cisplatin, an anti-thrombotic agent, a visualization agent, and an echogenic material.
30. The method of any one of claims 18-25 wherein the device is adapted for delivering the anti-scarring agent locally to tissue proximate to the device.
31. The method of any one of claims 18-25 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device.
32. The method of any one of claims 18-25 wherein the device comprises (a) about 0.01 μg to about 10 μg of the anti-scarring agent; (b) about 10 μg to about 10 mg of the anti-scarring agent; (c) about
10 mg to about 250 mg of the anti-scarring agent; (d) about 250 mg to about 1000 mg of the anti-scarring agent; or (e) about 1000 mg to about 2500 mg of the anti-scarring agent.
33. The method of any one of claims 18-25 wherein the device comprises (a) about 0.01 μg to about 1 μg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied; (b) about 1 μg to about 10 μg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied; (c) about 10 μg to about 250 μg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied; (d) about 250 μg to about 1000 μg of the anti- scarring agent per mm2 of device surface to which the anti-scarring agent is applied; or (e) about 1000 μg to about 2500 μg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
34. The method of any one of claims 18-25 wherein the device comprises (a) about 0.01 μg to about 100 μg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied; (b) about 0.01 μg to about 200 μg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied; or (c) about 0.01 μg to about 500 μg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
35. A method of making a device comprising: combining a medical device and an anti-scarring agent or a composition comprising an anti-scarring agent, wherein the agent inhibits scarring between the device and a host into which the device is implanted.
36. The method of claim 35 wherein the medical device is an intravascular device, a gastrointestinal stent, a tracheal and bronchial stent, a genital urinary stent, an ear and nose stent, an ear ventilation tube, an intraocular implant, a medical device for treating hypertropic scar or keloid, a vascular graft, a hemodialysis access device, a medical device comprising a film or a mesh, a glaucoma drainage device, a prosthetic heart valve or component thereof, a penile implant, an endotracheal or tracheostomy tube, a peritoneal dialysis catheter, a central nervous system shunt or pressure monitor device, an inferior vena cava filter, a gastrointestinal device, a central venous catheter, a ventricular assist device, a spinal implant, a surgical adhesion barrier.
37. The method of claim 35 or claim 36 wherein the anti- scarring agent is an antimicrobial compound.
38. The method of claim 37 wherein the antimicrobial compound is brefeldin A.
39. The method of claim 35 or claim 36 wherein the anti- scarring agent is selected from a histamine receptor antagonist, an alpha adrenergic receptor antagonist, an anti-psychotic compound, a CaM kinase Il inhibitor, a G protein agonist, an antibiotic selected from the group consisting of apigenin, ampicillin sodium salt, puromycin, an anti-microbial agent, a DNA topoisomerase inhibitor, a thromboxane A2 receptor inhibitor, a D2 dopamine receptor antagonist, a Peptidyl-Prolyl Cis/Trans lsomerase Inhibitor, a dopamine antagonist, an anesthetic compound, a clotting factor, a lysyl hydrolase inhibitor, a muscarinic receptor inhibitor, a superoxide anion generator, a steroid, an anti-proliferative agent, a diuretic, an anticoagulant, a cyclic GMP agonist, an adenylate cyclase agonist, an antioxidant, a nitric oxide synthase inhibitor, an anti-neoplastic agent, a DNA synthesis inhibitor, a DNA alkylating agent, a DNA methylation inhibitor, a NSAID agent, a peptidylglycine alpha-hydroxylating monooxygenase inhibitor, an MEK1/MEK 2 inhibitor, a NO synthase inhibitor, a retinoic acid receptor antagonist, an ACE inhibitor, a glycosylation inhibitor, an intracellular calcium influx inhibitor, an anti-emetic agent, an acetylcholinesterase inhibitor, an ALK-5 receptor antagonist, a RAR/RXT antagonist, an elF-2a inhibitor, an S-adenosyl-L-homocysteine hydrolase inhibitor, an estrogen agonist, a serotonin receptor inhibitor, an antithrombotic agent, a tryptase inhibitor, a pesticide, a bone mineralization promoter, a bisphosphonate compound selected from risedronate and an analogue or derivative thereof, an anti-inflammatory compound, a DNA methylation promoter, an anti-spasmodic agent, a protein synthesis inhibitor, an D-glucosidase inhibitor, a calcium channel blocker, a pyruvate dehydrogenase activator, a prostaglandin inhibitor, a sodium channel inhibitor, a serine protease inhibitor, an intracellular calcium flux inhibitor, a JAK2 inhibitor, an androgen inhibitor, an aromatase inhibitor, an anti-viral agent, a 5-HT inhibitor, an FXR antagonist, an actin polymerization and stabilization promoter, an AXOR12 agonist, an angiotensin Il receptor agonist, a platelet aggregation inhibitor, a CB1/CB2 receptor agonist, a norepinephrine reuptake inhibitor, a selective serotonin reuptake inhibitor, a reducing agent, and a immuno-modulator selected from Bay 11-7085, (-)- arctigenin, idazoxan hydrochloride.
40. The method of claim 35 or claim 36 wherein the anti- scarring agent is selected from an angiogenesis inhibitor, an apoptosis antagonist, an apoptosis activator, a beta 1 integrin antagonist, a beta tubulin inhibitor, a blocker of enzyme production in Hepatitis C, a Bruton's tyrosine kinase inhibitor, a calcineurin inhibitor, a caspase 3 inhibitor, a CC chemokine receptor antagonist, a cell cycle inhibitor, a cathepsin B inhibitor, a cathepsin K inhibitor, a cathepsin L inhibitor, a CD40 antagonist, a chemokine receptor antagonist, a chymase inhibitor, a collagenase antagonist, a CXCR antagonist, a cyclin dependent kinase inhibitor, a cyclooxygenase 1 inhibitor, a DHFR inhibitor, a cual integrin inhibitor, an elastase inhibitor, an elongation factor-1 alpha inhibitor, an endothelial growth factor antagonist, an endothelial growth factor receptor kinase inhibitor, an endotoxin antagonist, an epothilone and tubulin binder, an estrogen receptor antagonist, an FGF inhibitor, a farnexyl transferase inhibitor, a famesyltransferase inhibitor, an FLT-3 kinase inhibitor, an FGF receptor kinase inhibitor, a fibrinogen antagonist, a histone deacetylase inhibitor, an HMGCoA reductase inhibitor, an ICAM inhibitor, an IL, ICE, and IRAK antagonist, an lL-2 inhibitor, an immunosuppressant, an inosine monophosphate inhibitor, an integrin antagonist, an interleukin antagonist, an inhibitor of type III receptor tyrosine kinase, an irreversible inhibitor of enzyme methionine aminopeptidase type 2, an isozyme selective delta protein kinase C inhibitor, a JAK3 enzyme inhibitor, a JNK inhibitor, a kinase inhibitor, a kinesin antagonist, a leukotriene inhibitor and antagonist, a MAP kinase inhibitor, a matrix metalloproteinase inhibitor, an MCP-CCR2 inhibitor, an mTOR inhibitor, an mTOR kinase inhibitor, a microtubule inhibitor, an MIF inhibitor, a neurokinin antagonist, an NF kappa B inhibitor, a nitric oxide agonist, an ornithine decarboxylase inhibitor, a p38 MAP kinase inhibitor, a palmitoyl-protein thioesterase inhibitor, a PDGF receptor kinase inhibitor, a peroxisome proliferator-activated receptor (PPAR) agonist, a phosphatase inhibitor, a phosphodiesterase inhibitor, a PKC inhibitor, a platelet activating factor antagonist, a prolyl hydroxylase inhibitor, a polymorphonuclear neutrophil inhibitor, protein kinase B inhibitor, protein kinase C stimulant, purine nucleoside analogue, a purineoreceptor P2X antagonist, a Raf kinase inhibitor, reversible inhibitor of ErbB1 and ErbB2, ribonucleoside triphosphate reductase inhibitor, an SDF-1 antagonist, a sheddase inhibitor, an SRC inhibitor, a stromelysin inhibitor, an Syk kinase inhibitor, a telomerase inhibitor, a TGF beta inhibitor, a TNF-alpha antagonist or TACE inhibitor, a tumor necrosis factor antagonist, a Toll receptor inhibitor, a tubulin antagonist, a protein tyrosine kinase inhibitor, a VEGF inhibitor, and a vitamin D receptor agonist.
41. The method of claim 35 or claim 36 wherein the anti- scarring agent is selected from a retinoic acid receptor antagonist, a heat shock protein 90 antagonist, a steroid, a cell cycle inhibitor, a histone deacetylase inhibitor, an anti-microbial agent, an intracellular calcium flux inhibitor, an microtubule inhibitor, an HMGCoA reductase inhibitor, an actin polymerization and stabilization promoter, a tyrosine kinase inhibitor, a TGF beta inhibitor, a TNF-alpha antagonist, a TACE inhibitor, a calcineurin inhibitor, a peptidyl-prolyl cis/trans isomerase inhibitor, an apoptosis activator, an antimetabolite and antineoplastic agent, a TGF beta inhibitor, a DNA methylation promoter, and a PKC inhibitor.
42. The method of claim 35 or claim 36 wherein the anti- scarring agent is selected from ZD-6474, AP-23573, synthadotin, S-0885, aplidine, ixabepilone, IDN-5390, SB-2723005, ABT-518, combretastatin, anecortave acetate, SB-715992, temsirolimus, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, dolastatin 15, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17-DMAG, tacrolimus, loteprednol etabonate, juglone, prednisolone, puromycin, 3-BAABE, cladribine, mannose-6-phosphate, 5-azacytidine, Ly333531 (ruboxistaurin), and simvastatin.
43. The method of any one of claims 35-42 wherein the device comprises a coating wherein the coating comprises (a) the anti- scarring agent or (b) the anti-scarring agent and a polymer.
44. The method of any one of claims 35-42 wherein the device further comprises a polymer or a polymeric carrier.
45. The method of any one of claims 35-42 wherein the device further comprises a second pharmaceutically active agent.
46. The method of any one of claims 35-42 wherein the device further comprises at least one of an anti-inflammatory agent, an agent that inhibits infection, anthracycline, doxorubicin, mitoxantrone, fluoropyrimidine, 5-fluorouracil, a folic acid antagonist, methotrexate, podophylotoxin, etoposide, camptothecin, hydroxyurea, a platinum complex, cisplatin, an anti-thrombotic agent, a visualization agent, and an echogenic material.
47. The method of any one of claims 35-42 wherein the device is adapted for delivering the anti-scarring agent locally to tissue proximate to the device.
48. The method of any one of claims 35-42 wherein the anti-scarring agent is released into tissue in the vicinity of the device after deployment of the device.
49. The method of any one of claims 35-42 wherein the device comprises (a) about 0.01 μg to about 10 μg of the anti-scarring agent; (b) about 10 μg to about 10 mg of the anti-scarring agent; (c) about
10 mg to about 250 mg of the anti-scarring agent; (d) about 250 mg to about 1000 mg of the anti-scarring agent; or (e) about 1000 mg to about 2500 mg of the anti-scarring agent.
50. The method of any one of claims 35-42 wherein the device comprises (a) about 0.01 μg to about 1 μg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied; (b) about 1 μg to about 10 μg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied; (c) about 10 μg to about 250 μg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied; (d) about 250 μg to about 1000 μg of the anti- scarring agent per mm2 of device surface to which the anti-scarring agent is applied; or (e) about 1000 μg to about 2500 μg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
51. The method of any one of claims 35-42 wherein the device comprises (a) about 0.01 μg to about 100 μg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied; (b) about 0.01 μg to about 200 μg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied; or (c) about 0.01 μg to about 500 μg of the anti-scarring agent per mm2 of device surface to which the anti-scarring agent is applied.
52. A method for implanting a medical device comprising: (a) infiltrating a tissue of a host where the medical device is to be, or has been, implanted with i) an anti-scarring agent, ii) an anti-infective agent, iii) a polymer; iv) a composition comprising an anti-scarring agent and a polymer, v) a composition comprising an anti-infective agent and a polymer, or vi) a composition comprising an anti-scarring agent, an anti-infective agent and a polymer, and (b) implanting the medical device into the host.
53. The method of claim 52 wherein the medical device is an intravascular device, a gastrointestinal stent, a tracheal and bronchial stent, a genital urinary stent, an ear and nose stent, an ear ventilation tube, an intraocular implant, a medical device for treating hypertropic scar or keloid, a vascular graft, a hemodialysis access device, a medical device comprising a film or a mesh, a glaucoma drainage device, a prosthetic heart valve or component thereof, a penile implant, an endotracheal or tracheostomy tube, a peritoneal dialysis catheter, a central nervous system shunt or pressure monitor device, an inferior vena cava filter, a gastrointestinal device, a central venous catheter, a ventricular assist device, a spinal implant, a surgical adhesion barrier.
54. The method of claim 52 or claim 53 wherein the anti- scarring agent is an antimicrobial compound.
55. The method of claim 54 wherein the antimicrobial compound is brefeldin A.
56. The method of claim 52 or claim 53 wherein the anti- scarring agent is selected from a histamine receptor antagonist, an alpha adrenergic receptor antagonist, an anti-psychotic compound, a CaM kinase Il inhibitor, a G protein agonist, an antibiotic selected from the group consisting of apigenin, ampicillin sodium salt, puromycin, an anti-microbial agent, a DNA topoisomerase inhibitor, a thromboxane A2 receptor inhibitor, a D2 dopamine receptor antagonist, a Peptidyl-Prolyl Cis/Trans lsomerase Inhibitor, a dopamine antagonist, an anesthetic compound, a clotting factor, a lysyl hydrolase inhibitor, a muscarinic receptor inhibitor, a superoxide anion generator, a steroid, an anti-proliferative agent, a diuretic, an anti- coagulant, a cyclic GMP agonist, an adenylate cyclase agonist, an antioxidant, a nitric oxide synthase inhibitor, an anti-neoplastic agent, a DNA synthesis inhibitor, a DNA alkylating agent, a DNA methylation inhibitor, a NSAID agent, a peptidylglycine alpha-hydroxylating monooxygenase inhibitor, an MEK1/MEK 2 inhibitor, a NO synthase inhibitor, a retinoic acid receptor antagonist, an ACE inhibitor, a glycosylation inhibitor, an intracellular calcium influx inhibitor, an anti-emetic agent, an acetylcholinesterase inhibitor, an ALK-5 receptor antagonist, a RAR/RXT antagonist, an elF-2a inhibitor, an S-adenosyl-L-homocysteine hydrolase inhibitor, an estrogen agonist, a serotonin receptor inhibitor, an antithrombotic agent, a tryptase inhibitor, a pesticide, a bone mineralization promoter, a bisphosphonate compound selected from risedronate and an analogue or derivative thereof, an anti-inflammatory compound, a DNA methylation promoter, an anti-spasmodic agent, a protein synthesis inhibitor, an D-glucosidase inhibitor, a calcium channel blocker, a pyruvate dehydrogenase activator, a prostaglandin inhibitor, a sodium channel inhibitor, a serine protease inhibitor, an intracellular calcium flux inhibitor, a JAK2 inhibitor, an androgen inhibitor, an aromatase inhibitor, an anti-viral agent, a 5-HT inhibitor, an FXR antagonist, an actin polymerization and stabilization promoter, an AXOR12 agonist, an angiotensin Il receptor agonist, a platelet aggregation inhibitor, a CB1/CB2 receptor agonist, a norepinephrine reuptake inhibitor, a selective serotonin reuptake inhibitor, a reducing agent, and a immuno-modulator selected from Bay 11-7085, (-)- arctigenin, idazoxan hydrochloride.
57. The method of claim 52 or claim 53 wherein the anti- scarring agent is selected from an angiogenesis inhibitor, an apoptosis antagonist, an apoptosis activator, a beta 1 integrin antagonist, a beta tubulin inhibitor, a blocker of enzyme production in Hepatitis C, a Bruton's tyrosine kinase inhibitor, a calcineurin inhibitor, a caspase 3 inhibitor, a CC chemokine receptor antagonist, a cell cycle inhibitor, a cathepsin B inhibitor, a cathepsin K inhibitor, a cathepsin L inhibitor, a CD40 antagonist, a chemokine receptor antagonist, a chymase inhibitor, a collagenase antagonist, a CXCR antagonist, a cyclin dependent kinase inhibitor, a cyclooxygenase 1 inhibitor, a DHFR inhibitor, a cual integrin inhibitor, an elastase inhibitor, an elongation factor-1 alpha inhibitor, an endothelial growth factor antagonist, an endothelial growth factor receptor kinase inhibitor, an endotoxin antagonist, an epothilone and tubulin binder, an estrogen receptor antagonist, an FGF inhibitor, a farnexyl transferase inhibitor, a farnesyltransferase inhibitor, an FLT-3 kinase inhibitor, an FGF receptor kinase inhibitor, a fibrinogen antagonist, a histone deacetylase inhibitor, an HMGCoA reductase inhibitor, an ICAM inhibitor, an IL, ICE, and IRAK antagonist, an IL-2 inhibitor, an immunosuppressant, an inosine monophosphate inhibitor, an integrin antagonist, an interleukin antagonist, an inhibitor of type III receptor tyrosine kinase, an irreversible inhibitor of enzyme methionine aminopeptidase type 2, an isozyme selective delta protein kinase C inhibitor, a JAK3 enzyme inhibitor, a JNK inhibitor, a kinase inhibitor, a kinesin antagonist, a leukotriene inhibitor and antagonist, a MAP kinase inhibitor, a matrix metalloproteinase inhibitor, an MCP-CCR2 inhibitor, an mTOR inhibitor, an mTOR kinase inhibitor, a microtubule inhibitor, an MIF inhibitor, a neurokinin antagonist, an NF kappa B inhibitor, a nitric oxide agonist, an ornithine decarboxylase inhibitor, a p38 MAP kinase inhibitor, a palmitoyl-protein thioesterase inhibitor, a PDGF receptor kinase inhibitor, a peroxisome proliferator-activated receptor (PPAR) agonist, a phosphatase inhibitor, a phosphodiesterase inhibitor, a PKC inhibitor, a platelet activating factor antagonist, a prolyl hydroxylase inhibitor, a polymorphonuclear neutrophil inhibitor, protein kinase B inhibitor, protein kinase C stimulant, purine nucleoside analogue, a purineoreceptor P2X antagonist, a Raf kinase inhibitor, reversible inhibitor of ErbB1 and ErbB2, ribonucleoside triphosphate reductase inhibitor, an SDF-1 antagonist, a sheddase inhibitor, an SRC inhibitor, a stromelysin inhibitor, an Syk kinase inhibitor, a telomerase inhibitor, a TGF beta inhibitor, a TNF-alpha antagonist or TACE inhibitor, a tumor necrosis factor antagonist, a Toll receptor inhibitor, a tubulin antagonist, a protein tyrosine kinase inhibitor, a VEGF inhibitor, and a vitamin D receptor agonist.
58. The method of claim 52 or claim 53 wherein the anti- scarring agent is selected from a retinoic acid receptor antagonist, a heat shock protein 90 antagonist, a steroid, a cell cycle inhibitor, a histone deacetylase inhibitor, an anti-microbial agent, an intracellular calcium flux inhibitor, an microtubule inhibitor, an HMGCoA reductase inhibitor, an actin polymerization and stabilization promoter, a tyrosine kinase inhibitor, a TGF beta inhibitor, a TNF-alpha antagonist, a TACE inhibitor, a calcineurin inhibitor, a peptidyl-prolyl cis/trans isomerase inhibitor, an apoptosis activator, an antimetabolite and anti-neoplastic agent, a TGF beta inhibitor, a DNA methylation promoter, and a PKC inhibitor.
59. The method of claim 52 or claim 53 wherein the anti- scarring agent is selected from ZD-6474, AP-23573, synthadotin, S-0885, aplidine, ixabepilone, IDN-5390, SB-2723005, ABT-518, combretastatin, anecortave acetate, SB-715992, temsirolimus, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, dolastatin 15, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17-DMAG, tacrolimus, loteprednol etabonate, juglone, prednisolone, puromycin, 3-BAABE, cladribine, mannose-6-phosphate, 5-azacytidine, Ly333531 (ruboxistaurin), and simvastatin.
60. The method of any one of claims 52-59 wherein the anti-infective agent is selected from an anthracycline, doxorubicin, mitoxantrone, fluoropyrimidine, 5-fluorouracil, a folic acid antagonist, methotrexate, podophylotoxin, etoposide, camptothecin, hydroxyurea, a platinum complex, and cisplatin.
61. The method of any one of claims 52-59 wherein the composition comprises an anti-thrombotic agent.
62. The method of any one of claims 52-59 wherein the polymer is formed from reactants comprising a naturally occurring polymer; protein; carbohydrate; biodegradable polymer; nonbiodegradable polymer; collagen; methylated collagen; fibrinogen; thrombin; blood plasma; calcium salt; an antifibronolytic agent; fibrinogen analog; albumin; plasminogen; von Willebrands factor; factor VIII; hypoallergenic collagen; atelopeptidic collagen; crosslinked collagen; aprotinin; epsilon-amino-n-caproic acid; gelatin; protein conjugates; gelatin conjugates; a synthetic polymer; isocyanate-containing compound; a synthetic thiol-containing compound; a synthetic compound containing at least two thiol groups; a synthetic compound containing at least three thiol groups; a synthetic compound containing at least four thiol groups; a synthetic amino-containing compound; a synthetic compound containing at least two amino groups; a synthetic compound containing at least three amino groups; a synthetic compound containing at least four amino groups; a synthetic compound comprising a carbonyl-oxygen-succinimidyl group; a synthetic compound comprising at least two carbonyl-oxygen-succinimidyl groups; a synthetic compound comprising at least three carbonyl-oxygen-succinimidyl groups; a synthetic compound comprising at least four carbonyl-oxygen-succinimidyl groups; a synthetic polyalkylene oxide-containing compound; a synthetic compound comprising both polyalkylene oxide and biodegradable polyester blocks; a synthetic polyalkylene oxide-containing compound having reactive amino groups; a synthetic polyalkylene oxide-containing compound having reactive thiol groups; a synthetic polyalkylene oxide-containing compound having reactive carbonyl-oxygen-succinimidyl groups; a synthetic compound comprising a biodegradable polyester block; a synthetic polymer formed in whole or part from lactic acid or lactide; a synthetic polymer formed in whole or part from glycolic acid or glycolide; polylysine; (a) protein and (b) a compound comprising a polyalkylene oxide portion; (a) protein and (b) polylysine; (a) protein and (b) a compound having at least four thiol groups;
(a) protein and (b) a compound having at least four amino groups; (a) protein and (b) a compound having at least four carbonyl-oxygen- succinimide groups; (a) protein and (b) a compound having a biodegradable region formed from reactants selected from lactic acid, lactide, glycolic acid, glycolide, and epison-caprolactone; (a) collagen and (b) a compound comprising a polyalkylene oxide portion; (a) collagen and (b) polylysine; (a) collagen and (b) a compound having at least four thiol groups; (a) collagen and (b) a compound having at least four thiol groups; (a) collagen and (b) a compound having at least four amino groups; (a) collagen and (b) a compound having at least four carbonyl-oxygen-succinimide groups; (a) collagen and (b) a compound having a biodegradable region formed from reactants selected from lactic acid, lactide, glycolic acid, glycolide, and epison-caprolactone; (a) methylated collagen and (b) a compound comprising a polyalkylene oxide portion; (a) methylated collagen and (b) polylysine; (a) methylated collagen and (b) a compound having at least four thiol groups; (a) methylated collagen and (b) a compound having at least four amino groups; (a) methylated collagen and (b) a compound having at least four carbonyl-oxygen-succinimide groups; (a) methylated collagen and
(b) a compound having a biodegradable region formed from reactants selected from lactic acid, lactide, glycolic acid, glycolide, and epison- caprolactone; hyaluronic acid; a hyaluronic acid derivative; pentaerythritol poly(ethylene glycol)ether tetra-sulfhydryl of number average molecular weight between 3,000 and 30,000; pentaerythritol poly(ethylene glycol)ether tetra-amino of number average molecular weight between 3,000 and 30,000; or (a) a synthetic compound having a number average molecular weight between 3,000 and 30,000 and comprising a polyalkylene oxide region and multiple nucleophilic groups, and (b) a synthetic compound having a number average molecular weight between 3,000 and 30,000 and comprising a polyalkylene oxide region and multiple electrophilic groups.
63. A method for reducing or preventing surgical adhesions, comprising delivering a tissue-reactive polymeric composition to a site in need thereof to provide coated tissue, and delivering an anti-scarring agent to the coated tissue.
64. A method for reducing or preventing surgical adhesions, comprising delivering (a) an anti-scarring agent, or (b) a composition that comprises (i) an anti-scarring agent and (ii) a polymer and/or a compound that forms a polymer in situ.
65. The method of claim 63 or claim 64, wherein the site in need thereof is between a dural sleeve and paravertebral musculature in a patient post-laminectomy, a spinal nerve at a laminectomy site, a tissue around a spinal nerve at a laminectomy site, a site of a surgical disc resection, a site of a microdiscectomy, a site of a neurosurgical (brain) procedure, a spinal surgical site, an epidural tissue, a dural tissue, a gynecological site, a tissue surface of the pelvic side wall, a peritoneal cavity, a pelvic cavity, a site of a laparotomy, a site of an endoscopic procedure, a site of a hernia repair, a site of cholecystectomy, a site of a cardiac procedure, a site of cardiac transplant surgery, a site of cardiac vascular repair, a site of a heart valve replacement, a site of pericardial surgery, a site of an orthopedic surgical procedure, a site of a torn ligament, a site of a joint injury, a site of a tendon injury, a site of a cartilage injury, a site of a muscle injury, a site of a nerve injury, a site of a cosmetic surgical procedure, a site of a reconstructive surgical procedure, a site of a breast implant in a patient in need thereof, where the composition prevents surgical adhesions.
66. The method of any one of claims 63-65 wherein the anti-scarring agent is an antimicrobial compound.
67. The method of any one of claims 63-65 wherein the antimicrobial compound is brefeldin A.
68. The method of any one of claims 63-65 wherein the anti-scarring agent is selected from a histamine receptor antagonist, an alpha adrenergic receptor antagonist, an anti-psychotic compound, a CaM kinase Il inhibitor, a G protein agonist, an antibiotic selected from the group consisting of apigenin, ampicillin sodium salt, puromycin, an anti-microbial agent, a DNA topoisomerase inhibitor, a thromboxane A2 receptor inhibitor, a D2 dopamine receptor antagonist, a Peptidyl-Prolyl Cis/Trans lsomerase Inhibitor, a dopamine antagonist, an anesthetic compound, a clotting factor, a lysyl hydrolase inhibitor, a muscarinic receptor inhibitor, a superoxide anion generator, a steroid, an antiproliferative agent, a diuretic, an anticoagulant, a cyclic GMP agonist, an adenylate cyclase agonist, an antioxidant, a nitric oxide synthase inhibitor, an anti-neoplastic agent, a DNA synthesis inhibitor, a DNA alkylating agent, a DNA methylation inhibitor, a NSAID agent, a peptidylglycine alpha-hydroxylating monooxygenase inhibitor, an MEK1/MEK 2 inhibitor, a NO synthase inhibitor, a retinoic acid receptor antagonist, an ACE inhibitor, a glycosylation inhibitor, an intracellular calcium influx inhibitor, an anti-emetic agent, an acetylcholinesterase inhibitor, an ALK-5 receptor antagonist, a RAR/RXT antagonist, an elF-2a inhibitor, an S-adenosyl-L-homocysteine hydrolase inhibitor, an estrogen agonist, a serotonin receptor inhibitor, an antithrombotic agent, a tryptase inhibitor, a pesticide, a bone mineralization promoter, a bisphosphonate compound selected from risedronate and an analogue or derivative thereof, an anti-inflammatory compound, a DNA methylation promoter, an anti-spasmodic agent, a protein synthesis inhibitor, an D-glucosidase inhibitor, a calcium channel blocker, a pyruvate dehydrogenase activator, a prostaglandin inhibitor, a sodium channel inhibitor, a serine protease inhibitor, an intracellular calcium flux inhibitor, a JAK2 inhibitor, an androgen inhibitor, an aromatase inhibitor, an anti-viral agent, a 5-HT inhibitor, an FXR antagonist, an actin polymerization and stabilization promoter, an AXOR12 agonist, an angiotensin Il receptor agonist, a platelet aggregation inhibitor, a CB1/CB2 receptor agonist, a norepinephrine reuptake inhibitor, a selective serotonin reuptake inhibitor, a reducing agent, and a immuno-modulator selected from Bay 11-7085, (-)- arctigenin, idazoxan hydrochloride.
69. The method of any one of claims 63-65 wherein the anti-scarring agent is selected from an angiogenesis inhibitor, an apoptosis antagonist, an apoptosis activator, a beta 1 integrin antagonist, a beta tubulin inhibitor, a blocker of enzyme production in Hepatitis C, a Bruton's tyrosine kinase inhibitor, a calcineurin inhibitor, a caspase 3 inhibitor, a CC chemokine receptor.antagonist, a cell cycle inhibitor, a cathepsin B inhibitor, a cathepsin K inhibitor, a cathepsin L inhibitor, a CD40 antagonist, a chemokine receptor antagonist, a chymase inhibitor, a collagenase antagonist, a CXCR antagonist, a cyclin dependent kinase inhibitor, a cyclooxygenase 1 inhibitor, a DHFR inhibitor, a cual integrin inhibitor, an elastase inhibitor, an elongation factor-1 alpha inhibitor, an endothelial growth factor antagonist, an endothelial growth factor receptor kinase inhibitor, an endotoxin antagonist, an epothilone and tubulin binder, an estrogen receptor antagonist, an FGF inhibitor, a farnexyl transferase inhibitor, a famesyltransferase inhibitor, an FLT-3 kinase inhibitor, an FGF receptor kinase inhibitor, a fibrinogen antagonist, a histone deacetylase inhibitor, an HMGCoA reductase inhibitor, an ICAM inhibitor, an IL, ICE, and IRAK antagonist, an IL-2 inhibitor, an immunosuppressant, an inosine monophosphate inhibitor, an integrin antagonist, an interleukin antagonist, an inhibitor of type III receptor tyrosine kinase, an irreversible inhibitor of enzyme methionine aminopeptidase type 2, an isozyme selective delta protein kinase C inhibitor, a JAK3 enzyme inhibitor, a JNK inhibitor, a kinase inhibitor, a kinesin antagonist, a leukotriene inhibitor and antagonist, a MAP kinase inhibitor, a matrix metalloproteinase inhibitor, an MCP-CCR2 inhibitor, an mTOR inhibitor, an mTOR kinase inhibitor, a microtubule inhibitor, an MIF inhibitor, a neurokinin antagonist, an NF kappa B inhibitor, a nitric oxide agonist, an ornithine decarboxylase inhibitor, a p38 MAP kinase inhibitor, a palmitoyl-protein thioesterase inhibitor, a PDGF receptor kinase inhibitor, a peroxisome proliferator-activated receptor (PPAR) agonist, a phosphatase inhibitor, a phosphodiesterase inhibitor, a PKC inhibitor, a platelet activating factor antagonist, a prolyl hydroxylase inhibitor, a polymorphonuclear neutrophil inhibitor, protein kinase B inhibitor, protein kinase C stimulant, purine nucleoside analogue, a purineoreceptor P2X antagonist, a Raf kinase inhibitor, reversible inhibitor of ErbB1 and ErbB2, ribonucleoside triphosphate reductase inhibitor, an SDF- 1 antagonist, a sheddase inhibitor, an SRC inhibitor, a stromelysin inhibitor, an Syk kinase -inhibitor, a telomerase inhibitor, a TGF beta inhibitor, a TNF-alpha antagonist or TACE inhibitor, a tumor necrosis factor antagonist, a Toll receptor inhibitor, a tubulin antagonist, a protein tyrosine kinase inhibitor, a VEGF inhibitor, and a vitamin D receptor agonist.
70. The method of any one of claims 63-65 wherein the anti-scarring agent is selected from a retinoic acid receptor antagonist, a heat shock protein 90 antagonist, a steroid, a cell cycle inhibitor, a histone deacetylase inhibitor, an anti-microbial agent, an intracellular calcium flux inhibitor, an microtubule inhibitor, an HMGCoA reductase inhibitor, an actin polymerization and stabilization promoter, a tyrosine kinase inhibitor, a TGF beta inhibitor, a TNF-alpha antagonist, a TACE inhibitor, a calcineurin inhibitor, a peptidyl-prolyl cis/trans isomerase inhibitor, an apoptosis activator, an antimetabolite and anti-neoplastic agent, a TGF beta inhibitor, a DNA methylation promoter, and a PKC inhibitor.
71. The method of any one of claims 63-65 wherein the anti-scarring agent is selected from ZD-6474, AP-23573, synthadotin, S- 0885, aplidine, ixabepilone, IDN-5390, SB-2723005, ABT-518, combretastatin, anecortave acetate, SB-715992, terήsirolimus, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, dolastatin 15, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17-DMAG, tacrolimus, loteprednol etabonate, juglone, prednisolone, puromycin, 3-BAABE, cladribine, mannose-6-phosphate, 5-azacytidine, Ly333531 (ruboxistaurin), and simvastatin.
72. The method of any one of claims 63-71 wherein the composition further comprises an anti-infective agent selected from an anthracycline, doxorubicin, mitoxantrone, fluoropyrimidine, 5-fluorouracil, a folic acid antagonist, methotrexate, podophylotoxin, etoposide, camptothecin, hydroxyurea, a platinum complex, and cisplatin.
73. The method of any one of claims 63-71 wherein the composition further comprises an anti-thrombotic agent.
74. The method of any one of claims 63-71 wherein the composition comprises a naturally occurring polymer; protein; carbohydrate; biodegradable polymer; nonbiodegradable polymer; collagen; methylated collagen; fibrinogen; thrombin; blood plasma; calcium salt; an antifibronolytic agent; fibrinogen analog; albumin; plasminogen; von Willebrands factor; factor VIII; hypoallergenic collagen; atelopeptidic collagen; crosslinked collagen; aprotinin; epsilon-amino-n-caproic acid; gelatin; protein conjugates; gelatin conjugates; a synthetic polymer; isocyanate-containing compound; a synthetic thiol-containing compound; a synthetic compound containing at least two thiol groups; a synthetic compound containing at least three thiol groups; a synthetic compound containing at least four thiol groups; a synthetic amino-containing compound; a synthetic compound containing at least two amino groups; a synthetic compound containing at least three amino groups; a synthetic compound containing at least four amino groups; a synthetic compound comprising a carbonyl-oxygen- succinimidyl group; a synthetic compound comprising at least two carbonyl- oxygen-succinimidyl groups; a synthetic compound comprising at least three carbonyl-oxygen-succinimidyl groups; a synthetic compound comprising at least four carbonyl-oxygen-succinimidyl groups; a synthetic polyalkylene oxide-containing compound; a synthetic compound comprising both polyalkylene oxide and biodegradable polyester blocks; a synthetic polyalkylene oxide-containing compound having reactive amino groups; a synthetic polyalkylene oxide-containing compound having reactive thiol groups; a synthetic polyalkylene oxide-containing compound having reactive carbonyl-oxygen-succinimidyl groups; a synthetic compound comprising a biodegradable polyester block; a synthetic polymer formed in whole or part from lactic acid or lactide; a synthetic polymer formed in whole or part from glycolic acid or glycolide; polylysine; (a) protein and (b) a compound comprising a polyalkylene oxide portion; (a) protein and (b) polylysine; (a) protein and (b) a compound having at least four thiol groups; (a) protein and (b) a compound having at least four amino groups; (a) protein and (b) a compound having at least four carbonyl-oxygen-succinimide groups; (a) protein and (b) a compound having a biodegradable region formed from reactants selected from lactic acid, lactide, glycolic acid, glycolide, and epison-caprolactone; (a) collagen and (b) a compound comprising a polyalkylene oxide portion; (a) collagen and (b) polylysine; (a) collagen and (b) a compound having at least four thiol groups; (a) collagen and (b) a compound having at least four thiol groups; (a) collagen and (b) a compound having at least four amino groups; (a) collagen and (b) a compound having at least four carbonyl-oxygen-succinimide groups; (a) collagen and (b) a compound having a biodegradable region formed from reactants selected from lactic acid, lactide, glycolic acid, glycolide, and epison-caprolactone; (a) methylated collagen and (b) a compound comprising a polyalkylene oxide portion; (a) methylated collagen and (b) polylysine; (a) methylated collagen and (b) a compound having at least four thiol groups; (a) methylated collagen and (b) a compound having at least four amino groups; (a) methylated collagen and (b) a compound having at least four carbonyl-oxygen-succinimide groups; (a) methylated collagen and (b) a compound having a biodegradable region formed from reactants selected from lactic acid, lactide, glycolic acid, glycolide, and epison- caprolactone; hyaluronic acid; a hyaluronic acid derivative; pentaerythritol poly(ethylene glycol)ether tetra-sulfhydryl of number average molecular weight between 3,000 and 30,000; pentaerythritol poly(ethylene glycol)ether tetra-amino of number average molecular weight between 3,000 and 30,000; or (a) a synthetic compound having a number average molecular weight between 3,000 and 30,000 and comprising a polyalkylene oxide region and multiple nucleophilic groups, and (b) a synthetic compound having a number average molecular weight between 3,000 and 30,000 and comprising a polyalkylene oxide region and multiple electrophilic groups.
75. A method for treating or preventing inflammatory arthritis, comprising delivering to a patient in need thereof (a) an anti- scarring agent, or (b) a therapeutic composition comprising (i) a polymer and/or a compound that forms a polymer in situ and (ii) an anti-scarring agent.
76. The method of claim 75, wherein the inflammatory arthritis is osteoarthritis, primary osteoarthritis, secondary osteoarthritis, or rheumatoid arthritis.
77. The method of claim 75 or claim 76, wherein the agent or the composition is delivered intravenously, orally, by subcutaneous injection, by intramuscular injection, or intra-articularly.
78. The method of any one of claims 75-77 wherein the anti-scarring agent is an antimicrobial compound.
79. The method of any one of claims 75-77 wherein the antimicrobial compound is brefeldin A.
80. The method of any one of claims 75-77 wherein the anti-scarring agent is selected from a histamine receptor antagonist, an alpha adrenergic receptor antagonist, an anti-psychotic compound, a CaM kinase Il inhibitor, a G protein agonist, an antibiotic selected from the group consisting of apigenin, ampicillin sodium salt, puromycin, an anti-microbial agent, a DNA topoisomerase inhibitor, a thromboxane A2 receptor inhibitor, a D2 dopamine receptor antagonist, a Peptidyl-Prolyl Cis/Trans lsomerase Inhibitor, a dopamine antagonist, an anesthetic compound, a clotting factor, a lysyl hydrolase inhibitor, a muscarinic receptor inhibitor, a superoxide anion generator, a steroid, an anti-proliferative agent, a diuretic, an anticoagulant, a cyclic GMP agonist, an adenylate cyclase agonist, an antioxidant, a nitric oxide synthase inhibitor, an anti-neoplastic agent, a DNA synthesis inhibitor, a DNA alkylating agent, a DNA methylation inhibitor, a NSAID agent, a peptidylglycine alpha-hydroxylating monooxygenase inhibitor, an MEK1/MEK 2 inhibitor, a NO synthase inhibitor, a retinoic acid receptor antagonist, an ACE inhibitor, a glycosylation inhibitor, an intracellular calcium influx inhibitor, an anti-emetic agent, an acetylcholinesterase inhibitor, an ALK-5 receptor antagonist, a RAR/RXT antagonist, an elF-2a inhibitor, an S-adenosyl-L-homocysteine hydrolase inhibitor, an estrogen agonist, a serotonin receptor inhibitor, an antithrombotic agent, a tryptase inhibitor, a pesticide, a bone mineralization promoter, a bisphosphonate compound selected from risedronate and an analogue or derivative thereof, an anti-inflammatory compound, a DNA methylation promoter, an anti-spasmodic agent, a protein synthesis inhibitor, an D-glucosidase inhibitor, a calcium channel blocker, a pyruvate dehydrogenase activator, a prostaglandin inhibitor, a sodium channel inhibitor, a serine protease inhibitor, an intracellular calcium flux inhibitor, a JAK2 inhibitor, an androgen inhibitor, an aromatase inhibitor, an anti-viral agent, a 5-HT inhibitor, an FXR antagonist, an actin polymerization and stabilization promoter, an AXOR12 agonist, an angiotensin Il receptor agonist, a platelet aggregation inhibitor, a CB1/CB2 receptor agonist, a norepinephrine reuptake inhibitor, a selective serotonin reuptake inhibitor, a reducing agent, and a immuno-modulator selected from Bay 11-7085, (-)- arctigenin, idazoxan hydrochloride.
81. The method of any one of claims 75-77 wherein the anti-scarring agent is selected from an angiogenesis inhibitor, an apoptosis antagonist, an apoptosis activator, a beta 1 integrin antagonist, a beta tubulin inhibitor, a blocker of enzyme production in Hepatitis C, a Bruton's tyrosine kinase inhibitor, a calcineurin inhibitor, a caspase 3 inhibitor, a CC chemokine receptor antagonist, a cell cycle inhibitor, a cathepsin B inhibitor, a cathepsin K inhibitor, a cathepsin L inhibitor, a CD40 antagonist, a chemokine receptor antagonist, a chymase inhibitor, a collagenase antagonist, a CXCR antagonist, a cyclin dependent kinase inhibitor, a cyclooxygenase 1 inhibitor, a DHFR inhibitor, a cual integrin inhibitor, an elastase inhibitor, an elongation factor-1 alpha inhibitor, an endothelial growth factor antagonist, an endothelial growth factor receptor kinase inhibitor, an endotoxin antagonist, an epothilone and tubulin binder, an estrogen receptor antagonist, an FGF inhibitor, a farnexyl transferase inhibitor, a farnesyltransferase inhibitor, an FLT-3 kinase inhibitor, an FGF receptor kinase inhibitor, a fibrinogen antagonist, a histone deacetylase inhibitor, an HMGCoA reductase inhibitor, an ICAM inhibitor, an IL, ICE, and IRAK antagonist, an IL-2 inhibitor, an immunosuppressant, an inosine monophosphate inhibitor, an integrin antagonist, an interleukin antagonist, an inhibitor of type III receptor tyrosine kinase, an irreversible inhibitor of enzyme methionine aminopeptidase type 2, an isozyme selective delta protein kinase C inhibitor, a JAK3 enzyme inhibitor, a JNK inhibitor, a kinase inhibitor, a kinesin antagonist, a leukotriene inhibitor and antagonist, a MAP kinase inhibitor, a matrix metalloproteinase inhibitor, an MCP-CCR2 inhibitor, an mTOR inhibitor, an mTOR kinase inhibitor, a microtubule inhibitor, an MIF inhibitor, a neurokinin antagonist, an NF kappa B inhibitor, a nitric oxide agonist, an ornithine decarboxylase inhibitor, a p38 MAP kinase inhibitor, a palmitoyl-protein thioesterase inhibitor, a PDGF receptor kinase inhibitor, a peroxisome proliferator-activated receptor (PPAR) agonist, a phosphatase inhibitor, a phosphodiesterase inhibitor, a PKC inhibitor, a platelet activating factor antagonist, a prolyl hydroxylase inhibitor, a polymorphonuclear neutrophil inhibitor, protein kinase B inhibitor, protein kinase C stimulant, purine nucleoside analogue, a purineoreceptor P2X antagonist, a Raf kinase inhibitor, reversible inhibitor of ErbB1 and ErbB2, ribonucleoside triphosphate reductase inhibitor, an SDF-1 antagonist, a sheddase inhibitor, an SRC inhibitor, a stromelysin inhibitor, an Syk kinase inhibitor, a telomerase inhibitor, a TGF beta inhibitor, a TNF-alpha antagonist or TACE inhibitor, a tumor necrosis factor antagonist, a Toll receptor inhibitor, a tubulin antagonist, a protein tyrosine kinase inhibitor, a VEGF inhibitor, and a vitamin D receptor agonist.
82. The method of any one of claims 75-77 wherein the anti-scarring agent is selected from a retinoic acid receptor antagonist, a heat shock protein 90 antagonist, a steroid, a cell cycle inhibitor, a histone deacetylase inhibitor, an anti-microbial agent, an intracellular calcium flux inhibitor, an microtubule inhibitor, an HMGCoA reductase inhibitor, an actin polymerization and stabilization promoter, a tyrosine kinase inhibitor, a TGF beta inhibitor, a TNF-alpha antagonist, a TACE inhibitor, a calcineurin inhibitor, a peptidyl-prolyl cis/trans isomerase inhibitor, an apoptosis activator, an antimetabolite and anti-neoplastic agent, a TGF beta inhibitor, a DNA methylation promoter, and a PKC inhibitor.
83. The method of any one of claims 75-77 wherein the anti-scarring agent is selected from ZD-6474, AP-23573, synthadotin, S- 0885, aplidine, ixabepilone, IDN-5390, SB-2723005, ABT-518, combretastatin, anecortave acetate, SB-715992, temsirolimus, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin(A, thapsigargin, dolastatin 15, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17-DMAG, tacrolimus, loteprednol etabonate, juglone, prednisolone, puromycin, 3-BAABE, cladribine, mannose-6-phosphate, 5-azacytidine, Ly333531 (ruboxistaurin), and simvastatin.
84. The method of any one of claims 75-83 wherein the composition further comprises an anti-infective agent selected from an anthracycline, doxorubicin, mitoxantrone, fluoropyrimidine, 5-fluorouracil, a folic acid antagonist, methotrexate, podophylotoxin, etoposide, camptothecin, hydroxyurea, a platinum complex, and cisplatin.
85. The method of any one of claims 75-83 wherein the composition further comprises an anti-thrombotic agent.
86. The method of any one of claims 75-83 wherein the composition comprises a naturally occurring polymer; protein; carbohydrate; biodegradable polymer; nonbiodegradable polymer; collagen; methylated collagen; fibrinogen; thrombin; blood plasma; calcium salt; an antifibronolytic agent; fibrinogen analog; albumin; plasminogen; von Willebrands factor; factor VIII; hypoallergenic collagen; atelopeptidic collagen; crosslinked collagen; aprotinin; epsilon-amino-n-caproic acid; gelatin; protein conjugates; gelatin conjugates; a synthetic polymer; isocyanate-containing compound; a synthetic thiol-containing compound; a synthetic compound containing at least two thiol groups; a synthetic compound containing at least three thiol groups; a synthetic compound containing at least four thiol groups; a synthetic amino-containing compound; a synthetic compound containing at least two amino groups; a synthetic compound containing at least three amino groups; a synthetic compound containing at least four amino groups; a synthetic compound comprising a carbonyl-oxygen- succinimidyl group; a synthetic compound comprising at least two carbonyl- oxygen-succinimidyl groups; a synthetic compound comprising at least three carbonyl-oxygen-succinimidyl groups; a synthetic compound comprising at least four carbonyl-oxygen-succinimidyl groups; a synthetic polyalkylene oxide-containing compound; a synthetic compound comprising both polyalkylene oxide and biodegradable polyester blocks; a synthetic polyalkylene oxide-containing compound having reactive amino groups; a synthetic polyalkylene oxide-containing compound having reactive thiol groups; a synthetic polyalkylene oxide-containing compound having reactive carbonyl-oxygen-succinimidyl groups; a synthetic compound comprising a biodegradable polyester block; a synthetic polymer formed in whole or part from lactic acid or lactide; a synthetic polymer formed in whole or part from glycolic acid or glycolide; polylysine; (a) protein and (b) a compound comprising a polyalkylene oxide portion; (a) protein and (b) polylysine; (a) protein and (b) a compound having at least four thiol groups; (a) protein and (b) a compound having at least four amino groups; (a) protein and (b) a compound having at least four carbonyl-oxygen-succinimide groups; (a) protein and (b) a compound having a biodegradable region formed from reactants selected from lactic acid, lactide, glycolic acid, glycolide, and epison-caprolactone; (a) collagen and (b) a compound comprising a polyalkylene oxide portion; (a) collagen and (b) polylysine; (a) collagen and (b) a compound having at least four thiol groups; (a) collagen and (b) a compound having at least four thiol groups; (a) collagen and (b) a compound having at least four amino groups; (a) collagen and (b) a compound having at least four carbonyl-oxygen-succinimide groups; (a) collagen and (b) a compound having a biodegradable region formed from reactants selected from lactic acid, lactide, glycolic acid, glycolide, and epison-caprolactone; (a) methylated collagen and (b) a compound comprising a polyalkylene oxide portion; (a) methylated collagen and (b) polylysine; (a) methylated collagen and (b) a compound having at least four thiol groups; (a) methylated collagen and (b) a compound having at least four amino groups; (a) methylated collagen and (b) a compound having at least four carbonyl-oxygen-succinimide groups; (a) methylated collagen and (b) a compound having a biodegradable region formed from reactants selected from lactic acid, lactide, glycolic acid, glycolide, and epison- caprolactone; hyaluronic acid; a hyaluronic acid derivative; pentaerythritol poly(ethylene glycol)ether tetra-sulfhydryl of number average molecular _weight between 3,000 and 30,000; pentaerythritol poly(ethylene glycol)ether tetra-amino of number average molecular weight between 3,000 and 30,000; or (a) a synthetic compound having a number average molecular weight between 3,000 and 30,000 and comprising a polyalkylene oxide region and multiple nucleophilic groups, and (b) a synthetic compound having a number average molecular weight between 3,000 and 30,000 and comprising a polyalkylene oxide region and multiple electrophilic groups.
87. A method for reducing or preventing cartilage loss following an injury to a joint in a patient in need thereof, comprising delivering to the patient a) an anti-scarring agent or b) a composition comprising (i) an anti-scarring agent and (ii) a polymer and/or a compound that forms a polymer in situ.
88. The method of claim 87, wherein the injury is a cruciate ligament tear or a meniscal tear.
89. The method of claim 87 or claim 88 wherein the agent or the composition is delivered intra-articularly.
90. The method of any one of claims 87-89 wherein the anti-scarring agent is an antimicrobial compound.
91. The method of any one of claims 87-89 wherein the antimicrobial compound is brefeldin A.
92. The method of any one of claims 87-89 wherein the anti-scarring agent is selected from a histamine receptor antagonist, an alpha adrenergic receptor antagonist, an anti-psychotic compound, a CaM kinase Il inhibitor, a G protein agonist, an antibiotic selected from the group consisting of apigenin, ampicillin sodium salt, puromycin, an anti-microbial agent, a DNA topoisomerase inhibitor, a thromboxane A2 receptor inhibitor, a D2 dopamine receptor antagonist, a Peptidyl-Prolyl Cis/Trans lsomerase Inhibitor, a dopamine antagonist, an anesthetic compound, a clotting factor, a lysyl hydrolase inhibitor, a muscarinic receptor inhibitor, a superoxide anion generator, a steroid, an anti-proliferative agent, a diuretic, an anticoagulant, a cyclic GMP agonist, an adenylate cyclase agonist, an antioxidant, a nitric oxide synthase inhibitor, an anti-neoplastic agent, a DNA synthesis inhibitor, a DNA alkylating agent, a DNA methylation inhibitor, a NSAID agent, a peptidylglycine alpha-hydroxylating monooxygenase inhibitor, an MEK1/MEK 2 inhibitor, a NO synthase inhibitor, a retinoic acid receptor antagonist, an ACE inhibitor, a glycosylation inhibitor, an intracellular calcium influx inhibitor, an anti-emetic agent, an acetylcholinesterase inhibitor, an ALK-5 receptor antagonist, a RAR/RXT antagonist, an elF-2a inhibitor, an S-adenosyl-L-homocysteine hydrolase inhibitor, an estrogen agonist, a serotonin receptor inhibitor, an antithrombotic agent, a tryptase inhibitor, a pesticide, a bone mineralization promoter, a bisphosphonate compound selected from risedronate and an analogue or derivative thereof, an anti-inflammatory compound, a DNA methylation promoter, an anti-spasmodic agent, a protein synthesis inhibitor, an D-glucosidase inhibitor, a calcium channel blocker, a pyruvate dehydrogenase activator, a prostaglandin inhibitor, a sodium channel inhibitor, a serine protease inhibitor, an intracellular calcium flux inhibitor, a JAK2 inhibitor, an androgen inhibitor, an aromatase inhibitor, an anti-viral agent, a 5-HT inhibitor, an FXR antagonist, an actin polymerization and stabilization promoter, an AXOR12 agonist, an angiotensin Il receptor agonist, a platelet aggregation inhibitor, a CB1/CB2 receptor agonist, a norepinephrine reuptake inhibitor, a selective serotonin reuptake inhibitor, a reducing agent, and a immuno-modulator selected from Bay 11-7085, (-)- arctigenin, idazoxan hydrochloride.
93. The method of any one of claims 87-89 wherein the anti-scarring agent is selected from an angiogenesis inhibitor, an apoptosis antagonist, an apoptosis activator, a beta 1 integrin antagonist, a beta tubulin inhibitor, a blocker of enzyme production in Hepatitis C, a Bruton's tyrosine kinase inhibitor, a calcineurin inhibitor, a caspase 3 inhibitor, a CC chemokine receptor antagonist, a cell cycle inhibitor, a cathepsin B inhibitor, a cathepsin K inhibitor, a cathepsin L inhibitor, a CD40 antagonist, a chemokine receptor antagonist, a chymase inhibitor, a collagenase antagonist, a CXCR antagonist, a cyclin dependent kinase inhibitor, a cyclooxygenase 1 inhibitor, a DHFR inhibitor, a cual integrin inhibitor, an elastase inhibitor, an elongation factor-1 alpha inhibitor, an endothelial growth factor antagonist, an endothelial growth factor receptor kinase inhibitor, an endotoxin antagonist, an epothilone and tubulin binder, an estrogen receptor antagonist, an FGF inhibitor, a famexyl transferase inhibitor, a farnesyltransferase inhibitor, an FLT-3 kinase inhibitor, an FGF receptor kinase inhibitor, a fibrinogen antagonist, a histone deacetylase inhibitor, an HMGCoA reductase inhibitor, an ICAM inhibitor, an IL, ICE, and IRAK antagonist, an IL-2 inhibitor, an immunosuppressant, an inosine monophosphate inhibitor, an integrin antagonist, an interleukin antagonist, an inhibitor of type III receptor tyrosine kinase, an irreversible inhibitor of enzyme methionine aminopeptidase type 2, an isozyme selective delta protein kinase C inhibitor, a JAK3 enzyme inhibitor, a JNK inhibitor, a kinase inhibitor, a kinesin antagonist, a leukotriene inhibitor and antagonist, a MAP kinase inhibitor, a matrix metalloproteinase inhibitor, an MCP-CCR2 inhibitor, an mTOR inhibitor, an mTOR kinase inhibitor, a microtubule inhibitor, an MIF inhibitor, a neurokinin antagonist, an NF kappa B inhibitor, a nitric oxide agonist, an ornithine decarboxylase inhibitor, a p38 MAP kinase inhibitor, a palmitoyl-protein thioesterase inhibitor, a PDGF receptor kinase inhibitor, a peroxisome proliferator-activated receptor (PPAR) agonist, a phosphatase inhibitor, a phosphodiesterase inhibitor, a PKC inhibitor, a platelet activating factor antagonist, a prolyl hydroxylase inhibitor, a polymorphonuclear neutrophil inhibitor, protein kinase B inhibitor, protein kinase C stimulant, purine nucleoside analogue, a purineoreceptor P2X antagonist, a Raf kinase inhibitor, reversible inhibitor of ErbB1 and ErbB2, ribonucleoside triphosphate reductase inhibitor, an SDF-1 antagonist, a sheddase inhibitor, an SRC inhibitor, a stromelysin inhibitor, an Syk kinase inhibitor, a telomerase inhibitor, a TGF beta inhibitor, a TNF-alpha antagonist or TACE inhibitor, a tumor necrosis factor antagonist, a Toll receptor inhibitor, a tubulin antagonist, a protein tyrosine kinase inhibitor, a VEGF inhibitor, and a vitamin D receptor agonist.
94. The method of any one of claims 87-89 wherein the anti-scarring agent is selected from a retinoic acid receptor antagonist, a heat shock protein 90 antagonist, a steroid, a cell cycle inhibitor, a histone deacetylase inhibitor, an anti-microbial agent, an intracellular calcium flux inhibitor, an microtubule inhibitor, an HMGCoA reductase inhibitor, an actin polymerization and stabilization promoter, a tyrosine kinase inhibitor, a TGF beta inhibitor, a TNF-alpha antagonist, a TACE inhibitor, a calcineurin inhibitor, a peptidyl-prolyl cis/trans isomerase inhibitor, an apoptosis activator, an antimetabolite and anti-neoplastic agent, a TGF beta inhibitor, a DNA methylation promoter, and a PKC inhibitor.
95. The method of any one of claims 87-89 wherein the anti-scarring agent is selected from ZD-6474, AP-23573, synthadotin, S- 0885, aplidine, ixabepilone, IDN-5390, SB-2723005, ABT-518, combretastatin, anecortave acetate, SB-715992, temsirolimus, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, dolastatin 15, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17-DMAG, tacrolimus, loteprednol etabonate, juglone, prednisolone, puromycin, 3-BAABE, cladribine, mannose-6-phosphate, 5-azacytidine, Ly333531 (ruboxistaurin), and simvastatin.
96. The method of any one of claims 87-95 wherein the composition further comprises an anti-infective agent selected from an anthracycline, doxorubicin, mitoxantrone, fluoropyrimidine, 5-fluorouracil, a folic acid antagonist, methotrexate, podophylotoxin, etoposide, camptothecin, hydroxyurea, a platinum complex, and cisplatin.
97. The method of any one of claims 87-95 wherein the composition further comprises an anti-thrombotic agent.
98. The method of any one of claims 87-95 wherein the composition comprises a naturally occurring polymer; protein; carbohydrate; biodegradable polymer; nonbiodegradable polymer; collagen; methylated collagen; fibrinogen; thrombin; blood plasma; calcium salt; an antifibronolytic agent; fibrinogen analog; albumin; plasminogen; von Willebrands factor; factor VIII; hypoallergenic collagen; atelopeptidic collagen; crosslinked collagen; aprotinin; epsilon-amino-n-caproic acid; gelatin; protein conjugates; gelatin conjugates; a synthetic polymer; isocyanate-containing compound; a synthetic thiol-containing compound; a synthetic compound containing at least two thiol groups; a synthetic compound containing at least three thiol groups; a synthetic compound containing at least four thiol groups; a synthetic amino-containing compound; a synthetic compound containing at least two amino groups; a synthetic compound containing at least three amino groups; a synthetic compound containing at least four amino groups; a synthetic compound comprising a carbonyl-oxygen- succinimidyl group; a synthetic compound comprising at least two carbonyl- oxygen-succinimidyl groups; a synthetic compound comprising at least three carbonyl-oxygen-succinimidyl groups; a synthetic compound comprising at least four carbonyl-oxygen-succinimidyl groups; a synthetic polyalkylene oxide-containing compound; a synthetic compound comprising both polyalkylene oxide and biodegradable polyester blocks; a synthetic polyalkylene oxide-containing compound having reactive amino groups; a synthetic polyalkylene oxide-containing compound having reactive thiol groups; a synthetic polyalkylene oxide-containing compound having reactive carbonyl-oxygen-succinimidyl groups; a synthetic compound comprising a biodegradable polyester block; a synthetic polymer formed in whole or part from lactic acid or lactide; a synthetic polymer formed in whole or part from glycolic acid or glycolide; polylysine; (a) protein and (b) a compound comprising a polyalkylene oxide portion; (a) protein and (b) polylysine; (a) protein and (b) a compound having at least four thiol groups; (a) protein and (b) a compound having at least four amino groups; (a) protein and (b) a compound having at least four carbonyl-oxygen-succinimide groups; (a) protein and (b) a compound having a biodegradable region formed from reactants selected from lactic acid, lactide, glycolic acid, glycolide, and epison-caprolactone; (a) collagen and (b) a compound comprising a polyalkylene oxide portion; (a) collagen and (b) polylysine; (a) collagen and (b) a compound having at least four thiol groups; (a) collagen and (b) a compound having at least four thiol groups; (a) collagen and (b) a compound having at least four amino groups; (a) collagen and (b) a compound having at least four carbonyl-oxygen-succinimide groups; (a) collagen and (b) a compound having a biodegradable region formed from reactants selected from lactic acid, lactide, glycolic acid, glycolide, and epison-caprolactone; (a) methylated collagen and (b) a compound comprising a polyalkylene oxide portion; (a) methylated collagen and (b) polylysine; (a) methylated collagen and (b) a compound having at least four thiol groups; (a) methylated collagen and (b) a compound having at least four amino groups; (a) methylated collagen and (b) a compound having at least four carbonyl-oxygen-succinimide groups; (a) methylated collagen and (b) a compound having a biodegradable region formed from reactants selected from lactic acid, lactide, glycolic acid, glycolide, and epison- caprolactone; hyaluronic acid; a hyaluronic acid derivative; pentaerythritol poly(ethylene glycol)ether tetra-sulfhydryl of number average molecular weight between 3,000 and 30,000; pentaerythritol poly(ethylene glycol)ether tetra-amino of number average molecular weight between 3,000 and 30,000; or (a) a synthetic compound having a number average molecular weight between 3,000 and 30,000 and comprising a polyalkylene oxide region and multiple nucleophilic groups, and (b) a synthetic compound having a number average molecular weight between 3,000 and 30,000 and comprising a polyalkylene oxide region and multiple electrophilic groups.
99. A method for treating a vascular disease in a patient in need thereof, comprising delivering to the patient a) an anti-scarring agent or b) a composition comprising i) an anti-scarring agent and ii) a polymer and/or a compound that forms a polymer in situ.
100. The method of claim 99 wherein the vascular disease is stenosis, restenosis, or atherosclerosis.
101. The method of claim 99 or 100 wherein the agent or composition is delivered perivascularly.
102. The method of any one of claims 99-101 wherein the anti-scarring agent is an antimicrobial compound.
103. The method of any one of claims 99-101 wherein the antimicrobial compound is brefeldin A.
104. The method of any one of claims 99-101 wherein the anti-scarring agent is selected from a histamine receptor antagonist, an alpha adrenergic receptor antagonist, an anti-psychotic compound, a CaM kinase Il inhibitor, a G protein agonist, an antibiotic selected from the group consisting of apigenin, ampicillin sodium salt, puromycin, an anti-microbial agent, a DNA topoisomerase inhibitor, a thromboxane A2 receptor inhibitor, a D2 dopamine receptor antagonist, a Peptidyl-Prolyl Cis/Trans lsomerase Inhibitor, a dopamine antagonist, an anesthetic compound, a clotting factor, a lysyl hydrolase inhibitor, a muscarinic receptor inhibitor, a superoxide anion generator, a steroid, an anti-proliferative agent, a diuretic, an anticoagulant, a cyclic GMP agonist, an adenylate cyclase agonist, an antioxidant, a nitric oxide synthase inhibitor, an anti-neoplastic agent, a DNA synthesis inhibitor, a DNA alkylating agent, a DNA methylation inhibitor, a NSAID agent, a peptidylglycine alpha-hydroxylating monooxygenase inhibitor, an MEK1/MEK 2 inhibitor, a NO synthase inhibitor, a retinoic acid receptor antagonist, an ACE inhibitor, a glycosylation inhibitor, an intracellular calcium influx inhibitor, an anti-emetic agent, an acetylcholinesterase inhibitor, an ALK-5 receptor antagonist, a RAR/RXT antagonist, an elF-2a inhibitor, an S-adenosyl-L-homocysteine hydrolase inhibitor, an estrogen agonist, a serotonin receptor inhibitor, an antithrombotic agent, a tryptase inhibitor, a pesticide, a bone mineralization promoter, a bisphosphonate compound selected from risedronate and an analogue or derivative thereof, an anti-inflammatory compound, a DNA methylation promoter, an anti-spasmodic agent, a protein synthesis inhibitor, an D-glucosidase inhibitor, a calcium channel blocker, a pyruvate dehydrogenase activator, a prostaglandin inhibitor, a sodium channel inhibitor, a serine protease inhibitor, an intracellular calcium flux inhibitor, a JAK2 inhibitor, an androgen inhibitor, an aromatase inhibitor, an anti-viral agent, a 5-HT inhibitor, an FXR antagonist, an actin polymerization and stabilization promoter, an AXOR12 agonist, an angiotensin Il receptor agonist, a platelet aggregation inhibitor, a CB1/CB2 receptor agonist, a norepinephrine reuptake inhibitor, a selective serotonin reuptake inhibitor, a reducing agent, and a immuno-modulator selected from Bay 11-7085, (-)- arctigenin, idazoxan hydrochloride.
105. The method of any one of claims 99-101 wherein the anti-scarring agent is selected from an angiogenesis inhibitor, an apoptosis antagonist, an apoptosis activator, a beta 1 integrin antagonist, a beta tubulin inhibitor, a blocker of enzyme production in Hepatitis C, a Bruton's tyrosine kinase inhibitor, a calcineurin inhibitor, a caspase 3 inhibitor, a CC chemokine receptor antagonist, a cell cycle inhibitor, a cathepsin B inhibitor, a cathepsin K inhibitor, a cathepsin L inhibitor, a CD40 antagonist, a chemokine receptor antagonist, a chymase inhibitor, a collagenase antagonist, a CXCR antagonist, a cyclin dependent kinase inhibitor, a cyclooxygenase 1 inhibitor, a DHFR inhibitor, a cual integrin inhibitor, an elastase inhibitor, an elongation factor-1 alpha inhibitor, an endothelial growth factor antagonist, an endothelial growth factor receptor kinase inhibitor, an endotoxin antagonist, an epothilone and tubulin binder, an estrogen receptor antagonist, an FGF inhibitor, a famexyl transferase inhibitor, a farnesyltransferase inhibitor, an FLT-3 kinase inhibitor, an FGF receptor kinase inhibitor, a fibrinogen antagonist, a histone deacetylase inhibitor, an HMGCoA reductase inhibitor, an ICAM inhibitor, an IL, ICE, and IRAK antagonist, an IL-2 inhibitor, an immunosuppressant, an inosine monophosphate inhibitor, an integrin antagonist, an interleukin antagonist, an inhibitor of type III receptor tyrosine kinase, an irreversible inhibitor of enzyme methionine aminopeptidase type 2, an isozyme selective delta protein kinase C inhibitor, a JAK3 enzyme inhibitor, a JNK inhibitor, a kinase inhibitor, a kinesin antagonist, a leukotriene inhibitor and antagonist, a MAP kinase inhibitor, a matrix metalloproteinase inhibitor, an MCP-CCR2 inhibitor, an mTOR inhibitor, an mTOR kinase inhibitor, a microtubule inhibitor, an MIF inhibitor, a neurokinin antagonist, an NF kappa B inhibitor, a nitric oxide agonist, an ornithine decarboxylase inhibitor, a p38 MAP kinase inhibitor, a palmitoyl-protein thioesterase inhibitor, a PDGF receptor kinase inhibitor, a peroxisome proliferator-activated receptor (PPAR) agonist, a phosphatase inhibitor, a phosphodiesterase inhibitor, a PKC inhibitor, a platelet activating factor antagonist, a prolyl hydroxylase inhibitor, a polymorphonuclear neutrophil inhibitor, protein kinase B inhibitor, protein kinase C stimulant, purine nucleoside analogue, a purineoreceptor P2X antagonist, a Raf kinase inhibitor, reversible inhibitor of ErbB1 and ErbB2, ribonucleoside triphosphate reductase inhibitor, an SDF-1 antagonist, a sheddase inhibitor, an SRC. inhibitor, a stromelysin inhibitor, an Syk kinase inhibitor, a telomerase inhibitor, a TGF beta inhibitor, a TNF-alpha antagonist or TACE inhibitor, a tumor necrosis factor antagonist, a Toll receptor inhibitor, a tubulin antagonist, a protein tyrosine kinase inhibitor, a VEGF inhibitor, and a vitamin D receptor agonist.
106. The method of any one of claims 99-101 wherein the anti-scarring agent is selected from a retinoic acid receptor antagonist, a heat shock protein 90 antagonist, a steroid, a cell cycle inhibitor, a histone deacetylase inhibitor, an anti-microbial agent, an intracellular calcium flux inhibitor, an microtubule inhibitor, an HMGCoA reductase inhibitor, an actin polymerization and stabilization promoter, a tyrosine kinase inhibitor, a TGF beta inhibitor, a TNF-alpha antagonist, a TACE inhibitor, a calcineurin inhibitor, a peptidyl-prolyl cis/trans isomerase inhibitor, an apoptosis activator, an antimetabolite and anti-neoplastic agent, a TGF beta inhibitor, a DNA methylation promoter, and a PKC inhibitor.
107. The method of any one of claims 99-101 wherein the anti-scarring agent is selected from ZD-6474, AP-23573, synthadotin, S- 0885, aplidine, ixabepilone, IDN-5390, SB-2723005, ABT-518, combretastatin, anecortave acetate, SB-715992, temsirolimus, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, dolastatin 15, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17-DMAG, tacrolimus, loteprednol etabonate, juglone, prednisolone, puromycin, 3-BAABE, cladribine, mannose-6-phosphate, 5-azacytidine, Ly333531 (ruboxistaurin), and simvastatin.
-108. The method of any one oiclaims 99-107 wherein the composition further comprises an anti-infective agent selected from an anthracycline, doxorubicin, mitoxantrone, fluoropyrimidine, 5-fluorouracil, a folic acid antagonist, methotrexate, podophylotoxin, etoposide, camptothecin, hydroxyurea, a platinum complex, and cisplatin.
109. The method of any one of claims 99-107 wherein the composition further comprises an anti-thrombotic agent.
110. The method of any one of claims 99-107 wherein the composition comprises a naturally occurring polymer; protein; carbohydrate; biodegradable polymer; nonbiodegradable polymer; collagen; methylated collagen; fibrinogen; thrombin; blood plasma; calcium salt; an antifibronolytic agent; fibrinogen analog; albumin; plasminogen; von Willebrands factor; factor VIII; hypoallergenic collagen; atelopeptidic collagen; crosslinked collagen; aprotinin; epsilon-amino-n-caproic acid; gelatin; protein conjugates; gelatin conjugates; a synthetic polymer; isocyanate-containing compound; a synthetic thiol-containing compound; a synthetic compound containing at least two thiol groups; a synthetic compound containing at least three thiol groups; a synthetic compound containing at least four thiol groups; a synthetic amino-containing compound; a synthetic compound containing at least two amino groups; a synthetic compound containing at least three amino groups; a synthetic compound containing at least four amino groups; a synthetic compound comprising a carbonyl-oxygen- succinimidyl group; a synthetic compound comprising at least two carbonyl- oxygen-succinimidyl groups; a synthetic compound comprising at least three carbonyl-oxygen-succinimidyl groups; a synthetic compound comprising at least four carbonyl-oxygen-succinimidyl groups; a synthetic polyalkylene oxide-containing compound; a synthetic compound comprising both polyalkylene oxide and biodegradable polyester blocks; a synthetic polyalkylene oxide-containing compound having reactive amino groups; a synthetic polyalkylene oxide-containing compound having reactive thiol groups; a synthetic polyalkylene oxide-containing compound having reactive carbonyl-oxygen-succinimidyl groups; a synthetic compound comprising a biodegradable polyester block; a synthetic polymer formed in whole or part from lactic acid or lactide; a synthetic polymer formed in whole or part from glycolic acid or glycolide; polylysine; (a) protein and (b) a compound comprising a polyalkylene oxide portion; (a) protein and (b) polylysine; (a) protein and (b) a compound having at least four thiol groups; (a) protein and (b) a compound having at least four amino groups; (a) protein and (b) a compound having at least four carbonyl-oxygen-succinimide groups; (a) protein and (b) a compound having a biodegradable region formed from reactants selected from lactic acid, lactide, glycolic acid, glycolide, and epison-caprolactone; (a) collagen and (b) a compound comprising a polyalkylene oxide portion; (a) collagen and (b) polylysine; (a) collagen and (b) a compound having at least four thiol groups; (a) collagen and (b) a compound having at least four thiol groups; (a) collagen and (b) a compound having at least four amino groups; (a) collagen and (b) a compound having at least four carbonyl-oxygen-succinimide groups; (a) collagen and (b) a compound having a biodegradable region formed from reactants selected from lactic acid, lactide, glycolic acid, glycolide, and epison-caprolactone; (a) methylated collagen and (b) a compound comprising a polyalkylene oxide portion; (a) methylated collagen and (b) polylysine; (a) methylated collagen and (b) a compound having at least four thiol groups; (a) methylated collagen and (b) a compound having at least four amino groups; (a) methylated collagen and (b) a compound having at least four carbonyl-oxygen-succinimide groups; (a) methylated collagen and (b) a compound having a biodegradable region formed from reactants selected from lactic acid, lactide, glycolic acid, glycolide, and epison- caprolactone; hyaluronic acid; a hyaluronic acid derivative; pentaerythritol poly(ethylene glycol)ether tetra-sulfhydryl of number average molecular weight between 3,000 and 30,000; pentaerythritol poly(ethylene glycol)ether tetra-amino of number average molecular weight between 3,000 and 30,000; or (a) a synthetic compound having a number average molecular weight between 3,000 and 30,000 and comprising a polyalkylene oxide region and multiple nucleophilic groups, and (b) a synthetic compound having a number average molecular weight between 3,000 and 30,000 and comprising a polyalkylene oxide region and multiple electrophilic groups.
111. A composition comprising i) an anti-scarring agent and ii) a polymer or a compound that forms a polymer in situ.
112. The method of claim 111 wherein the anti-scarring agent is an antimicrobial compound.
113. The method of claim 111 or claim 112 wherein the antimicrobial compound is brefeldin A.
114. The method of any one of claims 111-113 wherein the anti-scarring agent is selected from a histamine receptor antagonist, an alpha adrenergic receptor antagonist, an anti-psychotic compound, a CaM kinase Il inhibitor, a G protein agonist, an antibiotic selected from the group consisting of apigenin, ampicillin sodium salt, puromycin, an anti-microbial agent, a DNA topoisomerase inhibitor, a thromboxane A2 receptor inhibitor, a D2 dopamine receptor antagonist, a Peptidyl-Prolyl Cis/Trans lsomerase Inhibitor, a dopamine antagonist, an anesthetic compound, a clotting factor, a lysyl hydrolase inhibitor, a muscarinic receptor inhibitor, a superoxide anion generator, a steroid, an antiproliferative agent, a diuretic, an anticoagulant, a cyclic GMP agonist, an adenylate cyclase agonist, an antioxidant, a nitric oxide synthase inhibitor, an antineoplastic agent, a DNA synthesis inhibitor, a DNA alkylating agent, a DNA methylation inhibitor, a NSAID agent, a peptidylglycine alpha-hydroxylating monooxygenase inhibitor, an MEK1/MEK 2 inhibitor, a NO synthase inhibitor, a retinoic acid receptor antagonist, an ACE inhibitor, a glycosylation inhibitor, an intracellular calcium influx inhibitor, an anti-emetic agent, an acetylcholinesterase inhibitor, an ALK-5 receptor antagonist, a RAR/RXT antagonist, an elF-2a inhibitor, an S-adenosyl-L-homocysteine hydrolase inhibitor, an estrogen agonist, a serotonin receptor inhibitor, an antithrombotic agent, a tryptase inhibitor, a pesticide, a bone mineralization promoter, a bisphosphonate compound selected from risedronate and an analogue or derivative thereof, an anti-inflammatory compound, a DNA methylation promoter, an anti-spasmodic agent, a protein synthesis inhibitor, an D-glucosidase inhibitor, a calcium channel blocker, a pyruvate dehydrogenase activator, a prostaglandin inhibitor, a sodium channel inhibitor, a serine protease inhibitor, an intracellular calcium flux inhibitor, a JAK2 inhibitor, an androgen inhibitor, an aromatase inhibitor, an anti-viral agent, a 5-HT inhibitor, an FXR antagonist, an actin polymerization and stabilization promoter, an AXOR12 agonist, an angiotensin Il receptor agonist, a platelet aggregation inhibitor, a CB1/CB2 receptor agonist, a norepinephrine reuptake inhibitor, a selective serotonin reuptake inhibitor, a reducing agent, and a immuno-modulator selected from Bay 11-7085, (-)- arctigenin, idazoxan hydrochloride.
115. The method of any one of claims 111-113 wherein the anti-scarring agent is selected from an angiogenesis inhibitor, an apoptosis antagonist, an apoptosis activator, a beta 1 integrin antagonist, a beta tubulin inhibitor, a blocker of enzyme production in Hepatitis C, a Bruton's tyrosine kinase inhibitor, a calcineurin inhibitor, a caspase 3 inhibitor, a CC chemokine receptor antagonist, a cell cycle inhibitor, a cathepsin B inhibitor, a cathepsin K inhibitor, a cathepsin L inhibitor, a CD40 antagonist, a chemokine receptor antagonist, a chymase inhibitor, a collagenase antagonist, a CXCR antagonist, a cyclin dependent kinase inhibitor, a cyclooxygenase 1 inhibitor, a DHFR inhibitor, a cual integrin inhibitor, an elastase inhibitor, an elongation factor-1 alpha inhibitor, an endothelial growth factor antagonist, an endothelial growth factor receptor kinase inhibitor, an endotoxin antagonist, an epothilone and tubulin binder, an estrogen receptor antagonist, an FGF inhibitor, a famexyl transferase inhibitor, a farnesyltransferase inhibitor, an FLT-3 kinase inhibitor, an FGF receptor kinase inhibitor, a fibrinogen antagonist, a histone deacetylase inhibitor, an HMGCoA reductase inhibitor, an ICAM inhibitor, an IL, ICE, and IRAK antagonist, an IL-2 inhibitor, an immunosuppressant, an inosine monophosphate inhibitor, an integrin antagonist, an interleukin antagonist, an inhibitor of type III receptor tyrosine kinase, an irreversible inhibitor of enzyme methionine aminopeptidase type 2, an isozyme selective delta protein kinase C inhibitor, a JAK3 enzyme inhibitor, a JNK inhibitor, a kinase inhibitor, a kinesin antagonist, a leukotriene inhibitor and antagonist, a MAP kinase inhibitor, a matrix metalloproteinase inhibitor, an MCP-CCR2 inhibitor, an mTOR inhibitor, an mTOR kinase inhibitor, a microtubule inhibitor, an MIF inhibitor, a neurokinin antagonist, an NF kappa B inhibitor, a nitric oxide agonist, an ornithine decarboxylase inhibitor, a p38 MAP kinase inhibitor, a palmitoyϊ-protein thioesterase inhibitor, a PDGF receptor kinase inhibitor, a peroxisome proliferator-activated receptor (PPAR) agonist, a phosphatase inhibitor, a phosphodiesterase inhibitor, a PKC inhibitor, a platelet activating factor antagonist, a prolyl hydroxylase inhibitor, a polymorphonuclear neutrophil inhibitor, protein kinase B inhibitor, protein kinase C stimulant, purine nucleoside analogue, a purineoreceptor P2X antagonist, a Raf kinase inhibitor, reversible inhibitor of ErbB1 and ErbB2, ribonucleoside triphosphate reductase inhibitor, an SDF-1 antagonist, a sheddase inhibitor, an SRC inhibitor, a stromelysin inhibitor, an Syk kinase inhibitor, a telomerase inhibitor, a TGF beta inhibitor, a TNF-alpha antagonist or TACE inhibitor, a tumor necrosis factor antagonist, a Toll receptor inhibitor, a tubulin antagonist, a protein tyrosine kinase inhibitor, a VEGF inhibitor, and a vitamin D receptor agonist.
116. The method of any one of claims 111-113 wherein the antirscarring agent is selected from a retinoic acid receptor antagonist, a heat shock protein 90 antagonist, a steroid, a cell cycle inhibitor, a histone deacetylase inhibitor, an anti-microbial agent, an intracellular calcium flux inhibitor, an microtubule inhibitor, an HMGCoA reductase inhibitor, an actin polymerization and stabilization promoter, a tyrosine kinase inhibitor, a TGF beta inhibitor, a TNF-alpha antagonist, a TACE inhibitor, a calcineurin inhibitor, a peptidyl-prolyl cis/trans isomerase inhibitor, an apoptosis activator, an antimetabolite and anti-neoplastic agent, a TGF beta inhibitor, a DNA methylation promoter, and a PKC inhibitor.
117. The method of any one of claims 111-113 wherein the anti-scarring agent is selected from ZD-6474, AP-23573, synthadotin, S- 0885, aplidine, ixabepilone, IDN-5390, SB-2723005, ABT-518, combretastatin, anecortave acetate, SB-715992, temsirolimus, adalimumab, erucylphosphocholine, alphastatin, etanercept, humicade, gefitinib, isotretinoin, radicicol, clobetasol propionate, homoharringtonine, trichostatin A, brefeldin A, thapsigargin, dolastatin 15, cerivastatin, jasplakinolide, herbimycin A, pirfenidone, vinorelbine, 17-DMAG, tacrolimus, loteprednol etabonate, juglone, prednisolone, puromycin, 3-BAABE, cladribine, mannose-6-phosphate, 5-azacytidine, Ly333531 (ruboxistaurin), and simvastatin.
118. The method of any one of claims 111-117 wherein the composition further comprises an anti-infective agent selected from an anthracycline, doxorubicin, mitoxantrone, fluoropyrimidine, 5-fluorouracil, a folic acid antagonist, methotrexate, podophylotoxin, etoposide, camptothecin, hydroxyurea, a platinum complex, and cisplatin.
119. The method of any one of claims 111-117 wherein the composition further comprises an anti-thrombotic agent.
120. The method of any one of claims 111-117 wherein the polymer is crosslinked.
121. The method of any one of claims 111-117 wherein the polymer reacts with a mammalian tissue.
122. The method of any one of claims 111-117 wherein the composition comprises a naturally occurring polymer; protein; carbohydrate; biodegradable polymer; nonbiodegradable polymer; collagen; methylated collagen; fibrinogen; thrombin; blood plasma; calcium salt; an antifibronolytic agent; fibrinogen analog; albumin; plasminogen; von Willebrands factor; factor VIII; hypoallergenic collagen; atelopeptidic collagen; crosslinked collagen; aprotinin; epsilon-amino-n-caproic acid; gelatin; protein conjugates; gelatin conjugates; a synthetic polymer; isocyanate-containing compound; a synthetic thiol-containing compound; a synthetic compound containing at least two thiol groups; a synthetic compound containing at least three thiol groups; a synthetic compound containing at least four thiol groups; a synthetic amino-containing compound; a synthetic compound containing at least two amino groups; a synthetic compound containing at least three amino groups; a synthetic compound containing at least four amino groups; a synthetic compound comprising a carbonyl-oxygen- succinimidyl group; a synthetic compound comprising at least two carbonyl- oxygen-succinimidyl groups; a synthetic compound comprising at least three carbonyl-oxygen-succinimidyl groups; a synthetic compound comprising at least four carbonyl-oxygen-succinimidyl groups; a synthetic polyalkylene oxide-containing compound; a synthetic compound comprising both polyalkylene oxide and biodegradable polyester blocks; a synthetic polyalkylene oxide-containing compound having reactive amino groups; a synthetic polyalkylene oxide-containing compound having reactive thiol groups; a synthetic polyalkylene oxide-containing compound having reactive carbonyl-oxygen-succinimidyl groups; a synthetic compound comprising a biodegradable polyester_block; a synthetic polymer formed in whole or part from lactic acid or lactide; a synthetic polymer formed in whole or part from glycolic acid or glycolide; polylysine; (a) protein and (b) a compound comprising a polyalkylene oxide portion; (a) protein and (b) polylysine; (a) protein and (b) a compound having at least four thiol groups; (a) protein and (b) a compound having at least four amino groups; (a) protein and (b) a compound having at least four carbonyl-oxygen-succinimide groups; (a) protein and (b) a compound having a biodegradable region formed from reactants selected from lactic acid, lactide, glycolic acid, glycolide, and epison-caprolactone; (a) collagen and (b) a compound comprising a polyalkylene oxide portion; (a) collagen and (b) polylysine; (a) collagen and (b) a compound having at least four thiol groups; (a) collagen and (b) a compound having at least four thiol groups; (a) collagen and (b) a compound having at least four amino groups; (a) collagen and (b) a compound having at least four carbonyl-oxygen-succinimide groups; (a) collagen and (b) a compound having a biodegradable region formed from reactants selected from lactic acid, lactide, glycolic acid, glycolide, and epison-caprolactone; (a) methylated collagen and (b) a compound comprising a polyalkylene oxide portion; (a) methylated collagen and (b) polylysine; (a) methylated collagen and (b) a compound having at least four thiol groups; (a) methylated collagen and (b) a compound having at least four amino groups; (a) methylated collagen and (b) a compound having at least four carbonyl-oxygen-succinimide groups; (a) methylated collagen and (b) a compound having a biodegradable region formed from reactants selected from lactic acid, lactide, glycolic acid, glycolide, and epison- caprolactone; hyaluronic acid; a hyaluronic acid derivative; pentaerythritol poly(ethylene glycol)ether tetra-sulfhydryl of number average molecular weight between 3,000 and 30,000; pentaerythritol poly(ethylene glycol)ether tetra-amino of number average molecular weight between 3,000 and 30,000; or (a) a synthetic compound having a number average molecular weight between 3,000 and 30,000 and comprising a polyalkylene oxide region and multiple nucleophilic groups, and (b) a synthetic compound having a number average molecular weight between 3,000 and 30,000 and comprising a polyalkylene oxide region and multiple electrophilic groups.
PCT/US2006/013030 2005-05-10 2006-03-31 Anti-scarring agents, therapeutic compositions, and use thereof WO2006135479A2 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US67929305P 2005-05-10 2005-05-10
US60/679,293 2005-05-10

Publications (2)

Publication Number Publication Date
WO2006135479A2 true WO2006135479A2 (en) 2006-12-21
WO2006135479A3 WO2006135479A3 (en) 2007-04-12

Family

ID=37532760

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2006/013030 WO2006135479A2 (en) 2005-05-10 2006-03-31 Anti-scarring agents, therapeutic compositions, and use thereof

Country Status (1)

Country Link
WO (1) WO2006135479A2 (en)

Cited By (57)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2008132233A1 (en) * 2007-05-01 2008-11-06 Kuros Biosurgery Ag Composition, methods and kits for prevention of adhesion
WO2014190047A1 (en) 2013-05-21 2014-11-27 Northwestern University Para-xylene films and therapeutic uses thereof
US8927004B1 (en) 2014-06-11 2015-01-06 Silver Bullet Therapeutics, Inc. Bioabsorbable substrates and systems that controllably release antimicrobial metal ions
US9011465B2 (en) 2004-10-08 2015-04-21 Covidien Lp Endoscopic surgical clip applier
US9089334B2 (en) 2008-08-29 2015-07-28 Covidien Lp Endoscopic surgical clip applier with connector plate
US9108051B2 (en) 2010-11-12 2015-08-18 Silver Bullet Therapeutics, Inc. Bone implant and systems that controllably releases silver
US9114197B1 (en) 2014-06-11 2015-08-25 Silver Bullett Therapeutics, Inc. Coatings for the controllable release of antimicrobial metal ions
US9173973B2 (en) 2006-07-20 2015-11-03 G. Lawrence Thatcher Bioabsorbable polymeric composition for a medical device
US9211205B2 (en) 2006-10-20 2015-12-15 Orbusneich Medical, Inc. Bioabsorbable medical device with coating
US9248254B2 (en) 2009-08-27 2016-02-02 Silver Bullet Therapeutics, Inc. Bone implants for the treatment of infection
US9393024B2 (en) 2010-02-25 2016-07-19 Covidien Lp Articulating endoscopic surgical clip applier
US9398917B2 (en) 2007-03-26 2016-07-26 Covidien Lp Endoscopic surgical clip applier
US9414844B2 (en) 2008-08-25 2016-08-16 Covidien Lp Surgical clip appliers
US9452242B2 (en) 2014-06-11 2016-09-27 Silver Bullet Therapeutics, Inc. Enhancement of antimicrobial silver, silver coatings, or silver platings
US9642627B2 (en) 2010-11-02 2017-05-09 Covidien Lp Self-centering clip and jaw
US9687247B2 (en) 2004-10-08 2017-06-27 Covidien Lp Apparatus for applying surgical clips
US9708348B2 (en) 2014-10-03 2017-07-18 Infinity Pharmaceuticals, Inc. Trisubstituted bicyclic heterocyclic compounds with kinase activities and uses thereof
US9717505B2 (en) 2010-07-28 2017-08-01 Covidien Lp Articulating clip applier cartridge
US9724864B2 (en) 2006-10-20 2017-08-08 Orbusneich Medical, Inc. Bioabsorbable polymeric composition and medical device
US9737310B2 (en) 2010-07-28 2017-08-22 Covidien Lp Articulating clip applier
US9751888B2 (en) 2013-10-04 2017-09-05 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
US9750500B2 (en) 2013-01-18 2017-09-05 Covidien Lp Surgical clip applier
US9763668B2 (en) 2004-10-08 2017-09-19 Covidien Lp Endoscopic surgical clip applier
US9775844B2 (en) 2014-03-19 2017-10-03 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
US9775624B2 (en) 2013-08-27 2017-10-03 Covidien Lp Surgical clip applier
US9775623B2 (en) 2011-04-29 2017-10-03 Covidien Lp Surgical clip applier including clip relief feature
US9821094B2 (en) 2014-06-11 2017-11-21 Silver Bullet Therapeutics, Inc. Coatings for the controllable release of antimicrobial metal ions
US9828377B2 (en) 2013-10-04 2017-11-28 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
US9848886B2 (en) 2013-01-08 2017-12-26 Covidien Lp Surgical clip applier
US9855043B2 (en) 2011-12-19 2018-01-02 Covidien Lp Jaw closure mechanism for a surgical clip applier
US9931124B2 (en) 2015-01-07 2018-04-03 Covidien Lp Reposable clip applier
US9968362B2 (en) 2013-01-08 2018-05-15 Covidien Lp Surgical clip applier
US10004502B2 (en) 2009-12-09 2018-06-26 Covidien Lp Surgical clip applier
US10160761B2 (en) 2015-09-14 2018-12-25 Infinity Pharmaceuticals, Inc. Solid forms of isoquinolinones, and process of making, composition comprising, and methods of using the same
US10159492B2 (en) 2012-05-31 2018-12-25 Covidien Lp Endoscopic clip applier
US10159491B2 (en) 2015-03-10 2018-12-25 Covidien Lp Endoscopic reposable surgical clip applier
US10166027B2 (en) 2006-10-17 2019-01-01 Covidien Lp Apparatus for applying surgical clips
US10231738B2 (en) 2008-08-29 2019-03-19 Covidien Lp Endoscopic surgical clip applier with wedge plate
US10258346B2 (en) 2007-04-11 2019-04-16 Covidien Lp Surgical clip applier
US10265435B2 (en) 2009-08-27 2019-04-23 Silver Bullet Therapeutics, Inc. Bone implant and systems and coatings for the controllable release of antimicrobial metal ions
US10287353B2 (en) 2016-05-11 2019-05-14 Huya Bioscience International, Llc Combination therapies of HDAC inhibitors and PD-1 inhibitors
US10292712B2 (en) 2015-01-28 2019-05-21 Covidien Lp Surgical clip applier with integrated cutter
US10349936B2 (en) 2011-12-29 2019-07-16 Covidien Lp Surgical clip applier with integrated clip counter
US10357250B2 (en) 2011-01-31 2019-07-23 Covidien Lp Locking cam driver and jaw assembly for clip applier
US10385131B2 (en) 2016-05-11 2019-08-20 Huya Bioscience International, Llc Combination therapies of HDAC inhibitors and PD-L1 inhibitors
US10390831B2 (en) 2015-11-10 2019-08-27 Covidien Lp Endoscopic reposable surgical clip applier
US10470765B2 (en) 2009-12-15 2019-11-12 Covidien Lp Surgical clip applier
US10660639B2 (en) 2012-05-04 2020-05-26 Covidien Lp Surgical clip applier with dissector
US10668167B2 (en) 2016-06-02 2020-06-02 Abbvie Inc. Glucocorticoid receptor agonist and immunoconjugates thereof
US10682135B2 (en) 2008-08-29 2020-06-16 Covidien Lp Endoscopic surgical clip applier
US10702278B2 (en) 2014-12-02 2020-07-07 Covidien Lp Laparoscopic surgical ligation clip applier
WO2020172113A1 (en) * 2019-02-18 2020-08-27 Lankenau Institute For Medical Research Scar reducing wound closure materials
US10759806B2 (en) 2016-03-17 2020-09-01 Infinity Pharmaceuticals, Inc. Isotopologues of isoquinolinone and quinazolinone compounds and uses thereof as PI3K kinase inhibitors
US10772970B2 (en) 2017-12-01 2020-09-15 Abbvie Inc. Glucocorticoid receptor agonist and immunoconjugates thereof
US10863992B2 (en) 2017-08-08 2020-12-15 Covidien Lp Endoscopic surgical clip applier
US10919914B2 (en) 2016-06-08 2021-02-16 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
WO2021231563A1 (en) * 2020-05-13 2021-11-18 Spiral Therapeutics Inc. Gelling solutions for administration of compounds to the inner ear

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8940742B2 (en) 2012-04-10 2015-01-27 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20050175703A1 (en) * 2003-11-20 2005-08-11 Angiotech International Ag Polymer compositions and methods for their use

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20050175703A1 (en) * 2003-11-20 2005-08-11 Angiotech International Ag Polymer compositions and methods for their use

Cited By (101)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9763668B2 (en) 2004-10-08 2017-09-19 Covidien Lp Endoscopic surgical clip applier
US10349950B2 (en) 2004-10-08 2019-07-16 Covidien Lp Apparatus for applying surgical clips
US9687247B2 (en) 2004-10-08 2017-06-27 Covidien Lp Apparatus for applying surgical clips
US9011465B2 (en) 2004-10-08 2015-04-21 Covidien Lp Endoscopic surgical clip applier
US10485538B2 (en) 2004-10-08 2019-11-26 Covidien Lp Endoscopic surgical clip applier
US9173973B2 (en) 2006-07-20 2015-11-03 G. Lawrence Thatcher Bioabsorbable polymeric composition for a medical device
US10166027B2 (en) 2006-10-17 2019-01-01 Covidien Lp Apparatus for applying surgical clips
US9211205B2 (en) 2006-10-20 2015-12-15 Orbusneich Medical, Inc. Bioabsorbable medical device with coating
US9724864B2 (en) 2006-10-20 2017-08-08 Orbusneich Medical, Inc. Bioabsorbable polymeric composition and medical device
US9398917B2 (en) 2007-03-26 2016-07-26 Covidien Lp Endoscopic surgical clip applier
US10363045B2 (en) 2007-03-26 2019-07-30 Covidien Lp Endoscopic surgical clip applier
US10258346B2 (en) 2007-04-11 2019-04-16 Covidien Lp Surgical clip applier
WO2008132233A1 (en) * 2007-05-01 2008-11-06 Kuros Biosurgery Ag Composition, methods and kits for prevention of adhesion
US9414844B2 (en) 2008-08-25 2016-08-16 Covidien Lp Surgical clip appliers
US10682135B2 (en) 2008-08-29 2020-06-16 Covidien Lp Endoscopic surgical clip applier
US10231738B2 (en) 2008-08-29 2019-03-19 Covidien Lp Endoscopic surgical clip applier with wedge plate
US9089334B2 (en) 2008-08-29 2015-07-28 Covidien Lp Endoscopic surgical clip applier with connector plate
US9545254B2 (en) 2008-08-29 2017-01-17 Covidien Lp Endoscopic surgical clip applier with connector plate
US10159484B2 (en) 2008-08-29 2018-12-25 Covidien Lp Endoscopic surgical clip applier with connector plate
US9358011B2 (en) 2008-08-29 2016-06-07 Covidien Lp Endoscopic surgical clip applier with connector plate
US11213298B2 (en) 2008-08-29 2022-01-04 Covidien Lp Endoscopic surgical clip applier with wedge plate
US10231735B2 (en) 2008-08-29 2019-03-19 Covidien Lp Endoscopic surgical clip applier
US11806021B2 (en) 2008-08-29 2023-11-07 Covidien Lp Endoscopic surgical clip applier with wedge plate
US10004548B2 (en) 2009-08-27 2018-06-26 Silver Bullet Therapeutics, Inc. Bone implants for the treatment of infection
US9889284B2 (en) 2009-08-27 2018-02-13 Silver Bullet Therapeutics, Inc. Bone implant and systems that controllably releases silver
US11925723B2 (en) 2009-08-27 2024-03-12 Silver Bullet Therapeutics, Inc. Bone implant and systems and coatings for the controllable release of antimicrobial metal ions
US11224471B2 (en) 2009-08-27 2022-01-18 Silver Bullet Therapeutics, Inc. Bone implants for the treatment of infection
US11020508B2 (en) 2009-08-27 2021-06-01 Silver Bullet Therapeutics, Inc. Bone implant and systems and coatings for the controllable release of antimicrobial metal ions
US10368929B2 (en) 2009-08-27 2019-08-06 Silver Bullet Therapeutics, Inc. Bone implants for the treatment of infection
US9248254B2 (en) 2009-08-27 2016-02-02 Silver Bullet Therapeutics, Inc. Bone implants for the treatment of infection
US10265435B2 (en) 2009-08-27 2019-04-23 Silver Bullet Therapeutics, Inc. Bone implant and systems and coatings for the controllable release of antimicrobial metal ions
US10758234B2 (en) 2009-12-09 2020-09-01 Covidien Lp Surgical clip applier
US10004502B2 (en) 2009-12-09 2018-06-26 Covidien Lp Surgical clip applier
US10470765B2 (en) 2009-12-15 2019-11-12 Covidien Lp Surgical clip applier
US9393024B2 (en) 2010-02-25 2016-07-19 Covidien Lp Articulating endoscopic surgical clip applier
US10271854B2 (en) 2010-02-25 2019-04-30 Covidien Lp Articulating endoscopic surgical clip applier
US11213299B2 (en) 2010-02-25 2022-01-04 Covidien Lp Articulating endoscopic surgical clip applier
US9737310B2 (en) 2010-07-28 2017-08-22 Covidien Lp Articulating clip applier
US9717505B2 (en) 2010-07-28 2017-08-01 Covidien Lp Articulating clip applier cartridge
US10568635B2 (en) 2010-07-28 2020-02-25 Covidien Lp Articulating clip applier
US11517322B2 (en) 2010-07-28 2022-12-06 Covidien Lp Articulating clip applier
US9642627B2 (en) 2010-11-02 2017-05-09 Covidien Lp Self-centering clip and jaw
US9789298B2 (en) 2010-11-12 2017-10-17 Silver Bullet Therapeutics, Inc. Bone implant and systems that controllably releases silver
US9108051B2 (en) 2010-11-12 2015-08-18 Silver Bullet Therapeutics, Inc. Bone implant and systems that controllably releases silver
US10357250B2 (en) 2011-01-31 2019-07-23 Covidien Lp Locking cam driver and jaw assembly for clip applier
US9775623B2 (en) 2011-04-29 2017-10-03 Covidien Lp Surgical clip applier including clip relief feature
US9855043B2 (en) 2011-12-19 2018-01-02 Covidien Lp Jaw closure mechanism for a surgical clip applier
US11278287B2 (en) 2011-12-29 2022-03-22 Covidien Lp Surgical clip applier with integrated clip counter
US10349936B2 (en) 2011-12-29 2019-07-16 Covidien Lp Surgical clip applier with integrated clip counter
US10660639B2 (en) 2012-05-04 2020-05-26 Covidien Lp Surgical clip applier with dissector
US10159492B2 (en) 2012-05-31 2018-12-25 Covidien Lp Endoscopic clip applier
US11026696B2 (en) 2012-05-31 2021-06-08 Covidien Lp Endoscopic clip applier
US9968362B2 (en) 2013-01-08 2018-05-15 Covidien Lp Surgical clip applier
US9848886B2 (en) 2013-01-08 2017-12-26 Covidien Lp Surgical clip applier
US10743886B2 (en) 2013-01-08 2020-08-18 Covidien Lp Surgical clip applier
US9750500B2 (en) 2013-01-18 2017-09-05 Covidien Lp Surgical clip applier
US10537329B2 (en) 2013-01-18 2020-01-21 Covidien Lp Surgical clip applier
US10434225B2 (en) 2013-05-21 2019-10-08 Northwestern University Para-xylene films and therapeutic uses thereof
WO2014190047A1 (en) 2013-05-21 2014-11-27 Northwestern University Para-xylene films and therapeutic uses thereof
EP2999427A4 (en) * 2013-05-21 2016-11-16 Univ Northwestern Para-xylene films and therapeutic uses thereof
US10682146B2 (en) 2013-08-27 2020-06-16 Covidien Lp Surgical clip applier
US9775624B2 (en) 2013-08-27 2017-10-03 Covidien Lp Surgical clip applier
US10329299B2 (en) 2013-10-04 2019-06-25 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
US9751888B2 (en) 2013-10-04 2017-09-05 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
US9828377B2 (en) 2013-10-04 2017-11-28 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
US10675286B2 (en) 2014-03-19 2020-06-09 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
US11541059B2 (en) 2014-03-19 2023-01-03 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
US9775844B2 (en) 2014-03-19 2017-10-03 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
US8927004B1 (en) 2014-06-11 2015-01-06 Silver Bullet Therapeutics, Inc. Bioabsorbable substrates and systems that controllably release antimicrobial metal ions
US9114197B1 (en) 2014-06-11 2015-08-25 Silver Bullett Therapeutics, Inc. Coatings for the controllable release of antimicrobial metal ions
US9452242B2 (en) 2014-06-11 2016-09-27 Silver Bullet Therapeutics, Inc. Enhancement of antimicrobial silver, silver coatings, or silver platings
US9821094B2 (en) 2014-06-11 2017-11-21 Silver Bullet Therapeutics, Inc. Coatings for the controllable release of antimicrobial metal ions
US8999367B1 (en) 2014-06-11 2015-04-07 Silver Bullet Therapeutics, Inc. Bioabsorbable substrates and systems that controllably release antimicrobial metal ions
US9708348B2 (en) 2014-10-03 2017-07-18 Infinity Pharmaceuticals, Inc. Trisubstituted bicyclic heterocyclic compounds with kinase activities and uses thereof
US10941162B2 (en) 2014-10-03 2021-03-09 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
US10253047B2 (en) 2014-10-03 2019-04-09 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
US10702278B2 (en) 2014-12-02 2020-07-07 Covidien Lp Laparoscopic surgical ligation clip applier
US9931124B2 (en) 2015-01-07 2018-04-03 Covidien Lp Reposable clip applier
US10765435B2 (en) 2015-01-07 2020-09-08 Covidien Lp Reposable clip applier
US10292712B2 (en) 2015-01-28 2019-05-21 Covidien Lp Surgical clip applier with integrated cutter
US11134956B2 (en) 2015-01-28 2021-10-05 Covidien Lp Surgical clip applier with integrated cutter
US10828044B2 (en) 2015-03-10 2020-11-10 Covidien Lp Endoscopic reposable surgical clip applier
US10159491B2 (en) 2015-03-10 2018-12-25 Covidien Lp Endoscopic reposable surgical clip applier
US10160761B2 (en) 2015-09-14 2018-12-25 Infinity Pharmaceuticals, Inc. Solid forms of isoquinolinones, and process of making, composition comprising, and methods of using the same
US11939333B2 (en) 2015-09-14 2024-03-26 Infinity Pharmaceuticals, Inc. Solid forms of isoquinolinones, and process of making, composition comprising, and methods of using the same
US11247995B2 (en) 2015-09-14 2022-02-15 Infinity Pharmaceuticals, Inc. Solid forms of isoquinolinones, and process of making, composition comprising, and methods of using the same
US11298135B2 (en) 2015-11-10 2022-04-12 Covidien Lp Endoscopic reposable surgical clip applier
US10390831B2 (en) 2015-11-10 2019-08-27 Covidien Lp Endoscopic reposable surgical clip applier
US10759806B2 (en) 2016-03-17 2020-09-01 Infinity Pharmaceuticals, Inc. Isotopologues of isoquinolinone and quinazolinone compounds and uses thereof as PI3K kinase inhibitors
US10385131B2 (en) 2016-05-11 2019-08-20 Huya Bioscience International, Llc Combination therapies of HDAC inhibitors and PD-L1 inhibitors
US11535670B2 (en) 2016-05-11 2022-12-27 Huyabio International, Llc Combination therapies of HDAC inhibitors and PD-L1 inhibitors
US10385130B2 (en) 2016-05-11 2019-08-20 Huya Bioscience International, Llc Combination therapies of HDAC inhibitors and PD-1 inhibitors
US10287353B2 (en) 2016-05-11 2019-05-14 Huya Bioscience International, Llc Combination therapies of HDAC inhibitors and PD-1 inhibitors
US10668167B2 (en) 2016-06-02 2020-06-02 Abbvie Inc. Glucocorticoid receptor agonist and immunoconjugates thereof
US10919914B2 (en) 2016-06-08 2021-02-16 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
US10863992B2 (en) 2017-08-08 2020-12-15 Covidien Lp Endoscopic surgical clip applier
US10772970B2 (en) 2017-12-01 2020-09-15 Abbvie Inc. Glucocorticoid receptor agonist and immunoconjugates thereof
EP3927383A1 (en) * 2019-02-18 2021-12-29 Lankenau Institute for Medical Research Scar reducing wound closure materials
WO2020172113A1 (en) * 2019-02-18 2020-08-27 Lankenau Institute For Medical Research Scar reducing wound closure materials
EP3927383A4 (en) * 2019-02-18 2022-11-09 Lankenau Institute for Medical Research Scar reducing wound closure materials
WO2021231563A1 (en) * 2020-05-13 2021-11-18 Spiral Therapeutics Inc. Gelling solutions for administration of compounds to the inner ear

Also Published As

Publication number Publication date
WO2006135479A3 (en) 2007-04-12

Similar Documents

Publication Publication Date Title
WO2006135479A2 (en) Anti-scarring agents, therapeutic compositions, and use thereof
US20050183731A1 (en) Polymer compositions and methods for their use
US20070208134A1 (en) Anti-scarring drug combinations and use thereof
US20070299043A1 (en) Anti-scarring drug combinations and use thereof
AU2004293071A1 (en) Polymer compositions and methods for their use
US20050149080A1 (en) Medical implants and anti-scarring agents
CN101420991A (en) Polymer compositions and methods for their use
US20050169958A1 (en) Medical implants and fibrosis-inducing agents
CN101420923A (en) Intravascular devices and fibrosis-inducing agents
WO2006121522A2 (en) Implantable sensors and pumps, anti-scarring agents, and therapeutic compositions
WO2006121521A2 (en) Soft tissue implants, anti-scarring agents, and therapeutic compositions

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application
NENP Non-entry into the national phase in:

Ref country code: DE

NENP Non-entry into the national phase in:

Ref country code: RU

122 Ep: pct application non-entry in european phase

Ref document number: 06799901

Country of ref document: EP

Kind code of ref document: A2